• Non ci sono risultati.

The Fas/FasL Signaling PathwayMaria Eugenia Guicciardi, Gregory J. Gores

N/A
N/A
Protected

Academic year: 2022

Condividi "The Fas/FasL Signaling PathwayMaria Eugenia Guicciardi, Gregory J. Gores"

Copied!
10
0
0

Testo completo

(1)

The Fas/FasL Signaling Pathway

Maria Eugenia Guicciardi, Gregory J. Gores

11

11.1

Introduction

Fas (also called CD95 or APO-1) belongs to the death receptor family, a subgroup of the tumor necrosis factor/nerve growth factor (TNF/NGF) receptor superfamily. These cell surface cytokine receptors are able to initiate an apoptotic signaling cascade after binding a group of structurally related ligands or specific antibodies [3, 40]. The members of this family are type I transmembrane proteins with a C- terminal intracellular tail, a membrane-spanning region, and an extracellular N-terminal domain.

Through interaction with the N-terminal domain, the receptors bind their cognate ligands (called death ligands), the majority of which are type II transmembrane proteins belonging to the TNF fam- ily of proteins, and comprised of an intracellular N-terminal domain, a transmembrane region, and a C-terminal extracellular tail. The signature features of the death receptors are represented by a highly homologous region in their extracellular domains containing one to five cysteine-rich repeats, and a 60- to 80-amino acid cytoplasmic sequence known as death domain (DD), which is required to initiate the death signal.

Engagement of death receptors results in initia- tion of the so-called extrinsic pathway of apoptosis, one of the two main signaling pathways leading to apoptotic cell death. The second one is generated by a mitochondrial dysfunction, and is referred to as the intrinsic pathway [23] (Fig. 11.1). Although both signaling pathways are sufficient to trigger apopto- sis, the two are not mutually exclusive and can be simultaneously activated in the same cell through crosstalk between pathways, especially in hepato- cytes.

Apoptosis is essential to preserve liver func- tion and health, as it ensures the efficient removal of unwanted cells (i.e., aged or virus-infected cells) in a highly controlled manner. Apoptotic cells are ultimately fragmented into membrane-bound, or- ganelle-containing corpses (apoptotic bodies),

which are readily engulfed by neighboring phago- cytes, mainly Kuppfer cells; this engulfment proc- ess may, under pathologic conditions, promote liver inflammation and damage by amplifying Fas-me- diated hepatocyte apoptosis through FasL produc- tion by the Kuppfer cells themselves (Fig. 11.2) [9].

Fig. 11.1. Apoptotic pathways. Apoptosis can be induced via the activation of the extrinsic or the intrinsic pathway. Death receptor engagement mediates the extrinsic pathway through the recruitment of adaptor proteins and the formation of the DISC. Initiator caspases (caspase-8 and -10) activated at the DISC directly cleave and activate the effector caspases (caspase-3, - 6, and -7) responsible for the degradation of cellular substrates.

Different stimuli, including growth factor deprivation, DNA damage, and UV light exposure, trigger the intrinsic pathway via the activation of proapoptotic members of the Bcl-2 family of protein (i.e., Bax, Bak), which cause mitochondrial dysfunction.

The action of the proapoptotic members of the Bcl-2 family can be antagonized by anti-apoptotic members of the same family (Bcl-2, Bcl-XL). Following mitochondrial dysfunction, caspase-9, another initiator caspase, is activated and, in turn, activates the effector caspases. See text for details

(2)

Although apoptosis in the liver can occur through activation of both the extrinsic and the intrinsic pathway, the extrinsic pathway seems to be by far the most relevant, likely due to the high level of ex- pression of death receptors in hepatic cells. In par- ticular, Fas is constitutively expressed by every cell type in the liver [15], rendering all liver cells sensi- tive to Fas-mediated apoptosis in vivo. Indeed, Fas- induced apoptosis plays a fundamental role in liver physiology by contributing to the elimination of se- nescent cells and maintaining liver homeostasis [1], as well as in pathologic conditions, by ensuring the removal of virus-infected or damaged cells [33, 41].

However, excessive or defective Fas-mediated apop- tosis leads to disease pathogenesis, such as liver fail- ure, fibrosis, and carcinogenesis. In this chapter, we review the molecular mechanisms and regulation of Fas signaling, and the role of the Fas/FasL system in the pathophysiology of the liver.

11.2

Fas (CD95/APO-1)

and Fas Ligand (FasL/CD95L)

11.2.1

Fas (CD95/APO-1)

Fas is a glycosylated cell-surface protein, ubiqui- tously expressed in various tissues, in particular in thymus, liver, heart, kidney, and in activated mature lymphocytes or virus-transformed lymphocytes.

Although soluble forms of the receptor also exist, whose functions are still largely unknown, the mem- brane-bound form is largely predominant and high- ly biologically active [10]. In order to avoid unneces- sary activation of the apoptotic pathway, Fas expres- sion and localization are tightly regulated through a variety of mechanisms. First of all, only a minimal amount of Fas is expressed on the plasma mem- brane in unstimulated cells, whereas the majority of the receptor localizes in the cytosol, in particular, in the Golgi complex and the trans-Golgi network [6, 66]. After a proapoptotic stimulus, Fas-containing vesicles translocate to the cell surface, increasing Fas expression on the plasma membrane and initiat- ing the apoptotic signal. This mechanism provides an effective tool to regulate the plasma membrane density of the death receptor, and avoid its sponta- neous activation [19, 66]. Fas can also be modulated at a post-translational level, by glycosylation of the receptor [60], as well as at the transcriptional level, by direct regulation of Fas expression. Indeed, a composite binding site for the transcription factor NF- κB is located in the Fas gene promoter [11], and a p53-responsive element has been identified within the first intron of the Fas gene, which cooperates with three sequences in the promoter to upregulate Fas receptor expression [20, 50, 51].

11.2.2

Fas Ligand (FasL/CD95L)

FasL (CD95L) is a type II transmembrane protein with homotrimeric structure, mainly expressed on the cell surface of activated T cells [70]. In the liver, the interaction between FasL-positive cytotoxic T lymphocytes and target cells, such as virus-infected cells or cancer cells, which usually overexpress Fas, represents a powerful tool to eliminate potentially toxic cells [7, 33, 41]. Kupffer cells, liver-specific phagocytes, can also express FasL and induce hepa- tocyte apoptosis [9, 52, 72]. Moreover, hepatocytes themselves overexpress FasL in certain pathological

Fig. 11.2. A vicious cycle of apoptosis. Schematic representa- tion of the amplification of Fas/CD95-mediated hepatocyte apoptosis by Kupffer cell-generated FasL/CD95L. Apoptotic hepatocytes fragment into apoptotic bodies. Engulfment of apoptotic bodies by Kupffer cells induces their activation and the production of FasL/CD95L, which, in turn, exacerbates the apoptotic damage

(3)

conditions, such as alcoholic hepatitis and Wilson's disease; this can bind Fas constitutively expressed on the same cell, and induce apoptosis via fratri- cide [21]. FasL also exists in a soluble, trimeric form generated after cleavage by a metalloprotease, but, similarly to the soluble form of Fas, its biological ac- tivity remains controversial [63]. Indeed, although the ability of soluble FasL to induce apoptosis has been documented, high serum levels of FasL often reported in hepatitis, AIDS and several types of tu- mor do not seem to correlate with increased apop- totic damage [74]. The explanation for this apparent contradiction has been provided by recent studies demonstrating that the apoptotic-inducing capac- ity of the soluble form is reduced by over 1,000-fold compared to the membrane-bound FasL [63, 65, 71].

Therefore, even elevated serum levels of soluble FasL might not be sufficient to cause significant apopto- sis. However, the binding of soluble FasL to some matrix proteins (i.e., fibronectin) has been shown to induce trimerization of the ligand, therefore effec- tively mimicking the membrane-bound form [2].

11.3

Fas/FasL Signaling

Engagement of Fas by either agonistic antibodies or FasL leads to the trimerization of the receptor, fol- lowed by recruitment of the adaptor molecule FADD (Fas-associated protein with death domain)/MORT- 1 (mediator of receptor-induced toxicity), a cytosol- ic protein with a C-terminal death domain, and a death effector domain at the N-terminus [78]. FADD

associates with the receptor through interaction of its death domains, while its death effector domain binds to a correspondent death effector domain or a caspase recruitment domain (CARD) in the pro- domain of inactive initiator caspases, such as pro- caspase-8 and procaspase-10. The resulting com- plex is called the death-inducing signaling complex (DISC) (Fig. 11.3). Recruitment and accumulation of procaspase-8 and/or -10 at the DISC results in self- processing, spontaneous activation of the caspase via autoproteolytic cleavage, and initiation of a pro- teolytic cascade. Procaspase-8 and -10 are proteo- lytically processed with similar kinetics, and both can initiate apoptosis independently of each other, as well as participate in the same apoptotic pathway [36, 79]. A family of proteins of viral origin called v-FLIPs (viral FLICE-inhibitory proteins) has been found to exert an anti-apoptotic activity by inhibit- ing the activation of effector caspases at the DISC.

v-FLIPs contain two death effector domains that en- able them to bind to FADD and block caspase-8 ac- tivation [46]. It has not yet been established whether they also inhibit caspase-10 activation. The human cellular homolog, c-FLIP (also called I-FLICE or Casper or Usurpin) [76], exists in a short and a long isoform, as a result of different splice variants. The short form, c-FLIP

S

, consists only of two death ef- fector domains, and structurally resembles v-FLIP.

The long form, c-FLIP

L

consists of two death effec- tor domains and a caspase-like domain, and closely resembles caspase-8, except that it contains an inac- tive enzymatic site, and, therefore, has no cysteine protease activity. Both forms of c-FLIP are recruited and bind to the DISC upon stimulation [62], but while c-FLIP

S

may competitively inhibit procaspase-

Fig. 11.3. The Fas/CD95 death-inducing signaling complex (DISC). Schematic rep- resentation of the DISC formed after the engagement of FasL/CD95L to Fas/CD95.

The death domain (DD) on the adapter protein FADD interacts with the receptor’s death domain, whereas the death effector domain (DED) binds the correspondent death effector domain in the pro-domain of the inactive initiator caspase-8 and/or -10. The short form of the inhibitor of ap- optosis cFLIP (cFLIPS) binds to the DED of FADD and prevents the recruitment of the initiator pro-caspases to the DISC, whereas the long form of cFLIP (cFLIPL) allows the recruitment of procaspase-8 to the DISC, but inhibits its processing into the active form. See text for details

(4)

8 recruitment to the DISC, c-FLIP

L

allows recruit- ment and even partial cleavage of procaspase-8.

However, by mechanisms yet to be clarified, c-FLIP

L

prevents further proteolytic processing of caspase-8 to generate the active subunits of the enzyme [37]

(Fig. 11.3).

Downstream of the DISC formation, activation of effector caspases, such as caspase-3, -6 and -7, which are ultimately responsible for the degrada- tion of key cellular components, can occur via two different signaling pathways. Based on the signaling pathway preferentially activated after Fas stimula- tion, cells have been classified into type I and type II [61] (Fig. 11.4). In type I cells, large amounts of caspase-8 are activated at the DISC, which, in turn, directly cleave and activate caspase-3. In these cells, prevention of mitochondrial dysfunction by over- expression of the anti-apoptotic proteins Bcl-2 or Bcl-X

L

does not block the activation of caspase-8 or caspase-3, nor does it inhibit apoptosis, suggesting a mitochondria-independent activation of a caspase cascade. In contrast, in type II cells, DISC forma-

tion is strongly reduced, and activation of caspases, including caspase-8, occurs mainly downstream of mitochondria, as both caspase activation and apop- tosis can be prevented by overexpression of Bcl-2 or Bcl-X

L

. Notably, Fas induces mitochondrial dysfunc- tion in both type I and type II cells, but only in type II cells are mitochondria essential for the execution of the apoptotic program, whereas in type I cells they likely function solely as amplifiers of the apoptotic signal [61]. Mitochondrial dysfunction during Fas signaling is initiated by caspase-8-mediated cleav- age of Bid, a pro-apoptotic, BH3-only member of the Bcl-2 family of proteins [39, 43]. The generated 15- kDa fragment (tBid) translocates to the mitochon- dria, and contributes to the formation of pores on the outer mitochondrial membrane, resulting in the release of apoptogenic factors, such as cytochrome c [39, 43], AIF (apoptosis-inducing factor) [73], and SMAC (second mitochondria-derived activator of caspases)/Diablo (direct IAP-binding protein with low pI) [14, 77]. In the cytosol, cytochrome c associ- ates with the cofactor Apaf-1 (apoptosis-activating factor 1) and procaspase-9, and forms a complex named apoptosome. Through an energy-requiring reaction, procaspase-9 in the apoptosome is proc- essed into the mature enzyme and, in turn, activates caspase-3, starting a caspase cascade downstream of the mitochondrium.

11.4

Fas/FasL in Liver Diseases

Dysregulation of hepatocyte apoptosis often associ- ates with liver diseases (Fig. 11.5). Downregulation of apoptosis leads to diseases associated with exces- sive cell growth, such as hepatocellular carcinoma.

On the contrary, excessive hepatocyte apoptosis is a feature of viral and autoimmune hepatitis, acute hepatic failure, cholestatic diseases, alcoholic and non-alcoholic hepatitis, chemotherapeutic-induced liver damage, as well as transplantation-associated liver damage, such as ischemia/reperfusion injury and graft rejection. The role of the Fas/FasL system in several human liver diseases associated with dis- ruption of apoptosis is described in greater detail in this section.

Fig. 11.4. Fas/CD95-mediated apoptotic pathways. Schematic representation of Fas-mediated apoptotic pathways in type I and type II cells. The amount of active caspase-8 generated at the DISC determines whether the cell activates a mitochondrial- independent (type I) or mitochondrial-dependent (type II) path- way of caspase activation and apoptosis. See text for details

(5)

11.4.1

Pathologic Conditions Associated with Reduced Fas-mediated Apoptosis

Hepatocellular Carcinoma

Hepatocellular carcinoma, the most common pri- mary malignancy of the liver, has multiple etiolo- gies, including environmental, nutritional, and met- abolic factors, as well as chronic viral infections. The role of Fas as an anticarcinogenic agent is suggested by the evidence that Fas-defective animals show increased risk of developing tumors. Consistently, Fas expression is downregulated in several tumors, including hepatocellular carcinoma, which origi- nate from tissue that previously expressed normal levels of Fas [28, 38]. Reductions of Fas expression of various extents have been described in hepatocar- cinomas, which inversely correlate with the severity of the disease. Indeed, the most significant reduc- tions in Fas expression are found in poorly differ- entiated, advanced carcinomas, and negatively cor- relate with patient survival [32, 53, 68]. The loss of Fas represents an advantageous adaptation for the cancer cell, because it allows the cell to survive the attack by FasL-expressing cytotoxic T lymphocytes and NK cells. In addition, tumor cells often express FasL, which enables them to actively kill the im- mune cells and create immune privileged sites [24, 27, 68]. Therefore, therapeutic approaches aimed to restore Fas expression and sensitivity to Fas-medi- ated apoptosis in tumor cells may be effective in the therapy of hepatocellular carcinomas. Indeed, several chemotherapeutic drugs induce tumor cell apoptosis by causing DNA damage and activation of the nuclear phosphoprotein p53, which, in turn, has been found to upregulate Fas and increase sen- sitivity to Fas-mediated apoptosis [50, 51]. However, a study recently performed using an experimental mouse model of chronic hepatitis-induced hepato- cellular carcinoma has demonstrated that inhibi- tion of FasL may actually reduce the development of cancer [54]. FasL has a proinflammatory activity [34, 64], and a status of continuous inflammation leads to massive cells loss and liver regeneration that, in turn, may significantly increase the chance of mutagenic events. Therefore, FasL is not only di- rectly responsible for hepatocyte killing, but is also indirectly involved in chronic liver dysplasia and hepatocellular carcinoma development by inducing an inflammatory response. Thus, early in the dis- ease process, inhibiting FasL and its inflammatory signaling would prevent the milieu necessary for carcinogenesis to occur. These new findings have to

be considered in order to design a better therapeutic approach for hepatocellular carcinomas.

11.4.2

Pathologic Conditions Associated with Excessive Fas-mediated Apoptosis

Viral Hepatitis

Viral hepatitis is mainly caused by infection with hepatitis B (HBV) or C virus (HCV). However, the virus itself has very mild cytopathic effects on the infected host cells, and the extensive tissue damage associated with viral hepatitis is generally the result of host immune response to viral antigen. During viral hepatitis, specific classes of cytotoxic T lym- phocytes (CTL) recognize and kill viral antigen-ex- pressing HBV- or HCV-infected hepatocytes to clear the virus from the liver. This causes the initial liver damage, which is subsequently exacerbated by the influx of antigen-non-specific inflammatory cells.

The killing of viral antigen-positive hepatocytes by CTL occurs via apoptosis, as demonstrated by the presence of apoptotic bodies, once referred to

Fig. 11.5. Role of Fas/CD95-mediated apoptosis in the patho- genesis of liver diseases. Schematic representation of the role of Fas/CD95-mediated apoptosis in liver health and disease. Under physiological conditions, the number of cells killed by apoptosis equals the number of cells produced by cell division. Imbalance between cell production and cell death leads to different liver diseases. See text for details

(6)

as Councilman bodies, in the liver of patients with viral hepatitis. In particular, Fas, although not the only apoptotic pathway involved, seems to play a key role in this process. Indeed, Fas expression is increased in the liver of patients with chronic hepa- titis B and C, and directly correlates with disease activity such as periportal and intralobular inflam- mation [21, 31, 42, 47, 49, 80]. It is not clear whether Fas expression is mainly regulated by virus-specific protein expression or by inflammatory cytokines, such as interleukin-1, generated after the first im- mune response. Areas of FasL-positive infiltrating mononuclear cells are also common in the liver of HBV- and HCV-infected patients, confirming the importance of the Fas/FasL system in the removal of infected cells by CTL during viral hepatitis [21, 31, 47, 49, 80]. Nevertheless, the role of the HBV and HCV proteins in Fas-mediated apoptosis remains controversial. The HBV X-gene product (HBx) has been shown to stimulate the apoptotic turnover of hepatocytes [75], as well as to activate NF- κB and c-Jun N-terminal kinase (JNK) pathways and there- fore to protect liver cells from apoptosis [13, 57].

Similarly, HCV proteins inhibit Fas-mediated ap- optosis and death in transgenic mice by preventing the release of cytochrome c from the mitochondria [44]. Therefore, hepatitis virus proteins may either sensitize the hepatocyte to Fas-induced apoptosis or inhibit apoptosis to maintain persistent infection.

Alcoholic Hepatitis

Although the pathogenesis of alcoholic hepatitis and cirrhosis is still poorly understood, apopto- sis certainly plays an important role both in vitro and in vivo. Apoptosis is a characteristic feature of experimental ethanol-induced liver injury [5, 22].

Moreover, hepatocyte apoptosis has recently been identified in liver biopsies of patients with alcoholic hepatitis, with a correlation with the disease sever- ity, being most abundant in patients with high bi- lirubin and AST levels, and grade 4 steatohepatitis [35, 56]. Among the several mechanisms proposed to explain alcohol-induced hepatocyte apoptosis, there is the activation of death receptor pathways, in particular, the Fas/FasL and TNF- α/TNF-R1 signal- ing. Patients with alcoholic hepatitis express higher levels of Fas and FasL in the hepatocytes compared to healthy subjects, which renders the cells more susceptible both to cytotoxic T lymphocyte-medi- ated apoptosis, and to cell death by autocrine and/

or paracrine mechanisms [56]. The increased ex- pression of Fas and FasL may result from TNF- α- induced activation of NF- κB, a transcription factor that can upregulate both these genes [11]. Indeed, TNF- α serum levels are elevated during alcoholic

hepatitis, and are directly involved in hepatocyte apoptosis [45]. Moreover, a recent study suggested that, in addition to a direct cytotoxic effect on the hepatocyte, the TNF- α/TNF-R1 system is required also for Fas-mediated cell death, as demonstrated by the increased resistance of TNF-R1/TNF-R2 double knockout mice to Fas-induced fulminant liver in- jury [12]. Thus, it appears that both Fas and TNF-R1 contribute to ethanol-mediated liver injury through a synergistic action in inducing hepatocyte apopto- sis.

Cholestatic Liver Disease

Cholestasis is defined as an impairment of bile flow through the liver. As a consequence, high concen- trations of bile acids accumulate within the hepato- cytes, causing tissue damage and liver failure. Sever- al studies demonstrated that one of the mechanisms by which bile acids, especially hydrophobic bile ac- ids, induce liver damage is by triggering hepatocyte apoptosis. Indeed, hydrophobic bile acids, such as deoxycholic and glycodeoxycholic acid, are able to cause hepatocyte apoptosis in vitro [16, 26, 30, 58, 66]. More remarkably, massive hepatocyte apoptosis is clearly detectable in the liver of bile duct-ligated mice, an animal model of extrahepatic cholestasis [48]. Although bile acids have detergent properties and could potentially damage the cell membranes, they actually need to be transported into the cell to trigger apoptosis, as cells lacking a functional bile acid transporter are resistant to bile acid-induced apoptosis [25]. It has been shown that elevated con- centrations of bile acids within the hepatocyte can induce Fas translocation from its intracellular loca- tions to the plasma membrane, where the increased surface density triggers its oligomerization and ini- tiates the apoptotic signal [66]. Indeed, bile acid-in- duced apoptosis largely occurs via a Fas-dependent, FasL-independent mechanism, both in vitro [16], and in vivo [48]. In addition, a recent study dem- onstrated that, in a model of chronic cholestasis, Fas-mediated cytoxicity promotes the development of liver fibrosis, the result of excessive deposition of extracellular matrix during the wound-healing re- sponse that follows a prolonged injury to the liver [8]. In the absence of Fas, long-term, bile duct-ligat- ed mice showed reduced markers of fibrosis, such as expression of α-smooth muscle actin and collagen deposition, as compared to Fas-expressing animals, suggesting that inhibition of Fas-mediated hepato- cyte apoptosis may prevent liver fibrogenesis.

Although Fas plays a major role in executing

bile acid-mediated apoptosis, other pathways are

also likely to be involved [48]. Recent reports have

demonstrated that, in the absence of Fas, bile acids

(7)

increase the transcription and oligomerization of another death receptor, TRAIL-R2/DR5, suggesting that bile acid cytotoxicity can also be mediated by TRAIL/TRAIL-R2 signaling pathway [29]. Both Fas and TRAIL-R2 signal apoptosis through activation of caspase-8/-10 and Bid, therefore targeted inhibi- tion of caspases or Bid could have therapeutic rel- evance in the treatment of cholestatic liver diseases.

Indeed, inhibition of Bid by injection of antisense oligonucleotides has already been demonstrated to reduce hepatocyte apoptosis and liver damage in bile duct-ligated mice [30].

Wilson's Disease

Wilson's disease is a genetic disorder caused by excessive copper storage in different organs and tissues, including the liver. Liver sections from pa- tients with Wilson's disease show significant hepa- tocyte apoptosis associated with upregulation of Fas and FasL on the hepatocyte cell membrane [69].

Similarly, hepatocyte apoptosis and Fas expression have been found to be increased in a model of cop- per overload in vitro [69]. As already suggested in alcoholic hepatitis, the simultaneous expression of Fas and FasL on the same cell membrane may pro- mote fratricide killing of neighboring cells. Copper accumulation within the hepatocyte causes oxida- tive stress, which, in turn, may promote Fas activa- tion and apoptosis [4]. The upregulation of Fas like- ly occurs via the activation of the tumor suppressor gene p53, which follows the oxidative stress-induced DNA damage. Indeed, treatment of hepatoma cells with copper results in a transient increase in p53 and Fas expression, the latter being a consequence of p53 transcriptional activity [55, 69]. Inhibition of either FasL or caspases effectively reduces apopto- sis with similar results, suggesting that Fas might be the only apoptotic signal involved in copper-in- duced apoptosis. Therefore therapies aimed to in- hibit either Fas or FasL or caspases might be useful in the treatment of Wilson's disease and could re- duce the need for transplantation in the acute form of this disease.

Non-alcoholic Steatohepatitis

Non-alcoholic steatohepatitis (NASH) is the most severe form of non-alcoholic fatty liver disease (NAFLD), characterized by the presence of mac- rovesicular steatosis along with inflammatory ac- tivity, and sometimes associated with fibrosis. The molecular mechanisms involved in tissue damage during NASH are poorly understood. However, it has recently been demonstrated that Fas expression, activation of caspase-3 and -7 and hepatocyte ap-

optosis are enhanced in the liver of NASH patients, and positively correlated with the biochemical and histopathologic markers of liver injury [17, 18]. Mi- tochondrial function is often impaired in the liver of subjects with NASH [59]. Activation of Fas results in mitochondrial dysfunction as a consequence of the activation of Bid and its translocation to the mi- tochondria. Moreover, mitochondrial dysfunction is associated with generation of reactive oxygen, which is also able to induce apoptosis, further ex- acerbating tissue injury and inflammation. Thus, Fas inhibition may be an effective therapy to reduce liver damage and prevent development of cirrhosis in NASH.

11.5 Conclusions

The Fas/FasL system plays a key role in maintaining liver homeostasis and function through regulation of cell death and survival. Indeed, several liver dis- eases are associated with either Fas overexpression or downregulation, which directly correlates with the onset of the disease and its severity. Hepatocyte apoptosis often represents the early stage of many liver diseases, independent of their etiology, and it occurs mainly through engagement of death recep- tors on the plasma membrane, especially Fas. Un- controlled hepatocyte apoptosis can progress into liver injury if the number of cells dying is signifi- cantly higher than the number of cells replaced by cell division (Fig. 11.5). Moreover, if the amount of apoptotic bodies overwhelms the clearance capac- ity by phagocytes, the apoptotic bodies undergo a process of autolysis resulting in the release of pro- inflammatory factors, which exacerbate the inflam- matory response and tissue damage. In addition, he- patic stellate cells are activated by the phagocytosis of apoptotic bodies, and generate a pro-fibrogenic response by promoting collagen deposition in the liver parenchyma and formation of fibrotic tissue.

In conclusion, therapeutic approaches aimed to

modulate Fas-mediated apoptosis may ultimately

be effective in reducing liver damage in several hu-

man liver diseases. Preliminary studies on animal

models of liver injury have already generated prom-

ising data regarding the feasibility and effectiveness

of genetic inhibition of Fas as a possible therapy to

prevent fulminant liver failure [67, 81].

(8)

Acknowledgments

This work was supported by grants from the Na- tional Institute of Health DK 41876 (to GJG) and the Mayo Foundation, Rochester, Minnesota.

Selected Reading

The following readings are suggested to help un- derstand the signal transduction through the death receptors and the fundamental role of Fas in liver physiology and pathophysiology.

Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF recep- tor superfamily: integrating mammalian biology. Cell 2001;104:487–501. [40]

Scaffidi C, Fulda S, Srinivasan A et al. Two CD95 (APO-1/Fas) sign- aling pathways. EMBO J 1998;17:1675–1687. [61]

Faubion WA, Gores GJ. Death receptors in liver biology and pathobiology. Hepatology 1999;29:1–4. [15]

Galle PR, Hofmann WJ, Walczak H et al. Involvement of the APO- 1/Fas (CD95) receptor and ligand in liver damage. J Exp Med 1995;182:1223–1230. [21]

References

1. Adachi M, Suematsu S, Kondo T et al. Targeted mutation in the Fas gene causes hyperplasia in peripheral lymphoid or- gans and in the liver. Nat Genet 1995;11:294–300.

2. Aoki K, Kurooka M, Chen J-J et al. Extracellular matrix inter- acts with soluble CD95L: retention and enhancement of cy- totoxicity. Nat Immunol 2001;2:333–337.

3. Ashkenazi A, Dixit VM. Apoptosis control by death and de- coy receptors. Curr Opin Cell Biol 1999;11:255–260.

4. Aust SD, Morehouse LA, Thomas CE. Role of metals in oxy- gen radical reactions. J Free Radic Biol Med 1985;1:3–25.

5. Benedetti A, Brunelli E, Risicate R et al. Subcellular chang- es and apoptosis induced by ethanol in rat liver. J Hepatol 1988;6:137–143.

6. Bennet M, MacDonald K, Chan S-W et al. Cell surface traffick- ing of Fas: a rapid mechanism of p53-mediated apoptosis.

Science 1998;282:290–293.

7. Berke G. The CTL's kiss of death. Cell 1995;81:9–12.

8. Canbay A, Higuchi H, Bronk SF et al. Fas enhances fibrogen- esis in the bile duct ligated mouse: a link between apoptosis and fibrosis. Gastroenterology 2002;123:1323–1330.

9. Canbay A, Feldstein AE, Higuchi H et al. Kupffer cell engulf- ment of apoptotic bodies stimulates death ligand and cy- tokine expression. Hepatology 2003;38:1188–1198.

10. Cascino I, Fiucci G, Papoff G et al. Three functional soluble forms of the human apoptosis-inducing Fas molecule are

produced by alternative splicing. J Immunol 1995;154:2706–

2713.

11. Chan H, Bartos DP, Owen-Schaub LB. Activation-depend- ent transcriptional regulation of the human Fas promoter requires NF-kappaB p50-p65 recruitment. Mol Cell Biol 1999;19:2098–2108.

12. Costelli P, Aoki P, Zingaro B et al. Mice lacking TNF recep- tors 1 and 2 are resistant to death and fulminant liver injury induced by agonistic anti-Fas antibody. Cell Death Differ 2003;10:997–1004.

13. Diao J, Khine AA, Sarangi F et al. X protein of hepatitis B virus inhibits Fas-mediated apoptosis and is associated with up-regulation of the SAPK/JNK pathway. J Biol Chem 2001;276:8328–8340.

14. Du C, Fang M, Li Y et al. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000;102:33–42.

15. Faubion WA, Gores GJ. Death receptors in liver biology and pathobiology. Hepatology 1999;29:1–4.

16. Faubion WA, Guicciardi ME, Miyoshi H et al. Toxic bile salts induce rodent hepatocyte apoptosis via direct activation of Fas. J Clin Invest 1999;103:137–145.

17. Feldstein AE, Canbay A, Angulo P et al. Hepatocyte apoptosis and Fas expression are prominent features of human non- alcoholic steatohepatitis. Gastroenterology 2003;125:437–

443.

18. Feldstein AE, Canbay A, Guicciardi ME et al. Diet associated hepatic steatosis sensitizes to Fas mediated liver injury in mice. J Hepatol 2003;39:978–983.

19. Feng G, Kaplowitz N. Colchicine protects mice from the lethal effect of an agonistic anti-Fas antibody. J Clin Invest 2000;105:329–339.

20. Friesen C, Herr I, Krammer PH et al. Involvement of the CD95 (APO-1/Fas) receptor/ligand system in drug-induced apop- tosis in leukemia cells. Nature Med 1996;2:574–580.

21. Galle PR, Hofmann WJ, Walczak H et al. Involvement of the APO-1/Fas (CD95) receptor and ligand in liver damage. J Exp Med 1995;182:1223–1230.

22. Goldin RD, Hunt NC, Clark J et al. Apoptotic bodies in a murine model of alcoholic liver disease. J Pathol 1993;171:73–76.

23. Green DR, Reed JC. Mitochondria and apoptosis. Science 1998;281:1309–1312.

24. Griffith TS, Brunner T, Fletcher SM et al. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science 1995;17:1189–1192.

25. Guicciardi ME, Faubion WA, Bronk SF et al. Mechanisms of bile acid-induced cell death. In: Andus T, Rogler G, Schlott- mann K et al, eds. Cytokines and cell homeostasis in the gastrointestinal tract. Dordrecht/Boston/London: Kluwer Academic Publishers, 2000:284–289.

26. Guicciardi ME, Gores GJ. Bile acid-mediated hepatocyte apoptosis and cholestatic liver disease. Digest Liver Dis 2002;34:387–392.

27. Hahne M, Rimoldi D, Schroter M et al. Melanoma cell expres- sion of Fas(Apo-1/CD95) ligand: implications for tumor im- mune escape. Science 1996;274:1363–1366.

(9)

28. Higaki K, Yano H, Kojiro M. Fas antigen expression and its relationship with apoptosis in human hepatocellular carci- noma and noncancerous tissues. Am J Pathol 1996;149:429–

437.

29. Higuchi H, Bronk SF, Takikawa Y et al. The bile acid glycoche- nodeoxycholate induces trail-receptor 2/DR5 expression and apoptosis. J Biol Chem 2001;276:38610–38618.

30. Higuchi H, Miyoshi H, Bronk SF et al. Bid antisense attenu- ates bile acid-induced apoptosis and cholestatic liver injury.

J Pharmacol Exp Ther 2001;299:866–873.

31. Hiramatsu N, Hayashi N, Katayama K et al. Immunohisto- chemical detection of Fas antigen in liver tissue of patients with chronic hepatitis C. Hepatology 1994;19:1354–1359.

32. Ito Y, Takeda T, Umeshita K et al. Fas antigen expression in hepatocellular carcinoma tissues. Oncol Rep 1998;5:41–44.

33. Kagi D, Vignaux F, Ledermann B et al. Fas and perforin path- ways as major mechanisms of T-cell-mediated cytotoxicity.

Science 1994;265:528–530.

34. Kang SM, Schneider DB, Lin Z et al. Fas ligand expression in islets of Langerhans does not confer immune privilege and instead targets them for rapid destruction. Nat Med 1997;3:738–743.

35. Kawahara H, Matsuda Y, Takase S. Is apoptosis involved in alcoholic hepatitis? Alcohol 1994;29:113–118.

36. Kischkel FC, Lawrence DA, Tinel A et al. Death recep- tor recruitment of endogenous caspase-10 and apop- tosis initiation in the absence of caspase-8. J Biol Chem 2001;276:46639–46646.

37. Krueger A, Baumann S, Krammer PH et al. FLICE-inhibitory proteins: regulators of death receptor-mediated apoptosis.

Mol Cell Biol 2001;21:8247–8254.

38. Leithauser F, Dhein J, Mechtersheimer G et al. Constitutive and induced expression of APO-1, a new member of the NGF/TNF receptor superfamily, in normal and neoplastic cells. Lab Invest 1993;69:415–429.

39. Li H, Zhu H, Xu CJ et al. Cleavage of BID by caspase 8 medi- ates the mitochondrial damage in the Fas pathway of apop- tosis. Cell 1998;94:491–501.

40. Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF re- ceptor superfamily: integrating mammalian biology. Cell 2001;104:487–501.

41. Lowin B, Hahne M, Mattmann C et al. Cytolytic T-cell cytotox- icity is mediated through perforin and Fas lytic pathyways.

Nature 1994;370:650–652.

42. Luo KX, Zhu YF, Zhang LX et al. In situ investigation of Fas/

FasL expression in chronic hepatitis B virus infection and its related liver diseases. J Viral Hep 1997;4:303–307.

43. Luo X, Budihardjo I, Zou H et al. Bid, a Bcl2 interacting pro- tein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 1998;94:481–490.

44. Machida K, Tsukiyama-Kohara K, Seike E et al. Inhibition of cytochrome c release in Fas-mediated signaling pathway in transgenic mice induced to express hepatitis C viral pro- teins. J Biol Chem 2001;276:12140–12146.

45. McClain C, Hill D, Schmidt J et al. Cytokines and alcoholic liver disease. Semin Liver Dis 1993;13:170–182.

46. Meinl E, Fickenscher H, Thome M et al. Anti-apoptotic strate- gies of lymphotropic viruses. Immunol Today 1998;19:474–

479.

47. Mita E, Hayashi N, Iio S et al. Role of Fas ligand in apoptosis induced by hepatitis C virus infection. Biochem Biophys Res Com 1994;204:468–474.

48. Miyoshi H, Rust C, Roberts PJ et al. Hepatocyte apoptosis after bile duct ligation in the mouse involves Fas. Gastroen- terology 1999;117:669–677.

49. Mochizuki K, Hayashi N, Hiramatsu N et al. Fas antigen ex- pression in liver tissue of patients with chronic hepatitis B. J Hepatol 1996;24:1–7.

50. Muller M, Strand S, Hug H et al. Drug-induced apoptosis in hepatoma cells is mediated by the CD95 (APO- 1/Fas) recep- tor/ligand system and involves activation of wild-type p53. J Clin Invest 1997;99:403–413.

51. Muller M, Wilder S, Bannasch D et al. p53 activates the CD95 (APO-1/Fas) gene in response to DNA damage by anticancer drugs. J Exp Med 1998;188:2033–2045.

52. Muschen M, Warskulat U, Peters-Regehr T et al. Involve- ment of CD95 (Apo-1/Fas) ligand expressed by rat Kupffer cells in hepatic immunoregulation. Gastroenterology 1999;116:666–677.

53. Nagao M, Nakajima Y, Hisanaga M et al. The alteration of Fas receptor and ligand system in hepatocellular carcinomas:

how do hepatoma cells escape from the host immune sur- veillance in vivo? Hepatology 1999;30:413–421.

54. Nakamoto Y, Kaneko S, Fan H et al. Prevention of hepato- cellular carcinoma development associated with chronic hepatitis by anti-Fas ligand antibody therapy. J Exp Med 2002;196:1105–1111.

55. Narayanan VS, Fitch CA, Levenson CW. Tumor suppressor protein p53 mRNA and subcellular localization are altered by changes in cellular copper in human Hep G2 cells. J Nutr 2001;131:1427–1432.

56. Natori S, Rust C, Stadheim LM et al. Hepatocyte apoptosis is a pathologic feature of human alcoholic hepatitis. J Hepatol 2001;34:248–253.

57. Pan J, Duan LX, Sun BS et al. Hepatitis B virus X protein pro- tects against anti-Fas-mediated apoptosis in human liver cells by inducing NF- B. J Gen Virol 2001;82:171–182.

58. Patel T, Bronk SF, Gores GJ. Increases of intracellular magne- sium promote glycodeoxycholate-induced apoptosis in rat hepatocytes. J Clin Invest 1994;94:2183–2192.

59. Perez-Carreras M, Del Hoyo P, Martin MA et al. Defective he- patic mitochondrial respiratory chain in patients with nonal- coholic steatohepatitis. Hepatology 2003;38:999–1007.

60. Peter ME, Hellbardt S, Schwartz-Albiez A et al. Cell surface sialylation plays a role in modulating sensitivity towards APO-1-mediated apoptotic cell death. Cell Death Differ 1995;2:163–171.

61. Scaffidi C, Fulda S, Srinivasan A et al. Two CD95 (APO-1/Fas) signaling pathways. EMBO J 1998;17:1675–1687.

(10)

62. Scaffidi C, Schmitz I, Krammer PH et al. The role of c-FLIP in modulation of CD95-induced apoptosis. J Biol Chem 1999;274:1541–1548.

63. Schneider P, Holler N, Bodmer JL et al. Conversion of mem- brane-bound Fas(CD95) ligand to its soluble form is associ- ated with downregulation of its proapoptotic activity and loss of liver toxicity. J Exp Med 1998;187:1205–1213.

64. Seino K, Kayagaki N, Okumura K et al. Antitumor effect of locally produced CD95 ligand. Nat Med 1997;3:165-170.

65. Shudo K, Kinoshita K, Imamura R et al. The membrane-bound but not the soluble form of human Fas ligand is responsible for its inflammatory activity. Eur J Immunol 2001;31:2504–

2511.

66. Sodeman T, Bronk SF, Roberts PJ et al. Bile salts mediate hepatocyte apoptosis by increasing cell surface trafficking of Fas. Am J Physiol Gastrointest Liver Physiol 2000;278:

G992–G999.

67. Song E, Lee SK, Wang J et al. RNA interference targeting Fas protects mice from fulminant hepatitis. Nature Med 2003;9:347–351.

68. Strand S, Hofmann WJ, Hug H et al. Lymphocyte apopto- sis induced by CD95 (APO-1/Fas) ligand-expressing tu- mor cells – a mechanism of immune evasion? Nature Med 1996;2:1361–1366.

69. Strand S, Hofmann WJ, Grambihler A et al. Hepatic failure and liver cell damage in acute Wilson's disease involve CD95 (APO-1/Fas) mediated apoptosis. Nat Med 1998;4:588–593.

70. Suda T, Takahashi T, Golstein P et al. Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell 1993;75:1169–1178.

71. Suda T, Hashimoto H, Tanaka M et al. Membrane Fas ligand kills human peripheral blood T lymphocytes, and soluble Fas ligand blocks the killing. J Exp Med 1997;186:2045–2050.

72. Sun Z, Wada T, Maemura K et al. Hepatic allograft-derived Kupffer cells regulate T cell response in rats. Liver Transpl 2003;9:489–497.

73. Susin SA, Zamzami N, Castedo M et al. Bcl-2 inhibits the mi- tochondrial release of an apoptogenic protease. J Exp Med 1996;184:1331–1341.

74. Tanaka M, Suda T, Haze K et al. Fas ligand in human serum.

Nature Med 1996;2:317–322.

75. Terradillos O, De La Coste A, Pollicino T et al. The hepatitis B virus X protein abrogates Bcl-2-mediated protection against Fas apoptosis in the liver. Oncogene 2002;21:377–386.

76. Tschopp J, Irmler M, Thome M. Inhibition of Fas death signals by FLIPs. Curr Opin Immunol 1998;10:552–558.

77. Verhagen AM, Ekert PG, Pakusch M et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000;102:43–

53.

78. Wallach D, Varfolomeev EE, Malinin NL et al. Tumor necrosis factor receptor and Fas signaling mechanisms. Annu Rev Im- munol 1999;17:331–367.

79. Wang J, Chun HJ, Wong W et al. Caspase-10 is an initiator caspase in death receptor signaling. Proc Natl Acad Sci USA 2001;98:13884–13888.

80. Yoneyama K, Goto T, Miura K et al. The expression of Fas and Fas ligand, and the effects of interferon in chronic liver dis- eases with hepatitis C virus. Hepatol Res 2002;24:327–337.

81. Zender L, Hutker S, Liedtke C et al. Caspase-8 small interfer- ing RNA prevents acute liver failure in mice. Proc Natl Acad Sci USA 2003;100:7797–7802.

Riferimenti

Documenti correlati

Si noti come in questo caso non siano presenti analogie tra la distribuzione delle velocità ultrasoniche (Figura 3.39 b) e la distribuzione delle temperature nei termogrammi. In

GEOtop is a hydrological model that calculates the energy and mass exchanges between soil, vegetation, and atmosphere, accounting for land cover, water redistribution, snow

Bensaou, “R-mac: Reser- vation medium access control protocol for wireless sensor networks,” in 32nd IEEE Conference on Local Computer Networks (LCN 2007), Oct 2007, pp.. Li,

According to the proposed design, the bridge deck will be made of GFRP pultruded profiles; the tower and stays will be of ordinary steel; stainless steel bolts and plates will be

In our juvenile model, we observed a comparable hepatic damage in males and females after 16 weeks of HFHC diet in spite of a sex related different time course of the alterations

List of abbreviations (alphabetic order): ACC: Acetyl-CoA carboxylase; AMPK: AMP-activated protein kinase; COL1A1/COL1A2: Collagen 1A1/Collagen 1A2; CPT-1:

Elimination of mother-to-child transmission of hepatitis B virus will require a comprehensive approach that includes prevention of hepatitis B virus infection in young

Decriminalising drug use and possession or providing alternatives to imprisonment for people who use drugs, and ensuring access to opioid agonist therapy, tuberculosis treatment,