11.02.2015 Views

barcelona . spain - European Association for the Study of the Liver

barcelona . spain - European Association for the Study of the Liver

barcelona . spain - European Association for the Study of the Liver

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

BARCELONA . SPAIN<br />

44 POSTGRADUATE COURSE SYLLABUS ALCOHOLIC LIVER DISEASE 45<br />

APRIL 18 - 19/2012 THE INTERNATIONAL LIVER CONGRESS TM 2012<br />

methods to diagnose this severe condition. The pattern <strong>of</strong> fibrosis is not homogeneous in all patients. It can<br />

be pericellular, perisinusoidal, and ‘chickenwire’. Eventually, bridging fibrosis develops and <strong>the</strong> subsequent<br />

<strong>for</strong>mation <strong>of</strong> regenerative nodules defines cirrhosis.<br />

FACTORS INFLUENCING FIBROSIS PROGRESSION IN ALD<br />

Although a positive correlation between cumulative alcohol intake and degree <strong>of</strong> liver fibrosis has been<br />

reported, extensive individual variability exists. The main environmental factors influencing <strong>the</strong> progression<br />

<strong>of</strong> liver fibrosis in patients with alcohol abuse include obesity and cigarette consumption. The role <strong>of</strong><br />

metabolic factors such as insulin resistance is unclear, yet recent reports suggest that it may favor fibrosis<br />

progression. Moreover, it is known that genetic factors influence <strong>the</strong> organ-specificity <strong>of</strong> alcohol-related<br />

harmful effects. Family studies, twin concordance studies, and interethnic variations in susceptibility<br />

suggest that genetic factors are important in determining disease risk.<br />

Genes encoding <strong>for</strong> alcohol-metabolizing enzymes and proteins involved in liver toxicity such as<br />

antioxidants and pro-inflammatory cytokines have been <strong>the</strong> main subject <strong>of</strong> investigations in genetic<br />

studies. The main enzymes involved in alcohol metabolism in humans are alcohol dehydrogenase (ADH),<br />

aldehyde dehydrogenase (ALDH), and cytochrome P450IIE1 (CYP2E1). Genetic factors influencing <strong>the</strong><br />

activity <strong>of</strong> <strong>the</strong>se enzymes and <strong>the</strong> rate <strong>of</strong> alcohol metabolism have been extensively studied. Because <strong>of</strong><br />

its fibrogenic potential, variations in <strong>the</strong> generation <strong>of</strong> acetaldehyde may explain individual differences after<br />

abusive alcohol consumption. Polymorphisms in genes encoding pro-inflammatory cytokines known to<br />

participate in <strong>the</strong> pathogenesis <strong>of</strong> ALD have also been examined, including TNF-α, NFκB, IL-1β and IL-1<br />

receptor antagonist, IL-2, IL-6 and IL-10. O<strong>the</strong>r studies have investigated <strong>the</strong> role <strong>of</strong> genetic variations <strong>of</strong><br />

factors involved in LPS-induced intracellular pathways including CD14 and TLR4. Patients with genetic<br />

variations <strong>of</strong> superoxide dismutase and gluthatione-S-transferase, powerful antioxidants, are a risk factor<br />

<strong>for</strong> developing severe ALD. Finally, a recent study indicates that variations <strong>of</strong> <strong>the</strong> patatin-like phospholipase<br />

domain-containing protein 3 (PNPLA3) strongly influence <strong>the</strong> development <strong>of</strong> advanced fibrosis among<br />

Caucasians. Despite <strong>the</strong> high number <strong>of</strong> studies assessing <strong>the</strong> role <strong>of</strong> gene variations in <strong>the</strong> susceptibility<br />

<strong>of</strong> ALD, a well-designed large study per<strong>for</strong>ming a genome wide association analysis is still lacking, and a<br />

genetic test capable <strong>of</strong> identifying patients <strong>the</strong> susceptibility to develop advanced ALD is lacking.<br />

Factors at diagnosis that predict <strong>the</strong> progression <strong>of</strong> ALD to cirrhosis are not well known. The main determinant<br />

<strong>of</strong> disease progression is <strong>the</strong> amount <strong>of</strong> alcohol intake and sustained abstinence. Thus, recent data suggest<br />

that higher alcohol intake is associated with a poor short-term prognosis. In addition, most observational<br />

studies indicate that age, female gender and overweight are risk factors <strong>for</strong> cirrhosis. The existence <strong>of</strong> a<br />

florid steatohepatitis at baseline is a major determinant <strong>of</strong> progression to cirrhosis and it also favors <strong>the</strong><br />

development <strong>of</strong> clinical complications.<br />

GENERAL MOLECULAR ASPECTS OF THE FIBROGENIC PROCESS<br />

Hepatic fibrosis is <strong>the</strong> excessive accumulation <strong>of</strong> fibrillar extracellular matrix (ECM) within <strong>the</strong> liver which<br />

occurs in <strong>the</strong> context <strong>of</strong> chronic liver diseases and is considered <strong>the</strong> result <strong>of</strong> <strong>the</strong> persistent activation <strong>of</strong> a<br />

‘wound healing’ response. In <strong>the</strong> setting <strong>of</strong> a chronic damage, activation <strong>of</strong> this process, evolved to protect<br />

<strong>the</strong> tissue from damage, results in <strong>the</strong> <strong>for</strong>mation <strong>of</strong> a fibrotic scar which eventually alters <strong>the</strong> cellular and<br />

functional balance <strong>of</strong> <strong>the</strong> organ. A key role in fibrogenesis is played by my<strong>of</strong>ibroblasts, which produce<br />

extracellular matrix and in general coordinate <strong>the</strong> ‘wound healing’ response (Figure 1). Hepatic stellate cells<br />

(HSC) are classically considered to be a major source <strong>of</strong> hepatic my<strong>of</strong>ibroblasts, but o<strong>the</strong>r cell types inside<br />

or outside <strong>the</strong> liver have been suggested to contribute to <strong>the</strong> expansion <strong>of</strong> <strong>the</strong> my<strong>of</strong>ibroblast population<br />

observed during injury, including portal fibroblasts and possibly cells recruited from <strong>the</strong> bone marrow. The<br />

possibility that epi<strong>the</strong>lial cells contribute to <strong>the</strong> my<strong>of</strong>ibroblast pool via a process <strong>of</strong> epi<strong>the</strong>lial-mesenchymal<br />

transition has also been recently hypo<strong>the</strong>sized, but its overall contribution to <strong>the</strong> fibrogenic process is<br />

debated. It is believed that <strong>the</strong> involvement <strong>of</strong> different populations <strong>of</strong> fibrogenic cells is dependent on <strong>the</strong><br />

etiology <strong>of</strong> liver damage and <strong>the</strong> resulting development <strong>of</strong> distinct spatial patterns <strong>of</strong> fibrosis. In alcoholic<br />

liver disease, fibrosis develops primarily in <strong>the</strong> pericentral areas, with a picture referred to as ‘pericellular<br />

fibrosis’. Although chronic liver diseases share most <strong>of</strong> <strong>the</strong> pr<strong>of</strong>ibrogenic mechanisms, <strong>the</strong> pathogenesis<br />

<strong>of</strong> fibrosis in ALD and in o<strong>the</strong>r conditions this conditions may have certain specificities. For this reason,<br />

several studies have recently focused on identify specific pathways leading to fibrosis in different etiologies<br />

<strong>of</strong> chronic liver disease.<br />

PATHOGENETIC MECHANISMS OF FIBROSIS SPECIFIC FOR ALD<br />

As discussed earlier, <strong>the</strong> spectrum <strong>of</strong> ALD ranges from liver steatosis to steatohepatitis, fibrosis, cirrhosis<br />

and hepatocellular carcinoma. The pathogenesis <strong>of</strong> steatosis includes increased fatty acid and triglyceride<br />

syn<strong>the</strong>sis and inhibited mitochondrial ß-oxidation <strong>of</strong> fatty acids and enhanced hepatic influx <strong>of</strong> free fatty<br />

acids from adipose tissue and <strong>of</strong> chylomicrones from <strong>the</strong> intestinal mucosa. Alcoholic fatty livers can<br />

develop inflammatory infiltrate (mainly composed by polymorphonuclear cells) and hepatocellular damage,<br />

a prerequisite to <strong>the</strong> progression to fibrosis and cirrhosis.<br />

Different pathogenic factors are implicating in <strong>the</strong> development <strong>of</strong> liver damage and fibrosis (Figure 2). In<br />

this respect, ALD represents a clear example <strong>of</strong> <strong>the</strong> importance <strong>of</strong> cell-cell interaction and cross-talk within<br />

<strong>the</strong> liver. In fact, hepatocytes, Kupffer cells, HSC, and inflammatory cells recreuited from <strong>the</strong> bloodstream<br />

all contribute to <strong>the</strong> pathogenesis <strong>of</strong> damage and <strong>the</strong> development <strong>of</strong> fibrosis.<br />

Products <strong>of</strong> alcohol metabolism may directly damage hepatocytes. Acetaldehyde, <strong>the</strong> major methanol<br />

metabolic product, binds to proteins and DNA resulting in functional alterations and protein adducts, which<br />

activate <strong>the</strong> immune system by <strong>for</strong>ming autoantigens. It also causes mitochondrial damage and impairs<br />

glutathione function, leading to oxidative stress and apoptosis. Acetaldehyde has been also shown to<br />

interfere with TGF-beta signaling via Smad3.<br />

In addition, reactive oxygen species (ROS) generation and <strong>the</strong> resulting lipid peroxidation play a<br />

major role in <strong>the</strong> pathogenesis <strong>of</strong> ASH and fibrosis. Main sources <strong>of</strong> ROS include CYP2E1-dependent<br />

MEOS, mitochondrial electron transport system <strong>of</strong> <strong>the</strong> respiratory chain, NADH-dependent cytochrome<br />

reductase and xanthine oxidase. Moreover, chronic alcohol intake markedly up-regulates CYP2E1, which<br />

metabolizes ethanol to acetaldehyde and parallels <strong>the</strong> generation <strong>of</strong> ROS and hydroxyl-ethyl radicals.<br />

Cytokines and inflammation: Several studies have indicated that inflammatory pathways are activated<br />

in <strong>the</strong> liver during ALD, including NF-κB and tumor necrosis factor-α (TNFα). Kupffer cells, activated<br />

lymphocytes, and macrophages infiltrating <strong>the</strong> liver tissue are sources <strong>of</strong> TNFα.<br />

More recently, a possible involvement <strong>of</strong> <strong>the</strong> chemokine system has been suggested by several experimental<br />

data in humans and in experimental models <strong>of</strong> ALD. In alcoholic hepatitis, hepatic expression <strong>of</strong> several<br />

chemokines has been found to be upregulated, and in particular expression <strong>of</strong> CXC components correlated<br />

with neutrophil infiltration and prognosis. These data identify a group <strong>of</strong> novel targets <strong>for</strong> novel <strong>the</strong>rapeutic<br />

approaches to <strong>the</strong> most severe <strong>for</strong>ms <strong>of</strong> alcoholic liver disease.<br />

Endotoxin and TLRs: Alcohol abuse results in changes in colonic microbiota and increased intestinal<br />

permeability, leading to endotoxemia. i.e. an increase in plasma lipopolysaccharide (LPS) levels, which is<br />

released in <strong>the</strong> circulation due to an increase in intestinal permeability. A pivotal aspect <strong>of</strong> this host-bacteria<br />

interaction is <strong>the</strong> recognition <strong>of</strong> TLR4 on Kupffer cells, that induce TNF-α production and contributes to liver<br />

injury. Recent studies have also shown that HSC also express several members <strong>of</strong> <strong>the</strong> toll-like receptor<br />

(TLR) subgroups, such as TLR4 and TLR9. TLR4 activation triggers multiple intracellular signaling pathways,<br />

including NF-κB, and induces <strong>the</strong> expression <strong>of</strong> several pr<strong>of</strong>ibrogenic cytokines. The resulting inflammatory<br />

milieu in <strong>the</strong> alcoholic liver leads to PMN infiltration, ROS <strong>for</strong>mation and hepatocellular damage, which drive<br />

fibrogenesis. Finally, impairment in <strong>the</strong> ubiquitin-proteasome pathway leads to hepatocellular injury and<br />

hepatic inclusions <strong>of</strong> aggregated cytokeratins (a.k.a. Mallory-Denk bodies).<br />

Following exposure to a hepatotoxic agent such as alcohol, hepatocytes activate several signaling pathways

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!