ArticlePDF Available

Abstract and Figures

With the increasing burden of liver cirrhosis, the most advanced stage of hepatic fibrosis, there is a need to better understand the pathological processes and mechanisms to target specific treatments to reverse or cease fibrosis progression. Antiviral therapy for hepatitis B and C has effectively treated underlying causes of chronic liver disease and has induced fibrosis reversal in some; however, this has not been targeted for the majority of aetiologies for cirrhosis including alcohol or nonalcoholic steatohepatitis. Fibrosis, characterised by the accumulation of extracellular matrix proteins, is caused by chronic injury from toxic, infectious, or metabolic causes. The primary event of fibrogenesis is increased matrix production and scar formation mediated by the hepatic stellate cell, which is the principal cell type involved. Experimental models using rodent and human cell lines of liver injury have assisted in better understanding of fibrogenesis, especially in recognising the role of procoagulant factors. This has led to interventional studies using anticoagulants in animal models with reversal of fibrosis as the primary endpoint. Though these trials have been encouraging, no antifibrotic therapies are currently licenced for human use. This literature review discusses current knowledge in the pathophysiology of hepatic fibrosis, including characteristics of the extracellular matrix, signalling pathways, and hepatic stellate cells. Current types of experimental models used to induce fibrosis, as well as up-to-date anticoagulant therapies and agents targeting the hepatic stellate cell that have been trialled in animal and human studies with antifibrotic properties, are also reviewed.
Content may be subject to copyright.
Creative Commons Attribution-Non Commercial 4.0 December 2019 • EUROPEAN MEDICAL JOURNAL
105
A Review of Liver Fibrosis and Emerging Therapies
Authors: Rooshi Nathwani, Benjamin H. Mullish, David Kockerling, Roberta
Forlano, Pinelopi Manousou, *Ameet Dhar
Integrative Systems Medicine and Digestive Disease, Imperial College London,
London, UK
*Correspondence to a.dhar@imperial.ac.uk
Disclosure: The authors have declared no conflicts of interest.
Received: 03.05.19
Accepted: 01.08.19
Keywords: Anticoagulation, antifibrotic therapy, fibrosis, hepatic fibrosis, hepatic stellate cell.
Citation: EMJ. 2019;4[4]:105-116.
Abstract
With the increasing burden of liver cirrhosis, the most advanced stage of hepatic fibrosis, there is a
need to better understand the pathological processes and mechanisms to target specific treatments
to reverse or cease fibrosis progression. Antiviral therapy for hepatitis B and C has eectively treated
underlying causes of chronic liver disease and has induced fibrosis reversal in some; however, this
has not been targeted for the majority of aetiologies for cirrhosis including alcohol or nonalcoholic
steatohepatitis. Fibrosis, characterised by the accumulation of extracellular matrix proteins, is caused
by chronic injury from toxic, infectious, or metabolic causes. The primary event of fibrogenesis is
increased matrix production and scar formation mediated by the hepatic stellate cell, which is the
principal cell type involved. Experimental models using rodent and human cell lines of liver injury have
assisted in better understanding of fibrogenesis, especially in recognising the role of procoagulant
factors. This has led to interventional studies using anticoagulants in animal models with reversal of
fibrosis as the primary endpoint. Though these trials have been encouraging, no antifibrotic therapies
are currently licenced for human use. This literature review discusses current knowledge in the
pathophysiology of hepatic fibrosis, including characteristics of the extracellular matrix, signalling
pathways, and hepatic stellate cells. Current types of experimental models used to induce fibrosis,
as well as up-to-date anticoagulant therapies and agents targeting the hepatic stellate cell that have
been trialled in animal and human studies with antifibrotic properties, are also reviewed.
INTRODUCTION
The burden of chronic liver disease continues
to grow, with 0.1% of the European population
aected by cirrhosis, the most advanced stage
of hepatic fibrosis.1 Although the aetiology of
the disease varies between countries,
fibrogenesis is the common pathological
mechanism that causes cirrhosis. Fibrosis
occurs following chronic liver injury from a
range of insults including toxins (alcohol),
infections (hepatitis B [HBV] and C viruses [HCV]),
and metabolic disease (nonalcoholic fatty liver
disease). Such insults drive inflammation, resulting
in increased synthesis and altered deposition
of extracellular matrix (ECM) components,
and impaired regeneration and wound healing
responses.2 This is a complex, dynamic process,
involving recruitment and activation of platelets,
inflammatory cells, hepatic stellate cells (HSC),
and other ECM-producing cells including portal
fibroblasts, hepatocytes, cholangiocytes, and
EUROPEAN MEDICAL JOURNAL • December 2019 EMJ EUROPEAN MEDICAL JOURNAL
106
bone marrow-derived cells.3 The end result,
cirrhosis, is defined by profound distortion of
hepatic microarchitecture, ultimately resulting
in the development of portal hypertension.4
Histologically, cirrhosis is characterised by
regenerative nodules of liver parenchyma
separated by, and encapsulated in, fibrotic
septa, with a clinical consequence of increased
mortality, morbidity, complications of portal
hypertension, and diminished quality of life.2,5 The
presence of hepatic fibrosis is a key predictor of
prognosis in chronic liver disease, independent
of aetiology.6 Generally this process evolves over
decades (usually 20–40 years), but it can be
rapidly progressive, as seen in children aected
by biliary atresia, drug-induced liver injury, HCV
co-infection with HIV, or HCV infection post
liver transplantation.7
Management of chronic liver disease has largely
focussed on aetiology-specific treatments;
however, significant progress has been made in
understanding the pathophysiology of fibrosis,
which has identified targets for potential
antifibrotic agents to either halt progression or
reverse fibrosis.
A search of the existing literature up to June
2019 was conducted using electronic databases
PubMed, Medline, and the Cochrane library, as well
as relevant guidelines, to present this literature
review, an overview of the current understanding
of the pathogenesis of hepatic fibrosis, in vitro and
in vivo models used in exploring pathogenesis,
and an update on proposed therapies.
PATHOPHYSIOLOGY
Extracellular Matrix
The ECM is a dynamic structural component of
the normal liver.8 It contains macromolecules
that provide the scaolding of the liver and act
as transducers of extracellular signals. In normal
liver tissue the ECM is composed of collagens,
glycoproteins, fibronectin, laminin, tenascin,
von Willebrand factor, and proteoglycans.9 It
is a component of Glisson’s capsule, portal
tracts, central veins, and the subendothelial
space of Disse.10 Various cellular sources of the
ECM have been identified but the main source
is the HSC. When liver injury is not severe,
neighbouring hepatocytes regenerate and
replace apoptotic and necrotic cells. However,
in chronic insult, the mechanism contributing
to fibrosis, this process fails and ECM proteins
take on the role of hepatocytes.11 The quantity
and quality of the ECM changes, increasing
up to 8-fold compared to a normal liver. There
is significant increase in collagen content and
proteoglycans, resulting in a higher density
interstitial type matrix.12 The ECM composition
also transforms, from one predominantly made
up of Type IV and VI collagen, glycoproteins,
proteoglycans, and laminin, to a matrix
consisting of Type I and III collagen and
fibronectin.10 These adaptations alter the local
microenvironment leading to subsequent
functional and physical restrictions on plasma
flow between sinusoids and hepatocytes, causing
impaired hepatic function.9
Signalling in Fibrosis
Fibrogenesis is a complex mechanism
involving an array of cellular and extracellular
signalling. Cytokine, chemokine, adipokine,
neuroendocrine, angiogenic, and nicotinamide
adenine dinucleotide phosphate-oxidase
(NADPH) signalling have all been found to play
important functions.13 The release of cytokines
in the context of fibrogenesis is controlled
in part by activation of metalloproteinases.
TGF-β1, platelet-derived growth factor (PDGF),
connective tissue growth factor (CTGF), TNF-α,
and vascular endothelial growth factor (VEGF)
are all involved in key mechanisms of fibrosis.9,1 4
TGF-α is particularly important in mediating
stellate cell activation to myofibroblasts and
stellate cell collagen production.14 VEGF and
PDGF control angiogenesis, which contributes
to ECM production, portal hypertension, and
regeneration postinjury.15 During acute or chronic
liver injury, platelets are the first cells recruited to
the injury site. They form aggregates in damaged
vasculature by converting fibrinogen into fibrin
and platelet α-granules rich in PDGF, TGF-α,
and VEGF.16
Chemokines, CCL2 (produced by Kuper cells and
HSC), and CCL5 are best recognised in fibrosis.17
CCL2 promotes HSC activation, facilitating
macrophage and monocyte recruitment into the
liver. Inhibition of CCL2, or its receptor CCR2, is
associated with reduced fibrosis in experimental
models.18 CCL5 and its receptors, CCR1 and CCR5,
are associated with fibrogenesis promotion.19
Creative Commons Attribution-Non Commercial 4.0 December 2019 • EUROPEAN MEDICAL JOURNAL
107
Other signalling molecules involved in
fibrogenesis include the adipokines, leptin, and
adiponectin, synthesised by HSC, as well as
cannabinoids and their respective receptors.
Cannabinoid receptor 1 is a profibrotic mediator;
however, cannabinoid receptor 2 has antifibrotic,
but proinflammatory, properties.13
Reactive oxygen species mediate fibrogenesis
by stimulating profibrogenic mediator release
from Kuper cells and activating HSC. Emerging
evidence suggests that NADPH also plays a role
in this process.20
Hepatic Stellate Cells
The HSC is the principal cell type involved in
fibrogenesis21 and its activation is the common
pathway leading to fibrosis. Developing a clear
understanding of factors that stimulate or inhibit
activation of HSC has helped guide development
of antifibrotic therapies. HSC lie in a quiescent
state in the subendothelial space of Disse and
comprise 15% of liver cells.8 In the normal liver,
they are the primary storage site for retinoids
and are derived from neural crest tissue because
of expression of neural crest markers, including
glial fibrillary acidic protein and nestin.22 These
cells are activated by proinflammatory cytokines
and protease activated receptor-1 ligation,
resulting in a myofibroblastic phenotype which is
proliferative, fibrogenic, and contractile.
Activation of HSC proceeds through an initiation
and perpetuation phase.13
Initiation refers to early changes that occur in
gene expression and phenotype, initiated by
mediators that render quiescent HSC responsive
to other stimuli. Characteristic to this phase is the
production of a fibrogenic, contractile phenotype
with induction of PDGF receptors.22 Initiation is
stimulated by oxidant stress signals, apoptotic
hepatocytes, lipopolysaccharides, and paracrine
stimuli from neighbouring cells including
cholangiocytes, Kuper cells, injured sinusoidal
endothelial cells, or other stellate cells.13 Initiation
activates stellate cells, while perpetuation is the
response to stimuli that maintains the activated
stellate cell in its myofibroblastic state to
allow fibrotic scar formation.23 These changes
include stellate cell proliferation, chemotaxis,
fibrogenesis, increased contractility, altered
matrix degeneration, and cytokine signalling.22
The release of a variety of mitogenic factors
causes proliferation of stellate cells. PDGF is a
potent mitogen upregulated during liver injury.24
Other molecules with mitogenic activity include
VEGF, thrombin, epidermal growth factor, TGF-α,
keratinocyte growth factor, and fibroblast growth
factor.25 Matrix metalloproteinase-2 (MMP-2)
acts as a stellate cell mitogen via activation
by discoidin domain receptor-2 binding to
fibrillar collagen.26
Activated stellate cells migrate towards areas of
hepatic injury during chemotaxis. Growth factors
within the liver which have chemoattractant
properties include PDGF, insulin growth factor-1,
endothelin-1, monocyte attractant protein-1, and
the chemokine receptor CXCR3;27 in contrast,
adenosine inhibits chemotaxis, allowing cells to
fix at injury sites.28
The primary event of fibrogenesis is increased
matrix production and scar formation mediated
by stellate cells.10 Type I collagen, the major
constituent of scar tissue, is upregulated post-
transcriptionally in stellate cells by the actions
of TGF-α via Smads, pivotal intracellular eector
proteins that mediate TGF-α signalling.29 CTGF
also promotes fibrogenesis.30 Other less potent
mediation of Type I collagen production include
angiotensin II, IL-1β, and TNF. 31
On activation the HSC contracts, which is
characterised by the increased expression of
α-smooth muscle actin, a contractile filament
protein.32 This results in impendence of sinusoidal
blood flow increasing portal resistance, and
eventually increasing portal pressure once
advanced fibrosis has developed. Progressive
development of intrahepatic shunting occurs
in conjunction with the constriction of
hepatic sinusoids. The process is mediated
by endothelin-1, as well as angiotensinogen
II and atrial natriuretic peptide. Nitric
oxide has an opposing eect, resulting in
sinusoidal relaxation.33
The concept of matrix degradation evolved
following evidence demonstrating that fibrosis
is reversible. This propagated the theory that
fibrosis is a dynamic balance between matrix
production and degradation.34 Initial matrix
degradation may be termed pathological,
corresponding to disruption of normal low-
density matrix of the subendothelial basement
EUROPEAN MEDICAL JOURNAL • December 2019 EMJ EUROPEAN MEDICAL JOURNAL
108
membrane, occurring in early stages of fibrosis.
This allows for normal matrix to be replaced by
a higher density pathological matrix, containing
fibrils. Matrix degrading collagenases, including
the MMP, break down Type IV collagen and are
central to this paradigm.35,36 Stellate cells and
Kuper cells are the primary sources of MMP-2
and MMP-9, respectively.35 Conversely, MMP-
1 degrades Type I collagen and is involved in
the restorative degradation of the pathological
matrix, thus, upregulation of this enzyme
would favour resolution of scar tissue. Tissue
inhibitors of metalloproteinases (TIMP) inhibit
the actions of collagenases, resulting in reduced
matrix degradation, and inhibit stellate cell
apoptosis, which prevents fibrosis resolution
and favours matrix accumulation.36 TIMP-1 and
2 are upregulated in progressive fibrosis, and
their sustained release from stellate cells is a key
determinant of progressive fibrosis.36,37
HSC are not only eectors of fibrosis but also
have a central role in amplifying inflammatory
signaling via toll-like receptors and cytokines
including monocyte chemotactic protein-1, CCL21,
CCR5, and Regulated upon Activation, Normal
T-cell Expressed, and Secreted (RANTES). They
demonstrate antigen presenting capabilities and
produce neutrophil chemoattractants.38,39
EXPERIMENTAL MODELS
OF LIVER INJURY
Techniques isolating and cultivating HSC have
represented a major advance in exploring the
complex mechanisms involved in fibrogenesis.
Rodent stellate cell lines have been characterised
but have now been superseded by human cell
lines.23 The most utilised human HSC lines include
the LX1 and LX2 cell lines. LX1 cell line is generated
by transformation with SV40 T antigen.40 The
LX2 cell line is generated by isolating stellate
cells from normal human livers and immortalising
them by culturing in low serum conditions.
When activated, these cells bear close similarity
to in vivo human activated HSC.41 The LX2 cell
line has been extensively validated and used
in a number of studies exploring the eects of
mediators on stellate cell activity.25 Other human
HSC lines are summarised in Table 1.40 Utilising
these cell lines therefore allows us the unique
opportunity to study factors that both inhibit
and activate HSC.
Animal models oer the opportunity to study
interactions of dierent cell types, gene
activation, protein-expressing profiles, and
signalling pathways, but as yet no animal model
exactly reproduces human hepatic fibrosis.42
Table 1: Summary table of human stellate cell lines used.
Human hepatic stellate cell line Derivation
LI90 Human hepatic epithelioid haemangioendothelioma.
TWNT-1
TWNT-4
Retrovirally-induced human telomerase reverse
transcriptase into LI90 cell line.
Human telomerase reverse transcriptase gene Normal human liver.
HSC-Li Normal human liver.
GREF-X Cirrhotic human liver.
LX1 Transformation with SV40 T antigen.
LX2 Isolating stellate cells from normal human livers and
immortalising them by culturing in low serum conditions.
Creative Commons Attribution-Non Commercial 4.0 December 2019 • EUROPEAN MEDICAL JOURNAL
109
A variety of agents, often carcinogenic, can be
employed to chemically induce fibrosis. The most
commonly used are carbon tetrachloride (CCl4)
and thioacetamide (TAA). Fibrosis generated in
this manner has both a degree of reproducibility
and similarity with human mechanisms and
pathways involved in hepatic fibrosis.43 They
also have the added advantage that they can
be used in conjunction with either transgenic or
wild-type mice to explore underlying molecular
mechanisms involved in hepatic fibrosis, or
can be used to evaluate the potential of novel
antifibrotic agents using strictly controlled
environmental and genetic conditions.43
Carbon Tetrachloride
CCL4 is the oldest and most common method of
inducing liver fibrosis in rodent models.44 It is a
halothane and induces hepatic injury when given
at repetitive low doses. It can be administered
in a variety of ways, but is most commonly
administered by intraperitoneal injection 3
times per week. CCl4 is bioactivated by oxidases.
This leads to a combination of eects including
CCl3 radical formation in the liver, which causes
hepatocyte damage via lipid peroxidation;
HSC activation; Kuper cell activation; TGF-β-1
upregulation; and increased oxidative stress.45,46
Histological alterations result in fatty change
with necrosis and intense necroinflammation in
centrilobular areas. This progresses to both septal
and nonseptal fibrosis resulting in extensive
fibrosis and cirrhosis. The fibrous septa are
classically thin and can regress with CCl4
withdrawal. The reproducibility and ease of
induction of fibrosis are its main advantages.
Disadvantages include heterogeneity in the
amounts of fibrosis produced between animals.32
Thioacetamide
TAA is a selective hepatotoxin. Chronic
administration not only induces liver fibrosis,
but can result in carcinogenesis, including
cholangiocarcinoma and hepatocellular
carcinoma. TAA is usually given in drinking
water over a period of 8–18 weeks to induce
liver fibrosis.47 Histologically, mild to moderate
amounts of fibrosis develop by 8 weeks
with elevated transaminases. By 12 weeks,
parenchymal damage occurs, with hepatocyte
swelling, necrosis, and proliferation. This results
in fibrous enlargement of the portal tracts, with
portal–portal and portal–central septa developing
resulting in cirrhosis. Histological similarities with
human viral hepatitis have led to its use as an
indirect model of both fibrosis secondary to HBV
and HCV infection. Withdrawal of TAA results in
resolution of fibrosis over an 8-week period. A
longer period of resolution, compared with CCl4
models, makes it a more suitable model when
evaluating potential antifibrotic therapies, and
the occurrence of regenerative nodules make it
more comparable to human cirrhosis.48
Dimethylnitrosamine
Dimethylnitrosamine (DMN) is a hepatotoxin
and a carcinogenic and mutagenic agent. DMN
is typically administered intraperitoneally three
times a week, with centrilobular and periportal
fibrosis characteristically developing after 3
weeks. Microscopically, this pattern of fibrosis is
seen in cirrhosis.49 DMN models induce HSC and
Kuper cells to express profibrotic cytokines
resulting in deposition of excessive ECM, the
primary pathogenesis of fibrosis, making it a
potentially useful animal model.50 However, DMN
has mutagenic and carcinogenic properties and
exposure can cause hepatocellular carcinoma.
This makes understanding fibrosis pathways
dicult to interpret but does allow the
pathogenesis of fibrosis to hepatocellular
carcinoma to be better understood.51
BILE DUCT LIGATION
Surgical ligation of the common bile duct causes
cholestasis and periportal inflammation. This
technique causes proliferation of biliary epithelial
cells and increases expression of fibrogenic
markers such as TIMP-1, Alpha-SMA, Type I
collagen, and TGF-β1.52 Although this model
aids portal fibrosis and portal myofibroblast
interrogation, bile duct ligation model use is
restricted due to the high frequency of gall
bladder perforation and bilio-peritoneum in
mice.53 Mortality risk is therefore significant and
this model is more suitable for short-term studies
investigating cholestasis-induced fibrosis.52,53
EUROPEAN MEDICAL JOURNAL • December 2019 EMJ EUROPEAN MEDICAL JOURNAL
110
Table 2: Summary table of anti-fibrotic strategies targeting hepatic stellate cell.
Agent Target Mechanisms
1. Reducing inflammation and immune responses before HSC activation.
Silymarin Oxidative stress mechanisms, NFκB
pathways, and PDGF signalling.
Inhibits oxidative stress and
subsequently stellate cell activation.
Glucocorticoids Immune mediators Reduction of inflammation.
Caeine A2A adenosine receptor Inhibition of A2A adenosine
reception expressed by
myofibroblasts and linked to matrix
production.
Curcumin Cannabinoid receptors Downregulation of cannabinoid 1
receptor, a profibrotic mediator.
Ursodeoxycholic acid Cholangiocytes Reduction in the cytotoxic eects of
bile acids and protects hepatocytes
against apoptosis.
2. Inhibition of HSC activation
Vitamin E Oxidative stress mechanisms Prevention of oxidative stress
implicated in fibrogenesis.
Thiazolindinediones PPAR Anti-inflammatory and antifibrotic
eects by inhibiting PDGF
expression. Antifibrotic eect has
been demonstrated in patients with
nonalcoholic fatty liver disease.
Oleoylethanolamide PPAR Same mechanism as
thiazolidinediones and but also
initiation of α-smooth muscle
expression.
Imatinib mesylate PDGF Suppression of PDGF and HSC
activation.
ACE inhibitors Renin–angiotensin system Downregulation of angiotensin II
receptors on HSC, responsible for
proliferation and contraction. Though
readily available, no large randomised
trials have been conducted in
humans.
Recombinant IL-22 Th22 receptors Inhibit HSC activation and suppresses
inflammatory cytokine release.
Thrombin and FXa PAR1 and 2 stellate cell activation Inhibition of PAR 1 and 2 stellate
mediated activation.
ROCK inhibitor GTP-binding protein Rho Inhibition of stellate cell activation.
3. Inhibiting response after HSC activation
GW6604 TGF-α Inhibit TGF-β1 signalling pathways.
Cytosporone B
NR4A1 gene agonist
TGF-α Inhibit TGF-β1 signalling pathways.
Bosentan Endothelin Endothelin antagonism reduces
stellate cell activation and
extracellular matrix production.
Halofuginone Collagen synthesis Inhibition of collagen Type I
synthesis by inhibition of SMAD3
phosphorylation.
Creative Commons Attribution-Non Commercial 4.0 December 2019 • EUROPEAN MEDICAL JOURNAL
111
Agent Target Mechanisms
Caspase inhibitors Caspase Inhibits eectors of apoptosis
signalling in hepatocytes.
Obeticholic acid Farnesoid-X receptor Improved integrity of hepatocytes,
reduction in HSC contractility and
reduction of collagen.
4. Promoting activated HSC into apoptosis
Gliotoxin NFκB Inhibition of NFκB pathways,
suppressing chronic hepatic
inflammation.
Sulfasalazine NFκB Inhibition of NFκB pathways,
suppressing chronic hepatic
inflammation.
Thalidomide NFκB Inhibition of NFκB pathways,
suppressing chronic hepatic
inflammation.
Melatonin NFκB Inhibition of NFκB pathways,
suppressing chronic hepatic
inflammation.
Cannabinoid 1 antagonist Cannabinoid 1 receptor Inhibition of collagen Type I
synthesis by inhibition of SMAD3
phosphorylation. Reduces cellular
proliferation and promotes
myofibroblast apoptosis.
Cannabinoid 2 agonist Cannabinoid 2 receptor Inhibits myofibroblast proliferation
and induces apoptosis.
Interferon NK Cells Promotes NK cell activity and
promotes HSC death.
Hepatocyte growth factor Myofibroblast Inhibits extracellular matrix producing
myofibroblasts.
Table 2 continued.
ATP-Binding Cassette Subfamily B
Member 4
The ATP-binding cassette subfamily B
member 4 (Abcb4-/-) gene encodes multidrug
resistance 3 (MDR3) protein, the canalicular
phosphatidylcholine lipid transporter. Altering
this gene by rendering it deficient causes
intrahepatic cholestasis and disease similar to
that of primary biliary cirrhosis. It functions to
protect cellular membranes facing the biliary
tree against bile acids and without
phosphatidylcholine there is biliary epithelial
and ductular damage, portal inflammation and
proliferation, and progressive portal fibrosis.54
Abcb4-/-knockout mice develop fibrosis between
4 and 8 weeks with TGF-β1 expression, HSC
activation at 4 weeks with collagen deposition,
and scarring at 8 weeks.55
These models have been used to study
primary biliary cirrhosis, cholestasis of
ACE: angiotensin-converting enzyme; FXa: Factor Xa; GTP: guanosine-5'-triphosphate; HSC: hepatic stellate cell;
NK: natural killer; NR4A1: nuclear receptor subfamily 4 group A member 1; PAR1: protease-activated receptor 1;
PAR2: protease-activated receptor 2; PDGF: platelet-derived growth factor; PPAR: peroxisome proliferator activated
receptors; ROCK: Rho kinase; SMAD3: SMAD Family Member 3.
Adapted from Ebrahimi H et al.58
EUROPEAN MEDICAL JOURNAL • December 2019 EMJ EUROPEAN MEDICAL JOURNAL
112
pregnancy drug-induced cholestasis, and
therapeutic interventions.56
ANTIFIBROTIC THERAPY
Trials of antifibrotic therapies in humans
have been limited compared to experimental
models in animals. This is partly due to the long
duration antifibrotic agents would need to be
administered to demonstrate a treatment
eect because of the time taken for the agent
to systemically accumulate. Trials are limited
by funding and concerns over side eects.
Furthermore, the historical need for histological
endpoints can limit recruitment. An ideal trial
would be one that has a short duration of
treatment, with a pre-existing clinical need for
routine liver histology.
Though significant progress has been made
in understanding the pathogenesis of fibrosis,
particularly the role of the HSC, no antifibrotic
therapy specifically targeting this cell type
responsible for increased matrix production and
scar formation is currently licensed for human
use. Several potential agents and antifibrotic
molecules such as silymarin, caeine, and
curcumin, summarised in Table 2, have shown
antifibrotic properties via targeting the HSC, but
are not routinely used in clinical practice.57,58
Silymarin, mainly consisting of silibinin, is a
flavonoid complex extracted from milk thistle.
It has been shown to diminish the following:
oxidative stress, lysis of hepatocytes, activation
of Kuper cells, and expression of α-SMA and
TGF-β1, all implicated in the pathogenesis
of fibrosis in rats treated with CCl4 to induce
fibrosis.59 Despite showing promise in animal
models with improved liver function tests, in
humans there is little evidence demonstrating
its clinical benefit. The De Avelar et al.60 meta-
analysis of six human studies demonstrated
that although alanine transaminase and
aspartate transaminase levels were reduced with
silymarin use, there was no clinically significant
benefit associated with this. To date, only one
randomised, double-blinded, placebo-controlled
study with biopsy-proven fibrosis investigating
the eect of silymarin has been conducted. This
showed that silymarin did not significantly reduce
nonalcoholic fatty liver disease activity scores,
although some improvement of fibrosis was
histologically seen compared to placebo. This
study was underpowered and did not include
other aetiologies of fibrosis.61
Several studies have reported the beneficial
eect of caeine against liver disease.62 The
major mechanism by which caeine exerts its
antifibrotic eect is largely attributable to caeine
being a pan antagonist of the adenosine receptor.
Liver myofibroblasts are profibrogenic and
express the A2A adenosine receptor; therefore,
blocking this receptor inhibits fibrogenesis.63 A
second proposed mechanism is that caeine
alters signalling and inflammation pathways in
fibrogenesis by reducing TGF-β expression, as
suggested in rodent models of fibrosis.64
Curcumin, a monomer extract from turmeric,
has not only shown anti-inflammatory and
antiproliferative properties but also antifibrotic
actions, as evidenced by its ability to protect
against fibrosis in CCl4 treated rats.65,66 Curcumin
also interferes with TGF-β signalling pathways
and PDGF receptors, leading to inhibition of
HSC activity. This is further achieved through
modulation of the cannabinoid receptor system
with downregulation of the cannabinoid receptor
1, inhibiting ECM expression by HSC.67
Anticoagulation
Evidence suggests that hepatic fibrogenesis
is associated with prothrombotic tendencies,
including factor V Leiden (FVL); this is
demonstrated by Wright et al.,68 who found that
FVL mutation increased the rate of fibrosis in HCV
infection. The coagulation proteins thrombin and
factor Xa (FXa) are also implicated through their
activation of HSC.69 Activation of the coagulation
system generates FXa, which in turn results in the
production of thrombin from its precursor protein,
prothrombin. Both thrombin and FXa activate
stellate cells via G-protein-coupled receptors
known as protease activated receptors (PAR).70
Duplantier et al.71 evaluated, in a rat model of
CCl4-induced chronic liver injury, the eect of
thrombin inhibition using a synthetic thrombin
antagonist SSR182289. The study demonstrated
a reduction in liver fibrosis and α-smooth
muscle actin expression, a marker of stellate cell
activation. Dhar et al.72 demonstrated that
administration of rivaroxaban, an FXa antagonist,
to mice that had been exposed to TAA for 8
weeks induced milder fibrosis, especially around
Creative Commons Attribution-Non Commercial 4.0 December 2019 • EUROPEAN MEDICAL JOURNAL
113
central veins, compared with control mice.
The overall mean percentage area of fibrosis
was significantly reduced, as was α-smooth
muscle actin expression. This eect is likely
because of FXa antagonists blocking PAR1 and
2-mediated stellate cell activation.72 Prolonged
administration of enoxaparin in a rat model of
cirrhosis, (induced using CCL4 or TAA), resulted
in an improvement in both portal hypertension
and liver fibrosis, possibly by potentiating fibrosis
regression, resulting in reduction of hepatic
vascular resistance and portal pressures.73 The
use of warfarin anticoagulation to reduce liver
fibrosis induced by CCl4 has been tested using a
transgenic mouse model of FVL-activated protein
C resistance. The progression of fibrosis in FVL
mice was compared to the control C57BL/6 mice
which had been anticoagulated with warfarin.
The results confirmed that the thrombophilic
mouse developed fibrosis at a faster rate than
the control mice, but warfarin anticoagulation
significantly reduced the rate of fibrosis seen in
both strains.74
A small study has suggested an ecacy of low-
molecular-weight heparin as an antifibrotic in
humans. Patients with HBV infection who were
treated with 3 weeks of heparin showed improved
serum levels of alanine aminotransferase and
bilirubin, with a reduction in serum hyaluronic
acid and Type IV collagen concentrations.75 Long-
term use of heparin as an antifibrotic agent in
humans may not be practical because of side
eects including osteopaenia, thrombocytopenia,
and idiopathic hepatitis. A multicentre Phase
II study evaluated the antifibrotic eect
of warfarin anticoagulation in transplanted
HCV patients with cirrhosis. Interim results
demonstrated a reduction in fibrosis scores
at 1-year post-transplantation in warfarinised
patients compared to those who were not.76
Angiotensin Converting
Enzyme Inhibitors
The role of angiotensin converting enzyme
(ACE) inhibitors as antifibrotic agents has been
explored with some success. Angiotensinogen
and angiotensin-1 are present in hepatocytes
and are highly activated in chronic liver disease.
Specifically, angiotensin II receptors are
upregulated during chronic liver injury resulting
in HSC activation. Activation of the renin–
angiotensin–aldosterone system also occurs
within the liver, triggering oxidative stress
and inflammatory cell release contributing to
fibrogenesis.77 The use of ACE inhibitors in
preventing HSC, and renin–angiotensin system
activation in preventing fibrogenesis, is a promising
treatment option. This has been supported in
animal studies, which demonstrated that ACE
inhibitor use in CCl4 treated rats ameliorated levels
of oxidative stress, hepatic inflammation, and
hepatic fibrosis.78 A meta-analysis by Kim et al.78
confirmed the benefits of their use in reducing
hepatic fibrosis in humans. This meta-analysis
only included studies in which intervention groups
used ACE inhibitors or angiotensin receptor
blockers and compared their eect to placebo.
Histological changes in fibrosis were the primary
outcome. ACE inhibitors lowered fibrosis scores,
serum fibrosis markers including TGF-β-1,
collagen Types I and IV, TIMP-1, and MMP-2.
Significantly, they were shown to be safe with no
significant dierences in renal function in those
who received ACE inhibitors versus those who
did not.78
Farnesoid X Receptor Agonists
The potent farnesoid X receptor (FXR) agonist
obeticholic acid has been shown to have
antifibrotic eects, especially in those with primary
biliary cirrhosis and nonalcoholic steatohepatitis,
histologically characterised by the presence of
fibrosis. FXR is present on HSC and is involved
in cellular regulation and activation. FXR
agonists, therefore, have the ability to inhibit HSC
activation and hepatic fibrogenesis.79 Obeticholic
acid use in TAA-treated rats decreased hepatic
inflammation and fibrogenesis, as well as portal
pressure and intrahepatic vascular resistance.
There was also decreased profibrotic cytokine
activity, assessed by TGF-β-1, CTGF, and
PDGF.80 The FLINT trial, a Phase IIb nonalcoholic
steatohepatitis study, compared those treated
with 72 weeks of obeticholic acid versus placebo
and fibrosis improvement was the primary
outcome of the study. It assessed for statistically
significant improvement in hepatic fibrosis
and fibrosis scores observed in the treatment
arm compared to those not being treated.79
Although the antifibrotic properties of FXR
agonists are significant, mild-to-moderate
side eects including pruritis, dyslipidaemia,
fatigue, headache, and gall stone disease have
been reported, which may limit their use in
clinical practice.79
EUROPEAN MEDICAL JOURNAL • December 2019 EMJ EUROPEAN MEDICAL JOURNAL
114
References
1. Pimpin L et al. Burden of liver disease
in Europe: Epidemiology and analysis
of risk factors to identify prevention
policies. J Hepatol. 2018;69(3):718-35.
2. Pinzani M. Pathophysiology of Liver
Fibrosis. Dig Dis. 2015;33(4):492-7.
3. Calvaruso V et al. Coagulation and
fibrosis in chronic liver disease. Gut.
2008;57(12):1722-7.
4. Wallace K et al. Liver Fibrosis.
Biochem. 2008;411:1-18.
5. Younossi ZA et al. Assessment of
utilities and health-related quality
of life in patients with chronic
liver disease. Am J Gastroenterol.
2001;96(2):579-83.
6. Lackner C, Tiniakos D. Fibrosis
and alcohol-related liver disease. J
Hepatol. 2019;70(2):294-304.
7. Bonnard P et al. Documented
rapid course of hepatic fibrosis
between two biopsies in patients
coinfected by HIV and HCV despite
high CD4 cell count. J Viral Hepat.
2007;14(11):806-11.
8. Wells RG. Cellular sources
of extracellular matrix in
hepatic fibrosis. Clin Liver Dis.
2008;12(4):759-68.
9. Schuppan D et al. Matrix as a
modulator of hepatic fibrogenesis.
Semin Liver Dis. 2001;21(3):351-72.
10. Hernandez-Gea V and Friendman SL.
Pathogenesis of liver fibrosis. Annu
Rev Pathol. 2010;6(1):425-56.
11. Yanguas SC et al. Experimental
models of liver fibrosis. Arch Toxicol.
2016;90(5):1025-48.
12. Gressner AM. The cell biology of
liver fibrogenesis – An imbalance
of proliferation, growth arrest and
apoptosis of myofibroblasts. Cell
Tissue Res. 1998;292(3):447-52.
13. Freidman SL. Evolving
challenges in hepatic fibrosis.
Nat Rev Gastroenterol Hepatol.
2010;7(8):425-36.
14. Higashi T. Hepatic stellate cells as key
target in liver fibrosis. Adv Drug Deliv
Rev. 2017;121:27-42.
15. Fernández M. Angiogenesis in liver
disease. J Hepatol. 2009;50(3):604-
20.
16. Henderson NC and Iredale JP. Liver
fibrosis: Cellular mechanisms of
progression and resolution. Clin Sci.
2007;112(5):265-80.
17. Seki E et al. Hepatic inflammation
and fibrosis: Functional links
and key pathways. Hepatology.
2015;61(3):1066-79.
18. Baeck C et al. Pharmacological
inhibition of the chemokine C-C
motif chemokine ligand 2 (monocyte
chemoattractant protein 1)
accelerates liver fibrosis regression
by suppressing Ly-6C(+) macrophage
infiltration in mice. Hepatology.
2014;59:1060-72.
19. Seki E et al. CCR1 and CCR5 promote
hepatic fibrosis in mice. J Clin Invest.
2009;119:1858-70.
20. Aoyama T et al. Nicotinamide
adenine dinucleotide phosphate
oxidase in experimental liver fibrosis:
GKT137831 as a novel potential
therapeutic agent. Hepatology.
2012;56(6):2316-27.
21. Carpino G et al. Alpha-SMA
expression in hepatic stellate
cells and quantitative analysis of
hepatic fibrosis in cirrhosis and in
recurrent chronic hepatitis after
liver transplantation. Dig Liver Dis.
2005;37(5):349-56.
22. Friedman SL. Molecular regulation of
hepatic fibrosis, an integrated cellular
response to tissue injury. J Biol Chem.
2000;275(4):2247-50.
23. Friedman SL. Mechanisms of Hepatic
Fibrogenesis. Gastroenterology.
2008;134(6):1655-69.
24. Ying HZ et al. PDGF signalling
pathway in hepatic fibrosis
pathogenesis and therapeutics
(review). Mol Med Rep.
2017;16(6):7879-89.
25. Friedman SL. Hepatic stellate
cells: Protean, multifunctional, and
enigmatic cells of the liver. Physiol
Rev. 2008;88(1):125-72.
26. Olaso E et al. Discoidin domain
receptor 2 regulates fibroblast
proliferation and migration through
the extracellular matrix in association
with transcriptional activation of
matrix metalloproteinase-2. J Biol
Chem. 2002;277(5):3606-13.
27. Tangkijvanich P et al. Wound-induced
migration of rat hepatic stellate cells
Thiazolidinediones
Thiazolidinediones, including pioglitazone and
rosiglitazone, are peroxisome proliferator-
activated receptors (PPAR) agonists and are
routinely used in the treatment of diabetes. They
are selective ligands for nuclear transcription
factor PPAR, expressed in HSC. PPAR activation
downregulates the ability to proliferate and
migrate in response to PDGF and reduces
expression of TGF-β-1, procollagen 1, fibronectin,
and TIMP, inhibiting the fibrogenic process.
Thiazolidinediones have been shown to be
eective antifibrotic agents in CCl4 treated
rats, with positive improvements in histology
and profibrotic cytokine profiling.81 A meta-
analysis of 8 randomised clinical trials, including
516 patients diagnosed radiologically or
histologically, concluded that thiazolidinedione
use for 2 years reversed advanced fibrosis
and improved fibrosis stage, even in those
without diabetes.82
CONCLUSION
Our understanding of the pathophysiology
of liver fibrosis continues to grow. Numerous
experimental studies have demonstrated
agents that may have therapeutic potential as
antifibrotic agents. Despite this, there are still few
candidates that have been successfully trialled
in human clinical trials that have demonstrated
a clear and significant antifibrotic eect.
More recently, potential therapies have shown
increasing promise and further studies are
required to ascertain whether these therapies
will be translated to clinical practice. At present,
removal of the insult resulting in fibrosis remains a
key strategy in its reduction and prevention.
Creative Commons Attribution-Non Commercial 4.0 December 2019 • EUROPEAN MEDICAL JOURNAL
115
is modulated by endothelin-1 through
rho-kinase-mediated alterations
in the acto-myosin cytoskeleton.
Hepatology. 2001;33(1):74-80.
28. Hashmi AZ et al. Adenosine inhibits
cytosolic signals and chemotaxis
in hepatic stellate cells. Am J
Physiol Gastrointest Liver Physiol.
2007;292(1):G395-401.
29. Xu F et al. TGF-β/SMAD pathway and
its regulation in hepatic fibrosis. J
Histochem Cytochem. 2016;64(3):157-
67.
30. Schuppan D et al. Liver fibrosis:
Direct antifibrotic agents and
targeted therapies. Matrix Biol.
2018;68-69:435-51.
31. Bataller R et al. NADPH oxidase
signal transduces angiotensin II in
hepatic stellate cells and is critical
in hepatic fibrosis. J Clin Invest.
2003;112(9):1383-94.
32. Rockey DC et al. Rat hepatic
lipocytes express smooth muscle
actin upon activation in vivo and in
culture. J Submicrosc Cytol Pathol.
1992;24(2):193-203.
33. Iwakiri Y et al. Vascular pathobiology
in chronic liver disease and cirrhosis –
Current status and future directions.
J Hepatol. 2014;61(4):912-24.
34. 34. Benyon RC et al. Extracellular
matrix degradation and the role of
hepatic stellate cells. Semin Liver Dis
2001;21:373-84.
35. Duarte S et al. Matrix
metalloproteinases in liver injury,
repair and fibrosis. Matrix Biol.
2015;0:147-56.
36. Iredale JP. Hepatic stellate cell
behavior during resolution of
liver injury. Semin Liver Dis.
2001;21(3):427-36.
37. Herbst H et al. Tissue inhibitor of
metalloproteinase-1 and -2 RNA
expression in rat and human liver
fibrosis. Am J Pathol. 1997;150:1647-
59.
38. 38. Bonacchi A et al. The chemokine
CCL21 modulates lymphocyte
recruitment and fibrosis in chronic
hepatitis C. Gastroenterology.
2003;125(4):1060-76.
39. Schwabe RF et al. Human hepatic
stellate cells express CCR5 and
RANTES to induce proliferation and
migration. Am J Physiol Gastrointest
Liver Physiol. 2003;285(5):G949-58.
40. Shang L. Human hepatic stellate
cell isolation and characterisation. J
Gastroenterol. 2018;53(1):6-17.
41. 41. Xu L. Human hepatic stellate
cell lines, LX-1 and LX-2: New tools
for analysis of hepatic fibrosis. Gut.
2005;54(1):142-51.
42. Yanguas SC et al. Experimental
models of liver fibrosis. Arch Toxicol.
2016;90(5):1025-48.
43. Weiler-Normann C et al. Mouse
models of liver fibrosis. Z
Gastroenterol. 2007;45(1):43-50.
44. Morrione TG. Factors influencing
collagen content in experimental
cirrhosis. Am J Pathol.
1949;25(2):273-85.
45. Hillebrandt S et al. Genome-wide
analysis of hepatic fibrosis in inbred
mice identifies the susceptibility
locus Hfib1 on chromosome 15.
Gastroenterology. 2002;123(6):2041-
51.
46. Kim KH. The antifibrotic eect of
TGF-beta1 siRNAs in murine model of
liver cirrhosis. Biochem Biophys Res
Commun. 2006;343(4):1072-78.
47. Delire B et al. Animal Models for
Fibrotic Liver Diseases: What We
Have, What We Need, and What Is
under Development. J Clin Transl
Hepatol. 2015;3(1):53-66.
48. Zimmerman T et al. Biochemical and
morphological studies on production
and regression of experimental liver
cirrhosis induced by thioacetamide
in Uje: WIST rats. Z Versuchstierkd.
1987;30(5-6):165-80.
49. Jenkins SA et al. A
dimethylnitrosamine-induced model
of cirrhosis and portal hypertension
in the rat. J Hepatol. 1985;1:489-99.
50. Kitamura K et al. Pathogenic
roles of tumor necrosis factor
receptor p55-mediated signals
in dimethylnitrosamine-induced
murine liver fibrosis. Lab Invest.
2002;82(5):571-83.
51. Magee PN et al. The production
of malignant primary hepatic
tumours in the rat by feeding
Dimethylnitrosamine. Br J Cancer.
1956;10(1):114-22.
52. Georgiev P et al. Characterization
of time-related changes after
experimental bile duct ligation. Br J
Surg. 2008;95(5):646-56.
53. Geerts AM et al. Comparison of
three research models of portal
hypertension in mice: Macroscopic,
histological and portal pressure
elevation. Int J Exp Pathol.
2008;89(4):251-63.
54. Oude Elferink RP and Paulusma CC.
Function and pathophysiological
importance of ABCB4 (MDR3
P-glycoprotein). Pflugers Arch.
2007;452(5):601-10.
55. Ikenaga N et al. A new Mdr2(-
/-) mouse model of sclerosing
cholangitis with rapid fibrosis
progression, early-onset portal
hypertension, and liver cancer. Am J
Pathol. 2015;185(2):325-34.
56. Trauner M et al. MDR3 (ABCB4)
defects: A paradigm for the genetics
of adult cholestatic syndromes.
Semin Liver Dis. 2007;27(1):77-98.
57. Bansal MB and Chamroonkul N.
Antifibrotics in liver disease: Are we
getting closer to clinical use? Hepatol
Int. 2019;13(1):25-39.
58. Ebrahimi H et al. New concepts on
reversibility and targeting of liver
fibrosis; A review article. Middle East
J Dig Dis. 2018;10(3):133-48.
59. Clichici S et al. Silymarin inhibits the
progression of fibrosis in the early
stages of liver injury in CCl₄-treated
rats. J Med Food. 2015;18(3):290-8.
60. De Avelar CR et al. Eect of silymarin
on biochemical indicators in patients
with liver disease: Systematic
review with meta-analysis. World J
Gastroenterol. 2017;23(27):5004-17.
61. Kheong W et al. A randomized
trial of silymarin for the treatment
of nonalcoholic steatohepatitis.
Clin Gastroenterol Hepatol.
2017;15(12):1940-9.
62. Modi AA et al. Increased caeine
consumption is associated with
reduced hepatic fibrosis. Hepatology.
2010;51(1):201-9.
63. Drano JA et al. How does coee
prevent liver fibrosis? Biological
plausibility for recent epidemiological
observations. Hepatology.
2014;60(2):464-7.
64. Shim SG et al. Caeine attenuates
liver fibrosis via defective adhesion
of hepatic stellate cells in cirrhotic
model. J Gastroenterol Hepatol.
2013;28(12):1877-84.
65. Goel A et al. Curcumin as
“Curecumin:” From kitchen to clinic.
Biochem Pharmacol. 2008:75(4):787-
809.
66. 66. Fu Y et al. Curcumin protects
the rat liver from CCl4-caused injury
and fibrogenesis by attenuating
oxidative stress and suppressing
inflammation. Mol Pharmocol.
2008;73(2):399-409.
67. Zhang Z et al. Curcumin modulates
cannabinoid receptors in liver fibrosis
in vivo and inhibits extracellular
matrix expression in hepatic stellate
cells by suppressing cannabinoid
receptor Type-1 in vitro. Eur J
Pharmacol. 2013:721(1-3):133-40.
68. Wright M et al. Factor V Leiden
polymorphism and the rate of fibrosis
development in chronic hepatitis C
virus infection. Gut. 2003;52(8):1206-
10.
69. Cheng JYK and Wong GLH.
Advances in the diagnosis and
treatment of liver fibrosis. Hepatoma
Res. 2017;3:156-69.
70. Coughlin SR. Thrombin signalling and
protease-activated receptors. Nature.
2000;407(6801):258-64.
71. Duplantier JG et al. A role of
thrombin in liver fibrosis. Gut.
2004;53(11):1682-7.
72. Dhar A et al. Thrombin and factor
Xa link the coagulation system with
liver fibrosis. BMC Gastroenterol.
2018;18:60.
EUROPEAN MEDICAL JOURNAL • December 2019 EMJ EUROPEAN MEDICAL JOURNAL
116
73. Cerini F et al. Enoxaparin reduces
hepatic vascular resistance and portal
pressure in cirrhotic rats. J Hepatol.
2016;64(4):834-42.
74. Anstee QM et al. Coagulation
status modulates murine hepatic
fibrogenesis: Implications for the
development of novel therapies. J
Thromb Haemost. 2008;6(8):1336-43.
75. Shi J et al. Eects of heparin on
liver fibrosis in patients with chronic
hepatitis B. World J Gastroenterol.
2003;9(7):1611-4.
76. Dhar et al. LP11: Warfarin
anticoagulation for liver fibrosis in
patients transplanted for hepatitis
C (WAFT-C): Results at one year. J
Hepatol. 2015;62(2):S268-9.
77. Reza HM et al. Angiotensin-
converting enzyme inhibitor prevents
oxidative stress, inflammation, and
fibrosis in carbon tetrachloride-
treated rat liver. Toxicol Mech
Methods. 2016;26(1):46-53.
78. Kim G et al. Renin-angiotensin system
inhibitors and fibrosis in chronic liver
disease: A systematic review. Hepatol
Int. 2016;10(5):819-28.
79. Abenavoli L et al. Obeticholic
acid: A new era in the treatment
of nonalcoholic fatty liver
disease. Pharmaceuticals (Basel).
2018;11(4):104.
80. Verbeke L et al. FXR agonist
obeticholic acid reduces hepatic
inflammation and fibrosis in
a rat model of toxic cirrhosis.
2016;6:33453.
81. Marra F. Thiazolidinediones and
hepatic fibrosis: don’t wait too long.
Gut. 2006;55(7):917-9.
82. Musso G et al. Thiazolidinediones
and advanced liver fibrosis in
nonalcoholic steatohepatitis: A
meta-analysis. JAMA Intern Med.
2017;177(5):633-40.
FOR REPRINT QUERIES PLEASE CONTACT: +44 (0) 1245 334450
... Genetik bozukluklar, kronik viral enfeksiyon (Hepatit B ve C virisü), toksinler (alkol kullanımı), immünopatolojik / otoimmün ataklar, vasküler metabolik bozukluklar, kolestaz, safra asidi bileşimindeki veya konsantrasyonundaki değişiklikler, venöz tıkanıklık, parazit enfeksiyonları gibi etiyolojik faktörlerden biri bile karaciğeri hepatik fibrozise yatkın hale getirebilir. Ayrıca endoplazmik retikulum stresi, mitokondriyal fonksiyonun değişmesi, oksidatif stresi provoke eden aşırı yağ birikimi ve alkolsüz yağlı karaciğer hastalığı hepatik fibrozisin başlaması ve ilerlemesine neden olabilir (27)(28)(29)(30)(31)(32)(38)(39)(40)(41)(42)(43). Bu etkenler iltihaplanmayı tetikler, ektraselüler matris (ECM) bileşenlerinin sentezinin ve birikiminin artmasına ve dolayısıyla rejenerasyonun ve yara iyileşme yanıtlarının bozulmasına neden olur. ...
... Trombositlerin aktivasyonu, inflamatuar hücreler, hepatik stellat hücreler (HSC) ve diğer ECM üreten hücreler fibroblastlar (kollajen tip 1,2), hepatositler, kolanjiyositler ve kemik iliğinden türetilen hücreler bu süreçte görev alır. Karaciğer fibrozisi geri dönüşümlü bir yara iyileşmesi olup hücre dışı matris (ECM) birikimi ile karakterize bir tepkimedir (27)(28)(29)(30)(31)37,41,(43)(44)(45)(46)(47). Hepatositlerin inflamasyona cevabı hepatik fibrozis fizyopatolojisinde belirleyici rol oynayan monositler ve makrofajlar gibi hem pro-hem de anti-inflamatuar hücrelerin salımını içerir. ...
... Oturmuş bir fibroziste intrahepatik venöz akışta ciddi kesinti sonucu portal hipertansiyon ve karaciğer yetmezliği ile klinikte artmış mortalite, morbidite ve yaşam kalitesinde ciddi azalma görülür. Hepatik fibroziste prognozu belirleyen anahtar noktalar altta yatan kronik karaciğer hastalığı ve bağımsız faktörlerdir (33,34,41). Kronik karaciğer hastalığının yönetimi büyük ölçüde etiyolojiye özgü tedavilere odaklanmıştır. ...
... Destruction of hepatocytes initiates fibrosis that results in the release of inflammatory cytokines and the activation of liver macrophages (Kupffer cells or KCs); activation and transformation of HSCs into fibrogenic and proliferative MFs; and leukocyte migration to the injury site. Activated HSCs release reactive oxygen species (ROS), cytokines, and chemokines in addition to fibers and a significant amount of ECM proteins, that is, Type I, III, and IV collagen, elastin, fibronectin, and laminin, as well as proteoglycans, which promote inflammation process [13], [15]. Liver fibrosis has numerous intricate mechanisms that involve both cellular and extracellular signaling [15], including transforming growth factor (TGF-β), signal transducer and activator of transcription (STAT3), Wnt/β-catenin, apoptotic, matrix metalloproteinases (MMPs) dan tissue Inhibitors of MMPs (TIMPs), Syndecan-1 (SDC1), and regulation of high-mobility group box 1 (HMGB1) pathways. ...
... Activated HSCs release reactive oxygen species (ROS), cytokines, and chemokines in addition to fibers and a significant amount of ECM proteins, that is, Type I, III, and IV collagen, elastin, fibronectin, and laminin, as well as proteoglycans, which promote inflammation process [13], [15]. Liver fibrosis has numerous intricate mechanisms that involve both cellular and extracellular signaling [15], including transforming growth factor (TGF-β), signal transducer and activator of transcription (STAT3), Wnt/β-catenin, apoptotic, matrix metalloproteinases (MMPs) dan tissue Inhibitors of MMPs (TIMPs), Syndecan-1 (SDC1), and regulation of high-mobility group box 1 (HMGB1) pathways. ...
Article
Full-text available
Liver fibrosis is a disease process that, without adequate treatment, can lead to liver failure and can be life-threatening. This disease is reversible and appropriate therapy can prevent further liver damage. Liver transplant therapy is the only treatment for an end-stage liver disease that works, but it has various obstacles and limitations in its implementation. Therefore, nowadays, mesenchymal stem cells (MSCs) have become a hope of therapy for liver fibrosis. Our literature review describes the pathomechanism of liver fibrosis and the steps of its resolution, accompanied by the possible role of MSCs in supporting the process. The activation of several complex pathways regulates liver fibrosis, and its resolution, involving Transforming Growth Factor (TGF)-β, signal transducer and activator of transcription-3, and Wnt/β-catenin signaling is involved in Hepatic Stellate Cells (HSCs) activation, which are precursors of myofibroblasts (MFs) and causes fibrosis. The presence of the High-mobility group box-1 pathway, which also induces the production of proinflammatory cytokines and the role of matrix metalloproteinases (MMPs)/tissue Inhibitors of MMPs s and Syndecan-1, is incorporated into the extracellular matrix (ECM). In repairing liver damage, four steps of liver fibrosis resolution are required, such as preventing further damage, restoring the intrahepatic balance of inflammation, removing and inactivating MFs, and ECM degradation associated with arresting the eight pathways of the fibrosis mechanism. MSCs can help resolve liver fibrosis and speed up wound healing, increase hepatocyte survival, and suppress HSCs activation by blocking fibrosis mechanism pathways such as TGF-β and pro-inflammatory factors such as tumor necrosis factor-alpha, interferon-gamma, IL-6, IL-17, and IL-23, in addition to an elevated level of an anti-inflammatory factor like IL-10.
... 18 A great deal of work has been done to understand the pathophysiology of fibrosis, which has led to the identification of prospective targets for antifibrotic drugs that could either slow down or reverse fibrosis. 19 Pitavastatin is a cholesterol-lowering agent (statin) that was approved in 2009. It is also one of the foundations for treating and preventing atherosclerotic cardiovascular disease. ...
Article
Full-text available
Liver fibrosis is a common chronic hepatic disease. This study aimed to investigate the effect of pitavastatin (Pit) against thioacetamide (TAA)‐induced liver fibrosis. Rats were divided into four groups: (1) control group; (2) TAA group (100 mg/kg, i.p.) three times weekly for 2 weeks; (3 and 4) TAA/Pit‐treated group, in which Pit was administered orally (0.4 and 0.8 mg/kg/day) for 2 weeks following TAA injections. TAA caused liver damage manifested by elevated serum transaminases, reduced albumin and histological alterations. Hepatic malondialdehyde (MDA) was increased, and glutathione (GSH) and superoxide dismutase (SOD) were decreased in TAA‐administered rats. TAA upregulated the inflammatory markers NF‐κB, NF‐κB p65, TNF‐α and IL‐6. Treatment with Pit ameliorated serum transaminases, elevated serum albumin and prevented histopathological changes in TAA‐intoxicated rats. Pit suppressed MDA, NF‐κB, NF‐κB p65, the inflammatory cytokines and PI3K mRNA in TAA‐intoxicated rats. In addition, Pit enhanced hepatic antioxidants and boosted the nuclear factor erythroid 2–related factor 2 (Nrf2) and heme oxygenase‐1 (HO‐1) mRNA. Moreover, immunohistological studies supported the ability of Pit to reduce liver fibrosis via suppressing p ‐AKT expression. In conclusion, Pit effectively prevents TAA‐induced liver fibrosis by attenuating oxidative stress and the inflammatory response. The hepatoprotective efficacy of Pit was associated with the upregulation of Nrf2/HO‐1 and downregulation of NF‐κB and PI3K/Akt signalling pathways.
... CCl4 treatment leads to a combination of effects causing hepatocyte damage via lipid peroxidation, HSCs activation, Kupffer cells activation, TGF-β1 upregulation and increased oxidative stress [54]. The achievement of the HF in our experimental conditions was validated by macroscopic and microscopic morphological analyses, biochemical evaluations, and alterations in the expression of the genes involved in the HF processes. ...
Article
Full-text available
Hepatic fibrosis (HF) is a major cause of liver-related disorders and together with cancer-associated fibroblasts can favor liver cancer development by modulating the tumor microenvironment. Advanced HF, characterized by an excess of extracellular matrix (ECM), is mediated by TGF-β1, that activates hepatic stellate cells (HSCs) and fibroblasts. A TGF-β1 receptor inhibitor, LY2157299 or Galunisertib (GLY), has shown promising results against chronic liver progression in animal models, and we show that it can be further improved by enhancing GLYs bioavailability through encapsulation in polymeric polygalacturonic-polyacrylic acid nanomicelles (GLY-NMs). GLY-NMs reduced HF in an in vivo rat model of liver fibrosis induced by intraperitoneal injection of CCl4 as shown by the morphological, biochemical, and molecular biology parameters of normal and fibrotic livers. Moreover, GLY-NM was able to induce recovery from HF better than free GLY. Indeed, the encapsulated drug reduces collagen deposition, hepatic stellate cells (HSCs) activation, prevents fatty degeneration and restores the correct lobular architecture of the liver as well as normalizes the serum parameters and expression of the genes involved in the onset of HF. In summary, GLY-NM improved the pharmacological activity of the free TGF-β1 inhibitor in the in vivo HF treatment and thus is a candidate as a novel therapeutic strategy.
... Attacking these PUFAs lead to propagate a chain reaction resulting in peroxidative damage of cytoplasmic membrane lipids, leading to progression of liver damage with subsequent fibrosis, cirrhosis, and hepatocellular carcinoma (6) . Though significant progress has been made, up to our knowledge there were no anti-fibrotic therapy specifically target HSCs which responsible for extracellular matrix deposition after being differentiated into fibroblasts and myofibroblast (7) . Some drugs and /or combination of two drugs are now in phase III clinical trials for attenuation of primary biliary cirrhosis such as Obetoticholic acid a farnesoids-X-receptor (FXR) agonist, Bezafibrate, an agonist of PPAR in phase III clinical trials with ursodeoxycholic acid (UDCA) and other combination therapy (8) . ...
Article
Full-text available
ABSTRACT: Liver fibrosis is considered now as one of the most spread disease worldwide. It is attributed to different underlying causative agents such as viral infections, ethanol-induced liver steatosis, and non-ethanol-induced hepatic steatosis, autoimmune and inherited disorders. Hepatic fibrosis was known to behave as tissue repair mechanism in which the initiation occurred through complicated series of interrelated and regulated signaling. These signals involved interactions between different types of cells. Among these cells are hepatocytes, non-parenchymal cells such as hepatic stellate cells (HSCs), liver sinusoidal endothelial cells, Kupffer cells, biliary epithelial cells, liver associated lymphocytes, and the non-resident infiltrating immune cells. current work was aimed to investigate the possible potential hepatopretective effects of krill oil alone and in combination with silymarin against Carbone tetrachloride-induced liver fibrosis/injury in white albino rats. Moreover, fifty white albino rats of both genders were utilized in this study. During such study liver fibrosis/damage was induced by intraperitoneal (I.P) injection of Carbone tetrachloride (CCl4) 50% in olive oil 1ml/kg twice weekly for 6 consecutive weeks in the induction group. Krill oil alone and in combination with silymarin was administered orally concurrently with I.P CCl4 for 6 consecutive weeks in the treatment groups. At the end of treatment period all animals were killed ,serum and tissue samples were collected for subsequent analyses. Serum levels of aminotransferases (ALT,AST), albumin , total serum bilirubin (T.S.B), and total anti-oxidant capacity were measured spectrophotometrically. In addition tissue level (content) of liver hudroxyproline content (Hyp) was determined by ELISA and relative liver weight percentage (R.L.W%) was also estimated.Results were significantly revealed that krill oil potentiate the hepatoprotective effects of silymarin against Carbone tetrachloride-induced liver fibrosis/injury.
... Carbon tetrachloride (CCl 4 ) is frequently used to induce liver failure, 40-42 but liver function occasionally occurs when CCl 4 administration is stopped. 42 Conversely, TAA damage persists for more than 2 months after withdrawal, and induces pathological characteristics similar to human chronic liver dysfunction. TAA-induced liver injury models are thus more suitable than CCl 4 for longitudinal studies. ...
Article
Full-text available
Background and aims: Efficacy evaluations with preclinical magnetic resonance imaging (MRI) are uncommon, but MRI in the preclinical phase of drug development provides information that is useful for longitudinal monitoring. The study aim was to monitor the protective effectiveness of silymarin with multiparameter MRI and biomarkers in a thioacetamide (TAA)-induced model of liver injury in rats. Correlation analysis was conducted to assess compare the monitoring of liver function by MRI and biomarkers. Methods: TAA was injected three times a week for 8 weeks to generate a disease model (TAA group). In the TAA and silymarin-treated (TAA-SY) groups, silymarin was administered three times weekly from week 4. MR images were acquired at 0, 2, 4, 6, and 8 weeks in the control, TAA, and TAA-SY groups. Results: The area under the curve to maximum time (AUCtmax) and T2* values of the TAA group decreased over the study period, but the serological markers of liver abnormality increased significantly more than those in the control group. In the TAA-SY group, MRI and serological biomarkers indicated attenuation of liver function as in the TAA group. However, pattern changes were observed from week 6 to comparable levels in the control group with silymarin treatment. Negative correlations between either AUCtmax or T2* values and the serological biomarkers were observed. Conclusions: Silymarin had hepatoprotective effects on TAA-induced liver injury and demonstrated the usefulness of multiparametric MRI to evaluate efficacy in preclinical studies of liver drug development.
... Phosphorylated Smad2 and Smad3 bind to Smad4 to form a heterotrimeric complex, which translocates to the nucleus and transcribes genes involved in ECM synthesis and deposition [13]. Due to the importance of TGF-β signaling, ECM synthesis, and HSC transformation in the pathophysiology of liver fibrosis, state-of-the-art antifibrotic strategies that target the ECM [14] and HSCs [15] and stem cell-based therapies [16] that target TGF-β1 [17] and enhance antifibrotic efficacy [18] are currently used to treat liver fibrosis. ...
Article
Full-text available
This study was undertaken to investigate the inhibitory effects of granulocyte-macrophage colony-stimulating factor (GM-CSF) on dimethylnitrosamine (DMN)-induced liver fibrosis in rats. Liver fibrosis was induced in Sprague-Dawley rats by injecting DMN intraperitoneally (at 10 mg/kg of body weight) daily for three consecutive days per week for 4 weeks. To investigate the effect of GM-CSF on disease onset, GM-CSF (50 μg/kg of body weight) was co-treated with DMN for 2 consecutive days per week for 4 weeks (4-week groups). To observe the effect of GM-CSF on the progression of liver fibrosis, GM-CSF was post-treated alone at 5–8 weeks after the 4 weeks of DMN injection (8-week groups). We found that DMN administration for 4 weeks produced molecular and pathological manifestations of liver fibrosis, that is, it increased the expressions of collagen type I, alpha-smooth muscle actin (α-SMA), and transforming growth factor-β1 (TGF-β1), and decreased peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. In addition, elevated serum levels of aspartate aminotransferase (AST), total bilirubin level (TBIL), and decreased albumin level (ALB) were observed. In both the 4-week and 8-week groups, GM-CSF clearly improved the pathological liver conditions in the gross and histological observations, and significantly recovered DMN-induced increases in AST and TBIL and decreases in ALB serum levels to normal. GM-CSF also significantly decreased DMN-induced increases in collagen type I, α-SMA, and TGF-β1 and increased DMN-induced decreases in PPAR-γ expression. In the DMN groups, survival decreased continuously for 8 weeks after DMN treatment for the first 4 weeks. GM-CSF showed a survival benefit when co-treated for the first 4 weeks but a marginal effect when post-treated for 5–8 weeks. In conclusion, co-treatment of GM-CSF showed therapeutic effects on DMN-induced liver fibrosis and survival rates in rats, while post-treatment efficiently blocked liver fibrosis.
... Attacking these PUFAs lead to propagate a chain reaction resulting in peroxidative damage of cytoplasmic membrane lipids, leading to progression of liver damage with subsequent fibrosis, cirrhosis, and hepatocellular carcinoma (6) . Though significant progress has been made, up to our knowledge there were no anti-fibrotic therapy specifically target HSCs which responsible for extracellular matrix deposition after being differentiated into fibroblasts and myofibroblast (7) . Some drugs and /or combination of two drugs are now in phase III clinical trials for attenuation of primary biliary cirrhosis such as Obetoticholic acid a farnesoids-X-receptor (FXR) agonist, Bezafibrate, an agonist of PPAR in phase III clinical trials with ursodeoxycholic acid (UDCA) and other combination therapy (8) . ...
Article
Full-text available
Liver fibrosis is considered now as one of the most spread disease worldwide. It is attributed to different underlying causative agents such as viral infections, ethanol-induced liver steatosis, and non-ethanol-induced hepatic steatosis, autoimmune and inherited disorders. Hepatic fibrosis was known to behave as tissue repair mechanism in which the initiation occurred through complicated series of interrelated and regulated signaling. These signals involved interactions between different types of cells. Among these cells are hepatocytes, non-parenchymal cells such as hepatic stellate cells (HSCs), liver sinusoidal endothelial cells, Kupffer cells, biliary epithelial cells, liver associated lymphocytes, and the non-resident infiltrating immune cells. current work was aimed to investigate the possible potential hepatopretective effects of krill oil alone and in combination with silymarin against Carbone tetrachloride-induced liver fibrosis/injury in white albino rats. Moreover, fifty white albino rats of both genders were utilized in this study. During such study liver fibrosis/damage was induced by intraperitoneal (I.P) injection of Carbone tetrachloride (CCl4) 50% in olive oil 1ml/kg twice weekly for 6 consecutive weeks in the induction group. Krill oil alone and in combination with silymarin was administered orally concurrently with I.P CCl4 for 6 consecutive weeks in the treatment groups. At the end of treatment period all animals were killed ,serum and tissue samples were collected for subsequent analyses. Serum levels of aminotransferases (ALT,AST), albumin , total serum bilirubin (T.S.B), and total anti-oxidant capacity were measured spectrophotometrically. In addition tissue level (content) of liver hudroxyproline content (Hyp) was determined by ELISA and relative liver weight percentage (R.L.W%) was also estimated.Results were significantly revealed that krill oil potentiate the hepatoprotective effects of silymarin against Carbone tetrachloride-induced liver fibrosis/injury.
... This complex translocates to the nucleus and transcribes genes involved in ECM synthesis and deposition 13 . Owing to the importance of TGF-β signaling, ECM synthesis, and HSCs transformation in liver brosis pathophysiology, several anti-brotic strategies that target the ECM 14 and HSCs 15 , and stem cell-based therapies 16 that target TGF-β1 17 and enhance anti-brotic e cacy 18 are the state-of-the-art therapeutic modalities for the treatment of liver brosis. ...
Preprint
Full-text available
Granulocyte-macrophage colony-stimulating factor (GM-CSF) exerts several therapeutic pharmacological effects but its role in liver fibrosis has not yet been studied. The current study investigates the inhibitory effects of GM-CSF on dimethylnitrosamine (DMN)-induced liver fibrosis in rats. In this study, liver fibrosis was induced in Sprague-Dawley rats by intraperitoneal injections of DMN (10 mg/kg of body weight) for three consecutive days per week for four weeks. To see the inhibitory effects on disease onset, GM-CSF (50 µg/kg of body weight) was injected for 2 consecutive days per week for 4 weeks along with DMN, while to see the therapeutic effects on disease progression, the GM-CSF injection was set forth at 4 weeks after the DMN injection. We found that DMN administration produced characteristics of molecular and pathological manifestations of liver fibrosis in rats including increased expressions of collagen I, alpha-smooth muscle actin (α-SMA), and transforming growth factor beta 1 (TGF-β1), and decreased PPAR-γ expression. Similarly, elevated serum levels of aspartate aminotransferase (AST), total bilirubin level (TBIL), and decreased albumin level (ALB) were observed. Treatment with GM-CSF improved the pathological liver conditions and significantly inhibited the elevated AST and TBIL, and increased ALB serum levels to normal. GM-CSF significantly decreased collagen I, α-SMA, and TGF-β1 expression and increased peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. In conclusion, GM-CSF reduced the DMN-induced rat liver fibrosis by inhibiting TGF-β1 signaling pathway.
Article
Full-text available
The main treatments for patients with nonalcoholic fatty liver disease (NAFLD) are currently based on lifestyle changes, including ponderal decrease and dietary management. However, a subgroup of patients with nonalcoholic steatohepatitis (NASH), who are unable to modify their lifestyle successfully, may benefit from pharmaceutical support. Several drugs targeting pathogenic mechanisms of NAFLD have been evaluated in clinical trials for the treatment of NASH. Farnesoid X receptor (FXR) is a nuclear key regulator controlling several processes of the hepatic metabolism. NAFLD has been proven to be associated with abnormal FXR activity. Obeticholic acid (OCA) is a first-in-class selective FXR agonist with anticholestatic and hepato-protective properties. Currently, OCA is registered for the treatment of primary biliary cholangitis. However, promising effects of OCA on NASH and its metabolic features have been reported in several studies.
Article
Full-text available
Currently, liver fibrosis and its complications are regarded as critical health problems. With the studies showing the reversible nature of liver fibrogenesis, scientists have focused on understanding the underlying mechanism of this condition in order to develop new therapeutic strategies. Although hepatic stellate cells are known as the primary cells responsible for liver fibrogenesis, studies have shown contributing roles for other cells, pathways, and molecules in the development of fibrosis depending on the etiology of liver fibrosis. Hence, interventions could be directed in the proper way for each type of liver diseases to better address this complication. There are two main approaches in clinical reversion of liver fibrosis; eliminating the underlying insult and targeting the fibrosis process, which have variable clinical importance in the treatment of this disease. In this review, we present recent concepts in molecular pathways of liver fibrosis reversibility and their clinical implications.
Article
Full-text available
The burden of liver disease in Europe continues to grow. We aimed to describe the epidemiology of liver diseases and their risk factors in European countries, identifying public health interventions that could impact on these risk factors to reduce the burden of liver disease. As part of the HEPAHEALTH project we extracted information on historical and current prevalence and mortality from national and international literature and databases on liver disease in 35 countries in the World Health Organization European region, as well as historical and recent prevalence data on their main determinants; alcohol consumption, obesity and hepatitis B and C virus infections. We extracted information from peer-reviewed and grey literature to identify public health interventions targeting these risk factors. The epidemiology of liver disease is diverse, with variations in the exact composition of diseases and the trends in risk factors which drive them. Prevalence and mortality data indicate that increasing cirrhosis and liver cancer may be linked to dramatic increases in harmful alcohol consumption in Northern European countries, and viral hepatitis epidemics in Eastern and Southern European countries. Countries with historically low levels of liver disease may experience an increase in non-alcoholic fatty liver disease in the future, given the rise of obesity across most European countries. Liver disease in Europe is a serious issue, with increasing cirrhosis and liver cancer. The public health and hepatology communities are uniquely placed to implement measures aimed at reducing their causes: harmful alcohol consumption, child and adult obesity, and chronic infection with hepatitis viruses, which will in turn reduce the burden of liver disease.
Article
Full-text available
Background: Thrombin activates hepatic stellate cells via protease-activated receptor-1. The role of Factor Xa (FXa) in hepatic fibrosis has not been elucidated. We aimed to evaluate the impact of FXa and thrombin in vitro on stellate cells and their respective inhibition in vivo using a rodent model of hepatic fibrosis. Methods: HSC-LX2 cells were incubated with FXa and/or thrombin in cell culture, stained for αSMA and relative gene expression and gel contraction calculated. C57BL/6 J mice were administered thioacetamide (TAA) for 8 weeks with Rivaroxaban (n = 15) or Dabigatran (n = 15). Control animals received TAA alone (n = 15). Fibrosis was scored and quantified using digital image analysis and hepatic tissue hydroxyproline estimated. Results: Stellate cells treated with FXa and thrombin demonstrated upregulation of procollagen, TGF-beta, αSMA and significant cell contraction (43.48%+/- 4.12) compared to culturing with FXa or thrombin alone (26.90%+/- 8.90, p = 0.02; 13.1%+/- 9.84, p < 0.001). Mean fibrosis score, percentage area of fibrosis and hepatic hydroxyproline content (2.46 vs 4.08, p = 0.008; 2.02% vs 3.76%, p = 0.012; 276.0 vs 651.3, p = 0.0001) were significantly reduced in mice treated with the FXa inhibitor compared to control mice. FXa inhibition was significantly more effective than thrombin inhibition in reducing percentage area of fibrosis and hepatic hydroxyproline content (2.02% vs 3.70%,p = 0.031; 276.0 vs 413.1,p = 0.001). Conclusions: FXa promotes stellate cell contractility and activation. Early inhibition of coagulation using a FXa inhibitor significantly reduces TAA induced murine liver fibrosis and may be a viable treatment for liver fibrosis in patients.
Article
Full-text available
The hepatic stellate cells (HSCs) localize at the space of Disse in the liver and have multiple functions. They are identified as the major contributor to hepatic fibrosis. Significant understanding of HSCs has been achieved using rodent models and isolated murine HSCs; as well as investigating human liver tissues and human HSCs. There is growing interest and need of translating rodent study findings to human HSCs and human liver diseases. However, species-related differences impose challenges on the translational research. In this review, we focus on the current information on human HSCs isolation methods, human HSCs markers, and established human HSC cell lines.
Article
Full-text available
The platelet‑derived growth factor (PDFG) signaling pathway exerts persistent activation in response to a variety of stimuli and facilitates the progression of hepatic fibrosis. Since this pathway modulates a broad spectrum of cellular processes, including cell growth, differentiation, inflammation and carcinogenesis, it has emerged as a therapeutic target for hepatic fibrosis and liver‑associated disorders. The present review exhibits the current knowledge of the role of the PDGF signaling pathway and its pathological profiles in hepatic fibrosis, and assesses the potential of inhibitors which have been investigated in the experimental hepatic fibrosis model, in addition to the clinical challenges associated with these inhibitors.
Article
Full-text available
Liver fibrosis is the center of diagnosis and management of essentially all chronic liver diseases. While liver biopsy examination still has a role in diagnosis and drug development, it is replaced by non-invasive assessments of liver biopsy in majority of the clinical scenarios. Radiological approaches, namely transient elastography, acoustic radiation force impulse imaging, shear wave elastography, magnetic resonance elastography provide accurate diagnosis of advanced fibrosis and cirrhosis. Serum test formulae based on common laboratory parameters or more specialized parameters including those commercially available panels FibroTest®, FibroMeter® and Enhanced Liver Fibrosis are also available. Combining different modalities may further improve the accuracy. The role of all these non-invasive assessments has been further expanded from diagnostic to prognostic, e.g. risk prediction of hepatocellular carcinoma (HCC) by LSM-HCC score. Treatment of liver fibrosis can be achieved by controlling the underlying diseases, with chronic viral hepatitis as the most established disease model. Currently there are multiple clinical trials evaluating different treatment options to improve fibrosis in patients with non-alcoholic fatty liver disease. Specific anti-fibrotic treatment targets e.g. direct downregulation of hepatic stellate cell, collagen synthesis inhibitors and transforming growth factor-β antagonists have been tested in laboratory and pending further studies in clinical settings.
Article
Histological fibrosis stage is one of the most important prognostic factors in compensated and decompensated alcohol-related liver disease (ALD). Morphological assessment of fibrosis is useful for patient stratification, enabling individualised management, and for evaluation of treatment effects in clinical studies. In contrast to most chronic liver diseases where fibrosis is portal-based, fatty liver disease (FLD) of alcoholic or non-alcoholic aetiology (NAFLD) is associated with a centrilobular pattern of injury which leads to perivenular fibrosis and/or pericellular fibrosis. Progression of FLD drives expansive pericellular fibrosis, linking vascular structures and paving the way for the development of cirrhosis. At the cirrhotic stage, ongoing tissue damage leads to increasing fibrosis severity due to parenchymal loss and proliferation of fibrous scars. Histologic fibrosis staging systems have been devised, based on topography and the extent of fibrosis, for most chronic liver diseases. The utility of histological staging is reflected in different risks associated with individual fibrosis stages which cannot be reliably distinguished by non-invasive fibrosis assessment. In contrast to NAFLD, ALD-specific staging systems that enable the standardised prognostication required for clinical management and trials are lacking. Although morphological similarities between NAFLD and ALD exist, differences in clinical and histological features may substantially limit the utility of established NAFLD-specific staging systems for prognostication in ALD. This review summarises morphological features of fibrosis in ALD and compares them to other chronic liver diseases, particularly NAFLD. ALD-related fibrosis is examined in the context of pathogenetic mechanisms of fibrosis progression, regression and clinical settings that need to be considered in future prognostically relevant ALD staging approaches.
Article
The process of wound healing in response to chronic liver injury leads to the development of liver fibrosis. Regardless of etiology, the profound impact of the degree of liver fibrosis on the prognosis of chronic liver diseases has been well demonstrated. While disease-specific therapy, such as treatments for viral hepatitis, has been shown to reverse liver fibrosis and cirrhosis in both clinical trials and real-life practice, subsets of patients do not demonstrate fibrosis regression. Moreover, where disease-specific therapies are not available, the need for antifibrotics exists. Increased understanding into the pathogenesis of liver fibrosis sets the stage to focus on antifibrotic therapies attempting to: (1) Minimize liver injury and inflammation; (2) Inhibit liver fibrogenesis by enhancing or inhibiting target receptor–ligand interactions or intracellular signaling pathways; and (3) Promote fibrosis resolution. While no antifibrotic therapies are currently available, a number are now being evaluated in clinical trials, and their use is becoming closer to reality for select subsets of patients.
Article
Liver fibrosis and in particular cirrhosis are the major causes of morbidity and mortality of patients with chronic liver disease. Their prevention or reversal have become major endpoints in clinical trials with novel liver specific drugs. Remarkable progress has been made with therapies that efficiently address the cause of the underlying liver disease, as in chronic hepatitis B and C. Highly effective antiviral therapy can prevent progression or even induce reversal in the majority of patients, but such treatment remains elusive for the majority of liver patients with advanced alcoholic or nonalcoholic steatohepatitis, genetic or autoimmune liver diseases. Moreover, drugs that would speed up fibrosis reversal are needed for patients with cirrhosis, since even with effective causal therapy reversal is slow or the disease may further progress. Therefore, highly efficient and specific antifibrotic agents are needed that can address advanced fibrosis, i.e., the detrimental downstream result of all chronic liver diseases. This review discusses targeted antifibrotic therapies that address molecules and mechanisms that are central to fibrogenesis or fibrolysis, including strategies that allow targeting of activated hepatic stellate cells and myofibroblasts and other fibrogenic effector cells. Focus is on collagen synthesis, integrins and cells and mechanisms specific including specific downregulation of TGFbeta signaling, major extracellular matrix (ECM) components, ECM-crosslinking, and extracellular matrix (ECM)-receptors such as integrins and discoidin domain receptors, ECM-crosslinking and methods for targeted delivery of small interfering RNA, antisense oligonucleotides and small molecules to increase potency and reduce side effects. With an increased understanding of the biology of the ECM and liver fibrosis and an improved preclinical validation, the translation of these approaches to the clinic is currently ongoing. Application to patients with liver fibrosis and a personalized treatment is tightly linked to the development of noninvasive biomarkers of fibrosis, fibrogenesis and fibrolysis.