ArticlePDF AvailableLiterature Review

Role of liver sinusoidal endothelial cells in nonalcoholic fatty liver disease

Authors:
  • Université de Paris

Abstract

Non-alcoholic fatty liver disease (NAFLD) and its complications are an expanding health problem associated with the metabolic syndrome. Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells localized at the interface between the blood derived from the gut and the adipose tissue on the one side, and other liver cells on the other side. In physiological conditions, LSECs are gatekeepers of liver homeostasis. LSECs display anti-inflammatory and anti-fibrogenic properties by preventing Kupffer cell and hepatic stellate cell activation and regulating intrahepatic vascular resistance and portal pressure. This review focusses on changes occurring in LSECs in NAFLD and on their consequences on NAFLD progression and complications. Capillarization, namely the loss of LSEC fenestrae, and LSEC dysfunction, namely the loss of the ability of LSECs to generate vasodilator agents in response to increased shear stress both occur early in NAFLD. These LSEC changes favour steatosis development and set the stage for NAFLD progression. At the stage of non-alcoholic steatohepatitis, altered LSECs release inflammatory mediators and contribute to the recruitment of inflammatory cells, thus promoting liver injury and inflammation. Altered LSECs also fail to maintain hepatic stellate cell quiescence and release fibrogenic mediators, including Hedgehog signalling molecules, promoting liver fibrosis. Liver angiogenesis is increased in NAFLD and contributes to liver inflammation and fibrosis, but also to hepatocellular carcinoma development. Thus, improving LSEC health appears to be a promising approach to prevent NAFLD progression and complications.
Role of liver sinusoidal endothelial cells in non-alcoholic fatty
liver disease
Adel Hammoutene
1,2
, Pierre-Emmanuel Rautou
1,3,4,5,
Summary
Non-alcoholic fatty liver disease (NAFLD) and its complications are an expanding health problem asso-
ciated with the metabolic syndrome. Liver sinusoidal endothelial cells (LSECs) are highly specialized
endothelial cells localized at the interface between the blood derived from the gut and the adipose tissue
on the one side, and other liver cells on the other side. In physiological conditions, LSECs are gatekeepers
of liver homeostasis. LSECs display anti-inflammatory and anti-fibrogenic properties by preventing
Kupffer cell and hepatic stellate cell activation and regulating intrahepatic vascular resistance and portal
pressure. This review focusses on changes occurring in LSECs in NAFLD and on their consequences on
NAFLD progression and complications. Capillarization, namely the loss of LSEC fenestrae, and LSEC dys-
function, namely the loss of the ability of LSECs to generate vasodilator agents in response to increased
shear stress both occur early in NAFLD. These LSEC changes favour steatosis development and set the
stage for NAFLD progression. At the stage of non-alcoholic steatohepatitis, altered LSECs release inflam-
matory mediators and contribute to the recruitment of inflammatory cells, thus promoting liver injury
and inflammation. Altered LSECs also fail to maintain hepatic stellate cell quiescence and release fibro-
genic mediators, including Hedgehog signalling molecules, promoting liver fibrosis. Liver angiogenesis is
increased in NAFLD and contributes to liver inflammation and fibrosis, but also to hepatocellular carci-
noma development. Thus, improving LSEC health appears to be a promising approach to prevent NAFLD
progression and complications.
Ó2019 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.
Introduction
Non-alcoholic fatty liver disease (NAFLD) encom-
passes a spectrum of conditions including simple
steatosis and non-alcoholic steatohepatitis
(NASH), defined as the association of steatosis,
hepatocellular damage, inflammation and varying
degrees of fibrosis.
1
NAFLD is associated with obe-
sity, insulin resistance and/or type 2 diabetes and
other metabolic abnormalities, collectively termed
the metabolic syndrome.
2,3
NAFLD is an expanding
health problem with an estimated global preva-
lence of 25%.
2,4
A recent modelling approach esti-
mated that NAFLD cases in the United States will
expand from 83 million in 2015, corresponding
to about 25% of the population, to 100 million in
2030, corresponding to more than 33% of the pop-
ulation.
4
While simple steatosis is generally
benign, NASH can progress to both cirrhosis and
end-stage liver disease. NASH is currently a lead-
ing cause of liver disease among adults awaiting
liver transplantation in Europe and in the United
States and is projected to become the most com-
mon indication for liver transplantation in the
next decade.
2,4
Importantly, patients with meta-
bolic syndrome and NASH also develop hepatocel-
lular carcinoma (HCC) in the absence of
cirrhosis.
5,6
Despite its prevalence and severity,
there is no approved therapy for NASH and avail-
able treatments only aim to control associated
conditions.
1
Understanding the mechanisms of
NAFLD, and in particular how simple steatosis pro-
gresses to NASH and then to cirrhosis and/or liver
cancer, is of the utmost importance.
The current view of the pathogenesis of NASH
centres on the response of hepatocytes to insulin
resistance and lipotoxicity. The immune system
and hepatic stellate cell activation are regarded
as secondary events.
1
The vascular endothelium,
representing the interface between blood and
other tissues of the body, is not only a physical
barrier but is implicated in different physiological
roles, such as haemostasis, metabolite transporta-
tion, inflammation, thrombosis, angiogenesis and
vascular tone. The liver endothelium is mainly
formed of liver sinusoidal endothelial cells (LSECs)
which are highly specialized endothelial cells at
the interface between blood derived from the vis-
ceral adipose tissue and the gut, on the one side,
and hepatic stellate cells and hepatocytes, on the
other side. LSECs have a unique phenotype in the
human body as they lack a basement membrane
and have a multitude of fenestrae organized into
sieves, that regulate the transport of macro-
molecules, including lipids and lipoproteins,
across the sinusoid.
7,8
This review will specifically
focus on the role of LSECs in the pathophysiology
of NAFLD and its complications.
Review JOURNAL
OF HEPATOLOGY
Keywords: Endothelium;
Steatosis; Capillarization;
Endothelial dysfunction;
Angiogenesis.
Received 5 January 2019;
received in revised form 10
2019; accepted 13 February
2019
1
Inserm, UMR-970, Paris Cardio-
vascular Research Center, PARCC,
Paris, France;
2
University Paris Descartes, Paris,
France;
3
INSERM, UMR1149, Centre de
Recherche sur l’Inflammation,
Paris, France;
4
University Paris Diderot, Paris,
France;
5
Service d’Hépatologie, Centre de
Référence des Maladies Vascu-
laires du Foie, DHU Unity, Pôle des
Maladies de l’Appareil Digestif,
Hôpital Beaujon, AP-HP, Clichy,
France
Corresponding author.
Address: Service d’hépatologie,
Hôpital Beaujon, 100 boulevard
du Général Leclerc, 92100 Cli-
chy, France. Tel.: +33 1 40 87 52
83; fax: +33 1 40 87 55 30.
E-mail address: pierre-emma-
nuel.rautou@inserm.fr
(P.-E. Rautou).
Journal of Hepatology 2019 vol. 70 j1278–1291
LSECs and simple steatosis
Role of LSECs in lipid transfer in the normal
liver
Dietary lipids present in the circulation have to be
transported through the vascular endothelium to
be metabolized by tissues. In physiological condi-
tions, LSECs are major regulators of the bidirec-
tional lipid exchange between the blood and the
liver parenchyma. First, LSEC fenestrae allow for
efficient transfer of lipoproteins, chylomicron
remnants (small lipoproteins derived from chy-
lomicrons generated by enterocytes from dietary
lipids), and other macromolecules, from the sinu-
soidal blood to the space of Disse, where they
are taken up by hepatocytes.
9–11
LSEC fenestrae
form a selective barrier for lipids. Indeed, older
studies using radiolabelled lipoproteins showed
that larger lipoproteins do not cross LSEC fenestrae
and remain in the lumen of the sinusoid.
12
More-
over, when LSECs lose their fenestrae following
vascular endothelial growth factor (VEGF) path-
way disruption,
11,13,14
uptake of fluorescently
labelled lipids is impaired.
11,15
Second, LSECs also
regulate lipid transfer through their high capacity
for endocytosis, as shown by their ability to
rapidly take-up oxidized or acetylated-low density
lipoproteins.
8,16,17
LSEC capillarization occurs early in NAFLD and
promotes steatosis
LSECs undergo morphological and functional
changes during liver steatosis.
18–21
One of the
most remarkable phenotypic changes is the loss
of fenestrae, also called defenestration or sinu-
soidal capillarization, associated with the forma-
tion of a basement membrane on the abluminal
surface of LSECs. Several independent groups
reported that sinusoidal capillarization appears
very early in NAFLD.
18–20
Miyao and colleagues
observed that defenestration begins after 1 week
of choline-deficient, L-amino acid-defined diet in
mice.
18
Similar observations were made in rats
fed a high-fat diet for 3 weeks.
20
Triggers for sinusoidal capillarization are not
fully identified, but we can speculate that exces-
sive dietary macronutrients, including lipids, car-
bohydrates, and gut microbiota-derived products
play a role.
19
Cogger and coworkers demon-
strated in mice challenged with several diets
varying in content of macronutrients and energy
that LSEC porosity and fenestrae frequency are
inversely correlated with dietary fat intake, while
fenestrae diameter is inversely correlated with
protein or carbohydrate intake.
19
In this study,
authors also found a negative correlation
between LSEC fenestrae (frequency, porosity and
diameter) and circulating free fatty acid (FFA)
levels.
19
In vitro studies suggested that defenes-
tration occurs following excessive lipid exposure.
For instance, exposure of human primary LSECs
to oxidized low-density lipoprotein (ox-LDL)
reduces the diameter and the porosity of the
fenestrae.
21
The effect of FFA on fenestrae has also
been tested in primary rat LSECs, but firm conclu-
sions cannot be drawn as the authors did not test
several concentrations of FFA but rather the pres-
ence vs. the absence of FFA, which does not ade-
quately mimic in vivo conditions.
22
Recent
evidence point to gut microbiota in the pathogen-
esis of NAFLD.
3,23
Cogger and colleagues showed
that LSEC fenestrae inversely correlated with the
abundance of Bacteroidetes in the gut and posi-
tively correlated with the abundance of Firmi-
cutes.
19
Moreover, it has been shown that a
single injection of endotoxin in rats induces a
decrease in both diameter and number of fenes-
trae suggesting that gut microbiota-derived prod-
ucts may contribute to LSEC capillarization,
although caution is needed since the concentra-
tion of endotoxin used in that study was high.
24
In turn, capillarization favours liver steatosis
(Fig. 1), as observed in mice deficient in plas-
malemma vesicle-associated protein (PLVAP), an
endothelial-specific integral membrane glycopro-
tein required for the formation of endothelial fen-
estrae.
25
These mice exhibit a pronounced
reduction in the number of LSEC fenestrae, associ-
ated with a decrease in sinusoidal permeability,
25
and spontaneously develop extensive steatosis.
25
These mice also have hyperlipoproteinemia and
increased triglyceridemia due to the retention of
chylomicron remnants in the blood. As mentioned
above, Vegfb
-/-
mice also exhibit a reduction in the
number of LSEC fenestrae and less uptake of
labelled oleic acid due to capillarization, but
steatosis was not evaluated.
15
A first hypothesis explaining this consequence
of capillarization on steatosis could be that
reduced LSEC permeability impairs the passage
of hepatocyte-derived very low-density lipopro-
tein toward the sinusoidal lumen, thus inducing
cholesterol and triglyceride retention in the liver.
However, these lipoproteins may escape the liver
through the lymphatic system.
26
As an alternative
explanation, Herrnberger et al. proposed that chy-
lomicron remnants originating from the blood,
and required for synthesis of very low-density
lipoprotein by hepatocytes, cannot reach hepato-
cytes due to LSEC capillarization; their absence
in hepatocytes might then stimulate de novo hep-
atic lipid synthesis and induce steatosis as a com-
pensatory mechanism.
25,27
However, there is no
available data to ascertain this hypothesis. Simi-
larly, Fraser and collaborators postulated that, fol-
lowing LSEC capillarization, chylomicron
remnants and dietary cholesterol no longer cross
the fenestrae to inhibit HMGCoA reductase, the
rate limiting enzyme for hepatocyte cholesterol
biosynthesis, consequently activating endogenous
cholesterol synthesis in hepatocytes.
28
Taken together, these data suggest that fea-
tures of metabolic syndrome are associated with
LSEC capillarization, which promotes steatosis
(Fig. 1).
Key point
LSEC capillarization and
dysfunction occur very
early in NAFLD progression
and contribute to dietary
induced steatosis.
JOURNAL
OF HEPATOLOGY
Journal of Hepatology 2019 vol. 70 j1278–1291 1279
LSECs dysfunction occurs early in NAFLD and
promotes steatosis
Liver steatosis is associated with an increased por-
tal pressure and increased intrahepatic vascular
resistance.
29–32
In patients, hepatic venous pres-
sure gradient correlates with the degree of steato-
sis.
29
Using Doppler flowmetry, Seifalian and
colleagues observed impaired sinusoidal perfusion
in human fatty liver grafts compared with normal
liver grafts.
33
Similar results were obtained in rab-
bits and rats with diet-induced steatosis, with an
inverse correlation between hepatic parenchymal
microcirculation and the severity of steatosis.
34,35
This increased intrahepatic vascular resistance
has a mechanical and a dynamic component. The
mechanical part is due to the compression of the
sinusoidal lumen by enlarged fat-laden hepato-
cytes.
29,36–38
The dynamic part is due to liver
endothelial dysfunction. Endothelial dysfunction
is a pathological condition, common to all vascular
beds, defined as the inability of blood vessels to
dilate in response to increased blood flow.
Endothelial dysfunction is generally indicated by
the loss of nitric oxide bioavailability due to eNOS
(also called NOS3) inhibition.
39
Several lines of
evidence show that LSEC dysfunction occurs in
fatty livers and is involved in the increased intra-
hepatic vascular resistance associated with steato-
sis.
40–43
eNOS activation and liver nitric oxide
content are reduced in mice and rats fed a high-
fat diet or a diet rich in saturated fatty acids for
4 weeks.
40,41
Isolated-perfused liver experiments
performed in these animals showed augmented
portal perfusion pressure and reduced vasodila-
tory response to acetylcholine, indicating liver
endothelial dysfunction. These changes were
observed in the absence of inflammation and
fibrosis, suggesting that endothelial dysfunction
is an early feature associated with steatosis in
NAFLD.
41,42
Several mechanisms could account for this liver
endothelial dysfunction associated with steatosis
(Fig. 2). First, LSECs dysfunction can be induced
by overabundance of lipids during steatosis. In
vitro experiments showed that stimulation of
human primary LSECs with ox-LDL downregulates
eNOS expression through the ox-LDL receptor,
LOX1.
21
In addition, exposure of primary LSECs
to palmitic acid also attenuates nitric oxide
bioavailability through peroxynitrite production
by NOX1, a nitric oxide consuming enzyme highly
expressed in LSECs of mice fed a high-fat diet.
44
Second, steatosis induces insulin resistance in
LSECs, leading to impairment of insulin-
dependent vasodilation.
41,45,46
This effect is due,
on the one hand, to the downregulation of eNOS
activity,
41
and on the other hand to the upregula-
tion of iNOS (also called NOS2), the inducible form
of NOS which can cause endothelial dysfunction
through increased nitro-oxidative stress.
46–48
Interestingly, V-PYRRO/NO – a diazeniumdiolate
ion metabolized in the liver that spontaneously
Chylomicron remnants
VLDL synthesis
Endogenous
cholesterol and
triglycerides
VLDL LDL
Peripheral
tissues
Fenestrae
Lumen
Space of Disse
Physiological conditions
LSEC
Chylomicron
remnants
VLDL synthesis
Endogenous
cholesterol and
triglycerides
synthesis
VLDL Lymphatic vessels
Metabolic syndrome
ox-LDL/FFA
Carbohydrates/proteins
Gut-microbiota products
Steatosis
Capillarization
Circulating
chylomicron
remnants
Hyperlipidemia
Lumen
Space of Disse Peripheral
circulation
Capillarization
LSEC
Hepatocyte
Exogenous
cholesterol and
triglycerides
VLDL
export
Exogenous
cholesterol and
triglycerides
Fig. 1. LSEC capillarization promotes steatosis. In physiological conditions, chylomicron remnants cross LSEC fenestrae and
provide cholesterol and triglycerides for VLDL synthesis. VLDL are then released by hepatocytes and reach blood flow through
fenestrae.In metabolic syndrome conditions, LSEC capillarization arises early in the course of NAFLD, possibly because of
exposure of LSECs to dietary macronutrients. In turn, LSEC capillarization promotes steatosis, possibly because capillarization
blocks the transfer of chylomicron remnants to hepatocytes, thus stimulating endogenous cholesterol and triglyceride
synthesis, as a compensatory mechanism for the synthesis of VLDL, which reach blood flow through the lymphatic system. FFA,
free fatty acids; LDL, low-density lipoprotein; LSECs, liver sinusoidal endothelial cells; NAFLD, non-alcoholic fatty liver disease;
ox-LDL, oxidized low-density lipoprotein; VLDL; very low-density lipoprotein.
Key point
Intrahepatic vascular
resistance is increased
even when steatosis is the
only histological feature of
NAFLD. This is due to the
combination of a compres-
sion of sinusoids by fat-
laden enlarged hepatocytes
and of a dysfunction of
LSECs due to reduced nitric
oxide bioavailability.
Review
1280 Journal of Hepatology 2019 vol. 70 j1278–1291
decomposes to nitric oxide with a very short half-
life at physiological pH and that triggers cyclic
guanosine 30,50-monophosphate (cGMP) synthesis
– improves hepatic microcirculation in mice with
steatosis induced by a high-fat diet.
49
Third, the
gut microbiota also seems to contribute to liver
endothelial dysfunction. Indeed, Garcia-Lezana
and colleagues demonstrated that restoration of
a healthy microbiota via faecal transplantation
normalizes portal hypertension by improving
intrahepatic vascular resistance and endothelial
dysfunction in rats.
43
In turn, LSEC dysfunction favours steatosis
(Fig. 2). Indeed, deficiency in nitric oxide in
eNos
/
mice results in massive fat droplet deposi-
tion and increases liver triglyceride content.
40,50
Nitric oxide contributes to the regulation of hep-
atic lipid content by limiting citrate synthesis in
mitochondria, which is involved in fatty acid pro-
duction.
50
Nitric oxide also attenuates synthesis of
fatty acids in isolated cultured rat hepatocytes by
nitrosylating acetyl-CoA
51
and activating AMP-
activated protein kinase,
52–54
an inhibitor of
glycerol-3-phosphate acyltransferase and thus of
triacylglycerol synthesis.
55,56
In addition, nitric
oxide also allows efficient fatty acid beta-
oxidation through s-nitrosylation of very long-
chain acyl-CoA dehydrogenase in hepatocytes.
57
Interestingly, therapies augmenting nitric oxide
availability in the liver ameliorate steatosis. The
V-PYRRO/NO or the improvement of nitric oxide/
cGMP signalling with the phosphodiesterase-5
inhibitor sildenafil protect against liver steatosis
in mice fed a high-fat diet.
40,58,59
Moreover, treat-
ment with simvastatin, a drug able to increase
expression and activity of eNOS expression in the
liver, decreases steatosis induced by a high-fat diet
in rats.
60
To summarize, steatosis is associated with LSEC
dysfunction which in turn worsens steatosis
(Fig. 2).
Angiogenesis and steatosis
Angiogenesis, defined as the formation of new
vessels from pre-existing vessels, is a key event
in the progression of NAFLD.
61–65
VEGF is the mas-
ter pro-angiogenic regulator of this process sup-
ported by activation of hypoxia inducible factors
(HIFs) in hypoxic areas.
64
Serum VEGF levels are higher in patients with
biopsy-proven steatosis than in healthy individu-
als.
62,63,65
In animal models, liver expression of
VEGF and CD105, an endothelial cell marker,
increase after 3 days of methionine- and choline-
deficient diet in obese and diabetic db/db trans-
genic mice and after 1 week of this diet in
C57BL6/J mice, before NASH appears.
63
However,
3 studies reported that new vessels develop in
the livers of patients with NASH, but not in
individuals with simple steatosis or normal
livers.
66–68
This suggests that molecular events
associated with upregulation of angiogenic factors
start early in the course of NAFLD, while angiogen-
esis appears later, as detailed below.
LSECs in NASH
LSECs contribute to oxidative stress in NASH
In response to lipotoxicity, hepatocytes generate
reactive oxygen species (ROS) and initiate a robust
inflammatory response that accentuates liver
injury.
1,3
In addition to hepatocytes,
1,3
the lipo-
toxic response also occurs in LSECs contributing
to ROS generation.
44,69
Indeed, ROS have been
detected not only in hepatocytes but also in sinu-
soidal cells in patients with NASH.
70
Circulating
lipids seem to account for the oxidative stress in
LSECs. Indeed, exposure of murine LSECs to palmi-
tic acid upregulates NOX1 expression, an enzyme
implicated in ROS production.
44
In addition, stim-
ulation of human primary LSECs with ox-LDL
increases ROS generation after binding to
LOX1.
21
This oxidative stress in LSECs contributes
to NASH. Indeed, mice with a global deficiency in
NOX1, which is highly expressed in LSECs in
NAFLD, had attenuated liver lesions when fed a
high-fat diet, as shown by lower serum ALT level
and lower hepatic cleaved caspase-3 expression.
44
Therefore, in NASH, ROS production takes place
not only in hepatocytes, but also to some extent
in LSECs, and seems to contribute to hepatocyte
injury.
Anti-inflammatory role of LSECs at initial stages
of NASH
Progression of simple steatosis to steatohepatitis is
accompanied by adhesion of leukocytes to the
sinusoidal endothelium followed by infiltration of
leukocytes within liver parenchyma to form
inflammatory foci.
71
Moderate and resolved
inflammatory responses are beneficial to the liver
as they promote the re-establishment of home-
ostasis, contribute to tissue repair and exert hep-
atoprotective effects.
72
However, chronic
inflammation, as seen in NASH, leads to death of
hepatocytes and causes damage to the liver
parenchyma.
72
In physiological conditions, LSECs
constitute a barrier regulating the entry of circulat-
ing leukocytes within liver parenchyma and play-
ing an anti-inflammatory role.
73–76
At early
stages of NAFLD progression, some evidence indi-
cates that LSECs also exhibit anti-inflammatory
functions.
40,77
Indeed, Tateya and colleagues ele-
gantly demonstrated that nitric oxide derived from
LSECs inhibits Kupffer cell activation in mice fed a
high-fat diet for a short period of time (8 weeks).
40
In vitro, both human and murine LSECs
exposed to FFA for a short period (16 hours) exhibit
a downregulation of pro-inflammatory chemoki-
nes involved in monocyte and macrophage
recruitment, through a MAPK dependent
pathway.
77
Key point
Molecular events associ-
ated with angiogenesis are
initiated during simple
steatosis, but angiogenesis
itself is only detected in
NASH.
JOURNAL
OF HEPATOLOGY
Journal of Hepatology 2019 vol. 70 j1278–1291 1281
LSECs promotes liver inflammation at more
advanced stages of NASH
As mentioned, LSEC alterations arise early in
NAFLD progression, prior to liver inflamma-
tion.
18,41
Indeed, LSEC capillarization precedes
Kupffer cell activation
18,41
and liver nitric oxide
content falls before liver NF-kB activation and
TNF
a
, IL-6 and ICAM-1 upregulation.
30,40,41
LSEC
capillarization and dysfunction are permissive
for establishment of liver inflammation. Indeed,
mice deficient in eNOS exhibit an accelerated
hepatic inflammatory response, while improving
nitric oxide/cGMP signalling with the
phosphodiesterase-5 inhibitor sildenafil or with
simvastatin prevents liver inflammation in
rodents fed a high-fat diet.
40,60
During NALFD progression, LSECs then acquire
a pro-inflammatory phenotype and functions
(Fig. 3). LSECs pro-inflammatory phenotype during
NASH is characterized by progressive overexpres-
sion of adhesion molecules including ICAM-1,
VCAM-1 and VAP-1 (AOC3) at the surface of LSECs,
as observed in mouse models of NASH.
68,78–82
LSECs also produce a number of pro-
inflammatory mediators in NASH, including TNF
a
,
IL-6, IL-1 and MCP1 (CCL2).
68,82–84
This pro-inflammatory phenotype of LSECs in
NASH is associated with pro-inflammatory func-
tions (Fig. 3). First, dysfunctional LSECs fail to
maintain Kupffer cell quiescence.
40
Second, the
release of inflammatory mediators by LSECs con-
tributes to the inflammatory response by activat-
ing neighbouring Kupffer cells, and by favouring
recruitment, adhesion and transmigration of blood
leukocytes.
82,85,86
The mechanisms of interaction
between leukocytes and LSECs in NASH have been
reviewed elsewhere in detail and are summarized
in Fig. 3.
80,87–89
LSECs’ expression of ICAM-1,
VCAM-1 and VAP-1 is crucial for these interactions
since in vivo and in vitro studies showed reduced
leukocyte adhesion to hepatic sinusoids when
these receptors are blocked or not functional.
80,90
Moreover, inhibition of the VCAM-1 ligand, VLA-
4 (or ITGA4), on monocytes using an anti-VLA-4
antibody inhibits adhesion and transendothelial
migration of monocytes across LSECs – from
wild-type mice fed a high-fat diet and from ob/ob
obese mice – and improves liver inflammation.
82
Steatosis
NO
Acetyl CoA
FenestraeLumen
Space of Disse
Physiological conditions
NO
eNOS
NO
Citrate
Fatty acids
Triglycerides
GPAT
Citrate
AMPK
Acetyl CoA
Metabolic syndrome
NO
Fatty acids
Triglycerides
Citrate
eNOS LSEC
dysfunction
↑iNOS
↑NOX1
ox-LDL, FFA
Insulin resistance IHVR
Peroxynit rites
Lumen
Space of Disse
Intrahepatic
vascular tone
LSEC LSEC
Hepatocyte
Fatty acids
β-oxidation
NO
ACC
AMPK
ACC
LSEC
dysfunction
Hepatocyte
enlargement
Nitrosylation
Citrate
Fatty acids
β-oxi dation
GPAT
X
X
NO
Nitrosylation
Fig. 2. LSEC dysfunction promotes steatosis. In physiological conditions, LSECs release NO which regulates intrahepatic
vascular tone on the one hand, and hepatic lipid metabolism on the other hand. NO limits hepatic lipid content by inhibiting
hepatic de novo lipogenesis, through a limitation of citrate synthesis in mitochondria, an inhibition of ACC and of GPAT, and by
promoting fatty acids beta-oxidation. In metabolic syndrome conditions, overabundance of lipids and insulin resistance lead to
downregulation of eNOS activity and to upregulation of iNOS and of NOX1 (a nitric oxide consuming enzyme), causing nitro-
oxidative stress through peroxynitrite production and eventually endothelial dysfunction. Reduced NO availability promotes
steatosis. Liver steatosis is associated with an increased intrahepatic vascular resistance which has a mechanical component,
due to the compression of the sinusoidal lumen by enlarged fat-laden hepatocytes, and a dynamic component, due to a liver
endothelial dysfunction. ACC, acetyl CoA carboxylase; AMPK, AMP-activated protein kinase; eNOS, endothelial nitric oxide
synthase; GPAT, glycerol-3-phosphate acyltransferase; LSECs, liver sinusoidal endothelial cells; IHRV; intrahepatic vascular
resistance; iNOS, inducible nitric oxide synthase; NO, nitric oxide; NOX1, NADPH oxidase 1.
Key point
Lipotoxicity and inflam-
mation induce endothelial
inflammation. Activated
LSECs release cytokines
and chemokines and over-
express adhesion mole-
cules, thus sustaining liver
inflammation.
Review
1282 Journal of Hepatology 2019 vol. 70 j1278–1291
Although the stimuli responsible for LSECs’
inflammatory phenotype and functions in NASH
are not firmly identified, several mediators are
potential candidates. This includes products
derived from the visceral adipose tissue, such as
ox-LDL, FFA and adipokines. Indeed, in vitro
studies showed that stimulation of LSECs with
ox-LDL and FFA (palmitate) activate NF-kB and
TLR-4, respectively.
21,44,91
Moreover, circulating
concentrations of several adipokines, including
TNF
a
and IL-6, are increased in the portal vein in
the context of metabolic syndrome, and may
contribute to LSECs inflammatory phenotype.
92
The gut microbiota also has an emerging role in
NASH as a source of inflammatory stimuli.
1,3
Increased intestinal permeability and elevated
plasma concentrations of lipopolysaccharide
(LPS)
93,94
observed in NASH may also contribute
to LSECs’ pro-inflammatory function.
83
Besides
mediators derived from the portal vein, hepato-
cytes and liver inflammatory cells also release
inflammatory mediators in NASH that can activate
LSECs.
1,95
To summarize, while LSECs play an anti-
inflammatory role in the initial stages of NAFLD,
a switch towards pro-inflammatory functions
occurs during the course of NAFLD development,
paving the way for NASH progression.
Angiogenesis in liver inflammation in NASH
Pathologic angiogenesis increases with
NASH
61,68,96,97
(Fig. 4). Indeed, several studies
reported the formation of new vessels in the liver
of patients with NASH. Moreover, serum VEGF and
sVEGFR1 levels are higher in patients with steato-
sis and biopsy-proven NASH than in healthy indi-
viduals.
62,66–68,98
Similarly, in animal models of
NASH, liver vasculature is disrupted and hepatic
expression of VEGF and CD105 is increased.
63
Several mechanisms trigger angiogenesis dur-
ing NASH. First, chronic inflammation promotes
angiogenesis. Indeed, chronic inflammation sus-
tains tissue hypoxia and induces transcription of
NF-κB
Lipotoxicity
Inflammation
Gut microbiota-
derived products
Activated Kupffer cell
Quiescent
Kupffer cell
Transendothelial
migration
Lymphocytes,
Monocytes, Neutrophils,
NK/NKT cells
Leukocytes
Chronic
inflammation
Space of Disse
Sinusoidal lumen
MCP1, TNFα,
IL-6, IL-1
NO
Endothelial dysfunction
VCAM-1, ICAM- 1, VAP-1
LSEC
Fig. 3. LSECs acquire pro-inflammatory functions in NASH. Lipotoxicity, inflammation and
gut microbiota-derived products induce LSECs’ inflammatory phenotype and function
mediated by NF-kB activation, which orchestrates the release of pro-inflammatory mediators
and the overexpression of adhesion molecules. Inflammatory mediators and LSEC dysfunction
promote Kupffer cell activation and leukocyte chemoattraction. Adhesion molecule overex-
pression allows adhesion and transendothelial migration of the recruited leukocytes in the
hepatic parenchyma. ICAM-1, intercellular adhesion molecule-1; IL-1, interleukin 1; IL-6,
interleukin 6; LSECs, liver sinusoidal endothelial cells; MCP1, monocyte chemoattractant
protein-1; NASH, non-alcoholic steatohepatitis; NF-kB, nuclear factor kappa B; NK, natural
killer; NO, nitric oxide; TNF
a
, tumor necrosis factor alpha; VAP-1, vascular adhesion protein-
1; VCAM-1, vascular cell adhesion molecule-1.
Steatosis Inflammation Fibrosis
Angiogenesis
Anti-angiogenic
therapies
Angiopoeitin-2
VEGF
Leptin
Hepatocytes injury
Inflammation
Fibrosis
Hypoxia
Anti-VEGFR2
Blocking
Angiopoietin2-Tie
interaction
(L1-10 peptibody)
Angiotensin-II
receptor
blockers
Hepatocytes- derived
microvesicles
Fig. 4. Angiogenesis in NASH. Hypoxia, liver injury, lipids, oxidative stress, inflammation and fibrosis induce the release of
pro-angiogenic factors, such as hepatocyte-derived microvesicles, VEGF and Angiopoietin-2, from parenchymal and non-
parenchymal cells including LSECs, promoting pathologic angiogenesis. Adipokines, such as leptin, also exhibit pro-angiogenic
activity contributing to pathologic angiogenesis in NASH. In turn, angiogenesis promotes liver inflammation and fibrosis as
shown by anti-angiogenic therapies which prevent liver inflammation and fibrosis in experimental models of NASH. LSECs,
liver sinusoidal endothelial cells; NASH, non-alcoholic steatohepatitis; VEGF, vascular endothelial growth factor; VEGFR2,
vascular endothelial growth factor receptor 2.
Key point
Inflammation promotes
angiogenesis that in turn
worsens liver inflamma-
tion as demonstrated by
the anti-inflammatory
effect of anti-angiogenic
therapies.
JOURNAL
OF HEPATOLOGY
Journal of Hepatology 2019 vol. 70 j1278–1291 1283
angiogenic genes modulated by HIF-1
a
.
61,99
Pro-
inflammatory mediators also elicit a direct angio-
genic response through the induction of HIF-1
a
transcriptional activity and VEGF production.
61
Moreover, cytokines and ROS released during
NASH can activate the MAPK/ERK pathway, a sig-
nalling pathway involved in cell migration and
angiogenesis.
61
Second, hepatocyte-derived
microvesicles link lipotoxicity with angiogenesis.
Indeed, hepatocytes exposed in vitro to excessive
amounts of saturated FFA, that mimics steatosis,
release microvesicles with a pro-angiogenic activ-
ity.
100
Likewise, mice fed a methionine- and
choline-deficient diet have high circulating levels
of hepatocyte-derived microvesicles able to
induce angiogenesis. Third, angiopoietin-2 is
another mechanism of liver angiogenesis in NASH.
Angiopoietins are key regulators of angiogenesis.
Although angiopoietins-1 and 2 contribute to vas-
cular stability and quiescence in physiological
conditions, angiopoietin-2 promotes pathological
angiogenesis in inflammatory conditions.
101
Lefere
and coworkers recently showed that serum
angiopoietin-2 levels are increased in patients
with NASH and correlate with liver steatosis,
inflammation and hepatocyte ballooning, but not
with liver fibrosis.
68
Similar findings were
observed with 2 murine models of NASH, namely
mice fed a methionine- and choline-deficient diet
and mice with neonatal injection of streptozotocin
followed by 16 weeks of western diet.
68
The main
source of hepatic angiopoietin-2 was LSECs.
68
Inhibiting angiopoietin-2 levels using the
angiopoietin-2/Tie2 receptor inhibiting peptibody
L1-10 reduced hepatic angiogenesis and normal-
ized vascular microarchitecture.
68
In turn, angiogenesis promotes inflammation
since various strategies of inhibition of angiogenesis
all improve liver inflammation (Fig. 4). Coulon and
colleagues showed in a mouse model of NASH that
treatment with anti-VEGFR2 antibody improves
liver vasculature and decreases liver inflammatory
gene expression, both using preventive and thera-
peutic approaches.
63
Lefere and colleagues showed
that blocking angiopoietin-2/Tie2 interaction with
the L1-10 peptibody also alleviates liver injury and
inflammation in mice fed a methionine- and
choline-deficient diet.
68
Importantly, this effect of
L1-10 therapy is at least partly mediatedby an effect
on LSECs since L1-10 treatment downregulates
VCAM-1, ICAM-1 and MCP1 expression in liver
endothelial cells isolated from mice fed a
methionine- and choline-deficient diet.
68
This
anti-inflammatory effect of anti-angiogenic treat-
ment is not specific for NASH, as it is observed in
most models of chronic liver disease, namely carbon
tetrachloride, and bile duct ligation.
102–106
To summarize, inflammation stimulates angio-
genesis that in turn worsens inflammation, as
shown by the anti-inflammatory effect of anti-
angiogenic therapies (Fig. 4).
LSECs in NASH-related liver fibrosis
Liver fibrosis is defined as the excessive deposi-
tion of extracellular matrix in liver parenchyma.
The main mechanism leading to liver fibrosis is a
long-standing wound healing process caused by
hepatocellular injury and inflammation and
mediated by hepatic stellate cell activation.
1,107
Hepatic stellate cells are nonparenchymal cells
close to LSECs, in the space of Disse, which store
retinoids in physiological conditions and shift
their phenotype to an activated myofibroblastic
state during liver injury and inflammation,
wherein they secrete large amounts of extracel-
lular matrix compounds.
107
As detailed above,
LSECs are major effectors of liver inflammation
in NASH, and consequently also promote hepatic
fibrosis. For example, LSECs overexpress VAP-1
during inflammation which, in addition to its
pro-inflammatory functions in NASH, is directly
involved in hepatic stellate cell activation.
80
Inhi-
bition or deficiency in VAP-1 in mice fed a
methionine- and choline-deficient diet or a
high-fat diet attenuates liver fibrosis.
80
LSECs
also contribute to liver fibrosis through capillar-
ization and endothelial dysfunction, as detailed
in the following sections.
LSEC capillarization promotes liver fibrosis
Capillarization is observed in patients and animal
models of NASH, preceding fibrosis,
18,41,108–112
but also promoting its development (Box 1).
Indeed, PLVAP deficient mice, displaying a pro-
nounced reduction in the number of LSEC fenes-
trae, spontaneously develop perisinusoidal liver
fibrosis.
25
Experiments performed using cultured LSECs
and hepatic stellate cells highlighted the impor-
tance of cross-talk between these cells types in
regulating each other’s phenotype. While healthy
LSECs maintain hepatic stellate cell quiescence,
capillarized LSECs lose this ability
112,113
(Fig. 5).
A vicious cycle between LSEC capillarization and
hepatic stellate cell activation then occurs during
the fibrotic process.
In NASH, ballooned hepatocytes produce
Hedgehog molecules.
114
The Hedgehog pathway
regulates capillarization, as inhibition of Hedge-
hog signalling prevents capillarization and par-
tially reverts the phenotype of LSECs from a
dedifferentiated state to their differentiated
LSECs alterations are observed in various dietary or genetic models of NAFLD, without
apparent specificity. Indeed, LSECs capillarization is observed in animals fed a choline-
deficient, L-amino acid-defined diet or a high-fat diet.1820 LSECs dysfunction can be induced
by a high-fat diet, or a rich saturated fatty acids diet, or a high-fat high-glucose and
high-fructose diet.40,41,43 Markers of LSECs inflammatory phenotype are observed in animals
fed a high-fat diet or a methionine and choline deficient diet.68,82 Markers of angiogenesis
appear after methionine and choline deficient diet in db/db obese mice and in C57BL/6 mice
and in rats fed a choline-deficient, L-amino acid-defined diet.63,129
Box 1. LSEC changes are not specific for certain animal models of NASH. LSEC, liver
sinusoidal endothelial cells; NASH, non-alcoholic steatohepatitis.
Key point
LSEC capillarization and
dysfunction precede liver
fibrosis and are permissive
for it, through the loss of
the ability of LSECs to
maintain quiescence of
hepatic stellate cells.
Review
1284 Journal of Hepatology 2019 vol. 70 j1278–1291
state.
115
LSECs are thus Hedgehog-sensitive cells,
but they are also Hedgehog producing cells. Simi-
larly, quiescent hepatic stellate cells are
Hedgehog-sensitive cells, while activated hepatic
stellate cells become Hedgehog-producing cells,
which are also able to release macrovesicles
loaded with Hedgehog signalling molecules that
interact with LSECs.
116,117
It is thus tempting to
speculate that during NASH, Hedgehog ligands
are released by hepatocytes and LSECs, thus
activating LSECs themselves, as well as quiescent
hepatic stellate cells, by autocrine and paracrine
effects. Activated hepatic stellate cells can then
secrete Hedgehog molecules, promoting LSEC
capillarization which in turn favours hepatic
stellate cell activation, promoting the fibrogenic
process.
LSECs dysfunction promotes liver fibrosis
Endothelial dysfunction appears very early in the
course of NAFLD and precedes fibrosis in animal
models of NASH.
30,40,41
Several lines of evidence
suggest that liver endothelial dysfunction
contributes to liver fibrosis. First, in rats fed a
high-fat diet, simvastatin increases liver eNOS
expression and ameliorates liver fibrosis.
60
Second, eNOS inhibition using L-NAME blocks
the ability of healthy LSECs to keep hepatic stellate
cells quiescent.
118,119
Third, an activator of soluble
guanylate cyclase, a receptor for nitric oxide, can
recapitulate the effect of healthy LSECs and
reverse hepatic stellate cell activation.
13
However,
a limitation of these studies is that most
approaches might not only act on endothelial
function but also on LSEC capillarization.
112,118
Indeed, although L-NAME blocks LSEC-induced
hepatic stellate cell quiescence, addition of a nitric
oxide donor does not directly reverse hepatic stel-
late cell activation in vitro,
13
suggesting that addi-
tional LSEC-derived factors could be responsible
for the reversion of activated hepatic stellate cells
to quiescence.
To summarize, these data demonstrate that
capillarization and LSEC dysfunction not only pre-
cede liver fibrosis, but also promote it (Fig. 5). In
their differentiated state, LSECs are able to main-
tain hepatic stellate cell quiescence, making differ-
entiated LSECs gatekeepers of fibrosis in NASH, as
in other chronic liver diseases.
Liver endothelial-to-mesenchymal transition: a
process promoting liver fibrosis?
Another important process that links endothelial
cells to organ fibrosis is endothelial-to-
mesenchymal transition, i.e. the mechanism by
which endothelial cells convert into myofibroblasts
and contribute to extracellular matrix deposi-
tion.
120,121
Endothelial-to-mesenchymal transition
occurs in various fibrotic cardiovascular and pul-
monary diseases.
120–122
In vitro studies showed
that healthy LSECs produce a modest amount of col-
lagen and fibronectin.
123
Capillarized LSECs secrete
fibrogenic factors, such as TGF-b1, and extracellular
matrix proteins, such as fibronectin and laminin,
that may be considered as an endothelial-to-
mesenchymal transition, as well as stimulating
activation of neighbouring hepatic stellate
cells.
8,124,125
In the liver disease field, only 1 study
has described endothelial-to-mesenchymal transi-
tion in vivo, in patients with alcohol- or hepatitis C
virus-related cirrhosis and in mice treated with car-
bon tetrachloride.
126
This process might also occur
during liver fibrosis in NASH, but further studies are
required.
Altered
LSEC
Fibronectine
Laminine
TGF-β
Hh molecules
Endothelial dysfunction
Capillarization
↑ Hh signaling
Hh molecules
Activated
HSC
Quiescent
HSC
ECM secretion
Space of Disse
Lumen
Hh molecules
NO
Fibrosis
Fig. 5. LSECs in NASH-related fibrosis. Healthy LSECs are gatekeepers of liver fibrosis by
maintaining HSCs quiescence through NO, while altered LSECs (following capillarization and
LSEC dysfunction) lose this ability. In addition, altered LSECs release profibrogenic molecules
such as TGF-b, Hedgehog molecules, laminin and fibronectin, which activate HSCs. Activated
HSCs produces Hedgehog molecules reinforcing their own activation and LSEC capillarization.
Activated HSCs then produce large amounts of extracellular matrix thus inducing liver fibrosis.
ECM, extracellular matrix; Hh, Hedgehog; HSCs, hepatic stellate cells; LSECs, liver sinusoidal
endothelial cell; NO, nitric oxide; TGF-b, transforming growth factor-b.
Stabilin
1/2
LYVE-1 CD32b
(SE-1)
ICAM-1Integrins
αvβ3
αvβ5
Higher
expression of
Integrins
αvβ3, αvβ5
Lower
expression of
ICAM-1
Lower adherence
capacity with
leukocytes
Higher adherence capacity with
cancerous cells
T-cells tolerance towards
cancer-associated
antigens
Immunosuppressive environment
Liver endothelial cell
Tumour progression
Fig. 6. Role of liver endothelial cells in HCC development in chronic liver diseases (not
specific for NAFLD). During HCC progression, endothelial cells sequentially lose their specific
markers including stabilin-1, stabilin-2, LYVE-1 and CD32b (SE-1). Conversely, endothelial
expression of integrins increases, facilitating adhesion of liver cancer cells. In parallel,
endothelial expression of ICAM-1 decreases, leading to a lower ability of leukocyte to adhere
and infiltrate HCC. Endothelial cells within HCC can also alter tumour-associated immune
responses via their ability to confer T cell tolerance towards cancer-associated antigens and
to create an immunosuppressive environment. ICAM-1, intercellular adhesion molecule-1;
LYVE-1, lymphatic vessel endothelial hyaluronic acid receptor 1; HCC, hepatocellular
carcinoma; NAFLD, non-alcoholic fatty liver disease.
JOURNAL
OF HEPATOLOGY
Journal of Hepatology 2019 vol. 70 j1278–1291 1285
Angiogenesis in NASH-related liver fibrosis
Liver angiogenesis correlates with liver fibrosis in
patients with NASH.
66,67
The mechanisms leading
to liver angiogenesis in NASH-related fibrosis
include those mentioned above, namely tissue
hypoxia,
61
hepatocyte-derived microvesi-
cles
100,127
and angiopoietin-2,
68
but also leptin.
Leptin concentrations are increased in the serum
of patients with NAFLD.
128
This adipocytokine
has both pro-angiogenic effects,
129
and direct
pro-fibrogenic effects, through the upregulation
of TGF-bin LSECs and Kupffer cells.
130
In turn, angiogenesis promotes liver fibrosis
since several approaches inhibiting liver angiogen-
esis prevent NASH-related fibrosis (Fig. 4). First, in
the study by Kitade and colleagues, neither angio-
genesis nor fibrosis were observed in the absence
of leptin signalling in a rat model of NASH.
129
Sec-
ond, blocking the release of pro-angiogenic
microvesicles from fat laden-hepatocytes or
inhibiting their binding to target cells protects
mice from steatohepatitis-induced pathologic
angiogenesis and results in a reduction in liver
fibrosis.
100
Third, Zhou and coworkers recently
showed that a specific deletion of the physiologi-
cal inhibitor of angiogenesis, prolyl-hydroxylase-
2, in endothelial cells results in an overexpression
of angiopoietin-2 and TGF-b1 in the liver and pro-
motes dietary-induced liver fibrosis in mice.
131,132
Whether this pro-fibrotic effect of endothelial
prolyl-hydroxylase-2 deficiency is directly
induced by promoting angiogenesis remains to
be demonstrated. Fourth, 2 studies reported that
inhibiting angiotensin-II receptor using telmisar-
tan or candesartan inhibits liver angiogenesis
and fibrosis in rats fed a choline-deficient, L-
amino acid-defined diet.
96,97
Finally, Lefere and
colleagues demonstrated that blocking angiogene-
sis by inhibiting the interaction between
angiopoietin-2/Tie2 using the L1-10 peptibody
improves liver fibrosis in preventive and therapeu-
tic strategies in mice fed a methionine- and
choline-deficient diet. Therapeutic application of
L1-10 peptibody also prevents liver fibrosis in dia-
betic mice with NASH (streptozotocin and western
diet model).
68
It should be noted that an anti-
fibrotic effect of anti-angiogenic treatments has
also been observed in models of chronic liver dis-
ease without NASH.
102–106,133–139
LSECs in NASH-induced HCC
In most cases, HCC develops on a background of
chronic liver disease (70–90% of all patients). The
role of liver endothelial cells in HCC development,
outside the NAFLD setting, has been reviewed
elsewhere and is summarized in Fig. 6.
8,76,140–143
Patients with metabolic syndrome and NAFLD
also develop HCC in the absence of underlying cir-
rhosis, suggesting oncogenic pathways specific for
NAFLD.
5,6
Adipokines and angiogenesis associated
with NAFLD seem to account – at least partly – for
this specific link between NAFLD and HCC.
Circulating concentrations of the adipokine
FABP4 are increased in patients with NAFLD with-
out HCC and correlate with liver inflammation
and fibrosis.
144
Interestingly, Laouirem and col-
leagues recently demonstrated that LSECs exposed
to conditions mimicking NAFLD – namely high con-
centrations of glucose, insulin, or VEGFA – release
FABP4. They also observed that FABP4 released by
LSECs exerts pro-oncogenic effects, since it induces
hepatocyte proliferation. In mice fed a high-fat diet,
specific inhibition of FABP4 reduces HCC growth.
145
We can speculate that FABP4 from LSECs not only
contributes to HCC progression but also to HCC
development in a NAFLD setting (Fig. 7).
In NAFLD, angiogenesis is highly stimulated
and promotes NAFLD-associated HCC, since vari-
ous inhibitors of angiogenesis all prevent HCC
development. First, leptin-mediated angiogenesis
has been demonstrated to be involved in HCC
development as neither angiogenesis nor HCC
develop in the absence of leptin signalling in
Zucker rats fed a choline-deficient, L-amino acid-
defined diet.
129
Second, Yoshiji and colleagues
showed that a conventional anti-angiogenic treat-
ment with sorafenib inhibits the appearance of
preneoplastic lesions in rats fed a choline-
deficient, L-amino acid-defined diet.
146
In this
study, authors also demonstrated that a treatment
combining low doses of sorafenib with the
angiotensin-II receptor inhibitor losartan also suc-
cessfully inhibited preneoplastic lesions.
146
Third,
Tamaki and colleagues demonstrated that inhibi-
tion of angiotensin-II receptor with telmisartan
inhibits HIF-1
a
activity and VEGF expression and
prevents HCC development in the liver of rats
fed a choline-deficient, L-amino acid-defined diet
for 48 weeks.
96
Finally, Lefere and coworkers
recently showed that therapeutic inhibition of
angiopoietin-2 alleviates steatohepatitis and pre-
vents NASH-associated HCC progression in mice.
68
Adipokines
FABP4
↑ Leptin
FABP4
↑ VEGF
↑ Angiopoeitin-2 ↑Angiogenesis
Anti-angiogenic
therapies
Angiotensin-II receptor blockers
(Losartan,Telmisartan)
Blocking Angiopoietin2-Tie
interaction (L1-10 peptibody)
Sorafenib
HCC development
and progression
Metabolic syndrome
NAFLD
Fig. 7. Role of liver endothelial cells in hepatocellular carcinoma development in the
NAFLD setting. Circulating concentrations of angiocrine factors, such as VEGF and
angiopoeitin-2, and adipokines, such as leptin are increased in NAFLD. These mediators
induce angiogenesis which promotes HCC growth. The adipokine FABP4 is released by adipose
tissue and endothelial cells and contributes to HCC development and progression. FABP4, fatty
acid binding protein 4; HCC, hepatocellular carcinoma; NAFLD, non-alcoholic fatty liver
disease.
Key point
Adipokines contribute to
HCC development in
NAFLD.
Key point
Liver angiogenesis corre-
lates with the severity of
liver fibrosis and promotes
its development. Once
established, fibrosis stimu-
lates angiogenesis by
increasing tissue hypoxia.
Blocking pathologic angio-
genesis prevents liver
fibrosis.
Key point
NALFD associated angio-
genesis promotes HCC.
Blocking pathologic angio-
genesis prevents HCC
development and
progression.
Review
1286 Journal of Hepatology 2019 vol. 70 j1278–1291
Gaps in knowledge and future directions
Even if our understanding of the role of LSECs in
NAFLD has progressed over the last years, several
aspects remain elusive. First, triggers responsible
for LSEC alterations in NAFLD are mostly unknown.
It has been suggested that mediators derived from
the visceral adipose tissue and the gut are respon-
sible, but this has not been convincingly estab-
lished. Indeed, available in vitro studies
considered each mediator individually and not in
combination, as in vivo in the portal venous
blood.
21,44,77
Second, mechanisms underlying
endothelial changes in NAFLD, including capillar-
ization, need to be defined which might provide
new therapeutic targets for NAFLD. Third, the role
of LSECs in NASH-related cirrhosis has not been
specifically investigated. Whether LSEC function
and phenotype differ in cirrhosis related to NASH
from cirrhosis related to other causes remains to
be assessed.
147
Fourth, although NAFLD is well rec-
ognized as favouring HCC development, we are still
at a very early stage of understanding how LSEC
changes might favour HCC development.
Conclusion
LSECs are gatekeepers of liver homeostasis in phys-
iological conditions. In NAFLD, sinusoidal endothe-
lial alterations, including capillarization and LSEC
dysfunction, occur early in disease progression, at
the stage of simple steatosis. These initial changes,
triggered by lipotoxicity, adipokines, inflammation
and gut microbiota-derived products are associ-
ated with a loss of the ability of LSECs to prevent
liver inflammation and fibrosis associated with
NASH. Indeed, altered LSECs fail to maintain Kupf-
fer cells and hepatic stellate cells in a quiescent
state. At the stage of NASH, altered LSECs con-
tribute to liver angiogenesis, inflammation, fibrosis
and HCC. Improving LSEC health has great thera-
peutic potential for NAFLD. The current challenge
is the identification of strategies to specifically tar-
get LSECs in order to modulate their activity.
Financial support
This work was supported by the ‘‘Institut National
de la Santé et de la Recherche Médicale” (ATIP
AVENIR), Paris Descartes University, the ‘‘Agence
Nationale pour la Recherche” (ANR-14-CE12-
0011, ANR-14-CE35-0022, ANR-18-CE14-0006-
01) and by the ‘‘Association Française pour l’Etude
du foie” (AFEF 2014). A.H. was supported by the
‘‘Ministère de l’Enseignement Supérieur et de la
Recherche”.
Conflict of interest
The authors declare no conflicts of interest that
pertain to this work.
Please refer to the accompanying ICMJE disclo-
sure forms for further details.
Authors’ contributions
A.H. and P-E.R wrote the manuscript.
Supplementary data
Supplementary data to this article can be found
online at https://doi.org/10.1016/j.jhep.2019.02.
012.
References
Author names in bold designate shared co-first authorship
[1] Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mecha-
nisms of NAFLD development and therapeutic strategies. Nat Med
2018;24:908–922. https://doi.org/10.1038/s41591-018-0104-9.
[2] Younossi ZM. Non-alcoholic fatty liver disease-a global public health
perspective. J Hepatol 2019;70:531–544. https://doi.org/10.1016/j.
jhep.2018.10.033.
[3] Marra F, Svegliati-Baroni G. Lipotoxicity and the gut-liver axis in NASH
pathogenesis. J Hepatol 2018;68:280–295. https://doi.org/10.1016/j.
jhep.2017.11.014.
[4] Estes C, Razavi H, Loomba R, Younossi Z, Sanyal AJ. Modeling the
epidemic of nonalcoholic fatty liver disease demonstrates an exponen-
tial increase in burden of disease. Hepatology 2018;67:123–133.
https://doi.org/10.1002/hep.29466.
[5] Paradis V, Zalinski S, Chelbi E, Guedj N, Degos F, Vilgrain V, et al.
Hepatocellular carcinomas in patients with metabolic syndrome often
develop without significant liver fibrosis: a pathological analysis.
Hepatology 2009;49:851–859. https://doi.org/10.1002/hep.22734.
[6] Kanwal F, Kramer JR, Mapakshi S, Natarajan Y, Chayanupatkul M,
Richardson PA, et al. Risk of hepatocellular cancer in patients with non-
alcoholic fatty liver disease 1828-1837.e2. Gastroenterology 2018;155.
https://doi.org/10.1053/j.gastro.2018.08.024.
[7] Zapotoczny B, Szafranska K, Kus E, Braet F, Wisse E, Chlopicki S, et al.
Tracking Fenestrae dynamics in live murine liver sinusoidal endothelial
cells. Hepatology 2018. https://doi.org/10.1002/hep.30232.
[8] Poisson J,Lemoinne S, Boulanger C, Durand F, Moreau R, Valla D, et al.
Liver sinusoidal endothelial cells: physiology and role in liver diseases.
J Hepatol 2017;66:212–227. https://doi.org/10.1016/j.
jhep.2016.07.009.
[9] Hilmer SN, Cogger VC, Fraser R, McLean AJ, Sullivan D, Le Couteur DG.
Age-related changes in the hepatic sinusoidal endothelium impede
lipoprotein transfer in the rat. Hepatology 2005;42:1349–1354. https://
doi.org/10.1002/hep.20937.
[10] Cogger VC, Hilmer SN, Sullivan D, Muller M, Fraser R, Le Couteur DG.
Hyperlipidemia and surfactants: the liver sieve is a link. Atheroscle-
rosis 2006;189:273–281. https://doi.org/10.1016/j.
atherosclerosis.2005.12.025.
[11] Carpenter B,Lin Y, Stoll S, Raffai RL, McCuskey R, Wang R. VEGF is
crucial for the hepatic vascular development required for lipoprotein
uptake. Development 2005;132:3293–3303. https://doi.org/10.1242/
dev.01902.
[12] Fraser R, Bosanquet AG, Day WA. Filtration of chylomicrons by the liver
may influence cholesterol metabolism and atherosclerosis. Atheroscle-
rosis 1978;29:113–123.
[13] Xie G, Wang X, Wang L, Wang L, Atkinson RD, Kanel GC, et al. Role of
differentiation of liver sinusoidal endothelial cells in progression and
regression of hepatic fibrosis in rats 918-927.e6. Gastroenterology
2012;142. https://doi.org/10.1053/j.gastro.2011.12.017.
[14] May D, Djonov V, Zamir G, Bala M, Safadi R, Sklair-Levy M, et al. A
transgenic model for conditional induction and rescue of portal
hypertension reveals a role of VEGF-mediated regulation of sinusoidal
fenestrations. PLoS One 2011;6. https://doi.org/10.1371/journal.
pone.0021478 e21478.
JOURNAL
OF HEPATOLOGY
Journal of Hepatology 2019 vol. 70 j1278–1291 1287
[15] Hagberg CE, Falkevall A, Wang X, Larsson E, Huusko J, Nilsson I, et al.
Vascular endothelial growth factor B controls endothelial fatty acid
uptake. Nature 2010;464:917–921. https://doi.org/
10.1038/nature08945.
[16] Van Berkel TJ, De Rijke YB, Kruijt JK. Different fate in vivo of oxidatively
modified low density lipoprotein and acetylated low density lipopro-
tein in rats. Recognition by various scavenger receptors on Kupffer and
endothelial liver cells. J Biol Chem 1991;266:2282–2289.
[17] Blomhoff R, Drevon CA, Eskild W, Helgerud P, Norum KR, Berg T.
Clearance of acetyl low density lipoprotein by rat liver endothelial
cells. Implications for hepatic cholesterol metabolism. J Biol Chem
1984;259:8898–8903.
[18] Miyao M, Kotani H, Ishida T, Kawai C, Manabe S, Abiru H, et al. Pivotal
role of liver sinusoidal endothelial cells in NAFLD/NASH progression.
Lab Invest 2015;95:1130–1144. https://doi.org/10.1038/labinvest.
2015.95.
[19] Cogger VC, Mohamad M, Solon-Biet SM, Senior AM, Warren A, O’Reilly
JN, et al. Dietary macronutrients and the aging liver sinusoidal
endothelial cell. Am J Physiol Heart Circ Physiol 2016;310:
H1064–H1070. https://doi.org/10.1152/ajpheart.00949.2015.
[20] Peng Qiuxian,Zhang Q,Xiao W, Shao M, Fan Q, Zhang H, et al.
Protective effects of Sapindus mukorossi Gaertn against fatty
liver disease induced by high fat diet in rats. Biochem Biophys
Res Commun 2014;450:685–691. https://doi.org/10.1016/j.bbrc.
2014.06.035.
[21] Zhang Q,Liu J, Liu J, Huang W, Tian L, Quan J, et al. oxLDL induces
injury and defenestration of human liver sinusoidal endothelial cells
via LOX1. J Mol Endocrinol 2014;53:281–293. https://doi.org/10.1530/
JME-14-0049.
[22] Hang T-C, Lauffenburger DA, Griffith LG, Stolz DB. Lipids promote
survival, proliferation, and maintenance of differentiation of rat liver
sinusoidal endothelial cells in vitro. Am J Physiol Gastrointest Liver
Physiol 2012;302:G375–G388. https://doi.org/10.1152/
ajpgi.00288.2011.
[23] Soderborg TK, Clark SE, Mulligan CE, Janssen RC, Babcock L, Ir D, et al.
The gut microbiota in infants of obese mothers increases inflammation
and susceptibility to NAFLD. Nat Commun 2018;9:4462. https://doi.
org/10.1038/s41467-018-06929-0.
[24] Dobbs BR, Rogers GW, Xing HY, Fraser R. Endotoxin-induced defenes-
tration of the hepatic sinusoidal endothelium: a factor in the patho-
genesis of cirrhosis? Liver 1994;14:230–233.
[25] Herrnberger L, Hennig R, Kremer W, Hellerbrand C, Goepferich A,
Kalbitzer HR, et al. Formation of fenestrae in murine liver sinusoids
depends on plasmalemma vesicle-associated protein and is required
for lipoprotein passage. PLoS One 2014;9. https://doi.org/10.1371/
journal.pone.0115005 e115005.
[26] Tanaka M, Iwakiri Y. The hepatic lymphatic vascular system: structure,
function, markers, and lymphangiogenesis. Cell Mol Gastroenterol
Hepatol 2016;2:733–749. https://doi.org/10.1016/j.
jcmgh.2016.09.002.
[27] Feingold KR, Grunfeld C. Introduction to Lipids and Lipoproteins. In: De
Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman
JM, editors. Endotext. South Dartmouth (MA): MDText.com, Inc.; 2000.
[28] Fraser R, Cogger VC, Dobbs B, Jamieson H, Warren A, Hilmer SN, et al.
The liver sieve and atherosclerosis. Pathology 2012;44:181–186.
https://doi.org/10.1097/PAT.0b013e328351bcc8.
[29] Francque S, Verrijken A, Mertens I, Hubens G, Van Marck E, Pelckmans
P, et al. Noncirrhotic human nonalcoholic fatty liver disease induces
portal hypertension in relation to the histological degree of steatosis.
Eur J Gastroenterol Hepatol 2010;22:1449–1457. https://doi.org/
10.1097/MEG.0b013e32833f14a1.
[30] Francque S,Laleman W, Verbeke L, Van Steenkiste C, Casteleyn C,
Kwanten W, et al. Increased intrahepatic resistance in severe steatosis:
endothelial dysfunction, vasoconstrictor overproduction and altered
microvascular architecture. Lab Invest 2012;92:1428–1439. https://
doi.org/10.1038/labinvest.2012.103.
[31] Mendes FD, Suzuki A, Sanderson SO, Lindor KD, Angulo P. Prevalence
and indicators of portal hypertension in patients with nonalcoholic
fatty liver disease 1028–1033.e2. Clin Gastroenterol Hepatol 2012;10
(1028–1033). https://doi.org/10.1016/j.cgh.2012.05.008.
[32] Baffy G. Origins of portal hypertension in nonalcoholic fatty liver
disease. Dig Dis Sci 2018;63:563–576. https://doi.org/10.1007/s10620-
017-4903-5.
[33] Seifalian AM, Chidambaram V, Rolles K, Davidson BR. In vivo demon-
stration of impaired microcirculation in steatotic human liver grafts.
Liver Transpl Surg 1998;4:71–77.
[34] Seifalian AM, Piasecki C, Agarwal A, Davidson BR. The effect of graded
steatosis on flow in the hepatic parenchymal microcirculation. Trans-
plantation 1999;68:780–784.
[35] Ijaz S, Yang W, Winslet MC, Seifalian AM. The role of nitric oxide in the
modulation of hepatic microcirculation and tissue oxygenation in an
experimental model of hepatic steatosis. Microvasc Res
2005;70:129–136. https://doi.org/10.1016/j.mvr.2005.08.001.
[36] Farrell GC, Teoh NC, McCuskey RS. Hepatic microcirculation in fatty
liver disease. Anat Rec (Hoboken) 2008;291:684–692. https://doi.org/
10.1002/ar.20715.
[37] Caldwell S, Lackner C. Perspectives on NASH Histology: Cellular
Ballooning. Ann Hepatol 2017;16:182–184. https://doi.org/10.5604/
16652681.1231550.
[38] Ijaz S, Yang W, Winslet MC, Seifalian AM. Impairment of hepatic
microcirculation in fatty liver. Microcirculation 2003;10:447–456.
https://doi.org/10.1038/sj.mn.7800206.
[39] Flammer AJ, Anderson T, Celermajer DS, Creager MA, Deanfield J, Ganz
P, et al. The assessment of endothelial function: from research into
clinical practice. Circulation 2012;126:753–767. https://doi.org/
10.1161/CIRCULATIONAHA.112.093245.
[40] Tateya S, Rizzo NO, Handa P, Cheng AM, Morgan-Stevenson V, Daum G,
et al. Endothelial NO/cGMP/VASP signaling attenuates Kupffer cell
activation and hepatic insulin resistance induced by high-fat feeding.
Diabetes 2011;60:2792–2801. https://doi.org/10.2337/db11-0255.
[41] Pasarín M, La Mura V, Gracia-Sancho J, García-Calderó H, Rodríguez-
Vilarrupla A, García-Pagán JC, et al. Sinusoidal endothelial dysfunction
precedes inflammation and fibrosis in a model of NAFLD. PLoS One
2012;7. https://doi.org/10.1371/journal.pone.0032785 e32785.
[42] Gonzalez-Paredes FJ,Hernández Mesa G, Morales Arraez D, Marcelino
Reyes R, Abrante B, Diaz-Flores F, et al. Contribution of cyclooxygenase
end products and oxidative stress to intrahepatic endothelial dysfunc-
tion in early non-alcoholic fatty liver disease. PLoS One 2016;11.
https://doi.org/10.1371/journal.pone.0156650 e0156650.
[43] García-Lezana T, Raurell I, Bravo M, Torres-Arauz M, Salcedo MT,
Santiago A, et al. Restoration of a healthy intestinal microbiota
normalizes portal hypertension in a rat model of nonalcoholic steato-
hepatitis. Hepatology 2018;67:1485–1498. https://doi.org/10.1002/
hep.29646.
[44] Matsumoto M,Zhang J, Zhang X, Liu J, Jiang JX, Yamaguchi K, et al. The
NOX1 isoform of NADPH oxidase is involved in dysfunction of
liver sinusoids in nonalcoholic fatty liver disease. Free Radical Biol
Med 2018;115:412–420. https://doi.org/10.1016/j.freeradbiomed.
2017.12.019.
[45] Pasarín M,Abraldes JG, Rodríguez-Vilarrupla A, La Mura V, García-
Pagán JC, Bosch J. Insulin resistance and liver microcirculation in a rat
model of early NAFLD. J Hepatol 2011;55:1095–1102. https://doi.org/
10.1016/j.jhep.2011.01.053.
[46] Pasarín M, Abraldes JG, Liguori E, Kok B, La Mura V. Intrahepatic
vascular changes in non-alcoholic fatty liver disease: Potential role of
insulin-resistance and endothelial dysfunction. World J Gastroenterol
2017;23:6777–6787. https://doi.org/10.3748/wjg.v23.i37.6777.
[47] Gunnett CA, Lund DD, Chu Y, Brooks RM, Faraci FM, Heistad DD. NO-
dependent vasorelaxation is impaired after gene transfer of inducible
NO-synthase. Arterioscler Thromb Vasc Biol 2001;21:1281–1287.
[48] Chauhan SD, Seggara G, Vo PA, Macallister RJ, Hobbs AJ, Ahluwalia A.
Protection against lipopolysaccharide-induced endothelial dysfunction
in resistance and conduit vasculature of iNOS knockout mice. FASEB J
2003;17:773–775. https://doi.org/10.1096/fj.02-0668fje.
[49] Kus E, Jasin
´ski K, Skórka T, Czyzynska-Cichon I, Chlopicki S. Short-term
treatment with hepatoselective NO donor V-PYRRO/NO improves
blood flow in hepatic microcirculation in liver steatosis in mice.
Pharmacol Rep 2018;70:463–469. https://doi.org/10.1016/j.
pharep.2017.11.019.
[50] Schild L, Dombrowski F, Lendeckel U, Schulz C, Gardemann A, Keilhoff
G. Impairment of endothelial nitric oxide synthase causes abnormal fat
and glycogen deposition in liver. Biochim Biophys Acta
2008;1782:180–187. https://doi.org/10.1016/j.bbadis.2007.12.007.
[51] Roediger WE, Hems R, Wiggins D, Gibbons GF. Inhibition of hepatocyte
lipogenesis by nitric oxide donor: could nitric oxide regulate lipid
synthesis? IUBMB Life 2004;56:35–40. https://doi.org/10.1080/
15216540310001649822.
Review
1288 Journal of Hepatology 2019 vol. 70 j1278–1291
[52] Winder WW, Hardie DG. Inactivation of acetyl-CoA carboxylase and
activation of AMP-activated protein kinase in muscle during exercise.
Am J Physiol 1996;270:E299–E304. https://doi.org/10.1152/ajpen-
do.1996.270.2.E299.
[53] Fu WJ, Haynes TE, Kohli R, Hu J, Shi W, Spencer TE, et al. Dietary L-
arginine supplementation reduces fat mass in Zucker diabetic fatty
rats. J Nutr 2005;135:714–721. https://doi.org/10.1093/jn/135.4.714.
[54] Zou M-H, Hou X-Y, Shi C-M, Kirkpatick S, Liu F, Goldman MH, et al.
Activation of 5’-AMP-activated kinase is mediated through c-Src and
phosphoinositide 3-kinase activity during hypoxia-reoxygenation of
bovine aortic endothelial cells. Role of peroxynitrite. J Biol Chem
2003;278:34003–34010. https://doi.org/10.1074/jbc.M300215200.
[55] Park H, Kaushik VK, Constant S, Prentki M, Przybytkowski E, Ruderman
NB, et al. Coordinate regulation of malonyl-CoA decarboxylase, sn-
glycerol-3-phosphate acyltransferase, and acetyl-CoA carboxylase by
AMP-activated protein kinase in rat tissues in response to exercise. J
Biol Chem 2002;277:32571–32577. https://doi.org/10.1074/jbc.
M201692200.
[56] Muoio DM, Seefeld K, Witters LA, Coleman RA. AMP-activated kinase
reciprocally regulates triacylglycerol synthesis and fatty acid oxidation
in liver and muscle: evidence that sn-glycerol-3-phosphate acyltrans-
ferase is a novel target. Biochem J 1999;338(Pt 3):783–791.
[57] Doulias P-T, Tenopoulou M, Greene JL, Raju K, Ischiropoulos H. Nitric
oxide regulates mitochondrial fatty acid metabolism through rever-
sible protein S-nitrosylation. Sci Signal 2013;6:rs1. https://doi.org/
10.1126/scisignal.2003252.
[58] Maslak E, Zabielski P, Kochan K, Kus K, Jasztal A, Sitek B, et al. The liver-
selective NO donor, V-PYRRO/NO, protects against liver steatosis and
improves postprandial glucose tolerance in mice fed high fat diet.
Biochem Pharmacol 2015;93:389–400. https://doi.org/10.1016/j.
bcp.2014.12.004.
[59] Kus K, Walczak M, Maslak E, Zakrzewska A, Gonciarz-Dytman A,
Zabielski P, et al. Hepatoselective nitric oxide (NO) donors, V-PYRRO/
NO and V-PROLI/NO, in nonalcoholic fatty liver disease: a comparison
of antisteatotic effects with the biotransformation and pharmacoki-
netics. Drug Metab Dispos 2015;43:1028–1036. https://doi.org/
10.1124/dmd.115.063388.
[60] Wang W, Zhao C, Zhou J, Zhen Z, Wang Y, Shen C. Simvastatin
ameliorates liver fibrosis via mediating nitric oxide synthase in rats
with non-alcoholic steatohepatitis-related liver fibrosis. PLoS ONE
2013;8. https://doi.org/10.1371/journal.pone.0076538 e76538.
[61] Coulon S,Heindryckx F, Geerts A, Van Steenkiste C, Colle I, Van
Vlierberghe H. Angiogenesis in chronic liver disease and its complica-
tions. Liver Int 2011;31:146–162. https://doi.org/10.1111/j.1478-
3231.2010.02369.x.
[62] Coulon S, Francque S, Colle I, Verrijken A, Blomme B, Heindryckx F,
et al. Evaluation of inflammatory and angiogenic factors in patients
with non-alcoholic fatty liver disease. Cytokine 2012;59:442–449.
https://doi.org/10.1016/j.cyto.2012.05.001.
[63] Coulon S, Legry V, Heindryckx F, Van Steenkiste C, Casteleyn C, Olievier
K, et al. Role of vascular endothelial growth factor in the pathophys-
iology of nonalcoholic steatohepatitis in two rodent models. Hepatol-
ogy 2013;57:1793–1805. https://doi.org/10.1002/hep.26219.
[64] Iwakiri Y, Shah V, Rockey DC. Vascular pathobiology in chronic liver
disease and cirrhosis - current status and future directions. J Hepatol
2014;61:912–924. https://doi.org/10.1016/j.jhep.2014.05.047.
[65] Tarantino G, Conca P, Pasanisi F, Ariello M, Mastrolia M, Arena A, et al.
Could inflammatory markers help diagnose nonalcoholic steatohepati-
tis? Eur J Gastroenterol Hepatol 2009;21:504–511. https://doi.org/
10.1097/MEG.0b013e3283229b40.
[66] Kitade M, Yoshiji H, Kojima H, Ikenaka Y, Noguchi R, Kaji K, et al.
Neovascularization and oxidative stress in the progression of non-
alcoholic steatohepatitis. Mol Med Rep 2008;1:543–548.
[67] Kitade M, Yoshiji H, Noguchi R, Ikenaka Y, Kaji K, Shirai Y, et al.
Crosstalk between angiogenesis, cytokeratin-18, and insulin resistance
in the progression of non-alcoholic steatohepatitis. World J Gastroen-
terol 2009;15:5193–5199.
[68] Lefere S, Van de Velde F, Hoorens A, Raevens S, Van Campenhout S,
Vandierendonck A, et al. Angiopoietin-2 promotes pathological angio-
genesis and is a novel therapeutic target in murine non-alcoholic fatty
liver disease. Hepatology 2018. https://doi.org/10.1002/hep.30294.
[69] Peters KM,Wilson RB, Borradaile NM. Non-parenchymal hepatic cell
lipotoxicity and the coordinated progression of non-alcoholic fatty
liver disease and atherosclerosis. Curr Opin Lipidol 2018. https://doi.
org/10.1097/MOL.0000000000000535.
[70] Seki S, Kitada T, Yamada T, Sakaguchi H, Nakatani K, Wakasa K. In situ
detection of lipid peroxidation and oxidative DNA damage in non-
alcoholic fatty liver diseases. J Hepatol 2002;37:56–62.
[71] Rinella ME. Nonalcoholic fatty liver disease: a systematic review. JAMA
2015;313:2263–2273. https://doi.org/10.1001/jama.2015.5370.
[72] Brenner C,Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals
in liver inflammation. J Hepatol 2013;59:583–594. https://doi.org/
10.1016/j.jhep.2013.03.033.
[73] Bissell DM, Wang SS, Jarnagin WR, Roll FJ. Cell-specific expression of
transforming growth factor-beta in rat liver. Evidence for autocrine
regulation of hepatocyte proliferation. J Clin Invest 1995;96:447–455.
https://doi.org/10.1172/JCI118055.
[74] Carambia A, Freund B, Schwinge D, Heine M, Laschtowitz A, Huber S,
et al. TGF-b-dependent induction of CD4
+
CD25
+
Foxp3
+
Tregs by liver
sinusoidal endothelial cells. J Hepatol 2014;61:594–599. https://doi.
org/10.1016/j.jhep.2014.04.027.
[75] Limmer A,Ohl J, Kurts C, Ljunggren HG, Reiss Y, Groettrup M, et al.
Efficient presentation of exogenous antigen by liver endothelial cells to
CD8+ T cells results in antigen-specific T-cell tolerance. Nat Med
2000;6:1348–1354. https://doi.org/10.1038/82161.
[76] Berg M,Wingender G, Djandji D, Hegenbarth S, Momburg F, Hämmer-
ling G, et al. Cross-presentation of antigens from apoptotic tumor cells
by liver sinusoidal endothelial cells leads to tumor-specific CD8+ T cell
tolerance. Eur J Immunol 2006;36:2960–2970. https://doi.org/10.1002/
eji.200636033.
[77] McMahan RH, Porsche CE, Edwards MG, Rosen HR. Free fatty acids
differentially downregulate chemokines in liver sinusoidal endothelial
cells: insights into non-alcoholic fatty liver disease. PLoS One 2016;11.
https://doi.org/10.1371/journal.pone.0159217 e0159217.
[78] Ip E, Farrell G, Hall P, Robertson G, Leclercq I. Administration of the
potent PPARalpha agonist, Wy-14,643, reverses nutritional fibrosis and
steatohepatitis in mice. Hepatology 2004;39:1286–1296. https://doi.
org/10.1002/hep.20170.
[79] Dela Peña A, Leclercq I, Field J, George J, Jones B, Farrell G. NF-kappaB
activation, rather than TNF, mediates hepatic inflammation in a murine
dietary model of steatohepatitis. Gastroenterology
2005;129:1663–1674. https://doi.org/10.1053/j.gastro.2005.09.004.
[80] Weston CJ,Shepherd EL,Claridge LC, Rantakari P, Curbishley SM,
Tomlinson JW, et al. Vascular adhesion protein-1 promotes liver
inflammation and drives hepatic fibrosis. J Clin Invest
2015;125:501–520. https://doi.org/10.1172/JCI73722.
[81] Tomita K, Tamiya G, Ando S, Ohsumi K, Chiyo T, Mizutani A, et al.
Tumour necrosis factor alpha signalling through activation of Kupffer
cells plays an essential role in liver fibrosis of non-alcoholic steato-
hepatitis in mice. Gut 2006;55:415–424. https://doi.org/10.1136/
gut.2005.071118.
[82] Miyach iY, Tsuchiy a K, Komiya C, Shiba K, Shimazu N, Yamaguchi S, et al.
Roles for cell-cell adhesion and contact in obesity-induced hepatic
myeloid cell accumulation and glucose intolerance. Cell Rep
2017;18:2766–2779. https://doi.org/10.1016/j.celrep. 2017.02.039.
[83] Wu J,Meng Z, Jiang M, Zhang E, Trippler M, Broering R, et al. Toll-like
receptor-induced innate immune responses in non-parenchymal liver
cells are cell type-specific. Immunology 2010;129:363–374. https://
doi.org/10.1111/j.1365-2567.2009.03179.x.
[84] Feder LS, Todaro JA, Laskin DL. Characterization of interleukin-1 and
interleukin-6 production by hepatic endothelial cells and macro-
phages. J Leukoc Biol 1993;53:126–132.
[85] Marra F, Tacke F. Roles for chemokines in liver disease. Gastroenterol-
ogy 2014;147(577–594). https://doi.org/10.1053/j.gastro.2014.06.043
e1.
[86] Roh Y-S, Seki E. Chemokines and chemokine receptors in the develop-
ment of NAFLD. Adv Exp Med Biol 2018;1061:45–53. https://doi.org/
10.1007/978-981-10-8684-7_4.
[87] Shetty S, Lalor PF, Adams DH. Lymphocyte recruitment to the liver:
molecular insights into the pathogenesis of liver injury and hepatitis.
Toxicology 2008;254:136–146. https://doi.org/10.1016/
j.tox.2008.08.003.
[88] Lalor PF, Shields P, Grant A, Adams DH. Recruitment of lymphocytes to
the human liver. Immunol Cell Biol 2002;80:52–64. https://doi.org/
10.1046/j.1440-1711.2002.01062.x.
[89] Lalor PF, Edwards S, McNab G, Salmi M, Jalkanen S, Adams DH. Vascular
adhesion protein-1 mediates adhesion and transmigration of lympho-
cytes on human hepatic endothelial cells. J Immunol
2002;169:983–992.
JOURNAL
OF HEPATOLOGY
Journal of Hepatology 2019 vol. 70 j1278–1291 1289
[90] Edwards S,Lalor PF, Nash GB, Rainger GE, Adams DH. Lymphocyte
traffic through sinusoidal endothelial cells is regulated by hepatocytes.
Hepatology 2005;41:451–459. https://doi.org/10.1002/hep.20585.
[91] Sutter AG, Palanisamy AP, Lench JH, Esckilsen S, Geng T, Lewin DNB,
et al. Dietary saturated fat promotes development of hepatic inflam-
mation through toll-like receptor 4 in mice. J Cell Biochem
2016;117:1613–1621. https://doi.org/10.1002/jcb.25453.
[92] Fontana L, Eagon JC, Trujillo ME, Scherer PE, Klein S. Visceral fat
adipokine secretion is associated with systemic inflammation in obese
humans. Diabetes 2007;56:1010–1013. https://doi.org/10.2337/db06-
1656.
[93] Harte AL,da Silva NF, Creely SJ, McGee KC, Billyard T, Youssef-Elabd
EM, et al. Elevated endotoxin levels in non-alcoholic fatty liver disease.
J Inflamm (Lond) 2010;7:15. https://doi.org/10.1186/1476-9255-7-15.
[94] Brun P, Castagliuolo I, Di Leo V, Buda A, Pinzani M, Palù G, et al.
Increased intestinal permeability in obese mice: new evidence in the
pathogenesis of nonalcoholic steatohepatitis. Am J Physiol Gastrointest
Liver Physiol 2007;292:G518–G525. https://doi.org/10.1152/
ajpgi.00024.2006.
[95] Kazankov K, Jørgensen SMD, Thomsen KL, Møller HJ, Vilstrup H, George
J, et al. The role of macrophages in nonalcoholic fatty liver disease and
nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol 2018.
https://doi.org/10.1038/s41575-018-0082-x.
[96] Tamaki Y, Nakade Y, Yamauchi T, Makino Y, Yokohama S, Okada M,
et al. Angiotensin II type 1 receptor antagonist prevents hepatic
carcinoma in rats with nonalcoholic steatohepatitis. J Gastroenterol
2013;48:491–503. https://doi.org/10.1007/s00535-012-0651-7.
[97] Yoshiji H, Kuriyama S, Noguchi R, Ikenaka Y, Kitade M, Kaji K, et al.
Angiotensin-II and vascular endothelial growth factor interaction plays
an important role in rat liver fibrosis development. Hepatol Res
2006;36:124–129. https://doi.org/10.1016/j.hepres.2006.07.003.
[98] Cayón A, Crespo J, Guerra AR, Pons-Romero F. Gene expression in obese
patients with non-alcoholic steatohepatitis. Rev Esp Enferm Dig
2008;100:212–218.
[99] Kietzmann T, Görlach A. Reactive oxygen species in the control of
hypoxia-inducible factor-mediated gene expression. Semin Cell Dev
Biol 2005;16:474–486. https://doi.org/10.1016/j.semcdb.2005.03.010.
[100] Povero D, Eguchi A, Niesman IR, Andronikou N, de Mollerat du Jeu X,
Mulya A, et al. Lipid-induced toxicity stimulates hepatocytes to release
angiogenic microparticles that require Vanin-1 for uptake by endothe-
lial cells. Sci Signal 2013;6:88. https://doi.org/10.1126/
scisignal.2004512.
[101] Kim M, Allen B, Korhonen EA, Nitschké M, Yang HW, Baluk P, et al.
Opposing actions of angiopoietin-2 on Tie2 signaling and FOXO1
activation. J Clin Invest 2016;126:3511–3525. https://doi.org/10.1172/
JCI84871.
[102] Tugues S, Fernandez-Varo G, Muñoz-Luque J, Ros J, Arroyo V, Rodés J,
et al. Antiangiogenic treatment with sunitinib ameliorates inflamma-
tory infiltrate, fibrosis, and portal pressure in cirrhotic rats. Hepatology
2007;46:1919–1926. https://doi.org/10.1002/hep.21921.
[103] Van Steenkiste C,Ribera J, Geerts A, Pauta M, Tugues S, Casteleyn C,
et al. Inhibition of placental growth factor activity reduces the severity
of fibrosis, inflammation, and portal hypertension in cirrhotic mice.
Hepatology 2011;53:1629–1640. https://doi.org/10.1002/hep.24238.
[104] Thabut D,Routray C, Lomberk G, Shergill U, Glaser K, Huebert R, et al.
Complementary vascular and matrix regulatory pathways underlie the
beneficial mechanism of action of sorafenib in liver fibrosis. Hepatol-
ogy 2011;54:573–585. https://doi.org/10.1002/hep.24427.
[105] Öztürk Akcora B, Storm G, Prakash J, Bansal R. Tyrosine kinase inhibitor
BIBF1120 ameliorates inflammation, angiogenesis and fibrosis in CCl4-
induced liver fibrogenesis mouse model. Sci Rep 2017;7:44545.
https://doi.org/10.1038/srep44545.
[106] Yang L, Kwon J, Popov Y, Gajdos GB, Ordog T, Brekken RA, et al.
Vascular endothelial growth factor promotes fibrosis resolution and
repair in mice. Gastroenterology 2014;146(1339–1350). https://doi.
org/10.1053/j.gastro.2014.01.061 e1.
[107] Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation.
Nat Rev Gastroenterol Hepatol 2017;14:397–411. https://doi.org/
10.1038/nrgastro.2017.38.
[108] Schaffner F, Poper H. Capillarization of hepatic sinusoids in man.
Gastroenterology 1963;44:239–242.
[109] Xu B, Broome U, Uzunel M, Nava S, Ge X, Kumagai-Braesch M, et al.
Capillarization of hepatic sinusoid by liver endothelial cell-reactive
autoantibodies in patients with cirrhosis and chronic hepatitis. Am J
Pathol 2003;163:1275–1289. https://doi.org/10.1016/S0002-9440(10)
63487-6.
[110] Sørensen KK, Simon-Santamaria J, McCuskey RS, Smedsrød B. Liver
sinusoidal endothelial cells. Compr Physiol 2015;5:1751–1774. https://
doi.org/10.1002/cphy.c140078.
[111] DeLeve LD, Wang X, Kanel GC, Atkinson RD, McCuskey RS. Prevention
of hepatic fibrosis in a murine model of metabolic syndrome with
nonalcoholic steatohepatitis. Am J Pathol 2008;173:993–1001. https://
doi.org/10.2353/ajpath.2008.070720.
[112] DeLeve LD. Liver sinusoidal endothelial cells in hepatic fibrosis.
Hepatology 2015;61:1740–1746. https://doi.org/10.1002/hep.27376.
[113] Maslak E, Gregorius A, Chlopicki S. Liver sinusoidal endothelial cells
(LSECs) function and NAFLD; NO-based therapy targeted to the liver.
Pharmacol Rep 2015;67:689–694. https://doi.org/10.1016/j.
pharep.2015.04.010.
[114] Rangwala F, Guy CD, Lu J, Suzuki A, Burchette JL, Abdelmalek MF, et al.
Increased production of sonic hedgehog by ballooned hepatocytes. J
Pathol 2011;224:401–410. https://doi.org/10.1002/path.2888.
[115] Xie G, Choi SS, Syn W-K, Michelotti GA, Swiderska M, Karaca G, et al.
Hedgehog signalling regulates liver sinusoidal endothelial cell capil-
larisation. Gut 2013;62:299–309. https://doi.org/10.1136/gutjnl-2011-
301494.
[116] Matz-Soja M, Gebhardt R. The many faces of Hedgehog signalling in the
liver: recent progress reveals striking cellular diversity and the
importance of microenvironments. J Hepatol 2014;61:1449–1450.
https://doi.org/10.1016/j.jhep.2014.06.041.
[117] Witek RP, Yang L, Liu R, Jung Y, Omenetti A, Syn W-K, et al. Liver cell-
derived microparticles activate hedgehog signaling and alter gene
expression in hepatic endothelial cells. Gastroenterology 2009;136
(320–330). https://doi.org/10.1053/j.gastro.2008.09.066 e2.
[118] Deleve LD, Wang X, Guo Y. Sinusoidal endothelial cells prevent rat
stellate cell activation and promote reversion to quiescence. Hepatol-
ogy 2008;48:920–930. https://doi.org/10.1002/hep.22351.
[119] Marrone G,Russo L, Rosado E, Hide D, García-Cardeña G, García-Pagán
JC, et al. The transcription factor KLF2 mediates hepatic endothelial
protection and paracrine endothelial-stellate cell deactivation induced
by statins. J Hepatol 2013;58:98–103. https://doi.org/10.1016/j.
jhep.2012.08.026.
[120] Li Y, Lui KO, Zhou B. Reassessing endothelial-to-mesenchymal transi-
tion in cardiovascular diseases. Nat Rev Cardiol 2018;15:445–456.
https://doi.org/10.1038/s41569-018-0023-y.
[121] Piera-Velazquez S, Mendoza FA, Jimenez SA. Endothelial to mesenchy-
mal transition (EndoMT) in the pathogenesis of human fibrotic
diseases. J Clin Med 2016:5. https://doi.org/10.3390/jcm5040045.
[122] Singh KK, Lovren F, Pan Y, Quan A, Ramadan A, Matkar PN, et al. The
essential autophagy gene ATG7 modulates organ fibrosis via regulation
of endothelial-to-mesenchymal transition. J Biol Chem
2015;290:2547–2559. https://doi.org/10.1074/jbc.M114.604603.
[123] Smedsrød B, De Bleser PJ, Braet F, Lovisetti P, Vanderkerken K, Wisse E,
et al. Cell biology of liver endothelial and Kupffer cells. Gut
1994;35:1509–1516.
[124] Wang R, Ding Q, Yaqoob U, de Assuncao TM, Verma VK, Hirsova P, et al.
Exosome adherence and internalization by hepatic stellate cells
triggers sphingosine 1-phosphate-dependent migration. J Biol Chem
2015;290:30684–30696. https://doi.org/10.1074/jbc.M115.671735.
[125] Wells RG. Cellular sources of extracellular matrix in hepatic fibrosis. Clin
Liver Dis 2008;12:759–768. https://doi.org/10.1016/j.cld.2008.07.008, viii.
[126] Ribera J, Pauta M, Melgar-Lesmes P, Córdoba B, Bosch A, Calvo M, et al.
A small population of liver endothelial cells undergoes endothelial-to-
mesenchymal transition in response to chronic liver injury. Am J
Physiol Gastrointest Liver Physiol 2017;313:G492–G504. https://doi.
org/10.1152/ajpgi.00428.2016.
[127] Lemoinne S, Thabut D, Housset C, Moreau R, Valla D, Boulanger CM,
et al. The emerging roles of microvesicles in liver diseases. Nat Rev
Gastroenterol Hepatol 2014;11:350–361. https://doi.org/10.1038/
nrgastro.2014.7.
[128] Huang X-D, Fan Y, Zhang H, Wang P, Yuan J-P, Li M-J, et al. Serum leptin
and soluble leptin receptor in non-alcoholic fatty liver disease. World J
Gastroenterol 2008;14:2888–2893.
[129] Kitade M, Yoshiji H, Kojima H, Ikenaka Y, Noguchi R, Kaji K, et al.
Leptin-mediated neovascularization is a prerequisite for progression of
nonalcoholic steatohepatitis in rats. Hepatology 2006;44:983–991.
https://doi.org/10.1002/hep.21338.
[130] Marra F, Bertolani C. Adipokines in liver diseases. Hepatology
2009;50:957–969. https://doi.org/10.1002/hep.23046.
[131] Zhou L-Y, Zeng H, Wang S, Chen J-X. Regulatory role of endothelial
PHD2 in the hepatic steatosis. Cell Physiol Biochem
2018;48:1003–1011. https://doi.org/10.1159/000491968.
Review
1290 Journal of Hepatology 2019 vol. 70 j1278–1291
[132] Ozer A, Bruick RK. Regulation of HIF by prolyl hydroxylases: recruit-
ment of the candidate tumor suppressor protein ING4. Cell Cycle
2005;4:1153–1156. https://doi.org/10.4161/cc.4.9.2040.
[133] Kantari-Mimoun C, Castells M, Klose R, Meinecke A-K, Lemberger UJ,
Rautou P-E, et al. Resolution of liver fibrosis requires myeloid cell-
driven sinusoidal angiogenesis. Hepatology 2015;61:2042–2055.
https://doi.org/10.1002/hep.27635.
[134] Taura K, De Minicis S, Seki E, Hatano E, Iwaisako K, Osterreicher CH,
et al. Hepatic stellate cells secrete angiopoietin 1 that induces
angiogenesis in liver fibrosis. Gastroenterology 2008;135:1729–1738.
https://doi.org/10.1053/j.gastro.2008.07.065.
[135] Yoshiji H, Kuriyama S, Yoshii J, Ikenaka Y, Noguchi R, Hicklin DJ, et al.
Vascular endothelial growth factor and receptor interaction is a
prerequisite for murine hepatic fibrogenesis. Gut 2003;52:1347–1354.
[136] Zhao S, Zhang Z, Yao Z, Shao J, Chen A, Zhang F, et al. Tetram-
ethylpyrazine attenuates sinusoidal angiogenesis via inhibition of
hedgehog signaling in liver fibrosis. IUBMB Life 2017;69:115–127.
https://doi.org/10.1002/iub.1598.
[137] Hennenberg M, Trebicka J, Kohistani Z, Stark C, Nischalke H-D, Krämer
B, et al. Hepatic and HSC-specific sorafenib effects in rats with
established secondary biliary cirrhosis. Lab Invest 2011;91:241–251.
https://doi.org/10.1038/labinvest.2010.148.
[138] Coch L, Mejias M, Berzigotti A, Garcia-Pras E, Gallego J, Bosch J, et al.
Disruption of negative feedback loop between vasohibin-1 and vascu-
lar endothelial growth factor decreases portal pressure, angiogenesis,
and fibrosis in cirrhotic rats. Hepatology 2014;60:633–647. https://doi.
org/10.1002/hep.26995.
[139] Chatterjee S. Reversal of vasohibin-driven negative feedback loop of
vascular endothelial growth factor/angiogenesis axis promises a novel
antifibrotic therapeutic strategy for liver diseases. Hepatology
2014;60:458–460. https://doi.org/10.1002/hep.27061.
[140] Yang ZF, Poon RTP. Vascular changes in hepatocellular carcinoma. Anat
Rec (Hoboken) 2008;291:721–734. https://doi.org/10.1002/ar.20668.
[141] Géraud C, Mogler C, Runge A, Evdokimov K, Lu S, Schledzewski K, et al.
Endothelial transdifferentiation in hepatocellular carcinoma: loss of
Stabilin-2 expression in peri-tumourous liver correlates with increased
survival. Liver Int 2013;33:1428–1440. https://doi.org/10.1111/
liv.12262.
[142] Wu LQ, Zhang WJ, Niu JX, Ye LY, Yang ZH, Grau GE, et al. Phenotypic
and functional differences between human liver cancer endothelial
cells and liver sinusoidal endothelial cells. J Vasc Res 2008;45:78–86.
https://doi.org/10.1159/000109079.
[143] Höchst B, Schildberg FA, Böttcher J, Metzger C, Huss S, Türler A, et al.
Liver sinusoidal endothelial cells contribute to CD8 T cell tolerance
toward circulating carcinoembryonic antigen in mice. Hepatology
2012;56:1924–1933. https://doi.org/10.1002/hep.25844.
[144] Milner K-L, van der Poorten D, Xu A, Bugianesi E, Kench JG, Lam KSL,
et al. Adipocyte fatty acid binding protein levels relate to inflammation
and fibrosis in nonalcoholic fatty liver disease. Hepatology
2009;49:1926–1934. https://doi.org/10.1002/hep.22896.
[145] Laouirem S, Sannier A, Norkowski E, Cauchy F, Doblas S, Rautou PE,
et al. Endothelial fatty liver binding protein 4: a new targetable
mediator in hepatocellular carcinoma related to metabolic syndrome.
Oncogene 2018. https://doi.org/10.1038/s41388-018-0597-1.
[146] Yoshiji H, Noguchi R, Namisaki T, Moriya K, Kitade M, Aihara Y, et al.
Combination of sorafenib and angiotensin-II receptor blocker attenu-
ates preneoplastic lesion development in a non-diabetic rat model of
steatohepatitis. J Gastroenterol 2014;49:1421–1429. https://doi.org/
10.1007/s00535-013-0906-y.
[147] Gracia-Sancho J, Marrone G, Fernández-Iglesias A. Hepatic microcircu-
lation and mechanisms of portal hypertension. Nat Rev Gastroenterol
Hepatol 2018. https://doi.org/10.1038/s41575-018-0097-3.
JOURNAL
OF HEPATOLOGY
Journal of Hepatology 2019 vol. 70 j1278–1291 1291
... It receives blood-borne free fatty acids (FFAs), lipoproteins and carbohydrates and utilizes these to produce new triglycerides (TGs), which are packaged and secreted into the bloodstream as very low-density lipoproteins (VLDL) 1 . Previous data suggest that the exchange of macromolecules between the blood and hepatocytes is facilitated by highly permeable liver sinusoidal endothelial cells (LSECs), which separate the sinusoidal vascular lumen from the microvilli of the hepatocytes [2][3][4][5] . LSECs typically harbor 50-300 nm-sized fenestrae or pores, which are organized in sieve plates 2,3 . ...
... Information about LSEC porosity and defenestration in individuals with MASLD is still scarce, even though a recent study reported less defenestration in liver biopsies from human individuals with MASH compared to those with MASLD, but without MASH 16 , indicating that defenestration preferentially takes place during an early stage of MASLD. It can be speculated that defenestration of LSEC reduces the export of VLDL from the liver (possibly contributing to hepatic steatosis) and prevents hepatic removal of chylomicron remnants from the bloodstream (possibly contributing to hyperlipidemia) 5,17 , thus triggering early development of MASLD; however, the molecular basis of LSEC defenestration and its contribution to MASLD, including genetic triggers of defenestration, are largely unknown 11 . ...
Article
Full-text available
Prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease, increases worldwide and associates with type 2 diabetes and other cardiometabolic diseases. Here we demonstrate that Sema3a is elevated in liver sinusoidal endothelial cells of animal models for obesity, type 2 diabetes and MASLD. In primary human liver sinusoidal endothelial cells, saturated fatty acids induce expression of SEMA3A , and loss of a single allele is sufficient to reduce hepatic fat content in diet-induced obese mice. We show that semaphorin-3A regulates the number of fenestrae through a signaling cascade that involves neuropilin-1 and phosphorylation of cofilin-1 by LIM domain kinase 1. Finally, inducible vascular deletion of Sema3a in adult diet-induced obese mice reduces hepatic fat content and elevates very low-density lipoprotein secretion. Thus, we identified a molecular pathway linking hyperlipidemia to microvascular defenestration and early development of MASLD.
... LSECs possess endothelial nitric oxide synthase (eNOS), which is responsible for the production of nitric oxide (NO), a key vasodilator molecule regulating sinusoidal flow within the liver [81]. In response to biomechanical, inflammatory, and immune-mediated signals, LSECs undergo distinct anatomical changes, including a loss of their fenestration and the development of basal lamina, negatively affecting molecular transport between portal blood and hepatocytes [82]. Moreover, structural changes in endothelial dysfunction are accompanied by diminished eNOS activity and NO generation as a hallmark of LSEC injury [81]. ...
Article
Full-text available
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a common condition with heterogeneous outcomes difficult to predict at the individual level. Feared complications of advanced MASLD are linked to clinically significant portal hypertension and are initiated by functional and mechanical changes in the unique sinusoidal capillary network of the liver. Early sinusoidal vasoregulatory changes in MASLD lead to increased intrahepatic vascular resistance and represent the beginning of portal hypertension. In addition, the composition and function of gut microbiota in MASLD are distinctly different from the healthy state, and multiple lines of evidence demonstrate the association of dysbiosis with these vasoregulatory changes. The gut microbiota is involved in the biotransformation of nutrients, production of de novo metabolites, release of microbial structural components, and impairment of the intestinal barrier with impact on innate immune responses, metabolism, inflammation, fibrosis, and vasoregulation in the liver and beyond. The gut–liver axis is a conceptual framework in which portal circulation is the primary connection between gut microbiota and the liver. Accordingly, biochemical and hemodynamic attributes of portal circulation may hold the key to better understanding and predicting disease progression in MASLD. However, many specific details remain hidden due to limited access to the portal circulation, indicating a major unmet need for the development of innovative diagnostic tools to analyze portal metabolites and explore their effect on health and disease. We also need to safely and reliably monitor portal hemodynamics with the goal of providing preventive and curative interventions in all stages of MASLD. Here, we review recent advances that link portal metabolomics to altered sinusoidal vasoregulation and may allow for new insights into the development of portal hypertension in MASLD.
... The reason for this may be that extensive resection of the liver parenchyma may result in a considerable reduction in the hepatic vascular bed, higher intrahepatic vascular resistance, PH, hyperperfusion of the remnant liver, and, ultimately, liver sinusoidal endothelial cell and hepatocyte injury. [7][8][9] Dysfunction of the liver sinusoidal endothelial cells can result in the release of inflammatory molecules and the subsequent recruitment of inflammatory cells, thus promoting liver injury and compensatory hepatocyte proliferation, resulting in hepatocarcinogenesis. 10 To the best of our knowledge, no study has focused on the influence of PHPH on patient outcomes. ...
Article
Full-text available
Aims Portal venous pressure frequently increases after a major hepatectomy. We aimed to assess the outcomes of patients with hepatocellular carcinoma (HCC) with different post‐hepatectomy portal pressure statuses and the predictive factors correlated with prognosis and post‐hepatectomy portal hypertension (PHPH). Methods Data from consecutive patients who underwent curative‐intent hepatectomy for HCC at the Eastern Hepatobiliary Surgery Hospital and Mengchao Hepatobiliary Hospital of Fujian Medical University between January 2008 and December 2016 were retrospectively analyzed. Patients were followed up until March 1, 2022. PHPH was defined as new‐onset portal hypertension (PH) in patients without preoperative PH within 1 month of hepatectomy. Patients were classified into two groups according to the presence or absence of PHPH: non‐PH and PHPH. The clinicopathological characteristics were compared between the two groups. Univariate and multivariate analyses were used to identify independent risk factors for early and late recurrence of HCC, overall survival (OS), and PHPH. Results Disease‐free survival rates were significantly higher in the non‐PH group ( n = 1068) than those in the PHPH group ( n = 423) (62.0%, 39.0%, and 31.0% vs. 46.2%, 24.5%, and 19.3% at 1, 3, and 5 years, respectively; all p < 0.001). The 1‐, 3‐, and 5‐year OS rates were 91.0%, 66.4%, and 51.4% in the non‐PH group and 80.0%, 48.9%, and 32.6% in the PHPH group, respectively (all p < 0.001). Multivariate analysis revealed that PHPH was independently associated with early recurrence of HCC and poor OS (hazard ratio [HR] 1.476, 95% confidence interval [CI] 1.279–1.704, p < 0.001 and HR 1.601, 95% CI 1.383–1.853, p < 0.001, respectively). Furthermore, hepatitis B virus infection, cirrhosis, preoperative alanine aminotransferase > 40 U/L, and major hepatectomy were identified as independent risk factors for PHPH. Conclusions PHPH was associated with early recurrence of HCC and poor OS but not with late recurrence. Therefore, PHPH may be an attractive target for therapeutic interventions and follow‐up surveillance to improve prognosis.
... Furthermore, altered LSECs contribute to hepatic angiogenesis, inflammation, fibrosis and HCC during the stages of NASH. Multiple pieces of evidence indicate that the dysfunction of LSECs in fatty livers is associated with increased intrahepatic vascular resistance related to steatosis [106]. ...
Article
Full-text available
Nonalcoholic fatty liver disease (NAFLD) is the liver component of a cluster of conditions, while its subtype, nonalcoholic steatohepatitis (NASH), emerges as a potentially progressive liver disorder that harbors the risk of evolving into cirrhosis and culminating in hepatocellular carcinoma (HCC). NASH and cardiovascular disease (CVD) have common risk factors, but compared to liver-related causes, the most common cause of death in NASH patients is CVD. Within the pharmacological armamentarium, statins, celebrated for their lipid-modulating prowess, have now garnered attention for their expansive therapeutic potential in NASH. Evidence from a plethora of studies suggests that statins not only manifest anti-inflammatory and antifibrotic properties but also impart a multifaceted beneficial impact on hepatic health. In this review, we used “statin”, “NAFLD”, “NASH”, and “CVD” as the major keywords and conducted a literature search using the PubMed and Web of Science databases to determine the safety and efficacy of statins in patients and animals with NASH and NAFLD, and the mechanism of statin therapy for NASH. Simultaneously, we reviewed the important role of the intestinal microbiota in statin therapy for NASH, as it is hoped that statins will provide new insights into modulating the harmful inflammatory microbiota in the gut and reducing systemic inflammation in NASH patients.
Article
Full-text available
Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells (ECs) that play an important role in liver development and regeneration. Additionally, it is involved in various pathological processes, including steatosis, inflammation, fibrosis and hepatocellular carcinoma. However, the rapid dedifferentiation of LSECs after culture greatly limits their use in vitro modeling for biomedical applications. In this study, we developed a highly efficient protocol to induce LSEC-like cells from human induced pluripotent stem cells (hiPSCs) in only 8 days. Using single-cell transcriptomic analysis, we identified several novel LSEC-specific markers, such as EPAS1, LIFR, and NID1, as well as several previously revealed markers, such as CLEC4M, CLEC1B, CRHBP and FCN3. These LSEC markers are specifically expressed in our LSEC-like cells. Furthermore, hiPSC-derived cells expressed LSEC-specific proteins and exhibited LSEC-related functions, such as the uptake of acetylated low density lipoprotein (ac-LDL) and immune complex endocytosis. Overall, this study confirmed that our novel protocol allowed hiPSCs to rapidly acquire an LSEC-like phenotype and function in vitro. The ability to generate LSECs efficiently and rapidly may help to more precisely mimic liver development and disease progression in a liver-specific multicellular microenvironment, offering new insights into the development of novel therapeutic strategies.
Article
Full-text available
Metabolic syndrome (MS) is becoming the leading risk factor for hepatocellular carcinoma (HCC). HCC development related to MS may occur in advanced or non-advanced liver fibrosis, suggesting specific molecular pathways. Among these pathways, basal inflammatory state and adipokines production are involved. The aim of this study was to evaluate the role of fatty acid-binding protein 4 (FABP4). In this study, we demonstrate the specific overexpression of FABP4 in human HCC samples from patients with MS compared to other risk factors for chronic liver disease with FABP4 expression restricted to peritumoral endothelial cells. In vitro, glucose, insulin, VEGFA and hypoxia upregulated endothelial FABP4, which was reversed by metformin through mTOR pathway inhibition. FABP4 exerts oncogenic effects on hepatoma cell lines by upregulating the angiogenesis gene signature and pathways involved in the cell cycle, leading to increased cell proliferation and migration, and downregulating HIF1 pathway; effects were reversed in the presence of a specific FABP4 inhibitor (BMS309403). We showed the role of microvesicles as FABP4 vectors between endothelial and tumor cells. In vivo, BMS309403 significantly reduces tumor growth in heterotopic and orthotopic xenografted mice model. In conclusion, this study demonstrates the emerging oncogenic role of liver endothelial cells through FABP4 in HCC related to MS, and highlights new anti-neoplastic mechanism of metformin.
Article
Full-text available
Maternal obesity is associated with increased risk for offspring obesity and non-alcoholic fatty liver disease (NAFLD), but the causal drivers of this association are unclear. Early colonization of the infant gut by microbes plays a critical role in establishing immunity and metabolic function. Here, we compare germ-free mice colonized with stool microbes (MB) from 2-week-old infants born to obese (Inf-ObMB) or normal-weight (Inf-NWMB) mothers. Inf-ObMB-colonized mice demonstrate increased hepatic gene expression for endoplasmic reticulum stress and innate immunity together with histological signs of periportal inflammation, a histological pattern more commonly reported in pediatric cases of NAFLD. Inf-ObMB mice show increased intestinal permeability, reduced macrophage phagocytosis, and dampened cytokine production suggestive of impaired macrophage function. Furthermore, exposure to a Western-style diet in Inf-ObMB mice promotes excess weight gain and accelerates NAFLD. Overall, these results provide functional evidence supporting a causative role of maternal obesity-associated infant dysbiosis in childhood obesity and NAFLD.
Article
Progressive changes in the hepatic perisinusoidal tissue space of Disse in chronic liver diseases ultimately lead to development of a basement membrane and capillarization of sinusoids. The transformation of an open circulation to a closed one decreases the effectiveness of hepatic circulation and aggravates hepatocellular insufficiency.
Article
The liver microcirculatory milieu, mainly composed of liver sinusoidal endothelial cells (LSECs), hepatic stellate cells (HSCs) and hepatic macrophages, has an essential role in liver homeostasis, including in preserving hepatocyte function, regulating the vascular tone and controlling inflammation. Liver microcirculatory dysfunction is one of the key mechanisms that promotes the progression of chronic liver disease (also termed cirrhosis) and the development of its major clinical complication, portal hypertension. In the present Review, we describe the current knowledge of liver microcirculatory dysfunction in cirrhotic portal hypertension and appraise the preclinical models used to study the liver circulation. We also provide a comprehensive summary of the promising therapeutic options to target the liver microvasculature in cirrhosis.
Article
Nonalcoholic fatty liver disease (NAFLD) and its inflammatory and often progressive subtype nonalcoholic steatohepatitis (NASH) are becoming the leading cause of liver-related morbidity and mortality worldwide, and a primary indication for liver transplantation. The pathophysiology of NASH is multifactorial and not yet completely understood; however, innate immunity is a major contributing factor in which liver-resident macrophages (Kupffer cells) and recruited macrophages play a central part in disease progression. In this Review, we assess the evidence for macrophage involvement in the development of steatosis, inflammation and fibrosis in NASH. In this process, not only the polarization of liver macrophages towards a pro-inflammatory phenotype is important, but adipose tissue macrophages, especially in the visceral compartment, also contribute to disease severity and insulin resistance. Macrophage activation is mediated by factors such as endotoxins and translocated bacteria owing to increased intestinal permeability, factors released from damaged or lipoapoptotic hepatocytes, as well as alterations in gut microbiota and defined nutritional components, including certain free fatty acids, cholesterol and their metabolites. Reflecting the important role of macrophages in NASH, we also review studies investigating drugs that target macrophage recruitment to the liver, macrophage polarization and their inflammatory effects as potential treatment options for patients with NASH.
Article
As the epidemics of obesity and type 2 diabetes mellitus increase worldwide, the prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing proportionately. The subtype of NAFLD which can be characterised as non-alcoholic steatohepatitis (NASH) is a potentially progressive liver disease that can lead to cirrhosis, hepatocellular carcinoma, liver transplantation, and death. NAFLD is also associated with extrahepatic manifestations such as chronic kidney disease, cardiovascular disease and sleep apnoea. NAFLD and NASH carry a large economic burden and create poor health-related quality of life. Despite this important burden, we are only beginning to understand its mechanisms of pathogenesis and the contribution of environmental and genetic factors to the risk of developing a progressive course of disease. Research is underway to identify appropriate non-invasive diagnostic methods and effective treatments. Although the risk of liver-related mortality is increased in patients with NAFLD and liver fibrosis stages F3 or F4, the leading cause of death is cardiovascular disease. Given the rapidly growing global burden of NAFLD and NASH, efforts must continue to find accurate non-invasive diagnostic and prognostic biomarkers, to develop effective treatments for individuals with advanced NASH and prevention methods for individuals at high risk of NAFLD and progressive liver disease.
Article
Angiogenesis contributes to the development of non-alcoholic steatohepatitis (NASH) and promotes inflammation, fibrosis and progression to hepatocellular carcinoma (HCC). Angiopoietin-2 (Ang-2) is a key regulator of angiogenesis. We aimed to investigate the role of Ang-2 and its potential as therapeutic target in NASH using human samples, in vivo mouse models and in vitro assays. Serum Ang-2 levels were determined in 104 obese patients undergoing bariatric surgery and concomitant liver biopsy. The effect of the Ang-2/Tie2 receptor inhibiting peptibody L1-10 was evaluated in the methionine-choline deficient (MCD) and streptozotocin-western diet NAFLD mouse models, and in vitro on endothelial cells and bone marrow-derived macrophages. The hepatic vasculature was visualized with μCT scans and scanning electron microscopy of vascular casts. Serum Ang-2 levels were increased in patients with histological NASH compared to patients with simple steatosis and correlated with hepatic CD34 immunoreactivity as a marker of hepatic angiogenesis. Serum and hepatic Ang-2 levels were similarly increased in mice with steatohepatitis. Both preventive and therapeutic L1-10 treatment reduced hepatocyte ballooning and fibrosis in MCD diet-fed mice, and was associated with reduced hepatic angiogenesis and normalization of the vascular micro-architecture. Liver-isolated endothelial cells and monocytes from MCD-fed L1-10-treated mice showed reduced expression of leukocyte adhesion and inflammatory markers, respectively, compared to cells from untreated MCD diet-fed mice. In the streptozotocin-western diet model, therapeutic Ang-2 inhibition was able to reverse NASH and attenuate HCC progression. In vitro, L1-10 treatment mitigated the increased cytokine production in LPS-stimulated endothelial cells, but not macrophages. Conclusion: Our findings provide evidence for Ang-2 inhibition as a new therapeutic strategy to target pathological angiogenesis in NASH. This article is protected by copyright. All rights reserved.
Article
Over the past 2 decades, nonalcoholic fatty liver disease (NAFLD) has grown from a relatively unknown disease to the most common cause of CLD in the world. In fact, 25% of the world's population is currently thought to have NAFLD. Non‐alcoholic steatohepatitis (NASH) is the subtype of NAFLD that can progress to cirrhosis, hepatocellular carcinoma, and death. NAFLD and NASH are found in not only adults—there is a high prevalence in children and adolescents. Due to NAFLD's close association with type 2 diabetes (T2DM) and obesity, the latest models predict the prevalence of NAFLD and NASH will increase, causing a tremendous clinical and economic burden and poor patient‐reported outcomes. Nonetheless, there is no accurate non‐invasive method to detect NASH and treatment is limited to life style modifications. To examine the state of NAFLD among different regions and understand the global trajectory of this disease, an international group of experts came together during 2017 AASLD Global NAFLD Forum. We provide a summary of this forum and an assessment of the current state of NAFLD and NASH worldwide. This article is protected by copyright. All rights reserved.
Article
The fenestrae of liver sinusoidal endothelial cells (LSEC) allow passive transport of solutes, macromolecules and particulate material between the sinusoidal lumen and the liver parenchymal cells. Until recently, fenestrae and fenestrae-associated structures were mainly investigated using electron microscopy on chemically fixed LSECs. Hence, the knowledge about their dynamic properties remained until to-date largely elusive. Recent progress in atomic force microscopy (AFM) has allowed the study of live cells in three-dimensions (X, Y & Z) over a prolonged time (t) and this at unprecedented speeds and resolving power. Hence, we employed those latest advances in AFM imaging on living LSECs. As a result, we were able to monitor as first the position, size and number of fenestrae and sieve plates using 4-D AFM (X, Y, Z & t) on intact LSEC in vitro. During those time-lapse experimentations, dynamic data were collected on the origin and morpho-functional properties of the filtration apparatus of LSECs. We present herein structural evidence on single laying and grouped fenestrae thereby elucidating their dynamic nature from formation to disappearance. We also collected data on the lifespan of fenestrae. More especially, the formation and closing of entire sieve plates were observed and recorded how the continuous rearrangement of sieve plates affect the structure of fenestrae within them. We observed also the dawn and rise of fenestrae-forming centres and defenestration centres in LSECs under different experimental conditions. Conclusion: No other multimodal biomedical high-resolution imaging technique to-date has allowed collecting new fine structural information on the lifespan, formation, and disappearance of LSEC fenestrae. By doing so, we gathered also evidence of three different pathways implemented in the loss of fenestrae that result in defenestrated LSECs. This article is protected by copyright. All rights reserved.
Article
Background: There are limited data on the risk of hepatocellular cancer (HCC) in patients with non-alcoholic fatty liver disease (NAFLD). We aimed to estimate the risk of incident HCC among patients with NAFLD. Methods: We conducted a retrospective cohort study from a total of 130 facilities in the Veterans Health Administration. Patients with NAFLD diagnosed between 1/1/2004 and 12/31/2008 were included and followed until HCC diagnosis, death or 12/31/2015. We also identified a gender and age-matched control cohort without NAFLD. We ascertained all new HCC cases from the Central Cancer Registry and manual chart reviews. We calculated incidence rates for HCC by NAFLD status as well as in subgroups of NAFLD patients. We used competing risk models to compare the risk of HCC in patients with vs. those without NAFLD. We reviewed electronic medical records of all HCC cases that developed in NAFLD patients without cirrhosis. Results: We compared 296,707 NAFLD patients with 296,707 matched controls. During 2,382,289 person-years [PY] of follow-up, 490 NAFLD patients developed HCC (0.21/1000 PY). HCC incidence was significantly higher among NAFLD patients vs. controls (0.02/1000 PY; hazard ratio, 7.62, 95% confidence interval=5.76-10.09). Among patients with NAFLD, those with cirrhosis had the highest annual incidence of HCC (10.6 /1000 PY). Among patients with NAFLD cirrhosis, HCC risk ranged from 1.6 to 23.7 per 1000 PY based on other demographic characteristics; the risk of HCC was the highest in older Hispanics with cirrhosis. In medical record reviews, 20% of NAFLD patients with HCC had no evidence of cirrhosis. Conclusions: Risk of HCC was higher in NAFLD patients than that observed in general clinical population. Most HCC cases in NAFLD developed in patients with cirrhosis. The absolute risk of HCC was higher than the accepted thresholds for HCC surveillance for most patients with NAFLD cirrhosis.