ArticlePDF AvailableLiterature Review

The non-invasive assessment of hepatic fibrosis

Authors:

Abstract and Figures

Hepatic disease accounts for approximately 2 million deaths/year worldwide. Liver fibrosis, as the last stage of numerous chronic liver diseases, is one of the most relevant prognostic factors. The liver biopsy with the histopathological examination is considered to be the “gold standard” for the identification and staging of the hepatic fibrosis. However, liver biopsy is known as an invasive investigation that has multiple limitations. Research studies conducted in the last few years focused on identifying non-invasive type methods for the evaluation of hepatic fibrosis; usually, there are 2 categories of such investigations: serologic tests and imaging techniques. This narrative review presents the non-invasive investigation methods used in the liver fibrosis evaluation. New molecular perspectives on fibrogenesis and fibrosis regression, as well as the appearance of therapeutic antifibrotic agents, justify the necessity of non-invasive tools to detect and grade liver fibrosis.
Content may be subject to copyright.
Review Article
The non-invasive assessment of hepatic
fibrosis
Gina Gheorghe
a
, Simona Bung
au
b
, Gabriela Ceobanu
c
,
M
ad
alina Ilie
a
, Nicolae Bacalbas
a
d
, Ovidiu Gabriel Bratu
e
,
Cosmin Mihai Vesa
f
, Mihnea-Alexandru G
aman
g,h,
*,
Camelia Cristina Diaconu
c,i
a
Department of Gastroenterology, University of Medicine and Pharmacy “Carol Davila”, 050474
Bucharest, Romania
b
Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea,
Romania
c
Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 014461 Bucharest,
Romania
d
Department of Obstetrics and Gynecology, University of Medicine and Pharmacy “Carol Davila”,
050474 Bucharest, Romania
e
Department of Urology, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest,
Romania
f
Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea,
410028, Oradea, Romania
g
University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania
h
Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni
Clinical Institute, Bucharest 022328, Romania
i
Department of Internal Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474
Bucharest, Romania
Received 18 May 2020; received in revised form 30 June 2020; accepted 10 August 2020
KEYWORDS
Acoustic radiation
force impulse
imaging;
Chronic liver disease;
Hepatic disease accounts for approximately 2 million deaths/year worldwide. Liver fibrosis, as
the last stage of numerous chronic liver diseases, is one of the most relevant prognostic fac-
tors. The liver biopsy with the histopathological examination is considered to be the “gold
standard” for the identification and staging of the hepatic fibrosis. However, liver biopsy is
known as an invasive investigation that has multiple limitations. Research studies conducted
* Corresponding author. Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute,
022328 Bucharest, Romania.
E-mail addresses: gheorghe_gina2000@yahoo.com (G. Gheorghe), simonabungau@gmail.com (S. Bung
au), gabriela.ceobanu@ymail.com
(G. Ceobanu), drmadalina@gmail.com (M. Ilie), nicolae_bacalbasa@yahoo.ro (N. Bacalbas
a), ovi78doc@yahoo.com (O.G. Bratu),
vcosmin2020@gmail.com (C.M. Vesa), mihneagaman@yahoo.com (M.-A. G
aman), drcameliadiaconu@gmail.com (C.C. Diaconu).
https://doi.org/10.1016/j.jfma.2020.08.019
0929-6646/Copyright ª2020, Formosan Medical Association. Published by Elsevier Taiwan LLC. This is an open access article under the CC
BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Available online at www.sciencedirect.com
ScienceDirect
journal homepage: www.jfma-online.com
Journal of the Formosan Medical Association (2021) 120, 794e803
Elastography;
FibroTest;
HepaScore
in the last few years focused on identifying non-invasive type methods for the evaluation of
hepatic fibrosis; usually, there are 2 categories of such investigations: serologic tests and im-
aging techniques. This narrative review presents the non-invasive investigation methods used
in the liver fibrosis evaluation. New molecular perspectives on fibrogenesis and fibrosis regres-
sion, as well as the appearance of therapeutic antifibrotic agents, justify the necessity of non-
invasive tools to detect and grade liver fibrosis.
Copyright ª2020, Formosan Medical Association. Published by Elsevier Taiwan LLC. This is an
open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
Introduction
Chronic liver disease is one of the main causes of morbidity/
mortality and it accounts for 2 million deaths worldwide each
year.
1
The most prevalent etiologies of chronic liver disease
consist of infections with hepatitis B/C viruses, alcohol-
related liver disease, and non-alcoholic fatty liver disease
(NAFLD).
1e3
Liver fibrosis is a response induced by chronic
liver injury, as it is in the viral hepatitis case, alcoholic liver
disease, autoimmune hepatitis, NAFLD, non-alcoholic stea-
tohepatitis (NASH) and cholestatic liver diseases.
4,5
Hepatic
fibrosis represents a serious health problem, as it may induce
and lead to advanced liver cirrhosis and hepatocellular car-
cinoma.
6
There is a strong relationship between liver fibrosis
and hepatocellular carcinoma (HCC). On one hand, the
presence of advanced liver fibrosis in patients with HCC may
increase the risk of complications of antineoplastic therapy
or even death (due to liver failure); on the other hand, he-
patic fibrosis is a risk factor for the recurrence of hepato-
cellular carcinoma after curative therapy.
7
Therefore,
detecting and staging liver fibrosis is essential in guiding the
management of patients diagnosed with HCC. A timely and
correct diagnosis of liver fibrosis is of paramount importance,
as current data show that it can be delayed if it is diagnosed in
an incipient stage.
6,7
The actual methods of diagnosis of liver
fibrosis consist of non-invasive tests which include serolog-
ical and imaging investigations, and liver biopsy, an invasive
method.
Pathophysiology of liver fibrosis
As already stated, liver fibrosis is considered as being the final
stage of hepatic chronic diseases.
8
The major factors involved
in fibrogenesis areinflammation and hepatocellular necrosis.
Massive necrosis and stromal collapse lead to the formation of
fibrous bands and nodules of regenerating hepatocytes,which
are usually large and irregular.
9
The development of regen-
erative nodules is a compensation mechanism aimed to
replace destroyed or non-functional hepatocytes.
9
Hepatic
fibrogenesis is based on the excessive synthesis of collagen
fibres. Under normal conditions, the synthesis of the extra-
cellular matrix is provided by hepatocytes, endothelial cells
and lipid-storing cells. In pathological circumstances, there is
a rise in the number of cells involved in collagen production,
an increase in the quantity of collagen produced by each cell
and also a change in extracellular matrix constituents, with
increased formation of mature type I collagen, laminin,
fibronectin and glycoproteins.
9
Of great importance in the development and progression
of hepatic fibrosis are the fibroblasts and Ito cells, also
named stellate cells, that change their phenotypic
expression and differentiate into myofibroblasts, which
have a main function in the collagenisation process of the
Disse space.
9
Hepatic fibrogenesis is conditioned, on one
hand, by the imbalance between cytokines that stimulate
hepatocyte regeneration (TGF-aetransforming growth
factor-a, EGF eepidermal growth factor, HGF ehepato-
cyte growth factor) and cytokines that inhibit regeneration
and stimulate fibrogenesis (TGF-beta1 etransforming
growth factor-beta1, HBGF eheparin-binding growth fac-
tor) and, on the other hand, by the disproportion between
the tissue inhibitors of metalloproteinases (TIMP) and the
matrix metalloproteinases (MMP).
9
In consequence, there will be a reduction in the activity
of MMP, which has anti-fibrotic and resorptive properties
and an intensification in the activity of TIMP.
9
The catabolic
of collagen fibres and the reversibility of fibrosis may be
possible when the levels of collagen are low, collagen fibres
have developed recently or when there is an increase of
MMP associated with a reduction of TIMP.
9,10
Advanced
fibrosis and formation of regenerating nodules are however
irreversible processes.
9,10
A schematic view of the patho-
physiology of liver fibrosis is presented in Fig. 1.
Assessment of liver fibrosis
Liver biopsy and histopathological examination is confirmed
as the gold standard in the case of the hepatic fibrosis eval-
uation and grading. The first percutaneous liver biopsy was
performed in 1883, in Germany, by Paul Ehrlich.
10
Over the
years, the evolution of liver puncture was obvious; nowadays,
there are many alternative procedures used in obtaining
samples of liver tissue: unguided biopsy, ultrasound-guided
biopsy, CT-guided biopsy, trans-jugular liver biopsy, laparo-
scopic biopsy or via laparotomy.
11
Despite significant prog-
ress, as clinical and experimental studies show, the liver
biopsy remains an invasive procedure that can be associated
with serious complications
12
such as: pain,
13
intraperitoneal
haemorrhage, intrahepatic and/or subcapsular haemor-
rhage, haemobilia, biliary peritonitis,abscesses, bacteremia,
sepsis,
14e16
pneumothorax, hemothorax, pleurisy, arterio-
venous fistulae, subcutaneous emphysema, side effects of
anesthesia, injury to other organs (lung, gall bladder, kidney,
colon),
17e19
or even death.
12,20
The contraindications of the liver biopsy are divided into
absolute contraindications and relative contraindications.
The absolute contraindications are as follows: significant
coagulopathy or thrombocytopenia; nonsteroidal anti-
inflammatory drugs use within last 7e10 days; patient’
impossibility to provide blood transfusion support or
Non-invasive assessment of hepatic fibrosis 795
decline blood transfusion; patient’ inability to cooperate
with the procedure; extrahepatic biliary obstruction;
vascular tumor/echinococcal cyst; suspected hemangioma;
incapacity to find an adequate biopsy site by ultrasound
and/or percussion. The relative contraindications of the
liver biopsy are hemophilia, ascites, morbid obesity,
amyloidosis, infection within the right pleural cavity or
infection below the right hemidiaphragm.
21
Furthermore,
because it evaluates only small segments of the liver and
not the whole hepatic structure, there can be
falseenegative errors. These limitations of liver biopsy led
to the emergence and development of some non-invasive
tools, very useful in liver fibrosis evaluation (e.g. serolog-
ical tests and imaging techniques).
22
Non-invasive tests aim to identify and stratify the level of
hepatic fibrosis by referencing certain histological scores,
such as the METAVIR score. This particular score was spe-
cifically coined for patients infected with hepatitis C virus
(HCV) and it evaluates the fibrosis level on a 5 points scale
23
(F0 eno fibrosis, F1 eportal fibrosis without septa, F2 e
few septa, F3 enumerous septa without cirrhosis, and F4 e
cirrhosis). The level of fibrosis is significant if the METAVIR
score F2.
23
Non-invasive tests can also be utilized to
monitor patients who are undergoing treatment with drugs
that induce hepatic lesions, such as methotrexate.
Fig. 2 shows the methods, generally available and
emerging, for evaluating liver fibrosis.
Serological tests
To evaluate the level of hepatic fibrosis, a variety of sero-
logical tests has been identified. Panels that utilize sero-
logical markers have been designed to increase the
predictive capacity.
These non-invasive tests require validation against liver
biopsy, which is the present gold standard for the diagnosis;
their diagnostic accuracy is evaluated using the area under
the receiver operator characteristic (AUROC), which com-
bines the sensitivity and specificity of the liver fibrosis
marker.
24
The most utilized panels at the moment are as fol-
lows
22,25
: aminotransferase-to-platelet ratio index (APRI),
FibroTest/FibroSure, Hepascore, and FibroSpect. Other
panels of serological tests which can be used in the evalu-
ation of liver fibrosis are: ActiTest, FIB-4 index, NAFLD
fibrosis score, PGA index, Fibro Index, Forns index, Fibro-
meter, BARD score, ELF, ALBI grade, Lok index, GUCI,
Bonacini-index (CDS), King’s score, Pohl index, VITRO score,
Zeng index.
26
These serological tests can differentiate be-
tween patients with or without significant fibrosis. The
main disadvantage is that they cannot accurately differ-
entiate between the particular levels of fibrosis.
27,28
So far,
none of the panels may be considered the standard
Figure 1 Schematic view of the pathophysiology of liver fibrosis.
Figure 2 Methods, generally available and emerging, for
evaluating liver fibrosis. Color figures are clearly marked as
being intended for: (II) color reproduction on the Web (free of
charge) and in grayscale in print (free of charge). Grayscale
versions of the figures are also supplied for printing purposes.
796 G. Gheorghe et al.
evaluation of hepatic fibrosis. Their usage depends on local
availability.
Serological tests may be classified into two
categories
22,27,28
:
Indirect tests reflect the changes in the hepatic func-
tion, but they do not allow the directly reflection of the
extracellular matrix metabolism (Table 1).
Direct tests reflect directly the extracellular matrix
metabolism. These fall under 3 subcategories: markers
associated with matrix deposition, markers associated
with matrix degradation and cytokines and chemokines
associated with fibrogenesis (Table 2).
Both quantitative and qualitative modifications of the
extracellular matrix pertain to hepatic fibrosis. Potential
biomarkers for the identification and stratification of he-
patic fibrosis include as follows: synthesis/degradation
products of collagen; enzymes which are involved in
biosynthesis/degradation matrix; proteoglycans/glycos-
aminoglycans; extracellular matrix glycoproteins.
22,28
He-
patic fibrosis evaluation markers were combined in
different panels to increase the predictive capacity of
these histopathological modifications.
19
APRI is a hepatic fibrosis evaluation score that was pri-
marily studied in HCV infected patients, human immuno-
deficiency virus (HIV) and HCV coinfected patients, and
patients with alcoholic liver disease/NAFLD.
29e33
According
to data from the literature, APRI sensitivity in detecting
hepatic fibrosis is 77%, while its specificity is 72%.
22
APRI
score is calculated using the following formula:
APRI Z[(ASTAST
ULN
)/platelet count]100, where AST
ULN
is the superior limit of the normal value of AST.
34
A meta-analysis that included 40 studies and 8739 pa-
tients with chronic hepatitis C showed that APRI had an
AUROC of 0.77, 0.80 and 0.83 for the diagnosis of significant
fibrosis (F2), severe fibrosis (F3) and cirrhosis, respec-
tively.
34
Another meta-analysis including 22 studies showed
similar results, with an AUROC of 0.76 for significant fibrosis
and 0.82 for cirrhosis.
35
FibroTest and FibroSure represent the same test which
has been commercialized under many names in the USA and
Europe. These tests require the age and sex of the patient,
but also values such as a-2-macroglobulin, a-2-globulin, g-
globulin, apolipoprotein A1, GGT, and total bilirubin.
36
ActiTest is a variant of FibroTest which further utilizes
ALT, thus reflecting hepatic fibrosis and necro-inflammatory
activity.
37
FibroTest sensitivity in detecting hepatic fibrosis
is 60e75%, while its specificity is 80e90%.
38
A meta-analysis
of 8 studies showed that FibroTest had a median AUROC of
0.84 for the diagnosis of advanced fibrosis and concluded
that FibroTest is an effective alternative to biopsy in pa-
tients diagnosed with chronic hepatitis C and B, ALD and
NAFLD.
39
HepaScore utilizes values such as bilirubin, g-glutamyl
transferase (GGT), hyaluronic acid, a-2-macroglobulin, sex
and age. This score has been tested in patients with alco-
holic liver disease, though it was not more accurate than
FibroTest.
40
It has proven to be accurate in patients with
HCV infection.
41
The AUROC for HepaScore was 0.85 for
significant fibrosis, 0.96 for advanced fibrosis and 0.94 for
cirrhosis.
42
FibroSpect II uses values of serum hyaluronic acid, a-2-
macroglobulin and tissue inhibitor of metalloproteinase-1
(TIMP-1). This score has proven useful in differentiating
Table 1 Indirect markers used in the liver fibrosis
evaluation.
Individual markers Serological panels
Serum aminotransferase levels
Platelet count coagulation parameters
GGT
Total bilirubin
A-2-macroglobulin
A-2-globulin (haptoglobin)
Proteomics and glycemic
APRI
FibroTest/FibroSure
ActiTest
Hepascore
FIB-4 index
NAFLD fibrosis score
PGA index
Fibro Index
Forns index
Fibro-meter
BARD score
Fibrospect II
ELF
ALBI grade
Lok index
GUCI
Bonacini-index
(CDS)
King’s score
Pohl index
VITRO score
Zeng index
GGT egamma-glutamyl transferase; APRI eaminotransferase-
to-platelet ratio index; FIB-4 efibrosis-4 index; NAFLD fibrosis
enon-alcoholic fatty liver disease; PGA eprothrombin time,
GGT, apolipoprotein A1; BARD eBMI, AST/ALT ratio, diabetes
mellitus; GUCI eGo
¨teborg University Cirrhosis Index.
Table 2 Direct markers used in the liver fibrosis
evaluation.
Markers
associated with
matrix
deposition
Markers
associated with
matrix
degradation
Cytokine and
chemokines associated
with hepatic fibrosis
PICP
PIIINP
Type I and IV
collagen
Laminin
Hyaluronic acid
YKL-40 (CHI3L1)
MMPs
TIMP-1 and TIMP-
2
TGF-a
TGF-b
PDGF
PICP eprocollagen type I carboxy-terminal peptide; PIIINP e
procollagen type III amino-terminal peptide; YKL-40 (inflam-
matory glycoprotein), also known as CHI3L1 echitinase-3-like
protein 1; MMP ematrix metalloproteinases; TIMP etissue in-
hibitors of metalloproteinases; TGF etransforming growth
factor; PDGF eplatelet-derived growth factor.
Non-invasive assessment of hepatic fibrosis 797
chronic HCV patients with moderate/severe fibrosis from
those with mild/no fibrosis.
43
The European Liver Fibrosis panel (ELF) utilizes hyal-
uronic acid level, amino-terminal pro-peptide of type III
collagen level and TIMP-1. The specificity of this score can
reach 95%.
44
Table 3 presents panels of serological tests
(and their components) utilized in the hepatic fibrosis
evaluation.
22
Regarding the correlation between liver fibrosis and HCC,
data from the literature suggest that serological tests cannot
determine the exact extent of liver fibrosis and therefore
cannot accurately stage it. However, a study of 464 patients
with surgically-treated HCC showed that the serological tests
which correlated best with the severity of liver fibrosis were
CDS and Lok’s index in hepatitis B virus-associated hepato-
cellular carcinoma, and FIB-4 and Lok’s index in hepatitis C
virus-associated HCC.
7
The best predictor for liver cirrhosis
was CDS.
7
The independent indicators of advanced liver
fibrosis included: multiple tumours, hepatitis C, low platelet
count and prolonged prothrombin time.
7
Imaging tests
Imaging tests are non-invasive investigation tools used to
detect and stage liver fibrosis.
45,46
They include conven-
tional investigations eultrasound (US), computed tomog-
raphy (CT), magnetic resonance imaging (MRI), as well as
newer techniques of imaging (ultrasound elastography and
magnetic resonance elastography).
22
Morphologic assessment of liver fibrosis can be per-
formed using conventional US, CT and MRI. However, these
investigations are characterized by unreliability and low
sensitivity, as the morphologic features they describe are
usually absent in the early stages of fibrosis.
47
Imaging signs
of liver fibrosis include morphological changes of the liver
(nodular hepatic surface, caudal lobe hypertrophy, het-
erogeneous parenchyma, changes in liver size), increase in
diameter and tortuosity of the hepatic artery, reduction of
hepatic vein diameter, and dilated venous system, as well
as the dilation of portal, splenic and superior mesenteric
veins, and spleen modification (splenomegaly).
48
The
development of gastro-esophageal, para-esophageal, left
and short gastric, umbilical and abdominal wall varices,
splenorenal and retroperitoneal shunts are also signing of
advanced hepatic disease. Because these are imaging
changes seen merely in end-stage liver fibrosis, conven-
tional investigations demonstrate their inaccuracy for
staging liver fibrosis over its entire spectrum of severity.
49
Another limitation of conventional imaging is due to the
fact that many morphologic features are subjective, lead-
ing to differences of opinion between observers as found
within studies.
50
An emerging imaging technique is positron emission to-
mography (PET), the most commonly used radiopharma-
ceutical for PET examination being F-fluoro-2-deoxy-D-
glucose (F-FDG). Having a superior spatial resolution, F-FDG
PET/CT can assess liver fibrosis by quantification of hepatic
glucose metabolism, as concluded by a study published by
Verloh et al.
51
Elastography is a newer technology. There are two types
of ultrasound elastography: shear wave elastography (SWE)
and strain elastography (SE), also named real time elastog-
raphy (Hi-RTE). SWE provides a quantitative measure of
stiffness by using acoustic/mechanical pulses induced by the
ultrasound machine.
22
Among SWE methods: ultrasound e
transient elastography (TE), acoustic radiation force impulse
imaging (ARFI), two-dimensional (2D) shear wave elastog-
raphy (SWE). The first SWE system was Transient elastog-
raphy (Fibroscan) and it is considered the most widely SWE-
based technique in the non-invasive liver fibrosis diagnosis.
49
However, TE does not offer morphologic imaging guidance,
as the ultrasound detector is one-dimensional (1D).
Compared to transient elastography, both ARFI and 2D-SWE
Table 3 Panels of indirect serologic tests used in the
hepatic fibrosis evaluation.
Serological
test
Components
APRI AST, platelet count
FibroTest/
FibroSure
a-2-macroglobulin, a-2-globulin
(haptoglobin), g-globulin, apolipoprotein
A1, GGT, total bilirubin
ActiTest ALT, a-2-macroglobulin, a-2-globulin
(haptoglobin), gglobulin, apolipoprotein
A1, GGT, total bilirubin
HepaScore Bilirubin, GGT, hyaluronic acid, a-2-
macroglobulin, age, gender
FIB-4 index Platelet count, ALT, AST, age
NAFLD fibrosis
score
BMI, blood glucose levels, aminotransferase
levels, platelet count, albumin, age
PGA index Prothrombin index, GGT, apolipoprotein A1
FibroIndex Platelet count, AST, gglobulin
FornsIndex GGT, cholesterol, platelet count, age
Fibrometer Platelet count, prothrombin index, AST, a-2-
macroglobulin, hyaluronic acid, blood urea
nitrogen, age
BARD score BMI, AST/ALT ratio, DM presence
FibroSpect II Serum hyaluronic acid, tissue inhibitor of
metalloproteinase-1 (TIMP-1) and a-2-
macroglobulin
ELF Hyaluronic acid level, amino-terminal pro-
peptide of type III collagen level, and TIMP-1
ALBI grade Bilirubin, albumin
Lok index Platelet count, AST, ALT, INR
GUCI Platelet count, AST, prothrombin index
Bonacini-
index (CDS)
ALT/AST ratio, INR, platelet count
King’s score Age, platelet count, AST, INR
Pohl index Platelet count, AST, ALT
VITRO score Platelet count, von Willebrand factor
antigen
Zeng index Age, a-2-Macroglobulin, GGT, and
hyaluronic acid levels
AST easpartate aminotransferase; GGT egamma-glutamyl
transferase; ALT ealanine aminotransferase; BMI ebody mass
index; TIMP-1 etissue inhibitor of metalloproteinase-1; APRI e
aminotransferase-to-platelet ratio index; FIB-4 efibrosis-4;
NAFLD enon-alcoholic fatty liver disease; PGA eprothrombin
time, GGT, apolipoprotein A1; BARD eBMI, AST/ALT ratio,
diabetes mellitus; DM ediabetes mellitus; ELF eEuropean Liver
Fibrosis panel; CDS ecirrhosis discriminant score, GUCI e
Go
¨teborg University Cirrhosis Index.
798 G. Gheorghe et al.
are imaging techniques incorporated in US scanners. ARFI
method can be done in real-time imaging, so that the larger
blood vessels and masses can be distinguished and avoided.
45
2D-SWE technique is currently the newest SWE-based
method; its concrete role in monitoring and staging liver
fibrosis needs to be demonstrated through further studies.
48
Both ARFI and 2D-SWE are technologic advancements over
TE, with lower failure rates; nevertheless, a real benefit in
the diagnostic accuracy of one technique over the others for
liver fibrosis staging has not been clearly established yet.
Comparison between different types of shear wave elas-
tography is depicted in Table 4 .
50e60
Hi-RTE is technically different from SWE methods; in this
case, the evaluation of the tissue stiffness is obtained after
manual compression. In a study conducted by Tatsumi, 119
patients with chronic liver disease underwent Hi-RTE; the
results were compared with TE and serum markers. Hi-RTE
showed a negative correlation with stages of fibrosis and TE
findings, thus suggesting RTE’s superior performance
compared to TE.
61
Another study, conducted by Colombo
et al., evaluated 45 patients with chronic liver disease and
27 normal subjects and compared three elastographic
methods: TE, ARFI, and Hi-RTE. The AUROCs for predicting
significant fibrosis (F2) for TE, RTE, and ARFI were 0.89,
0.75, and 0.81, respectively. In this study, TE was superior to
RTE, with no significant difference between TE and ARFI nor
between ARFI and RTE. The AUROC values for predicting
liver cirrhosis (FZ4) for TE, RTE, and ARFI showed similar
values (0.92, 0.85, and 0.93, respectively).
62
MRE (magnetic resonance elastography) is a non-invasive
procedure used to measure the liver viscoelastic proper-
ties.
63
As a consequence of hepatic fibrosis, liver stiffness
increases, and it can be evaluated using the propagation of
mechanical waves measurement. MRE has proven its supe-
riority to transient elastography through the capacity to
scan the entire organ and also because it can be performed
in patients with ascites or obesity. Multiple studies have
shown that the diagnostic performance of MRE is higher to
that of ARFI and TE, one major characteristic being its
possibility to diagnose accurately mild fibrosis.
64,65
In a
meta-analysis which included 12 studies, MRE was proven to
have a high accuracy for the diagnosis of significant or
advanced fibrosis and cirrhosis, independent of the BMI and
the etiology of chronic liver disease.
63
Because MRE as-
sesses the whole liver, sampling errors are limited and
interobserver variability is reduced. Also, MRE allows the
regional distribution characterization of the hepatic
fibrosis, a fact that may be useful in underlying and diag-
nosis of liver disease (e.g. primary sclerosing cholangitis).
There are some limitations of MRE, such as high cost,
restricted availability, long time of examination and its
reliance on patient’ cooperation for breath-holds.
63e65
Sequential algorithms
Evidence provided by a clinical study conducted on 183
subjects suffering from chronic hepatitis C has shown that
Table 4 Comparison of the different types of shear wave elastography.
46e55
Transient elastography (TE) Acoustic radiation force impulse
imaging (ARFI)
2D shear wave elastography (2D-
SWE)
Mechanism Uses shear wave imaging to estimate
liver stiffness
Uses conventional hepatic
Ultrasonography to assess liver
stiffness
- Combines ultrasound images with
radiation force induced into the
liver
- Measure shear waves propagation
in real time
Advantages - Accurately diagnoses cirrhosis
(fibrosis stage 4)
- Distinguishes advanced fibrosis from
minimal or no fibrosis
- TE can be used by physicians at the
bedside
- Inexpensive
- Portable
- Short procedure time (<5 min)
- Immediate results
- Reproducible
- Accurately diagnoses early liver
fibrosis in chronic liver disease
patients
- It is performed in real-time
imaging
- Ability to select the area to be
assessed, avoiding large vessels or
ribs
- Can be applied in patients with
obesity and/or ascites
- High accuracy and precision
- Low failure rate
- Good applicability
- Adjustable location of interest
depending on the operator
- Failure rate is significantly lower
than that of TE
Disadvantages - The exact measurement locations are
not recorded
- Cannot evaluate liver parenchyma for
hepatic disease or masses
- Less reliable in patients with obesity,
narrow intercostal spaces, and/or
ascites
- High technical failure rate (6e23%)
- Requirement for patient fasting
- Requirement for patient fasting
- Relatively high expense for
deploying at multiple sites
- More expensive
- Less available and validated by
current studies compared with TE
- Requires more expertise to
perform
- More expensive
- Requires more operator expertise
- Restricted availability
- Insufficient evidence concerning
the diagnostic performance
Non-invasive assessment of hepatic fibrosis 799
an association of TE and biochemical tests offered favour-
able diagnostic results in identifying advanced fibrosis and
clinically significant fibrosis.
66
In the case of concordance
between FibroScan and FibroTest, the obtained results
revealed a favourable ratio of liver biopsy for significant
fibrosis (F2) in 84% of cases, an excellent ratio for severe
fibrosis (F3) in 95% of cases and for liver cirrhosis in 94% of
cases.
66
Based on the findings of a clinical research carried
out on 235 patients suffering from chronic hepatitis C, was
designed the Fibropaca algorithm that included APRI,
FibroTest and the Forns Index.
67
The association of the
three tests allowed the proper classification for 81.3% of
the subjects included in the research.
67
A set of three algorithms, Sequential algorithms for
fibrosis evaluation (SAFE) biopsy, comprising Fibrotest and
APRI, was designed by Sebastiani et al.
68
The algorithms
were applied in assessing the F2 stage for subjects with
normal or high values of liver transaminases as well as for
diagnosing F4. Using SAFE biopsy influenced the decrease in
the necessity for liver biopsy with 50%.
68
A captivating al-
gorithm in examining great numbers of people is a serum
non-invasive assay as the first examination test, succeeded
by shear wave elastography.
69
Combining transient elas-
tography and blood fibrosis assays offers the potential for a
precise diagnostic of advanced liver fibrosis. A new algo-
rithm was designed
70
comprising an imaging procedure
FibroMeterVCTE and a serum assay, easy liver fibrosis test
(eLIFT). The eLIFT is a score that integrates age, gender,
AST, prothrombin time, gamma-glutamyl transferase and
platelets, being designed to identify advanced cirrhosis.
The FibroMeterVCTE represents a procedure of vibration
controlled transient elastography (VCTE). In the research,
eLIFT was carried out first, succeeded by FibroMeterVCTE if
eLIFT did not identify advanced fibrosis. The association of
FibroMeterVCTE and eLIFT presented a sensitivity of 76.1%
in identifying advanced fibrosis and for identifying cirrhosis
a sensitivity of 92.1%.
70
Emerging technologies egenetic and
microbiome signature scores
Genetic differences among people determine distinctive
predisposition for the occurrence of liver fibrosis, ac-
counting approximately half of the phenotypic variation in
a chronic liver disease like NAFLD.
69
High risk of fibrosis or
steatosis is connected with particular gene polymorphisms.
A research performed on 515 subjects suffering from NAFLD
revealed an association of steatosis with variations of
TM6SF2 and PNPLA3 genes.
70
The TM6SF2, PNPLA3 and
MBOAT7 gene variations can be associated with hepatocyte
lesions as demonstrated by the research findings.
70,71
The
MBOAT7 polymorphism was correlated to liver fibrosis while
TM6SF2 variant was correlated to fat accumulation. High
risk of steatosis and fibrosis was connected with the PNPLA3
polymorphism. The discovery of a genetic signature
comprising 7 predictive (single nucleotide polymorphisms)
SNPs able to detect Caucasian subjects suffering from
chronic hepatitis C presenting high risk of cirrhosis deter-
mined the establishing of a cirrhosis risk score using genetic
variants.
72
Other developing procedures comprise the
usage of microRNAs, fragments of RNA that adjust gene
expression; miRNA122 being correlated in NAFLD subjects
with liver fibrosis.
69
Imaging techniques are evolving, of
them three-dimensional (3D) MRE has the potential to
determine shear wave propagation in several plans, thus
keeping away from mathematical suppositions character-
istic to 3D methods.
69,70
It was noticed that particular gut flora was connected to
an increased risk of liver cirrhosis.
73
The risk of developing
obesity, NAFLD and gastrointestinal malignancies is corre-
lated with the presence of a dysbiotic microbiome.
74e78
Moreover, particular gut bacteria may transform choline
in trimethylamine, accentuating the risk of evolution to
non-alcoholic steatohepatitis (NASH).
74
A team of re-
searchers set a microbiome or gut bacteria signature score
comprising 37 bacterial species that had promising poten-
tial in differentiating mild/moderate NAFLD from severe
NAFLD.
74
Conclusions
Non-invasive liver fibrosis evaluation is an important topic
of recent research. Since needle liver biopsy is usually
associated with significant risks/limitations, different
scores (based on various biochemical markers or hepatic
tissue stiffness) were elaborated. Non-invasive serologic
tests are classified into direct tests (which, as the name
implies, directly evaluate the metabolism of the extracel-
lular matrix) and indirect tests that reflect the changes in
liver function, caused by liver fibrosis. With a sensitivity
and specificity that can reach up to 95%, serological tests
may be useful non-invasive methods in detecting liver
fibrosis.
22
As well, non-invasive imaging tests include con-
ventional morphologic investigations (US, CT or MRI) and
newer imaging techniques (US/MRI elastography). Both
these non-invasive types of tests have the advantage of
being more accessible and avoiding complications that may
occur after liver biopsy. They are also recommended to be
performed in patients for whom liver biopsy is contra-
indicated. Sequential algorithms may be efficient for
detecting advanced fibrosis, using a combination of elas-
tography methods with direct or indirect serum tests. New
methods related to genetic signature scores appear, based
on combinations of microbiome signature scores or SNPs,
characteristic for dysbiotic gut flora, having right differ-
entiating capacity in determining/anticipating liver
fibrosis.
Author contributions
Conceptualization: Gina Gheorghe and Gabriela Ceobanu.
Data curation: M
ad
alina Ilie. Formal Analysis: Cosmin Mihai
Vesa. Investigation: Gabriela Ceobanu, M
ad
alina Ilie, Nic-
olae Bacalbas
a and Camelia Cristina Diaconu. Methodology:
Simona Bung
au and Ovidiu Gabriel Bratu. Software: Cosmin
Mihai Vesa. Supervision: Simona Bung
au and Camelia Cris-
tina Diaconu. Validation: Gina Gheorghe and Camelia Cris-
tina Diaconu. Visualization: Ovidiu Gabriel Bratu and
Cosmin Mihai Vesa. Writingdoriginal draft: Gina Gheorghe,
Simona Bung
au and Cosmin Mihai Vesa. Writingdreview
and editing: Simona Bung
au, Mihnea-Alexandru G
aman
and Camelia Cristina Diaconu. All authors have read and
800 G. Gheorghe et al.
agreed to the published version of the manuscript. Gina
Gheorghe, Camelia Cristina Diaconu, Simona Bung
au, Nic-
olae Bacalbas
a and Cosmin Mihai Vesa contributed equally
to the writing of this paper and share first authorship. All
authors approved the submission of the final version of the
manuscript.
Declaration of competing interest
The authors have no conflicts of interest relevant to this
article.
References
1. Moon AM, Singal AG, Tapper EB. Contemporary epidemiology of
chronic liver disease and cirrhosis. Clin Gastroenterol Hepatol
2019;S1542e3565:30849e63. https:
//doi.org/10.1016/j.cgh.2019.07.060. PMID: 31401364.
2. Zaha DC, Vesa C, Uivarosan D, Bratu O, Fratila O, Tit DM, et al.
Influence of inflammation and adipocyte biochemical markers
on the components of metabolic syndrome. Exp Ther Med
2020;20:121e8. https://doi.org/10.3892/etm.2020.8663.
PMID: 32509004.
3. Pennisi G, Celsa C, Spatola F, Dallio M, Federico A, Petta S.
Pharmacological therapy of non-alcoholic fatty liver disease:
what drugs are available now and future perspectives. Int J
Environ Res Public Health 2019;16:E4334. https://doi.or-
g/10.3390/ijerph16224334. PMID: 31703268.
4. Kup
cova
´V, Fedele
sova
´M, Bulas J, Kozmonova
´P, Turecky
´L.
Overview of the pathogenesis, genetic, and non-invasive clin-
ical, biochemical, and scoring methods in the assessment of
NAFLD. Int J Environ Res Public Health 2019;16:E3570. https:
//doi.org/10.3390/ijerph16193570. PMID: 31554274.
5. Greenaway C, Makarenko I, Abou Chakra CN, Alabdulkarim B,
Christensen R, Palayew A, et al. The effectiveness and cost-
effectiveness of hepatitis C screening for migrants in the
EU/EEA: a systematic review. Int J Environ Res Public Health
2018;15:E2013. https://doi.org/10.3390/ijerph15092013.
PMID: 30223539.
6. Aydın MM, Akc¸alı KC. Liver fibrosis. Turk J Gastroenterol 2018;
29:14e21. https://doi.org/10.5152/tjg.2018.17330. PMID:
29391303.
7. Ho SY, Liu PO, Hsu CY, Hsia CY, Su CW, He YJ, et al. Current
noninvasive liver reserve models do not predict histological
fibrosis severity in hepatocellular carcinoma. Sci Rep 2018;8:
15074. https://doi.org/10.1038/s41598-018-33536-2. PMID:
30305679.
8. Toosi AE. Liver fibrosis: causes and methods of assessment. A
review. Rom J Intern Med 2015;53:304e14. https:
//doi.org/10.1515/rjim-2015-0039. PMID: 26939206.
9. Henderson NC, Iredale JP. Liver fibrosis: cellular mechanisms
of progression and resolution. Clin Sci (Lond) 2007;112:
265e80. https://doi.org/10.1042/CS20060242. PMID:
17261089.
10. Rockey DC, Friedman SL. Hepatic fibrosis and cirrhosis. Zakim
and Boyer’s hepatology ea textbook of liver disease. 5th ed.
Saunders Elsevier; 2006. p. 87e109.
11. Bravo A, Sheth SG, Chopra S. Approach to liver biopsy. UpTo-
Date. 2020. https://www.uptodate.com/contents/approach-
to-liver-biopsy?. [Accessed 3 April 2020].
12. Abdel-Daim MM, El-Tawil OS, Bungau SG, Atanasov AG. Appli-
cations of antioxidants in metabolic disorders and degenera-
tive diseases: mechanistic approach. Oxid Med Cell Longev
2019;2019:4179676. https://doi.org/10.1155/2019/4179676.
PMID: 31467632.
13. Popa AR, Bungau S, Vesa CM, Bondar AC, Pantis C, Maghiar O,
et al. Evaluating the efficacy of the treatment with benfoti-
amine and a-lipoic acid in distal symmetric painful diabetic
polyneuropathy. Rev Chim 2019;70:3108e14. https:
//doi.org/10.37358/RC.19.9.7498.
14. Cao B, Wu J, Xu C, Chen Y, Xie Q, Ouyang L, et al. The accu-
mulation and metabolism characteristics of rare earth ele-
ments in SpragueeDawley rats. Int J Environ Res Public Health
2020;17:E1399. https://doi.org/10.3390/ijerph17041399.
PMID: 32098119.
15. Dallio M, Diano N, Masarone M, Gravina AG, Patane
`V,
Romeo M, et al. Chemical effect of bisphenol A on non-
alcoholic fatty liver disease. Int J Environ Res Public Health
2019;16:E3134. https://doi.org/10.3390/ijerph16173134.
PMID: 31466361.
16. Elhelaly AE, Albasher G, Alfarrajah S, Almeer R, Bahbah EI,
Fouda MMA, et al. Protective effects of hesperidin and diosmin
against acrylamide-induced liver, kidney and brain oxidative
damage in rats. Environ Sci Pollut Res 2019;26:35151e62.
https://doi.org/10.1007/s11356-019-06660-3. PMID:31686333.
17. Lin MH, Chiu SY, Chang PH, Lai YL, Chen PC, Ho WC. Hyper-
lipidemia and statins use for the risk of new diagnosed sarco-
penia in patients with chronic kidney: a population-based
study. Int J Environ Res Public Health 2020;17:E1494. https:
//doi.org/10.3390/ijerph17051494. PMID: 32110901.
18. Spinu AD, Bratu OG, Marcu DR, Stanciu AE, Gherghiceanu F,
Ionita-Radu F, et al. Underactive bladder ean underestimated
entity. J Mind Med Sci 2020;7:23e8. https:
//doi.org/10.22543/7674.71.P2328.
19. Moisi MI, Rus M, Bungau S, Zaha CD, Uivarosan D, Fratila O,
et al. Acute coronary syndromes in chronic kidney disease:
clinical and therapeutic characteristics. Medicina (Kaunas)
2020;56:E118. https://doi.org/10.3390/medicina56030118.
PMID: 32182690.
20. Rosato V, Masarone M, Dallio M, Federico A, Aglitti A,
Persico M. NAFLD and extra-hepatic comorbidities: current
evidence on a multi-organ metabolic syndrome. Int J Environ
Res Public Health 2019;16:E3415. https://doi.org/10.3390/i-
jerph16183415. PMID: 31540048.
21. Gherlan G, Calistru P. Evaluarea fibrozei hepatice emetode
invazive sau neinvazive. Rom J Infect Dis 2007;10:130e4.
22. Curry MP, Afdhal NH. Noninvasive assessment of hepatic
fibrosis: overview of serologic and radiographic tests.UpTo-
Date. 2020. https://www.uptodate.com/contents/noninvasive-
assessment-of-hepatic-fibrosis-overview-of-serologic-and-
radiographic-tests?. [Accessed 27 March 2020].
23. Bedossa P, Poynard T. An algorithm for the grading of activity in
chronic hepatitis C. The METAVIR Cooperative Study Group.
Hepatology 1996;24:289e93. https://doi.or-
g/10.1002/hep.510240201. PMID: 8690394.
24. Poynard T, Halfon P, Castera L, Munteanu M, Imbert-Bismut F,
Ratziu V, et al. Standardization of ROC curve areas for diag-
nostic evaluation of liver fibrosis markers based on preva-
lences of fibrosis stages. Clin Chem 2007;53(9):1615e22.
https://doi.org/10.1373/clinchem.2007.085795.PMID:
17634213.
25. Jang HJ, Min JH, Lee JE, Shin KS, Kim KH, Cho SY. Assessment
of liver fibrosis with gadoxetic acid-enhanced MRI: comparisons
with transient elastography, ElastPQ, and serologic fibrosis
markers. Abdom Radiol (NY) 2019;44:2769e80. https:
//doi.org/10.1007/s00261-019-02041-z. PMID: 31041497.
26. Soresi M, Giannitrapani L, Cervello M, Licata A, Montalto G.
Non invasive tools for the diagnosis of liver cirrhosis. World J
Gastroenterol 2014;20(48):18131e50. https:
//doi.org/10.3748/wjg.v20.i48.18131. PMID: 25561782.
27. Chou R, Wasson N. Blood tests to diagnose fibrosis or cirrhosis
in patients with chronic hepatitis C virus infection: a system-
atic review. Ann Intern Med 2013;158:807e20. https:
Non-invasive assessment of hepatic fibrosis 801
//doi.org/10.7326/0003-4819-158-11-201306040-00005. PMID:
23732714.
28. Vesa CM, Popa AR, Bungau S, Daina LG, Buhas C, Judea-Pusta CT,
et al. Exploration of insulin sensitivity, insulin resistance, early
insulin secretion and b-cell function, and their relationship with
glycated hemoglobin level in normal weight patients with newly
diagnosed type 2 diabetes mellitus. Rev Chim 2019;70:4217e23.
https://doi.org/10.37358/RC.19.12.7735.
29. Caste
´ra L, Vergniol J, Foucher J, Le Bail B, Chanteloup E,
Haaser M, et al. Prospective comparison of transient elastog-
raphy, Fibrotest, APRI, and liver biopsy for the assessment of
fibrosis in chronic hepatitis C. Gastroenterology 2005;128:
343e50. https://doi.org/10.1053/j.gastro.2004.11.018. PMID:
15685546.
30. Lieber CS, Weiss DG, Morgan TR, Paronetto F. Aspartate
aminotransferase to platelet ratio index in patients with
alcoholic liver fibrosis. Am J Gastroenterol 2006;101:1500e8.
https://doi.org/10.1111/j.1572-0241.2006.00610.x. PMID:
16863553.
31. Singal AG, Thomassen LV, Gretch DR, Shuhart MC. Use of the
AST to platelet ratio index in HCV/HIV co-infected patients.
Aliment Pharmacol Ther 2011;33:566e77. https:
//doi.org/10.1111/j.1365-2036.2010.04560.x. PMID:
21205257. PMCID: PMC3552516.
32. Angulo P, Bugianesi E, Bjornsson ES, Charatcharoenwitthaya P,
Mills PR, Barrera F, et al. Simple noninvasive systems predict
long-term outcomes of patients with nonalcoholic fatty liver
disease. Gastroenterology 2013;145:782e9. https:
//doi.org/10.1053/j.gastro.2013.06.057. PMID: 23860502.
33. Pan X, Kaminga AC, Chen J, Luo M, Luo J. Fetuin-A and fetuin-B
in non-alcoholic fatty liver disease: a meta-analysis and meta-
regression. Int J Environ Res Public Health 2020;17:E2735.
https://doi.org/10.3390/ijerph17082735. PMID: 32326594.
34. Lin ZH, Xin YN, Dong QJ, Wang Q, Jiang XJ, Zhan SH, et al.
Performance of the aspartate aminotransferase-to-platelet
ratio index for the staging of hepatitis C-related fibrosis: an
updated meta-analysis. Hepatology 2011;53(3):726e36. https:
//doi.org/10.1002/hep.24105. PMID: 21319189.
35. Shaheen AA, Myers RP. Diagnostic accuracy of the aspartate
aminotransferase-to-platelet ratio index for the prediction of
hepatitis C-related fibrosis: a systematic review. Hepatology
2007;46:912e21. https://doi.org/10.1186/1471-230X-12-14.
PMID: 22333407.
36. Imbert-Bismut F, Ratziu V, Pieroni L, Charlotte F, Benhamou Y,
Poynard T, MULTIVIRCGroup. Biochemicalmarkers of liverfibrosis
in patients with hepatitis C virus infection: a prospective study.
Lancet 2001;357:1069e75. https://doi.org/10.1016/S0140-
6736(00)04258-6. PMID: 11297957.
37. Halfon P, Imbert-Bismut F, Messous D, Antoniotti G,
Benchetrit D, Cart-Lamy P, et al. A prospective assessment of
the inter-laboratory variability of biochemical markers of
fibrosis (FibroTest) and activity (ActiTest) in patients with
chronic liver disease. Comp Hepatol 2002;1:3. https:
//doi.org/10.1186/1476-5926-1-3. PMID: 12537583.
38. Salkic NN, Jovanovic P, Hauser G, Brcic M. FibroTest/Fibrosure
for significant liver fibrosis and cirrhosis in chronic hepatitis B:
a meta-analysis. Am J Gastroenterol 2014;109:796e809.
https://doi.org/10.1038/ajg.2014.21. PMID: 24535095.
39. Poynard T, Morra R, Halfon P, Castera L, Ratziu V, Imbert-
Bismut F, et al. Meta-analyses of FibroTest diagnostic value in
chronic liver disease. BMC Gastroenterol 2007;7:40. https:
//doi.org/10.1186/1471-230X-7-40. PMID: 17937811.
40. Naveau S, Gaude
´G, AsnaciosA, Agostini H, Abella A, Barri-Ova N,
et al. Diagnostic and prognostic values of noninvasive biomarkers
of fibrosis in patients with alcoholic liver disease. Hepatology
2009;49:97e105. https://doi.org/10.1002/hep.22576. PMID:
19053048.
41. Becker L, Salameh W, Sferruzza A, Zhang K, ng Chen R, Malik R,
et al. Validation of hepascore, compared with simple indices of
fibrosis, in patients with chronic hepatitis C virus infection in
United States. Clin Gastroenterol Hepatol 2009;7:696e701.
https://doi.org/10.1016/j.cgh.2009.01.010. PMID: 19514117.
42. Adams LA, Bulsara M, Rossi E, DeBoer B, Speers D, George J,
et al. Hepascore: an accurate validated predictor of liver
fibrosis in chronic hepatitis C infection. Clin Chem 2005;
51(10):1867e73. https://doi.org/10.1373/clin-
chem.2005.048389. PMID: 16055434.
43. Mehta P, Ploutz-Snyder R, Nandi J, Rawlins SR, Sanderson SO,
Levine RA. Diagnostic accuracy of serum hyaluronic acid,
FIBROSpect II, and YKL-40 for discriminating fibrosis stages in
chronic hepatitis C. Am J Gastroenterol 2008;103:928e36.
https://doi.org/10.1111/j.1572-0241.2007.01761.x. PMID:
18371145.
44. Rosenberg WM, Voelker M, Thiel R, Becka M, Burt A,
Schuppan D, et al. Serum markers detect the presence of liver
fibrosis: a cohort study. Gastroenterology 2004;127:1704e13.
https://doi.org/10.1053/j.gastro.2004.08.052. PMID:
15578508.
45. Kang JS, Lee MH. Noninvasive diagnostic and prognostic
assessment tools for liver fibrosis and cirrhosis in patients with
chronic liver disease. IntechOpen 2017. https:
//doi.org/10.5772/intechopen.68317.
46. Li H, Guo M, An Z, Meng J, Jiang J, Song J, et al. Prevalence
and risk factors of metabolic associated fatty liver disease in
Xinxiang, China. Int J Environ Res Public Health 2020;17:
E1818. https://doi.org/10.3390/ijerph17061818. PMID:
32168920. PMCID: PMC7143027.
47. Horowitz JM, Venkatesh SK, Ehman RL, Haveri K, Kamath P,
Ohliger MA, et al. Evaluation of hepatic fibrosis: a review from
the society of abdominal radiology disease focus panel. Abdom
Radiol (NY) 2017;42:2037e53. https://doi.org/10.1007/s00261-
017-1211-7. PMID: 28624924.
48. Jiang H, Zheng T, Duan T, Chen J, Song B. Non-invasive in vivo
imaging grading of liver fibrosis. J Clin Transl Hepatol 2018;6:
198e207. https://doi.org/10.14218/JCTH.2017.00038. PMID:
29951365.
49. Smith AD, Porter KK, Elkassem AA, Sanyal R, Lockhart ME.
Current imaging techniques for noninvasive staging of hepatic
fibrosis. AJR: Am J Roentgenol 2019;213:1e13. https:
//doi.org/10.2214/AJR.19.21144. PMID: 30973773.
50. Tang A, Cloutier G, Szeverenyi NM, Sirlin CB. Ultrasound elas-
tography and MR elastography for assessing liver fibrosis: Part
1, principles and techniques. AJR: Am J Roentgenol 2015;205:
22e32. https://doi.org/10.2214/AJR.15.14552. PMID:
25905647.
51. Verloh N, Einspieler I, Utpatel K, Menhart K, Brunner S,
Hofheinz F, et al. In vivo confirmation of altered hepatic
glucose metabolism in patients with liver fibrosis/cirrhosis by
18
F-FDG PET/CT. EJNMMI Res 2018;8:98. https:
//doi.org/10.1186/s13550-018-0452-y. PMID: 30414009.
52. Myers RP, Pomier-Layrargues G, Kirsch R, Pollett A, Beaton M,
Levstik M, et al. Discordance in fibrosis staging between liver
biopsy and transient elastography using the FibroScan XL probe.
J Hepatol 2012;56:564e70. https://doi.org/10.1016/j.j-
hep.2011.10.007. PMID: 22027584.
53. Friedrich-Rust M, Hadji-Hosseini H, Kriener S, Herrmann E,
Sircar I, Kau A, et al. Transient elastography with a new probe
for obese patients for non-invasive staging of non-alcoholic
steatohepatitis. Eur Radiol 2010;20:2390e6. https:
//doi.org/10.1007/s00330-010-1820-9. PMID: 20526777.
54. Naveau S, Lamouri K, Pourcher G, Njike
´-Nakseu M, Ferretti S,
Courie R, et al. The diagnostic accuracy of transient elastog-
raphy for the diagnosis of liver fibrosis in bariatric surgery
candidates with suspected NAFLD. Obesity Surg 2014;24:
802 G. Gheorghe et al.
1693e701. https://doi.org/10.1007/s11695-014-1235-9. PMID:
24841950.
55. Piotrowski D, Sa
˛czewska-Piotrowska A, Jaroszewicz J,
Boro
n-Kaczmarska A. Lymphocyte-to-monocyte ratio as the
best simple predictor of bacterial infection in patients with
liver cirrhosis. Int J Environ Res Public Health 2020;17:
E1727. https://doi.org/10.3390/ijerph17051727.PMID:
32155772.
56. Nightingale K. Acoustic radiation force impulse (ARFI) imaging:
a review. Curr Med Imaging Rev 2011;7:328e39. https:
//doi.org/10.2174/157340511798038657. PMID: 22545033.
57. Srinivasa Babu A, Wells ML, Teytelboym OM, Mackey JE,
Miller FH, Yeh BM, et al. Elastography in chronic liver disease:
modalities, techniques, limitations, and future directions.
Radiographics 2016;36:1987e2006. https:
//doi.org/10.1148/rg.2016160042. PMID: 27689833.
58. Sirli R, Bota S, SporeaI, Jurchis A, Popescu A, Gradinaru-Tasc
au O,
et al. Liver stiffness measurements by means of supersonic shear
imaging in patients without known liver pathology. Ultrasound
Med Biol 2013;39:1362e7. https://doi.org/10.1016/j.ul-
trasmedbio.2013.03.021. PMID: 23743106.
59. Papastergiou V, Tsochatzis E, Burroughs AK. Non-invasive
assessment of liver fibrosis. Ann Gastroenterol 2012;25:
218e31. PMID: 24714123. PMCID: PMC3959378.
60. Huwart L, Sempoux C, Vicaut E, Salameh N, Annet L, Danse E,
et al. Magnetic resonance elastography for the noninvasive
staging of liver fibrosis. Gastroenterology 2008;135:32e40.
https://doi.org/10.1053/j.gastro.2008.03.076. PMID: 18471441.
61. Tatsumi C, Kudo M, Ueshima K, Kitai S, Takahashi S, Inoue T,
et al. Noninvasive evaluation of hepatic fibrosis using serum
fibrotic markers, transient elastography (FibroScan) and real-
time tissue elastography. Intervirology 2008;51(Suppl. 1):
27e33. https://doi.org/10.1159/000122602. PMID:
Q18544945Q.
62. Colombo S, Buonocore M, Del Poggio A, Jamoletti C, Elia S,
Mattiello M, et al. Head-to-head comparison of transient
elastography (TE), real-time tissue elastography (RTE), and
acoustic radiation force impulse (ARFI) imaging in the diagnosis
of liver fibrosis. J Gastroenterol 2012;47:461e9. https:
//doi.org/10.1007/s00535-011-0509-4. PMID: Q22223175Q.
63. Singh S, Venkatesh SK, Wang Z, Miller FH, Motosugi U, Low RN, et al.
Diagnostic performance of magnetic resonance elastography in
staging liver fibrosis: a systematic review and meta-analysis of in-
dividual participant data. Clin Gastroenterol Hepatol 2015;13:
440e51. https://doi.org/10.1016/j.cgh.2014.09.046.PMID:
25305349.
64. Guo Y, Parthasarathy S, Goyal P, McCarthy RJ, Larson AC,
Miller FH. Magnetic resonance elastography and acoustic ra-
diation force impulse for staging hepatic fibrosis: a meta-
analysis. Abdom Imaging 2015;40:818e34. https:
//doi.org/10.1007/s00261-014-0137-6. PMID: 24711064.
65. Caste
´ra L, Vergniol J, Foucher J, Le Bail B, Chanteloup E,
Haaser M, et al. Prospective comparison of transient elastog-
raphy, Fibrotest, APRI, and liver biopsy for the assessment of
fibrosis in chronic hepatitis C. Gastroenterology 2005;128:
343e50. https://doi.org/10.1053/j.gastro.2004.11.018. PMID:
15685546.
66. Bourliere M, Penaranda G, Renou C, Botta-Fridlund D, Tran A,
Portal I, et al. Validation and comparison of indexes for fibrosis
and cirrhosis prediction in chronic hepatitis C patients: pro-
posal for a pragmatic approach classification without liver bi-
opsies. J Viral Hepat 2006;13:659e70. https:
//doi.org/10.1111/j.1365-2893.2006.00736.x. PMID:
16970597.
67. Sebastiani G, Vario A, Guido M, Noventa F, Plebani M, Pistis R,
et al. Stepwise combination algorithms of non-invasive markers
to diagnose significant fibrosis in chronic hepatitis C. J Hepatol
2006;44:686e93. https://doi.org/10.1016/j.j-
hep.2006.01.007. PMID: 16490278.
68. Loomba R, Adams LA. Advances in non-invasive assessment of
hepatic fibrosis. Gut 2020;69:1343e52. https:
//doi.org/10.1136/gutjnl-2018-317593. PMID: 32066623.
69. Boursier J, de Ledinghen V, Leroy V, Anty R, Francque S,
Salmon D, et al. A stepwise algorithm using an at-a-glance first-
line test for the non-invasive diagnosis of advanced liver
fibrosis and cirrhosis. J Hepatol 2017;66:1158e65. https:
//doi.org/10.1016/j.jhep.2017.01.003. PMID: 28088581.
70. Krawczyk M, Rau M, Schattenberg JM, Bantel H, Pathil A,
Demir M, et al. Combined effects of the PNPLA3 rs738409,
TM6SF2 rs58542926, and MBOAT7 rs641738 variants on NAFLD
severity: a multicenter biopsy-based study. J Lipid Res 2017;
58:247e55. https://doi.org/10.1194/jlr.P067454. PMID:
27836992.
71. Huang H, Shiffman ML, Friedman S, Venkatesh R, Bzowej N,
Abar OT, et al. A 7 gene signature identifies the risk of devel-
oping cirrhosis in patients with chronic hepatitis C. Hepatology
2007;46:297e306. https://doi.org/10.1002/hep.21695. PMID:
17461418.
72. Gheorghe G, Stoian AP, Gaman MA, Socea B, Neagu TP,
Stanescu AMA, et al. The benefits and risks of antioxidant
treatment in liver diseases. Rev Chim 2019;70:651e5. https:
//doi.org/10.37358/RC.19.2.6977.
73. Chen YM, Liu Y, Zhou RF, Chen XL, Wang C, Tan XY, et al. As-
sociations of gut-flora-dependent metabolite trimethylamine-
N-oxide, betaine and choline with nonalcoholic fatty liver
disease in adults. Sci Rep 2016;6:19076. https:
//doi.org/10.1038/srep19076. PMID: 26743949.
74. Loomba R, Seguritan V, Li W, Long T, Klitgord N, Bhatt A,
et al. Gut microbiome-based metagenomic signature for
non-invasive detection of advanced fibrosis in human
nonalcoholic fatty liver disease. Cell Metab 2017;25:
1054e62. https://doi.org/10.1016/j.cmet.2017.04.001.
PMID: 28467925.
75. Tseng CH, Wu CY. The gut microbiome in obesity. J Formos Med
Assoc 2019;118:S3e9. https:
//doi.org/10.1016/j.jfma.2018.07.009. PMID: 30057153.
76. Yang YJ, Ni YH. Gut microbiota and pediatric obesity/non-
alcoholic fatty liver disease. J Formos Med Assoc 2019;118:
S55e61. https://doi.org/10.1016/j.jfma.2018.11.006. PMID:
30509561.
77. Chen MX, Wang SY, Kuo CH, Tsai IL. Metabolome analysis for
investigating host-gut microbiota interactions. J Formos Med Assoc
2019;118:S10e22. https://doi.org/10.1016/j.jfma.2018.09.007.
PMID: 30269936.
78. Weng MT,Chiu YT,Wei PY,Chiang CW, FangHL, Wei SC. Microbiota
and gastrointestinal cancer. JFormosMedAssoc2019;118:
S32e41. https://doi.org/10.1016/j.jfma.2019.01.002.PMID:
30655033.
Non-invasive assessment of hepatic fibrosis 803
... For patients not successfully treated, subsequent tests are useful in assessing ibrosis progression. These tests are also used in patients with other chronic liver diseases, such as non-alcoholic fatty liver disease and primary sclerosing cholangitis [1,3]. ...
... These tests are often used to differentiate patients with signi icant ibrosis (F2 to F4) from those with minimal or absent ibrosis (F0 to F1). Many of these tests have been evaluated in speci ic populations (often patients with chronic hepatitis C virus -HCV), which should be considered when attempting to generalize results to other populations [1,3]. ...
... In patients with chronic hepatitis C [3], evaluating ibrosis progression can be valuable for several reasons: ...
Article
Full-text available
Aim: This study examines the pathological outcomes of chronic liver injuries, with a focus on liver fibrosis. It emphasizes understanding the structural changes within the liver that may lead to cirrhosis and functional impairments, crucial for developing targeted antifibrotic therapies. Methods: Our approach reviews existing literature detailing the use of traditional diagnostic methods—biochemical and serological tests alongside liver biopsies. Additionally, we evaluate the reliability and efficacy of non-invasive techniques such as serological test panels and imaging examinations. These methods are compared to understand their viability as supplementary or alternative diagnostic tools to liver biopsy. Significance: Liver fibrosis, if unmanaged, can progress to severe conditions such as cirrhosis and hepatocellular carcinoma, making it vital to understand its progression and treatment options. This study underscores the need for precise and non-invasive diagnostic tools in the clinical management of liver fibrosis, providing insight into the progression of chronic liver diseases and potential therapeutic targets. Conclusion and future perspectives: The research confirms that while liver biopsy remains the definitive method for staging liver fibrosis, its risks and limitations necessitate the use of enhanced non-invasive diagnostic techniques. These methods have shown promising results in accuracy and are critical for broadening clinical applications and patient safety. It is recommended that the scientific community continue to develop and validate non-invasive diagnostic tools. Enhancing the accuracy and reliability of these tools can provide a cost-effective, accessible, and safer alternative for large-scale screening and management of liver fibrosis in asymptomatic populations. Additionally, integrating advancements in radiologic and serological markers can further refine these diagnostic methods, improving overall patient outcomes.
... The other popular and thoroughly tested noninvasive techniques for determining liver fibrosis are transient elastography (TE) (Fibroscan), an ultrasoundbased procedure (14). This technique has been used to diagnose liver stiffness and fibrosis in the past (15,16). Thus, monitoring the stiffness of the liver over time can show how some liver disorders, including NASH, progress (17). ...
... Fibroscan is an ultrasound-based technique and one of the most extensively used and well-validated non-invasive methods for the assessment of liver fibrosis. Diagnosis of liver stiffness and fibrosis by this method was reported previously (15,16). Thus, serial evaluation of liver stiffness can provide evidence about the progression of liver diseases like NASH. ...
Article
Full-text available
Background: Most prevalent chronic liver disease in developed and developing nations is non-alcoholic fatty liver disease. From fatty liver, which often has benign, non-progressive clinical history, to non-alcoholic steatohepatitis, a more serious variant of fatty liver that can lead to cirrhosis and end-stage liver disease, non-alcoholic fatty liver disease encompasses broad spectrum of diseases. The gold standard for determining extent of hepatic fibrosis is still liver biopsy; however, number of noninvasive tests have been established to make diagnosis and assess effectiveness of treatment. Objective: Aim of study was to assess effectiveness of the combination of fibroscan and noninvasive biochemical tests and scoring systems for assessing liver fibrosis. Subjects and Methods: Cross-sectional prospective study conducted at outpatient clinic of Baghdad Gastroenterology and Hepatology Teaching Hospital from October 2018-March 2020. One hundred patients with fatty liver selected and subjected to specific questionnaires. Results: One hundred patients with fatty liver studied; fifty-five (55%) males, forty-five (45%) females. Mean age was (45 ±12.24) years, females were significantly older. Mean stiffness score was 11.7(SD:5.29) KPa. Forty-six (46%) patients had advanced fibrosis. The Positive correlation between FibroScan examination results and the noninvasive scores results was significant, as well as there was significant positive correlation between age and stiffness score and significant negative correlation between platelet count and stiffness score. Conclusions: This study showed that half patients showed advanced fibrosis, highlighting the need for early detection and management of fatty liver patients. Implementation of FibroScan with noninvasive fibrosis scoring tools has been shown to be helpful in this situation.
... Hepatic fibrosis is a pathological repair process in which multiple pathogenic factors stimulate and activate HSC, leading to abnormal proliferatsion of connective tissue in the liver, destruction of liver structure, and abnormal physiological function changes. When the structure and function of liver cells are damaged, it can cause liver cell swelling, increased index of liver, and increased membrane permeability, leading to the infiltratsion of permeable enzymes into the blood [71]. ALT and AST are the most sensitive indicators reflecting the stability of ...
Article
Full-text available
Objective This study aimed to evaluate the intervention effect of curcumin on hepatic fibrosis in rodent models through systematic review and meta-analysis, in order to provide meaningful guidance for clinical practice. Methods A systematic retrieval of relevant studies on curcumin intervention in rats or mice hepatic fibrosis models was conducted, and the data were extracted. The outcome indicators included liver cell structure and function related indicators, such as alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), albumin (ALB), ratio of albumin to globulin (A/G), total bilirubin (TBIL), bax protein, bcl-2 protein and index of liver, as well as the relevant indicators for evaluating the degree of hepatic fibrosis, such as hyaluronic acid (HA), laminin (LN), type I collagen (Collagen I), type III collagen (Collagen III), type III procollagen (PCIII), type III procollagen amino terminal peptide (PIIINP), type IV collagen (IV-C), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), α-Smooth muscle actin (α-SMA), hydroxyproline (HYP), platelet derived factor-BB (PDGF-BB), connective tissue growth factor (CTGF) and transforming growth factor-β1 (TGF-β1), and oxidative stress-related indicators, such as superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px). These results were then analyzed by meta-analysis. Studies were evaluated for methodological quality using the syrcle’s bias risk tool. Results A total of 59 studies were included in the meta-analysis, and the results showed that curcumin can reduce the levels of ALT, AST, ALP, TBIL, bax protein, and index of liver in hepatic fibrosis models. It can also reduce HA, LN, Collagen I, Collagen III, PCIII, PIIINP, IV-C, TNF-α, α-SMA, HYP, PDGF-BB, CTGF, TGF-β1 and MDA, and increase the levels of ALB, A/G, SOD, and GSH-Px in the hepatic fibrosis models. However, the effects of curcumin on bcl-2 protein, IL-6 in hepatic fibrosis models and index of liver in mice were not statistically significant. Conclusion The analysis results indicate that curcumin can reduce liver cell apoptosis by maintaining the stability of liver cell membrane, inhibit the activation and proliferation of hepatic stellate cells by reducing inflammatory response, and alleviate tissue peroxidation damage by clearing oxygen free radicals.
... The etiology of the diseases is broad, including accumulation of toxins, alcohol abuse for a prolonged time, infections, autoimmune diseases, metabolic disorders, as well as genetic disorders. CLD is an extremely common clinical condition, and the focus is on common etiologies, clinical manifestations and management 6 . Over the past three decades, experts have extensively explored the topic of drug-induced liver injury in patients with liver disease [7][8][9] . ...
Article
Full-text available
Introduction. Chronic liver disease is an important public health problem with a major negative impact on all aspects of patients’ lives. The consequences are directly related to the chronic liver disease, but also to the administered treatment. The objective of the study was to evaluate the quality of life in patients with chronic liver diseases by using SF-LDQOL questionnaire, including complementary questions related to the use of nutritional supplements. Materials and methods. A cross-sectional analysis on the quality of life of patients with a confirmed diagnosis of chronic liver disease (CLD), using the SF-LDQOL short form questionnaire, on a group of 40 patients was conducted. Data on CLD patients (age, sex, disease activity, risk factors, major clinical symptoms, use of hepatoprotective nutritional supplements (NS)) were collected and analyzed. Results. The mean age of participants was 68.34±14.45 years. The CLD impact on the general health status and quality of life was moderate to mild in the studied group. In most cases, the linear frequency shows an increasing trend for minimal impact. Regarding the use of nutritional supplements (NS), more than 80% of patients with CLD used them, especially those with hepatoprotective effects. The use of NS had a positive impact on the quality of life. Conclusions. The SF-LDQOL questionnaire, supplemented with questions about the use of NS, is a useful tool for physicians and pharmacists to use as a routine assessment of the quality of life in CLD patients, while also assessing the impact of hepatoprotective supplements consumption.
... Biopsy is considered the gold-standard method for diagnosing liver disease and fibrosis; however, it can lead to severe bleeding and pain owing to its invasiveness (83). In addition, biopsy is limited to a small area of the liver and results in sampling errors, thereby leading to a potentially inaccurate diagnosis of heterogeneously distributed liver diseases (84). Therefore, a non-invasive diagnostic method is required for accurate and safe assessment of the extent of liver disease and fibrosis. ...
Article
Full-text available
MicroRNAs are small non-coding RNAs with a length of 20-24 nucleotides. They bind to the 3'-untranslated region of target genes to induce the degradation of target mRNAs or inhibit their translation. Therefore, they are involved in the regulation of development, apoptosis, proliferation, differentiation and other biological processes (including hormone secretion, signaling and viral infections). Chronic diseases in children may be difficult to treat and are often associated with malnutrition resulting from a poor diet. Consequently, further complications, disease aggravation and increased treatment costs impose a burden on patients and their families. Existing evidence suggests that microRNAs are involved in various chronic non-neoplastic diseases in children. The present review discusses the roles of microRNAs in five major chronic diseases in children, namely, diabetes mellitus, congenital heart diseases, liver diseases, bronchial asthma and epilepsy, providing a theoretical basis for them to become therapeutic biomarkers in chronic pediatric diseases.
Article
Full-text available
Background: Hepatitis C and B virus infections significantly contribute to global chronic liver disease mortality. Objectives: This study explores the role of serum markers (AST/ALT ratio, APRI Score, FIB-4 Score, and Forns index) in non-invasively assessing liver damage in patients with chronic hepatitis C and B. Methods: In this single-center, retrospective, observational study, we analyzed data from 327 patients to establish correlations between serological markers and fibrosis grade using Spearman's correlation. Receiver operator characteristic (ROC) analysis evaluated the ability of these markers to predict advanced fibrosis. Results: In hepatitis B and C cohorts, all markers show significant positive correlations with liver fibrosis (P < 0.001). FIB-4 and the Forns index exhibit moderate correlation (Spearman’s rho 0.48), while AST/ALT and APRI score show mild correlation (Spearman’s rho 0.21 and 0.31). In hepatitis C, the Forns index (0.814) and FIB-4 (0.80) outperform other markers. In hepatitis B, Forns (AUC = 0.73), APRI (AUC = 0.68), and FIB-4 (AUC = 0.68) demonstrate significant predictive ability. Conclusions: FIB-4 and the Forns index hold clinical significance as fibrosis biomarkers in the management of chronic viral hepatitis. FIB-4 is a universal marker, while the interpretation of the Forns index requires consideration of the etiology of chronic viral hepatitis.
Article
Full-text available
Hepatitis C virus (HCV) infection has been a major global health concern, with a significant impact on public health. In recent years, there have been remarkable advancements in our understanding of HCV and the development of novel therapeutic agents. The Saudi Society for the Study of Liver Disease and Transplantation formed a working group to develop HCV practice guidelines in Saudi Arabia. The methodology used to create these guidelines involved a comprehensive review of available evidence, local data, and major international practice guidelines regarding HCV management. This updated guideline encompasses critical aspects of HCV care, including screening and diagnosis, assessing the severity of liver disease, and treatment strategies. The aim of this updated guideline is to assist healthcare providers in the management of HCV in Saudi Arabia. It summarizes the latest local studies on HCV epidemiology, significant changes in virus prevalence, and the importance of universal screening, particularly among high-risk populations. Moreover, it discusses the promising potential for HCV elimination as a public health threat by 2030, driven by effective treatment and comprehensive prevention strategies. This guideline also highlights evolving recommendations for advancing disease management, including the treatment of HCV patients with decompensated cirrhosis, treatment of those who have previously failed treatment with the newer medications, management in the context of liver transplantation and hepatocellular carcinoma, and treatment for special populations.
Article
Full-text available
Objectives: Serological tests and elastography have recently been used more commonly for the detection of liver fibrosis. The aim of this study was to compare the histopathologically confirmed liver fibrosis stage with serological tests and point shear wave elastography (pSWE) in chronic hepatitis B (CHB) patients. Materials and Methods: Patients who underwent liver biopsy for CHB in the infectious diseases clinic were included. Demographic characteristics, laboratory results, and pSWE measurements were recorded retrospectively. pSWE measurements were evaluated according to the guidelines of the European Federation of Ultrasound Societies in Medicine and Biology. Results: Thirty-five patients were included in the study, 23 (65.7%) of whom were male. The mean age was 47.2±12.6. Significant fibrosis was found in 15 patients (42.9%) on histopathological evaluation. The mean pSWE value of patients with mild fibrosis was 1.6±0.5 m/sec and with significant fibrosis was 2.2±0.5 m/ sec. Significant fibrosis risk was shown to be associated with mean pSWE values (p=0.002) and the accuracy rate was calculated 62.8% (the area under the curve: 0.807). When the cut-off value of pSWE was taken as 1.77 m/sec to determine significant fibrosis (likelihood ratio: 2.67), the sensitivity was 80% and the specificity was 70% (p=0.002). The correlation between pSWE median values with age (r=0.452, p<0.01), body mass index (r=0.673, p<0.01), grade of steatosis (r=0.534, p<0.01), and stage of fibrosis (r=0.633, p<0.01) was calculated. Conclusion: pSWE is promising; however, it is thought that the method will develop further if pSWE is used more frequently in clinical practice.
Article
Full-text available
The magnitude of the effect of fetuin-A and fetuin-B on non-alcoholic fatty liver disease (NAFLD) remains undefined. Therefore, the aim of this study was to synthesize previous findings to obtain a reliable estimation of this relationship. This study was registered in PROSPERO with the number CRD42019126314. Studies published not later than March 2019, examining the relationship between fetuin-A, fetuin-B, and NAFLD, were identified by a systematic search in the electronic databases of the Web of Science, PubMed, Embase, and Cochrane Library. Pooled estimates of standardized mean difference (SMD), calculated using the random-effects model in a meta-analysis, were applied to estimate the strength of the association between fetuin-A, fetuin-B, and NAFLD. Thirty publications were identified and analyzed based on specified inclusion criteria. Collectively, they consisted of 3800 NAFLD participants and 3614 controls. Compared with the controls, significant higher values of the fetuin-A (SMD = 0.83, 95% CI: 0.59 to 1.07, Z = 6.82, p < 0.001) and fetuin-B (SMD = 0.18, 95% CI: 0.02 to 0.33, Z = 2.27, p = 0.023) were observed in NAFLD patients. Meanwhile, in the subgroup analysis, the effect value of fetuin-A in the NASH group was significantly higher than that in the NAFL group (p = 0.036). The findings of this study suggest that elevated fetuin-A and fetuin-B may independently indicate the occurrence of NAFLD. Nevertheless, further research is needed to confirm these results.
Article
Full-text available
Metabolic syndrome is associated with increased risk of cardiovascular disease. This study investigated the correlation between adipocyte and inflammation biomarkers, and metabolic syndrome and its components. The study included 80 patients with normal body-mass index and 80 obese patients. The groups were assessed for serum values of adiponectin, leptin and highly sensitive C reactive protein (hsCRP), the homeostatic model assessment of insulin resistance (HOMA-IR), as well as the influence of these biochemical markers on the prevalence of metabolic syndrome and its components. Leptin, HOMA-IR and hsCRP had statistically significant (P<0.01) higher values in the group of obese subjects, while adiponectin had statistically significant (P<0.01) lower values. The prevalence of metabolic syndrome was 35% in the obese group and 5% in the normal weight group. Adiponectin and HOMA-IR were the variables significantly associated with metabolic syndrome (P<0.01), adiponectin/HOMA-IR ratio and leptin/adiponectin ratio were also associated with metabolic syndrome (P<0.01). No relationship was found between metabolic syndrome and hsCRP. Adiponectin and adiponectin/HOMA-IR were associated with all the components of metabolic syndrome and they can be useful to identify patients with high risk of diabetes mellitus and cardiovascular disease.
Article
Full-text available
Introduction. The concept of underactive bladder is relatively new. Currently there is no generally accepted definition of this pathology. Diagnosis depends on urodynamic findings, and symptoms are usually rare and intricated with the symptoms of other urinary pathology. Matherials and methods. This review examines the current literature on underactive bladder regarding pathology, definition, diagnosis, current guidelines, and any further potential medical developments. Conclusions. Underactive bladder is a poorly understood pathologic condition. Only since 2002 has there been any consensus regarding the definition. The diagnosis relies only on urodynamics; clinical diagnosis is a challenge even for a consultant; and treatment does not seem to alleviate much of the suffering. This disease remains underrecognized and undertreated. More research is needed to identify less invasive diagnosis tools and treatment for this pathology.
Article
Full-text available
Metabolic associated fatty liver disease (MAFLD) is recognized as the liver disease component of metabolic syndrome, which is mainly related to insulin resistance and genetic susceptibility. It is the most prevalent chronic liver disease worldwide. With rapid lifestyle transitions, its prevalence worldwide is increasing, and tremendous challenges in controlling this pandemic are arising. The objective of this study was to investigate the prevalence and risk factors of MAFLD in rural areas of Xinxiang, Henan in 2017. We conducted a cross-sectional analysis of rural inhabitants aged 20-79 years in Xinxiang, Henan in 2017, using cluster random sampling (N = 9140). Physical examinations were conducted at local clinics from April to June 2017. After overnight fasting, all participants underwent physical examinations, blood routine tests, biochemical examinations, and liver ultrasound and completed questionnaires. We investigated the crude and age-adjusted MAFLD prevalence and analyzed the characteristics of those with, and without, MAFLD, using logistic regression. Approximately 2868 (31.38%) participants were diagnosed with MAFLD. The overall age-adjusted MAFLD prevalence was 29.85% (men: 35.36%; women: 26.49%). The MAFLD prevalence increased with age, and peaked at the 50-59-year age group, and then began to decline. Higher body mass index, waist circumference, percentage of lymphocytes, levels of hemoglobin, platelet count, triglyceride, fasting plasma glucose, and serum uric acid were independently and positively correlated with MAFLD; In contrary, active physical activity and high-density lipoprotein cholesterol were negatively correlated with MAFLD. In summary, the MAFLD prevalence in the study population was 29.85%. Higher body mass index, waist circumference, percentage of lymphocytes, levels of hemoglobin, platelet count, triglyceride, fasting plasma glucose, and serum uric acid were risk factors for MAFLD.
Article
Full-text available
This study evaluated the clinical characteristics of the acute coronary syndromes (ACS) in chronic kidney disease (CKD) patients and established prognostic values of the biomarkers and echocardiography. 273 patients admitted to the cardiology department of the Clinical County Emergency Hospital of Oradea, Romania, with ACS diagnosis were studied. Two study groups were formed according to the presence of CKD (137 patients with ACS + CKD and 136 with ACS without CKD). Kidney Disease: Improving Global Outcomes (KDIGO) threshold was used to assess the stages of CKD. Data regarding the medical history, laboratory findings, biomarkers, echocardiography, and coronary angiography were analysed for both groups. ACS parameters were represented by ST-segment elevation myocardial infarction (STEMI), which revealed a greater incidence in subjects without CKD (43.88%); non-ST-segment elevation myocardial infarction (NSTEMI), characteristic for the CKD group (28.47%, with statistically significance p = 0.04); unstable angina and myocardial infarction with nonobstructive coronary arteries (MINOCA). Diabetes mellitus, chronic heart failure, previous stroke, and chronic coronary syndrome were more prevalent in the ACS + CKD group (56.93%, p < 0.01; 41.61%, p < 0.01; 18.25%, p < 0.01; 45.26%, p < 0.01). N-terminal pro b-type natriuretic peptide (NT-proBNP) was statistically higher (p < 0.01) in patients with CKD; Killip class 3 was evidenced more frequently in the same group (p < 0.01). Single-vessel coronary artery disease (CAD) was statistically more frequent in the ACS without CKD group (29.41%, p < 0.01) and three-vessel CAD or left main coronary artery disease (LMCA) were found more often in the ACS + CKD group (27.01%, 14.6%). Extension of the CAD in CKD subjects revealed an increased prevalence of the proximal CAD, and the involvement of various coronary arteries is characteristic in these patients. Biomarkers and echocardiographic elements can outline the evolution and outcomes of ACS in CKD patients.
Article
Full-text available
Background and aim: The aim of this study was to assess the diagnostic performance of new morphology-related indices and Child–Turcotte–Pugh (CTP) and Model for End-Stage Liver Disease (MELD) scores during hospitalization in predicting the onset of bacterial infection in patients with liver cirrhosis. Material and methods: A total of 171 patients (56.9% males; median age 59 years; total number of hospitalizations 209) with liver cirrhosis were included in this observational study. The diagnosis of cirrhosis was made on the basis of clinical, biochemical, ultrasonic, histological, and endoscopic findings. The neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), modified aspartate aminotransferase-to-platelet ratio index (APRI), aspartate aminotransferase-to-alanine aminotransferase ratio (AAR), Fibrosis-4 index (FIB-4), platelet-to-lymphocyte ratio (PLR), neutrophil-to-monocyte ratio (NMR), and CTP and MELD scores were calculated for the cases of patients with cirrhosis. Results: Bacterial infection was diagnosed in 60 of the 209 (28.7%) hospitalizations of patients with cirrhosis. The most common infections were urinary tract infection (UTI), followed by pneumonia and sepsis. The more severe the liver failure, the greater the bacterial infection prevalence and mortality. Patients with decompensated liver cirrhosis were infected more often than subjects with compensated cirrhosis (50.0% vs. 12.9%, p = 0.003). The calculated MELD score, CTP, NLR, LMR, AAR, monocyte count, and C-reactive protein (CRP) concentration were also related to the bacterial infection prevalence, and mortality areas under the curve (AUC) were 0.629, 0.687, 0.606, 0.715, 0.610, 0.648, and 0.685, respectively. The combined model with two variables (LMR and CTP) had the best AUC of 0.757. The most common bacteria isolated from patients with UTI were Escherichia coli, Enterococcus faecalis, and Klebsiella pneumonia. Gram-negative bacteria were also responsible for spontaneous bacterial peritonitis (SBP), and together with gram-positive streptococci and staphylococci, these microorganisms were isolated from blood cultures of patients with sepsis. Significant differences were found between CTP classification, MELD score, NLR, LMR, AAR, CRP, and PLR in patients with cirrhosis with, or without, bacterial infection. Conclusions: Bacterial infection prevalence is relatively high in patients with liver cirrhosis. Although all analyzed scores, including the LMR, NLR, aspartate aminotransferase (AST)/alanine aminotransferase (ALT), CRP, CTP, and MELD, allowed the prediction of bacterial occurrence, the LMR had the highest clinical utility, according to the area under the curve (AUC) and odds ratio (OR).
Article
Full-text available
The current study aims to investigate the influence of five rare earth elements (REEs) (i.e., lanthanum (La), cerium (Ce), praseodymium (Pr), neodymium (Nd), and gadolinium (Gd)) on the growth of Sprague-Dawley (SD) rats, and to explore the accumulation characteristics of REEs in tissues and organs with different doses as well as the detoxification and elimination of high-dose REEs. Fifty healthy male SD rats (140~160 g) were randomly divided into five groups and four of them were given gavage of sodium citrate solution with REEs in different doses, one of which was the control group. Hair, blood, and bone samples along with specific viscera tissue samples from the spleen and the liver were collected for detection of REEs by Inductively Coupled Plasma-Mass Spectrometry (ICP-MS). Treated rats expressed higher concentrations of REEs in the bones, the liver, and spleen samples than the control group (P < 0.05). Few differences were found in relative abundance of La, Ce, Pr, Nd, and Gd in the hair and the liver samples, although different administration doses were given. The relative abundance of Ce in bone samples was significantly lower in the low-dose group and control group, whereas the relative abundance of La and Pr in the bone samples were highest among all groups. Although in the REEs solution, which was given to rats in high-dose group, the La element had a higher relative abundance than Ce element, it ended up with higher Ce element relative abundance than La element in the spleen samples. REEs had a hormetic effect on body weight gain of SD rats. The accumulation of the measured REEs were reversible to low concentrations in the blood and hair, but non-reversible in the bones, the spleen, and the liver. Different tissues and organs can selectively absorb and accumulate REEs. Further inter-disciplinary studies about REEs are urgently needed to identify their toxic effects on both ecosystems and organisms.
Article
Full-text available
Background: Previous research found that statins, in addition to its efficiency in treating hyperlipidemia, may also incur adverse drug reactions, which mainly include myopathies and abnormalities in liver function. Aim: This study aims to assess the risk for newly onset sarcopenia among patients with chronic kidney disease using statins. Material and Method: In a nationwide retrospective population-based cohort study, 75,637 clinically confirmed cases of chronic kidney disease between 1997 and 2011were selected from the National Health Insurance Research Database of Taiwan. The selection of the chronic kidney disease cohort included a discharge diagnosis with chronic kidney disease or more than 3 outpatient visits with the diagnosis of chronic kidney disease found within 1 year. After consideration of patient exclusions, we finally got a total number of 67,001 cases of chronic kidney disease in the study. The Cox proportional hazards model was used to perform preliminary analysis on the effect of statins usage on the occurrence of newly diagnosed sarcopenia; the Cox proportional hazards model with time-dependent covariates was conducted to take into consideration the individual temporal differences in medication usage, and calculated the hazard ratio (HR) and 95% confidence interval after controlling for gender, age, income, and urbanization. Results: Our main findings indicated that patients with chronic kidney disease who use statins seem to effectively prevent patients from occurrences of sarcopenia, high dosage of statins seem to show more significant protective effects, and the results are similar over long-term follow-up. In addition, the risk for newly diagnosed sarcopenia among patients with lipophilic statins treatment was lower than that among patients with hydrophilic statins treatment. Conclusion: It seems that patients with chronic kidney disease could receive statin treatment to reduce the occurrence of newly diagnosed sarcopenia. Additionally, a higher dosage of statins could reduce the incidence of newly diagnosed sarcopenia in patients with chronic kidney disease.
Article
Full-text available
The non-alcoholic fatty liver disease (NAFLD) is rapidly becoming the most common cause of chronic liver disease as well as the first cause of liver transplantation. NAFLD is commonly associated with metabolic syndrome (MetS), and this is the most important reason why it is extremely difficult to treat this disease bearing in mind the enormous amount of interrelationships between the liver and other systems in maintaining the metabolic health. The treatment of NAFLD is a key point to prevent NASH progression to advanced fibrosis, to prevent cirrhosis and to prevent the development of its hepatic complications (such as liver decompensation and HCC) and even extrahepatic one. A part of the well-known healthy effect of diet and physical exercise in this setting it is important to design the correct pharmaceutical strategy in order to antagonize the progression of the disease. In this regard, the current review has the scope to give a panoramic view on the possible pharmacological treatment strategy in NAFLD patients.
Article
Liver fibrosis should be assessed in all individuals with chronic liver disease as it predicts the risk of future liver-related morbidity and thus need for treatment, monitoring and surveillance. Non-invasive fibrosis tests (NITs) overcome many limitations of liver biopsy and are now routinely incorporated into specialist clinical practice. Simple serum-based tests (eg, Fibrosis Score 4, non-alcoholic fatty liver disease Fibrosis Score) consist of readily available biochemical surrogates and clinical risk factors for liver fibrosis (eg, age and sex). These have been extensively validated across a spectrum of chronic liver diseases, however, tend to be less accurate than more ‘complex’ serum tests, which incorporate direct measures of fibrogenesis or fibrolysis (eg, hyaluronic acid, N-terminal propeptide of type three collagen). Elastography methods quantify liver stiffness as a marker of fibrosis and are more accurate than simple serum NITs, however, suffer increasing rates of unreliability with increasing obesity. MR elastography appears more accurate than sonographic elastography and is not significantly impacted by obesity but is costly with limited availability. NITs are valuable for excluding advanced fibrosis or cirrhosis, however, are not sufficiently predictive when used in isolation. Combining serum and elastography techniques increases diagnostic accuracy and can be used as screening and confirmatory tests, respectively. Unfortunately, NITs have not yet been demonstrated to accurately reflect fibrosis change in response to treatment, limiting their role in disease monitoring. However, recent studies have demonstrated lipidomic, proteomic and gut microbiome profiles as well as microRNA signatures to be promising techniques for fibrosis assessment in the future.