ArticlePDF Available

Abstract and Figures

Background/aim: Silybin is the main component of silymarin with antioxidant, anti-inflammatory and cytoprotective actions. Our aim was to compare the effect of silybin used as single substance, silybin-phosphatidylcholine complex (SilPho), and derivatives of silybin (MannpSil, GalpSil, GlcpSil, LactpSil) on MKN28 and HepG2 cell viability and cell death, in vitro, after induction of oxidative stress. Materials and methods: Oxidative stress was induced by incubating HepG2 and MKN28 cells with xanthine oxidase in the presence of its substrate xanthine. Cell viability was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazoliumbromide assay. Determination of Malondialdehyde (MDA) in MKN28 cells was performed by high-performance liquid chromatography. Quantitative analysis of apoptotic cells was carried-out using annexin. Results: SilPho and new silybin glycoconjugates did not affect cell viability, while silybin induced about 50% cell death in both MKN28 and in HepG2 cells. Pre-treatment of cells with silybin and new silybin glycoconjugates (before oxidative stress induction) did not affect cell viability, while SilPho had a protective effect. Exposure of MKN28 cells to oxidative stress caused a two-fold increase in cellular MDA concentration compared to untreated cells. Moreover, pre-treatment with SilPho, but not with silybin, significantly prevented oxidative stress-induced increase in cellular Malondialdehyde. Moreover, silybin induced apoptosis potentiated by oxidative stress, while SilPho did not induce any effect. Oxidative stress caused cell death primarily by necrosis, antagonized by SilPho. Conclusion: The protective effect of SilPho is partially due to inhibition of radical oxidative species.
Content may be subject to copyright.
Editorial Office: International Institute of Anticancer Research,
DELINASIOS G.J & CO G.P., Kapandriti, POB 22, Attiki 19014,
Greece. Fax: 0030-22950-53389; Tel: 0030-22950-52945
e-mail: journals@iiar-anticancer.org
Dear Sir/Madam:
Enclosed are the galley proofs of your article for IN VIVO.
We would like to call your attention to the following:
1. Please read thoroughly, correct, and return the proofs to the Editorial
Office within 24 hours.
2. Proofs should be returned preferably by e-mail or fax. Delays in the
return of these proofs will necessitate the publication of your paper
in a later issue of the journal.
3. Please read the entire manuscript carefully to verify that no changes
in meaning have been introduced into the text through language
improvements or editorial corrections.
4. Corrections should be limited to typographical errors.
5. Should you require reprints, PDF file, online open access, issues or
special author rate subscriptions, please fill the attached reprint order
form.
6. If you opt for online open access publication of your paper, it will
instantly upon publication be available to read and reproduce free
of charge.
7. Should you require information about your article (publication date,
volume, page numbers, etc) please call: +30-22950-52945 or send
an e-mail to journals@iiar-anticancer.org.
8. Please provide your complete address (not P.O.B.), telephone and
fax numbers for the delivery of reprints and issues.
9. Please feel free to contact us with any queries that you may have
(Tel./Fax: +30-22950-53389 or +30-22950-52945, e-mail:
journals@iiar-anticancer.org).
Thank you for taking the time to study these guidelines.
I greatly appreciate your cooperation and your valuable contribution to
this journal.
Yours sincerely,
J.G. Delinasios
Managing Editor
Enclosures
in vivo International Journal of Experimental and Clinical
Pathophysiology and Drug Research
ISSN (print): 0258-851X, ISSN (online): 1791-7549
Abstract. Background: Silybin is the main component of
silymarin with antioxidant, anti-inflammatory and
cytoprotective actions. Aim: To compare the effect of silybin
used as single substance, silybin–phosphatidylcholine
complex (SilPho), and derivatives of silybin (MannpSil,
GalpSil, GlcpSil, LactpSil) on MKN28 and HepG2 cell
viability and cell death, in vitro, after the induction of
oxidative stress. Materials and Methods: Oxidative stress
was induced by incubating HepG2 and MKN28 cells with
xanthine oxidase in the presence of its substrate xanthine.
Cell viability was determined by the 3-(4,5-dimethylthiazol-
2-yl)-2,5-diphenyl tetrazoliumbromide assay. Determination
of Malondialdehyde in MKN28 cells was performed by High
Performance Liquid Chromatography. Quantitative analysis
of apoptotic cells was carried out using annexin. Results:
SilPho and new silybin glycoconjugates did not affect cell
viability, while silybin induced about 50% cell death in both
MKN28 and in HepG2 cells. The pre-treatment of cells with
silybin and new silybin glycoconjugates (before oxidative
stress induction) did not affect cell viability, while SilPho
had a protective effect. Exposure of MKN28 cells to oxidative
stress caused a twofold increase in cellular MDA
concentration compared to untreated cells. Moreover,
pretreatment with SilPho but not with silybin significantly
prevented oxidative stress-induced increase in cellular
Malondialdehyde. Moreover, silybin induced apoptosis
potentiated by oxidative stress, while SilPho did not induce
any effect. Oxidative stress caused cell death primarily by
necrosis, antagonized by SilPho. Conclusion: The protective
effect of SilPho is partially due to inhibition of radical
oxidative species.
Silybin is the main component of silymarin (famous
antioxidant) with an increasing number of effects (1).
Silibinin is a semipurified, commercially available fraction
of silymarin: Silibinin is an approximately 1:1 mixture of
two diastereoisomeric compounds, silybin A and silybin B
(2). Therefore, purified silybin and silibinin are practically
synonymous (2).
The main effects attributed both in vitro and in vivo to
silybin are related to its antioxidant, anti-inflammatory and
cytoprotective actions (3-5). Silybin is also considered a
chemopreventive and cancer-protective agent because it
modulates a series of mitogenic signaling and cell-cycle
regulators (6, 7), mediating a pro-apoptotic effect (8, 9).
Both bioavailability and therapeutic efficacy of silybin in
vivo are rather limited by low water solubility, low
bioavailability, and poor intestinal absorption (10). To
improve these pharmacological limitations, a silybin
phytosomecomplex (silybin plus phosphatidylcholine;
SilPho) has been co-formulated with vitamin E [Realsil
(RA), Istituto Biochimico Italiano, Lorenzini S.p.a., Italy]
(11, 12). Pharmacokinetic analyses indicated that the
bioavailability of silybinphytosome is much higher than that
of silymarin, and in this pharmaceutical preparation, silybin
is widely distributed in plasma and tissues, which include the
liver, lung, stomach, skin, and prostate (13, 14).
In vivo, silymarin and silybin have been used as
therapeutic herbal products for treatment of acute and
chronic liver diseases: in particular, alcoholic liver disease
and cirrhosis (15-17), nonalcoholic fatty liver disease (18)
and hepatic fibrosis (19, 20). In animals, silybinphytosome
complex reduces oxidative stress, lipid peroxidation,
collagen accumulation and consequently liver damage (19).
1
Correspondence to: A. Federico, MD, Ph.D., Department of
Clinical and Experimental Medicine, Second University of Naples,
Via Pansini 580131 Naples, Italy. Tel: +39 0815666723, Fax: +39
0815666837, e-mail: alessandro.federico@unina2.it
Key Words: Silybin, cell necrosis, oxidative stress.
in vivo 29: xxx-xxx (2015)
Silybin–Phosphatidylcholine Complex Protects Human
Gastric and Liver Cells from Oxidative Stress
ALESSANDRO FEDERICO1, MARCELLO DALLIO1, GIOVANNI DI FABIO2,
ARMANDO ZARRELLI2, SILVIA ZAPPAVIGNA3, PAOLA STIUSO3,
CONCETTA TUCCILLO1, MICHELE CARAGLIA3and CARMELA LOGUERCIO1
Departments of 1Clinical and Experimental Medicine, and 3Biochemistry, Biophysics
and General Pathology, Second University of Naples, Naples, Italy;
2Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
No: 3272-F
Please mark the appropriate
section for this paper
Experimental
Clinical
Epidemiological
0258-851X/2015 $2.00+.40
In men, RA ameliorates some serum and histological
parameters of liver damage and fibrosis (18).
Recently, Zarrelli et al. obtained new 9''–phosphodiester
silybin conjugates with different mono- and di-saccharide
labels through the anomeric hydroxyl group in order to
enhance the biological efficacy of the derivatives by
increasing their in vivo stability, binding affinity, and overall
uptake (21). These silybin derivatives have water solubility
well above that of silybin. Despite a large series of studies
reported in literature, confusion about the different actions
of silybin exists. Therefore the following merit investigation:
i) if the effects of silybin are similar in different cell lines of
different histogenesis; ii) the influence of the concentrations
used in different experimental models; c) the effects of the
different silybin derivatives.
In the present study, we compared the effect of silybin
used as a single agent or as SilPho, and different silybin
derivatives on MKN28 and HepG2 cell death in vitro after
the induction of oxidative stress. We used two cell lines
(MKN28 and HepG2) to verify the results obtained there by
excluding the possibility of interference of the type of cell
on the results.
Materi al s and Methods
Materials. Silybin was a gift from Indena (Milan, Italy). SilPho was
provided by Istituto Biochimico Italiano (G. Lorenzini S.p.A. Milan,
Italy). Silybin derivatives were synthesized according to Zarrelli et
al. (21): in detail, an efficient synthetic procedure leads to new
9''–phosphodiestersilybin conjugates with different mono- and di-
saccharide labels through the anomeric hydroxyl group. In this
approach a suitable 9''-phosphoramidite was used as silybin building
block and 1-OH full protected mono- and di-saccharide derivatives
chosen as sugar starting materials (21). The new silybin conjugates
were: silybin-900-phosphoryl-D-mannopyranoside (MannpSil),
silybin-900-phosphoryl-D-galactopyranoside (GalpSil), silybin-900-
phosphoryl-D-glucopyranoside (GlcpSil), silybin-900-phosphoryl-
D-Lactopyranoside (LactpSil).
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazoliumbromide
(MTT) assay was purchased by Sigma (Milan, Italy). Annexin
Apoptosis Detection Kit was obtained from BD Biosciences (San
Diego, CA, USA). Dulbecco’s modified Eagle’s medium
(DMEM):F12, penicillin, streptomycin, fetal bovine serum, L-
glutamine and trypsin/EDTA were obtained from Life Technologies
Inc. (Gaithersburg, MD, USA).
Agent preparation. Pure silybin was dissolved in dimethyl sulfoxide
(DMSO) and used at final concentration of 10, 25, 50, 75, 100 and
200 μM. SilPho was dissolved in DMSO to achieve final
concentrations of silybin similar to those employed for testing
silybin alone (10, 25, 50, 75, 100 and 200 μM). Silybin derivatives
were dissolved in water and used at final concentration of 10, 25,
50, 75, 100 and 200 μM.
Cell culture. HepG2 cells were derived from human hepatocellular
carcinoma (22) and MKN28 cells were derived from a human well-
differentiated gastric tubular adenocarcinoma and showing gastric-
type differentiation (23) (Cell Bank Interlab Cell Line Collection,
IST Genova, Italy). HepG2 and MKN28 cells were grown as
monolayer in DMEM supplemented with 10% fetal calf serum and
1% antibiotic-antimycotic solution (Life Technologies Inc.) at 37˚C
in a humidified atmosphere of 5% CO2 in air. Cytotoxicity
experiments were conducted using serum-free medium.
Induction of oxidative stress. Oxidative stress was induced by
incubating HepG2 and MKN28 cells with xanthine oxidase (XO; 10-
100 mU/ml) in the presence of its substrate xanthine (X; 1 mM) for
periods of up to 3 hours. Exposure of cells in culture to X-XO (1 mM
+ 50 mM) for 2 hours causes significant cell injury (50%) and this
has been demonstrated to be due to generation of radical oxidative
species (ROS) and in particular of OH produced from H2O2 by iron
catalyzed Fenton reaction (22).
We examined the effect of silybin, SilPho, and the new silybin
glycoconjugates (MannpSil, GalpSil, GlcpSil, LactpSil) on X-XO-
induced cell damage. Cells were incubated with serum-free medium
(Control) for 1-48 hours; with serum-free medium for 1-48 hours and
then with X-XO (1 mM + 50 mM) for 2 hours (X-XO control); with
silybin, SilPho, and new silybin glycoconjugates (10-200 μM) for 1-
48 hours and then, after washing, with X-XO (1 mM + 50 mM) for
2 hours.
Cell viability. Cell viability was determined by the MTT assay.
Briefly, 10 μl of MTT (5mg/ml saline) were added to each well, and
treated cells were incubated for 90 min at 37˚C and centrifuged for
five minutes. After aspiration of supernatant, cells were lysed and
solubilised by addition of 100 μl of 0.04N HCl containing
isopropanol. The absorbance of each sample was analyzed at 540
nm. Cell viability (%) was calculated by dividing the absorbance of
samples obtained from cells incubated with test drugs by the
absorbance of samples obtained from cells incubated with tissue
culture medium only (control) and multiplying this ratio by 100.
Data are presented as the mean±standard deviation (SD) of three
experiments run in duplicate.
Determination of lipid peroxidation. Malondialdehyde (MDA) is
considered a presumptive biomarker for lipid peroxidation in live
organisms and cultured cells (24). Determination of MDA in
MKN28 cells was performed by High Performance Liquid
Chromatography (HPLC) with fluorimetric detection, according to
the method of Bergamo and colleagues (25). Cellular pellets were
extracted with 250 ml of Milli-Q water in an ultrasonic bath for 30
minutes after the addition of 250 ml of cold 10% tricloracetic acid
(TCA). Samples were vigorously mixed (three minutes) and
centrifuged (5 minutes, 10000 × g). The supernatant was added to
700 ml of thiobarbituric acid prepared using thiobarbituric acid in 2
M acetate buffer at pH 3, degassing by a vacuum pump (5 minutes),
and flushing the final solution with nitrogen for 10 minutes. The
mixtures were degassed and then incubated for 30 minutes at 90˚C.
At the end of the incubation period, samples were cooled,
centrifuged (5 minutes, 10000 × g) to remove particulate material
and, finally, sample aliquots (20 ml) were analyzed by HPLC.
Quantification of MDA was obtained from a calibration curve
constructed by injecting increasing amounts of standard MDA.
MDA concentration was expressed as mg/106cells.
Quantitative analysis of apoptotic cells by flow cytometry.
Quantitative analysis of apoptotic cells with and without treatment
in vivo 29: xxx-xxx (2015)
2
of silybin was carried out using the Annexin Apoptotic Detection
Kit II (BD Biosciences).
Briefly, MKN-28 cells were treated with silybin alone or SilPho
at the doses previously described for 24 h with or without X-XO-
induced cell damage. Cells were harvested, washed twice with cold
Phosphate Buffered Saline (PBS) and then resuspended in 1X
binding buffer at a density of 1×106 cells/ml. Cellular pellets were
subjected to annexin and propidium iodide staining at room
Federico et al: Silybin–Phosphatidylcholine Complex Combats Andoxidative Stress
3
Figure 1. Evaluation of cell viability in MKN28 (A) and HepG2 (B) cultured cells after incubation with silybin, silybin–phosphatidylcholine complex
(SilPho), silybin-900-phosphoryl-D-mannopyranoside (MannpSil), silybin-900-phosphoryl-D-galactopyranoside (GalpSil), silybin-900-phosphoryl-
D-glucopyranoside (GlcpSil), silybin-900-phosphoryl-D-Lactopyranoside (LactpSil), before and after induction of oxidative stress with xanthine-
xanthine oxidase (X-XO). The data are reported as means±SD of three experiments. The concentration of agents utilized was 50 μg and the time of
observation was 24 hours. *p<0.01 vs. Control and SilPho alone.
temperature for 15 minutes in the dark and analyzed by flow
cytometry within 1 hour after The addition of 400 μl of 1X binding
buffer. Apoptotic cells, stained with annexin and propidium iodide,
were analyzed by fluorescence activated cell sorting using a Cell
Quest 3.4 software (FACS Calibur; BD Biosciences, San Jose, CA,
USA). The apoptotic cells stained with annexin exhibited green
fluorescence, whereas the cells stained with propidium iodide
exhibited red and green fluorescence.
Experiments were conducted as it follows: I: Evaluation of
silybin, SilPho, MannpSil, GalpSil, GlcpSil and LactpSil toxicity in
MKN28 and HepG2 cultured cells under basal conditions; II:
evaluation of cell viability after the induction of oxidative stress;
III: evaluation of cell viability in MKN28 and HepG2 cultured cells
after incubation with silybin, SilPho, MannpSil, GalpSil, GlcpSil
and LactpSil and subsequent induction of oxidative stress; IV:
determination of MDA as a marker showing the induction of
oxidative stress in cultured cells after incubation with silybin alone
and with SilPho; V: quantitative analysis of apoptotic cells with and
without treatment of silybin and SilPho.
Statistical analysis. Data are expressed as the mean±SD.
Significance of differences was assessed by one-way analysis of
variance (ANOVA) and, when the F value was significant, by
Tukey-Kramer test for multiple comparisons or by Student’s t-test
for comparison between two means. Differences were considered to
be significantly different if p<0.05.
Results
Effect of oxidative stress on MKN 28 and HepG2 cell
viability. Oxidative stress was induced by incubating MKN28
and HepG2 cells with XO (10-100 mU/ml) in the presence
of its substrate (1 mM) for periods of up to 3 hours. Two-
hour incubation with X-XO (1 mM and 10-100 mU/ml)
caused a dose-dependent and significant reduction in cell
viability, as assessed by the MTT assay (Figure 1). For the
subsequent experiments, a concentration of X-XO of 1 mM
plus 50 mU/ml was selected that led to a decrease in cell
viability close to 60%.
Effect of silybin, SilPho, and new silybin glycoconjugates on
X-XO induced cell damage. Underbasal conditions, the
incubation of MKN28 and HepG2 cells with silybin,SilPho,
and new silybin glycoconjugates led to two different results
(Figure 1). SilPho and new silybin glycoconjugates did not
affect cell viability, while silybin induced cell death of about
50%, even at the lower dose used, both of MKN28 and
HepG2 cells. The pre-treatment of cells with silybin and new
silybin glycoconjugates (before X-XO incubation) did not
affect cell viability, while SilPho had a protective effect
(Figure 1). In Figure 1, the concentration of molecules
utilized and the time of observation reported are 50 μg and
24 hours, respectively. With the exception of the SilPho (see
later), the same results have been verified at 10, 25, 50, 75,
100, 200 μM and at 1-48 hours of observation (data not
shown).
As the oxidative damage induced by X-XO and the effect
of pre-treatment were similar in MKN28 and HepG2 cells
we decided to perform the following experiments only in
MKN28 cell line.
Effect of silybin and of SilPho on X-XO induced lipid
peroxidation in MKN28 cells. ROS-induced cell damage is
associated with cell membrane disruption due to lipid
in vivo 29: xxx-xxx (2015)
4
Figure 2. Effect of silybin (A) and silybin–phosphatidylcholine complex
(SilPho) (B) on Malondialdehyde (MDA) in MKN28 (A) and HepG2 (B)
cell before and after induction of oxidative stress with xanthine-xanthine
oxidase (X-XO). The data are reported as means±SD of three
experiments. *p<0.05 and **p<0.01 vs. 0 μg/ml.
peroxidation. Therefore, we hypothesized that silybin and
SilPho might prevent lipid peroxidation caused by ROS
generated by X-XO. In this light, we evaluated whether
pretreatment with silybin and SilPho was able to counteract
X-XO-increased cellular MDA, a marker of lipid
peroxidation. Exposure of MKN28 cells to X-XO caused
an approximately two-fold increase in cellular MDA
concentration compared with untreated cells (Figure 2).
Moreover, pretreatment with SilPho (Figure 2B) (25-100
μg) but not with silybin (10-50 μg) (Figure 2A)
significantly prevented X-XO-induced increase of cell
MDA. These results suggest that the protective effect of
SilPho was partially due to inhibition of ROS-induced lipid
peroxidation.
Data regarding new silybin glycoconjugates are not
reported because experimental results were similar to those
obtained with silybin.
Effect of silybin and SilPho on MKN28 cell apoptosis and
necrosis before and after induction of oxidative stress with
X-XO. The pre-incubation with SilPho showed a dose-
dependent protective effect (Table I). These effects are,
almost in part, explained by the results obtained by flow
cytometry (FACS). In fact, as reported in Table II, we
found that the two assessed substances differently affected
cell vitality. Under basal conditions, silybin induced
apoptosis and SilPho did not induce any effect. Oxidative
stress caused cell death primarily by inducing cell
necrosis. The concomitant presence of silybin and
oxidative stress enhanced the ability of the latterto induce
apoptosis. SilPho had no effects on apoptosis, but
significantly counteracted cell necrosis. The increase of
silybin and SilPho concentrations up to 100 μg did not
modify these results.
Discussion
Several reports have been published on silymarin and its
flavonolignan obtained from the seeds of milk thistle (Silybum
marianum) constituents regarding their liver-protective,
antioxidant, and free-radical scavenging activities (1, 3-5).
Silybin acts, both in vitro and in vivo, as a radical scavenger by
increasing the levels of superoxide dismutase and glutathione
peroxidase and by reducing MDA and 4-hydroxynonenal (26),
markers of lipid peroxidation. Similarly, vitamin E and
phospholipids are well-known antioxidants and the
conjugation of these three substances without any alteration in
their stability enhances antioxidant action (27, 28). The
conjugation of silybin with phospholipids was performed in
order to modify its solubility and absorption in vivo. In fact,
while silybin has very low solubility in water, its conjugation
with other substances allowed its intravenous administration
and enhanced its oral bioavailability (11). In vivo, the complex
Federico et al: Silybin–Phosphatidylcholine Complex Combats Andoxidative Stress
5
Table I. Cell viability on treatment with silybin–phosphatidylcholine complex (SilPho) and xanthine-xanthine oxidase (X-XO). SilPho protects against
X-XO-induced damage in MKN28 and HepG2 cells.
Cell viability (%)
0 μg/ml 10 μg/ml 25 μg/ml 50 μg/ml 75 μg/ml 100 μg/ml 200 μg/ml
MKN28
Control 100 - - - - - -
X-XO 60 - - - - - -
SilPho - 85 80 83 84 83 80
SilPho+X-XO - 61 64 70 67 68 66
HepG2
Control 100 - - - - - -
X-XO 60 - - - - - -
SilPho - 88 85 89 85 88 84
SilPho+X-XO - 60 63 71 68 66 65
Table II. Cell deathwith pre-incubation of cells with silybin and
silybin–phosphatidylcholine complex (SilPho) under basal conditions
and after the induction of oxidative stress.
Apoptosis (%) Necrosis (%)
Basal 2.99 1.65
Silybin, 25 μg/ml 7.52* 6.38*
SilPho, 25 μg/ml 2.89 2.22
Oxidative stress 5.50* 12.97*
Oxidative stress + silybin, 25 μg/ml 22.05* 6.40*
Oxidative stress+ SilPho, 25 μg/ml 3.69 8.41*
*p<0.05 vs. basal.
of silybin with phospholids and vitamin E (RA) is rapidly
absorbed, with a blood peak concentration at 2 hours and a
large inter-organ distribution (14).
The new silybin derivatives obtained by Zarrelli et al. have
a higher water solubility than that of silybin, with enhanced
biological efficacy, binding affinity, and overall uptake (21).
In the present study, we assessed the effects of all these
compounds on cell viability and evaluated whether silybin or
SilPho pre-treatments were able to counteract X-XO-induced
increase of intracellular MDA. Exposure of MKN-28 cells to
X-XO caused an approximately two-fold increase in MDA
level as compared to untreated cells. Moreover, pretreatment
with SilPho and silybin prevented X-XO-induced
intracellular MDA increase. This suggests that the protective
and antioxidant effect of SilPho and silybin is, at least in
part, due to inhibition of ROS-mediated lipid peroxidation.
In vitro studies revealed that flavonoids can have
considerable antioxidant activity in a wide range of chemical
oxidation systems (29, 30). In our study, silybin and SilPho
exhibited powerful spontaneous antioxidant capacity in
human gastric and liver cells. Moreover, we evaluated the
protective effect on X-XO induced injury in MKN-28 cells
line measuring cell viability, and we found that only SilPho
had a dose-dependent protective effect. It is likely that
phospholipids have a protective effect against X-XO-induced
cell death by stabilizing plasma membranes.
In our experimental system, cell death induced by
oxidative stress followed two different patterns. The first led
to necrosis, a typical consequence of acute metabolic
perturbation, and the second to apoptosis, the consequence
of programmed death (31). Silybin enhanced X-XO-induced
apoptosis and reduced X-XO-mediated necrosis, whereas
SilPho significantly counteracted only cell necrosis.
Previously, we demonstrated that RA induced a
normalization of circulating lipids in patients with non-
alcoholic steatohepatitis, probably by improving liver
function (32).
In conclusion, our results show that both silybin and
SilPho act as antioxidants in an in vitro cell system, reducing
MDA levels induced by oxidative stress. Moreover, SilPho
protects MKN-28 cells from X-XO-induced cell death, being
more active than silybinin protecting cells from oxidative
stress.
Con fli ct of Interest S tatement
All of authors have declared no personal or family conflicts of
interest in regard to this study. This study was not funded.
References
1 Gazák R, Walterová D and Kren V: Silybin and silymarin-new
and emerging applications in medicine. Curr Med Chem 14:
315-338, 2007.
2 Kroll DJ, Shaw HS and Oberlies NH: Milk Thistle
nomenclature: why it matters in cancer research and
pharmacokinetic studies. Integr Cancer Ther 6: 110-119, 2007.
3 Kravchenko LV, Morozov SV and Tutel'yan VA: Effects of
flavonoids on the resistance of microsomes to lipid peroxidation
in vitro and ex vivo. Bull Exp Biol Med 136: 572-575, 2006.
4 Schumann J, Prockl J, Kiemer AK, Vollmar AM, Bang R and
Tiegs G: Silibinin protects mice from T-cell-dependent liver
injury. J Hepatol 39: 333-340, 2003.
5 Trappoliere M, Caligiuri A, Schmid M, Bertolani C, Failli P,
Vizzutti F, Novo E, di Manzano C, Marra F, Loguercio C and
Pinzani M: Silybin, a component of sylimarin, exerts anti-
inflammatory and anti-fibrogenic effects on human hepatic
stellate cells. J Hepatol 50: 1102-1111, 2009.
6 Mallikarjuna G, Dhanalakshmi S, Singh RP, Agarwal C and
Agarwal R: Silibinin protects against photocarcinogenesis via
modulation of cell cycle regulators, mitogen-activated protein
kinases, and Akt signaling. Cancer Res 64: 6349-6356, 2004.
7 Tyagi A, Agarwal C, Harrison G, Glode LM and Agarwal R:
Silibinin causes cell cycle arrest and apoptosis in human bladder
transitional cell carcinoma cells by regulating CDKI-CDK-cyclin
cascade, and caspase 3 and PARP cleavages. Carcinogenesis 25:
1711-1720, 2004.
8 Yoo HG, Jung SN, Hwang YS, Park JS, Kim MH, Jeong M, Ahn
SJ, Ahn BV, Shin BAPark RK and Jung YD: Involvement of
NFkB and caspases in silibinin-induced apoptosis of endothelial
cells. Int J Mol Med 13: 81-86, 2004.
9 Sharma G, Singh RP, Chan DC and Agarwal R: Silibinin induces
growth inhibition and apoptotic cell death in human lung
carcinoma cells. Anticancer Res 23: 2649-2655, 2003.
10 Wen Z, Dumas TE, Schrieber SJ, Hawke RL, Fried MW and
SmithPC: Pharmacokinetics and metabolic profile of free,
conjugated, and total silymarin flavonolignans in human plasma
after oral administration of milk thistle extract. Drug Metab
Dispos 36: 65-72, 2008.
11 Yanyu X,Yunmei S, Zhipeng C and Qineng P: The preparation
of silybin-phospholipid complex and the study on its
pharmacokinetics in rats. Int J Pharm 307: 77-82, 2006.
12 Jia LJ, Zhang DR, Li ZY, Feng FF, Wang YC, Dai WT, Duan CX
and Zhang Q: Preparation and characterization of silybin-loaded
nanostructured lipid carriers. Drug Deliv 17: 11-18, 2010.
13 Flaig TW, Gustafson DL, Su LJ, Zirrolli JA, Crighton F,
Harrison GS, Pierson AS, Agarwal R and Glodé LM: A phase I
and pharmacokinetic study of silybin–phytosome in prostate
cancer patients. Invest New Drugs 25: 139-146, 2007.
14 Kidd PM: Bioavailability and activity of phytosome complexes
from botanical polyphenols: the silymarin, curcumin, green tea,
and grape seed extracts. Altern Med Rev 14: 226-246, 2009.
15 Lirussi F, Beccarello A, Zanette G, De Monte A, Donadon V,
Velussi M and Crepaldi G: Silybin-beta-cyclodextrin in the
treatment of patients with diabetes mellitus and alcoholic liver
disease. Efficacy study of a new preparation of an anti-oxidant
agent. Diabetes Nutr Metab 15: 222-231, 2002.
16 Lucena MI, Andrade RJ, de la Cruz JP, Rodriguez-Mendizabal
M, Bianco E and Sánchez de la Cuesta F: Effects of silymarin
MZ-80 on oxidative stress in patients with alcoholic cirrhosis.
Results of a randomized, double-blind, placebo-controlled
clinical study. Int J ClinPharmacol Ther 40: 2-8, 2002.
17 Valenzuela A, Lagos C, Schmidt K and Videla LA: Silymarin
pro¬tection against hepatic lipid peroxidation induced by acute
in vivo 29: xxx-xxx (2015)
6
ethanol intoxication in the rat. Biochem Pharmacol 34: 2209-
2212, 1985.
18 Loguercio C, Andreone P, Brisc C, Brisc MC, Bugianesi E,
Chiaramonte M, Cursaro C, Danila M, de Sio I, Floreani A,
Freni MA, Grieco A, Groppo M, Lazzari R, Lobello S, Lorefice
E, Margotti M, Miele L, Milani S, Okolicsanyi L, Palasciano G,
Portincasa P, Saltarelli P, Smedile A, Somalvico F, Spadaro A,
Sporea I, Sorrentino P, Vecchione R, Tuccillo C, Del Vecchio
Blanco C and Federico A: Silybin combined with
phosphatidylcholine and vitamin E in patients with nonalcoholic
fatty liver disease: A randomized controlled trial. Free Radic
Biol Med 52: 1658-1665, 2012.
19 Di Sario A, Bendie E and Taffetani S: RealSil, a new sylibin-
phosphatidylcholine complex, reduces dimethylnitrosamine-
induced hepatic fibrosis in the rat. J Hepatol 38: P76, 2003.
20 Jia JD, Bauer M, Cho JJ, Ruehl M, Milani S, Boigk G, Riecken
EO and Schuppan D: Antifibrotic effect of silymarin in rat
secondary biliary fibrosis is mediated by down-regulation of
procollagen alpha1(I) and TIMP-1. J Hepatol 35: 392-398, 2001.
21 Zarrelli A, Romanucci V, Tuccillo C, Federico A, Loguercio C,
Gravante R and Di Fabio G: New Silibinin glyco-conjugates:
Synthesis and evaluation of antioxidant properties. Bioorg Med
Chem Lett 24: 5147-5149, 2014.
22 Alía M, Ramos S, Mateos R, Bravo L and Goya L: Quercetin
protects human hepatoma cell line (HepG2) against oxidative
stress induced by tert-butyl hydroperoxide.Toxicol Appl
Pharmacol 212: 110-118, 2006.
23 Romano M, Razandi M, Sekhon S, Krause WJ and Ivey KJ:
Human cell line for study of damage to gastric epithelial cells
in vitro. J Lab Clin Med 111: 430-440, 1988.
24 Ohkawa H, Ohishi N and Tagi K: Assay for lipid peroxides in
animal tissues by thiobarbituric acid reaction. Anal Biochem 95:
351-358, 1979.
25 Bergamo P, Fedele E, Balestrieri M, Abrescia P and Ferrara L:
Measurement of malondialdehyde levels in food by high-
performance liquid chromatography with fluorometric detection.
J Agric Food Chem 46: 2171-2176, 1998.
26 Wang YK, Hong YJ and Huang ZQ: Protective effects of silybin
on human umbilical vein endothelial cell injury induced by
H2O2 in vitro. Vascul Pharmacol 43: 198-206, 2005.
27 Cheng Y, Zhao Q, Liu X, Araki S, Zhang S and Miao J:
Phosphatidylcholine-specific phospholipase C, p53 and ROS in
the association of apoptosis and senescence in vascular
endothelial cells. FEBS Lett 580: 4911-4915, 2006.
28 Gavazza MB and Catalá A: The effect of alpha-tocopherol on
lipid peroxidation of microsomes and mitochondria from rat
testis. Prostaglandins Leukot Essent Fatty Acids 74: 247-254,
2006.
29 Chen JW, Zhu ZQ, Hu TX and Zhu DY: Structure–activity
relationship of natural flavonoids in hydroxyl radical-scavenging
effects. Acta Pharmacol Sin 23: 667-672, 2002.
30 Dugas AJ Jr, Castañeda-Acosta J, Bonin GC, Price KL, Fischer
NH and Winston GW: Evaluation of the total peroxyl radical-
scavenging capacity of flavonoids: structure-activity
relationships. J Nat Prod 63: 327-331, 2002.
31 Kim R, Emi M and Tanabe K: The Role of apoptosis in cancer
cell survival and therapeutic outcome. Cancer Biol Ther 5: 1429-
1442, 2006.
32 Stiuso P, Scognamiglio I, Murolo M, Ferranti P, De Simone C,
Rizzo MR, Tuccillo C, Caraglia M, Loguercio C and Federico
A: Serum oxidative stress markers and lipidomic profile to detect
NASH patients responsive to an antioxidant treatment: a pilot
study. Oxid Med Cell Longev 2014: 169216, 2014.
Received May 15, 2015
Revised July 6, 2015
Accepted July 8, 2015
Federico et al: Silybin–Phosphatidylcholine Complex Combats Andoxidative Stress
7
July 8, 2015
Dr.
A. Federico
Re: Your manuscript No. 3272-F entitled «Silybin–Phosphatidylcholine...»
Dear
Dr
Referring to your above manuscript for publication in in vivo, please allow us to use this form letter in reply:
1. Referee’s recommendations:
<Urgent to be published immediately.
<<Accepted in the presented form.
<<Accepted with minor changes.
<Accepted with grammatical or language corrections.
<<Remarks:
2. Excess page charges.
<<Your article has approx. xxx printed pages and is in excess of the allotted number by approx. xx
printed pages. The charges are EURO xxx per excess page, totalling EURO xxx
We ask you to confirm acceptance of these charges.
<<Your article includes xxx pages with color figures. The charges are EURO xxx per color page,
totalling EURO xxxx
<<Our invoice will be sent by e-mail to the corresponding author.
3. <Your article will appear in Volume 29, Issue No. 5, 2015
4. <Please order your reprints, pdf or online open access, now. This will facilitate our prompt planning of
future issues and rapid publication of your article. Reprints will be delivered by rapid one-day delivery
within one month from publication.
We would appreciate your prompt reply.
With many thanks,
Yours sincerely,
J.G. Delinasios
Managing Editor
EDITORIAL OFFICE: INTERNATIONAL INSTITUTE OF ANTICANCER RESEARCH
DELINASIOS G.J. & CO G.P., Kapandriti, P.O.B. 22, Attiki 19014, Greece. Tel. : 0030-22950-52945;
Tel & Fax:0030-22950-53389; e-mail: journals@iiar-anticancer.org
International Journal of Experimental and Clinical Pathophysiology and Drug Research
ISSN (print): 0258-851X, ISSN (online): 1791-7549
Please type or print the requested information on the reprint order form and return it to the Editorial Office
by fax or e-mail. Fees for reprints, PDF file, or online open access must be paid for in advance.
If your paper is subject to charges for excess pages or color plates, please add these charges to the payment for
reprints. The reprints are not to be sold.
PRICE LIST FOR REPRINTS WITHOUT COVER
Online Number of copies requested (prices are in Euro)
Page Open PDF 50 100 200 300 400 500 1000 1500 2000 3000
length Access File
Fee*Fee
(Euro)(Euro)
1-4pp 400 175 140 285 337 388 453 504 851 1135 1470 2038
5-8 500 225 150 388 453 530 595 672 1083 1445 1832 2554
9-12 600 277 160 504 569 659 737 827 1341 1780 2219 3096
13-16 700 354 180 659 737 840 943 1046 1625 2141 2657 3676
17-20 800 419 200 788 879 982 1098 1227 1883 2451 3044 4244
*Online open access of an article published in 2015 is accompanied by a complimentary online
subscription to in vivo.
For reprints with cover: Please add EURO 80.00 per 100 copies.
Postage: Please add 5% on the reprint prices.
Reprint Order Form
Of my paper No. 3272-F comprising 7printed pages, entitled
«Silybin–Phosphatidylcholine...»
accepted for publication in in vivo Vol. 29 No. 5
<<
I require a total of copies at EURO:
<<
I do not require reprints.
<<
Please send me a PDF file of the article at EURO:
<<
Please provide Online Open Access of the article at the Stanford University Highwire Press website
(at Euro ) immediately upon publication, and enter my complimentary online subscription to in vivo.
<<
Please send me a copy of the issue containing my paper at EURO 25.00.
<<
Please enter my personal subscription to in vivo at the special Author’s price of
EURO 190.00 (
<<
print;
<<
online) (
<<
Year: 2015).
<<
A check for the above amounts payable to DELINASIOS G.J. & CO G.P., is enclosed.
<<
Please send an invoice to (Billing Name and Address):
VAT number (For EC countries):
Name:
Address: Signature:
Country: City:
Postal code: e-mail:
Tel: Fax:
<<
Please send reprints to (Complete Address and Tel. no.):
Editorial Office: International Institute of Anticancer Research,
DELINASIOS G.J. & CO G.P., Kapandriti, P.O.B. 22, Attiki 19014, Greece
Fax: +30-22950-53389; Tel: +30-22950-52945; e-mail: journals@iiar-anticancer.org
in vivo International Journal of Experimental and Clinical
Pathophysiology and Drug Research
ISSN (print): 0258-851X, ISSN (online): 1791-7549
... Phosphatidylcholine (PC), a polyunsaturated fatty acid, is a component of biological membranes and can be obtained in the diet from various resources such as egg yolk, mustard, sunflower, and soybean [9]. PC has been found to have anti-inflammatory [10], antioxidant [11,12], and anti-Alzheimer's [13] effects. The role of PC on oxidative stress has been reported in cell and animal experiments [11,12], but the role of PC in kidney and liver cells based on AGEs damage have not been reported. ...
... PC has been found to have anti-inflammatory [10], antioxidant [11,12], and anti-Alzheimer's [13] effects. The role of PC on oxidative stress has been reported in cell and animal experiments [11,12], but the role of PC in kidney and liver cells based on AGEs damage have not been reported. ...
Article
Full-text available
Background and Objectives: Receptors of the advanced glycation products (RAGE) are activated to promote cell death and contributes to chronic diseases such as diabetes and inflammation. Advanced glycation end products (AGEs), which interact with RAGE are complex compounds synthesized during diabetes development and are presumed to play a significant role in pathogenesis of diabetes. Phosphatidylcholine (PC), a polyunsaturated fatty acid found in egg yolk, mustard, and soybean, is thought to exert anti-inflammatory activity. We investigated the effects of PC on AGEs-induced hepatic and renal cell injury. Materials and Methods: In this study, we evaluated cytokine and NF-κB/MAPK signal pathway activity in AGEs induced human liver (HepG2) cells and human kidney (HK2) cells with and without PC treatment. Results: PC reduced RAGE expression and attenuated levels of inflammatory cytokines and NF-kB/MAPK signaling. Moreover, cells treated with PC exhibited a significant reduction in cytotoxicity, oxidative stress, and inflammatory factor levels. Conclusions: These findings suggest that PC could be an effective functional material for hepatic and renal injury involving with oxidative stress caused by AGEs during diabetic conditions.
... Согласно данным доклинических исследований, в такой комбинации не только значительно увеличивается биодоступность силибинина, но и обеспечивается взаимопотенцирование антиоксидантного эффекта, достигается коррекция митохондриальной дисфункции при НАСГ, уменьшается выраженность поражения печени при амиодарон-индуцированном стеатозе [79][80][81]. Отмечается, что добавление фосфатидилхолина компенсирует нежелательный проапоптогенный эффект силибинина в условиях оксидативного стресса [80]. ...
... Согласно данным доклинических исследований, в такой комбинации не только значительно увеличивается биодоступность силибинина, но и обеспечивается взаимопотенцирование антиоксидантного эффекта, достигается коррекция митохондриальной дисфункции при НАСГ, уменьшается выраженность поражения печени при амиодарон-индуцированном стеатозе [79][80][81]. Отмечается, что добавление фосфатидилхолина компенсирует нежелательный проапоптогенный эффект силибинина в условиях оксидативного стресса [80]. ...
Article
Full-text available
В клинической практике нередко используются одновременно несколько гепатопротективных средств в виде отдельных препаратов или фиксированных комбинаций, выпускаемых фармацевтической промышленностью. Комбинированное применение может обеспечивать как усиление того или иного фармакологического эффекта, так и расширение спектра гепатотропного действия. Наиболее часто в комбинациях изучаются и применяются эссенциальные фосфолипиды, глицирризиновая кислота, урсодезоксихолевая кислота, силибинин и S-аденозилметионин. В работе обсуждены патогенетические обоснования комбинированного использования гепатотропных лекарственных средств и прове-ден анализ результатов доклинических и клинических исследований, посвященных использованию комбинаций при различных заболеваниях печени. Fixed-dose drug products as well as non-fi xed hepatoprotective drug combinations are commonly used in modern clinical practice. Combined and concurrent drug use makes it possible to augment the pharmacological eff ects of individual agents, or extend the range of their potential indications. The drugs most commonly considered for combination therapy include essential phospholipids, glycyrrhizinic acid, ursodeoxycholic acid, silibinin, and S-adenosylmethionine. This paper discusses the rationale for combined use of liver- targeting drugs from a pathogenetic viewpoint, and provides a review of the evidence from clinical trials on combined pharmacotherapy for liver disease.
... Silybin exhibits the ability to deactivate pro-inflammatory signals associated with NF-κB activation, which is implicated in the synthesis of cytokines like TNF-α, IL-1, and IL-6 (Polyak et al., 2010). This effect is attributed to its dual capability as a free radical scavenger and inhibitor of lipid peroxidation, as demonstrated in both in vitro and in vivo studies (Nencini et al., 2007 andFederico et al., 2015). According to Bhattacharya (2011), silymarin inhibits the inflammatory process by impeding the migration of neutrophils and Kupfer cells. ...
... Silybin exhibits the ability to deactivate pro-inflammatory signals associated with NF-κB activation, which is implicated in the synthesis of cytokines like TNF-α, IL-1, and IL-6 (Polyak et al., 2010). This effect is attributed to its dual capability as a free radical scavenger and inhibitor of lipid peroxidation, as demonstrated in both in vitro and in vivo studies (Nencini et al., 2007 andFederico et al., 2015). According to Bhattacharya (2011), silymarin inhibits the inflammatory process by impeding the migration of neutrophils and Kupfer cells. ...
... Previous reports have shown that obesity promotes fatty acid accumulation and inflammation in the liver, leading to liver injury (Marengo et al., 2016). Obesity is associated with an increased risk of liver cancer development and progression (Federico et al., 2015). Serum levels of pro-tumor cytokines and adipokines (e.g., leptin, IL-6, VEGF, IL-8, resistin and endolipin) are higher in obese males than in overweight (OW) and normal weight (NW) males (Steppan et al., 2001;Ramirez et al., 2012). ...
Article
Full-text available
Nonalcoholic steatohepatitis (NASH) is a clinical syndrome with pathological changes that are similar to those of alcoholic hepatitis without a history of excessive alcohol consumption. It is a specific form of nonalcoholic fatty liver disease (NAFLD) that is characterized by hepatocyte inflammation based on hepatocellular steatosis. Further exacerbation of NASH can lead to cirrhosis, which may then progress to hepatocellular carcinoma (HCC). There is a lack of specific and effective treatments for NASH and NASH-driven HCC, and the mechanisms of the progression of NASH to HCC are unclear. Therefore, there is a need to understand the pathogenesis and progression of these diseases to identify new therapeutic approaches. Currently, an increasing number of studies are focusing on the utility of natural products in NASH, which is likely to be a promising prospect for NASH. This paper reviews the possible mechanisms of the pathogenesis and progression of NASH and NASH-derived HCC, as well as the potential therapeutic role of natural products in NASH and NASH-derived HCC.
... However, the use of silibinin is limited by its low solubility in water (0.4 mg/L) [22] that involves also in a low bioavailability [23,24]. Several research groups are involved in solving these problems using a pro-drug approach with the insertion of phosphate groups that considerably increases the water solubility of drug [25][26][27] or through the use of liposomes [28]. In addition to silymarin, the fruit contains other active substances with a protective action such as flavonoids, antioxidants, proteins, oleic, and linolenic acids, but above all sterols. ...
Article
Full-text available
The use of Silybum marianum L. for therapeutic purposes has been known since ancient times. Its phytocomplex reduces transaminases and other biohumoral indices in the course of liver disease and also in hepato-renal syndrome. In particular, the flavonolignan component has shown properties that would partially explain the ability of the phytocomplex to induce a certain regeneration of liver cells, stimulate the cellular elimination of toxins and reduce the inflammatory component, present in fatty, alcoholic and hormonal therapies with steroids. S. marianum is also successfully used in the treatment of patients with symptomatic chronic hepatitis, with complete disappearance of clinical symptoms, such as asthenia, loss of appetite, severe meteorism, dyspepsia, and with normalization of transaminases. The same results can be obtained in patients undergoing heavy chemotherapy cycles. Modern herbal medicine uses it in decoction or infusion, however with some caution in patients suffering from hypertension, due to the presence of tyramine. In addition, the extracts of the roots have antioxidant, diuretic and febrifugal properties and those of the leaves have aperitif properties. It is therefore interesting to provide a picture of the different non-flavonolignanic components (terpenes, steroids and essential oils) of the plant and their properties, which have perhaps been wrongly neglected over the past few years.
... Owing to its phenolic existence, it can give electrons to stabilize free radicals and reactive oxygen molecules [63]. Silymarin also modulates intracellular glutathione, which inhibits membranes from lipid peroxidation [64]. Silymarin also has antiviral effects as it affects RNA and DNA synthesis [65]. ...
Article
Full-text available
COVID-19 is known as one of the deadliest pandemics of the century. The rapid spread of this deadly virus at incredible speed has stunned the planet and poses a challenge to global scientific and medical communities. Patients with COVID-19 are at an increased risk of co-morbidities associated with liver dysfunction and injury. Moreover, hepatotoxicity induced by antiviral therapy is gaining importance and is an area of great concern. Currently, alternatives therapies are being sought to mitigate hepatic damage, and there has been growing interest in the research on bioactive phytochemical agents (nutraceuticals) due to their versatility in health benefits reported in various epidemiological studies. Therefore, this review provides information and summarizes the juncture of antiviral, immunomodulatory, and hepatoprotective nutraceuticals that can be useful during the management of COVID-19.
... Furthermore, it was recently revealed that the treatment of breast cancer cells with silibinin results in the induction of apoptosis and the attenuation of both miR-21 and miR-155 levels [61] (Figure 2 and Table 3). Silibinin and its derivative (e.g., glyco-conjugated silibinin) have antioxidant activities and can protect gastric and hepatocellular carcinoma cells from xanthine oxidase-induced oxidative stress [62,63]. Therefore, it is still further required to investigate the effects of silibinin on different cancer cell types. ...
Article
Full-text available
Cancer is a global health concern and one of the main causes of disease-related death. Even with considerable progress in investigations on cancer therapy, effective anti-cancer agents and regimens have thus far been insufficient. There has been compelling evidence that natural phytochemicals and their derivatives have potent anti-cancer activities. Plant-based anti-cancer agents, such as etoposide, irinotecan, paclitaxel, and vincristine, are currently being applied in medical treatments for patients with cancer. Further, the efficacy of plenty of phytochemicals has been evaluated to discover a promising candidate for cancer therapy. For developing more effective cancer therapy, it is required to apprehend the molecular mechanism deployed by natural compounds. MicroRNAs (miRNAs) have been realized to play a pivotal role in regulating cellular signaling pathways, affecting the efficacy of therapeutic agents in cancer. This review presents a feature of phytochemicals with anti-cancer activity, focusing mainly on the relationship between phytochemicals and miRNAs, with insights into the role of miRNAs as the mediators and the regulators of anti-cancer effects of phytochemicals.
Article
Objectives Contamination of surface waters is a major health threat for all living creatures. Some types of blue-green algae that naturally occur in fresh water, are able to produce various toxins, like Microcystins (MCs). Microcystin-leucine arginine (MC-LR) produced by Microcystis aeruginosa is the most toxic and abundant isoforms of MCs, and it causes hepatotoxicity. The present article reviews preclinical experiments examined different treatments, including herbal derivatives, dietary supplements and drugs against MC-LR hepatotoxicity. Methods We searched scientific databases Web of Science, Embase, Medline (PubMed), Scopus, and Google Scholar using relevant keywords to find suitable studies until November 2023. Results MC-LR through Organic anion transporting polypeptide superfamily transporters (OATPs) penetrates and accumulates in hepatocytes, and it inhibits protein phosphatases (PP1 and PP2A). Consequently, MC-LR disturbs many signaling pathways and induces oxidative stress thus damages cellular macromolecules. Some protective agents, especially plants rich in flavonoids, and natural supplements, as well as chemoprotectants were shown to diminish MC-LR hepatotoxicity. Conclusion The reviewed agents through blocking the OATP transporters (nontoxic nostocyclopeptide-M1, captopril, and naringin), then inhibition of MC-LR uptake (naringin, rifampin, cyclosporin-A, silymarin and captopril), and finally at restoration of PPAse activity (silybin, quercetin, morin, naringin, rifampin, captopril, azo dyes) exert hepatoprotective effect against MC-LR.
Article
Prodrugs are ingenious derivatives of therapeutic agents designed to improve the pharmacokinetic profile of the drug. Here, we report an efficient and regioselective solid phase approach for obtaining new prodrugs of 9“-silybins conjugated with 3'-ribonucleotide units (uridine and adenosine) as pro-moieties. Uridine and adenosine conjugates were obtained in good yields (41-50%), beginning with silibinin and its diastereomers (silybin A and silybin B), using a NovaSyn® support functionalized with an ad hoc linker, which allowed selective detachment of only the desired products. As expected, the solubility of both uridine and adenosine conjugates was higher than that of the parental natural product (5 mg/mL and 3 mg/mL for uridine and adenosine, respectively). Our investigations revealed that uridine conjugates were quickly cleaved by RNase A, releasing silybin drugs, even at low enzyme concentrations. No toxic effects were found for any ribonucleotide conjugate on differentiated neuroblastoma SH-SY5Y cells when tested at increasing concentrations. All results strongly encourage further investigations of uridine-silybin prodrugs as potential therapeutic agents for both oral and intravenous administration. The present synthetic approach represents a valuable strategy to the future design of new prodrugs with modified nucleoside pro-moieties to modulate the pharmacokinetics of silybins or different natural products with strong pharmacological activities but poor bioavailability. 2009 Elsevier Ltd. All rights reserved.
Article
Full-text available
Liver steatosis can evolve to steatohepatitis (NASH) through a series of biochemical steps related to oxidative stress in hepatocytes. Antioxidants, such as silybin, have been proposed as a treatment of patients with nonalcoholic fatty liver disease (NAFLD) and NASH. In this study, we evaluated, in patients with histologically documented NASH, the oxidant/antioxidant status and lipid "fingerprint" in the serum of NASH patients, both in basal conditions and after 12 months of treatment with silybin-based food integrator Realsil (RA). The oxidant/antioxidant status analysis showed the presence of a group of patients with higher basal severity of disease (NAS scores 4.67 ± 2.5) and a second group corresponding to borderline NASH (NAS scores = 3.8 ± 1.5). The chronic treatment with RA changed the NAS score in both groups that reached the statistical significance only in group 2, in which there was also a significant decrease of serum lipid peroxidation. The lipidomic profile showed a lipid composition similar to that of healthy subjects with a restoration of the values of free cholesterol, lysoPC, SM, and PC only in group 2 of patients after treatment with RA. Conclusion. These data suggest that lipidomic and/or oxidative status of serum from patients with NASH could be useful as prognostic markers of response to an antioxidant treatment.
Article
Full-text available
The only currently recommended treatment for nonalcoholic fatty liver disease (NAFLD) is lifestyle modification. Preliminary studies of silybin showed beneficial effects on liver function. Realsil (RA) comprises the silybin phytosome complex (silybin plus phosphatidylcholine) coformulated with vitamin E. We report on a multicenter, phase III, double-blind clinical trial to assess RA in patients with histologically documented NAFLD. Patients were randomized 1:1 to RA or placebo (P) orally twice daily for 12 months. Prespecified primary outcomes were improvement over time in clinical condition, normalization of liver enzyme plasma levels, and improvement of ultrasonographic liver steatosis, homeostatic model assessment (HOMA), and quality of life. Secondary outcomes were improvement in liver histologic score and/or decrease in NAFLD score without worsening of fibrosis and plasma changes in cytokines, ferritin, and liver fibrosis markers. We treated 179 patients with NAFLD; 36 were also HCV positive. Forty-one patients were prematurely withdrawn and 138 patients analyzed per protocol (69 per group). Baseline patient characteristics were generally well balanced between groups, except for steatosis, portal infiltration, and fibrosis. Adverse events (AEs) were generally transient and included diarrhea, dysgeusia, and pruritus; no serious AEs were recorded. Patients receiving RA but not P showed significant improvements in liver enzyme plasma levels, HOMA, and liver histology. Body mass index normalized in 15% of RA patients (2.1% with P). HCV-positive patients in the RA but not the P group showed improvements in fibrogenesis markers. This is the first study to systematically assess silybin in NAFLD patients. Treatment with RA but not P for 12 months was associated with improvement in liver enzymes, insulin resistance, and liver histology, without increases in body weight. These findings warrant further investigation.
Article
A number of discoveries have clarified the molecular mechanism of apoptosis, thus clarifying the link between apoptosis and therapeutic outcome. Even though apoptosis is thought to play a major role in anticancer therapy, the clinical relevance of induction of apoptosis remains uncertain, particularly in solid tumors. Induction of apoptosis by anticancer agents has been shown to correlate with tumor response, however, non-apoptotic forms of cell death, such as autophagy and extrinsic senescence, have also been shown to contribute to the overall tumor response. Cellular damage induces growth arrest and tumor suppression by inducing apoptosis, necrosis, and senescence; the mechanism of cell death depends on the magnitude of DNA damage following exposure to various concentrations of anticancer agents. Apoptosis-resistant cells and transduction pathways which inhibit apoptosis can induce non-apoptotic mechanisms of cell death and senescence, thereby preserving the antitumor effect of some anticancer agents. Heterogeneic antitumor responses include various cell types of cell death, depending on the degree of cellular or DNA damage incurred by cancer cells. As a new therapeutic strategy, alternative types of cell death might be exploited to control and eradicate cancer cells. This review discusses the clinical significance of apoptosis, as well as the potential contribution of other types of cell death to overall tumor sensitivity in the hopes that new therapeutic strategies might follow.
Article
Background/Aims: Silymarin reduces hepatic collagen accumulation by 35% in rats with secondary biliary cirrhosis. The aim of the present study was to explore its antifibrotic mechanism.Methods: Thirty female adult Wistar rats were allocated to (1) bile duct occlusion, (2) bile duct occlusion and oral silymarin at 50 mg/kg per day, and (3) sham operation and oral silymarin at 50 mg/kg per day. Steady-state mRNA levels for procollagen α1(I), tissue inhibitor of metalloproteinases-1 (TIMP-1), and transforming growth factor (TGF) β1 were determined by multi-probe ribonuclease protection assay.Results: After 6 weeks of bile duct occlusion, liver collagen content was increased 12-fold, when compared with the sham-operated controls. These animals displayed 17-, 6.5- and 16-fold higher transcript levels for procollagen α1(I), TIMP-1 and TGFβ1 (P
Article
A sensitive and reproducible HPLC assay with fluorometric detection was used to measure the malondialdehyde (MDA) concentration in food (butter, margarine, oil, fish, and meat tissue). Samples were homogenized in water supplemented with butylated hydroxytoluene. Proteins were precipitated with ice-cold 5% trichloroacetic acid and removed by centrifugation. The supernatant was incubated in a 0.28% thiobarbituric acid (TBA) mixture from which the oxygen was depleted. Optimal incubation time and temperature, for the TEA treatment, were found to be 30 min and 90 degrees C, respectively. The MDA-TBA adduct was fractionated by reverse phase HPLC and detected by fluorescence (lambda(EX) = 515 nn; lambda(EM) = 543 nm). Elution was performed at 1 mL/min flow rate with a mixture of acetonitrile and sodium phosphate at pH 7 (15:85 v/v). The described sample preparation procedure minimizes the lipid oxidation and provides high sensitivity (0.01 pmol of MDA), reproducibility, and specificity.
Article
Nanostructured lipid carriers (NLC) are a new generation of lipid nanoparticles, which are produced by controlled mixing of solid lipids with spatially incompatible liquid lipids leading to special nanostructures with improved drug incorporation and release properties. In this study, silybin-loaded nanostructured lipid carriers with various liquid lipid content were successfully prepared by the method of emulsion evaporation at a high temperature and solidification at a low temperature. The size and morphology of nanoparticles were significantly influenced by the liquid lipid content. As the liquid lipid content increased to 20 wt%, the obtained particles showed distinguished smaller size. Compared with solid lipid nanoparticles (SLN), NLC presented improved drug loading capacity which increased with increasing the liquid lipid content. The differential scanning calorimetry (DSC) and X-ray diffraction (XRD) analysis indicated that the incorporation of liquid lipids could interfere with the crystallization of solid lipids. The drug in vitro release behavior from NLC displayed a biphasic drug release pattern with burst release at the initial stage and prolonged release afterwards, and the successful controlled release rate can be achieved by controlling the liquid lipid content.