Вы находитесь на странице: 1из 6

Physiological Responses of a Natural Antioxidant

Victor J. Johnson1
Flavonoid Mixture, Silymarin, in BALB/c Mice: Quanren He1
III. Silymarin Inhibits T-Lymphocyte Function at Low Doses but Stimulates Marcin F. Osuchowski1, 2
Inflammatory Processes at High Doses Raghubir P. Sharma2
Original Paper

Abstract were observed but only the CD4+ population in mice treated
with 10 mg/kg of silymarin was significantly different from con-
Silymarin is a mixture of bioactive flavonoids isolated from Milk trol. Functional examination of secondary lymphoid cells re-
Thistle (Silybum marianum). Crude extracts from this plant have vealed that phytohemagglutinin-induced T-lymphocyte proli-
been used for centuries as a natural remedy and silymarin is now feration was increased in the lowest dose group only. B-lympho-
effectively used in the treatment of inflammatory liver toxicity cyte blastogenesis induced by lipopolysaccharide was increased
and disease in humans. In vitro studies show that silymarin can following exposure to 10 and 50 mg/kg of silymarin. Similarly,
inhibit the production and damage caused by tumor necrosis fac- expression of TNFa, inducible nitric oxide synthase, IL-1b and
tor a (TNFa) and is a potent antioxidant both in vitro and in vivo. IL-6 mRNA were increased dose-dependently. The expression of
Such findings suggest silymarin may impact the immune system IL-2 and IL-4 were reduced in mice treated with 10 and 50 mg/kg
but little information exists following in vivo exposure. There- of silymarin although only the 10 mg/kg group was significantly
fore, we tested the hypothesis that exposure to silymarin will different from control. The results indicate that in vivo parenteral
modulate the inflammatory immune response. Male BABL/c exposure to silymarin results in suppression of T-lymphocyte
mice (6/group) were treated intraperitoneally once daily for five function at low doses and stimulation of inflammatory processes
days with 0, 10, 50 or 250 mg/kg of silymarin. Silymarin exposure at higher doses. Further studies investigating the effects of sily-
44 did not produce any signs of overt toxicity or any changes in rela- marin on the immune system are warranted.
tive organ weights. Flow cytometric examination of splenic lym-
phocyte populations showed that the absolute number of CD3+ Key words
T-lymphocytes was reduced in the 10 and 50 mg/kg groups al- Silymarin ´ inflammation ´ natural product ´ T-lymphocyte ´ cyto-
though significance was evident only in the 10 mg/kg group. kine ´ Silybum marianum ´ Asteraceae
Concomitant decreases in CD4+ and CD8+ T-cell populations

Introduction for years in the treatment of human liver toxicity and chronic liv-
er diseases. Pharmacological studies have indicated that even
Silymarin is a mixture of bioactive flavonoids isolated from the high doses of silymarin are not overtly toxic to patients receiving
seeds and fruits of milk thistle (Silybum marianum). This mixture therapy [4]. The biological actions of this mixture have been at-
has been recognized as a powerful antioxidant displaying hepa- tributed to several mechanisms including membrane stabiliza-
toprotective [1], chemopreventive [2] and immunomodulatory tion and inhibition of lipid peroxidation [5], elevation of gluta-
[3] properties. Crude extracts from milk thistle have been used thione and superoxide dismutase (SOD) [6] as well as free radical

Affiliation
1
Department of Physiology and Pharmacology, College of Veterinary Medicine,
The University of Georgia, Athens, GA, USA
2
Animal Anatomy, University Warmia and Mazury, Olsztyn, Poland

Correspondence
Dr. Raghubir P. Sharma ´ Department of Physiology and Pharmacology ´ College of Veterinary Medicine ´
The University of Georgia ´ Athens, GA 30602-7389, USA ´ Phone: +1-706-542-2788 ´ Fax: +1-706-542-3015 ´
E-mail: rpsharma@vet.uga.edu

Received May 28, 2002 ´ Accepted July 28, 2002


Bibliography
Planta Med 2003; 69: 44±49 ´  Georg Thieme Verlag Stuttgart ´ New York ´ ISSN 0032-0943
scavenging. The increasing use of silymarin as a natural compo- Treatment of assay groups
nent in nearly 200 natural medications warrants further investi- Silymarin was purchased from Sigma-Aldrich Chemical Compa-
gation on the toxicology of this mixture at a subclinical level, ny (St. Louis, MO). This product consists of a mixture of seven
namely the immune system. isomers including toxifolin (4 %), silichristin (27.9 %), silidianin
(2.9 %), silybin A (19.3 %), silybin B (31.3 %), isosilybin A (8.2 %)
Several in vitro studies address the effects of silymarin on com- and isosilybin B (2.3 %) as determined by high-performance li-
ponents of the immune system. Lang et al. [7] demonstrated quid chromatography with 277 nm detection. Male BALB/c
that exposure of peripheral blood cells to silymarin resulted in mice received daily intraperitoneal (i. p.) injections for 5 conse-
increased mitogen-induced lymphocyte proliferation but de- cutive days of phosphate buffered saline (PBS, vehicle control)
creased antibody-dependent cellular cytotoxicity, natural killer or 10, 50, 250 mg/kg of silymarin (Sigma, St. Louis, MS) as a sus-
cell activity and reduced the CD8+ T-cell population. Stimulation pension in PBS. This route of exposure has been used previously
of the basal rate of proliferation was also observed in splenocytes for the investigation of the hepatoprotective effects of silymar-
from silymarin-treated rats [8]. In contrast, Meroni et al. [9] in [15] and we recently employed the same protocol to examine
showed a dose-dependent inhibition of mitogen-induced lym- the effects of silymarin on normal liver [16]. One day following

Original Paper
phocyte blastogenesis following in vitro exposure to silybin, an the final injection, mice were euthanized by decapitation, trunk
active component of silymarin. In vitro exposure to silymarin blood collected and the spleen was aseptically excised and
was shown to restore function and maturation of peritoneal weighed. Peripheral blood was used for the determination of
macrophage from patients undergoing continuous ambulatory erythrocyte and leukocyte populations. Single cell suspensions
peritoneal dialysis [10]. In addition, silibinin is capable of indu- were prepared from the spleen as reported previously [17] and
cing HL-60 cell differentiation into a monocyte lineage [11]. used for phenotypic and functional analysis of lymphocyte po-
These studies suggest that silymarin may have differential ef- pulations.
fects on lymphocytes versus macrophage/monocyte type cells.
Mitogen-induced lymphocyte proliferation
Silymarin has also been shown to affect the production and re- The proliferative response of splenic lymphocytes from mice
lease of cytokines. Silymarin potently inhibited the expression of treated with silymarin in vivo was examined using the lympho-
tumor necrosis factor a (TNFa) in mouse skin exposed to tumor cyte blastogenesis assay described earlier [17]. Cultures were
promoters [12]. Silybin displays similar anti-inflammatory actions stimulated with concanavalin A (ConA; 5 mg/ml), phytohemag-
resulting in a dose-dependent inhibition of lipopolysaccharide glutinin (PHA-P; 10 mg/ml) and LPS (50 mg/ml) for 48 hours at
(LPS)-induced TNFa production in RAW264.7 macrophage cells 37 8C with 5 % CO2, and subsequently pulsed with [methyl-
3
[13]. Recent evidence demonstrates that the mechanism of TNFa H]thymidine (25 mCi/ml, 6.7 Ci/mmol, DuPont NEN Products,
inhibition is through decreased activation of nuclear factor kB Boston, MA) for an additional 18 hours. Proliferative responses
(NFkB) in cells exposed to silymarin or its components [3], [14]. It were expressed as net disintegrations per minute (DPM) and
is likely that the free radical scavenging properties of silymarin are stimulation indices (DPM with mitogen/DPM without mitogen). 45
responsible for the observed inhibition of NFkB.
Flow cytometric phenotyping of splenic lymphocyte
The vast majority of literature regarding the immune system ef- populations
fects of silymarin involved pathological or noxious stimuli. There Three-color flow cytometry was used to determine the preval-
is a lack of information regarding the immunological effects of ence of specific lymphocyte populations in the spleen as de-
this flavonoid mixture on the normal healthy immune system. scribed earlier [17]. Labeled cells were fixed in 0.5 % formalin in
The objective of the present study was to determine the effects PBS and acquired (20 000 events) using an EPICS XL-MCL flow
of sub-acute treatment with silymarin on the representation cytometer (Coulter Cytometry, Hialeah, FL) equipped with a 488
and function of lymphocytes in the peripheral immune system nm argon ion laser and Lysis II acquisition software. Analysis was
of healthy BALB/c mice. performed using WinMDI flow analysis package.

Analysis of mRNA expression


Materials and Methods Splenocytes (2.5 ” 106/ml) were stimulated with 10 mg/ml PHA-P
or 1 mg/ml LPS for six hours following which total RNA was ex-
Animal care and handling tracted using TRIzol (Invitrogen, Carlsbad, CA) according to
Inbred male BALB/c mice (Harlan, Indianapolis, IN), 7 ± 8 weeks manufacturer's protocol. Reverse-transcriptase polymerase
of age and an average body weight of 20 g were used. Mice were chain reaction (RT-PCR) was used to analyze the expression of
randomly assigned to treatment cages (6/cage) and allowed to mRNA for IL-2 and IL-4 in PHA±stimulated cultures and TNFa,
acclimate for one week in the University of Georgia Animal Re- IL-1b, IL-6 and iNOS in LPS-stimulated cultures. The expression
sources facility maintained at 21 8C with a 12-hour light/dark cy- of b-actin (internal control) was examined in both treatments.
cle. Rodent chow (Harlan Teklad, Madison, WI) and distilled wa- The conditions for reverse transcription and PCR steps were per-
ter were supplied ad libitum. Food and water consumption as formed as previously reported [17] with the exception of primer
well as body weight gain were monitored daily over the treat- sets (see Table 1). Cycle number was optimized to achieve ampli-
ment period. The care and treatment of the mice were in accord- fication within the linear range. Amplification products were
ance with the guidelines established by the Public Health Service electrophoretically separated and documented using a Kodak
Policy on Humane Care and Use of Laboratory Animals and were DC290 digital camera. The resulting images were digitized and
approved by the Institutional Animal Care and Use Committee. quantified using UN-SCAN-IT software (Silk Scientific, Inc.,

Johnson VJ et al. Physiological Responses of ¼ Planta Med 2003; 69: 44 ± 49


Table 1 Primer sets and amplifications conditions*

Cytokine Primer Sequence Annealing Temperature (8C) Cycle Number

IL-2 sense 5¢ CTCGCATCCTGTGTCACATT 3¢ 54 35


antisense 5¢ ATCCTGGGGAGTTTCAGGTT 3¢
IL-4 sense 5¢ TCAACCCCCAGCTAGTTGTC 3¢ 54 35
antisense 5¢ GGAGCTCACTCTCTGTGGTG 3¢
TNFa sense 5¢ CTCTTCAAGGGACAAGGCTG 3¢ 56 31
antisense 5¢ CGGACTCCGCAAAGTCTAAG 3¢
IL-1b sense 5¢ GCAACTGTTCCTGAACTCA 3¢ 55 25
antisense 5¢ CTCGGAGCCTGTAGTGCAG 3¢
IL-6 sense 5¢ TTCCATCCAGTTGCCTTCTT 3¢ 54 33
antisense 5¢ CAGAATTGCCATTGCACAAC 3¢
iNOS sense 5¢ CACCTTGGAGTTCACCCAGT 3¢ 52 32
Original Paper

antisense 5¢ ACCACTCGTACTTGGGATGC 3¢
b-actin sense 5¢ ATGGATGACGATATCGCT 3¢ 56 25
antisense 5¢ ATGAGGTAGTCTGTCAGGT 3¢

* Thermal cycles consisted of denaturation at 94 8C for 15 seconds, annealing (see above) for 15 seconds and extension at 72 8C for 30 seconds followed by a final extension at 72 8C for 2 minutes.

Orem, UT) and the pixel values for each cytokine were normal- Lymphocyte populations and functions
ized to that of b-actin. Splenic cellularity was not changed by silymarin treatment. Phe-
notypic analysis of lymphocyte populations revealed that sily-
Statistical analysis marin caused a decrease in CD3+ T-lymphocytes in the spleen
All statistical analyses were performed using the SAS statistical (Fig. 1a). Interestingly, the decrease was only significant at
software (SAS Institute, Cary, NC). Treatment effects were ana- 10 mg/kg of silymarin. T-lymphocyte populations were the
lyzed using one way analysis of variance (ANOVA) followed by same as control in the higher dose groups. The CD45+ B-lympho-
Fisher's PLSD. A value of P < 0.05 was considered significant un- cyte population remained unaffected by silymarin treatment
less indicated otherwise. (Fig. 1b). Further separation of the T-lymphocyte pool indicated
that the decrease in the 10 mg/kg group was mainly in the CD4+
T-helper cell population as shown in Fig. 1c. There was a decrease
Results in the CD8+ T-suppressor/cytotoxic population (Fig. 1d) although
46 statistical significance was not achieved.
Toxicity
No overt signs of clinical toxicity or behavioral alterations were Lymphocyte proliferation also proved to be more sensitive to low
observed in mice treated for 5 consecutive days with silymarin. doses of silymarin than to high doses as shown in Table 3. Silymar-
Treatment with silymarin did result in a significant decrease in in dose-dependently increased the basal rate of splenocyte proli-
food intake although no affect on body weight gain was evident feration although the changes were not significant. PHA-P-induced
among the dose groups (data not shown). Consistent with the T-lymphocyte proliferation was increased by 46 % in mice treated
lack of overt toxicity, silymarin treatment did not change relative with 10 mg/kg of silymarin. B-lymphocyte proliferation induced
organ weights as shown in Table 2. A small, although significant, by LPS was increased by 65 % and 35 % in mice treated with 10 and
increase in erythrocytes was seen in the highest treatment group 50 mg/kg of silymarin, respectively. The proliferative response to
(Table 2). Peripheral blood leukocyte counts were not affected by ConA was not changed although the increase in basal proliferation
silymarin treatment.

Table 2 The effects of daily treatment with silymarin for 5 days on relative organ weights and blood cellularity in BALB/c mice

Relative organ weights (g/100 g BW)


Treatment Spleen Thymus Liver Kidney RBCb/mm3 (” 10 ± 6) WBCc/mm3 (” 10 ± 3)
(mg/kg/day)

Control 0.41  0.02a 0.23  0.03 5.62  0.22 1.57  0.04 5.31  0.59 4.54  1.09
10 0.40  0.03 0.26  0.05 5.54  0.11 1.58  0.03 6.55  0.68 5.21  1.50
50 0.38  0.02 0.26  0.04 5.22  0.09 1.58  0.04 5.57  0.45 4.45  0.90
250 0.47  0.03 0.22  0.02 5.24  0.18 1.56  0.02 7.72  0.72* 6.24  0.60

a
Mean  SEM (n = 5 for control; n = 6 for all other groups).
b
Red blood cells.
c
White blood cells (leucocytes).
* Significantly different from control group at P < 0.05 (Fisher's PLSD).

Johnson VJ et al. Physiological Responses of ¼ Planta Med 2003; 69: 44 ± 49


Fig. 1 Altered splenic lymphocyte phenotype in male
BALB/c mice following short-term treatment with silymarin.
Mice were treated i. p. for 5 days with silymarin following
which flow cytometric analysis of CD3+ T-lymphocyte (a),
CD45+/B220 B-lymphocyte (b), CD4+ T-helper lymphocyte
(c) and CD8+ T-cytotoxic/suppressor lymphocyte (d) popula-
tions was performed. Data expressed as absolute number of
positive cells per spleen. Mean  SEM (n = 6). * indicates
significantly different from control group at P < 0.05.

Original Paper
Table 3 The effects of daily treatment with silymarin for 5 days on basal and mitogen induced lymphocyte blastogenesis in BALB/c mice

Con A PHA-P LPS


Treatment No Mitogen Net DPMb SIc Net DPM SI Net DPM SI
(mg/kg/day) DPMa (” 10 ± 3) (” 10 ± 3) (” 10 ± 3) (” 10 ± 3)

Control 1.6  0.2d 231.7  6.6 144.6  4.6 41.1  8.1 25.7  5.6 42.1  4.2 26.3  3.0
10 2.0  0.2 231.8  4.9 115.7  2.7# 59.9  6.3* 30.0  3.5 69.4  2.7** 34.8  1.5
50 2.0  0.4 235.0  6.2 120.5  3.5# 51.4  8.8 26.4  5.0 56.8  6.1* 29.1  3.4
250 2.8  0.8 222.5  3.8 80.7  1.5# 49.7  7.6 18.0  3.0 35.9  1.0 13.0  0.4# 47
a
DPM = disintegrations/minute.
b
Net DPM = DPM with mitogen - DPM without mitogen.
c
SI = Stimulation Index = DPM with mitogen ˜ DPM without mitogen.
d
Mean  SEM (n = 5)
Significantly different from control; * P < 0.05, ** P < 0.01, #P < 0.001 (Fisher's PLSD).

resulted in a decrease in the ConA induced stimulation index for the treated mice. The purpose of this study was to determine the
proliferation in the silymarin treated animals. effects of silymarin on the immune system. Mice treated with a
low dose (10 mg/kg) displayed a decrease in the T-lymphocyte
Cytokine gene expression was also sensitive to silymarin treat- population in the spleen and decreased functionality of these
ment. PHA-induced expression of IL-2 and IL-4 was decreased cells. In contrast, mice treated with a high dose of silymarin
significantly in the 10 mg/kg group only (Fig. 2). In contrast, (250 mg/kg) showed an increased responsiveness to the inflam-
the expression of LPS-induced pro-inflammatory cytokines matory agent LPS.
was increased by silymarin in a dose-dependent manner as
shown in Fig. 3. TNFa and IL-6 expressions were increased in Only low dose treatment with silymarin resulted in a decrease in
mice treated with 50 mg/kg of silymarin and TNFa, IL-6, IL-1b the T-lymphocyte population in the spleen, specifically that of
and iNOS gene expressions were increased in the 250 mg/kg the CD4+ T-helper cell population. Lang et al. [7] showed a de-
group. creasing effect of this flavonoid on peripheral T-lymphocytes al-
though the target population was CD8+ T-lymphocytes. The CD8+
T-cytotoxic/suppressor population was not significantly de-
Discussion creased in this study. Analysis of T-lymphocyte gene expression
also showed suppression at low doses. The effect of silymarin on
Silymarin at the dose range and treatment duration used in the IL-2 and IL-4 mRNA expression may be due to a decrease in T-
present study did not produce any signs of overt clinical toxicity lymphocytes in the spleen or a decrease in responsiveness to
in male BALB/c mice. Despite the drop in caloric intake, there stimulation as reported by Meroni et al. [9]. Interestingly, sily-
were no effects on body weight gain or relative organ weight in marin treatment elevated the proliferative response of T-lym-

Johnson VJ et al. Physiological Responses of ¼ Planta Med 2003; 69: 44 ± 49


In contrast to the inhibitory effects of short-term low dose
treatment with silymarin, high doses promoted the inflamma-
tory response in BALB/c mice. Splenocytes from silymarin treat-
ed mice (250 mg/kg group) showed increased expression of
TNFa, IL-1b, IL-6 and iNOS in response to LPS activation. These
results are paradoxical in that they seem to contradict many of
the in vitro studies demonstrating a decrease in NFkB activation
[3], [14] and NFkB-mediated gene expression [12], [13] follow-
ing exposure to silymarin. The duration of treatment may be
responsible for the difference noted above. Silymarin is a pow-
erful antioxidant that in short-term (hours) in vitro experi-
ments will scavenge the reactive oxygen species produced by
inflammatory agents. Treatment with silymarin in vivo has
also been shown to have antioxidant effects mainly in the form
Original Paper

of restoration of cellular thiol status and bolstering of cellular


glutathione pools [18].

Of particular interest to the present research is the effect of si-


lymarin on the expression of superoxide dismutase (SOD). Lang
et al. [19] demonstrated that silymarin markedly induced SOD
expression and activity in lymphocytes and erythrocytes ex-
posed in vitro. SOD is an antioxidant enzyme that catalyzes the
dismutation of superoxide radical to H2O2, a potent activator of
NFkB [20]. Contrary to the antioxidant role of SOD, over-expres-
sion of Cu/Zn-SOD [20] and Mn-SOD [21] resulted in activation
of NFkB and gene expression. It is likely that in the absence of
changes in the H2O2 metabolizing enzymes, over-expression of
Fig. 2 Decreased PHA-induced expression of T-lymphocyte cytokines
IL-2 and IL-4 in splenocytes from silymarin treated BALB/c mice. Mice SOD leads to excess H2O2 and subsequent NFkB activation. No
were treated with silymarin i. p. for 5 consecutive days following which evidence exists suggesting that silymarin influences the activ-
splenocyte cultures were stimulated with PHA (10 mg/ml) and total ity of catalase or glutathione peroxidase and thus the meta-
RNA was extracted and RT-PCR was used to determine relative changes bolism of H2O2. Reactive oxygen species including superoxide
in the expression of IL-2 (a) and IL-4 (b). Mean  SEM (n = 6). Signifi-
and H2O2 are integral to LPS-mediated signal transduction
cantly different from control group at * P < 0.05 and ** P < 0.01.
48 [22]. It is possible that in our system increased SOD activity in
lymphocytes from silymarin treated mice would result in high-
phocytes to PHA-P. This indicates that there may be a compensa- er generation of H2O2 thus leading to more potent activation of
tory response elicited to correct the silymarin-induced deficit in NFkB. This represents a plausible explanation for the observed
T-lymphocyte numbers and/or function. increase in inflammatory gene expression in response to LPS

Fig. 3 Silymarin treatment promotes LPS-induced expres-


sion of inflammatory genes in BABL/c mice. Mice were treat-
ed with silymarin i. p. for 5 consecutive days following which
splenocytes were treated with LPS (1 mg/ml) and total RNA
was extracted and RT-PCR was used to determine relative
changes in the expression of TNFa (a), IL-1b (b), iNOS (c)
and IL-6 (d). Mean  SEM (n = 6). Significantly different
from control group at * P < 0.05, ** P < 0.005 and *** P <
0.001.

Johnson VJ et al. Physiological Responses of ¼ Planta Med 2003; 69: 44 ± 49


stimulation. Further experimentation is needed to confirm this 9
Meroni PL, Barcellini W, Borghi MO, Vismara A, Ferraro G, Ciani D, Za-
hypothesis. nussi C. Silybin inhibition of human T-lymphocyte activation. Interna-
tional Journal of Tissue Reactions 1988; 10: 177 ± 81
10
Tager M, Dietzmann J, Thiel U, Hinrich Neumann K, Ansorge S. Re-
The purpose of the present study was to determine the physio- storation of the cellular thiol status of peritoneal macrophages from
logical impact of short-term treatment with silymarin on the im- CAPD patients by the flavonoids silibinin and silymarin. Free Radical
mune system. The results indicate that silymarin exerts differen- Research 2001; 34: 137 ± 51
11
Kang SN, Lee MH, Kim KM, Cho D, Kim TS. Induction of human pro-
tial effects depending on the dose administered. We observed an myelocytic leukemia HL-60 cell differentiation into monocytes by sili-
inhibitory effect on T-lymphocytes at the lowest dose but this ef- binin: involvement of protein kinase C. Biochemical Pharmacology
fect disappeared with increased dose. The average daily oral dose 2001; 61: 1487 ± 95
12
for humans taking silymarin is between 500 and 1000 mg/day Zi X, Mukhtar H, Agarwal R. Novel cancer chemopreventive effects of a
flavonoid antioxidant silymarin: inhibition of mRNA expression of an
(7 ± 15 mg/kg for 70 kg person). The observed decrease in T-cell endogenous tumor promoter TNFa. Biochemical and Biophysical Re-
responses in mice exposed to low doses may be beneficial in di- search Communications 1997; 239: 334 ± 9
sease states characterized by T-cell mediated damage as in pri- 13
Cho JY, Kim PS, Park J, Yoo ES, Baik KU, Kim Y, Park MH. Inhibition of
mary biliary cirrhosis [23]. At higher doses, our data indicate tumor necrosis factor-a production in lipopolysaccharide-stimulated

Original Paper
RAW264.7 cells from Amorpha fruticosa. Journal of Ethnopharmacolo-
that silymarin treatment may increase the ability of the immune gy 2000; 70: 127 ± 33
system to fight bacterial infections, thereby adding to the grow- 14
Saliou C, Rihn B, Cillard J, Okamoto T, Packer L. Selective inhibition of
ing list of therapeutic application for this flavonoid mixture. NF-kB activation by the flavonoid hepatoprotector silymarin in
HepG2: Evidence for different activating pathways. FEBS Letters
1998; 440: 8 ± 12
15
Letteron P, Labbe G, Degott C, Berson A, Fromenty B, Delaforge M, Lar-
Acknowledgements rey D, Pessayre D. Mechanism for the protective effects of silymarin
against carbon tetrachloride-induced lipid peroxidation and hepato-
This study was supported in part by a grant from the National In- toxicity in mice. Evidence that silymarin acts both as an inhibitor of
metabolic activation and as a chain-breaking antioxidant. Biochemi-
stitutes of Health TW01009. cal Pharmacology 1990; 39: 2027 ± 34
16
He Q, Osuchowski MF, Johnson VJ, Sharma RP. Physiological responses
of a natural antioxidant flavonoid, silymarin, in BALB/c mice: I. Induc-
References tion of transforming growth factor b1 and c-myc in liver with marginal
effects on other genes. Planta Medica2002; 68: 676 ± 9
17
1
Johnson VJ, Sharma RP. Gender-dependent immunosuppression fol-
Hahn G, Lehmann HD, Kurten M, Uebel H, Vogel G. On the pharmaco- lowing subacute exposure to fumonisin B1. International Immuno-
logy and toxicology of silymarin, and antihepatotoxic active principle pharmacology 2001; 1: 2023 ± 34
from Silybum marianum. Arzneimittel-Forschung/Drug Research 18
Valenzuela A, Aspillaga M, Vial S, Guerra R. Selectivity of silymarin on
1968; 18: 698 ± 704 the increase of the glutathione content in different tissues of the rat.
2
Lahiri-Chatterjee M, Katiyar SK, Mohan RR, Agarwal R. A flavonoid an- Planta Medica 1989; 55: 420 ± 2
tioxidant, silymarin, affords exceptionally high protection against tu- 19
Lang I, Deak G, Muzes G, Pronai L, Feher J. Effect of the natural biofla-
mor promotion in the SNECAR mouse skin tumorogenisis model. Can- vonoid antioxidant silymarin on superoxide dismutase (SOD) activity
cer Research 1999; 59: 622 ± 32 49
3
and expression in vitro. Biotechnology Therapeutics 1993; 4: 263 ± 70
Manna SK, Mukhopadhyay A, Van NT, Aggarwal BB. Silymarin sup- 20
Schmidt KN, Amstad P, Cerutti P, Baeuerle PA. The roles of hydrogen
presses TNF-induced activation of NK-kB, c-Jun N-terminal kinase, peroxide and superoxide as messengers in the activation of transcrip-
and apoptosis. The Journal of Immunology 1999; 163: 6800 ± 9 tion factor NF-kappa B. Chemistry and Biology 1995; 2: 13 ± 22
4
Ferenci P, Dragosics B, Dittrich H, Frank H, Benda L, Lochs H, Meryn S, 21
Li JJ, Oberley LW, Fan M, Colburn NH. Inhibition of AP-1 and NF-kB by
Base W, Schneider B. Randomized controlled trial of silymarin treat- manganese-containing superoxide dismutase in human breast cancer
ment in patients with cirrhosis of the liver. Journal of Hepatology cells. FASEB Journal 1998; 12: 1713 ± 23
1989; 9: 105 ± 13 22
Han YJ, Kwon YG, Chung HT, Lee SK, Simmons RL, Billiar TR, Kim YM.
5
Greimel A, Koch H. Silymarin ± An inhibitor of horseradish peroxidase. Antioxidant enzymes suppress nitric oxide production through the in-
Experimentia 1977; 33: 1417 ± 8 hibition of NF-kappaB activation: Role of H2O2 and nitric oxide in in-
6
Lang I, Deak G, Muzes G, Pronai L, Feher J. Effect of natural bioflavo- ducible nitric oxide synthase expression in macrophages. Nitric Oxide
noid antioxidant silymarin on superoxide dismutase (SOD) activity 2001; 5: 504 ± 13
and expression in vitro. Biotechnology therapeutics 1993; 4: 263 ± 70 23
Kita H, Lian ZX, Van de Water J, He XS, Matsumura S, Kaplan M, Luketic
7
Lang I, Deak GY, Nekam K, Muzes GY, Gonzalez-Cabello R, Gergely P, V, Coppel RL, Ansari AA, Gershwin ME. Identification of HLA-A2-re-
Feher J. Hepatoprotective and immunomodulatory effects of antioxi- stricted CD8(+) cytotoxic T cell responses in primary biliary cirrhosis:
dant therapy. Acta Medica Hungaria 1988; 45: 287 ± 95 T cell activation is augmented by immune complexes cross-presented
8
Agoston M, Cabello RG, Blazovics A, Feher J, Vereckei A. The effect of by dendritic cells. Journal of Experimental Medicine 2002; 195: 113 ±
amiodarone and/or antioxidant treatment on splenocyte blast trans- 23
formation. Clinica Chimica Acta 2001; 303: 87 ± 94

Johnson VJ et al. Physiological Responses of ¼ Planta Med 2003; 69: 44 ± 49

Вам также может понравиться