Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 21221, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34707245

ABSTRACT

Dengue is an arthropod-borne viral disease that has become endemic and a global threat in many countries with no effective antiviral drug available currently. This study showed that flavonoids: silymarin and baicalein could inhibit the dengue virus in vitro and were well tolerated in Vero cells with a half-maximum cytotoxic concentration (CC50) of 749.70 µg/mL and 271.03 µg/mL, respectively. Silymarin and baicalein exerted virucidal effects against DENV-3, with a selective index (SI) of 10.87 and 21.34, respectively. Baicalein showed a better inhibition of intracellular DENV-3 progeny with a SI of 7.82 compared to silymarin. Baicalein effectively blocked DENV-3 attachment (95.59%) to the Vero cells, while silymarin prevented the viral entry (72.46%) into the cells, thus reducing viral infectivity. Both flavonoids showed promising antiviral activity against all four dengue serotypes. The in silico molecular docking showed that silymarin could bind to the viral envelope (E) protein with a binding affinity of - 8.5 kcal/mol and form hydrogen bonds with the amino acids GLN120, TRP229, ASN89, and THR223 of the E protein. Overall, this study showed that silymarin and baicalein exhibited potential anti-DENV activity and could serve as promising antiviral agents for further development against dengue infection.


Subject(s)
Antiviral Agents/toxicity , Dengue Virus/drug effects , Flavanones/toxicity , Silymarin/toxicity , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Dengue Virus/physiology , Flavanones/pharmacology , Inhibitory Concentration 50 , Protein Binding , Silymarin/pharmacology , Vero Cells , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects
2.
Neurotoxicology ; 78: 64-70, 2020 05.
Article in English | MEDLINE | ID: mdl-32084436

ABSTRACT

Silymarin is a phytotherapeutic agent derived from the species Silybum marianum (Asteraceae), commonly is known as milk thistle, and traditionally used as a hepatoprotective; however, recent studies have proposed its use in order to promote lactogenesis, but there are few reports of its effects on the development of offspring. Thus, the objective of this study was to evaluate the effect of silymarin treatment during pregnancy and breastfeeding on the sensory-somatic-motor development and adult behavior of F1-generation Swiss mice. The pregnant females of the parental generation were distributed in four experimental groups and treated orally with doses of 100, 200 or 300 mg/kg of silymarin, with a control group receiving vehicle - vegetable oil (VEH), to obtain the F1-generation. At the end of lactation, the parental generation were submitted to euthanasia. Body mass evolution was determined in both generations. The sensory-motor development of the offspring (F1-generation) was evaluated, and one male pup from each litter was followed up for an analysis of adult behavior. In the F1 analysis, no differences between the groups were observed in initial development from the sensory-somatic-motor analysis performed during the 1st to 21st postnatal days. In the behavioral evaluation of adults from the F1 generation, all the groups from dams treated with silymarin in open field (OF) analysis showed a decrease in the time spent in the periphery and an increase in the time spent in the center, but the ambulation observed by the number of quadrant crossed showed no difference. In addition, during OF, the 100 and 200 mg/kg groups presented an increase in fecal bolus compared with the VEH group. There was a decrease in immobility time in the forced swimming test in the 300 mg/kg group compared to the VEH group. Regarding the memory and learning test, the groups did not differ in their recognition scores. The results of this study using an animal model indicate that treatment with silymarin during pregnancy and breastfeeding does not promote significant morpho-functional changes in the offspring in their initial development and adult behavior, indicating the safety of its use during gestation and lactation.


Subject(s)
Behavior, Animal/drug effects , Maternal Behavior/drug effects , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/psychology , Silymarin/toxicity , Animals , Body Weight/drug effects , Female , Lactation , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced
3.
Nat Prod Res ; 34(24): 3540-3544, 2020 Dec.
Article in English | MEDLINE | ID: mdl-30856005

ABSTRACT

Silymarin prepared from the fruits of Silybum marianum (L.) Gaertn. (Asteraceae) has long been used for the treatment of liver disorders. This study was carried out to evaluate the protective effect of the fruit extract of white-flowered S. marianum variety albiflorum Eig. (WSE) against paracetamol-induced liver toxicity in rats. Silyhermin, isosilandrins A/B were identified as the major flavonolignans in WSE. Cytotoxic activities of WSE and isolated flavonolignans compared to silymarin were carried out using sulforhodamine B assay. WSE, silyhermin and isosilandrins had no obvious harmful effect on normal human cell line compared to silymarin with IC50 values 78.95, 84.34, 72.14 and 16.83 µg/ml, respectively. The hepatoprotective activity of WSE at dose 50 mg/kg was comparable to silymarin (100 mg/kg). These data were supplemented with histopathological studies on liver sections. The hepatoprotective effects of WSE on oxidative stress induced by administration of paracetamol are probably associated with its antioxidant properties.


Subject(s)
Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Plant Extracts/pharmacology , Protective Agents/pharmacology , Silybum marianum/chemistry , Acetaminophen/toxicity , Animals , Cell Line , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Egypt , Fruit/chemistry , Humans , Lignans/analysis , Lignans/chemistry , Lignans/pharmacology , Male , Oxidative Stress/drug effects , Plant Extracts/chemistry , Plants, Medicinal/chemistry , Protective Agents/chemistry , Rats, Wistar , Silymarin/toxicity
4.
Bioorg Med Chem ; 25(17): 4845-4854, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28756013

ABSTRACT

To investigate the effects of alkylation at 5-OH and 20-OH of 2,3-dehydrosilybin on prostate cancer cell proliferation, the synthetic approaches to 5- or/and 20-O-alkyl-2,3-dehydrosilybins, through a multi-step sequence from commercially available silybin, have been successfully developed. The first three reactions in the syntheses were completed through a one-pot procedure by managing anaerobic and aerobic conditions. With these synthetic methods in hand, twenty-one 2,3-dehydrosilybins, including seven 20-O-alkyl, seven 5,20-O-dialkyl, and seven 5-O-alkyl-2,3-dehydrosilybins, have been achieved for the evaluation of their biological profiles. Our WST-1 cell proliferation assay data indicate that nineteen out of the twenty-one 2,3-dehydrosilybins possess significantly improved antiproliferative potency as compared with silybin toward both androgen-sensitive (LNCaP) and androgen-insensitive prostate cancer cell lines (PC-3 and DU145). 5-O-Alkyl-2,3-dehydrosilybins were identified as the optimal subgroup that can consistently inhibit cell proliferation in three prostate cancer cell models with all IC50 values lower than 8µM. Our flow cytometry-based assays also demonstrate that 5-O-heptyl-2,3-dehydrosilybin effectively arrests the cell cycle in the G0/G1 phase and activates PC-3 cell apoptosis.


Subject(s)
Antineoplastic Agents/chemical synthesis , Silymarin/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Silymarin/chemical synthesis , Silymarin/toxicity , Structure-Activity Relationship
5.
AAPS PharmSciTech ; 18(8): 3236-3246, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28577126

ABSTRACT

Silibinin (SB) and pomegranate oil (PO) present therapeutic potential due to antioxidant activity, but the biological performance of both bioactives is limited by their low aqueous solubility. To overcome this issue, the aim of the present investigation was to develop nanocapsule suspensions with PO as oil core for SB encapsulation, as well as assess their toxicity in vitro and radical scavenging activity. The nanocapsule suspensions were prepared by interfacial deposition of preformed polymer method. SB-loaded PO-based nanocapsules (SBNC) showed an average diameter of 157 ± 3 nm, homogenous size distribution, zeta potential of -14.1 ± 1.7 mV, pH of 5.6 ± 0.4 and SB content close to 100%. Similar results were obtained for the unloaded formulation (PONC). The nanocapsules controlled SB release at least 10 times as compared with free SB in methanolic solution. The SBNC scavenging capacity in vitro was statistically higher than free SB (p < 0.05). Cell viability in monocytes and lymphocytes was kept around 100% in the treatments with SBNC and PONC, while the SB and the PO caused a decrease around 30% at 50 µM (SB) and 724 µg/mL (PO). Protein carbonyls and DNA damage were minimized by SB and PO nanoencapsulation. Lipid peroxidation occurred in nanocapsule treatments regardless of the SB presence, which may be attributed to PO acting as substrate in reaction. The free compounds also caused lipid peroxidation. The results show that SBNC and PONC presented adequate physicochemical characteristics and low toxicity against human blood cells. Thereby, this novel nanocarrier may be a promising formulation for therapeutic applications.


Subject(s)
Cytotoxins/chemistry , Free Radical Scavengers/chemistry , Lythraceae , Nanocapsules/chemistry , Silymarin/chemistry , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cytotoxins/toxicity , Dose-Response Relationship, Drug , Free Radical Scavengers/toxicity , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/physiology , Lipid Peroxidation/drug effects , Nanocapsules/toxicity , Plant Oils/chemistry , Plant Oils/toxicity , Silybin , Silymarin/toxicity , Solubility
6.
Curr Med Chem ; 23(34): 3925-3950, 2016.
Article in English | MEDLINE | ID: mdl-27557939

ABSTRACT

BACKGROUND: The generic name "flavonolignan" was created in 1968 for a relatively small class of naturally occurring hybrid molecules biogenetically originated from ubiquitous flavonoids and lignans (phenylpropanoids). The first group of flavonolignans was extracted from Silybum marianum that has long been used for hepatoprotection. Recently, the medicinal merit of flavonolignans has been extended to the prostate cancer management. METHODS: Systematic interpretation and summarization of the relevant literature. RESULTS: Over forty naturally occurring flavonolignans have so far been obtained from various plants. Certain flavonolignans have been demonstrated by in vitro cell-based and in vivo animal-based experiments, and human clinical studies i) to possess effective chemopreventive function against various tumor promoters; ii) to show the anti-angiogenic efficacy; iii) to have potential in treating prostate cancer; iv) to sensitize prostate tumors to chemotherapeutic agents through down-regulation of P-glycoprotein and other mechanisms; and v) to be used by prostate cancer patients to protect or treat the hepatotoxicity caused by several chemotherapies. Certain flavonolignans can synergize with well-established chemotherapeutic agents for prostate cancer. CONCLUSION: This review provides a systematic and in-depth overview of the promise and potential of flavonolignans in prostate cancer management, which covers their chemopreventive effect, chemotherapeutic treatment, mechanisms of actions, synthetic derivatives, structure-activity relationships, and the difference in inhibiting prostate cancer cell proliferation between certain flavonoligans and their respective flavonoid counterpart. This summarization aims to provide valuable insights into further and rational development of flavonolignans for prostate cancer management by interpreting the data reported in the literature.


Subject(s)
Flavonolignans/therapeutic use , Prostatic Neoplasms/drug therapy , Apoptosis , Flavonolignans/chemistry , Flavonolignans/metabolism , Flavonolignans/toxicity , Humans , Male , Plant Extracts/chemistry , Plants, Medicinal/chemistry , Plants, Medicinal/metabolism , Prostatic Neoplasms/pathology , Silybin , Silymarin/chemistry , Silymarin/therapeutic use , Silymarin/toxicity , Structure-Activity Relationship
7.
Mater Sci Eng C Mater Biol Appl ; 65: 164-71, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-27157739

ABSTRACT

PAMAM-grafted TiO2 nanotubes (PAMAM-TiO2NT) have been synthesized and evaluated as new drug nanocarriers, using curcumin (CUR), methotrexate (MTX), and silibinin (SIL) as model therapeutic compounds. TiO2NT were surface-modified using a silane coupling agent and subsequently conjugated with PAMAM dendrimer of the third generation. The characterization of PAMAM-TiO2NT nanomaterials was performed by FTIR, TEM, N2 adsorption-desorption isotherms, XRD, and TGA techniques, which accounted for a 2.6wt.% of PAMAM grafting in the prepared materials. The drug loading capacity, drug release properties, and cytotoxicity of PAMAM-TiO2NT showed a significant improvement compared to pristine TiO2NT, thus revealing the promising properties of these new materials for drug delivery purposes.


Subject(s)
Dendrimers/chemistry , Drug Carriers/chemistry , Nanotubes/chemistry , Pharmaceutical Preparations/chemistry , Titanium/chemistry , Cell Survival/drug effects , Curcumin/chemistry , Curcumin/metabolism , Curcumin/toxicity , Drug Liberation , HeLa Cells , Humans , Methotrexate/chemistry , Methotrexate/metabolism , Methotrexate/toxicity , Microscopy, Electron, Transmission , Pharmaceutical Preparations/metabolism , Silybin , Silymarin/chemistry , Silymarin/metabolism , Silymarin/toxicity , Spectroscopy, Fourier Transform Infrared
8.
Environ Sci Pollut Res Int ; 23(17): 17226-35, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27221463

ABSTRACT

Benzo[a]pyrene (B[a]P) is an environmental toxicant and endocrine disruptor. Therefore, the aim of the present study was to investigate the toxicity of B[a]P in testis of rats and also to study the role of silymarin and thymoquinone (TQ) as natural antioxidants in the alleviation of such toxicity. Data of the present study showed that levels of testosterone, estrogen and progesterone were significantly decreased after treatment of rats with B[a]P. In addition, B[a]P caused downregulation of the expressions of steroidogenic enzymes including CYP17A1 and CP19A1, and decreased the activity of 17-ß hydroxysteroid dehydrogenase (17ß-HSD). Moreover, B[a]P decreased the activities of antioxidant enzymes including catalase (CAT), glutathione peroxidase (GPX) and superoxide dismutase (SOD), and significantly increased free radicals levels in testis of male rats. However, pretreatment of rats with silymarin prior to administration of B[a]P was found to restore the level of free radicals, antioxidant status, and activities of steroidogenic enzymes to their normal levels in testicular tissues. Moreover, histopathological finding showed that silymarin recovered the abnormalities occurred in tubules caused by B[a] P in testis of rats. On the other hand, TQ showed pro-oxidant effects and did not ameliorate the toxic effects of B[a] P on the testicular tissue since it decreased antioxidant enzymes activities and inhibited the protein expression of CYP11A1 and CYP21A2 compared to control rats. Moreover, TQ decreased the levels of testosterone, estrogen, and progesterone either in the presence or absence of B[a]P. It is concluded that B[a]P decreased testosterone levels, inhibited antioxidant enzymes activities, caused downregulation of CYP isozymes involved in steroidogenesis, and increased free radical levels in testis. Moreover, silymarin was more effective than TQ in restoring organism health and alleviating the deleterious effects caused by B[a]P in the testis of rats. Due to its negative impact, it is highly recommended to limit the use of TQ as a dietary supplement since millions of people in the Middle East are using it to improve their health.


Subject(s)
Antioxidants/toxicity , Benzo(a)pyrene/toxicity , Benzoquinones/toxicity , Environmental Pollutants/toxicity , Reproduction/drug effects , Silymarin/toxicity , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Biomarkers/analysis , Catalase/metabolism , Free Radicals/metabolism , Glutathione Peroxidase/metabolism , Male , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Testis/drug effects , Testosterone/metabolism
9.
J Photochem Photobiol B ; 156: 61-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26851710

ABSTRACT

Silymarin, a standardized extract of the seeds of the milk thistle (Silybum marianum) and its major component, silybin, is now used as an active component in a broad spectrum of dietary supplements, cosmetics and dermatological preparations. However, despite its use in skin products, there are no published data to exclude its phototoxic potential. The primary purpose of this study was to examine the phototoxicity of silymarin and its flavonolignans, silybin, isosilybin, silychristin, silydianin and 2,3-dehydrosilybin by validated 3T3 NRU assay. Further, we compared the validated biological system Balc/c 3T3 cell line with other cell models, particularly normal human dermal fibroblasts (NHDF), normal human epidermal keratinocytes (NHEK) and the human keratinocyte cell line (HaCaT). The results showed that silymarin and the flavonolignans silybin, isosilybin, silychristin and silydianin had no phototoxicity towards any of the cells used. In contrast, 2,3-dehydrosilybin was identified as a compound with phototoxic potential. Further study is needed to evaluate the health risks associated with 2,3-dehydrosilybin use in skin preparations.


Subject(s)
Silymarin/toxicity , Ultraviolet Rays , 3T3 Cells , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred BALB C
10.
Toxicol In Vitro ; 29(2): 337-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25500126

ABSTRACT

The cytotoxic effect of monensin, narasin and salinomycin followed by their co-action with silybin in the cell line cultures of human hepatoma (HepG2), chicken hepatoma (LMH) or rat myoblasts (L6) have been investigated. The effective concentration of the studied ionophoric polyethers has been assessed within two biochemical endpoints: mitochondrial activity (MTT assay) and membrane integrity (LDH assay) after 24h incubation of each compound and farther, the cytotoxicity influenced in course of their interaction with silybin was determined. The most affected endpoints were found for inhibition of mitochondrial activity of the hepatoma cell lines and their viability depended on concentration of the ionophoric polyether, as well as on the cell line tested. The rat myoblasts were more sensitive target for cellular membrane damage when compared to inhibition of mitochondrial activity. An interaction between the ionophoric polyethers and silybin resulted a considerable cytotoxicity decrease within all studied cell lines; the combination index (CI) showed differences of interaction mode and dependence on cell culture, concentration of silybin, as well as the assay used. The obtained results are of interest in respect to recent findings on applicability of salinomycin and monensin for human therapy.


Subject(s)
Monensin/toxicity , Pyrans/toxicity , Silymarin/toxicity , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Chickens , Drug Interactions , Hep G2 Cells , Humans , Myoblasts/drug effects , Rats , Silybin
11.
Curr Drug Targets ; 14(14): 1659-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24093748

ABSTRACT

Osteoporosis is a progressive disease of the skeleton characterised by bone fragility due to a reduction in bone mass and possibly to alteration in bone architecture that lead to a propensity to fracture with minimum trauma. Most osteoporotic fractures occur at locations rich in trabecular or cancellous bone and usually related to post menopausal women. Recently, silymarin received attention due to its alternative beneficial effect on bone formation. It is a mixture of flavonoids with powerful antioxidant properties. This review focuses on the use of milk thistle or silymarin for the treatment of osteoporosis that may be related to fracture bone. Silymarin shows potent antioxidant herb that may modulate multiple genes in favour of helping to build bone and prevent bone loss. In the mouse fracture healing model, silymarin supplementation improved tibial healing with elevated BMD and serum levels of ALP and osteocalcin. Silymarin also demonstrated clear estrogenic antiosteoporotic effects in bone structure. Silymarin appears to play a crucial role to prevent bone loss and might regulate osteogenesis and may be beneficial for fracture healing. If silymarin is considered for the use of post menopausal women, it may be used for the treatment of osteoporosis. It would be of great benefit to postmenopausal women to develop an oestrogen antagonist that is as potent and efficacious as oestrogen in preventing bone loss without the major side effect associated with HRT.


Subject(s)
Fracture Healing/drug effects , Fractures, Bone/drug therapy , Osteoporosis/drug therapy , Plant Preparations/therapeutic use , Silybum marianum/chemistry , Silymarin/therapeutic use , Animals , Bone Density/drug effects , Fractures, Bone/etiology , Fractures, Bone/metabolism , Fractures, Bone/pathology , Humans , Osteoporosis/complications , Osteoporosis/metabolism , Osteoporosis/pathology , Plant Preparations/administration & dosage , Plant Preparations/toxicity , Receptors, Estrogen/metabolism , Silymarin/administration & dosage , Silymarin/toxicity
12.
Cancer Lett ; 339(1): 102-6, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23879966

ABSTRACT

Small-cell lung carcinoma (SCLC) has a dismal prognosis in part because of multidrug resistance (MDR). Silibinin is a flavonolignan extracted from milk thistle (Silybum marianum), extracts of which are used in traditional medicine. We tested the effects of silibinin on drug-sensitive (H69) and multi-drug resistant (VPA17) SCLC cells. VPA17 cells did not show resistance to silibinin (IC50 = 60 µM for H69 and VPA17). Flow cytometry analysis after incubation in 30 µM silibinin showed no changes in cell cycle phases in VPA17 or H69 cells compared with untreated cells. Silibinin (30 µM) incubation was pro-apoptotic in VPA17 cells after > 3 days, as measured by ELISA of BUdR labeled DNA fragments. Apoptosis was also indicated by an increase in caspase-3 specific activity and decrease in survivin in VPA17 MDR cells. VPA17 cells had increased Pgp-mediated efflux of calcein acetoxymethyl ester (calcein AM); however, this was inhibited in cells pre-incubated in silibinin for 5 days. Pre-incubation of VPA17 cells in 30 µM silibinin for 5 days also reversed resistance to etoposide (IC50 = 5.50 to 0.65 µM) and doxorubicin (IC50 = 0.625 to 0.035 µM). The possible synergistic relationship between silibinin and chemotherapy drugs was determined by exposure of VPA17 cells to 1:1 ratios of their respective IC50 values, with serial dilutions at 0.25 to 2.0 × IC50 and calculation of the combination index (CI). Silibinin and etoposide showed synergism (CI = 0.46 at ED50), as did silibinin and doxorubicin (CI = 0.24 at ED50). These data indicate that in SCLC, silibinin is pro-apoptotic, reverses MDR and acts synergistically with chemotherapy drugs. Silibinin, a non-toxic natural product may be useful in the treatment of drug-resistant SCLC.


Subject(s)
Antioxidants/pharmacology , Drug Resistance, Neoplasm , Lung Neoplasms/metabolism , Silymarin/pharmacology , Small Cell Lung Carcinoma/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Antioxidants/toxicity , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , DNA Fragmentation/drug effects , Drug Resistance, Multiple , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Silybin , Silymarin/toxicity , Survivin
13.
Bioorg Med Chem ; 21(13): 3919-26, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23673225

ABSTRACT

Silymarin, an extract of the seeds of milk thistle (Silybum marianum), is used as an herbal remedy, particularly for hepatoprotection. The main chemical constituents in silymarin are seven flavonolignans. Recent studies explored the non-selective methylation of one flavonolignan, silybin B, and then tested those analogues for cytotoxicity and inhibition of both cytochrome P450 (CYP) 2C9 activity in human liver microsomes and hepatitis C virus infection in a human hepatoma (Huh7.5.1) cell line. In general, enhanced bioactivity was observed with the analogues. To further probe the biological consequences of methylation of the seven major flavonolignans, a series of 7-O-methylflavonolignans were generated. Optimization of the reaction conditions permitted selective methylation at the phenol in the 7-position in the presence of each metabolite's 4-5 other phenolic and/or alcoholic positions without the use of protecting groups. These 7-O-methylated analogues, in parallel with the corresponding parent compounds, were evaluated for cytotoxicity against Huh7.5.1 cells; in all cases the monomethylated analogues were more cytotoxic than the parent compounds. Moreover, parent compounds that were relatively non-toxic and inactive or weak inhibitors of hepatitis C virus infection had enhanced cytotoxicity and anti-HCV activity upon 7-O-methylation. Also, the compounds were tested for inhibition of major drug metabolizing enzymes (CYP2C9, CYP3A4/5, UDP-glucuronsyltransferases) in pooled human liver or intestinal microsomes. Methylation of flavonolignans differentially modified inhibitory potency, with compounds demonstrating both increased and decreased potency depending upon the compound tested and the enzyme system investigated. In total, these data indicated that monomethylation modulates the cytotoxic, antiviral, and drug interaction potential of silymarin flavonolignans.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Silybum marianum/chemistry , Silymarin/chemistry , Silymarin/pharmacology , Antiviral Agents/isolation & purification , Antiviral Agents/toxicity , Aryl Hydrocarbon Hydroxylases/metabolism , Cell Line , Cell Survival/drug effects , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Glucuronosyltransferase/metabolism , Hepatitis C/drug therapy , Humans , Methylation , Microsomes/drug effects , Microsomes/enzymology , Microsomes/metabolism , Silymarin/isolation & purification , Silymarin/toxicity
14.
J Med Chem ; 55(11): 5231-42, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22624880

ABSTRACT

A codrug of the anti-Alzheimer drug tacrine and the natural product silibinin was synthesized. The codrug's biological and pharmacological properties were compared to an equimolar mixture of the components. The compound showed potent acetyl- and butyrylcholinesterase inhibition. In a cellular hepatotoxicity model, analyzing the influence on viability and mitochondria of hepatic stellate cells (HSC), the toxicity of the codrug was markedly reduced in comparison to that of tacrine. Using a neuronal cell line (HT-22), a neuroprotective effect against glutamate-induced toxicity could be observed that was absent for the 1:1 mixture of components. In subsequent in vivo experiments in rats, in contrast to the effects seen after tacrine treatment, after administration of the codrug no hepatotoxicity and no induction of the cytochrome P450 system were noticed. In a scopolamine-induced cognitive impairment model using Wistar rats, the codrug was as potent as tacrine in reversing memory dysfunction. The tacrine-silibinin codrug shows high AChE and BChE inhibition, neuroprotective effects, lacks tacrine's hepatotoxicity in vitro and in vivo, and shows the same pro-cognitive effects in vivo as tacrine, being superior to the physical mixture of tacrine and silibinin in all these regards.


Subject(s)
Cholinesterase Inhibitors/chemistry , Cognition/drug effects , Liver/drug effects , Neuroprotective Agents/chemistry , Silymarin/chemistry , Tacrine/analogs & derivatives , Tacrine/chemistry , Acetylcholinesterase/metabolism , Allosteric Regulation , Animals , Butyrylcholinesterase/metabolism , Cell Count , Cell Line , Cell Survival/drug effects , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/toxicity , Eels , Female , Glutamic Acid/pharmacology , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Horses , Lipid Peroxidation/drug effects , Liver/cytology , Maze Learning/drug effects , Mice , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/toxicity , Rats , Rats, Wistar , Silybin , Silymarin/pharmacology , Silymarin/toxicity , Tacrine/pharmacology , Tacrine/toxicity
15.
Pharm Biol ; 49(4): 408-15, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21391842

ABSTRACT

OBJECTIVE: Punarnavashtak kwath (PNK) is a classical Ayurvedic formulation, mentioned in Ayurvedic literature Bhaishajya Ratnavali, for hepatic disorders and asthma. This study investigated the hepatoprotective activity of PNK to validate the traditional use of this formulation. MATERIALS AND METHODS: PNK was prepared in the laboratory according to the method given in Ayurvedic literature. Phytochemical screening was performed to determine the presence of phytoconstituents. Hepatoprotective activity was evaluated against CCl(4)-induced hepatotoxicity in rats and by its effect on the HepG2 cell line. RESULTS: Preliminary phytochemical screening revealed the presence of alkaloids, tannins, flavonoids, saponins, and a bitter principle in PNK. Administration of PNK produced significant hepatoprotective effect as demonstrated by decreased levels of serum liver marker enzymes such as aspartate transaminase, serum alanine transaminase, serum alkaline phosphatase, and serum bilirubin and an increase in protein level. Thiopentone-induced sleeping time was also decreased in the PNK-treated animals compared with the CCl(4)-treated group. It also showed antioxidant activity by increase in activity of glutathione, superoxide dismutase, and catalase and by a decrease in thiobarbituric acid reactive substance level compared with the CCl(4)-treated group. Results of a histopathological study also support the hepatoprotective activity of PNK. Investigation carried out on the HepG2 cell line depicted significant increase in viability of cells exposed to PNK as compared with CCl(4)-treated cells. DISCUSSION AND CONCLUSION: It can be concluded that PNK protects hepatocytes from CCl(4)-induced liver damages due to its antioxidant effect on hepatocytes. An in vitro study on HepG2 cell lines also supports its protective effect.


Subject(s)
Antioxidants/pharmacology , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Cytoprotection/drug effects , Liver/drug effects , Phytotherapy , Plant Extracts/pharmacology , Alanine Transaminase/blood , Animals , Antioxidants/analysis , Antioxidants/toxicity , Aspartate Aminotransferases/blood , Female , Hep G2 Cells , Humans , Male , Medicine, Ayurvedic , Mice , Plant Extracts/analysis , Plant Extracts/toxicity , Plants, Medicinal , Rats , Rats, Wistar , Silymarin/pharmacology , Silymarin/toxicity
16.
Toxicol In Vitro ; 25(1): 21-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20828605

ABSTRACT

The effect of a standardised dry extract from Silybum marianum (HEPAR-PASC®) on the enzyme kinetics of cytochrome-P450 isoenzymes (CYP) was investigated with primary human hepatocytes and human liver microsomes in order to assess the potential for drug-drug interactions. A cytotoxic effect on hepatocytes was observed at concentrations at and above 50 µg/ml. The EC(50) value was calculated to be 72.0 µg/ml. Therefore, the chosen test concentrations for CYP induction on human hepatocytes were 50, 10, and 1.5 µg/ml, which allowed for interpretation of the clinical significance of the data with a range of 50-1-fold c(max) at maximal recommended doses. No induction was observed at the lowest concentration of 1.5 µg/ml, which is close to c(max). The extract did not induce CYP 3A4 at any of the tested concentrations. A low or marginal induction of 1A2, 2B6, and 2E1 at the maximum concentration of 50 µg/ml was observed. CYP inhibition on human microsomes was tested at concentrations of 150, 15, and 1.5 µg/ml. No or minor CYP inhibition was observed for all CYPs tested at the lowest concentration of 1.5 µg/ml, i.e. CYPs 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4. At concentrations of 15 and 150 µg/ml the extract significantly inhibited CYP 2B6, 2C8, 2C9, 2C19, 2E1, and 3A4. In these cases, K(i) values were determined. All K(i) values exceeded c(max) by at least a factor of 10-fold. According to FDA regulations 1>c(max)/K(i)>0.1 indicates, that drug-drug interactions are possible for CYPs 2C8, and 2C9, but not likely, and are remote for CYPs 2C19, 2D6, and 3A4.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/drug effects , Herb-Drug Interactions , Plant Extracts/toxicity , Seeds/chemistry , Silybum marianum/chemistry , Silymarin/toxicity , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/biosynthesis , Enzyme Induction/drug effects , Enzyme Inhibitors/toxicity , Hepatocytes/enzymology , Humans , Inhibitory Concentration 50 , Kinetics , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Plant Extracts/chemistry , Silybin , Silymarin/chemistry
17.
Mutagenesis ; 25(3): 223-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20032005

ABSTRACT

Silybin (SB), a constituent of the medicinal plant Silybum marianum, is reported to be a potent hepatoprotective agent, but little is currently known regarding its genotoxicity, mutagenicity and potential chemopreventive properties. In this study, we evaluated the ability of SB to induce DNA migration and micronuclei (MN) formation in human hepatoma cells (HepG2). Also, possible preventive effects of SB on MN formation induced by three different mutagens, bleomycin (BLEO), benzo[a]pyrene (B[a]P) and aflatoxin B(1) (AFB(1)), were studied. To clarify the possible mechanism of SB antimutagenicity, three treatment protocols were applied: pretreatment, in which SB was added before the application of the mutagens; simultaneous treatment, in which SB was added during treatment and post-treatment, in which SB was added after the application of the mutagens. At concentrations up to 100 microM, SB was non-genotoxic, while at a concentration of 200 microM, SB induced DNA migration, generated oxidized DNA bases, reduced cell viability, decreased the replicative index of the cells and induced oxidative stress. It is noteworthy that SB was able to reduce the genotoxic effect induced by B[a]P, BLEO and AFB(1) in pretreatment and simultaneous treatments but had no significant effect on DNA damage induction in post-treatment. Taken together, our findings indicate that SB presents anti-genotoxic activity in vitro, which suggests potential use as a chemopreventive agent.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Mutagens/toxicity , Silymarin/toxicity , Aflatoxin B1/toxicity , Benzo(a)pyrene/toxicity , Bleomycin/toxicity , Cell Death/drug effects , Cell Division/drug effects , Cell Line, Tumor , DNA/metabolism , DNA Damage , DNA-Formamidopyrimidine Glycosylase/metabolism , Endonucleases/metabolism , Escherichia coli Proteins/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Micronuclei, Chromosome-Defective/drug effects , Mutagens/chemistry , Reactive Oxygen Species/metabolism , Silybin , Silymarin/chemistry
18.
Acta Pharmacol Sin ; 31(1): 118-26, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20023692

ABSTRACT

AIM: Silibinin (SB), silydianin (SD), and silychristin (SC) are components of silymarin. These compounds can be used to protect the skin from oxidative stress induced by ultraviolet (UV) irradiation and treat it. To this end, the absorption of silymarin constituents via the skin was examined in the present report. METHODS: Transport of SB, SD, and SC under the same thermodynamic activity through and into the skin and the effects of pH were studied in vitro using a Franz diffusion assembly. RESULTS: The lipophilicity increased in the order of SC

Subject(s)
Silymarin/pharmacokinetics , Skin Absorption , Administration, Cutaneous , Animals , Antioxidants/isolation & purification , Antioxidants/pharmacokinetics , Antioxidants/toxicity , Diffusion Chambers, Culture , Female , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Mice , Mice, Nude , Permeability , Silybin , Silymarin/chemistry , Silymarin/isolation & purification , Silymarin/toxicity , Skin Irritancy Tests , Thermodynamics , Time Factors
19.
Eur J Pharmacol ; 589(1-3): 1-7, 2008 Jul 28.
Article in English | MEDLINE | ID: mdl-18619590

ABSTRACT

Silybin is a flavonoid with antioxidant and free radical scavenging abilities. Silybin also acts as an iron chelator by binding Fe (III). The present study was undertaken to assess the biological effects of silybin on T leukemia cells in the presence or absence of iron and compare its effects with a well-known iron chelator; desferrioxamine. In these experiments, we studied the growth capacity of Jurkat while varying iron availability in the environment. Desferrioxamine significantly inhibited growth and proliferation of Jurkat cells, blocking treated cells in the G0/G1 phase and inducing apoptosis. In contrast, silybin showed a bimodal effect, inducing cell proliferation at lower concentrations whereas inhibition of DNA synthesis and significant cell death was observed at higher concentrations. Chelation of Fe totally abrogated antiproliferative, cytotoxic and apoptotic effects of desferrioxamine on Jurkat cells. Conversely, the silybin-Fe complex had no appreciable effect on its antiproliferative and cytotoxic activities. The cytotoxic effect of desferrioxamine was also prevented in iron-loaded Jurkat cells; however, the effect of silybin on the growth and viability of iron-loaded cells was similar to the effect of its iron complex on untreated Jurkat cells. Despite the Fe chelating activity of silybin that suggests its possible application in chelation therapy of chronic iron overload, the biological effects of silybin on Jurkat cells are different than those of desferrioxamine, probably due to antioxidant activity of silybin, which causes pro-oxidant effect via iron-catalyzed oxidation with the subsequent generation of reactive oxygen species.


Subject(s)
Antioxidants/pharmacology , Cell Proliferation/drug effects , Deferoxamine/pharmacology , Iron Chelating Agents/pharmacology , Iron/metabolism , Leukemia, T-Cell/pathology , Antioxidants/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , DNA Replication/drug effects , Deferoxamine/toxicity , Dose-Response Relationship, Drug , Humans , Iron Chelating Agents/toxicity , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Silybin , Silymarin/pharmacology , Silymarin/toxicity , Time Factors
20.
Med Res Rev ; 28(5): 746-72, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17979145

ABSTRACT

Medicinal plants have been traditionally used for treating liver diseases since centuries. Several leads from plant sources have been found as potential hepatoprotective agents with diverse chemical structures. Although, a big list of hepatoprotective phytomolecules was reported in the scientific literature, only a few were potent against various types of liver damages. Of which, silymarin, andrographolide, neoandrographolide, curcumin, picroside, kutkoside, phyllanthin, hypophyllanthin, and glycyrrhizin have largely attracted the scientific community. This review focuses discussion on the chemistry, biological activity, mode of action, toxicity, and future prospects of these leads.


Subject(s)
Cytokines/metabolism , Liver Diseases/drug therapy , Liver/drug effects , Plant Extracts/pharmacology , Protective Agents/pharmacology , Animals , Cinnamates/chemistry , Cinnamates/metabolism , Cinnamates/pharmacology , Cinnamates/toxicity , Curcumin/chemistry , Curcumin/metabolism , Curcumin/pharmacology , Curcumin/toxicity , Diterpenes/chemistry , Diterpenes/metabolism , Diterpenes/pharmacology , Diterpenes/toxicity , Glucosides/chemistry , Glucosides/metabolism , Glucosides/pharmacology , Glucosides/toxicity , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/metabolism , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/toxicity , Humans , Lignans/chemistry , Lignans/metabolism , Lignans/pharmacology , Lignans/toxicity , Liver/metabolism , Liver Diseases/metabolism , Phytotherapy , Plant Extracts/metabolism , Plant Extracts/therapeutic use , Plant Extracts/toxicity , Plants , Protective Agents/chemistry , Protective Agents/metabolism , Protective Agents/therapeutic use , Protective Agents/toxicity , Silymarin/chemistry , Silymarin/metabolism , Silymarin/pharmacology , Silymarin/toxicity , Tetrahydronaphthalenes/chemistry , Tetrahydronaphthalenes/metabolism , Tetrahydronaphthalenes/pharmacology , Tetrahydronaphthalenes/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...