Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.667
Filter
1.
Balkan Med J ; 41(3): 193-205, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38700358

ABSTRACT

Background: Paclitaxel (PAX) is a widely used chemotherapy drug for various cancer types but often induces significant toxicity in multiple organ systems. Silymarin (SIL), a natural flavonoid, has shown therapeutic potential due to its multiple benefits. Aims: To evaluate the therapeutic efficacy of SIL in mitigating liver and kidney damage induced by PAX in rats, focusing on oxidative stress, inflammation, and apoptosis pathways. Study Design: Experimental animal model. Methods: The study included 28 male Wistar rats aged 12-14 weeks weighing 270-300 g. The rats were divided into four groups: control, SIL, PAX, and PAX + SIL, with seven in each group. The rats received intraperitoneal (i.p.) injections at a dose of 2 mg per kilogram of body weight of PAX for 5 successive days, followed by oral gavage with 200 mg/kg body mass of SIL for 10 uninterrupted days. We examined the effect of SIL on specific serum biochemical parameters using an autoanalyzer and rat-specific kits. The spectrophotometric methods was used to investigate oxidative stress indicators in kidney and liver tissues. Aquaporin-2 (AQP-2), B-cell lymphoma-2 (Bcl-2), cysteine aspartate-specific protease-3 (caspase-3), interleukin-6 (IL-6), nuclear factor kappa B (NF-κB), and streptavidin-biotin staining were used to assess immunoreactivity in PAX-induced liver and kidney injury models. Results: SIL treatment significantly reduced serum levels of alanine aminotransferase, aspartate aminotransferase, creatinine, urea, and C-reactive protein, indicating its effectiveness in treating PAX-induced liver and kidney injury. SIL treatment significantly reduced oxidative stress by increasing essential antioxidant parameters, such as superoxide dismutase, catalase, glutathione peroxidase, and glutathione. It also reduced malondialdehyde levels in liver and kidney tissues of SIL-PAX groups (p < 0.05). SIL administration reduced NF-κB, caspase-3, and IL-6 expression while increasing Bcl-2 and AQP2 levels in liver and kidney tissues of rats treated with SIL and PAX (p < 0.05). Conclusion: Our findings indicate the potential of SIL to alleviate PAX-induced liver and kidney damage in rats by reducing oxidative stress, inflammation, and apoptotic processes.


Subject(s)
Apoptosis , Inflammation , Oxidative Stress , Paclitaxel , Rats, Wistar , Silymarin , Animals , Oxidative Stress/drug effects , Rats , Male , Apoptosis/drug effects , Inflammation/drug therapy , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Silymarin/pharmacology , Silymarin/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/etiology , Antioxidants/pharmacology , Antioxidants/therapeutic use , Liver/drug effects , Kidney/drug effects , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology
2.
Pak J Pharm Sci ; 37(1): 139-145, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38741410

ABSTRACT

Liposomes, a nanoscale carrier, plays an important role in the delivery of drug, affects the in vivo efficacy of drugs. In this paper, silymarin(SM)-loaded liposomes was optimized using the response surface method (RSM), with entrapment efficiency (EE%) as an index. The formulation was optimized as follow: lecithin (7.8mg/mL), SM/lecithin (1/26) and lecithin/cholesterol (10/1). The optimized SM liposomes had a high EE (96.58 ±3.06%), with a particle size of 290.3 ±10.5nm and a zeta potential of +22.98 ±1.73mV. In vitro release tests revealed that SM was released in a sustained-release manner, primarily via diffusion mechanism. In vitro cytotoxicity studies demonstrated that the prepared SM liposomes had stronger inhibitory effects than the model drug. Overall, these results indicate that this liposome system is suitable for intravenous delivery to enhance the antitumor effects of SM.


Subject(s)
Lecithins , Liposomes , Particle Size , Silymarin , Silymarin/pharmacology , Silymarin/chemistry , Silymarin/administration & dosage , Humans , Lecithins/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Drug Liberation , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol/chemistry , Chemistry, Pharmaceutical , Drug Compounding
3.
Mol Biol Rep ; 51(1): 529, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637422

ABSTRACT

BACKGROUND: TGF-ß1 and SMAD3 are particularly pathogenic in the progression of renal fibrosis. AIM: This study aimed to evaluate the kidney protective potentials of silymarin (SM) and exosomes of mesenchymal stem cells against the nephrotoxin thioacetamide (TAA) in rats. METHODS: 32 female rats were randomly assigned into four groups: the control group, the TAA group, the TAA + SM group, and the TAA + Exosomes group. The kidney homogenates from all groups were examined for expression levels of TGF-ß receptors I and II using real-time PCR, expression levels of collagen type I and CTGF proteins using ELISA, and the expression levels of nuclear SMAD2/3/4, cytoplasmic SMAD2/3, and cytoplasmic SMAD4 proteins using the western blot technique. RESULTS: Compared to the control group, the injection of TAA resulted in a significant increase in serum levels of urea and creatinine, gene expression levels of TßRI and TßRII, protein expression levels of both collagen I and CTGF proteins, cytoplasmic SMAD2/3 complex, and nuclear SMAD2/3/4 (p-value < 0.0001), with significantly decreased levels of the co-SMAD partner, SMAD4 (p-value < 0.0001). Those effects were reversed considerably in both treatment groups, with the superiority of the exosomal treatment regarding the SMAD proteins and the expression levels of the TßRI gene, collagen I, and CTGF proteins returning to near-control values (p-value > 0.05). CONCLUSION: Using in vitro and in vivo experimental approaches, the research discovered a reno-protective role of silymarin and exosomes of BM-MSCs after thioacetamide-induced renal fibrosis in rats, with the advantage of exosomes.


Subject(s)
Exosomes , Kidney Diseases , Silymarin , Rats , Female , Animals , Transforming Growth Factor beta/metabolism , Thioacetamide/toxicity , Thioacetamide/metabolism , Silymarin/pharmacology , Exosomes/metabolism , Fibrosis , Transforming Growth Factor beta1/metabolism , Kidney Diseases/pathology , Collagen Type I/metabolism , Smad Proteins/metabolism
4.
Nutrients ; 16(8)2024 Apr 14.
Article in English | MEDLINE | ID: mdl-38674860

ABSTRACT

Silymarin, salvianolic acids B, and puerarin were considered healthy food agents with tremendous potential to ameliorate non-alcoholic fatty liver disease (NAFLD). However, the mechanisms by which they interact with gut microbiota to exert benefits are largely unknown. After 8 weeks of NAFLD modeling, C57BL/6J mice were randomly divided into five groups and fed a normal diet, high-fat diet (HFD), or HFD supplemented with a medium or high dose of Silybum marianum extract contained silymarin or polyherbal extract contained silymarin, salvianolic acids B, and puerarin for 16 weeks, respectively. The untargeted metabolomics and 16S rRNA sequencing were used for molecular mechanisms exploration. The intervention of silymarin and polyherbal extract significantly improved liver steatosis and recovered liver function in the mice, accompanied by an increase in probiotics like Akkermansia and Blautia, and suppressed Clostridium, which related to changes in the bile acids profile in feces and serum. Fecal microbiome transplantation confirmed that this alteration of microbiota and its metabolites were responsible for the improvement in NAFLD. The present study substantiated that alterations of the gut microbiota upon silymarin and polyherbal extract intervention have beneficial effects on HFD-induced hepatic steatosis and suggested the pivotal role of gut microbiota and its metabolites in the amelioration of NAFLD.


Subject(s)
Depsides , Diet, High-Fat , Dietary Supplements , Gastrointestinal Microbiome , Isoflavones , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Silymarin , Animals , Gastrointestinal Microbiome/drug effects , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/drug therapy , Diet, High-Fat/adverse effects , Isoflavones/pharmacology , Male , Mice , Silymarin/pharmacology , Benzofurans/pharmacology , Liver/metabolism , Liver/drug effects , Disease Models, Animal , Bile Acids and Salts/metabolism , Plant Extracts/pharmacology
5.
Cell Signal ; 119: 111186, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38643945

ABSTRACT

Breast cancer is one of the most common cancers threatening women's health. Our previous study found that silibinin induced the death of MCF-7 and MDA-MB-231 human breast cancer cells. We noticed that silibinin-induced cell damage was accompanied by morphological changes, including the increased cell aspect ratio (cell length/width) and decreased cell area. Besides, the cytoskeleton is also destroyed in cells treated with silibinin. YAP/TAZ, a mechanical signal sensor interacted with extracellular pressure, cell adhesion area and cytoskeleton, is also closely associated with cell survival, proliferation and migration. Thus, the involvement of YAP/TAZ in the cytotoxicity of silibinin in breast cancer cells has attracted our interests. Excitingly, we find that silibinin inhibits the nuclear translocation of YAP/TAZ in MCF-7 and MDA-MB-231 cells, and reduces the mRNA expressions of YAP/TAZ target genes, ACVR1, MnSOD and ANKRD. More importantly, expression of YAP1 gene is negatively correlated with the survival of the patients with breast cancers. Molecular docking analysis reveals high probabilities for binding of silibinin to the proteins in the YAP pathways. DARTS and CETSA results confirm the binding abilities of silibinin to YAP and LATS. Inhibiting YAP pathway either by addition of verteporfin, an inhibitor of YAP/TAZ-TEAD, or by transfection of si-RNAs targeting YAP or TAZ further enhances silibinin-induced cell damage. While enhancing YAP activity by silencing LATS1/2 or overexpressing YAPS127/397A, an active form of YAP, attenuates silibinin-induced cell damage. These findings demonstrate that inhibition of the YAP/TAZ pathway contributes to cytotoxicity of silibinin in breast cancers, shedding lights on YAP/TAZ-targeted cancer therapies.


Subject(s)
Adaptor Proteins, Signal Transducing , Breast Neoplasms , Signal Transduction , Silybin , Silymarin , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Humans , Silybin/pharmacology , Silymarin/pharmacology , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction/drug effects , MCF-7 Cells , Cell Line, Tumor , Phosphoproteins/metabolism , Trans-Activators/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Cell Survival/drug effects , Molecular Docking Simulation , Cell Proliferation/drug effects , Verteporfin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects
6.
Environ Toxicol ; 39(6): 3746-3759, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38546352

ABSTRACT

BACKGROUND: Aluminum (Al) is a ubiquitous element with proven nephrotoxicity. Silymarin (SM) is a mixture of polyphenolic components extracted from Silybum marianum and exhibited protective influences. However, SM bioactivity can be enhanced by its incorporation in chitosan (CS) through the use of nanotechnology. This work proposed to assess the protective influence of SM and its loaded chitosan nanoparticles (SM-CS-NPs) on aluminum chloride (AlCl3)-induced nephrotoxicity. METHODS: Six groups were created randomly from 42 male Wistar rats and each one contains 7 rats (n = 7). Group I, acted as a control and received water. Group II received SM (15 mg/kg/day) and group III administered with SM-CS-NPs (15 mg/kg/day). Group IV received AlCl3 (34 mg/kg) and groups V and VI were treated with SM and SM-CS-NPs with AlCl3 respectively for 30 days. RESULTS: AlCl3 administration significantly elevated TBARS, H2O2, and kidney function levels besides LDH activity. Whereas GSH, CAT, SOD, GPx, GST, and GR values were all substantially reduced along with protein content, and ALP activity. Additionally, significant alterations in lipid profile, hematological parameters, and renal architecture were observed. Moreover, TNF-α, TGF-ß, and MMP9 gene expression were upregulated in kidney tissues. The administration of SM or its nanoparticles followed by AlCl3 intoxication attenuated renal dysfunction replenished the antioxidant system, and downregulated TNF-α, TGF-ß, and MMP9 gene expression in renal tissues compared to the AlCl3 group. CONCLUSION: SM-CS-NPs have more pronounced appreciated protective effects than SM and have the proficiency to balance oxidant/antioxidant systems in addition to their anti-inflammatory effect against AlCl3 toxicity.


Subject(s)
Kidney , Nanoparticles , Oxidative Stress , Protective Agents , Rats, Wistar , Silymarin , Animals , Oxidative Stress/drug effects , Male , Silymarin/pharmacology , Nanoparticles/chemistry , Nanoparticles/toxicity , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Protective Agents/pharmacology , Protective Agents/chemistry , Aluminum Chloride/toxicity , Hyperlipidemias/drug therapy , Hyperlipidemias/chemically induced , Rats , Antioxidants/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Aluminum/toxicity
7.
BMC Vet Res ; 20(1): 108, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38500117

ABSTRACT

BACKGROUND: Camel milk and silymarin have many different beneficial effects on several animal species. Meanwhile, Aflatoxins are mycotoxins with extraordinary potency that pose major health risks to several animal species. Additionally, it has been documented that aflatoxins harm the reproductive systems of a variety of domestic animals. The present design aimed to investigate the impact of aflatoxin B1 (AFB1) on rat body weight and reproductive organs and the ameliorative effects of camel milk and silymarin through measured serum testosterone, testes pathology, and gene expression of tumor necrosis factor (TNF-α), luteinizing hormone receptor (LHR), and steroidogenic acute regulatory protein (StAR) in the testes. A total of sixty mature male Wister white rats, each weighing an average of 83.67 ± 0.21 g, were used. There were six groups created from the rats. Each division had ten rats. The groups were the control (without any treatment), CM (1 ml of camel milk/kg body weight orally), S (20 mg silymarin/kg b. wt. suspension, orally), A (1.4 mg aflatoxin/kg diet), ACM (aflatoxin plus camel milk), and AS (aflatoxin plus silymarin). RESULTS: The results indicated the positive effects of camel milk and silymarin on growth, reproductive organs, and gene expression of TNF-α, LHR, and StAR with normal testicular architecture. Also, the negative effect of AFB1 on the rat's body weight and reproductive organs, as indicated by low body weight and testosterone concentration, was confirmed by the results of histopathology and gene expression. However, these negative effects were ameliorated by the ingestion of camel milk and silymarin. CONCLUSION: In conclusion, camel milk and silymarin could mitigate the negative effect of AFB1 on rat body weight and reproductive organs.


Subject(s)
Aflatoxins , Silymarin , Male , Rats , Animals , Aflatoxin B1/toxicity , Aflatoxin B1/metabolism , Silymarin/pharmacology , Camelus , Milk , Tumor Necrosis Factor-alpha/metabolism , Rats, Wistar , Testis/metabolism , Testosterone/metabolism , Body Weight
8.
Behav Brain Res ; 464: 114929, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38428646

ABSTRACT

This study evaluated the effects of citicoline and silymarin nanomicelles (SMnm) in repeated restraint stress (RRS). METHOD: Mice were exposed to RRS for four consecutive days, 2 hrs. daily. On day 5 of the study, SMnm (25 and 50 mg/kg, i.p.) and citicoline (25 and 75 mg/kg), and a combination of them (25 mg/kg, i.p.) were initiated. On day 18, anxiety-like behavior, behavioral despair, and exploratory behavior were evaluated. The prefrontal cortex (PFC) and the hippocampus were dissected measuring brain-derived neurotrophic factor (BDNF), cAMP response element-binding protein (CREB), and tumor necrosis factor-alpha (TNF-α) through Western Blot and ELISA, respectively. RESULTS: In RR-exposed mice, anxiety-like behavior in the elevated plus maze (EPM) was enhanced by reductions in open arm time (OAT%) P < 0.001, and open arm entry (OAE%) P < 0.001. In the forced swimming test (FST), the immobility increased P < 0.001 while the swimming and climbing reduced P < 0.001. In the open field test (OFT), general motor activity was raised P < 0.05. Further, body weights reduced P < 0.001, and tissue BDNF and pCREB expressions decreased P < 0.001 while TNF-α increased P < 0.001. Conversely, SMnm, citicoline and their combination could reduce anxiety-like behavior P < 0.001. The combination group reduced the depressive-like behaviors P < 0.001. Moreover, body weights were restored P < 0.001. Besides, BDNF and pCREB expressions increased while TNF-α reduced, P < 0.001. CONCLUSION: The combination synergistically improved emotion-like behaviors, alleviating the inflammation and upregulating the hippocampal BDNF-mediated CREB signaling pathway.


Subject(s)
Antidepressive Agents , Silymarin , Mice , Animals , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Cytidine Diphosphate Choline/metabolism , Cytidine Diphosphate Choline/pharmacology , Silymarin/pharmacology , Silymarin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Hippocampus/metabolism , Body Weight , Depression/metabolism
9.
Eur J Pharm Biopharm ; 198: 114272, 2024 May.
Article in English | MEDLINE | ID: mdl-38537909

ABSTRACT

Three-dimensional liver bioprinting is an emerging technology in the field of regenerative medicine that aids in the creation of functional tissue constructs that can be used as transplantable organ substitutes. During transplantation, the bioprinted donor liver must be protected from the oxidative stress environment created by various factors during the transplantation procedure, as well as from drug-induced damage from medications taken as part of the post-surgery medication regimen following the procedure. In this study, Silymarin, a flavonoid with the hepatoprotective properties were introduced into the GelMA bioink formulation to protect the bioprinted liver against hepatotoxicity. The concentration of silymarin to be added in GelMA was optimised, bioink properties were evaluated, and HepG2 cells were used to bioprint liver tissue. Carbon tetrachloride (CCl4) was used to induce hepatotoxicity in bioprinted liver, and the effect of this chemical on the metabolic activities of HepG2 cells was studied. The results showed that Silymarin helps with albumin synthesis and shields liver tissue from the damaging effects of CCl4. According to gene expression analysis, CCl4 treatment increased TNF-α and the antioxidant enzyme SOD expression in HepG2 cells while the presence of silymarin protected the bioprinted construct from CCl4-induced damage. Thus, the outcomes demonstrate that the addition of silymarin in GelMA formulation protects liver function in toxic environments.


Subject(s)
Acrylamides , Chemical and Drug Induced Liver Injury , Liver Transplantation , Silymarin , Humans , Silymarin/metabolism , Silymarin/pharmacology , Carbon Tetrachloride , Gelatin , Plant Extracts/chemistry , Living Donors , Liver , Antioxidants/metabolism , Chemical and Drug Induced Liver Injury/metabolism
10.
Pestic Biochem Physiol ; 199: 105801, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38458692

ABSTRACT

Atrazine is a widely applied herbicide to improve crop yield and maintain general health. It has been reported to impair thyroid function and architecture in experimental animals. Alterations in thyroid hormones disrupt normal body function and metabolism. Silymarin, a hepatoprotective flavonolignan, was found to improve thyroid function and body metabolism. Additionally, garlic displays several protective effects on body organs. Therefore, this study explored the prophylactic impact of natural compounds comprising silymarin and garlic extract on disrupted thyroid function, hepatic iodothyronine deiodinase type 1, and metabolic parameters in atrazine-intoxicated male rats. We found that daily pre- and co-treatment of atrazine-intoxicated male rats with silymarin (100 mg/kg, p.o) and/or garlic extract (10 mg/kg, p.o) significantly improved thyroid activation and hepatic functionality as evidenced by the re-establishment of T3, T3/T4, and TSH values as well as ALT and AST activities. Interestingly, individual or concurrent supplementation of the atrazine group with silymarin and garlic extract prevented the down-regulation in hepatic iodothyronine deiodinase type 1. These effects were coupled with the repletion of serum and hepatic antioxidants and the amelioration of lipid peroxidation. In addition, current natural products markedly alleviated weight gain, dyslipidemia, hyperglycemia, glucose intolerance, and insulin resistance. Notably, a cocktail of silymarin and garlic extract exerted superior protection against atrazine-triggered deterioration of thyroid, hepatic, and metabolic functioning to individual treatments. Present findings pinpoint the prophylactic and synergistic influence of silymarin and garlic extract combinatorial regimen on thyroid activation and body metabolism via enhancing antioxidant potential, maintaining hepatic function, and iodothyronine deiodinase type 1.


Subject(s)
Atrazine , Garlic , Silymarin , Rats , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Garlic/metabolism , Atrazine/toxicity , Silymarin/pharmacology , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology , Iodide Peroxidase/metabolism , Iodide Peroxidase/pharmacology , Liver
11.
Int J Biol Macromol ; 264(Pt 2): 130679, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38462110

ABSTRACT

Breast cancer is a major cause of death in women worldwide leading to requirement of new therapeutic strategies. Silymarin demonstrated the anti-cancer activity however, due to low bioavailability its use is restricted. This study aimed to improve the solubility of silymarin by developing a silymarin loaded zein nanoparticles (SLNPs) which was stabilized by beta cyclodextrin. Comprehensive physiochemical characterization studies based on DLS, FTIR, UV-Vis Spectroscopy, FE-SEM, TEM, XRD, DSC, NMR and TGA confirmed the successful synthesis of SLNPs via an anti-solvent precipitation method. FE-SEM and TEM images demonstrated the uniform size and spherical shape of nanoparticles with encapsulation and loading efficiencies of 84.32 ± 1.9 % and 15.25 ± 2.4 % respectively. The zein protein interaction with silymarin, and ß-cyclodextrin was shown to be beneficial via the use of molecular simulations and binding energy calculations. Cellular studies demonstrated dose and time dependent cytotoxicity of SLNPs on MCF-7 breast cancer cell. FACS, qRT-PCR and Western blotting showed Bax (pro-apoptotic) upregulation while Bcl-2 (anti-apoptotic) downregulation. Our findings suggest that these loaded nanoparticles are more efficient than pure drug, enhancing its bioavailability and paving the path for developing it as a promising nutraceutical to treat breast cancer.


Subject(s)
Breast Neoplasms , Nanoparticles , Silymarin , Zein , Female , Humans , Silymarin/pharmacology , Silymarin/chemistry , Zein/chemistry , Molecular Docking Simulation , Breast Neoplasms/drug therapy , Nanoparticles/chemistry , Particle Size
12.
Phytother Res ; 38(5): 2572-2593, 2024 May.
Article in English | MEDLINE | ID: mdl-38475999

ABSTRACT

It is suggested that supplementation with silymarin (SIL) has beneficial impacts on kidney and liver functions. This systematic review and dose-response meta-analysis assessed the impact of SIL administration on certain hepatic, renal, and oxidative stress markers. A systematic search was conducted in various databases to identify relevant trials published until January 2023. Randomized controlled trials (RCTs) that evaluated the effects of SIL on kidney and liver markers were included. A random-effects model was used for the analysis and 41 RCTs were included. The pooled results indicated that SIL supplementation led to a significant reduction in serum levels of alkaline phosphatase, alanine transaminase, creatinine, and aspartate aminotransferase, along with a substantial elevation in serum glutathione in the SIL-treated group compared to their untreated counterparts. In addition, there was a nonsignificant decrease in serum levels of gamma-glutamyl transferase, malondialdehyde (MDA), total bilirubin, albumin (Alb), total antioxidant capacity, and blood urea nitrogen. Sub-group analyses revealed a considerable decline in MDA and Alb serum values among SIL-treated participants with liver disease in trials with a longer duration (≥12 weeks). These findings suggest that SIL may ameliorate certain liver markers with potential hepatoprotective effects, specifically with long-term and high-dose supplementation. However, its nephroprotective effects and impact on oxidative stress markers were not observed. Additional high-quality RCTs with longer durations are required to determine the clinical efficacy of SIL supplementation on renal and oxidative stress markers.


Subject(s)
Dietary Supplements , Kidney , Liver , Oxidative Stress , Silymarin , Silymarin/pharmacology , Humans , Kidney/drug effects , Liver/drug effects , Oxidative Stress/drug effects , Antioxidants/pharmacology , Randomized Controlled Trials as Topic , Dose-Response Relationship, Drug , Biomarkers/blood
13.
Phytomedicine ; 128: 155493, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38484626

ABSTRACT

BACKGROUND: ID3 (inhibitor of DNA binding/differentiation-3) is a transcription factor that enables metastasis by promoting stem cell-like properties in endothelial and tumor cells. The milk thistle flavonolignan silibinin is a phytochemical with anti-metastatic potential through largely unknown mechanisms. HYPOTHESIS/PURPOSE: We have mechanistically investigated the ability of silibinin to inhibit the aberrant activation of ID3 in brain endothelium and non-small cell lung cancer (NSCLC) models. METHODS: Bioinformatic analyses were performed to investigate the co-expression correlation between ID3 and bone morphogenic protein (BMP) ligands/BMP receptors (BMPRs) genes in NSCLC patient datasets. ID3 expression was assessed by immunoblotting and qRT-PCR. Luciferase reporter assays were used to evaluate the gene sequences targeted by silibinin to regulate ID3 transcription. In silico computational modeling and LanthaScreen TR-FRET kinase assays were used to characterize and validate the BMPR inhibitory activity of silibinin. Tumor tissues from NSCLC xenograft models treated with oral silibinin were used to evaluate the in vivo anti-ID3 effects of silibinin. RESULTS: Analysis of lung cancer patient datasets revealed a top-ranked positive association of ID3 with the BMP9 endothelial receptor ACVRL1/ALK1 and the BMP ligand BMP6. Silibinin treatment blocked the BMP9-induced activation of the ALK1-phospho-SMAD1/5-ID3 axis in brain endothelial cells. Constitutive, acquired, and adaptive expression of ID3 in NSCLC cells were all significantly downregulated in response to silibinin. Silibinin blocked ID3 transcription via BMP-responsive elements in ID3 gene enhancers. Silibinin inhibited the kinase activities of BMPRs in the micromolar range, with the lower IC50 values occurring against ACVRL1/ALK1 and BMPR2. In an in vivo NSCLC xenograft model, tumoral overexpression of ID3 was completely suppressed by systematically achievable oral doses of silibinin. CONCLUSIONS: ID3 is a largely undruggable metastasis-promoting transcription factor. Silibinin is a novel suppressor of ID3 that may be explored as a novel therapeutic approach to interfere with the metastatic dissemination capacity of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Inhibitor of Differentiation Proteins , Lung Neoplasms , Neoplasm Proteins , Silybin , Silybin/pharmacology , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Humans , Animals , Cell Line, Tumor , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/genetics , Silymarin/pharmacology , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Xenograft Model Antitumor Assays , Bone Morphogenetic Protein 6 , Silybum marianum/chemistry , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Female
14.
Int J Mol Sci ; 25(4)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38396727

ABSTRACT

Silymarin has ameliorated obesity, type 2 diabetes (T2DM), and insulin resistance (IR) in combination with standard therapy, diet, or exercise in recent studies. Obesity and IR are the main risk factors for developing T2DM and other metabolic disorders. Today, there is a need for new strategies to target IR in patients with these metabolic diseases. In the present longitudinal study, a group of non-diabetic insulin-resistant women with type 1 and type 2 obesity were given silymarin for 12 weeks, with no change in habitual diet and physical activity. We used the Homeostatic Model Assessment for Insulin Resistance Index (HOMA-IR) to determine IR at baseline and after silymarin treatment (t = 12 weeks). We obtained five timepoint oral glucose tolerance tests, and other biochemical and clinical parameters were analyzed before and after treatment. Treatment with silymarin alone significantly reduced mean fasting plasma glucose (FPG) and HOMA-IR levels at 12 weeks compared to baseline values (p < 0.05). Mean fasting plasma insulin (FPI), total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), triglycerides (Tg), indirect bilirubin, and C-reactive protein (CRP) levels decreased compared to baseline values, although changes were non-significant. The overall results suggest that silymarin may offer a therapeutic alternative to improve IR in non-diabetic individuals with obesity. Further clinical trials are needed in this type of patient to strengthen the results of this study.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Silymarin , Female , Humans , Blood Glucose/metabolism , Body Mass Index , Cholesterol, HDL , Diabetes Mellitus, Type 2/metabolism , Insulin , Longitudinal Studies , Obesity/drug therapy , Obesity/metabolism , Triglycerides , Silymarin/pharmacology , Silymarin/therapeutic use
15.
Inflammopharmacology ; 32(2): 949-963, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38372848

ABSTRACT

BACKGROUND: Owing to the rich phytochemical content of Silymarin, it may effectively manage inflammation and oxidative stress. We, therefore, aimed to examine the existing evidence on the effect of Silymarin consumption on inflammation and oxidative stress factors by conducting a systematic review and meta-analysis of randomized controlled trials. METHODS: A systematic literature search up to September 2023 was completed in PubMed/Medline, Scopus, and Web of Science, to identify eligible RCTs. Heterogeneity tests of the selected trials were performed using the I2 statistic. Random effects models were assessed based on the heterogeneity tests, and pooled data were determined as weighted mean differences with a 95% confidence interval. RESULTS: Fifteen RCTs were included in this meta-analysis. Our findings showed that Silymarin consumption significantly decreased CRP (WMD, - 0.50 mg/L; 95% CI, (- 0.95 to - 0.04); p = 0.03), MDA (WMD, - 1.19 nmol/mL; 95% CI, (- 1.99 to - 0.38); p = 0.004), and IL-6 (WMD, - 0.44 pg/ml; 95% CI, (- 0.75 to - 0.12); p = 0.006). Silymarin consumption had no significant effects on IL-10, TAC, and GSH. A significant non-linear relationship was observed between the duration of the intervention and MDA changes. CONCLUSIONS: Silymarin can help reduce inflammation in patients with diabetes and thalassemia by reducing MDA as an oxidative stress marker and CRP and IL-6 as inflammatory markers.


Subject(s)
Silymarin , Adult , Humans , Biomarkers/metabolism , Dietary Supplements , Inflammation/drug therapy , Interleukin-6 , Oxidative Stress , Silymarin/pharmacology , Silymarin/therapeutic use
16.
Poult Sci ; 103(5): 103567, 2024 May.
Article in English | MEDLINE | ID: mdl-38417302

ABSTRACT

Improving productive performance is a daily challenge in the poultry industry. Developing cost-effective additives and strategies that improve performance in antibiotic-free poultry production is critical to maintaining productivity and efficiency. This study evaluates the influence of a commercially available phytogenic feed additive (CA-PFA, that comprises silymarin, betaine and curcumin extracts as main ingredients) and silymarin on commercial broilers' productive performance and liver function with and without carbon tetrachloride (CCl4)-induced liver damage. The experiment was conducted in a completely randomized design, with six treatments, eight replicates, and eight birds per replicate in 18 one-day-old male broilers (Cobb Vantress 500) each; under a 3 × 2 factorial arrangement (3 diets x 2 levels of CCl4, 0 and 1 mL/kg body weight orally). The experimental treatments included 3 diets, commercially recommended doses of CA-PFA (500 mg/kg of feed; this dose provides 70 mg/kg of silymarin, besides the other active ingredients included in the formulation), silymarin (250 mg/kg of feed, containing 28% of active ingredient; this dose provides 70 mg/kg of silymarin as active ingredient) and an additive-free basal diet as a control. A standard commercial silymarin was used as a reference due to its well-known and extensively studied hepatoprotective properties that can mitigate the negative effects of CCl4 in the liver. The data were analyzed as a 2-way ANOVA, and the means showing significant (P ≤ 0.05) differences were then compared using the Post-Hoc Tukey HSD test. No interaction was detected between factors. Exposure to CCl4 had a noticeable detrimental effect on alertness, productive performance, and liver function of broilers without a significant increase in mortality. Including CA-PFA in the diet improved productive performance compared to the basal diet from day 21 to the end of the trial, on day 42. While no influence in feed intake was detected for any treatment, CA-PFA improved body weight gain (BWG) and feed conversion ratio (FCR) significantly (P < 0.05) from day 21 to the end of the trial in healthy and CCl4-exposed birds. The results show that CA-PFA supplementation improves performance parameters in broilers with and without CCl4-induced liver damage, when compared to a basal diet and the addition of a standard commercial silymarin product.


Subject(s)
Animal Feed , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury , Chickens , Diet , Dietary Supplements , Poultry Diseases , Silymarin , Animals , Silymarin/administration & dosage , Silymarin/pharmacology , Animal Feed/analysis , Male , Diet/veterinary , Dietary Supplements/analysis , Poultry Diseases/chemically induced , Poultry Diseases/prevention & control , Chemical and Drug Induced Liver Injury/veterinary , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/etiology , Betaine/administration & dosage , Betaine/pharmacology , Random Allocation , Curcumin/administration & dosage , Curcumin/pharmacology , Liver/drug effects
17.
Cell Mol Biol (Noisy-le-grand) ; 70(1): 62-66, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38372113

ABSTRACT

We aimed to explore the role of silymarin and mitogen-activated protein kinase (MAPK) pathway in the regulation of proliferation and invasion of non-small cell lung cancer cells. Non-small cell lung cancer cells were cultured and divided into groups and treated with drugs, and A blank control group was set up. The concentration of silymarin in the experimental group was 10 mg/L, 20 mg/L and 40 mg/L, respectively, which were recorded as groups A, B and C, and three repeated experiments were performed in each group. Absorbance (A value), survival rate and number of invasions were measured at 490 nm 24 h and 48 h after treatment, and the protein expression levels of MMP-2, MMP-9, p-p38, p-JNK and p-ERK 1/2 of cells in each group were detected. There were differences in the A value (control group > Group A > Group B > Group C), cell survival rate (control group < group A < group B < group C) and the number of cell invasions (control group > Group A > Group B > group C) at 24h and 48h among all groups (P<0.05). After 24h of administration, the mRNA expression of MMP-2 and MMP-9, P-P38 and P-JNK protein expression were significantly different among groups, and the control group was > group A > Group B > group C (P<0.05). There were no significant differences in protein expression levels of p38, JNK, ERK 1/2 and P-ERK 1/2 among all groups (P>0.05). Silymarin may inhibit the proliferation and invasion of non-small cell lung cancer cells by inhibiting the activity of MAPK pathway, and the higher the concentration, the more obvious the inhibition effect, which provides a basis for further research and treatment of non-small cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Silymarin , Humans , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Silymarin/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Cell Proliferation , p38 Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System
18.
J Cosmet Dermatol ; 23(5): 1816-1827, 2024 May.
Article in English | MEDLINE | ID: mdl-38193246

ABSTRACT

BACKGROUND: The purpose of this study was to investigate the protective effect of Silibinin-loaded polymeric micelles from human hair against UV-B radiation. METHODS: Eight formulations with different concentrations of Silibinin, Pluronic F-127, and Labrasol-Labrafil were made by a solvent evaporation method, and the selected formulation was chosen by examining their properties like particle size and loading efficiency. Six groups of human hair, including a group that received the selected formulation, were exposed to UV-B radiation and by calculating its factors such as peak-to-valley roughness, RMS roughness, FTIR, and the amount of protein loss, the protective effect of the selected formulation was judged. RESULTS: According to the results, the loading efficiency and particle size of the selected formulation were 45.34% and 43.19 nm. The Silibinin release profile had two parts, fast and slow, which were suitable for creating a drug depot on hair. Its zeta potential also confirmed the minimum electrostatic interference between the formulation and hair surface. The zeta potential of selected formulation was -5.9 mv. Examination of AFM images showed that the selected formulation was able to prevent the increase in peak-to-valley roughness and RMS roughness caused by UV-B radiation. RMS roughness after 600 h of UV radiation in Groups 5 and 6 was significantly lower than the negative control group and the amount of this factor did not differ significantly between 0 and 600, so it can be concluded that the selected formulation containing Silibinin and the positive control group was able to prevent the increase of RMS roughness and hair destruction. In other hands, the two positive control groups and the selected formulation containing Silibinin were able to effectively reduce hair protein loss. CONCLUSION: Silibinin-loaded polymeric micelles were able to effectively protect hair from structural and chemical changes caused by UV-B radiation.


Subject(s)
Hair , Micelles , Particle Size , Silybin , Ultraviolet Rays , Humans , Ultraviolet Rays/adverse effects , Silybin/pharmacology , Silybin/administration & dosage , Silybin/chemistry , Hair/drug effects , Hair/radiation effects , Silymarin/pharmacology , Silymarin/administration & dosage , Silymarin/chemistry , Polymers/chemistry , Drug Liberation/radiation effects , Antioxidants/pharmacology , Antioxidants/administration & dosage , Drug Carriers/chemistry , Drug Carriers/radiation effects
19.
Planta Med ; 90(4): 298-304, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38219733

ABSTRACT

Silymarin, a widely-used hepatoprotective agent, has shown antitumor properties in both in vitro and animal studies. Currently, there is limited knowledge regarding silymarin's antitelomerase effects on human colorectal cancer and hepatocyte carcinoma cells. In this study, we investigated the antiproliferative and antitelomerase effects of silymarin on four human colorectal cancer and HepG2 hepatocyte carcinoma cell lines. The cell viability and telomerase activity were assessed using MTT and the telomerase repeat amplification protocol assay, respectively. We also investigated the effects of silymarin on the expression of human telomerase reverse transcriptase and its promoter methylation in HepG2 cells by real-time RT-PCR and methylation-specific PCR, respectively. Silymarin treatment inhibited cell proliferation and telomerase activity in all cancer cells. After 24 h of treatment, silymarin exhibited IC50 values ranging from 19 - 56.3 µg/mL against these cancer cells. A 30-min treatment with silymarin at the IC50 concentration effectively inhibited telomerase activity in cell-free extracts of both colorectal cancer and hepatocyte carcinoma cells. Treatment of HepG2 cells with 10 and 30 µg/mL of silymarin for 48 h resulted in a decrease in human telomerase reverse transcriptase expression to 75 and 35% of the level observed in the untreated control (p < 0.01), respectively. Treatment with silymarin (10, 30, and 60 µg/mL) for 48 h did not affect human telomerase reverse transcriptase promoter methylation in HepG2 cells. In conclusion, our findings suggest that silymarin inhibits cancer cell growth by directly inhibiting telomerase activity and downregulating its human telomerase reverse transcriptase catalytic subunit. However, silymarin did not affect human telomerase reverse transcriptase promoter methylation at the concentrations of 10 - 60 µg/mL used in this study.


Subject(s)
Carcinoma, Hepatocellular , Colorectal Neoplasms , Liver Neoplasms , Silymarin , Telomerase , Animals , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Silymarin/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Telomerase/genetics , Telomerase/metabolism , Cell Line, Tumor , Colorectal Neoplasms/drug therapy
20.
Mol Biol Rep ; 51(1): 118, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38227082

ABSTRACT

BACKGROUND: Ovarian cancer is one of the most lethal gynecological cancers among women worldwide. Cisplatin (Cis) is an effective chemotherapeutic agent used to treat several types of cancer. Silymarin (SLM) is an extract of medicinal plant Silybum marianum (milk thistle) with anti-inflammatory, anti-angiogenesis, antioxidant, and anticancer properties used alone or in combination with other drugs. OBJECTIVE: This study aimed to explore the effects of co-treatment with SLM and Cis on A2780 human ovarian cancer cell lines. METHODS: In this study, A2780 cells were treated with various concentrations of SLM and Cis, separately and in combination. Cell cytotoxicity, scratch, clonogenic, and flow-cytometry assays were accomplished to estimate cell viability, migration, colony formation, and apoptosis, respectively. Real-time PCR was utilized to determine the expression levels of miR-155 and miR-27a. RESULTS: SLM significantly reduced the proliferation of A2780 cells in a concentration- and time-dependent manner. Combination treatment with SLM and Cis was more potent than either single treatment in reducing viability, suppressing migration, inhibiting colony formation, and promoting the induction of apoptosis. Additionally, gene expression analysis revealed a significant decline in the expression levels of miR-155 and miR-27a in response to all separate and combined treatments, and co-treatment was more effective than individual treatments in altering miRNAs expression. CONCLUSION: Based on our findings, SLM boosts the anticancer activity of Cis and mitigates its side effects. Thus, the co-treatment of SLM and Cis can be proposed as a promising therapeutic strategy for further investigation.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Silymarin , Female , Humans , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Silymarin/pharmacology , Cell Line, Tumor , Cell Proliferation , Apoptosis , MicroRNAs/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...