1
|
Koeberle SC, Thürmer M, Su F, Werner M, Grander J, Hofer L, Gollowitzer A, Xuan LL, Benscheid FJ, Bonyadi Rad E, Zarrelli A, Di Fabio G, Werz O, Romanucci V, Lupp A, Koeberle A. Silybin A from Silybum marianum reprograms lipid metabolism to induce a cell fate-dependent class switch from triglycerides to phospholipids. Theranostics 2025; 15:2006-2034. [PMID: 39897559 PMCID: PMC11780512 DOI: 10.7150/thno.99562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/25/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Silybum marianum is used to protect against degenerative liver damage. The molecular mechanisms of its bioactive component, silybin, remained enigmatic, although membrane-stabilizing properties, modulation of membrane protein function, and metabolic regulation have been discussed for decades. Methods: Experiments were performed with hepatocyte cell lines and primary monocytes in vitro under both basal and stressed conditions, and in mice in vivo. Quantitative lipidomics was used to detect changes in phospholipids and triglycerides. Key findings were confirmed by Western blotting, quantitative PCR, microscopy, enzyme activity assays, metabolic flux studies, and functional relationships were investigated using selective inhibitors. Results: We show that specifically the stereoisomer silybin A decreases triglyceride levels and lipid droplet content, while enriching major phospholipid classes and maintaining a homeostatic phospholipid composition in human hepatocytes in vitro and in mouse liver in vivo under normal and pre-disease conditions. Conversely, in cell-based disease models of lipid overload and lipotoxic stress, silybin treatment primarily depletes triglycerides. Mechanistically, silymarin/silybin suppresses phospholipid-degrading enzymes, induces phospholipid biosynthesis to varying degrees depending on the conditions, and down-regulates triglyceride remodeling/biosynthesis, while inducing complex changes in sterol and fatty acid metabolism. Structure-activity relationship studies highlight the importance of the 1,4-benzodioxane ring configuration of silybin A in triglyceride reduction and the saturated 2,3-bond of the flavanonol moiety in phospholipid accumulation. Enrichment of hepatic phospholipids and intracellular membrane expansion are associated with a heightened biotransformation capacity. Conclusion: Our study deciphers the structural features of silybin contributing to hepatic lipid remodeling and suggests that silymarin/silybin protects the liver in individuals with mild metabolic dysregulation, involving a lipid class switch from triglycerides to phospholipids, whereas it may be less effective in disease states associated with severe metabolic dysregulation.
Collapse
Affiliation(s)
- Solveigh C. Koeberle
- Institute of Pharmaceutical Sciences/Pharmacognosy and Excellence Field BioHealth, University of Graz, 8010 Graz, Austria
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Maria Thürmer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Fengting Su
- Institute of Pharmaceutical Sciences/Pharmacognosy and Excellence Field BioHealth, University of Graz, 8010 Graz, Austria
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Markus Werner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Laura Hofer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Loc Le Xuan
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Felix J. Benscheid
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Ehsan Bonyadi Rad
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Armando Zarrelli
- Department of Chemical Sciences, University of Napoli Federico II, I-80126 Naples, Italy
| | - Giovanni Di Fabio
- Department of Chemical Sciences, University of Napoli Federico II, I-80126 Naples, Italy
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Valeria Romanucci
- Department of Chemical Sciences, University of Napoli Federico II, I-80126 Naples, Italy
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Andreas Koeberle
- Institute of Pharmaceutical Sciences/Pharmacognosy and Excellence Field BioHealth, University of Graz, 8010 Graz, Austria
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
2
|
Alnuqaydan AM. Riddelline from Tamarix articulate as a potential anti-bacterial lead compound for novel antibiotics discovery: A comprehensive computational and toxicological studies. PLoS One 2024; 19:e0310319. [PMID: 39541292 PMCID: PMC11563397 DOI: 10.1371/journal.pone.0310319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 08/28/2024] [Indexed: 11/16/2024] Open
Abstract
Tamarix articulate from the Tamaricaece family is a halophytic plant. This plant is commonly called Athal or Tamarix in different Arabic and Asian countries. Due to the high load of polyphenolic phytochemicals, the plant has been used as a therapeutic option against several diseases for decades. The plant is an anti-inflammatory, anti-bacterial, anti-viral, anti-cancer, anti-oxidant, and anti-inflammatory. In this work, the 222 phytochemical compounds of T. articulate from our previous study are used in different bioinformatic and biophysics techniques to explore their biological potency against different anti-bacterial, anti-cancer and anti-viral targets. By doing so, it was found that Riddelline ranked as the best binding molecule of biological macromolecules selected herein in particular the bacterial targets. The binding energy value of the compound for the KdsA enzyme was -14.64 kcal/mol, KdsB (-13.09 kcal/mol), MurC (-13.67 kcal/mol), MurD (-13.54 kcal/mol), MurF (-14.20 kcal/mol), Polo-like kinase 1 (Plk1) (-12.34 kcal/mol), Bcl-2 protein (-13.39 kcal/mol), SARS-CoV-2 main protease enzyme (-12.67 kcal/mol), and Human T cell leukemia virus protease (-13.67 kcal/mol). The mean Rg value of KdsA-Riddelline complex and KdsA-FPE complex is 32.67 Å, and average RMSD of KdsA-Riddelline complex and KdsA-FPE complex is 2.31 Å, respectively. The binding energy complexes was found to be dominated by van der Waals (-71.98 kcal/mol for KdsA-Riddelline complex and -65.09 kcal/mol for KdsA-FPE complex). The lead compound was also unveiled to show favorable druglike properties and pharmacokinetics. Together, the data suggest the good anti-bacterial activities of the T. articulate phytochemicals and thus can be subjected to experimental in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Abdullah M. Alnuqaydan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
3
|
Rivas-Caceres RR, Khazaei R, Ponce-Covarrubias JL, Di Rosa AR, Ogbuagu NE, Estrada GT, Zigo F, Gorlov IF, Slozhenkina MI, Mosolov AA, Lackner M, Elghandour MMMY. Effects of dietary Silybum marianum powder on growth performance, egg and carcass characteristics, immune response, intestinal microbial population, haemato-biochemical parameters and sensory meat quality of laying quails. Poult Sci 2024; 103:104036. [PMID: 39079329 PMCID: PMC11340560 DOI: 10.1016/j.psj.2024.104036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/15/2024] [Accepted: 06/23/2024] [Indexed: 08/25/2024] Open
Abstract
The study aimed to assess the effects of different dietary Silybum marianum (SM, milk thistle) powder levels on growth performance, productivity, immunity, small intestine, haemato-biochemical parameters, meat quality, and egg and carcass characteristics of laying quails. The experimental subjects consisted of one hundred and eight 43-day-old quails divided into 3 treatments (0, 0.75, and 1.50% SM) with 4 replicates each. The egg characteristics and growth performance of the quails were evaluated. Quails were euthanized for evaluation of carcasses, microbiota, and sensory characteristics of meat. Blood samples were analyzed for haematology and biochemical profile. SM at 0.75% and 1.50% significantly (P < 0.05) increased feed intake, enhanced egg characteristics (number, weight, width, length, volume, weight of egg yolk, and eggshell thickness), jejunum and ileum length, spleen weight, lactobacillus population, sensory characteristics of meat, red blood cell (RBC), hemoglobin, erythrocytic indices, concentration of albumin, globulin and thyroid stimulating hormone (TSH). SM at 0.75% and 1.50% decreased (P < 0.05) carcass weight (abdominal fat, heart, neck, and pancreas), feed conversion ratio (FCR) based on eggs produced, percentages of heterophils and lymphocytes, concentration of lactate dehydrogenase, population of coliforms clostridia, and Escherichia coli. Aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, and creatine kinase were not significantly (P > 0.05) altered by 0.75% and 1.50% SM. SM at both levels (0.75% and 1.50%) may improve growth, egg characteristics, immune response, intestinal morphology and microbiota, meat quality and erythropoiesis, and also lead to decreased cholesterol in laying quails. Economics can be improved, too. The authors recommend adding 1.0% of SM to quail diet.
Collapse
Affiliation(s)
- Raymundo Rene Rivas-Caceres
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez 32310, Chihuahua, México
| | - Roshanak Khazaei
- Department of Animal Science, Rasht Branch, Islamic Azad University, Rasht, 4147654919, Iran
| | - José Luis Ponce-Covarrubias
- Escuela Superior de Medicina Veterinaria y Zootecnia No. 3, Universidad Autónoma de Guerrero (UAGro), Técpan de Galeana 40900, Guerrero, México
| | - Ambra Rita Di Rosa
- Department of Veterinary Sciences, University of Messina, Messina, 98168, Italy
| | - Ngozi Ejum Ogbuagu
- Department of Veterinary Physiology, Ahmadu Bello University Zaria, 810107, Nigeria
| | - Gustavo Tirado Estrada
- Departamento de Ingenierías, Instituto Tecnológico El Llano Aguascalientes (ITEL)/Tecnológico Nacional de México (TecNM), El Llano 20330, Aguascalientes, México
| | - František Zigo
- Department of Animal Nutrition and Husbandry, University of Veterinary Medicine and Pharmacy, 041 81 Košice, Slovakia
| | - Ivan F Gorlov
- Volga Region Research Institute of Manufacture and Processing of Meat and Milk Production, Volgograd 400131, Russia
| | - Marina I Slozhenkina
- Volga Region Research Institute of Manufacture and Processing of Meat and Milk Production, Volgograd 400131, Russia
| | - Aleksandr A Mosolov
- Volga Region Research Institute of Manufacture and Processing of Meat and Milk Production, Volgograd 400131, Russia
| | - Maximilian Lackner
- Department of Industrial Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria
| | - Mona M M Y Elghandour
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Estado de México, 50000, México.
| |
Collapse
|
4
|
Lai W, Zhang J, Sun J, Min T, Bai Y, He J, Cao H, Che Q, Guo J, Su Z. Oxidative stress in alcoholic liver disease, focusing on proteins, nucleic acids, and lipids: A review. Int J Biol Macromol 2024; 278:134809. [PMID: 39154692 DOI: 10.1016/j.ijbiomac.2024.134809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Oxidative stress is one of the important factors in the development of alcoholic liver disease. The production of reactive oxygen species and other free radicals is an important feature of alcohol metabolism in the liver and an important substance in liver injury. When large amounts of ROS are produced, the homeostasis of the liver REDOX system will be disrupted and liver injury will be caused. Oxidative stress can damage proteins, nucleic acids and lipids, liver dysfunction. In addition, damaging factors produced by oxidative damage to liver tissue can induce the occurrence of inflammation, thereby aggravating the development of ALD. This article reviews the oxidative damage of alcohol on liver proteins, nucleic acids, and lipids, and provides new insights and summaries of the oxidative stress process. We also discussed the relationship between oxidative stress and inflammation in alcoholic liver disease from different perspectives. Finally, the research status of antioxidant therapy in alcoholic liver disease was summarized, hoping to provide better help for learning and developing the understanding of alcoholic liver disease.
Collapse
Affiliation(s)
- Weiwen Lai
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiahua Zhang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiawei Sun
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tianqi Min
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Jincan He
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd, Science City, Guangzhou 510663, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
5
|
Yavuz A, Küçük A, Ergörün Aİ, Dursun AD, Yiğman Z, Alkan M, Arslan M. Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation. Exp Ther Med 2024; 27:242. [PMID: 38655036 PMCID: PMC11036365 DOI: 10.3892/etm.2024.12530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/27/2023] [Indexed: 04/26/2024] Open
Abstract
Sepsis is a systemic inflammatory response syndrome that develops in the host against microorganisms. This response develops away from the primary infection site and results in end-organ damage. The present study aimed to investigate the protective and therapeutic effects on lung and kidney tissue of silymarin (S) and dexmedetomidine (DEX) applied 1 h before and after sepsis induced by the cecal ligation and puncture (CLP) method in rats. A total of 62 rats was randomly divided into eight groups: i) Control (n=6); ii) cecal perforation (CLP; n=8); iii) S + CLP (n=8; S + CLP; S administered 1 h before CPL); iv) CLP + S (n=8; S administered 1 h after CLP); v) DEX + CLP (n=8; D + CLP; DEX administered 1 h before CLP); vi) CLP + D (n=8; DEX administered 1 h after CLP); vii) SD + CLP (n=8; S and DEX administered 1 h before CLP) and viii) CLP + SD (n=8; S and DEX administered 1 h after CLP). After the cecum filled with stool, it was tied with 3/0 silk under the ileocecal valve and the anterior surface of the cecum was punctured twice with an 18-gauge needle. A total of 100 mg/kg silymarin and 100 µg/kg DEX were administered intraperitoneally to the treatment groups. Lung and kidney tissue samples were collected to evaluate biochemical and histopathological parameters. In the histopathological examination, all parameters indicating kidney injury; interstitial edema, peritubular capillary dilatation, vacuolization, ablation of tubular epithelium from the basement membrane, loss of brush border in the proximal tubule epithelium, cell swelling and nuclear defragmentation; were increased in the CLP compared with the control group. Silymarin administration increased kidney damage, including ablation of tubular epithelium from the basement membrane, compared with that in the CLP group. DEX significantly reduced kidney damage compared with the CLP and silymarin groups. The co-administration of DEX + silymarin decreased kidney damage, although it was not as effective as DEX-alone. To conclude, intraperitoneal DEX ameliorated injury in CLP rats. DEX + silymarin partially ameliorated injury but silymarin administration increased damage. As a result, silymarin has a negative effects with this dosage and DEX has a protective effect. In the present study, it was determined that using the two drugs together had a greater therapeutic effect than silymarin and no differences in the effects were not observed any when the application times of the agents were changed.
Collapse
Affiliation(s)
- Aydin Yavuz
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ayşegül Küçük
- Department of Physiology, Faculty of Medicine, Kutahya Health Science University, Kutahya 43020, Turkey
| | - Aydan İremnur Ergörün
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ali Doğan Dursun
- Department of Physiology, Faculty of Medicine, Atılım University, Ankara 06830, Turkey
| | - Zeynep Yiğman
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
- Neuroscience and Neurotechnology Center of Excellence, Gazi University, Ankara 06510, Turkey
| | - Metin Alkan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Mustafa Arslan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
- Life Sciences Application and Research Center, Gazi University, Ankara 06830, Turkey
- Laboratory Animal Breeding and Experimental Research Center, Gazi University, Ankara 06510, Turkey
| |
Collapse
|
6
|
Jaffar HM, Al‐Asmari F, Khan FA, Rahim MA, Zongo E. Silymarin: Unveiling its pharmacological spectrum and therapeutic potential in liver diseases-A comprehensive narrative review. Food Sci Nutr 2024; 12:3097-3111. [PMID: 38726410 PMCID: PMC11077231 DOI: 10.1002/fsn3.4010] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 05/12/2024] Open
Abstract
Liver diseases, encompassing conditions such as cirrhosis, present a substantial global health challenge with diverse etiologies, including viral infections, alcohol consumption, and non-alcoholic fatty liver disease (NAFLD). The exploration of natural compounds as therapeutic agents has gained traction, notably the herbal remedy milk thistle (Silybum marianum), with its active extract, silymarin, demonstrating remarkable antioxidant and hepatoprotective properties in extensive preclinical investigations. It can protect healthy liver cells or those that have not yet sustained permanent damage by reducing oxidative stress and mitigating cytotoxicity. Silymarin, a natural compound with antioxidant properties, anti-inflammatory effects, and antifibrotic activity, has shown potential in treating liver damage caused by alcohol, NAFLD, drug-induced toxicity, and viral hepatitis. Legalon® is a top-rated medication with excellent oral bioavailability, effective absorption, and therapeutic effectiveness. Its active component, silymarin, has antioxidant and hepatoprotective properties, Eurosil 85® also, a commercial product, has lipophilic properties enhanced by special formulation processes. Silymarin, during clinical trials, shows potential improvements in liver function, reduced mortality rates, and alleviation of symptoms across various liver disorders, with safety assessments showing low adverse effects. Overall, silymarin emerges as a promising natural compound with multifaceted hepatoprotective properties and therapeutic potential in liver diseases.
Collapse
Affiliation(s)
- Hafiza Madiha Jaffar
- University Institute of Diet & Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
| | - Fahad Al‐Asmari
- Department of Food and Nutrition Sciences, College of Agricultural and Food SciencesKing Faisal UniversityAl‐AhsaSaudi Arabia
| | - Faima Atta Khan
- University Institute of Diet & Nutritional Sciences, Faculty of Allied Health SciencesThe University of LahoreLahorePakistan
- Department of Food Science, Faculty of Life SciencesGovernment College UniversityFaisalabadPakistan
| | - Muhammad Abdul Rahim
- Department of Food Science, Faculty of Life SciencesGovernment College UniversityFaisalabadPakistan
- Department of Food Science & Nutrition, Faculty of Medicine and Allied Health SciencesTimes InstituteMultanPakistan
| | - Eliasse Zongo
- Laboratoire de Recherche et d'Enseignement en Santé et Biotechnologies AnimalesUniversité Nazi BONIBobo DioulassoBurkina Faso
| |
Collapse
|
7
|
Islam Shawon S, Nargis Reyda R, Qais N. Medicinal herbs and their metabolites with biological potential to protect and combat liver toxicity and its disorders: A review. Heliyon 2024; 10:e25340. [PMID: 38356556 PMCID: PMC10864916 DOI: 10.1016/j.heliyon.2024.e25340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
The liver is an essential organ that helps the body with immunity, metabolism, and detoxification, among other functions. Worldwide, liver illnesses are a leading cause of mortality and disability. There are few effective treatment choices, but they frequently have unfavorable side effects. Investigating the potential of medicinal plants and their bioactive phytoconstituents in the prevention and treatment of liver disorders has gained more attention in recent years. An assessment of the hepatoprotective potential of medicinal plants and their bioactive secondary metabolites is the goal of this thorough review paper. To determine their hepatoprotective activity, these plants were tested against liver toxicity artificially induced in rats, mice and rabbits by chemical agents such as carbon tetrachloride (CCl4), paracetamol (PCM), thioacetamide (TAA), N-nitrosodiethylamine, d-galactosamine/lipopolysaccharide, antitubercular medicines (rifampin, isoniazid) and alcohol. To find pertinent research publications published between 1989 and 2022, a comprehensive search of electronic bibliographic databases (including Web of Science, SpringerLink, ScienceDirect, Google Scholar, PubMed, Scopus, and others) was carried out. The investigation comprised 203 plant species from 81 families in total. A thorough discussion was mentioned regarding the hepatoprotective qualities of plants belonging to several families, such as Fabaceae, Asteraceae, Lamiaceae, and Euphorbiaceae. The plant groups Asteraceae and Fabaceae were the most frequently shown to have hepatoprotective properties. The phytochemical constituents namely flavonoids, phenolic compounds, and alkaloids exhibited the highest frequency of hepatoprotective action. Also, some possible mechanism of action of some active constituents from medicinal plants was discussed in brief which were found in some studies. In summary, the information on medicinal plants and their potentially hepatoprotective bioactive phytoconstituents has been consolidated in this review which emphasizes the importance of further research to explore the efficacy and safety of these natural remedies for various liver ailments.
Collapse
Affiliation(s)
- Shahparan Islam Shawon
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Rashmia Nargis Reyda
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Nazmul Qais
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| |
Collapse
|
8
|
Kim TH, Heo SY, Chandika P, Kim YM, Kim HW, Kang HW, Je JY, Qian ZJ, Kim N, Jung WK. A literature review of bioactive substances for the treatment of periodontitis: In vitro, in vivo and clinical studies. Heliyon 2024; 10:e24216. [PMID: 38293511 PMCID: PMC10826675 DOI: 10.1016/j.heliyon.2024.e24216] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/16/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024] Open
Abstract
Periodontitis is a common chronic inflammatory disease of the supporting tissues of the tooth that involves a complex interaction of microorganisms and various cell lines around the infected site. To prevent and treat this disease, several options are available, such as scaling, root planning, antibiotic treatment, and dental surgeries, depending on the stage of the disease. However, these treatments can have various side effects, including additional inflammatory responses, chronic wounds, and the need for secondary surgery. Consequently, numerous studies have focused on developing new therapeutic agents for more effective periodontitis treatment. This review explores the latest trends in bioactive substances with therapeutic effects for periodontitis using various search engines. Therefore, this study aimed to suggest effective directions for therapeutic approaches. Additionally, we provide a summary of the current applications and underlying mechanisms of bioactive substances, which can serve as a reference for the development of periodontitis treatments.
Collapse
Affiliation(s)
- Tae-Hee Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
| | - Seong-Yeong Heo
- Jeju Marine Research Center, Korea Institute of Ocean Science & Technology (KIOST), Jeju, 63349, Republic of Korea
| | - Pathum Chandika
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Hyun-Woo Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Department of Marine Biology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Hyun Wook Kang
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 48513, Republic of Korea
| | - Jae-Young Je
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Major of Human Bioconvergence, School of Smart Healthcare, Pukyong National University, Busan, 48513, Republic of Korea
| | - Zhong-Ji Qian
- College of Food Science and Technology, School of Chemistry and Environment, Guangdong Ocean University, Zhanjiang, 524088, China
- Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Shenzhen, 518108, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524025, China
| | - Namwon Kim
- Ingram School of Engineering, Texas State University, San Marcos, TX, 78666, USA
- Materials Science, Engineering, and Commercialization (MSEC), Texas State University, San Marcos, TX, 78666, USA
| | - Won-Kyo Jung
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 48513, Republic of Korea
| |
Collapse
|
9
|
Bai Y, Wang L, TingYang, Wang L, Ge W. Silymarin ameliorates peritoneal fibrosis by inhibiting the TGF-β/Smad signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2379-2391. [PMID: 37052642 DOI: 10.1007/s00210-023-02450-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/24/2023] [Indexed: 04/14/2023]
Abstract
Peritoneal dialysis (PD) is the mainstay of treatment for renal failure replacement therapy. Although PD has greatly improved the quality of life of end-stage renal disease (ESRD) patients, long-term PD can lead to ultrafiltration failure, which in turn causes peritoneal fibrosis (PF). Silymarin (SM) is a polyphenolic flavonoid isolated from the milk thistle (Silybum marianum) species that has a variety of pharmacological actions, including antioxidant, anti-inflammatory, antiviral, and anti-fibrotic pharmacological activities. However, the effect of SM on PF and its potential mechanisms have not been clarified. The aim of this study was to investigate the preventive effect of SM on PF in vitro and in vivo as well as elucidate the underlying mechanisms. We established PF mouse models and human pleural mesothelial cell fibrosis in vitro by intraperitoneal injection of high-glucose peritoneal dialysis solution (PDS) or transforming growth factor-β1 (TGF-β1), and evaluated the effect of SM on peritoneal fibrosis in vivo and in vitro. We found that SM alleviated peritoneal dysfunction. Meanwhile, SM inhibited the expression of fibrotic markers (TGF-β1, collagen I, fibronectin) and restored the expression of E-cadherin, BMP-7 in PF mice and TGF-β1-treated Met-5A cells. Furthermore, SM markedly down-regulated the expression of TGF-β1, p-Smad2, and p-Smad3 and up-regulated the expression of smad7. In conclusion, these findings suggested that SM may be an efficient and novel therapy for the prevention of PF through inhibition of TGF-β/Smad signaling.
Collapse
Affiliation(s)
- Yingwen Bai
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Lulu Wang
- Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu Province, China
| | - TingYang
- Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu Province, China
| | - Lingyun Wang
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Weihong Ge
- Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu Province, China.
| |
Collapse
|
10
|
Wang Y, Chen Q, Wu S, Sun X, Yin R, Ouyang Z, Yin H, Wei Y. Amelioration of ethanol-induced oxidative stress and alcoholic liver disease by in vivo RNAi targeting Cyp2e1. Acta Pharm Sin B 2023; 13:3906-3918. [PMID: 37719371 PMCID: PMC10502278 DOI: 10.1016/j.apsb.2023.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/16/2022] [Accepted: 12/10/2022] [Indexed: 09/19/2023] Open
Abstract
Alcoholic liver disease (ALD) results from continuous and heavy alcohol consumption. The current treatment strategy for ALD is based on alcohol withdrawal coupled with antioxidant drug intervention, which is a long process with poor efficacy and low patient compliance. Alcohol-induced CYP2E1 upregulation has been demonstrated as a key regulator of ALD, but CYP2E1 knockdown in humans was impractical, and pharmacological inhibition of CYP2E1 by a clinically relevant approach for treating ALD was not shown. In this study, we developed a RNAi therapeutics delivered by lipid nanoparticle, and treated mice fed on Lieber-DeCarli ethanol liquid diet weekly for up to 12 weeks. This RNAi-based inhibition of Cyp2e1 expression reduced reactive oxygen species and oxidative stress in mouse livers, and contributed to improved ALD symptoms in mice. The liver fat accumulation, hepatocyte inflammation, and fibrosis were reduced in ALD models. Therefore, this study suggested the feasibility of RNAi targeting to CYP2E1 as a potential therapeutic tool to the development of ALD.
Collapse
Affiliation(s)
- Yalan Wang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Qiubing Chen
- Department of Urology, Frontier Science Centre for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Shuang Wu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Xinyu Sun
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Runting Yin
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Zhen Ouyang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Hao Yin
- Department of Urology, Frontier Science Centre for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- RNA Institute, Wuhan University, Wuhan 430072, China
- Wuhan Research Centre for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430010, China
| | - Yuan Wei
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
11
|
Tao G, Liu F, Jin Z, Liu B, Wang H, Li D, Tang W, Chen Y, He Q, Qin S. A strategy of local hydrogen capture and catalytic hydrogenation for enhanced therapy of chronic liver diseases. Theranostics 2023; 13:2455-2470. [PMID: 37215568 PMCID: PMC10196827 DOI: 10.7150/thno.80494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/08/2023] [Indexed: 05/24/2023] Open
Abstract
Background: Chronic liver diseases (CLD) frequently derive from hepatic steatosis, inflammation and fibrosis, and become a leading inducement of cirrhosis and hepatocarcinoma. Molecular hydrogen (H2) is an emerging wide-spectrum anti-inflammatory molecule which is able to improve hepatic inflammation and metabolic dysfunction, and holds obvious advantages in biosafety over traditional anti-CLD drugs, but existing H2 administration routes cannot realize the liver-targeted high-dose delivery of H2, severely limiting its anti-CLD efficacy. Method: In this work, a concept of local hydrogen capture and catalytic hydroxyl radical (·OH) hydrogenation is proposed for CLD treatment. The mild and moderate non-alcoholic steatohepatitis (NASH) model mice were intravenously injected with PdH nanoparticles firstly, and then daily inhaled 4% hydrogen gas for 3 h throughout the whole treatment period. After the end of treatment, glutathione (GSH) was intramuscularly injected every day to assist the Pd excretion. Results: In vitro and in vivo proof-of-concept experiments have confirmed that Pd nanoparticles can accumulate in liver in a targeted manner post intravenous injection, and play a dual role of hydrogen captor and ·OH filter to locally capture/store the liver-passing H2 during daily hydrogen gas inhalation and rapidly catalyze the ·OH hydrogenation into H2O. The proposed therapy significantly improves the outcomes of hydrogen therapy in the prevention and treatment of NASH by exhibiting a wide range of bioactivity including the regulation of lipid metabolism and anti-inflammation. Pd can be mostly eliminated after the end of treatment under the assistance of GSH. Conclusion: Our study verified a catalytic strategy of combining PdH nanoparticles and hydrogen inhalation, which exhibited enhanced anti-inflammatory effect for CLD treatment. The proposed catalytic strategy will open a new window to realize safe and efficient CLD treatment.
Collapse
Affiliation(s)
- Geru Tao
- Key Laboratory of Major Diseases and Hydrogen Medical Translational Applications in Universities of Shandong Province & Key Laboratory of Hydrogen Biomedical Research of Health Commission of Shandong Province, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
- Taishan Institute for Hydrogen Biomedical Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
| | - Feng Liu
- Key Laboratory of Major Diseases and Hydrogen Medical Translational Applications in Universities of Shandong Province & Key Laboratory of Hydrogen Biomedical Research of Health Commission of Shandong Province, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
- Taishan Institute for Hydrogen Biomedical Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
| | - Zhaokui Jin
- School of Biomedical Engineering, Guangzhou Medical University, Guangdong, 511495 China
| | - Boyan Liu
- Key Laboratory of Major Diseases and Hydrogen Medical Translational Applications in Universities of Shandong Province & Key Laboratory of Hydrogen Biomedical Research of Health Commission of Shandong Province, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
- Taishan Institute for Hydrogen Biomedical Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
| | - Hao Wang
- Key Laboratory of Major Diseases and Hydrogen Medical Translational Applications in Universities of Shandong Province & Key Laboratory of Hydrogen Biomedical Research of Health Commission of Shandong Province, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
- Taishan Institute for Hydrogen Biomedical Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
| | - Daosheng Li
- Pathology Department of Tai'an City Central Hospital, Tai'an 271016, China
| | - Wei Tang
- Key Laboratory of Human-Machine-Intelligence Synergic System, Research Center for Neural Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Yuan Chen
- Key Laboratory of Major Diseases and Hydrogen Medical Translational Applications in Universities of Shandong Province & Key Laboratory of Hydrogen Biomedical Research of Health Commission of Shandong Province, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
- Taishan Institute for Hydrogen Biomedical Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
| | - Qianjun He
- Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Shenzhen Research Institute, Shanghai Jiao Tong University, Shenzhen 518057, China
| | - Shucun Qin
- Key Laboratory of Major Diseases and Hydrogen Medical Translational Applications in Universities of Shandong Province & Key Laboratory of Hydrogen Biomedical Research of Health Commission of Shandong Province, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
- Taishan Institute for Hydrogen Biomedical Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271000, China
| |
Collapse
|
12
|
Natural and synthetic compounds for glioma treatment based on ROS-mediated strategy. Eur J Pharmacol 2023:175537. [PMID: 36871663 DOI: 10.1016/j.ejphar.2023.175537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/08/2023] [Accepted: 01/23/2023] [Indexed: 03/06/2023]
Abstract
Glioma is the most frequent and most malignant tumor of the central nervous system (CNS),accounting for about 50% of all CNS tumor and approximately 80% of the malignant primary tumors in the CNS. Patients with glioma benefit from surgical resection, chemo- and radio-therapy. However these therapeutical strategies do not significantly improve the prognosis, nor increase survival rates owing to restricted drug contribution in the CNS and to the malignant characteristics of glioma. Reactive oxygen species (ROS) are important oxygen-containing molecules that regulate tumorigenesis and tumor progression. When ROS accumulates to cytotoxic levels, this can lead to anti-tumor effects. Multiple chemicals used as therapeutic strategies are based on this mechanism. They regulate intracellular ROS levels directly or indirectly, resulting in the inability of glioma cells to adapt to the damage induced by these substances. In the current review, we summarize the natural products, synthetic compounds and interdisciplinary techniques used for the treatment of glioma. Their possible molecular mechanisms are also presented. Some of them are also used as sensitizers: they modulate ROS levels to improve the outcomes of chemo- and radio-therapy. In addition, we summarize some new targets upstream or downstream of ROS to provide ideas for developing new anti-glioma therapies.
Collapse
|
13
|
Lin S, Huang L, Wu Y, Huang L, Wu P, Huang T, Li Z, Hu Y. Uncovering the protective mechanism of Pien-Tze-Huang in rat with alcoholic liver injury based on cytokines analysis and untargeted metabonomics. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1217:123626. [PMID: 36753840 DOI: 10.1016/j.jchromb.2023.123626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 12/17/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Pien-Tze-Huang (PTH) is a well-known traditional Chinese patent medicine with excellent liver-protection effect. However, the mechanism of hepatoprotective action has not yet been entirely elucidated. The aim of this study was to investigate the mechanism of protective effect of PTH on alcohol-induced liver injury in rats using cytokine analysis and untargeted metabolomics approaches. An alcoholic liver disease (ALD) model with SD rats was established, and PTH was administered according to the prescribed dose. The hepatoprotective effect of PTH was evaluated by pathological observation of liver tissue and changes in biochemical index activity and cytokines in serum. Serum samples were analyzed by ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UHPLC-QTOF/MS), and differentially expressed metabolites were screened by multivariate statistical analysis. KEGG combined with metabolic pathway analysis were used to evaluate the underlying metabolic pathways. Results showed liver histopathology injury was attenuated. The levels of IL-6, TNF-α and NF-κB were significantly decreased in rats intervened with PTH groups, suggesting that it may alleviate inflammation via suppressing the inflammatory cytokines signaling pathway. Eighty differentially expressed metabolites were found and identified. Pathway analysis indicated that the hepatoprotective effects of PTH occurred through the regulation of inflammatory cytokines signaling pathway, primary bile acid biosynthesis, vitamin B6 metabolism pathway, cholesterol metabolism, and tyrosine metabolism. PTH showed favorable hepatoprotective effect through multiple pathways. This study has great importance in fully revealing the mechanism of hepatoprotective action and can help improve the clinical application of PTH.
Collapse
Affiliation(s)
- Shouer Lin
- School of Pharmacy, Fujian Medical university, Fuzhou, Fujian, 350122, China; Physical and Chemical Analysis Department, Fujian Provincial Center For Disease Control and Prevention, Fujian Provincial Key Laboratory of Zoonosis Research, Fuzhou, Fujian, 350001, China
| | - Lingyi Huang
- School of Pharmacy, Fujian Medical university, Fuzhou, Fujian, 350122, China
| | - Youjia Wu
- School of Pharmacy, Fujian Medical university, Fuzhou, Fujian, 350122, China
| | - Liying Huang
- School of Pharmacy, Fujian Medical university, Fuzhou, Fujian, 350122, China.
| | - Pingping Wu
- School of Pharmacy, Fujian Medical university, Fuzhou, Fujian, 350122, China
| | - Tingxuan Huang
- School of Pharmacy, Fujian Medical university, Fuzhou, Fujian, 350122, China
| | - Zhenyue Li
- School of Pharmacy, Fujian Medical university, Fuzhou, Fujian, 350122, China
| | - Yuhan Hu
- School of Pharmacy, Fujian Medical university, Fuzhou, Fujian, 350122, China
| |
Collapse
|
14
|
Yue S, Wang S, Liu X, Bian X, Ding C, Wu T, Li D, Zhou J. Ameliorative effect of silymarin on the quality of frozen-thawed boar spermatozoa. Reprod Domest Anim 2023; 58:298-306. [PMID: 36269155 DOI: 10.1111/rda.14286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 11/28/2022]
Abstract
Although Silymarin (SMN) has powerful antioxidant properties, little is known about its effects on the quality of frozen-thawed boar sperm. The present study aimed to evaluate the influences of SMN added to the thawing extender on boar sperm parameters essential for fertilization. The frozen-thawed semen was diluted in a Modena thawing extender supplemented with different concentrations of SMN (0, 5, 10, 20 and 50 μM respectively), and then the changes in quality parameters, antioxidant capacity, mitochondrial function and in vitro fertilization (IVF) capability of frozen-thawed sperm were assessed. Here we demonstrated that the motility, plasma membrane integrity and acrosomal integrity of frozen-thawed sperm improved efficiently by SMN (p < .05). In antioxidant parameters evaluation, the tROS level and MDA content of frozen-thawed spermatozoa were reduced in the 20 μM SMN group, while the T-AOC activity significantly increased (p < .05), indicating that the supplementation with SMN can promote the antioxidant capacity of frozen-thawed boar sperm. Besides, we also discovered that the addition of SMN significantly upregulated ATP content and enhanced the mitochondrial activity of sperm. More interestingly, SMN promoted the activities of mitochondrial respiratory chain complexes (MRCC) I, II, III and IV in frozen-thawed sperm significantly. Functionally, the higher penetration rate and increased total efficiency of fertilization were observed in the 20 μM SMN group. In summary, supplementation with SMN in the thawing medium ameliorates the quality of frozen-thawed boar sperm by enhancing mitochondrial respiratory capacity, producing large amounts of ATP and regulating ROS formation.
Collapse
Affiliation(s)
- Shunli Yue
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shunwei Wang
- Reproductive Medicine Center, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Xue Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiaoqi Bian
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Chang Ding
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Tong Wu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Dantong Li
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Jiabo Zhou
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| |
Collapse
|
15
|
Chae YJ, Heo H, Woo CW, Kim ST, Kwon JI, Choi MY, Sung YS, Kim KW, Kim JK, Choi Y, Woo DC. Preclinical Long-term Magnetic Resonance Imaging Study of Silymarin Liver-protective Effects. J Clin Transl Hepatol 2022; 10:1167-1175. [PMID: 36381105 PMCID: PMC9634766 DOI: 10.14218/jcth.2021.00499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/09/2022] [Accepted: 03/17/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Efficacy evaluations with preclinical magnetic resonance imaging (MRI) are uncommon, but MRI in the preclinical phase of drug development provides information that is useful for longitudinal monitoring. The study aim was to monitor the protective effectiveness of silymarin with multiparameter MRI and biomarkers in a thioacetamide (TAA)-induced model of liver injury in rats. Correlation analysis was conducted to assess compare the monitoring of liver function by MRI and biomarkers. METHODS TAA was injected three times a week for 8 weeks to generate a disease model (TAA group). In the TAA and silymarin-treated (TAA-SY) groups, silymarin was administered three times weekly from week 4. MR images were acquired at 0, 2, 4, 6, and 8 weeks in the control, TAA, and TAA-SY groups. RESULTS The area under the curve to maximum time (AUCtmax) and T2* values of the TAA group decreased over the study period, but the serological markers of liver abnormality increased significantly more than those in the control group. In the TAA-SY group, MRI and serological biomarkers indicated attenuation of liver function as in the TAA group. However, pattern changes were observed from week 6 to comparable levels in the control group with silymarin treatment. Negative correlations between either AUCtmax or T2* values and the serological biomarkers were observed. CONCLUSIONS Silymarin had hepatoprotective effects on TAA-induced liver injury and demonstrated the usefulness of multiparametric MRI to evaluate efficacy in preclinical studies of liver drug development.
Collapse
Affiliation(s)
- Yeon Ji Chae
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hwon Heo
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chul-Woong Woo
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Sang-Tae Kim
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Jae-Im Kwon
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Monica Young Choi
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yu Sub Sung
- Clinical Research Center, Asan Medical Center, Seoul, Republic of Korea
| | - Kyung Won Kim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Kon Kim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoonseok Choi
- Medical Research Institute, Gangneung Asan Hospital, Gangneung-si, Gangwon-do, Republic of Korea
- Correspondence to: Dong Cheol Woo, Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea. ORCID: https://orcid.org/0000-0001-8202-015X. Tel: +82-2-3010-4155, Fax: +82-10-5559-7102, E-mail: ; Yoonseok Choi, Medical Research Institute, Gangneung Asan Hospital, University of Ulsan College of Medicine 38, Bangdong-gil, Sacheon-myeon, Gangneung-si, Gangwon-do 25440, Korea. ORCID: https://orcid.org/0000-0002-8478-2999. Tel: +82-33-610-4799, Fax: +82-33-610-3089, E-mail:
| | - Dong-Cheol Woo
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Correspondence to: Dong Cheol Woo, Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea. ORCID: https://orcid.org/0000-0001-8202-015X. Tel: +82-2-3010-4155, Fax: +82-10-5559-7102, E-mail: ; Yoonseok Choi, Medical Research Institute, Gangneung Asan Hospital, University of Ulsan College of Medicine 38, Bangdong-gil, Sacheon-myeon, Gangneung-si, Gangwon-do 25440, Korea. ORCID: https://orcid.org/0000-0002-8478-2999. Tel: +82-33-610-4799, Fax: +82-33-610-3089, E-mail:
| |
Collapse
|
16
|
Mirzaei N, Jahanian Sadatmahalleh S, Rouholamin S, Nasiri M. A randomized trial assessing the efficacy of Silymarin on endometrioma-related manifestations. Sci Rep 2022; 12:17549. [PMID: 36266431 PMCID: PMC9584967 DOI: 10.1038/s41598-022-22073-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 10/10/2022] [Indexed: 01/13/2023] Open
Abstract
To study the effect of silymarin on the Interleukin-6 (IL-6) level, size of endometrioma lesion, pain, sexual function, and Quality of Life (QoL) in women diagnosed with endometriosis. This randomized, double-blind placebo-controlled clinical trial was performed on 70 women with endometriosis which was divided into two groups of intervention and control. The intervention was 140 mg silymarin (or matching placebo) administered twice daily for 12 weeks. The volume of endometrioma lesions, the level of IL-6 concentration in serum, pain, sexual function, and QoL were analyzed before and after the intervention. The means of endometrioma volume (P = 0.04), IL-6 (P = 0.002), and pain (P < 0.001) were reduced significantly in the silymarin group after intervention. However, the QoL and female sexual function did not improve substantially in the two groups (P > 0.05). Silymarin significantly reduced interleukin-6 levels, sizes of endometrioma lesions, and pain-related symptoms. The trial has been registered in the Iranian Registry of Clinical Trials (IRCT20150905023897N5) on 4th February 2020 (04/02/2020) ( https://en.irct.ir/trial/42215 ) and the date of initial participant enrollment was 2nd March 2020 (02/03/2020).
Collapse
Affiliation(s)
- Negin Mirzaei
- grid.412266.50000 0001 1781 3962Department of Reproductive Health and Midwifery, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Al-Ahmad Highway, Nasr Bridge, Tehran, 14115-111 Iran
| | - Shahideh Jahanian Sadatmahalleh
- grid.412266.50000 0001 1781 3962Department of Reproductive Health and Midwifery, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Al-Ahmad Highway, Nasr Bridge, Tehran, 14115-111 Iran
| | - Safoura Rouholamin
- grid.411036.10000 0001 1498 685XDepartment of Obstetrics and Gynecology, Faculty of Medical Sciences, Isfahan University of Medical Sciences, Hezar-Jerib Ave., Isfahan, 81746 73461 Iran
| | - Malihe Nasiri
- grid.411600.2Department of Basic Sciences, Faculty of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Li P, Hu J, Zhao H, Feng J, Chai B. Multi-Omics Reveals Inhibitory Effect of Baicalein on Non-Alcoholic Fatty Liver Disease in Mice. Front Pharmacol 2022; 13:925349. [PMID: 35784718 PMCID: PMC9240231 DOI: 10.3389/fphar.2022.925349] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/06/2022] [Indexed: 12/20/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, whose etiology is poorly understood. Accumulating evidence indicates that gut microbiota plays an important role in the occurrence and progression of various human diseases, including NAFLD. In this study, NAFLD mouse models were established by feeding a high-fat diet (HFD). Baicalein, a natural flavonoid with multiple biological activities, was administered by gavage, and its protective effect on NAFLD was analyzed by histopathological and blood factor analysis. Gut microbiota analysis demonstrated that baicalein could remodel the overall structure of the gut microbiota from NAFLD model mice, especially Anaerotruncus, Lachnoclostridium, and Mucispirillum. Transcriptomic analysis showed baicalein restored the expressions of numerous genes that were upregulated in hepatocytes of NAFLD mice, such as Apoa4, Pla2g12a, Elovl7, Slc27a4, Hilpda, Fabp4, Vldlr, Gpld1, and Apom. Metabolomics analysis proved that baicalein mainly regulated the processes associated with lipid metabolism, such as alpha-Linolenic acid, 2-Oxocarboxylic acid, Pantothenate and CoA biosynthesis, and bile secretion. Multi-omics analysis revealed that numerous genes regulated by baicalein were significantly correlated with pathways related to lipid metabolism and biosynthesis and secrection of bile acid, and baicalein might affect lipid metabolism in liver via regulating the ecological structure of gut microbiota in NAFLD mice. Our results elucidated the correlated network among diet, gut microbiota, metabolomic, and transcriptional profiling in the liver. This knowledge may help explore novel therapeutic approaches against NAFLD.
Collapse
Affiliation(s)
- Ping Li
- Institute of Loess Plateau, Shanxi University, Taiyuan, China
- Department of Biological Science and Technology, Jinzhong University, Jinzhong, China
| | - Jianran Hu
- Department of Biological Science and Technology, Jinzhong University, Jinzhong, China
| | - Hongmei Zhao
- Department of Biological Science and Technology, Jinzhong University, Jinzhong, China
| | - Jing Feng
- Department of Gastroenterology, Shanxi Provincial People’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Baofeng Chai
- Institute of Loess Plateau, Shanxi University, Taiyuan, China
- *Correspondence: Baofeng Chai,
| |
Collapse
|
18
|
Local Insulin-Derived Amyloidosis Model Confronted with Silymarin: Histological Insights and Gene Expression of MMP, TNF-α, and IL-6. Int J Mol Sci 2022; 23:ijms23094952. [PMID: 35563343 PMCID: PMC9101448 DOI: 10.3390/ijms23094952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
Amyloidosis is a heterogeneous group of protein deposition diseases associated with the presence of amyloid fibrils in tissues. Analogs of insulin that are used for treating diabetic patients (including regular insulin) can form amyloid fibrils, both in vitro and in vivo as reported in patients. The main purpose of this study was the induction of localized insulin-generated amyloidosis and the observation of silymarin effects on this process. In order to obtain amyloid structures, regular insulin was incubated at 37 °C for 24 h. Congo red absorbance and transmission electron microscopy images validated the formation of amyloid fibrils. Those fibrils were then injected subcutaneously into rats once per day for 6, 12 or 18 consecutive days in the presence or absence of silymarin, and caused development of firm waxy masses. These masses were excised and stained with Hematoxylin and Eosin, Congo red and Thioflavin S. Histological examination showed adipose cells and connective tissue in which amyloid deposition was visible. Amyloids decreased in the presence of silymarin, and the same effect was observed when silymarin was added to normal insulin and injected subsequently. Furthermore, plasma concentrations of MMP2, TNF-α, and IL-6 inflammatory factors were measured, and their gene expression was locally assessed in the masses by immunohistochemistry. All three factors increased in the amyloidosis state, while silymarin had an attenuating effect on their plasma levels and gene expression. In conclusion, we believe that silymarin could be effective in counteracting insulin-generated local amyloidosis.
Collapse
|
19
|
Borymska W, Zych M, Dudek S, Kaczmarczyk-Sedlak I. Silymarin from Milk Thistle Fruits Counteracts Selected Pathological Changes in the Lenses of Type 1 Diabetic Rats. Nutrients 2022; 14:1450. [PMID: 35406062 PMCID: PMC9003010 DOI: 10.3390/nu14071450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/12/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023] Open
Abstract
Diabetes is a metabolic disease affecting many tissues and organs. The main etiological factor for diabetic complications is hyperglycemia and subsequent pathologies, such as oxidative stress. One of the organs susceptible to the development of diabetic complications is the eye with all of its elements, including the lens. The aim of this study was to evaluate the effect of silymarin, an extract obtained from milk thistle fruit husks, on the oxidative stress markers in the lenses of type 1 diabetic rats. The study was performed on male rats in which type 1 diabetes was induced with 60 mg/kg streptozotocin injection. Diabetic animals were treated via an intragastric tube with silymarin at 50 and 100 mg/kg doses for four weeks. Multiple oxidative stress and polyol pathway-related parameters were measured in the lenses, and auxiliary biochemical tests in the serum were conducted. Diabetes induced severe pathological changes both in the lenses and the serum, and silymarin counteracted several of them. Nevertheless, the qualitative analyses encompassing all tested parameters indicate that silymarin slightly improved the overall state of diabetic animals. Upon the obtained results, it can be concluded that silymarin reveals a faint positive effect on the lenses in type 1 diabetic rats.
Collapse
Affiliation(s)
- Weronika Borymska
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (M.Z.); (S.D.); (I.K.-S.)
| | | | | | | |
Collapse
|
20
|
El-Fadaly AA, Afifi NA, El-Eraky W, Salama A, Abdelhameed MF, El-Rahman SSA, Ramadan A. Fisetin alleviates thioacetamide-induced hepatic fibrosis in rats by inhibiting Wnt/β-catenin signaling pathway. Immunopharmacol Immunotoxicol 2022; 44:355-366. [PMID: 35255766 DOI: 10.1080/08923973.2022.2047198] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Liver fibrosis is a chronic wound-healing response to liver injury of various origins and represents a major health problem. OBJECTIVE The current study endeavored to investigate the repressing effect of fisetin on hepatic fibrosis induced by thioacetamide (TAA) in rats. MATERIALS AND METHODS Rats were injected with TAA (200 mg/kg) intraperitoneally twice per week for 6 weeks to induce liver fibrosis. Fisetin (50 and 100 mg/kg/day) or silymarin (50 mg/kg/day) were given orally on a daily basis along with TAA. Liver function parameters, oxidative stress, inflammatory and fibrogenic biomarkers as well as wnt3a, β-catenin, glycogen synthase kinase 3 (GSK-3β) and cyclin D1 were estimated. Histoapthological and immunohistochemical examinations were performed. RESULTS Fisetin restored normal liver functions, increased reduced glutathione (GSH) level and decreased malondialdehyde (MDA), as well as inflammatory biomarkers including; tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6). Additionally, it lessened transforming growth factor β1 (TGF-β1), collagen I and tissue inhibitor of metalloproteinase-1 (TIMP-1) levels as well as elevated matrix metalloproteinase-9 (MMP-9) hepatic content. Furthermore, fisetin significantly suppressed wnt3a gene expression associated with decreased β-catenin and increased GSK-3β levels. Moreover, fisetin decreased the progress of histologic hepatic fibroplasia and diminished hepatic expression of α-SMA and cyclin D1. CONCLUSION Fisetin curbed liver fibrosis and exhibited superior activity over silymarin through inhibition of hepatic stellate cells (HSCs) activation and proliferation via suppressing the Wnt/β-catenin pathway, modulating MMP-9 and TIMP-1, and inhibiting multiple profibrogenic factors, besides its antioxidant and anti-inflammatory effects. Therefore, fisetin is a promising therapeutic candidate for hepatic fibrosis.
Collapse
Affiliation(s)
| | - Nehal A Afifi
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Wafaa El-Eraky
- Department of Pharmacology, National Research Centre, Cairo, Egypt
| | - Abeer Salama
- Department of Pharmacology, National Research Centre, Cairo, Egypt
| | | | - Sahar S Abd El-Rahman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - A Ramadan
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
21
|
Koltai T, Fliegel L. Role of Silymarin in Cancer Treatment: Facts, Hypotheses, and Questions. J Evid Based Integr Med 2022; 27:2515690X211068826. [PMID: 35018864 PMCID: PMC8814827 DOI: 10.1177/2515690x211068826] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/20/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
The flavonoid silymarin extracted from the seeds of Sylibum marianum is a mixture of 6 flavolignan isomers. The 3 more important isomers are silybin (or silibinin), silydianin, and silychristin. Silybin is functionally the most active of these compounds. This group of flavonoids has been extensively studied and they have been used as hepato-protective substances for the mushroom Amanita phalloides intoxication and mainly chronic liver diseases such as alcoholic cirrhosis and nonalcoholic fatty liver. Hepatitis C progression is not, or slightly, modified by silymarin. Recently, it has also been proposed for SARS COVID-19 infection therapy. The biochemical and molecular mechanisms of action of these substances in cancer are subjects of ongoing research. Paradoxically, many of its identified actions such as antioxidant, promoter of ribosomal synthesis, and mitochondrial membrane stabilization, may seem protumoral at first sight, however, silymarin compounds have clear anticancer effects. Some of them are: decreasing migration through multiple targeting, decreasing hypoxia inducible factor-1α expression, inducing apoptosis in some malignant cells, and inhibiting promitotic signaling among others. Interestingly, the antitumoral activity of silymarin compounds is limited to malignant cells while the nonmalignant cells seem not to be affected. Furthermore, there is a long history of silymarin use in human diseases without toxicity after prolonged administration. The ample distribution and easy accessibility to milk thistle-the source of silymarin compounds, its over the counter availability, the fact that it is a weed, some controversial issues regarding bioavailability, and being a nutraceutical rather than a drug, has somehow led medical professionals to view its anticancer effects with skepticism. This is a fundamental reason why it never achieved bedside status in cancer treatment. However, in spite of all the antitumoral effects, silymarin actually has dual effects and in some cases such as pancreatic cancer it can promote stemness. This review deals with recent investigations to elucidate the molecular actions of this flavonoid in cancer, and to consider the possibility of repurposing it. Particular attention is dedicated to silymarin's dual role in cancer and to some controversies of its real effectiveness.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
22
|
Hamad Shareef S, Abdel Aziz Ibrahim I, Alzahrani AR, Al-Medhtiy MH, Ameen Abdulla M. Hepatoprotective effects of methanolic extract of green tea against Thioacetamide-Induced liver injury in Sprague Dawley rats. Saudi J Biol Sci 2022; 29:564-573. [PMID: 35002452 PMCID: PMC8716963 DOI: 10.1016/j.sjbs.2021.09.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/03/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Since ancient times, herbal medicines have been applied in the treatment of cancer. Tea, derivative from the dried leaves of Camellia sinensis (L.) Kuntze plant is the most popular beverage globally after water and is available in various forms. Green tea has been expansively investigated for its beneficial properties of cancer prevention and therapy. The goal of the research: The current study was conducted to evaluate the hepaprotective character of methanolic green tea extract and its mechanism of action contrary to thioacetamide (TAA)-produced liver fibrosis of Sprague Dawley rats. MATERIALS AND METHODS Thirty rodents were equally placed in 5 clusters including normal control, TAA group as a positive control, silymarin as standard drug control, and treatment groups consisting of high dose and a low dose Camellia sinensis. Rats in experimental clusters by mouth fed with C. sinensis at 250 mg/kg or 500 mg/kg daily for 2 months. After 60 days, all rats were sacrificed. Blood specimens were gathered for liver biochemical examination. Livers of all groups were dissected out and subjected to histopathological examination through the Hematoxylin and Eosin stain, Masson trichrome, and immunohistochemistry stains (PCNA). Liver tissue homogenate was also analyzed for antioxidant activity parameters. RESULTS Gross morphological examination showed a regular liver architecture in C. sinensis fed collections compared to the TAA sets. Histology of rat's liver fed with C. sinensis showed an important decrease in the liver index with hepatic cells propagation, mild cellular injury, and immunostaining showed significant down-expression of proliferating cell nuclear antigen (PCNA). TAA produced liver fibrosis through a significant increase in serum alanine transferase, aspartate aminotransferase, alkaline phosphatase, and bilirubin. Total protein and albumin also decreased in the TAA group. Moreover, the reduction of antioxidant enzyme activity including superoxide dismutase and catalase as well as the increase in malondialdehyde was detected in the TAA control group. Meanwhile, an abnormal level of liver biochemical parameters was restored closer to the normal levels in serum of the C. sinensis-fed clusters. In addition, C. sinensis fed assemblies showed elevated antioxidative enzymes activity with a reduction in malondialdehyde level comparable to the levels in silymarin-treated rats. CONCLUSIONS Green tea potentially inhibited the progression of liver cirrhosis, down -regulation of PCNA proliferation, prevented oxidation of hepatocytes, recovered SOD and CAT enzymes, condensed MDA and reduced cellular inflammation.
Collapse
Affiliation(s)
- Suhayla Hamad Shareef
- Department of Medical Microbiology, College of Science, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
- Department of Biology, College of Education, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdullah R. Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Morteta H. Al-Medhtiy
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, University of Kufa, Iraq
| | - Mahmood Ameen Abdulla
- Department of Medical Microbiology, College of Science, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
23
|
de Souza Basso B, Haute GV, Ortega-Ribera M, Luft C, Antunes GL, Bastos MS, Carlessi LP, Levorse VG, Cassel E, Donadio MVF, Santarém ER, Gracia-Sancho J, Rodrigues de Oliveira J. Methoxyeugenol deactivates hepatic stellate cells and attenuates liver fibrosis and inflammation through a PPAR-ɣ and NF-kB mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114433. [PMID: 34280502 DOI: 10.1016/j.jep.2021.114433] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/02/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Studies have shown interest in nutraceuticals for the prevention of liver diseases. Methoxyeugenol, is a molecule found in foods, such as nutmeg (Myristica fragrans Houtt.) and Brazilian red propolis. These two sources of methoxyeugenol, propolis and nutmeg, are used in folk medicine for the treatment of hepatic and gastrointestinal disorders, although little is known about their effects on the prevention of liver fibrosis. Natural PPAR (Peroxisome proliferator-activated receptor) agonists would represent unique molecules for therapy, considering the lack of therapeutics to treat liver fibrosis in chronic liver disease. Thus, investigation on new alternatives are necessary, including the search for natural compounds from renewable and sustainable sources. Liver fibrosis is a pathological process characterized by an exacerbated cicatricial response in the hepatic tissue, which compromises liver function. Therefore, inhibition of HSC (hepatic stellate cell) activation and hepatocyte damage are considered major strategies for the development of new anti-fibrotic treatments. AIM OF THE STUDY This study aimed to investigate the effects of methoxyeugenol treatment on HSC phenotype modulation in human and murine cells, hepatocyte damage prevention, and protective effects in vivo, in order to evaluate its therapeutic potential for liver fibrosis prevention. METHODS We investigated the effects of methoxyeugenol in (i) in vitro models using human and murine HSC and hepatocytes, and (ii) in vivo models of CCl4 (carbon tetrachloride) -induced liver fibrosis in mice. RESULTS We herein report that methoxyeugenol decreases HSC activation through the activation of PPAR-ɣ, ultimately inducing a quiescent phenotype highlighted by an increase in lipid droplets, loss of contraction ability, and a decrease in the proliferative rate and mRNA expression of fibroblast markers. In addition, methoxyeugenol prevented hepatocytes from oxidative stress damage. Moreover, in mice submitted to chronic liver disease through CCl4 administration, methoxyeugenol decreased the inflammatory profile, liver fibrosis, mRNA expression of fibrotic genes, and the inflammatory pathway signaled by NF-kB (Nuclear factor kappa B). CONCLUSION We propose methoxyeugenol as a novel and potential therapeutic approach to treat chronic liver disease and fibrosis.
Collapse
Affiliation(s)
- Bruno de Souza Basso
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Gabriela Viegas Haute
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Martí Ortega-Ribera
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Lab, IDIBAPS Biomedical Research Institute - CIBEREHD, Barcelona, Spain
| | - Carolina Luft
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Géssica Luana Antunes
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Matheus Scherer Bastos
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Leonardo Pfeiff Carlessi
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vitor Giancarlo Levorse
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo Cassel
- School of Technology, Pontifical Catholic University of Rio Grande do Sul - PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcio Vinícius Fagundes Donadio
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eliane Romanato Santarém
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Lab, IDIBAPS Biomedical Research Institute - CIBEREHD, Barcelona, Spain
| | - Jarbas Rodrigues de Oliveira
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
24
|
Abstract
Significance: As the central metabolic organ, the liver is exposed to a variety of potentially cytotoxic, proinflammatory, profibrotic, and carcinogenic stimuli. To protect the organism from these deleterious effects, the liver has evolved a number of defense systems, which include antioxidant substrates and enzymes, anti-inflammatory tools, enzymatic biotransformation systems, and metabolic pathways. Recent Advances: One of the pivotal systems that evolved during phylogenesis was the heme catabolic pathway. Comprising the important enzymes heme oxygenase and biliverdin reductase, this complex pathway has a number of key functions including enzymatic activities, but also cell signaling, and DNA transcription. It further generates two important bile pigments, biliverdin and bilirubin, as well as the gaseous molecule carbon monoxide. These heme degradation products have potent antioxidant, immunosuppressive, and cytoprotective effects. Recent data suggest that the pathway participates in the regulation of metabolic and hormonal processes implicated in the pathogenesis of hepatic and other diseases. Critical Issues: This review discusses the impact of the heme catabolic pathway on major liver diseases, with particular focus on the involvement of cellular targeting and signaling in the pathogenesis of these conditions. Future Directions: To utilize the biological consequences of the heme catabolic pathway, several unique therapeutic strategies have been developed. Research indicates that pharmaceutical, nutraceutical, and lifestyle modifications positively affect the pathway, delivering potentially long-term clinical benefits. However, further well-designed studies are needed to confirm the clinical benefits of these approaches. Antioxid. Redox Signal. 35, 734-752.
Collapse
Affiliation(s)
- Libor Vítek
- Fourth Department of Internal Medicine, and Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
25
|
Yan J, Nie Y, Luo M, Chen Z, He B. Natural Compounds: A Potential Treatment for Alcoholic Liver Disease? Front Pharmacol 2021; 12:694475. [PMID: 34290612 PMCID: PMC8287649 DOI: 10.3389/fphar.2021.694475] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Excessive alcohol intake is a direct cause of alcoholic liver disease (ALD). ALD usually manifests as fatty liver in the initial stage and then develops into alcoholic hepatitis (ASH), fibrosis and cirrhosis. Severe alcoholism induces extensive hepatocyte death, liver failure, and even hepatocellular carcinoma (HCC). Currently, there are few effective clinical means to treat ALD, except for abstinence. Natural compounds are a class of compounds extracted from herbs with an explicit chemical structure. Several natural compounds, such as silymarin, quercetin, hesperidin, and berberine, have been shown to have curative effects on ALD without side effects. In this review, we pay particular attention to natural compounds and developing clinical drugs based on natural compounds for ALD, with the aim of providing a potential treatment for ALD.
Collapse
Affiliation(s)
- Junbin Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yunmeng Nie
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Minmin Luo
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiyun Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Beihui He
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Key Laboratory of Integrative Chinese and Western Medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
26
|
Gad D, El-Shora H, Fraternale D, Maricchiolo E, Pompa A, Dietz KJ. Bioconversion of Callus-Produced Precursors to Silymarin Derivatives in Silybum marianum Leaves for the Production of Bioactive Compounds. Int J Mol Sci 2021; 22:2149. [PMID: 33670070 PMCID: PMC7926748 DOI: 10.3390/ijms22042149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/02/2022] Open
Abstract
The present study aimed to investigate the enzymatic potential of Silybum marianum leaves to bioconvert phenolic acids produced in S. marianum callus into silymarin derivatives as chemopreventive agent. Here we demonstrate that despite the fact that leaves of S. marianum did not accumulate silymarin themselves, expanding leaves had the full capacity to convert di-caffeoylquinic acid to silymarin complex. This was proven by HPLC separations coupled with electrospray ionization mass spectrometry (ESI-MS) analysis. Soaking the leaf discs with S. marianum callus extract for different times revealed that silymarin derivatives had been formed at high yield after 16 h. Bioconverted products displayed the same retention time and the same mass spectra (MS or MS/MS) as standard silymarin. Bioconversion was achieved only when using leaves of a specific age, as both very young and old leaves failed to produce silymarin from callus extract. Only medium leaves had the metabolic capacity to convert callus components into silymarin. The results revealed higher activities of enzymes of the phenylpropanoid pathway in medium leaves than in young and old leaves. It is concluded that cotyledon-derived callus efficiently produces compounds that can be bio-converted to flavonolignans in leaves tissue of S. marianum.
Collapse
Affiliation(s)
- Dina Gad
- Botany and Microbiology Department, Faculty of Science, Menoufia University, Shebin EL-Koum 32511, Egypt
- Biochemistry and Physiology of Plants, Faculty of Biology W5, Bielefeld University, 33501 Bielefeld, Germany;
| | - Hamed El-Shora
- Botany Department, Faculty of Science, Mansoura University, Mansoura 35511, Egypt;
| | - Daniele Fraternale
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo” Via Donato Bramante, 28, 61029 Urbino, Italy; (D.F.); (E.M.)
| | - Elisa Maricchiolo
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo” Via Donato Bramante, 28, 61029 Urbino, Italy; (D.F.); (E.M.)
| | - Andrea Pompa
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo” Via Donato Bramante, 28, 61029 Urbino, Italy; (D.F.); (E.M.)
| | - Karl-Josef Dietz
- Biochemistry and Physiology of Plants, Faculty of Biology W5, Bielefeld University, 33501 Bielefeld, Germany;
| |
Collapse
|