1
|
Bahari H, Shahraki Jazinaki M, Rashidmayvan M, Taheri S, Amini MR, Malekahmadi M. The effects of silymarin consumption on inflammation and oxidative stress in adults: a systematic review and meta-analysis. Inflammopharmacology 2024; 32:949-963. [PMID: 38372848 DOI: 10.1007/s10787-023-01423-6] [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: 09/23/2023] [Accepted: 12/24/2023] [Indexed: 02/20/2024]
Abstract
BACKGROUND Owing to the rich phytochemical content of Silymarin, it may effectively manage inflammation and oxidative stress. We, therefore, aimed to examine the existing evidence on the effect of Silymarin consumption on inflammation and oxidative stress factors by conducting a systematic review and meta-analysis of randomized controlled trials. METHODS A systematic literature search up to September 2023 was completed in PubMed/Medline, Scopus, and Web of Science, to identify eligible RCTs. Heterogeneity tests of the selected trials were performed using the I2 statistic. Random effects models were assessed based on the heterogeneity tests, and pooled data were determined as weighted mean differences with a 95% confidence interval. RESULTS Fifteen RCTs were included in this meta-analysis. Our findings showed that Silymarin consumption significantly decreased CRP (WMD, - 0.50 mg/L; 95% CI, (- 0.95 to - 0.04); p = 0.03), MDA (WMD, - 1.19 nmol/mL; 95% CI, (- 1.99 to - 0.38); p = 0.004), and IL-6 (WMD, - 0.44 pg/ml; 95% CI, (- 0.75 to - 0.12); p = 0.006). Silymarin consumption had no significant effects on IL-10, TAC, and GSH. A significant non-linear relationship was observed between the duration of the intervention and MDA changes. CONCLUSIONS Silymarin can help reduce inflammation in patients with diabetes and thalassemia by reducing MDA as an oxidative stress marker and CRP and IL-6 as inflammatory markers.
Collapse
Affiliation(s)
- Hossein Bahari
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mostafa Shahraki Jazinaki
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Rashidmayvan
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research Center, Gonabad University of Medical Science, Gonabad, Iran
| | - Shaghayegh Taheri
- Department of Clinical Biochemistry, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Reza Amini
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Malekahmadi
- Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Gupta J, Jalil AT, Riyad Muedii ZAH, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Farhood B. The Radiosensitizing Potentials of Silymarin/Silibinin in Cancer: A Systematic Review. Curr Med Chem 2024; 31:6992-7014. [PMID: 37921180 DOI: 10.2174/0109298673248404231006052436] [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: 02/09/2023] [Revised: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Although radiotherapy is one of the main cancer treatment modalities, exposing healthy organs/tissues to ionizing radiation during treatment and tumor resistance to ionizing radiation are the chief challenges of radiotherapy that can lead to different adverse effects. It was shown that the combined treatment of radiotherapy and natural bioactive compounds (such as silymarin/silibinin) can alleviate the ionizing radiation-induced adverse side effects and induce synergies between these therapeutic modalities. In the present review, the potential radiosensitization effects of silymarin/silibinin during cancer radiation exposure/radiotherapy were studied. METHODS According to the PRISMA guideline, a systematic search was performed for the identification of relevant studies in different electronic databases of Google Scholar, PubMed, Web of Science, and Scopus up to October 2022. We screened 843 articles in accordance with a predefined set of inclusion and exclusion criteria. Seven studies were finally included in this systematic review. RESULTS Compared to the control group, the cell survival/proliferation of cancer cells treated with ionizing radiation was considerably less, and silymarin/silibinin administration synergistically increased ionizing radiation-induced cytotoxicity. Furthermore, there was a decrease in the tumor volume, weight, and growth of ionizing radiation-treated mice as compared to the untreated groups, and these diminutions were predominant in those treated with radiotherapy plus silymarin/ silibinin. Furthermore, the irradiation led to a set of biochemical and histopathological changes in tumoral cells/tissues, and the ionizing radiation-induced alterations were synergized following silymarin/silibinin administration (in most cases). CONCLUSION In most cases, silymarin/silibinin administration could sensitize the cancer cells to ionizing radiation through an increase of free radical formation, induction of DNA damage, increase of apoptosis, inhibition of angiogenesis and metastasis, etc. However, suggesting the use of silymarin/silibinin during radiotherapeutic treatment of cancer patients requires further clinical studies.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, U.P., India
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Andrés Alexis Ramírez-Coronel
- Psychometry and Ethology Laboratory, Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellin, Colombia
- Educational Statistics Research Group (GIEE), National University of Education, Cuenca, Ecuador
| | | | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
3
|
Singh M, Kadhim MM, Turki Jalil A, Oudah SK, Aminov Z, Alsaikhan F, Jawhar ZH, Ramírez-Coronel AA, Farhood B. A systematic review of the protective effects of silymarin/silibinin against doxorubicin-induced cardiotoxicity. Cancer Cell Int 2023; 23:88. [PMID: 37165384 PMCID: PMC10173635 DOI: 10.1186/s12935-023-02936-4] [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: 01/17/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023] Open
Abstract
PURPOSE Although doxorubicin chemotherapy is commonly applied for treating different malignant tumors, cardiotoxicity induced by this chemotherapeutic agent restricts its clinical use. The use of silymarin/silibinin may mitigate the doxorubicin-induced cardiac adverse effects. For this aim, the potential cardioprotective effects of silymarin/silibinin against the doxorubicin-induced cardiotoxicity were systematically reviewed. METHODS In this study, we performed a systematic search in accordance with PRISMA guideline for identifying all relevant studies on "the role of silymarin/silibinin against doxorubicin-induced cardiotoxicity" in different electronic databases up to June 2022. Sixty-one articles were obtained and screened based on the predefined inclusion and exclusion criteria. Thirteen eligible papers were finally included in this review. RESULTS According to the echocardiographic and electrocardiographic findings, the doxorubicin-treated groups presented a significant reduction in ejection fraction, tissue Doppler peak mitral annulus systolic velocity, and fractional shortening as well as bradycardia, prolongation of QT and QRS interval. However, these echocardiographic abnormalities were obviously improved in the silymarin plus doxorubicin groups. As well, the doxorubicin administration led to induce histopathological and biochemical changes in the cardiac cells/tissue; in contrast, the silymarin/silibinin co-administration could mitigate these induced alterations (for most of the cases). CONCLUSION According to the findings, it was found that the co-administration of silymarin/silibinin alleviates the doxorubicin-induced cardiac adverse effects. Silymarin/silibinin exerts its cardioprotective effects via antioxidant, anti-inflammatory, anti-apoptotic activities, and other mechanisms.
Collapse
Affiliation(s)
- Mandeep Singh
- Department of Physical Education, University of Jammu, Srinagar, Jammu, India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq
- Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellín, Colombia
- Educational Statistics Research Group (GIEE), National University of Education, Cuenca, Ecuador
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Bello M. Structural basis of Nrf2 activation by flavonolignans from silymarin. J Mol Graph Model 2023; 119:108393. [PMID: 36525840 DOI: 10.1016/j.jmgm.2022.108393] [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: 08/10/2022] [Revised: 11/08/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Several properties of silymarin (SM) extract have been attributed to their three major flavonolignans (silybin, silychristin, and silydianin) and their 2,3-dehydro derivatives (2,3-dehydrosilybin, 2,3-dehydrosilychristin, and 2,3-dehydrosilydianin). Experimental findings have suggested that the antioxidative and protective activities of these compounds could be due to their ability to activate nuclear factor erythroid 2-related factor 2 (Nrf2). The mechanism by which SM compounds exert their effect has been suggested to be by disrupting the complex between Nrf2 and Kelch-like ECH-associated protein 1 (Keap1). However, information about the structural and energetic basis of the inhibitory mechanism of SM compounds on the Nrf2-Keap1 pathway is lacking. We evaluated the binding properties of SM compounds because experimental findings have pointed to them as potential activators of Nrf2. Our study combined docking and molecular dynamics (MD) simulations with the Poisson-Boltzmann and generalized Born and surface area (MMPBSA and MMGBSA) methods and quantum mechanics-molecular mechanics (QMMM) calculations to investigate Keap1-ligand interactions. Our results predicted that silybinA and 2,3-dehydrosilybin bind to Keap1, forming interactions with the same pockets as those observed for the cocrystallized Keap1-Cpd16 complex but with more favorable binding free energies. These findings indicate that both natural compounds are potential activators of Nrf2.
Collapse
Affiliation(s)
- Martiniano Bello
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón, s/n, Col. Casco de Santo Tomas, Ciudad de México, 11340, Mexico.
| |
Collapse
|
5
|
DuBreuil DM, Lai X, Zhu K, Chahyadinata G, Perner C, Chiang BM, Battenberg A, Sokol CL, Wainger BJ. Phenotypic screen identifies the natural product silymarin as a novel anti-inflammatory analgesic. Mol Pain 2023; 19:17448069221148351. [PMID: 36526437 PMCID: PMC9893088 DOI: 10.1177/17448069221148351] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/01/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Sensory neuron hyperexcitability is a critical driver of pathological pain and can result from axon damage, inflammation, or neuronal stress. G-protein coupled receptor signaling can induce pain amplification by modulating the activation of Trp-family ionotropic receptors and voltage-gated ion channels. Here, we sought to use calcium imaging to identify novel inhibitors of the intracellular pathways that mediate sensory neuron sensitization and lead to hyperexcitability. We identified a novel stimulus cocktail, consisting of the SSTR2 agonist L-054,264 and the S1PR3 agonist CYM5541, that elicits calcium responses in mouse primary sensory neurons in vitro as well as pain and thermal hypersensitivity in mice in vivo. We screened a library of 906 bioactive compounds and identified 24 hits that reduced calcium flux elicited by L-054,264/CYM5541. Among these hits, silymarin, a natural product derived from milk thistle, strongly reduced activation by the stimulation cocktail, as well as by a distinct inflammatory cocktail containing bradykinin and prostaglandin E2. Silymarin had no effect on sensory neuron excitability at baseline, but reduced calcium flux via Orai channels and downstream mediators of phospholipase C signaling. In vivo, silymarin pretreatment blocked development of adjuvant-mediated thermal hypersensitivity, indicating potential use as an anti-inflammatory analgesic.
Collapse
Affiliation(s)
- Daniel M DuBreuil
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Genomic Medicine Unit, Sanofi, Waltham, MA, USA
| | - Xiaofan Lai
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kevin Zhu
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Gracesenia Chahyadinata
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Caroline Perner
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Universitätsmedizin Greifswald, Germany
| | - Brenda M Chiang
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Ashley Battenberg
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Caroline L Sokol
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Brian J Wainger
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston MA, USA
| |
Collapse
|
6
|
Garcia-Manieri JAA, Correa VG, Backes E, de Sá-Nakanishi AB, Bracht L, Comar JF, Corrêa RCG, Peralta RM, Bracht A. A Critical Appraisal of the Most Recent Investigations on the Hepatoprotective Action of Brazilian Plants. PLANTS (BASEL, SWITZERLAND) 2022; 11:3481. [PMID: 36559593 PMCID: PMC9785989 DOI: 10.3390/plants11243481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Conventional treatments for liver diseases are often burdened by side effects caused by chemicals. For minimizing this problem, the search for medicines based on natural products has increased. The objective of this review was to collect data on the potential hepatoprotective activity of plants of the Brazilian native flora. Special attention was given to the modes of extraction, activity indicators, and identification of the active compounds. The databases were Science direct, Pubmed, and Google Academic. Inclusion criteria were: (a) plants native to Brazil; (b) studies carried out during the last 15 years; (c) high-quality research. A fair number of communications met these criteria. Various parts of plants can be used, e.g., fruit peels, seeds, stem barks, and leaves. An outstanding characteristic of the active extracts is that they were mostly obtained from plant parts with low commercial potential, i.e., by-products or bio-residues. The hepatoprotective activities are exerted by constituents such as flavonoids, phenolic acids, vitamin C, phytosterols, and fructose poly- and oligosaccharides. Several Brazilian plants present excellent perspectives for the obtainment of hepatoprotective formulations. Very important is the economical perspective for the rural producers which may eventually increase their revenue by selling increasingly valued raw materials which otherwise would be wasted.
Collapse
Affiliation(s)
| | - Vanesa Gesser Correa
- Departamento de Bioquímica, Universidade Estadual de Maringá, Maringá 87020-900, Brazil
| | - Emanueli Backes
- Departamento de Bioquímica, Universidade Estadual de Maringá, Maringá 87020-900, Brazil
| | | | - Lívia Bracht
- Departamento de Bioquímica, Universidade Estadual de Maringá, Maringá 87020-900, Brazil
| | | | - Rúbia Carvalho Gomes Corrêa
- Programa de Pós-Graduação em Tecnologias Limpas, Instituto Cesumar de Ciência, Tecnologia e Inovação—ICETI, Universidade Cesumar—UNICESUMAR, Maringá 87050-900, Brazil
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Rosane Marina Peralta
- Departamento de Bioquímica, Universidade Estadual de Maringá, Maringá 87020-900, Brazil
| | - Adelar Bracht
- Departamento de Bioquímica, Universidade Estadual de Maringá, Maringá 87020-900, Brazil
| |
Collapse
|
7
|
El-Garhy O, Soudy FA, Alharbi YM, Alshanbari FA, Almujaydil MS, Alhomaid RM, Ahmed-Farid OA, Mohamed SA, El-Garhy HAS, Barakat H, EL Nagar AG. Dietary Supplementation of Silybum marianum Seeds Improved Growth Performance and Upregulated Associated Gene Expression of Muscovy Ducklings ( Cairina moschata). Antioxidants (Basel) 2022; 11:2300. [PMID: 36421486 PMCID: PMC9687607 DOI: 10.3390/antiox11112300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 04/01/2024] Open
Abstract
The effect of feeding on diets supplemented with Silybum marianum L. dry seeds (SMS) on growth performance, mortality percentage, biochemical parameters, the expression profile of related genes, and genotoxic effect in Muscovy ducklings was evaluated during a brooding period of 4 weeks. Two hundred and forty one-day-old Muscovy ducks were randomly assigned to four treatment groups (60 ducklings/group), the first group fed on basal diet with no additives (control), and the second (4 g kg-1), third (8 g kg-1), and fourth (12 g kg-1) groups fed the basal diet supplemented with 0, 4, 8, and 12 g kg-1 diet SMS, respectively. A substantial improvement in live body weight (LBW), body weight gain (BWG), and growth rate (GR), and a decrease in feed conversion ratios (FCR) and mortality rate were shown in ducks fed a diet supplemented with either 8 g kg-1 or 12 g kg-1 SMS compared to the other groups. Relevant improvements in liver function, oxidative stress markers, purinergic cell energy, and brain appetite were recorded on ducklings fed diets supplemented with SMS. Moreover, diets which included 8 or 12 g kg-1 SMS positively upregulated the expression of growth hormone gene (GH) and antioxidant genes (SOD1, SOD2, and CAT). These results are consistent with the increase in liver activity SOD and CAT enzymes, resulting in less DNA fragmentation. Consequently, all the aforementioned improvements in biochemical parameters and gene expression profiling may explain the superiority of the treated ducklings compared with the control group. Conclusively, the SMS could be used as a natural feed additive to promote health status and improve the growth performance of small grower ducks during the brooding period.
Collapse
Affiliation(s)
- Osama El-Garhy
- Department of Animal Production, Faculty of Agriculture at Moshtohor, Benha University, Moshtohor 13736, Egypt
| | - Fathia A. Soudy
- Genetics and Genetic Engineering Department, Faculty of Agriculture, Benha University, Moshtohor 13736, Egypt
| | - Yousef M. Alharbi
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Fahad A. Alshanbari
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Mona S. Almujaydil
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Raghad M. Alhomaid
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Omar A. Ahmed-Farid
- Physiology Department, National Organization for Drug Control and Research (NODCAR), Giza 12553, Egypt
| | - Shereen A. Mohamed
- Genetics and Genetic Engineering Department, Faculty of Agriculture, Benha University, Moshtohor 13736, Egypt
| | - Hoda A. S. El-Garhy
- Genetics and Genetic Engineering Department, Faculty of Agriculture, Benha University, Moshtohor 13736, Egypt
| | - Hassan Barakat
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
- Food Technology Department, Faculty of Agriculture, Benha University, Moshtohor 13736, Egypt
| | - Ayman G. EL Nagar
- Department of Animal Production, Faculty of Agriculture at Moshtohor, Benha University, Moshtohor 13736, Egypt
| |
Collapse
|
8
|
Xu S, Jiang X, Jia X, Jiang X, Che L, Lin Y, Zhuo Y, Feng B, Fang Z, Li J, Wang J, Ren Z, Wu D. Silymarin Modulates Microbiota in the Gut to Improve the Health of Sow from Late Gestation to Lactation. Animals (Basel) 2022; 12:ani12172202. [PMID: 36077922 PMCID: PMC9454421 DOI: 10.3390/ani12172202] [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: 08/03/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
Inflammatory responses reduce milk production in lactating sows. Silymarin may modulate inflammatory reactions. Here, we aimed to verify whether dietary silymarin supplementation could alleviate inflammatory responses in lactating sows through microbiota change in the gut. We also investigated how silymarin impacts inflammatory response in lactating sows. One hundred and ten sows were randomly assigned to a control diet (basal diet) or treatment diet (basal diet and 40 g/d silymarin) from the 108th day of gestation to weaning. Blood, milk, and feces from sows were collected for analysis. It was shown in the results that dietary silymarin supplementation decreased the level of pro-inflammatory cytokine IL-1β (p < 0.05) on the 18th day of lactation in the blood of the sows. Dietary silymarin supplementation tended to decrease (p = 0.06) somatic cell count in the colostrum of sows. Dietary silymarin supplementation reduced the gut bacterial community and the richness of the gut microbial community (p < 0.01) using 16S rRNA gene sequencing. The fecal microbes varied at different taxonomic levels in the lactating sows with silymarin supplementation. The most representative changes included an increase in the relative abundance of Fibrobacteres and Actinobacteria (p < 0.05) and tended to reduce the relative abundance of Spirochaetaes and Tenericutes (p = 0.09, 0.06) at the phylum level. It is suggested that dietary silymarin supplementation in late gestation until lactation has anti-inflammatory effects in lactation sow, which could be associated with the modulation of gut microbiota.
Collapse
Affiliation(s)
- Shengyu Xu
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: (S.X.); (D.W.)
| | - Xiaojun Jiang
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinlin Jia
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuemei Jiang
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lianqiang Che
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Lin
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yong Zhuo
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengfeng Fang
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jian Li
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jianping Wang
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - De Wu
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: (S.X.); (D.W.)
| |
Collapse
|
9
|
Bond KH, Sims-Lucas S, Oxburgh L. Targets for Renal Carcinoma Growth Control Identified by Screening FOXD1 Cell Proliferation Pathways. Cancers (Basel) 2022; 14:cancers14163958. [PMID: 36010951 PMCID: PMC9406217 DOI: 10.3390/cancers14163958] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary FOXD1 regulates the proliferation of clear cell renal cell carcinoma (ccRCC) cells, and ccRCC cells in which FOXD1 has been inactivated do not form tumors efficiently in an animal model. Reproducing growth inhibition in tumor cells by inhibiting FOXD1 pathways presents a possible therapeutic approach for ccRCC and other cancers. We have established an analysis strategy to identify FOXD1-regulated target pathways that may be therapeutically tractable, and compounds that modulate these pathways were selected for testing. Targets in three pathways were identified: FOXM1, PME1, and TMEM167A, which were inhibited by compounds FDI-6, AMZ-30, and silibinin, respectively. The effects of these compounds on the growth of tumor cells from patients cultured in a novel 3D tumor-replica culture environment revealed that FDI-6 and silibinin had strong growth inhibitory effects. This investigation informs new therapeutic targets to control ccRCC tumor growth, and provides a strategy to compare the responsiveness of individual patient tumor replicas to growth-inhibitory compounds. Abstract Clinical association studies suggest that FOXD1 is a determinant of patient outcome in clear cell renal cell carcinoma (ccRCC), and laboratory investigations have defined a role for this transcription factor in controlling the growth of tumors through regulation of the G2/M cell cycle transition. We hypothesized that the identification of pathways downstream of FOXD1 may define candidates for pharmacological modulation to suppress the G2/M transition in ccRCC. We developed an analysis pipeline that utilizes RNA sequencing, transcription factor binding site analysis, and phenotype validation to identify candidate effectors downstream from FOXD1. Compounds that modulate candidate pathways were tested for their ability to cause growth delay at G2/M. Three targets were identified: FOXM1, PME1, and TMEM167A, which were targeted by compounds FDI-6, AMZ-30, and silibinin, respectively. A 3D ccRCC tumor replica model was used to investigate the effects of these compounds on the growth of primary cells from five patients. While silibinin reduced 3D growth in a subset of tumor replicas, FDI-6 reduced growth in all. This study identifies tractable pathways to target G2/M transition and inhibit ccRCC growth, demonstrates the applicability of these strategies across patient tumor replicas, and provides a platform for individualized patient testing of compounds that inhibit tumor growth.
Collapse
Affiliation(s)
- Kyle H. Bond
- Rogosin Institute, Room 2-43, 310 East 67th St., New York, NY 10065, USA
| | - Sunder Sims-Lucas
- Children’s Hospital of Pittsburgh, Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Leif Oxburgh
- Rogosin Institute, Room 2-43, 310 East 67th St., New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
10
|
The role of protein kinases as key drivers of metabolic dysfunction-associated fatty liver disease progression: New insights and future directions. Life Sci 2022; 305:120732. [PMID: 35760093 DOI: 10.1016/j.lfs.2022.120732] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD), proposed in 2020 is a novel term for non-alcoholic fatty liver disease (NAFLD) which was coined for the first time in 1980. It is a leading cause of the most chronic liver disease and hepatic failure all over the world, and unfortunately, with no licensed drugs for treatment yet. The progress of the disease is driven by the triggered inflammatory process, oxidative stress, and insulin resistance in many pathways, starting with simple hepatic steatosis to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and liver cancer. Protein kinases (PKs), such as MAPK, ErbB, PKC, PI3K/Akt, and mTOR, govern most of the pathological pathways by acting on various downstream key points in MAFLD and regulating both hepatic gluco- lipo-neogenesis and inflammation. Therefore, modulating the function of those potential protein kinases that are effectively involved in MAFLD might be a promising therapeutic approach for tackling this disease. In the current review, we have discussed the key role of protein kinases in the pathogenesis of MAFLD and performed a protein-protein interaction (PPI) network among the main proteins of each kinase pathway with MAFLD-related proteins to predict the most likely targets of the PKs in MAFLD. Moreover, we have reported the experimental, pre-clinical, and clinical data for the most recent investigated molecules that are activating p38-MAPK and AMPK proteins and inhibiting the other PKs to improve MAFLD condition by regulating oxidation and inflammation signalling.
Collapse
|
11
|
Systematic Optimization of Solid Lipid Nanoparticles of Silybin for Improved Oral Drug Delivery by Box-Behnken Design: In Vitro and In Vivo Evaluations. J Pharm Innov 2022. [DOI: 10.1007/s12247-022-09637-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
12
|
Patel A, Perl A. Redox Control of Integrin-Mediated Hepatic Inflammation in Systemic Autoimmunity. Antioxid Redox Signal 2022; 36:367-388. [PMID: 34036799 PMCID: PMC8982133 DOI: 10.1089/ars.2021.0068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/20/2022]
Abstract
Significance: Systemic autoimmunity affects 3%-5% of the population worldwide. Systemic lupus erythematosus (SLE) is a prototypical form of such condition, which affects 20-150 of 100,000 people globally. Liver dysfunction, defined by increased immune cell infiltration into the hepatic parenchyma, is an understudied manifestation that affects up to 20% of SLE patients. Autoimmunity in SLE involves proinflammatory lineage specification in the immune system that occurs with oxidative stress and profound changes in cellular metabolism. As the primary metabolic organ of the body, the liver is uniquely capable to encounter oxidative stress through first-pass derivatization and filtering of waste products. Recent Advances: The traffic of immune cells from their development through recirculation in the liver is guided by cell adhesion molecules (CAMs) and integrins, cell surface proteins that tightly anchor cells together. The surface expression of CAMs and integrins is regulated via endocytic traffic that is sensitive to oxidative stress. Reactive oxygen species (ROS) that elicit oxidative stress in the liver may originate from the mitochondria, the cytosol, or the cell membrane. Critical Issues: While hepatic ROS production is a source of vulnerability, it also modulates the development and function of the immune system. In turn, the liver employs antioxidant defense mechanisms to protect itself from damage that can be harnessed to serve as therapeutic mechanisms against autoimmunity, inflammation, and development of hepatocellular carcinoma. Future Directions: This review is aimed at delineating redox control of integrin signaling in the liver and checkpoints of regulatory impact that can be targeted for treatment of inflammation in systemic autoimmunity. Antioxid. Redox Signal. 36, 367-388.
Collapse
Affiliation(s)
- Akshay Patel
- Division of Rheumatology, Department of Medicine, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Microbiology and Immunology, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Andras Perl
- Division of Rheumatology, Department of Medicine, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Microbiology and Immunology, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
13
|
Xu Y, Li J, He B, Feng T, Liang L, Huang X. In vitro Dissolution Testing and Pharmacokinetic Studies of Silymarin Solid Dispersion After Oral Administration to Healthy Pigs. Front Vet Sci 2022; 9:815198. [PMID: 35300217 PMCID: PMC8921073 DOI: 10.3389/fvets.2022.815198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
We evaluated the pharmacokinetics of silymarin solid dispersion in pigs to determine whether silybin bioavailability would be increased over that of a silymarin premix. In vitro dissolution testing was conducted using dissolution apparatus 1 (baskets) at 100 rpm at 37 ± 0.5°C in pH 1.2 HCl, pH 6.8 phosphate, and pH 4.3 acetate buffers containing 0.5% Tween-80. In vivo pharmacokinetics were studied using 16 healthy pigs (Yorkshire × Landrace) that were randomly assigned to two groups. Silymarin as solid dispersion and premix dosage forms were administered directly by stomach tubes at 50 mg kg−1 silybin. In vitro dissolution of silybin for the premix was 35.02, 35.90, and 38.70% in these buffers, respectively. In contrast, silybin dissolution in solid dispersions was increased to 82.92, 87.48, and 99.70%, respectively. Silymarin solid dispersion administered at a single dose resulted in a peak concentration (Cmax) of 1,190.02 ± 246.97 ng ml−1 with the area under the curve (AUC0−∞) at 1,299.19 ± 67.61 ng ml−1 h. These parameters for the premix groups were 411.35 ± 84.92 ng ml−1 and 586.82 ± 180.99 ng ml−1 h, respectively. The Cmax and AUC0−∞ values for the solid dispersion were about twice that of the premix and were consistent with the in vitro dissolution data.
Collapse
|
14
|
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: 37] [Impact Index Per Article: 12.3] [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
|
15
|
Fatima S, Kumari A, Dwivedi VP. Advances in adjunct therapy against tuberculosis: Deciphering the emerging role of phytochemicals. MedComm (Beijing) 2021; 2:494-513. [PMID: 34977867 PMCID: PMC8706769 DOI: 10.1002/mco2.82] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
Eastern countries are a major source of medicinal plants, which set up a rich source of ethnopharmacologically known medicines used in the treatment of various diseases. These traditional medicines have been known as complementary, alternative, or nonconventional therapy across globe for ages. Tuberculosis (TB) poses a huge global burden and leads to maximum number of deaths due to an infectious agent. Treatment of TB using Directly Observed Treatment Short-course (DOTS) therapy comprises multiple antibiotics is quite lengthy and causes serious side-effects in different organs. The length of the TB treatment leads to withdrawal from the patients, which paves the way for the emergence of drug resistance in the bacterial population. These concerns related to therapy need serious and immediate interventions. Traditional medicines using phytochemicals has shown to provide tremendous potential in TB treatment, mainly in the eradication of Mycobacterium tuberculosis (M.tb), increasing natural immunity, and managing the side effects of anti-TB drugs. This review describes the antituberculosis potential of selected ethnopharmacologically important phytochemicals as potential immune-modulator and as an adjunct-therapy in TB. This review will be a useful reference for researchers working on ethnopharmacology and will open the door for the discovery of novel agents as an adjunct-therapy to tuberculosis.
Collapse
Affiliation(s)
- Samreen Fatima
- Immunobiology GroupInternational Centre for Genetic Engineering and BiotechnologyNew DelhiIndia
| | - Anjna Kumari
- Immunobiology GroupInternational Centre for Genetic Engineering and BiotechnologyNew DelhiIndia
| | - Ved Prakash Dwivedi
- Immunobiology GroupInternational Centre for Genetic Engineering and BiotechnologyNew DelhiIndia
| |
Collapse
|
16
|
Kim SH, Choo GS, Yoo ES, Woo JS, Lee JH, Han SH, Jung SH, Kim HJ, Jung JY. Silymarin inhibits proliferation of human breast cancer cells via regulation of the MAPK signaling pathway and induction of apoptosis. Oncol Lett 2021; 21:492. [PMID: 33968208 PMCID: PMC8100955 DOI: 10.3892/ol.2021.12753] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/16/2021] [Indexed: 12/30/2022] Open
Abstract
Silymarin is a purified mixture of four isomeric flavonoids extracted from the seeds and fruit of the milk thistle plant, Silybum marianus (L.). Silymarin exhibits a wide variety of biological effects and is commonly used in traditional medicine. Therefore, the anticancer effects of silymarin on human breast cancer cells were investigated to determine its pharmacological mechanisms in vitro and in vivo. The viability and proliferation of MDA-MB- 231 and MCF-7 breast cancer cells were investigated using MTT and wound healing assays. Silymarin decreased the viability and proliferation of MDA-MB-231 and MCF-7 cells in a concentration-dependent manner. The number of apoptotic bodies, as shown by DAPI staining, was increased in a concentration-dependent manner, indicating that silymarin induces apoptosis. Additionally, changes in the expression levels of apoptosis-related proteins were demonstrated in human breast cancer cells using western blotting. Silymarin increased the levels of Bax, cleaved poly-ADP ribose polymerase, cleaved caspase-9 and phosphorylated (p-)JNK, and decreased the levels of Bcl-2, p-P38 and p-ERK1/2. Furthermore, the inhibitory effects of silymarin on MCF-7 tumor growth were investigated. In mice treated with silymarin for 3 weeks (25 and 50 mg/kg), MCF-7 tumor growth was inhibited without organ toxicity. In MCF-7 tumors, silymarin induced apoptosis and decreased p-ERK1/2 levels, as assessed using a TUNEL assay and immunohistochemistry. These results indicated that silymarin inhibited breast cancer cell proliferation both in vitro and in vivo by modulating the MAPK signaling pathway. Therefore, silymarin may potentially be used as a chemo-preventive or therapeutic agent.
Collapse
Affiliation(s)
- Sung-Hyun Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| | - Gang-Sik Choo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| | - Eun-Seon Yoo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| | - Joong-Seok Woo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| | - Jae-Han Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| | - So-Hee Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| | - Soo-Hyun Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| | - Hyeong-Jin Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 340-702, Republic of Korea
| |
Collapse
|
17
|
Kesharwani SS, Jain V, Dey S, Sharma S, Mallya P, Kumar VA. An overview of advanced formulation and nanotechnology-based approaches for solubility and bioavailability enhancement of silymarin. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
18
|
Scaria B, Sood S, Raad C, Khanafer J, Jayachandiran R, Pupulin A, Grewal S, Okoko M, Arora M, Miles L, Pandey S. Natural Health Products (NHP's) and Natural Compounds as Therapeutic Agents for the Treatment of Cancer; Mechanisms of Anti-Cancer Activity of Natural Compounds and Overall Trends. Int J Mol Sci 2020; 21:E8480. [PMID: 33187200 PMCID: PMC7697102 DOI: 10.3390/ijms21228480] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Most cancer therapeutics, such as tubulin-targeting chemotherapy drugs, cause cytotoxic, non-selective effects. These harmful side-effects drastically reduce the cancer patient's quality of life. Recently, researchers have focused their efforts on studying natural health products (NHP's) which have demonstrated the ability to selectively target cancer cells in cellular and animal models. However, the major hurdle of clinical validation remains. NHP's warrant further clinical investigation as a therapeutic option since they exhibit low toxicity, while retaining a selective effect. Additionally, they can sensitize cancerous cells to chemotherapy, which enhances the efficacy of chemotherapeutic drugs, indicating that they can be utilized as supplemental therapy. An additional area for further research is the investigation of drug-drug interactions between NHP's and chemotherapeutics. The objectives of this review are to report the most recent results from the field of anticancer NHP research, and to highlight the most recent advancements in possible supplemental therapeutic options.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Siyaram Pandey
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada; (B.S.); (S.S.); (C.R.); (J.K.); (R.J.); (A.P.); (S.G.); (M.O.); (M.A.); (L.M.)
| |
Collapse
|
19
|
Tuli HS, Mittal S, Aggarwal D, Parashar G, Parashar NC, Upadhyay SK, Barwal TS, Jain A, Kaur G, Savla R, Sak K, Kumar M, Varol M, Iqubal A, Sharma AK. Path of Silibinin from diet to medicine: A dietary polyphenolic flavonoid having potential anti-cancer therapeutic significance. Semin Cancer Biol 2020; 73:196-218. [PMID: 33130037 DOI: 10.1016/j.semcancer.2020.09.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/11/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
In the last few decades, targeting cancer by the use of dietary phytochemicals has gained enormous attention. The plausible reason and believe or mind set behind this fact is attributed to either lesser or no side effects of natural compounds as compared to the modern chemotherapeutics, or due to their conventional use as dietary components by mankind for thousands of years. Silibinin is a naturally derived polyphenol (a flavonolignans), possess following biochemical features; molecular formula C25H22O10, Molar mass: 482.44 g/mol, Boiling point 793 °C, with strikingly high antioxidant and anti-tumorigenic properties. The anti-cancer properties of Silibinin are determined by a variety of cellular pathways which include induction of apoptosis, cell cycle arrest, inhibition of angiogenesis and metastasis. In addition, Silibinin controls modulation of the expression of aberrant miRNAs, inflammatory response, and synergism with existing anti-cancer drugs. Therefore, modulation of a vast array of cellular responses and homeostatic aspects makes Silibinin an attractive chemotherapeutic agent. However, like other polyphenols, the major hurdle to declare Silibinin a translational chemotherapeutic agent, is its lesser bioavailability. After summarizing the chemistry and metabolic aspects of Silibinin, this extensive review focuses on functional aspects governed by Silibinin in chemoprevention with an ultimate goal of summarizing the evidence supporting the chemopreventive potential of Silibinin and clinical trials that are currently ongoing, at a single platform.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Sonam Mittal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | | | - Sushil Kumar Upadhyay
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Tushar Singh Barwal
- Department of Zoology, Central University of Punjab, Bathinda, 151 001, Punjab, India
| | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda, 151 001, Punjab, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai, 400 056, Maharastra, India
| | - Raj Savla
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai, 400 056, Maharastra, India
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur, India
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla, TR48000, Turkey
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research (Formerly Faculty of Pharmacy), Jamia Hamdard (Deemed to be University), Delhi, India
| | - Anil Kumar Sharma
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India.
| |
Collapse
|
20
|
Darvishi-Khezri H, Kosaryan M, Karami H, Salehifar E, Mahdavi M, Alipour A, Aliasgharian A. Can Use of Silymarin Improve Inflammatory Status in Patients with β-Thalassemia Major? A Crossover, Randomized Controlled Trial. Complement Med Res 2020; 28:123-130. [PMID: 32971524 DOI: 10.1159/000509829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/29/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND In β-thalassemia major (β-TM) patients, iron overload is one of the main causes of inflammation. This study investigated whether use of silymarin could improve inflammatory status in patients with β-TM and iron overload, through a placebo-controlled, crossover study. METHODS Silymarin (140 mg, 3 times a day) or placebo were prescribed to all patients (n = 82) for 12 weeks, and after a 2-week washout period, patients were crossed over to the other group. The efficacy of silymarin was assessed by measuring serum C-reactive protein (CRP) (mg/dL), interleukin (IL)-6 (pg/mL), and IL-10 (pg/mL). RESULTS Sixty-nine patients completed the study. Data analysis showed that compared to the placebo, silymarin could decrease CRP, IL-6, and raise IL-10 significantly (the p values for all variables were <0.001). Cohen's d for CRP adjusted according to the baseline CRP value was -1.72, the 95% confidence interval (CI) -2.12 to -1.33. The adjusted Cohen's d equal to -1.12, 95% CI -1.48 to -0.76, and 0.78, 95% CI 0.43-1.12, were also estimated for IL-6 and IL-10, respectively. CONCLUSION The results of the current study demonstrate that the combination of iron chelation therapy with silymarin can improve inflammatory status in patients with β-TM in the clinical setting.
Collapse
Affiliation(s)
- Hadi Darvishi-Khezri
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehrnoush Kosaryan
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Karami
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ebrahim Salehifar
- Phamaceutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran,
| | - Mohammadreza Mahdavi
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abbas Alipour
- Department of Community Medicine, Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Aily Aliasgharian
- Medical Microbiology, Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
21
|
Lazzeroni M, Petrangolini G, Legarreta Iriberri JA, Pascual Avellana J, Tost Robusté D, Cagnacci S, Macis D, Aristarco V, Bonanni B, Morazzoni P, Johansson H, Riva A. Development of an HPLC-MS/MS Method for the Determination of Silybin in Human Plasma, Urine and Breast Tissue. Molecules 2020; 25:molecules25122918. [PMID: 32599946 PMCID: PMC7356828 DOI: 10.3390/molecules25122918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 11/01/2022] Open
Abstract
Silybin is a flavonolignan extracted from Silybum marianum with chemopreventive activity against various cancers, including breast. This study was designed to develop an HPLC-MS/MS method for the determination of silybin in human plasma, urine and breast tissue in early breast cancer patients undergoing Siliphos® supplementation, an oral silybin-phosphatidylcholine complex. The determination of silybin was carried out by liquid-liquid extraction with methyl-tert-butyl ether (MTBE); total silybin concentration was determined by treating the samples with β-glucuronidase, while for the determination of free silybin, the hydrolytic step was omitted. Naringenin and naproxen were selected as internal standards. The detection of the analyte was carried out by mass spectrometry and by chromatography. The HPLC-MS/MS method was evaluated in terms of selectivity, linearity, limit of quantification, precision and accuracy, and carryover. The method proved to be selective, linear, precise and accurate for the determination of silybin. To the best of our knowledge, this presents the first analytical method with the capacity to quantify the major bioactive components of milk thistle in three different biological matrices with a lower limit of quantification of 0.5 ng/mL for plasma. Silybin phosphatidylcholine, taken orally, can deliver high blood concentrations of silybin, which selectively accumulates in breast tumor tissue.
Collapse
Affiliation(s)
- Matteo Lazzeroni
- IEO-European Institute of Oncology, Division of Cancer Prevention and Genetics, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.L.); (S.C.); (D.M.); (V.A.); (B.B.); (H.J.)
| | - Giovanna Petrangolini
- Research and Development Department, Indena S.p.A., Viale Ortles 12, 20139 Milan, Italy; (P.M.); (A.R.)
- Correspondence:
| | - José Antonio Legarreta Iriberri
- Bioanalysis Department, Kymos Pharma Services S.L., Ronda Can Fatjó 7-B, 08290 Cerdanyola del Vallès, Spain; (J.A.L.I.); (J.P.A.); (D.T.R.)
| | - Jaume Pascual Avellana
- Bioanalysis Department, Kymos Pharma Services S.L., Ronda Can Fatjó 7-B, 08290 Cerdanyola del Vallès, Spain; (J.A.L.I.); (J.P.A.); (D.T.R.)
| | - Digna Tost Robusté
- Bioanalysis Department, Kymos Pharma Services S.L., Ronda Can Fatjó 7-B, 08290 Cerdanyola del Vallès, Spain; (J.A.L.I.); (J.P.A.); (D.T.R.)
| | - Sara Cagnacci
- IEO-European Institute of Oncology, Division of Cancer Prevention and Genetics, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.L.); (S.C.); (D.M.); (V.A.); (B.B.); (H.J.)
| | - Debora Macis
- IEO-European Institute of Oncology, Division of Cancer Prevention and Genetics, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.L.); (S.C.); (D.M.); (V.A.); (B.B.); (H.J.)
| | - Valentina Aristarco
- IEO-European Institute of Oncology, Division of Cancer Prevention and Genetics, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.L.); (S.C.); (D.M.); (V.A.); (B.B.); (H.J.)
| | - Bernardo Bonanni
- IEO-European Institute of Oncology, Division of Cancer Prevention and Genetics, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.L.); (S.C.); (D.M.); (V.A.); (B.B.); (H.J.)
| | - Paolo Morazzoni
- Research and Development Department, Indena S.p.A., Viale Ortles 12, 20139 Milan, Italy; (P.M.); (A.R.)
| | - Harriet Johansson
- IEO-European Institute of Oncology, Division of Cancer Prevention and Genetics, IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.L.); (S.C.); (D.M.); (V.A.); (B.B.); (H.J.)
| | - Antonella Riva
- Research and Development Department, Indena S.p.A., Viale Ortles 12, 20139 Milan, Italy; (P.M.); (A.R.)
| |
Collapse
|
22
|
Chi C, Zhang C, Liu Y, Nie H, Zhou J, Ding Y. Phytosome-nanosuspensions for silybin-phospholipid complex with increased bioavailability and hepatoprotection efficacy. Eur J Pharm Sci 2020; 144:105212. [PMID: 31923602 DOI: 10.1016/j.ejps.2020.105212] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/30/2019] [Accepted: 01/06/2020] [Indexed: 12/15/2022]
Abstract
Silybin, a natural compound for treating liver disease, has been shown to provide diverse biological activities such as anticancer, antioxidant and hepatoprotective. However, it is still challenging to develop silybin product due to its poor aqueous solubility and limited gastrointestinal absorption. In order to improve the low bioavailability of silybin, a novel formulation of phytosome-nanosuspensions for silybin shielding termed as SPCs-NPs, has been developed herein for hepatoprotection efficacy. We found that SPCs-NPs formulation not only possessed an increased in vitro dissolution rate but also improved plasma concentration in the in vivo pharmacokinetic study. Moreover, SPCs-NPs was provided with more potent hepatoprotective effects in pharmacodynamic assessments. Moreover, physicochemical features including interactions between silybin and phospholipid, and crystalline variation of the optimized SPCs-NPs formulation were confirmed by using Fourier-transform infrared spectrometry (FTIR), 1H nuclear magnetic resonance spectroscopy (H-NMR), differential scanning calorimetry (DSC), and powder X-ray diffraction spectroscopy (PXRD) respectively. Overall, the interesting finding of this study suggested that SPCs-NPs could be applied as a promising formulation for a higher drug bioavailability and better hepatoprotection efficacy.
Collapse
Affiliation(s)
- Cheng Chi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Chenshuang Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Haichen Nie
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Station Mall Drive, West Lafayette, IN 47907, United States
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| | - Yang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| |
Collapse
|
23
|
Sieniawska E, Maciejewska-Turska M, Świątek Ł, Xiao J. Plant-based Food Products for Antimycobacterial Therapy. EFOOD 2020. [DOI: 10.2991/efood.k.200418.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
24
|
The Importance of Natural Antioxidants in the Treatment of Spinal Cord Injury in Animal Models: An Overview. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3642491. [PMID: 32676138 PMCID: PMC7336207 DOI: 10.1155/2019/3642491] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/04/2019] [Indexed: 02/06/2023]
Abstract
Patients with spinal cord injury (SCI) face devastating health, social, and financial consequences, as well as their families and caregivers. Reducing the levels of reactive oxygen species (ROS) and oxidative stress are essential strategies for SCI treatment. Some compounds from traditional medicine could be useful to decrease ROS generated after SCI. This review is aimed at highlighting the importance of some natural compounds with antioxidant capacity used in traditional medicine to treat traumatic SCI. An electronic search of published articles describing animal models of SCI treated with natural compounds from traditional medicine was conducted using the following terms: Spinal Cord Injuries (MeSH terms) AND Models, Animal (MeSH terms) AND [Reactive Oxygen Species (MeSH terms) AND/OR Oxidative Stress (MeSH term)] AND Medicine, Traditional (MeSH terms). Articles reported from 2010 to 2018 were included. The results were further screened by title and abstract for studies performed in rats, mice, and nonhuman primates. The effects of these natural compounds are discussed, including their antioxidant, anti-inflammatory, and antiapoptotic properties. Moreover, the antioxidant properties of natural compounds were emphasized since oxidative stress has a fundamental role in the generation and progression of several pathologies of the nervous system. The use of these compounds diminishes toxic effects due to their high antioxidant capacity. These compounds have been tested in animal models with promising results; however, no clinical studies have been conducted in humans. Further research of these natural compounds is crucial to a better understanding of their effects in patients with SCI.
Collapse
|
25
|
Yu Y, Li LF, Tao J, Zhou XM, Xu C. Silibinin induced apoptosis of human epidermal cancer A431 cells by promoting mitochondrial NOS. Free Radic Res 2019; 53:714-726. [DOI: 10.1080/10715762.2019.1603376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Yu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Lan-fang Li
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jing Tao
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Xiao-mian Zhou
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Cheng Xu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, Shenyang, PR China
| |
Collapse
|
26
|
Radko L, Cybulski W. The decrease of lasalocid residue in the edible tissues by silymarin supplementation of chicken diet. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 2019; 36:722-728. [PMID: 30973080 DOI: 10.1080/19440049.2019.1584406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Widespread use of coccidiostats, in spite of beneficial control of protozoan infections in poultry, implies a risk of residues in edible tissues, and there is increasing interest in the development of strategies for prevention of veterinary drugs residue in food-producing animals. The aim of this study is assigned to clarify the impact of silymarin addendum in the diet on lasalocid concentration in the liver and breast muscles from the broiler. Four groups of chickens received a feed with lasalocid at levels between 75 and 200 mg kg-1. Other four groups received a feed with lasalocid (75-200 mg kg-1) plus silymarin. Significant differences of lasalocid concentrations between the liver and breast muscles were observed. Moreover, the chickens from the groups supplemented with silymarin shown significant decreases of lasalocid concentrations in the analysed tissues. The herbal substance did not counteract the ionophore in the treatment of coccidiosis and did not change biochemical parameters of blood. These findings suggest that silymarin might be used in chicken feeding in order to reduce the risk from lasalocid contamination of the broiler edible tissues.
Collapse
Affiliation(s)
- Lidia Radko
- a Department of Pharmacology and Toxicology , National Veterinary Research Institute , Pulawy , Poland
| | - Wojciech Cybulski
- a Department of Pharmacology and Toxicology , National Veterinary Research Institute , Pulawy , Poland
| |
Collapse
|
27
|
Saad MA, Rastanawi AA, El-Yamany MF. Alogliptin abates memory injuries of hepatic encephalopathy induced by acute paracetamol intoxication via switching-off autophagy-related apoptosis. Life Sci 2018; 215:11-21. [DOI: 10.1016/j.lfs.2018.10.069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/22/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
|
28
|
Nahari E, Razi M. Silymarin amplifies apoptosis in ectopic endometrial tissue in rats with endometriosis; implication on growth factor GDNF, ERK1/2 and Bcl-6b expression. Acta Histochem 2018; 120:757-767. [PMID: 30195499 DOI: 10.1016/j.acthis.2018.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/29/2018] [Accepted: 08/16/2018] [Indexed: 12/25/2022]
Abstract
The present prospective study was done to evaluate the effect of silymarin (SMN) on endometriotic-like legions establishment and growth in experimentally-induced endometriosis. For this purpose, the experimental endometriosis was induced in 12 rats and then the animals subdivided into endometriosis-sole and SMN (50 mg kg-1, orally)+endometriosis groups. Following 28 days, the legions establishment, size, Glial cell line-derived neurotrophic factor (GDNF), gfrα1, B Cell Lymphoma 6 (Bcl-6b), Bcl-2, extracellular regulator kinase (ERK1/2) expression ratios, angiogenesis, the apoptosis and fibrosis indices were investigated. The SMN significantly (P < 0.05) decreased the enometriotic-like legions establishment and size, decreased mRNA levels of GDNF, gfrα1, Bcl-6b and Bcl-2 and remarkably diminished GDNF, gfrα1, Bcl-6b and Bcl-2-positive cells distribution/mm2 of tissue versus endometriosis-sole group. The SMN + endometriosis group exhibited a significant (P < 0.05) enhancement in ERK1/2 expression and represented diminished vascularized area and increased apoptosis and fibrosis indices, as well. In conclusion, the SMN by down-regulating GDNF and its receptor gfrα1 expression inhibits GDNF-gfrα1 complex generation and consequently suppresses Bcl-6b expression. Moreover, the SMN by enhancing the ERK1/2 expression and by suppressing the Bcl-2 expression promotes the apoptosis pathway. Finally, the SMN by down-regulating the angiogenesis ratio accelerates apoptosis and consequently induces severe fibrosis in endometriotic-like legions.
Collapse
Affiliation(s)
- Elaheh Nahari
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Mazdak Razi
- Department of Basic Sciences, Faculty of Veterinary Medicine, P.O. BOC: 1177, Urmia University, Urmia, Iran.
| |
Collapse
|
29
|
Gabrielová E, Bartošíková L, Nečas J, Modrianský M. Cardioprotective effect of 2,3-dehydrosilybin preconditioning in isolated rat heart. Fitoterapia 2018; 132:12-21. [PMID: 30385403 DOI: 10.1016/j.fitote.2018.10.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/23/2018] [Accepted: 10/28/2018] [Indexed: 12/11/2022]
Abstract
2,3-dehydrosilybin (DHS) is a minor component of silymarin, Silybum marianum seed extract, used in some dietary supplements. One of the most promising activities of this compound is its anticancer and cardioprotective activity that results, at least partially, from its cytoprotective, antioxidant, and chemopreventive properties. The present study investigated the cardioprotective effects of DHS in myocardial ischemia and reperfusion injury in rats. Isolated hearts were perfused by the Langendorff technique with low dose DHS (100 nM) prior to 30 min of ischemia induced by coronary artery occlusion. After 60 min of coronary reperfusion infarct size was determined by triphenyltetrazolium staining, while lactatedehydrogenase activity was evaluated in perfusate samples collected at several timepoints during the entire perfusion procedure. Signalosomes were isolated from a heart tissue after reperfusion and involved signalling proteins were detected. DHS reduced the extent of infarction compared with untreated control hearts at low concentration; infarct size as proportion of ischemic risk zone was 7.47 ± 3.1% for DHS versus 75.3 ± 4.8% for ischemia. This protective effect was comparable to infarct limitation induced by ischemic preconditioning (22.3 ± 4.5%). Selective inhibition of Src-family kinases with PP2 (4-Amino-3-(4-chlorophenyl)-1-(t-butyl)-1H-pyrazolo[3,4-d]pyrimidine) abrogated the protection afforded by DHS. This study provides experimental evidence that DHS can mediate Src-kinase-dependent cardioprotection against myocardial damage produced by ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Eva Gabrielová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Lenka Bartošíková
- Department of Physiology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Jiří Nečas
- Department of Physiology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Martin Modrianský
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic.
| |
Collapse
|
30
|
Dupuis ML, Conti F, Maselli A, Pagano MT, Ruggieri A, Anticoli S, Fragale A, Gabriele L, Gagliardi MC, Sanchez M, Ceccarelli F, Alessandri C, Valesini G, Ortona E, Pierdominici M. The Natural Agonist of Estrogen Receptor β Silibinin Plays an Immunosuppressive Role Representing a Potential Therapeutic Tool in Rheumatoid Arthritis. Front Immunol 2018; 9:1903. [PMID: 30174672 PMCID: PMC6107853 DOI: 10.3389/fimmu.2018.01903] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/01/2018] [Indexed: 12/22/2022] Open
Abstract
Estrogens, in particular 17β-estradiol (E2), have a strong influence on the immune system and also affect pathological conditions such as autoimmune diseases. The biological effects of E2 are mediated by two intracellular receptors, i.e., estrogen receptor (ER)α and ERβ, which function as ligand-activated nuclear transcription factors producing genomic effects. Immune cells express both ERα and ERβ that play a complex role in modulating inflammation. Phytoestrogens display estrogen-like effects. Among them, silibinin, the major active constituent of silymarin extracted by the milk thistle (Silybum marianum), has been suggested to have an ERβ selective binding. Silibinin is known to have anti-inflammatory, hepatoprotective, and anticarcinogenic effects; however, the role of silibinin in modulating human immune responses and its impact on autoimmunity remains unclear. Aim of this study was to dissect the ability of the ERβ natural ligand silibinin to modulate T cell immunity, taking into account possible differences between females and males, and to define its possible role as therapeutic tool in immune-mediated diseases. To this purpose, female and age-matched male healthy subjects and patients with active rheumatoid arthritis (RA) were recruited. We evaluated the ability of silibinin to modulate ERβ expression in T lymphocytes and its effects on T cell functions (i.e., apoptosis, proliferation, and cytokine production). We also analyzed whether silibinin was able to modulate the expression of microRNA-155 (miR-155), which strongly contributes to the pathogenesis of RA driving aberrant activation of the immune system. We demonstrated that silibinin upregulated ERβ expression, induced apoptosis, inhibited proliferation, and reduced expression of the pro-inflammatory cytokines IL-17 and TNF-α, through ERβ binding, in T lymphocytes from female and male healthy donors. We obtained similar results in T lymphocytes from patients with active RA in term of apoptosis, proliferation, and cytokine production. In addition, we found that silibinin acted as an epigenetic modifier, down-modulating the expression of miR-155. In conclusion, our data demonstrated an immunosuppressive role of silibinin, supporting its application in the treatment of autoimmune diseases as drug, but also as dietary nutritional supplement, opening new perspective in the field of autoimmune disease management.
Collapse
Affiliation(s)
- Maria Luisa Dupuis
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Fabrizio Conti
- Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Angela Maselli
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Teresa Pagano
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Ruggieri
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Anticoli
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Fragale
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Gabriele
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Fulvia Ceccarelli
- Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Guido Valesini
- Rheumatology Unit, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Elena Ortona
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Pierdominici
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
31
|
Safarpoor M, Ghaedi M, Asfaram A, Yousefi-Nejad M, Javadian H, Zare Khafri H, Bagherinasab M. Ultrasound-assisted extraction of antimicrobial compounds from Thymus daenensis and Silybum marianum: Antimicrobial activity with and without the presence of natural silver nanoparticles. ULTRASONICS SONOCHEMISTRY 2018; 42:76-83. [PMID: 29429729 DOI: 10.1016/j.ultsonch.2017.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 06/08/2023]
Abstract
The present study is devoted to prepare a new antibacterial and antifungal agent based on in situ-synthesized silver nanoparticles at room temperature using Rosmarinus officinalis (R. officinalis) leaf extract. The Ag-NPs characterization by UV-visible, SEM, TEM and XRD revealed that the particles sizes were in the range of 10-33 nm. In this study, hydroalcoholic extracts were used with ultrasonic method. Ultrasonication has recently received attention as a novel bioprocessing tool for process intensification in many areas of downstream processing. The antimicrobial activities of T. daenensis and S. marianum extracts with and without the presence of Ag-NPs were investigated at concentrations from 12.5 to 50 mg/mL against Staphylococcus aureus (S. aureus, Gram-positive organism) and Escherichia coli (E. coli, Gram-negative organism), and fungal strains were Aspergillus oryzae (A. oryzae) and Candida albicans (C. albicans). Antimicrobial activity determined using agar disc diffusion method revealed that the activities of Ag-NPs/T. daenensis were superior to Ag-NPs/S. marianum and extracts (T. daenensis and S. marianum). The medicinal plant extract can be used to synthesize the Ag-NPs as an eco-friendly and inexpensive method in large scale. The results showed that the prepared Ag-NPs/extracts as good antibacterial and antifungal agents can be potentially applied against rapidly increasing of antibiotic resistance.
Collapse
Affiliation(s)
| | - Mehrorang Ghaedi
- Department of Chemistry, Yasouj University, Yasouj 75918-74831, Iran.
| | - Arash Asfaram
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Hamedreza Javadian
- Universitat Politècnica de Catalunya, Department of Chemical Engineering, ETSEIB, Diagonal 647, 08028 Barcelona, Spain
| | | | - Marzieh Bagherinasab
- Department of Animal Science, Kermanshah University, Kermanshah 67156-85438, Iran
| |
Collapse
|
32
|
Hashemi Jabali NS, Mahdavi AH, Ansari Mahyari S, Sedghi M, Akbari Moghaddam Kakhki R. Effects of milk thistle meal on performance, ileal bacterial enumeration, jejunal morphology and blood lipid peroxidation in laying hens fed diets with different levels of metabolizable energy. J Anim Physiol Anim Nutr (Berl) 2018; 102:410-420. [PMID: 28608581 DOI: 10.1111/jpn.12747] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/20/2017] [Indexed: 11/30/2022]
Abstract
This study was conducted to evaluate the effects of different levels of milk thistle meal on performance, blood biochemical indices, ileal bacterial counts and intestinal histology in laying hens fed diets containing different levels of metabolizable energy. A total number of 200 Leghorn laying hens (Hy-Line W-36) were randomly assigned to eight experimental treatments with five cage replicates of five birds each. Dietary treatments consisted of four levels of milk thistle meal (0%, 15%, 30% and 60%) and two levels of AMEn (11.09 and 12.34 MJ/kg) fed over a period of 80 days. In vitro studies revealed that the total phenolic component of milk thistle meal was 470.64 mg gallic acid equivalent/g of the sample, and its antioxidant activity for inhibiting the 2-2-diphenyl-1-picrichydrazyl free radical and reducing ferric ions was about 21% higher than that of butylated hydroxyltoluene (p < .05). Diets containing high level of AMEn led to improved egg production (p < .05), egg weight (p < .05), egg mass (p < .01) and feed conversion ratio (p < .01). In addition, offering diets containing high energy significantly enhanced (p < .01) serum triglyceride and malondialdehyde (MDA) concentrations as well as jejunal villus height. Dietary supplementation of 3% milk thistle meal resulted in the best feed conversion ratio (p < .05), reduction of ileal Escherichia coli enumeration (p < .01) and an enhancement in the villus height-to-crypt depth ratio (p < .05). Furthermore, feeding incremental levels of this meal led to remarkable decrease in serum cholesterol, triglyceride and MDA (p < .01) concentrations while significant increase in blood high-density lipoprotein content and goblet cell numbers (p < .05). The present findings indicate that milk thistle meal with high antioxidant and antibacterial properties in laying hen diets may improve health indices and productive performance.
Collapse
Affiliation(s)
- N S Hashemi Jabali
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - A H Mahdavi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - S Ansari Mahyari
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - M Sedghi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | | |
Collapse
|
33
|
Papackova Z, Heczkova M, Dankova H, Sticova E, Lodererova A, Bartonova L, Poruba M, Cahova M. Silymarin prevents acetaminophen-induced hepatotoxicity in mice. PLoS One 2018; 13:e0191353. [PMID: 29342206 PMCID: PMC5771617 DOI: 10.1371/journal.pone.0191353] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022] Open
Abstract
Acetaminophen or paracetamol (APAP) overdose is a common cause of liver injury. Silymarin (SLM) is a hepatoprotective agent widely used for treating liver injury of different origin. In order to evaluate the possible beneficial effects of SLM, Balb/c mice were pretreated with SLM (100 mg/kg b.wt. per os) once daily for three days. Two hours after the last SLM dose, the mice were administered APAP (300 mg/kg b.wt. i.p.) and killed 6 (T6), 12 (T12) and 24 (T24) hours later. SLM-treated mice exhibited a significant reduction in APAP-induced liver injury, assessed according to AST and ALT release and histological examination. SLM treatment significantly reduced superoxide production, as indicated by lower GSSG content, lower HO-1 induction, alleviated nitrosative stress, decreased p-JNK activation and direct measurement of mitochondrial superoxide production in vitro. SLM did not affect the APAP-induced decrease in CYP2E1 activity and expression during the first 12 hrs. Neutrophil infiltration and enhanced expression of inflammatory markers were first detected at T12 in both groups. Inflammation progressed in the APAP group at T24 but became attenuated in SLM-treated animals. Histological examination suggests that necrosis the dominant cell death pathway in APAP intoxication, which is partially preventable by SLM pretreatment. We demonstrate that SLM significantly protects against APAP-induced liver damage through the scavenger activity of SLM and the reduction of superoxide and peroxynitrite content. Neutrophil-induced damage is probably secondary to necrosis development.
Collapse
Affiliation(s)
- Zuzana Papackova
- Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Veterinary Science, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czech Republic
- * E-mail:
| | - Marie Heczkova
- Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Helena Dankova
- Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Sticova
- Clinical and Transplant Pathology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Alena Lodererova
- Clinical and Transplant Pathology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lenka Bartonova
- Clinical and Transplant Pathology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Poruba
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Monika Cahova
- Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
34
|
Darvishi-Khezri H, Salehifar E, Kosaryan M, Karami H, Alipour A, Shaki F, Aliasgharian A. The impact of silymarin on antioxidant and oxidative status in patients with β-thalassemia major: A crossover, randomized controlled trial. Complement Ther Med 2017; 35:25-32. [DOI: 10.1016/j.ctim.2017.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/06/2017] [Accepted: 08/07/2017] [Indexed: 02/08/2023] Open
|
35
|
Molavi O, Narimani F, Asiaee F, Sharifi S, Tarhriz V, Shayanfar A, Hejazi M, Lai R. Silibinin sensitizes chemo-resistant breast cancer cells to chemotherapy. PHARMACEUTICAL BIOLOGY 2017; 55:729-739. [PMID: 28027688 PMCID: PMC6130726 DOI: 10.1080/13880209.2016.1270972] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/30/2016] [Accepted: 12/03/2016] [Indexed: 05/22/2023]
Abstract
CONTEXT Multiple drug resistance is the major obstacle to conventional chemotherapy. Silibinin, a nontoxic naturally occurring compound, has anticancer activity and can increase the cytotoxic effects of chemotherapy in various cancer models. OBJECTIVE To evaluate the effects of silibinin on enhancing the sensitivity of chemo-resistant human breast cell lines to doxorubicin (DOX) and paclitaxel (PAC). MATERIALS AND METHODS The cells were treated with silibinin (at 50 to 600 μM concentrations) and/or chemo drugs for 24 and 48 h, then cell viability and changes in oncogenic proteins were determined by MTT assay and Western blotting/RT-PCR, respectively. Flow cytometry was used to study apoptosis in the cells receiving different treatments. The antitumorigenic effects of silibinin (at 200 to 400 μM concentration) were evaluated by mammosphere assay. RESULTS Silibinin exerted significant growth inhibitory effects with IC50 ranging from 200 to 570 μM in different cell lines. Treatment of DOX-resistant MDA-MB-435 cells with silibinin at 200 μM reduced DOX IC50 from 71 to 10 μg/mL and significantly suppressed the key oncogenic pathways including STAT3, AKT, and ERK in these cells. Interestingly treatment of DOX-resistant MDA-MB-435 cells with silibinin at 400 μM concentration for 48 h induced a 50% decrease in the numbers of colonies as compared with DMSO-treated cells. Treatment of PAC-resistant MCF-7 cells with silibinin at 400 μM concentration generated synergistic effects when it was used in combination with PAC at 250 nM concentration (CI = 0.81). CONCLUSION Silibinin sensitizes chemo-resistant cells to chemotherapeutic agents and can be useful in treating breast cancers.
Collapse
Affiliation(s)
- Ommoleila Molavi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- CONTACT Ommoleila MolaviDepartment of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, 111-Faculty of Pharmacy, Tabriz University, Tabriz, Iran
| | - Farzaneh Narimani
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farshid Asiaee
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Sharifi
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shayanfar
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Raymond Lai
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
36
|
Arafa Keshk W, Zahran SM, Katary MA, Abd-Elaziz Ali D. Modulatory effect of silymarin on nuclear factor-erythroid-2-related factor 2 regulated redox status, nuclear factor-κB mediated inflammation and apoptosis in experimental gastric ulcer. Chem Biol Interact 2017; 273:266-272. [PMID: 28648817 DOI: 10.1016/j.cbi.2017.06.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/07/2017] [Accepted: 06/21/2017] [Indexed: 01/17/2023]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) consumption has been commonly associated with gastric mucosal lesions including gastric ulcer. Silymarin (SM) is a flavonoid mixture with anti-oxidant and anti-inflammatory activities which explain its protective role against hepatic and renal injuries. However, its impact on gastric ulcer has not yet been elucidated. Thus we went further to investigate the potential protective effects of SM against indomethacin-induced gastric injury in rats. Pretreatment with SM (50 mg/kg orally) attenuated the severity of gastric mucosal damage as evidenced by decreasing ulcer index (UI) and ulcer score, improvement of disturbed histopathologicl features to be insignificant with those induced by the reference anti-ulcer drug. Pretreatment with SM also suppressed gastric inflammation by decreasing myeloperoxidase activity, tumer necrosis factor-α (TNF- α) and interleukin 6 (IL6) levels along with nuclear factor kappa B p65 (NF-κB) expression. Meanwhile, SM prevent gastric oxidative stress via inhibition of lipid peroxides formation, enhancement of glutathione peroxidase, superoxide dismutase activities and up-regulation of nuclear factor-erythroid-2-related factor 2 (Nrf2), the redox-sensitive master regulator of oxidative stress signaling. In conclusion, the results herein revealed that SM has a gastro-protective effect which is mediated via suppression of gastric inflammation, oxidative stress, increased the anti-oxidant and the cyto-protective defense mechanisms.
Collapse
Affiliation(s)
- Walaa Arafa Keshk
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Egypt.
| | - Samer Mahmoud Zahran
- Biochemistry Department, Faculty of Pharmacy and Drug Manufacturing, Pharos University, Alexandria, Egypt
| | - Mohamed Alaa Katary
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhur University, Egypt
| | - Darin Abd-Elaziz Ali
- Department of Histopathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
37
|
Reina M, Martínez A. Silybin interacting with Cu 4 , Ag 4 and Au 4 clusters: Do these constitute antioxidant materials? COMPUT THEOR CHEM 2017. [DOI: 10.1016/j.comptc.2017.03.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
38
|
Amawi H, Ashby CR, Tiwari AK. Cancer chemoprevention through dietary flavonoids: what's limiting? CHINESE JOURNAL OF CANCER 2017. [PMID: 28629389 PMCID: PMC5477375 DOI: 10.1186/s40880-017-0217-4] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Flavonoids are polyphenols that are found in numerous edible plant species. Data obtained from preclinical and clinical studies suggest that specific flavonoids are chemo-preventive and cytotoxic against various cancers via a multitude of mechanisms. However, the clinical use of flavonoids is limited due to challenges associated with their effective use, including (1) the isolation and purification of flavonoids from their natural resources; (2) demonstration of the effects of flavonoids in reducing the risk of certain cancer, in tandem with the cost and time needed for epidemiological studies, and (3) numerous pharmacokinetic challenges (e.g., bioavailability, drug–drug interactions, and metabolic instability). Currently, numerous approaches are being used to surmount some of these challenges, thereby increasing the likelihood of flavonoids being used as chemo-preventive drugs in the clinic. In this review, we summarize the most important challenges and efforts that are being made to surmount these challenges.
Collapse
Affiliation(s)
- Haneen Amawi
- Department of Pharmacology and Systems Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43560, USA
| | - Charles R Ashby
- Pharmaceutical Sciences, College of Pharmacy, St. John's University, Queens, NY, 11432, USA
| | - Amit K Tiwari
- Department of Pharmacology and Systems Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43560, USA. .,Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
39
|
|
40
|
Polachi N, Bai G, Li T, Chu Y, Wang X, Li S, Gu N, Wu J, Li W, Zhang Y, Zhou S, Sun H, Liu C. Modulatory effects of silibinin in various cell signaling pathways against liver disorders and cancer – A comprehensive review. Eur J Med Chem 2016; 123:577-595. [DOI: 10.1016/j.ejmech.2016.07.070] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 12/23/2022]
|
41
|
Al-Rasheed N, Faddah L, Al-Rasheed N, Bassiouni YA, Hasan IH, Mahmoud AM, Mohamad RA, Yacoub HI. Protective Effects of Silymarin, Alone or in Combination with Chlorogenic Acid and/or Melatonin, Against Carbon Tetrachloride-induced Hepatotoxicity. Pharmacogn Mag 2016; 12:S337-45. [PMID: 27563222 PMCID: PMC4971954 DOI: 10.4103/0973-1296.185765] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the hepatoprotective effects of silymarin (SIL), alone and combined with chlorogenic acid (CA) and/or melatonin (ME), using a rat model of carbon tetrachloride (CCl4)-induced injury. MATERIALS AND METHODS Hepatotoxicity was induced by a single dose of CCl4 (1 ml/kg, IP). One day after, rats were received SIL (200 mg/kg) alone or in combination with CA (60 mg/kg) and/or ME (20 mg/kg) for 21 days. RESULTS SIL significantly decreased serum alanine aminotransferase, inflammatory cytokines, and vascular endothelial growth factor levels. Histological alterations, fibrogenesis, oxidative DNA damage, inflammatory mediators, and caspase-3 activity were significantly attenuated in SIL treated CCl4-intoxicated rats. On the other hand, cytochrome P450 2E1 activity showed a significant decrease in the liver of CCl4-intoxicated rats, an effect that was reversed following treatment with SIL. All beneficial effects of SIL were markedly potentiated when combined with CA and/or ME. CONCLUSIONS These data indicate that SIL, alone and combined with CA and/or ME, protected the liver against CCl4-induced hepatotoxicity via attenuating inflammation, oxidative DNA damage, apoptosis, and fibrotic changes. The significantly intensified hepatoprotective effects of SIL when combined with both CA and ME suggest a possible synergism. These synergistic effects need to be further confirmed using detailed studies. SUMMARY Silymarin, chlorogenic acid and melatonin possess in vivo hepatoprotective activitySilymarin, chlorogenic acid and melatonin attenuate fibrogenesis, oxidative DNA damage, inflammation and apoptosisChlorogenic acid and melatonin enhance the hepatoprotective effect of silymarin. Abbreviations used: SIL: silymarin, CA: chlorogenic acid, ME: melatonin, CCl4: carbon tetrachloride, CYP2E1, cytochrome P450 2E1, ALT: alanine aminotransferase, IL-6: interleukin 6, IFN-γ: interferon gamma, VEGF: vascular endothelial growth factor, TNF-α: tumor necrosis factor alpha, CRP: C-reactive protein, 8-OxodG: 8-Oxo-2'-deoxyguanosine, TGF-B1: transforming growth factor beta 1, HSCs: hepatic stellate cells.
Collapse
Affiliation(s)
- Nouf Al-Rasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Laila Faddah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nawal Al-Rasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Yieldez A Bassiouni
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Iman H Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ayman M Mahmoud
- Department of Zoology, Division of Physiology, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Raeesa A Mohamad
- Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hazar I Yacoub
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
42
|
Lesson From Comparison of CAM Use by Women With Female-Specific Cancers to Others: It's Time to Focus on Interaction Risks With CAM Therapies. Integr Cancer Ther 2016; 6:313-44. [DOI: 10.1177/1534735407309257] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
There has been no examination as to whether the prevalence of complementary and alternative medicine (CAM) use, as well as personal factors associated with CAM use and predictive of CAM use for women with female-specific cancers, is similar to those in other diagnostic groups. The purpose of this review is to compare CAM use and personal factors associated with and predictive of CAM use by women with female-specific cancers to samples of other diagnostic groups. If it is the case that CAM use is similar across various types of samples, then it may be unnecessary to continue to study detailed CAM use by those in separate diagnostic groups and instead focus energies on the examination of CAM therapies that may have risks for interaction with conventional therapies, such as biologically based therapies. The researcher concludes that we are now in an era in which we need to use our restricted time, human resources, and finances to examine biologically based CAM use that may carry high risks for interactions or toxicities for specific groups under examination, rather than examine global CAM use, unless the situation warrants such all-inclusive study.
Collapse
|
43
|
Šťastník O, Jůzl M, Karásek F, Štenclová H, Nedomová Š, Pavlata L, Mrkvicová E, Doležal P, Jarošová A. The effect of feeding milk thistle seed cakes on quality indicators of broiler chickens meat. POTRAVINARSTVO 2016. [DOI: 10.5219/579] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
This study was conducted to evaluate the effect of feeding milk thistle (Silybum marianum L.) seed cakes at dose 5% and 15% in feed mixture on quality indicators of broiler chickens meat. The used milk thistle seed cakes contained 3.73% of flavonolignans and 129.83 mg.kg-1 of cyanidin-3-glucoside. A 150 cockerels of Ross 308 were divided into three equal groups. The chickens were fattened on conventional deep litter system. The experimental groups received feed mixtures containing 5% of milk thistle seed cakes (MT5), 15% of milk thistle seed cakes (MT15) and third group was control - without milk thistle seed cakes (C). The trial lasted 37 days. At the end of trial was observed significant higher average weight of chickens (2,320.31 g) in control group. Compare to that the experimental group MT5 achieved significant lower mean bodyweight 2,166.69 g. From the perspective of fattening was decreased growth of chickens where a higher percentage of milk thistle seed cakes (MT15). The group MT15 was up to 420 g lower slaughter weight compared to the control group. This was probably due to the higher content of fiber in the feed. At the end of experiment 15 birds were selected randomly from each group, weighed and slaughtered. Feathers were removed and chickens were eviscerated. Carcass yield was calculated for each group like as percentage of live weight. The MT5 and MT15 group had significantly higher breast meat tenderness that the control group. Initial pH1 was highest in group with its middle addition of milk thistle seed cakes (MT5). Significant differences were not observed between control and group MT15. Breast meat was rated as the best in parameter flavour in control and MT15 group. The thigh meat was evaluated significantly best for colour parameter in MT15 group. Fibreness was rated as the finest in MT15 group. The addition of milk thistle seed cakes do not worsened sensory characteristic of breast or thigh meat of broilers and reflects optimal sensory quality traits.
Collapse
|
44
|
Potential Effects of Silymarin and Its Flavonolignan Components in Patients with β-Thalassemia Major: A Comprehensive Review in 2015. Adv Pharmacol Sci 2016; 2016:3046373. [PMID: 26997953 PMCID: PMC4779508 DOI: 10.1155/2016/3046373] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/21/2016] [Accepted: 01/27/2016] [Indexed: 02/08/2023] Open
Abstract
Major β-thalassemia (β-TM) is one of the most common inherited hemolytic types of anemia which is caused as a result of absent or reduced synthesis of β-globin chains of hemoglobin. This defect results in red blood cells lysis and chronic anemia that can be treated by multiple blood transfusions and iron chelation therapy. Without iron chelation therapy, iron overload will cause lots of complications in patients. Antioxidant components play an important role in the treatment of the disease. Silymarin is an antioxidant flavonoid isolated from Silybum marianum plant. In the present study, we reviewed clinical and experimental studies investigating the use of silymarin prior to September 1, 2015, using PubMed, ISI Web of Knowledge, Science Direct, Scopus, Ovid, and Cochrane Library databases and we evaluated the potential effects of silymarin on controlling the complications induced by iron overload in patients with β-TM. Based on the results of the present study, we can conclude that silymarin may be useful as an adjuvant for improving multiple organ dysfunctions.
Collapse
|
45
|
Amniattalab A, Malekinejad H, Rezabakhsh A, Rokhsartalab-Azar S, Alizade-Fanalou S. Silymarin: A Novel Natural Agent to Restore Defective Pancreatic β Cells in Streptozotocin (STZ)-induced Diabetic Rats. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2016; 15:493-500. [PMID: 27980584 PMCID: PMC5149036 DOI: pmid/27980584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This study aimed to investigate the potency of silymarin (SMN) and melatonin (MEL) on restoring the pancreatic cells in streptozotocin (STZ)-induced diabetic rats. Male Wistar rats were divided into five groups, including: control (C), untreated diabetic (D), SMN-treated diabetic (50 mg/Kg, orally), MEL-treated diabetic (10 mg/Kg, i.p.), and SMN plus MEL-treated diabetic rats. Diabetes was induced by injection of STZ (50 mg/Kg, i.p.). The blood glucose and insulin levels were measured. After the 28 days treatment period, antioxidant status was analyzed by determination of total antioxidant capacity (TAC) in the liver and serum. The histopathological changes in the pancreatic islets were examined by histochemical staining and enumeration of cells. Although none of the test compounds reduced the blood glucose level to normal concentration, however SMN alone and in combination with MEL was able to decline it significantly (P<0.05) after 28 days administration. Both SMN and MEL could recover the diabetes-reduced TAC values. Moreover, the diabetes-induced cellular vacuolation and cells depletion were improved by the SMN treatment. Our data suggest that the SMN and MEL treatment was able to normalize the antioxidant status, while only SMN administration could restore the cells of Langerhans islets in diabetic rats.
Collapse
Affiliation(s)
- Amir Amniattalab
- Department of Pathology, Islamic Azad University, Urmia Branch, Urmia, Iran.
| | - Hassan Malekinejad
- Department of pharmacology and toxicology, Faculty of Veterinary medicine, Urmia University, Iran.
- Department of pharmacology and toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iarn.
- Corresponding author: E-mail:
| | - Aysa Rezabakhsh
- Department of Pharmacology and toxicology, Faculty of Pharmacy, University of Medical sciences, Tabriz, Iran.
| | - Shirin Rokhsartalab-Azar
- Department of pharmacology and toxicology, Faculty of Veterinary medicine, Urmia University, Iran.
| | - Shahin Alizade-Fanalou
- Department of pharmacology and toxicology, Faculty of Veterinary medicine, Urmia University, Iran.
| |
Collapse
|
46
|
Abstract
This review is to describe synergistic effects of various combinations of dietary natural products including curcumin, quercetin, soybean isoflavones, silibinin, and EGCG that have potential for the treatment of prostate cancer. These data can provide valuable insights into the future rational design and development of synergistic and/or hybrid agents for potential treatment of prostate cancer.
Collapse
|
47
|
Gabrielová E, Zholobenko AV, Bartošíková L, Nečas J, Modriansky M. Silymarin Constituent 2,3-Dehydrosilybin Triggers Reserpine-Sensitive Positive Inotropic Effect in Perfused Rat Heart. PLoS One 2015; 10:e0139208. [PMID: 26418338 PMCID: PMC4856230 DOI: 10.1371/journal.pone.0139208] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/10/2015] [Indexed: 02/02/2023] Open
Abstract
2,3-dehydrosilybin (DHS) is a minor flavonolignan component of Silybum marianum seed extract known for its hepatoprotective activity. Recently we identified DHS as a potentially cardioprotective substance during hypoxia/reoxygenation in isolated neonatal rat cardiomyocytes. This is the first report of positive inotropic effect of DHS on perfused adult rat heart. When applied to perfused adult rat heart, DHS caused a dose-dependent inotropic effect resembling that of catecholamines. The effect was apparent with DHS concentration as low as 10 nM. Suspecting direct interaction with β-adrenergic receptors, we tested whether DHS can trigger β agonist-dependent gene transcription in a model cell line. While DHS alone was unable to trigger β agonist-dependent gene transcription, it enhanced the effect of isoproterenol, a known unspecific β agonist. Further tests confirmed that DHS could not induce cAMP accumulation in isolated neonatal rat cardiomyocytes even though high concentrations (≥ 10 μM) of DHS were capable of decreasing phosphodiesterase activity. Pre-treatment of rats with reserpine, an indole alkaloid which depletes catecholamines from peripheral sympathetic nerve endings, abolished the DHS inotropic effect in perfused hearts. Our data suggest that DHS causes the inotropic effect without acting as a β agonist. Hence we identify DHS as a novel inotropic agent.
Collapse
Affiliation(s)
- Eva Gabrielová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Aleksey Vladimirovich Zholobenko
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Lenka Bartošíková
- Department of Physiology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Jiří Nečas
- Department of Physiology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Martin Modriansky
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| |
Collapse
|
48
|
Gillessen A, Herrmann WA, Kemper M, Morath H, Mann K. [Effect of silymarin on liver health and quality of life. Results of a non-interventional study]. MMW Fortschr Med 2015; 156 Suppl 4:120-6. [PMID: 26153591 DOI: 10.1007/s15006-014-3758-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Many drugs are known to have hepatotoxic side effects. The effect of silymarin on liver function and liver-injury-impaired quality of life under daily practice conditions in patients with elevated values of liver enzymes was evaluated in the present non-interventional study. METHOD Patients with drug-induced elevated aminotransferase levels and indication for silymarin (Legalon forte) treatment for 2 to 3 months were documented prospectively over 4 months. At baseline, after 2 and 4 months, respectively, the following parameters were documented: alanine transaminase (ALT), aspartate transaminase (AST), γ-glutamyltransferase (GGT), alkaline phosphatase, total bilirubin, presence of liver-related skin symptoms and discoloured urine, severity of liver-related symptoms and quality of life. RESULTS In total, 190 patients (53.2% male, median age 60.0 years [range 19-81]) from 48 centres participated in the non-interventional study. Among potentially hepatotoxic drugs, analgesics/anti-inflammatory drugs were used most frequently (45.8%). These drugs have been administered for a median period of 2.8 years (range 0.0-26.1). At baseline, all patients had elevated levels of ALT, AST or GGT. Fatigue, flatulence, upper abdominal discomfort, lethargy, and joint complaints were the most severe liver-related symptoms and prevalent in over 62% of patients. Quality of life was affected in 88.7% of patients. Significant reductions were achieved in all documented laboratory parameters (p < 0.001), leading to marked improvement in liver-related symptoms and increased quality of life already after 2 months. The percentage of patients with liver enzymes in the normal range increased considerably within 4 months. No adverse drug reactions were observed. CONCLUSIONS Silymarin is a safe and efficacious treatment option for patients with elevated liver enzymes. A benefit in terms of liver-related symptoms as well as quality of life and performance was demonstrated already after 2 months of treatment.
Collapse
|
49
|
The protective effect of Capparis ovata on 6-mercaptopurine-induced hepatotoxicity and oxidative stress in rats. J Pediatr Hematol Oncol 2015; 37:290-4. [PMID: 25411867 DOI: 10.1097/mph.0000000000000288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Capparis ovata is a member of Capparidacaeae family has been used in phytomedicine with a lot of positive effects such as an antioxidative, antihyperlipidemic, anti-inflammatory, and antihepatotoxic agent. The aim of this study was to research the protective effect of C. ovata on 6-mercaptopurine (6-MP) induced to hepatotoxicity and oxidative stress in rats. The rats were divided into 4 groups: control, 6-MP, C. ovataovate, and 6-MP + C. ovata. A complete blood count was performed, liver function test and antioxidant enzymes levels such as superoxide dismutase, glutathione peroxidase, catalase, and malondialdehyde were measured in blood before and after a 14-day test period. White blood cell and platelet counts were lower in the 6-MP group than other 3 groups (P < 0.005). Hepatic transaminase levels were higher in 6-MP group than the 3 groups (P < 0.05). Superoxide dismutase, glutathione peroxidase, and CAT levels were lower and malondialdehyde was higher in blood samples in 6-MP group than other 3 groups (P < 0.005). In conclusion, our tests were showed that C. ovata may be useful in patients receiving 6-MP therapy to prevent hepatotoxicity and in order to maintain uninterrupted therapy possibly reducing the risk of relapse. Although additional studies ensure that Capparis does not affect 6-MP antileukemic activity. We believe these results are important contribution to the literature.
Collapse
|
50
|
Silymarin as a Natural Antioxidant: An Overview of the Current Evidence and Perspectives. Antioxidants (Basel) 2015; 4:204-47. [PMID: 26785346 PMCID: PMC4665566 DOI: 10.3390/antiox4010204] [Citation(s) in RCA: 362] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 02/06/2015] [Accepted: 03/09/2015] [Indexed: 12/16/2022] Open
Abstract
Silymarin (SM), an extract from the Silybum marianum (milk thistle) plant containing various flavonolignans (with silybin being the major one), has received a tremendous amount of attention over the last decade as a herbal remedy for liver treatment. In many cases, the antioxidant properties of SM are considered to be responsible for its protective actions. Possible antioxidant mechanisms of SM are evaluated in this review. (1) Direct scavenging free radicals and chelating free Fe and Cu are mainly effective in the gut. (2) Preventing free radical formation by inhibiting specific ROS-producing enzymes, or improving an integrity of mitochondria in stress conditions, are of great importance. (3) Maintaining an optimal redox balance in the cell by activating a range of antioxidant enzymes and non-enzymatic antioxidants, mainly via Nrf2 activation is probably the main driving force of antioxidant (AO) action of SM. (4) Decreasing inflammatory responses by inhibiting NF-κB pathways is an emerging mechanism of SM protective effects in liver toxicity and various liver diseases. (5) Activating vitagenes, responsible for synthesis of protective molecules, including heat shock proteins (HSPs), thioredoxin and sirtuins and providing additional protection in stress conditions deserves more attention. (6) Affecting the microenvironment of the gut, including SM-bacteria interactions, awaits future investigations. (7) In animal nutrition and disease prevention strategy, SM alone, or in combination with other hepatho-active compounds (carnitine, betaine, vitamin B12, etc.), might have similar hepatoprotective effects as described in human nutrition.
Collapse
|