1
|
Al-Haideri M. Silymarin suppresses proliferation and PD-L1 expression in colorectal cancer cells and increases inflammatory CD8+ cells in tumor-bearing mice. Clin Res Hepatol Gastroenterol 2024; 48:102425. [PMID: 39048076 DOI: 10.1016/j.clinre.2024.102425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/15/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Silymarin as an herbal medicine has shown anticancer effects on tumor cells, while having low toxicity in normal cells. In this study, the effects of Silymarin on proliferation and apoptosis of colorectal cancer cells and its impact on immune response against cancer cells were evaluated in vitro and in vivo. METHODS AND MATERIALS The effect of Silymarin on CT-26 and Caco-2 cells proliferation and apoptosis were demonstrated by MTT assay and PI staining. A subcutaneous tumor of colorectal cancer was developed. Silymarin and Doxorubicin were administrated by intravenous injection. qRT-PCR analyses was performed on blood samples and tumor tissues. Spleen tissue was used to evaluate CD8+ T cell immune responses. Histological study was carried out on tumor tissues. RESULTS Silymarin showed anti-proliferative effects on CT-26 and Caco-2 cells. The markers of immunogenic cell death (Calreticulin exposure, ATP secretion, and HMGB1 secretion) significantly increased in both cell lines in the presence of silymarin. The expression of genes related to cell proliferation particularly β-Catenin and Cycline D1, and also anti-apoptotic ones such as Bcl-2 significantly reduced in mice treated with Silymarin while the expression of pro-apoptotic Bax increased. The RNA level of PD-L1 decreased in tumor tissues exposed by Silymarin. Moreover, the number of CTLs increased in the spleen of mice treated with Silymarin in comparison with untreated mice. Decreased tumor size and also survival of colorectal cancer cells in Silymarin-treated mice were observed in histological analysis. CONCLUSION Silymarin treatment showed a suppressive role on colorectal cancer cells almost as much as Doxorubicin. Our study indicated that having a low toxicity profile, cost-effectiveness, and availability of raw materials, plant-derived Silymarin can be a good candidate for further investigation to treat CRC.
Collapse
Affiliation(s)
- Maysoon Al-Haideri
- School of medicine, Pharmacy Department, University of Kurdistan Hawlêr, Erbil, Kurdistan, Iraq.
| |
Collapse
|
2
|
Ali W, Chen Y, Gandahi JA, Qazi IH, Sun J, Wang T, Liu Z, Zou H. Cross-Talk Between Selenium Nanoparticles and Cancer Treatment Through Autophagy. Biol Trace Elem Res 2024; 202:2931-2940. [PMID: 37817045 DOI: 10.1007/s12011-023-03886-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023]
Abstract
Autophagy is commonly referred as self-eating and a complex cellular process that is involved in the digestion of protein and damaged organelles through a lysosome-dependent mechanism, and this mechanism is essential for maintaining proper cellular homeostasis. Selenium is a vital trace element that plays essential functions in antioxidant defense, redox state control, and range of particular metabolic processes. Selenium nanoparticles have become known as a promising agent for biomedical use, because of their high bioavailability, low toxicity, and degradability. However, and in recent years, they have attracted the interest of researchers in developing anticancer nano-drugs. Selenium nanoparticles can be used as a potential therapeutic agent or in combination with other agents to act as carriers for the development of new treatments. More intriguingly, selenium nanoparticles have been extensively shown to impact autophagy signaling, allowing selenium nanoparticles to be used as possible cancer treatment agents. This review explored the connections between selenium and autophagy, followed by developments and current advances of selenium nanoparticles for autophagy control in various clinical circumstances. Furthermore, this study examined the functions and possible processes of selenium nanoparticles in autophagy regulation, which may help us understand how selenium nanoparticles regulate autophagy for the potential cancer treatment.
Collapse
Affiliation(s)
- Waseem Ali
- College of Veterinary Medicine, Yangzhou University Yangzhou, Yangzhou, Jiangsu, 225009, People's Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Yan Chen
- College of Veterinary Medicine, Yangzhou University Yangzhou, Yangzhou, Jiangsu, 225009, People's Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Jameel Ahmed Gandahi
- College of Veterinary Medicine, Yangzhou University Yangzhou, Yangzhou, Jiangsu, 225009, People's Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Izhar Hyder Qazi
- College of Veterinary Medicine, Yangzhou University Yangzhou, Yangzhou, Jiangsu, 225009, People's Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Jian Sun
- College of Veterinary Medicine, Yangzhou University Yangzhou, Yangzhou, Jiangsu, 225009, People's Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University Yangzhou, Yangzhou, Jiangsu, 225009, People's Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University Yangzhou, Yangzhou, Jiangsu, 225009, People's Republic of China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China.
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University Yangzhou, Yangzhou, Jiangsu, 225009, People's Republic of China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China.
| |
Collapse
|
3
|
Zarei Shandiz S, Erfani B, Hashemy SI. Protective effects of silymarin in glioblastoma cancer cells through redox system regulation. Mol Biol Rep 2024; 51:723. [PMID: 38833199 DOI: 10.1007/s11033-024-09658-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Glioblastoma multiforme, a deadly form of brain tumor, is characterized by aggressive growth and poor prognosis. Oxidative stress, a disruption in the balance between antioxidants and oxidants, is a crucial factor in its pathogenesis. Silymarin, a flavonoid extracted from milk thistle, has shown therapeutic potential in inhibiting cancer cell growth, promoting apoptosis, and reducing inflammation. It also regulates oxidative stress. This study aims to investigate the regulatory effects of silymarin on oxidative stress parameters, especially the transcription factor Nrf2 and its related enzymes in GBM cancer cells, to develop a new anti-cancer compound with low toxicity. METHODS AND RESULTS First, the cytotoxicity of silymarin on U-87 MG cells was investigated by MTT and the results showed an IC50 of 264.6 μM. Then, some parameters of the redox system were measured with commercial kits, and the obtained results showed that silymarin increased the activity of catalase and superoxide dismutase enzymes, as well as the total antioxidant capacity levels; while the malondialdehyde level that is an indicator of lipid peroxidation was decreased by this compound. The expression level of Nrf2 and HO-1 and glutaredoxin and thioredoxin enzymes were checked by real-time PCR method, and the expression level increased significantly after treatment. CONCLUSIONS Our findings suggest that silymarin may exert its cytotoxic and anticancer effects by enhancing the Nrf2/HO-1 pathway through antioxidant mechanisms in U-87 MG cells.
Collapse
Affiliation(s)
- Sara Zarei Shandiz
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Bahareh Erfani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Hamed RA, Talib WH. Targeting cisplatin resistance in breast cancer using a combination of Thymoquinone and Silymarin: an in vitro and in vivo study. PHARMACIA 2024; 71:1-19. [DOI: 10.3897/pharmacia.71.e117997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Background: Breast cancer (BC) is considered the most diagnosed cancer among women globally. This is because of its high possibility of metastasis and high resistance to chemotherapy. Cisplatin is a platinum-based antitumor agent that is used to treat various types of cancer. However, the main obstacle to using this drug is drug resistance. Drug resistance is a cause of most relapses of cancer which eventually lead to death. Nowadays, combining natural products is a trend to overcome drug resistance. Thymoquinone (TQ) is a natural phytochemical that exists mainly in blackseed. It has been used in medicine for decades, especially as an anticancer agent. Silymarin is a milk thistle compound that exhibits anticancer, hepatoprotective, and neuroprotective activity. Hence, the combination of TQ and silymarin could be a probable solution to treat cancer and reduce chemoresistance.
Methods: This study tested this combination on cisplatin-sensitive (EMT6/P) and cisplatin-resistant (EMT6/CPR) mouse mammary cell lines. Apoptotic and antiproliferative activity was assessed for TQ and silymarin in vitro using caspase-3 and [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide] (MTT) assays, respectively. An in vivo study was performed to evaluate the effect of TQ and silymarin combination in mice inoculated with EMT6/P and EMT6/CPR cells. The safety profile was also examined using creatinine and liver enzyme assays.
Results: In vitro, the TQ and silymarin combination synergized in both cell lines. Also, this combination caused apoptosis induction at a higher rate than the single treatment in both cell lines. In vivo, TQ and silymarin combination resulted in a remarkable reduction in tumor size and enhanced the cure rate in mice implanted with EMT6/P and EMT6/CPR cell lines. According to the safety profile results, TQ and silymarin combination was safe.
Conclusion: In conclusion, the combination of TQ and silymarin provides a promising solution in treating BC resistant to cisplatin by inducing apoptosis. Further studies are needed to define the exact anticancer mechanisms of this combination.
Collapse
|
5
|
Zhou YM, Dong XR, Xu D, Tang J, Cui YL. Therapeutic potential of traditional Chinese medicine for interstitial lung disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116952. [PMID: 37487964 DOI: 10.1016/j.jep.2023.116952] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/26/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Interstitial lung disease (ILD) is a chronic lung dysfunction disease with a poor prognosis and poor recovery. The clinically used therapeutic drugs, such as glucocorticoids and immunosuppressants, have no significant therapeutic effect and are accompanied with severe side effects. In recent years, considerable progress has been made in exploring and applying natural herb components for treating ILD. Traditional Chinese Medicine (TCM) possesses innate, non-toxic characteristics and offers advantages in preventing and treating pulmonary ailments. However, a comprehensive study of TCM on ILD therapy has not yet been reviewed. AIM OF THE REVIEW This review aimed to provide a comprehensive summary of the monomer components, total extracts, and prescriptions of TCM for ILD therapy, elucidating their molecular mechanisms to serve as a reference in treating ILD. MATERIALS AND METHODS The literature information was searched in the PubMed, Web of Science databases. The search keywords included 'interstitial lung disease', 'lung fibrosis' or 'pulmonary fibrosis', and 'traditional Chinese medicine', 'traditional herbal medicine', or 'herb medicine'. RESULTS The active components of single herbs, such as alkaloids, flavonoids, terpenoids, phenols, and quinones, have potential therapeutic effects on ILD. The active extracts and prescriptions were also summarized and analyzed. The herbs, Glycyrrhiza uralensis Fisch. (Gancao), Astragalus membranaceus Fisch. Bunge. (Huangqi) and Angelicasinensis (Oliv.) Diels (Danggui), play significant roles in the treatment of ILD. The mechanisms involve the inhibition of inflammatory factor release, anti-oxidative injury, and interference with collagen production, etc. CONCLUSION: This review examines the therapeutic potential of TCM for ILD and elucidates its molecular mechanisms, demonstrating that mitigating inflammation and oxidative stress, modulating the immune system, and promoting tissue repair are efficacious strategies for ILD therapy. The depth research will yield both theoretical and practical implications.
Collapse
Affiliation(s)
- Yan-Ming Zhou
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Xin-Ran Dong
- The Second Hospital of Tianjin Medical University, Tianjin, 300211, PR China
| | - Dong Xu
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China.
| | - Jie Tang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
6
|
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
|
7
|
Pferschy-Wenzig EM, Kunert O, Thumann T, Moissl-Eichinger C, Bauer R. Characterization of metabolites from milk thistle flavonolignans generated by human fecal microbiota. PHYTOCHEMISTRY 2023; 215:113834. [PMID: 37648045 DOI: 10.1016/j.phytochem.2023.113834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 09/01/2023]
Abstract
Silymarin, a mixture of diastereomeric and regioisomeric flavonolignans from milk thistle (Silybum marianum (L.) Gaertn.) fruits, is known to possess a panel of pharmacological activities. However, due to low water solubility and extensive phase II metabolism, the oral bioavailability of the flavonolignans is limited. Since their interaction with gut microbiome is likely due to their predominantly fecal excretion route, the biotransformation of milk thistle flavonolignans by gut microorganisms was studied. A 1:1 mixture of the two main silymarin flavonolignans silybins A and B was incubated in human fecal suspension from one donor for 24 h under anoxic conditions. Purification of the incubate allowed to isolate and structurally elucidate the two main metabolites as (2R, 3R)-2-{4-[2-(3,4-dihydroxy-phenyl)-(1R)-1-hydroxymethyl-ethoxy]-3-hydroxy-phenyl}-3,5,7-trihydroxy-chroman-4-one (a product of demethylation and dioxane ring cleavage) and demethylsilybin B. Furthermore, silymarin was incubated with human fecal suspension, and its biotransformation was monitored by means of LC-HRMS metabolite profiling. Apart from the two isolated and structurally elucidated metabolites, several types of biotransformation products could be annotated, including demethylation products, reduction/ring cleavage products, products of demethylation plus reduction/ring cleavage, as well as several low molecular weight aromatic metabolites. The potential pharmacological activities of these gut microbial metabolites deserve closer examination in the future.
Collapse
Affiliation(s)
- Eva-Maria Pferschy-Wenzig
- University of Graz, Institute of Pharmaceutical Sciences, Beethovenstraße 8, 8010, Graz, Austria; BioTechMed- Graz, Mozartgasse 12/II, 8010, Graz, Austria.
| | - Olaf Kunert
- University of Graz, Institute of Pharmaceutical Sciences, Beethovenstraße 8, 8010, Graz, Austria.
| | - Timo Thumann
- University of Graz, Institute of Pharmaceutical Sciences, Beethovenstraße 8, 8010, Graz, Austria.
| | - Christine Moissl-Eichinger
- Medical University Graz, Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Neue Stiftingtalstraße 6 (MC1.B.)/III, 8010, Graz, Austria; BioTechMed- Graz, Mozartgasse 12/II, 8010, Graz, Austria.
| | - Rudolf Bauer
- University of Graz, Institute of Pharmaceutical Sciences, Beethovenstraße 8, 8010, Graz, Austria; BioTechMed- Graz, Mozartgasse 12/II, 8010, Graz, Austria.
| |
Collapse
|
8
|
Rahnama S, Tehrankhah ZM, Mohajerani F, Mohammadi FS, Yeganeh ZY, Najafi F, Babashah S, Sadeghizadeh M. Milk thistle nano-micelle formulation promotes cell cycle arrest and apoptosis in hepatocellular carcinoma cells through modulating miR-155-3p /SOCS2 /PHLDA1 signaling axis. BMC Complement Med Ther 2023; 23:337. [PMID: 37749575 PMCID: PMC10521506 DOI: 10.1186/s12906-023-04168-5] [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: 06/19/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) is a prevalent form of liver cancer that causes significant mortality in numerous individuals worldwide. This study compared the effects of milk thistle (MT) and nano-milk thistle (N-MT) on the expression of the genes that participate in apoptosis and cell cycle pathways in Huh-7 and HepG2 cells. METHODS IC50 values of MT and N-MT were determined using the MTT assay. Huh-7 and HepG2 cell lines (containing mutant and wild-type TP53 gene, respectively) were incubated with MT and N-MT for 24h and 48h and the impact of MT and N-MT on the proliferation of these cell lines was evaluated through a comparative analysis. Cell cycle and apoptosis were assessed by flow cytometry after 24h and 48h treatment in the cell lines mentioned. Real-time PCR was used to analyze miR-155-3p, PHLDA1, SOCS2, TP53, P21, BAX, and BCL-2 expression in the cell lines that were being treated. RESULTS N-MT reduces cancer cell growth in a time and concentration-dependent manner, which is more toxic compared to MT. Huh-7 was observed to have IC50 values of 2.35 and 1.7 μg/ml at 24h and 48h, and HepG2 was observed to have IC50 values of 3.4 and 2.6 μg/ml at 24 and 48h, respectively. N-MT arrested Huh-7 and HepG2 cells in the Sub-G1 phase and induced apoptosis. N-MT led to a marked reduction in the expression of miR-155-3p and BCL-2 after 24h and 48h treatments. Conversely, PHLDA1, SOCS2, BAX, and P21 were upregulated in the treated cells compared to untreated cells, which suggests that milk thistle has the potential to regulate these genes. N-MT reduced the expression of TP53 in Huh-7 cells after mentioned time points, while there was a significant increase in the expression of the TP53 gene in HepG2 cells. No gene expression changes were observed in MT-treated cells after 24h and 48h. CONCLUSION N-MT can regulate cancer cell death by arresting cell cycle and inducing apoptosis. This occurs through the alteration of apoptotic genes expression. A reduction in the expression of miR-155-3p and increase in the expression of SOCS2 and PHLDA1 after N-MT treatment showed the correlation between miR-155-3p and PHLDA1/SOCS2 found in bioinformatics analysis. While N-MT increased TP53 expression in HepG2, reduced it in Huh-7. The findings indicate that N-MT can function intelligently in cancer cells and can be a helpful complement to cancer treatment.
Collapse
Affiliation(s)
- Saghar Rahnama
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Moazezi Tehrankhah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Mohajerani
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Faezeh Shah Mohammadi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Yousefi Yeganeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farhood Najafi
- Department of Resin and Additives, Institute for Color Science and Technology, Tehran, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
9
|
Li W, Xu X. Advances in mitophagy and mitochondrial apoptosis pathway-related drugs in glioblastoma treatment. Front Pharmacol 2023; 14:1211719. [PMID: 37456742 PMCID: PMC10347406 DOI: 10.3389/fphar.2023.1211719] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumor of the central nervous system (CNS). It is a leading cause of death among patients with intracranial malignant tumors. GBM exhibits intra- and inter-tumor heterogeneity, leading to drug resistance and eventual tumor recurrence. Conventional treatments for GBM include maximum surgical resection of glioma tissue, temozolomide administration, and radiotherapy, but these methods do not effectively halt cancer progression. Therefore, development of novel methods for the treatment of GBM and identification of new therapeutic targets are urgently required. In recent years, studies have shown that drugs related to mitophagy and mitochondrial apoptosis pathways can promote the death of glioblastoma cells by inducing mitochondrial damage, impairing adenosine triphosphate (ATP) synthesis, and depleting large amounts of ATP. Some studies have also shown that modern nano-drug delivery technology targeting mitochondria can achieve better drug release and deeper tissue penetration, suggesting that mitochondria could be a new target for intervention and therapy. The combination of drugs targeting mitochondrial apoptosis and autophagy pathways with nanotechnology is a promising novel approach for treating GBM.This article reviews the current status of drug therapy for GBM, drugs targeting mitophagy and mitochondrial apoptosis pathways, the potential of mitochondria as a new target for GBM treatment, the latest developments pertaining to GBM treatment, and promising directions for future research.
Collapse
|
10
|
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
|
11
|
Chen D, Lu H, Ma Y, Huang Y, Zhang T, Fan S, Lin W, Huang Y, Jin H, Ruan Y, Xu JF, Pi J. Trends and recent progresses of selenium nanoparticles as novel autophagy regulators for therapeutic development. Front Nutr 2023; 10:1116051. [PMID: 36819694 PMCID: PMC9931911 DOI: 10.3389/fnut.2023.1116051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Autophagy, one of the major intracellular degradation systems, plays an important role in maintaining normal cellular physiological functions and protecting organisms from different diseases. Selenium (Se), an essential trace element, is involved in many metabolic regulatory signaling events and plays a key role in human health. In recent years, selenium nanoparticles (Se NPs) have attracted increasing attentions in biomedical field due to their low toxicity, high bioavailability and high bioactivity. Taking the advantage of their advanced biological activities, Se NPs can be used alone as potential therapeutic agents, or combine with other agents and served as carriers for the development of novel therapeutics. More interestingly, Se NPs have been widely reported to affect autophagy signaling, which therefor allow Se NPs to be used as potential therapeutic agents against different diseases. Here, this review suggested the relationships between Se and autophagy, followed by the trends and recent progresses of Se NPs for autophagy regulation in different diseased conditions. More importantly, this work discussed the roles and potential mechanisms of Se NPs in autophagy regulating, which might enhance our understanding about how Se NPs regulate autophagy for potential disease treatment. This work is expected to promote the potential application of Se NPs as novel autophagy regulators, which might benefit the development of novel autophagy associated therapeutics.
Collapse
Affiliation(s)
- Dongsheng Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hongmei Lu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yuhe Ma
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yuhe Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Tangxin Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Shuhao Fan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Wensen Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yifan Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yongdui Ruan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,*Correspondence: Yongdui Ruan,
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China,Jun-Fa Xu,
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China,Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China,Jiang Pi,
| |
Collapse
|
12
|
Wang Y, Yuan AJ, Wu YJ, Wu LM, Zhang L. Silymarin in cancer therapy: Mechanisms of action, protective roles in chemotherapy-induced toxicity, and nanoformulations. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
13
|
Mechanistic Insights into the Pharmacological Significance of Silymarin. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165327. [PMID: 36014565 PMCID: PMC9414257 DOI: 10.3390/molecules27165327] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 12/29/2022]
Abstract
Medicinal plants are considered the reservoir of diverse therapeutic agents and have been traditionally employed worldwide to heal various ailments for several decades. Silymarin is a plant-derived mixture of polyphenolic flavonoids originating from the fruits and akenes of Silybum marianum and contains three flavonolignans, silibinins (silybins), silychristin and silydianin, along with taxifolin. Silybins are the major constituents in silymarin with almost 70–80% abundance and are accountable for most of the observed therapeutic activity. Silymarin has also been acknowledged from the ancient period and is utilized in European and Asian systems of traditional medicine for treating various liver disorders. The contemporary literature reveals that silymarin is employed significantly as a neuroprotective, hepatoprotective, cardioprotective, antioxidant, anti-cancer, anti-diabetic, anti-viral, anti-hypertensive, immunomodulator, anti-inflammatory, photoprotective and detoxification agent by targeting various cellular and molecular pathways, including MAPK, mTOR, β-catenin and Akt, different receptors and growth factors, as well as inhibiting numerous enzymes and the gene expression of several apoptotic proteins and inflammatory cytokines. Therefore, the current review aims to recapitulate and update the existing knowledge regarding the pharmacological potential of silymarin as evidenced by vast cellular, animal, and clinical studies, with a particular emphasis on its mechanisms of action.
Collapse
|
14
|
Abderrezag N, Montenegro ZJS, Louaer O, Meniai AH, Cifuentes A, Ibáñez E, Mendiola JA. One-step sustainable extraction of Silymarin compounds of wild Algerian milk thistle (Silybum marianum) seeds using Gas Expanded Liquids. J Chromatogr A 2022; 1675:463147. [PMID: 35640448 DOI: 10.1016/j.chroma.2022.463147] [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: 03/09/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/25/2022]
Abstract
This work reports the application of Gas Expanded Liquid (GXL) extraction to concentrate the flavonolignan fraction (silymarin) and taxifolin from Silybum marianum seeds, which have proven to be highly valuable health-promoting compounds. GXL using green solvents was used to isolate silymarin with the objective of replacing conventional methods. In one hand, the effect of different compositions of solvents, aqueous ethanol (20%, 50% or 80% (v/v)) at different CO2/liquid (25, 50 and 75%) ratios, on the GXL extraction was investigated. The obtained extracts have been chemically and functionally characterized by means of UHPLC-ESI-MS/MS (triple quadrupole) and in-vitro assays such as anti-inflammatory, anti-cholinergic and antioxidant. Results revealed that the operating conditions influenced the extraction yield, the total phenolic content and the presence of the target compounds. The best obtained yield was 55.97% using a ternary mixture of solvents composed of CO2:EtOH:H2O (25:60:15) at 40 °C and 9 MPa in 160 min. Furthermore, the results showed that obtained extracts had significant antioxidant and anti-inflammatory activities (with best IC50 value of 8.80 µg/mL and 28.52 µg/mL, respectively) but a moderate anti-cholinesterase activity (with best IC50 value of 125.09 µg/mL). Otherwise, the concentration of silymarin compounds in extract can go up to 59.6% using the present one-step extraction method without further purification, being silybinA+B the predominant identified compound, achieving value of 545.73 (mg silymarin/g of extract). The obtained results demonstrate the exceptional potential of GXL to extract high-added values molecules under sustainable conditions from different matrices.
Collapse
Affiliation(s)
- Norelhouda Abderrezag
- Laboratory of Environmental Processes Engineering, University of Salah Boubnider Constantine 3, Ali Mendjli, 25000 Constantine, Algeria; Profesora Facultad de Ingeniería Agroindustrial, Universidad de Nariño (UdeNar), Pasto, Colombia
| | | | - Ouahida Louaer
- Laboratory of Environmental Processes Engineering, University of Salah Boubnider Constantine 3, Ali Mendjli, 25000 Constantine, Algeria
| | - Abdeslam-Hassen Meniai
- Laboratory of Environmental Processes Engineering, University of Salah Boubnider Constantine 3, Ali Mendjli, 25000 Constantine, Algeria
| | - Alejandro Cifuentes
- Foodomics Laboratory, Bioactivity and Food Analysis Department, Institute of Food Science Research CIAL (CSIC-UAM), Nicolas Cabrera 9, 28049 Madrid, Spain
| | - Elena Ibáñez
- Foodomics Laboratory, Bioactivity and Food Analysis Department, Institute of Food Science Research CIAL (CSIC-UAM), Nicolas Cabrera 9, 28049 Madrid, Spain
| | - Jose A Mendiola
- Foodomics Laboratory, Bioactivity and Food Analysis Department, Institute of Food Science Research CIAL (CSIC-UAM), Nicolas Cabrera 9, 28049 Madrid, Spain.
| |
Collapse
|
15
|
Mi XJ, Choi HS, Perumalsamy H, Shanmugam R, Thangavelu L, Balusamy SR, Kim YJ. Biosynthesis and cytotoxic effect of silymarin-functionalized selenium nanoparticles induced autophagy mediated cellular apoptosis via downregulation of PI3K/Akt/mTOR pathway in gastric cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:154014. [PMID: 35247670 DOI: 10.1016/j.phymed.2022.154014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 02/08/2022] [Accepted: 02/25/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Silymarin, a blend of flavonolignans isolated from plant Silybum marianum L., has long been used as an herbal medicine. Biogenic routes especially the plant-based synthesis of selenium nanoparticles (SeNPs) is safe, eco-friendly, nontoxic and being considered as one of the best strategies for treatment of cancer. PURPOSE Silymarin-mediated green synthesis of SeNPs and their possibility as an anticancer agent have not been reported to date. Therefore, our present study was aimed to synthesize and characterize the selenium mediated silymarin nanoparticles (Si-SeNPs) from silymarin and investigate their possibility as an anticancer agent. METHODS The physicochemical characteristics of Si-SeNPs were analyzed using various analytical techniques, such as HPLC, field emission-transmission electron microscope, energy-dispersive X-ray spectrometer, and thermogravimetric analysis. The underlying molecular mechanism were evaluated using AGS gastric cancer cells. RESULTS Compared with silymarin, the Si-SeNPs exhibited significantly increased cytotoxic effect of AGS cells without exhibiting toxicity on normal cells. Real time PCR and western blotting analysis indicated that Si-SeNPs induced expression of Bax/Bcl-2, cytochrome c, and cleavage of caspase proteins, which is associated with mitochondria-mediated apoptosis signaling in AGS cells. Moreover, agonist assay using PI3K activator indicated that Si-SeNPs-inhibited PI3K/AKT/mTOR pathways were significantly associated as an autophagy and apoptosis signaling in AGS cells. CONCLUSION Our study demonstrated the improved anticancer efficacy of Si-SeNPs- induced apoptosis and autophagy pathways, and therefore recommended Si-SeNPs as a novel anticancer agent after in vivo studies.
Collapse
Affiliation(s)
- Xiao-Jie Mi
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
| | - Han Sol Choi
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
| | - Haribalan Perumalsamy
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea; Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Rajeshkumar Shanmugam
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha University, SIMATS, Chennai 600077, TN, India
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha University, SIMATS, Chennai 600077, TN, India
| | - Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University, Gwangjin-gu, Seoul, 05006, Republic of Korea.
| | - Yeon-Ju Kim
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
16
|
Isolation and Characterization of a Flavonoid and a Neolignan from
Silybum marianum
: In‐vitro Cytotoxic Evaluation. ChemistrySelect 2022. [DOI: 10.1002/slct.202200502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
17
|
Amer ME, Amer MA, Othman AI, Elsayed DA, El-Missiry MA, Ammar OA. Silymarin inhibits the progression of Ehrlich solid tumor via targeting molecular pathways of cell death, proliferation, angiogenesis, and metastasis in female mice. Mol Biol Rep 2022; 49:4659-4671. [PMID: 35305227 DOI: 10.1007/s11033-022-07315-2] [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: 12/30/2021] [Accepted: 03/01/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Plant-derived phytochemicals have been reported to exert anticancer activity. This study investigated the antitumor role of silymarin (Silybum marianum) (SMN) and its molecular targets in Ehrlich solid tumor xenografts in vivo. METHODS AND RESULTS Female Swiss albino mice were divided into three groups (of five animals each) that were engrafted with Ehrlich tumor (ET) cells with or without SMN treatment. The 3rd groups treated with DMSO only vehicle control group. A significant reduction in animal body mass and tumor volume/weight were observed in xenografted mice treated with SMN. SMN modulated oxidative stress in tumors while enhancing the antioxidant levels in mouse serum. SMN activated both mitochondrial and death receptor-related apoptosis pathways and induced cell cycle arrest, marked by a significant downregulation of cyclin D1 in SMN-treated tumors. Significant decreases in RNA content and protein expression levels of Ki-67 and proliferating cell nuclear antigen were observed in ET cells. Additionally, SMN downregulated vascular endothelial growth factor and nuclear factor-kappa B levels indicating anti-angiogenesis activity of this agent. SMN upregulated the expression of E-cadherin in tumor tissue suggesting, that SMN has potential ability to inhibit metastasis. Tumor tissue from SMN-treated animals showed a remarkable degeneration and reduction in the neoplastic cell density. CONCLUSIONS The anticancer effect was associated with apparent apoptosis in neoplastic cells with abundance of multifocal necrotic areas. SMN was found to inhibit ET growth via enhancing apoptosis, inhibition of cell division and reduction in angiogenesis in vivo. Hypothetical scheme of SMN antitumor effects (mechanism of signaling) in solid ET in vivo. SMN anticancer effect may be mediated by molecular mediators that affect proliferation, cell cycle activity, apoptotic pathways, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Maggie E Amer
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt.
| | - Maher A Amer
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt
| | - Azza I Othman
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt
| | - Doaa A Elsayed
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura, Egypt
| | | | - Omar A Ammar
- Basic Science Department, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
18
|
Melim C, Magalhães M, Santos AC, Campos EJ, Cabral C. Nanoparticles as phytochemical carriers for cancer treatment: News of the last decade. Expert Opin Drug Deliv 2022; 19:179-197. [PMID: 35166619 DOI: 10.1080/17425247.2022.2041599] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The development and application of novel therapeutic medicines for the treatment of cancer are of vital importance to improve the disease's outcome and survival rate. One noteworthy treatment approach is the use of biologically active compounds present in natural products. Even though these phytocompounds present anti-inflammatory, antioxidant, and anticancer properties, their use is limited essentially due to poor systemic delivery, low bioavailability, and water solubility concerns. To make full use of the anticancer potential of natural products, these limitations need to be technologically addressed. In this sense, nanotechnology emerges as a promising drug delivery system strategy. AREAS COVERED In this review, the benefits and potential of nanodelivery systems for natural products encapsulation as promising therapeutic approaches for cancer, which were developed during the last decade, are highlighted. EXPERT OPINION The nanotechnology area has been under extensive research in the medical field given its capacity for improving the therapeutic potential of drugs by increasing their bioavailability and allowing a targeted delivery to the tumor site. Thereby, the nanoencapsulation of phytocompounds can have a direct impact on the recognized therapeutic activity of natural products towards cancer.
Collapse
Affiliation(s)
- Catarina Melim
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, 3000-548 Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
| | - Mariana Magalhães
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, 3000-548 Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Ana Cláudia Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Elisa Julião Campos
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, 3000-548 Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| | - Célia Cabral
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, 3000-548 Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal.,Centre for Functional Ecology, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| |
Collapse
|
19
|
Yassin NYS, AbouZid SF, El-Kalaawy AM, Ali TM, Almehmadi MM, Ahmed OM. Silybum marianum total extract, silymarin and silibinin abate hepatocarcinogenesis and hepatocellular carcinoma growth via modulation of the HGF/c-Met, Wnt/β-catenin, and PI3K/Akt/mTOR signaling pathways. Biomed Pharmacother 2022; 145:112409. [PMID: 34781148 DOI: 10.1016/j.biopha.2021.112409] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has been identified as one of the most deadly malignancies with limited therapeutic efficacy worldwide. However, understanding the molecular mechanisms of crosstalk between signaling pathways in HCC and predicting cancer cell responses to targeted therapeutic interventions remain to be challenge. Thus, in this study, we aimed to evaluate the anticancerous efficacy of Silybum marianum total extract (STE), silymarin (Sm), and silibinin (Sb) against experimentally-induced HCC in rats. In vitro investigations were also performed and the anticancer effects against HCC cell lines (HepG2 and Huh7) were confirmed. Wistar rats were given diethylnitrosamine (DEN)/2-acetylaminofluorene (AAF)/carbon tetrachloride (CCl4) and were orally treated with STE (200 mg/kg body weight (bw)), Sm (150 mg/kg bw), and Sb (5 mg/kg bw) every other day from the 1st or 16th week to the 25th week of DEN/AAF/CCl4 injection. Treatment with STE, Sm, and Sb inhibited the growth of cancerous lesions in DEN/AAF/CCl4-treated rats. This inhibition was associated with inhibition of Ki-67 expression and repression of HGF/cMet, Wnt/β-catenin, and PI3K/Akt/mTOR signaling pathways. STE, Sm, and Sb improved liver function biomarkers and tumor markers (AFP, CEA, and CA19.9) and increased total protein and albumin levels in serum. STE, Sm, and Sb treatment was also noted to reduce the hepatic production of lipid peroxides, increase hepatic glutathione content, and induce the activities of hepatic antioxidant enzymes in DEN/AAF/CCl4-treated rats. These results indicate that STE, Sm, and Sb exert anti-HCC effects through multiple pathways, including suppression of Ki-67 expression and HGF/cMet, Wnt/β-catenin, and PI3K/Akt/mTOR pathways and enhancement of antioxidant defense mechanisms.
Collapse
Affiliation(s)
- Nour Y S Yassin
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Sameh F AbouZid
- Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University for Sustainable Development, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt
| | - Asmaa M El-Kalaawy
- Department of Pharmacology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Tarek M Ali
- Department of Physiology, College of Medicine, Taif University, P. O. Box 11099, Taif 21944, Saudi Arabia
| | - Mazen M Almehmadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P. O. Box 11099, Taif 21944, Saudi Arabia
| | - Osama M Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt.
| |
Collapse
|
20
|
Yassin NYS, AbouZid SF, El-Kalaawy AM, Ali TM, Elesawy BH, Ahmed OM. Tackling of Renal Carcinogenesis in Wistar Rats by Silybum marianum Total Extract, Silymarin, and Silibinin via Modulation of Oxidative Stress, Apoptosis, Nrf2, PPAR γ, NF- κB, and PI3K/Akt Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7665169. [PMID: 34630852 PMCID: PMC8497111 DOI: 10.1155/2021/7665169] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022]
Abstract
The present work was designed to assess the efficacy of Silybum marianum total extract (STE), silymarin (Sm), and silibinin (Sb) against experimentally induced renal carcinogenesis in male Wistar rats and their roles in regulating oxidative stress, inflammation, apoptosis, and carcinogenesis. The diethylnitrosamine (DEN)/2-acetylaminofluorene (AAF)/carbon tetrachloride (CCl4)-administered rats were orally treated with STE (200 mg/kg b.w.), Sm (150 mg/kg b.w.), and Sb (5 mg/kg b.w.) every other day either from the 1st week or from the 16th week of carcinogen administration to the end of 25th week. The treatments with STE, Sm, and Sb attenuated markers of toxicity in serum, decreased kidney lipid peroxidation (LPO), and significantly reinforced the renal antioxidant armory. The biochemical results were further confirmed by the histopathological alterations. The treatments also led to suppression of proinflammatory mediators such as NF-κβ, p65, Iκβα, and IL-6 in association with inhibition of the PI3K/Akt pathway. Furthermore, they activated the expressions of PPARs, Nrf2, and IL-4 in addition to downregulation of apoptotic proteins p53 and caspase-3 and upregulation of antiapoptotic mediator Bcl-2. The obtained data supply potent proof for the efficacy of STE, Sm, and Sb to counteract renal carcinogenesis via alteration of varied molecular pathways.
Collapse
Affiliation(s)
- Nour Y. S. Yassin
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Sameh F. AbouZid
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Asmaa M. El-Kalaawy
- Department of Pharmacology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Tarek M. Ali
- Department of Physiology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Basem H. Elesawy
- Department of Pathology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Osama M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
21
|
Xia T, Li J, Ren X, Liu C, Sun C. Research progress of phenolic compounds regulating IL-6 to exert antitumor effects. Phytother Res 2021; 35:6720-6734. [PMID: 34427003 DOI: 10.1002/ptr.7258] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/08/2021] [Accepted: 08/08/2021] [Indexed: 02/06/2023]
Abstract
Cytokine therapy, which activates the host immune system, has become an important and novel therapeutic approach to treat various cancers. Recent studies have shown that IL-6 is an important cytokine that regulates the homeostasis in vivo. However, excessive IL-6 plays a pathological role in a variety of acute and chronic inflammatory diseases, especially in cancer. IL-6 can transmit signals through JAK/STAT, RAS /MAPK, PI3K/ Akt, NF-κB, and other pathways to promote cancer progression. Phenolic compounds can effectively regulate the level of IL-6 in tumor cells and improve the tumor microenvironment. This article focuses on the phenolic compounds through the regulation of IL-6, participate in the prevention of cancer, inhibit the proliferation of cancer cells, reduce angiogenesis, improve therapeutic efficacy, and reduce side effects and other aspects. This will help to further advance research on cytokine therapy to reduce the burden of cancer and improve patient prognosis. However, current studies are mostly limited to animal and cellular experiments, and high-quality clinical studies are needed to further determine their antitumor efficacy in humans.
Collapse
Affiliation(s)
- Tingting Xia
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin Ren
- Clinical Medical Colleges, Weifang Medical University, Weifang, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
22
|
Elsawy H, Alzahrani AM, Alfwuaires M, Sedky A, El-Trass EE, Mahmoud O, Abdel-Moneim AM, Khalil M. Analysis of silymarin-modulating effects against acrylamide-induced cerebellar damage in male rats: Biochemical and pathological markers. J Chem Neuroanat 2021; 115:101964. [PMID: 33965515 DOI: 10.1016/j.jchemneu.2021.101964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/16/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Acrylamide (ACR) is a well-proven neurotoxin and potential food carcinogen in humans and rodent models. Silymarin (SIL) is a flavonoid mixture isolated from seeds, leaves, and fruits of Silymarin marianum (milk thistle) that possesses a free-radical scavenging effect. OBJECTIVE In this work, the primary focus was to investigate the efficacy of SIL to mitigate ACR-induced subacute neurotoxic effects and oxidative changes in rat cerebellum. METHODS Adult male rats were treated intraperitoneally with ACR (50 mg/kg) with or without SIL (160 mg/kg). The neuropathology and biochemical parameters viz. lipid peroxidation (measured as levels of malondialdehyde or MDA), catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), serotonin (5-hydroxytryptamine; 5-HT), dopamine (DA), and cathepsin D (CTSD) in the cerebellum have been evaluated. RESULTS The data showed that ACR induced redox disruptions as measured by increased MDA levels and inhibition of CAT, SOD, and GPx antioxidant enzyme activities. Besides, cerebellar monoamine neurotransmitters, 5-HT and DA, were depleted in ACR-treated rats. Furthermore, ACR administration caused a significant elevation of CTSD activity, indicating that ACR could trigger apoptosis or apoptosis-like death. At the tissue level, cerebellar cortex sections from ACR-treated animals were characterized by severe neuronal damage. The administration of SIL to ACR-treated rats remarkably alleviated all the aforementioned ACR-induced effects. CONCLUSION SIL has a potent therapeutic effect against ACR-induced cerebellar neurotoxicity in experimental rats via the attenuation of oxidative/antioxidative responses and the inhibition of CTSD-activity.
Collapse
Affiliation(s)
- Hany Elsawy
- Department of Chemistry, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa, 31982, Saudi Arabia; Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Abdullah M Alzahrani
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa, 31982, Al-Ahsa, Saudi Arabia.
| | - Manal Alfwuaires
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa, 31982, Al-Ahsa, Saudi Arabia.
| | - Azza Sedky
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa, 31982, Al-Ahsa, Saudi Arabia; Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Eman E El-Trass
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt.
| | - Omar Mahmoud
- Essex Partnership University NHS Foundation Trust: Harlow, Essex, Great Britain, United Kingdom.
| | - Ashraf M Abdel-Moneim
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa, 31982, Al-Ahsa, Saudi Arabia; Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Mahmoud Khalil
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt; Department of Biological Sciences, Faculty of Science, Beirut Arab University, Lebanon.
| |
Collapse
|
23
|
Faisal Z, Mohos V, Fliszár-Nyúl E, Valentová K, Káňová K, Lemli B, Kunsági-Máté S, Poór M. Interaction of silymarin components and their sulfate metabolites with human serum albumin and cytochrome P450 (2C9, 2C19, 2D6, and 3A4) enzymes. Biomed Pharmacother 2021; 138:111459. [PMID: 33706132 DOI: 10.1016/j.biopha.2021.111459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Silymarin is a mixture of flavonolignans isolated from the fruit of milk thistle (Silybum marianum (L.) Gaertner). Milk thistle extract is the active ingredient of several medications and dietary supplements to treat liver injury/diseases. After the oral administration, flavonolignans are extensively biotransformed, resulting in the formation of sulfate and/or glucuronide metabolites. Previous studies demonstrated that silymarin components form stable complexes with serum albumin and can inhibit certain cytochrome P450 (CYP) enzymes. Nevertheless, in most of these investigations, silybin was tested; while no or only limited information is available regarding other silymarin components and metabolites. In this study, the interactions of five silymarin components (silybin A, silybin B, isosilybin A, silychristin, and 2,3-dehydrosilychristin) and their sulfate metabolites were examined with human serum albumin and CYP (2C9, 2C19, 2D6, and 3A4) enzymes. Our results demonstrate that each compound tested forms stable complexes with albumin, and certain silymarin components/metabolites can inhibit CYP enzymes. Most of the sulfate conjugates were less potent inhibitors of CYP enzymes, but 2,3-dehydrosilychristin-19-O-sulfate showed the strongest inhibitory effect on CYP3A4. Based on these observations, the simultaneous administration of high dose silymarin with medications should be carefully considered, because milk thistle flavonolignans and/or their sulfate metabolites may interfere with drug therapy.
Collapse
Affiliation(s)
- Zelma Faisal
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, Pécs H-7624, Hungary; János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary.
| | - Violetta Mohos
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, Pécs H-7624, Hungary; János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary.
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, Pécs H-7624, Hungary; János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary.
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Kristýna Káňová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; University of Chemistry and Technology Prague, Technická 5, 166 28 Prague, Czech Republic.
| | - Beáta Lemli
- János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary; Institute of Organic and Medicinal Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary.
| | - Sándor Kunsági-Máté
- János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary; Institute of Organic and Medicinal Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary.
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Szigeti út 12, Pécs H-7624, Hungary; János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary.
| |
Collapse
|
24
|
Zhao M, Chen S, Ji X, Shen X, You J, Liang X, Yin H, Zhao L. Current innovations in nutraceuticals and functional foods for intervention of non-alcoholic fatty liver disease. Pharmacol Res 2021; 166:105517. [PMID: 33636349 DOI: 10.1016/j.phrs.2021.105517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023]
Abstract
As innovations in global agricultural production and food trading systems lead to major dietary shifts, high morbidity rates from non-alcoholic fatty liver disease (NAFLD), accompanied by elevated risk of lipid metabolism-related complications, has emerged as a growing problem worldwide. Treatment and prevention of NAFLD and chronic liver disease depends on the availability of safe, effective, and diverse therapeutic agents, the development of which is urgently needed. Supported by a growing body of evidence, considerable attention is now focused on interventional approaches that combines nutraceuticals and functional foods. In this review, we summarize the pathological progression of NAFLD and discuss the beneficial effects of nutraceuticals and the active ingredients in functional foods. We also describe the underlying mechanisms of these compounds in the intervention of NAFLD, including their effects on regulation of lipid homeostasis, activation of signaling pathways, and their role in gut microbial community dynamics and the gut-liver axis. In order to identify novel targets for treatment of lipid metabolism-related diseases, this work broadly explores the molecular mechanism linking nutraceuticals and functional foods, host physiology, and gut microbiota. Additionally, the limitations in existing knowledge and promising research areas for development of active interventions and treatments against NAFLD are discussed.
Collapse
Affiliation(s)
- Mengyao Zhao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China
| | - Shumin Chen
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaoguo Ji
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xin Shen
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Jiangshan You
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xinyi Liang
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China.
| | - Liming Zhao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China; School of Life Science, Shandong University of Technology, Zibo, Shandong 255000, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China.
| |
Collapse
|
25
|
Ali AM, El-Tawil OS, Al-Mokaddem AK, Abd El-Rahman SS. Promoted inhibition of TLR4/miR-155/ NF kB p65 signaling by cannabinoid receptor 2 agonist (AM1241), aborts inflammation and progress of hepatic fibrosis induced by thioacetamide. Chem Biol Interact 2021; 336:109398. [PMID: 33503444 DOI: 10.1016/j.cbi.2021.109398] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/04/2020] [Accepted: 01/22/2021] [Indexed: 12/20/2022]
Abstract
The endocannabinoid system plays a pivotal role, whether it is promoting or dampening hepatic fibrosis. This study investigated the role of Cannabinoid receptor 2 (CB2) activation by the synthetic analog (AM1241) on revoking the progress of liver fibrosis. Thioacetamide (TAA) was used to induce liver fibrosis in rats for three weeks followed by its concurrent administration with AM1241 at two different doses for another three weeks. Markers for liver function and oxidative stress, hepatic TNF-α, IL-1β and IL-6, qRT-PCR expression of Toll like receptor 4 (TLR4), TGF-β1, α-SMA and microRNA-155 (miR-155) genes, Western blot for protein levels of Vimentin and E-cadherin, immunohistochemical expression of NFκB p65 and histopathology of liver tissue were all investigated. AM1241 administration significantly maintained liver function markers and decreased; malondialdehyde, Vimentin, TLR4, TGF-β1, α-SMA and miR-155 genes expression, NFκB p65 immune-expression and pro-inflammatory cytokines (TNF-α, IL-1β and IL-6). Additionally, AM1241 significantly increased E-Cadherin level, GSH and SOD content. Histologically, AM1241 limited fibroplasia extension, and broke the itinerary of bridging fibrosis. In conclusion, activation of the CB2 receptors by AM1241 promoted liver regeneration and overrun the progression of liver fibrosis through; inhibition of TLR4/miR-155/NFκB p65 pathway, suppression of pro-inflammatory IL-6, IL-1β and TNF-α, reducing TGF-β1, α-SMA, Vimentin and up-regulating E-Cadherin.
Collapse
Affiliation(s)
- Alaa M Ali
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Osama S El-Tawil
- Department of Toxicology, Forensic Medicine and Veterinary Regulations, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Asmaa K Al-Mokaddem
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | | |
Collapse
|
26
|
Meresman GF, Götte M, Laschke MW. Plants as source of new therapies for endometriosis: a review of preclinical and clinical studies. Hum Reprod Update 2020; 27:367-392. [PMID: 33124671 DOI: 10.1093/humupd/dmaa039] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Given the disadvantages and limitations of current endometriosis therapy, there is a progressive increase in studies focusing on plant-derived agents as a natural treatment option with the intention of achieving high efficiency, avoiding adverse effects and preserving the chance for successful pregnancy. The heterogeneity of these studies in terms of evaluated agents, applied approaches and outcomes illustrates the need for an up-to-date summary and critical view on this rapidly growing field in endometriosis research. OBJECTIVE AND RATIONALE This review provides a comprehensive overview of plant-derived agents and natural treatment strategies that are under preclinical or clinical investigation and critically evaluates their potential for future endometriosis therapy. SEARCH METHODS An English language PubMed literature search was performed using variations of the terms 'endometriosis', 'natural therapy', 'herb/herbal', 'plant', 'flavonoid', 'polyphenol', 'phytochemical', 'bioactive', 'Kampo' and 'Chinese medicine'. It included both animal and human studies. Moreover, the Clinicaltrials.gov database was searched with the term 'endometriosis' for clinical trials on plant-derived agents. No restriction was set for the publication date. OUTCOMES Natural therapies can be assigned to three categories: (i) herbal extracts, (ii) specific plant-derived bioactive compounds and (iii) Chinese herbal medicine (CHM). Agents of the first category have been shown to exert anti-proliferative, anti-inflammatory, anti-angiogenic and anti-oxidant effects on endometrial cells and endometriotic lesions. However, the existing evidence supporting their use in endometriosis therapy is quite limited. The most studied specific plant-derived bioactive compounds are resveratrol, epigallocatechin-3-gallate, curcumin, puerarin, ginsenosides, xanthohumol, 4-hydroxybenzyl alcohol, quercetin, apigenin, carnosic acid, rosmarinic acid, wogonin, baicalein, parthenolide, andrographolide and cannabinoids, with solid evidence about their inhibitory activity in experimental endometriosis models. Their mechanisms of action include pleiotropic effects on known signalling effectors: oestrogen receptor-α, cyclooxygenase-2, interleukin-1 and -6, tumour necrosis factor-α, intercellular adhesion molecule-1, vascular endothelial growth factor, nuclear factor-kappa B, matrix metalloproteinases as well as reactive oxygen species (ROS) and apoptosis-related proteins. Numerous studies suggest that treatment with CHM is a good choice for endometriosis management. Even under clinical conditions, this approach has already been shown to decrease the size of endometriotic lesions, alleviate chronic pelvic pain and reduce postoperative recurrence rates. WIDER IMPLICATIONS The necessity to manage endometriosis as a chronic disease highlights the importance of identifying novel and affordable long-term safety therapeutics. For this purpose, natural plant-derived agents represent promising candidates. Many of these agents exhibit a pleiotropic action profile, which simultaneously inhibits fundamental processes in the pathogenesis of endometriosis, such as proliferation, inflammation, ROS formation and angiogenesis. Hence, their inclusion into multimodal treatment concepts may essentially contribute to increase the therapeutic efficiency and reduce the side effects of future endometriosis therapy.
Collapse
Affiliation(s)
- Gabriela F Meresman
- Institute of Biology and Experimental Medicine (IBYME-CONICET), C1428ADN Buenos Aires, Argentina
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
27
|
Wang X, Zhang Z, Wu SC. Health Benefits of Silybum marianum: Phytochemistry, Pharmacology, and Applications. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:11644-11664. [PMID: 33045827 DOI: 10.1021/acs.jafc.0c04791] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Silybum marianum (SM), a well-known plant used as both a medicine and a food, has been widely used to treat various diseases, especially hepatic diseases. The seeds and fruits of SM contain a flavonolignan complex called silymarin, the active compounds of which include silybin, isosilybin, silychristin, dihydrosilybin, silydianin, and so on. In this review, we thoroughly summarize high-quality publications related to the pharmacological effects and underlying mechanisms of SM. SM has antimicrobial, anticancer, hepatoprotective, cardiovascular-protective, neuroprotective, skin-protective, antidiabetic, and other effects. Importantly, SM also counteracts the toxicities of antibiotics, metals, and pesticides. The diverse pharmacological activities of SM provide scientific evidence supporting its use in both humans and animals. Multiple signaling pathways associated with oxidative stress and inflammation are the common molecular targets of SM. Moreover, the flavonolignans of SM are potential agonists of PPARγ and ABCA1, PTP1B inhibitors, and metal chelators. At the end of the review, the potential and perspectives of SM are discussed, and these insights are expected to facilitate the application of SM and the discovery and development of new drugs. We conclude that SM is an interesting dietary medicine for health enhancement and drug discovery and warrants further investigation.
Collapse
Affiliation(s)
- Xin Wang
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Qingdao, Shandong 266109, People's Republic of China
| | - Zhen Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Qingdao, Shandong 266109, People's Republic of China
| | - Shuai-Cheng Wu
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Qingdao, Shandong 266109, People's Republic of China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, People's Republic of China
| |
Collapse
|
28
|
Wang C, He C, Lu S, Wang X, Wang L, Liang S, Wang X, Piao M, Cui J, Chi G, Ge P. Autophagy activated by silibinin contributes to glioma cell death via induction of oxidative stress-mediated BNIP3-dependent nuclear translocation of AIF. Cell Death Dis 2020; 11:630. [PMID: 32801360 PMCID: PMC7429844 DOI: 10.1038/s41419-020-02866-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
Induction of lethal autophagy has become a strategy to eliminate glioma cells, but it remains elusive whether autophagy contributes to cell death via causing mitochondria damage and nuclear translocation of apoptosis inducing factor (AIF). In this study, we find that silibinin induces AIF translocation from mitochondria to nuclei in glioma cells in vitro and in vivo, which is accompanied with autophagy activation. In vitro studies reveal that blocking autophagy with 3MA, bafilomycin A1 or by knocking down ATG5 with SiRNA inhibits silibinin-induced mitochondrial accumulation of superoxide, AIF translocation from mitochondria to nuclei and glioma cell death. Mechanistically, silibinin activates autophagy through depleting ATP by suppressing glycolysis. Then, autophagy improves intracellular H2O2 via promoting p53-mediated depletion of GSH and cysteine and downregulation of xCT. The increased H2O2 promotes silibinin-induced BNIP3 upregulation and translocation to mitochondria. Knockdown of BNIP3 with SiRNA inhibits silibinin-induced mitochondrial depolarization, accumulation of mitochondrial superoxide, and AIF translocation from mitochondria to nuclei, as well as prevents glioma cell death. Furthermore, we find that the improved H2O2 reinforces silibinin-induced glycolysis dysfunction. Collectively, autophagy contributes to silibinin-induced glioma cell death via promotion of oxidative stress-mediated BNIP3-dependent nuclear translocation of AIF.
Collapse
Affiliation(s)
- Chongcheng Wang
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Chuan He
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Shan Lu
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Xuanzhong Wang
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Lei Wang
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Shipeng Liang
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Xinyu Wang
- Department of Radiotherapy, Second Hospital of Jilin University, 130021, Changchun, China
| | - Meihua Piao
- Department of Anesthesiology, First Hospital of Jilin University, 130021, Changchun, China
| | - Jiayue Cui
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, 130021, Changchun, China
| | - Guangfan Chi
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 130021, Changchun, China
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China.
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China.
| |
Collapse
|
29
|
Izzo AA. An updated PTR virtual issue on the pharmacology of the nutraceutical curcumin. Phytother Res 2020; 34:671-673. [PMID: 32077178 DOI: 10.1002/ptr.6635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Angelo A Izzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
30
|
Knecht K, Kinder D, Stockert A. Biologically-Based Complementary and Alternative Medicine (CAM) Use in Cancer Patients: The Good, the Bad, the Misunderstood. Front Nutr 2020; 6:196. [PMID: 32039227 PMCID: PMC6992534 DOI: 10.3389/fnut.2019.00196] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
As complementary and alternative medicine (CAM) becomes more popular, it is being used in cancer patients to aid in recovery or to treat symptoms associated with the current chemotherapy. Numerous papers exist that discuss patients using CAM with cancer chemotherapy and their outcomes-both positive and negative. However, in the case of the negative outcomes, the reason for the dangers or interactions with drugs are not made clear. Indeed, many chemotherapy regimens are rendered less effective by the well-meaning but uninformed patient or their family members and friends. Similarly, reports of positive outcomes with CAM and chemotherapy provide a strong basis for further research, but do not identify specific mechanisms of action. These small clinical studies and in vitro studies identify a necessary area for further research and provide a much needed, although often rejected, alternative look at whole treatment plans. Careful review of the available information and evaluation of the nature of the CAM effects are necessary to combat the misunderstanding and sometimes unwarranted claims over CAM use. This mini review will explore some of the commonly used CAM agents and their mechanisms of interactions with other treatments. Suggestions as to which agents can be safe and when to use them will be an integral part of this review.
Collapse
Affiliation(s)
- Kathryn Knecht
- Loma Linda University School of Pharmacy, Loma Linda University, Loma Linda, CA, United States
| | - David Kinder
- Department of Pharmaceutical and Biomedical Sciences, Raabe College of Pharmacy, Ohio Northern University, Ada, OH, United States
| | - Amy Stockert
- Department of Pharmaceutical and Biomedical Sciences, Raabe College of Pharmacy, Ohio Northern University, Ada, OH, United States
| |
Collapse
|
31
|
Antal DS, Ardelean F, Avram S, Pavel IZ, Danciu C, Soica C, Dehelean C. Flavonolignans: One Step Further in the Broad-Spectrum Approach of Cancer. Anticancer Agents Med Chem 2020; 20:1817-1830. [PMID: 31976848 DOI: 10.2174/1871520620666200124112649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/11/2019] [Accepted: 12/24/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The small chemical class of flavonolignans encompasses unique hybrid molecules with versatile biological activities. Their anticancer effects have received considerable attention, and a large body of supporting evidence has accumulated. Moreover, their ability to interact with proteins involved in drug resistance, and to enhance the effects of conventional chemotherapeutics in decreasing cell viability make them influential partners in addressing cancer. OBJECTIVE The review provides an outline of the various ways in which flavonolignans advance the combat against cancer. While the main focus falls on flavonolignans from milk thistle, attention is drawn to the yet, underexplored potential of less known flavonolignan subgroups derived from isoflavonoids and aurones. METHODS Proceeding from the presentation of natural flavonolignan subtypes and their occurrence, the present work reviews these compounds with regard to their molecular targets in cancer, anti-angiogenetic effects, synergistic efficacy in conjunction with anticancer agents, reversal of drug resistance, and importance in overcoming the side effects of anticancer therapy. Recent advances in the endeavor to improve flavonolignan bioavailability in cancer are also presented. CONCLUSIONS Significant progress has been achieved in detailing the molecular mechanisms of silybin and its congeners in experimental models of cancer. The availability of novel formulations with improved bioavailability, and data from phase I clinical trials in cancer patients provide an encouraging basis for more extensive trials aimed at evaluating the benefits of Silybum flavonolignans in cancer management. On the other hand, further research on the antitumor efficacy of iso-flavonolignans and other subtypes of flavonolignans should be pursued.
Collapse
Affiliation(s)
- Diana S Antal
- Department of Pharmaceutical Botany, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Florina Ardelean
- Department of Pharmaceutical Botany, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Stefana Avram
- Department of Pharmacognosy, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ioana Z Pavel
- Department of Pharmacognosy, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Corina Danciu
- Department of Pharmacognosy, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Codruta Soica
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Cristina Dehelean
- Department of Toxicology, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|