1
|
Keshavarz M, Ghorbani M, Shamsizadeh F, Namdari H, Salimi V, Rezaei F. Effects and Mechanisms of Silibinin on Influenza A/H1N1 Pathogenesis in a Mouse Model. J Trop Med 2025; 2025:6618423. [PMID: 39850538 PMCID: PMC11756948 DOI: 10.1155/jotm/6618423] [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: 01/26/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
Silymarin is a polyphenolic flavonoid extracted from milk thistle. It has potent immunomodulatory effects and can inhibit the replication of influenza A virus (IAV). The present study aimed to determine the inflammatory and anti-inflammatory cytokine secretion patterns in mice before and after silibinin treatment. For this, bronchoalveolar lavage (BAL) fluids were collected from the thoracic cavity 5 days after the intervention, and viral quantification was performed using TaqMan Real-time PCR. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate IFN-γ and IL-10 levels in serum and BAL samples. Finally, pathological damage to lung tissue was assessed by pathologists. The results reveal that silibinin pretreatment exhibits a dose-dependent immunomodulatory effect on IFN-γ and IL-10 levels. After the virus challenge, silibinin reduced immune cell infiltration in mouse BAL fluid. These data similarly suggest a remarkable immunomodulatory effect of silibinin. Silibinin also decreased lung damage following the virus challenge in the post-treatment group, but its lung protective properties seem to be due to a different mechanism than when it was administered before infection. Finally, high doses of silibinin (post-treatment) significantly reduced viral load in BAL fluid compared to the virus challenge group. These results support the idea that therapies aimed at moderating immune and inflammatory responses are essential to decrease the mortality rate caused by IAV infection. Silibinin has strong immunomodulatory properties, can inhibit IAV infection, and reduces lung tissue damage in a dose-dependent manner.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohsen Ghorbani
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Shamsizadeh
- Department of Parasitology and Mycology, School of Paramedicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Haideh Namdari
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- School of Public Health, National Influenza Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
El-Banna AA, Ibrahim RS. Metabolic profiling of milk thistle different organs using UPLC-TQD-MS/MS coupled to multivariate analysis in relation to their selective antiviral potential. BMC Complement Med Ther 2024; 24:115. [PMID: 38454377 PMCID: PMC10921647 DOI: 10.1186/s12906-024-04411-7] [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: 09/09/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
INTRODUCTION Silybum marianum commonly known as milk thistle is one of the most imperative medicinal plants due to its remarkable pharmacological activities. Lately, the antiviral activities of S. marianum extract have been studied and it showed effectiveness against many viruses. OBJECTIVE Although most previous studies were concerned mainly with silymarin content of the fruit, the present study provides comprehensive comparative evaluation of S. marianum different organs' chemical profiles using UPLC-MS/MS coupled to chemometrics to unravel potentially selective antiviral compounds against human coronavirus (HCoV-229E). METHODOLOGY UPLC-ESI-TQD-MS/MS analysis was utilized to establish metabolic fingerprints for S. marianum organs namely fruits, roots, stems and seeds. Multivariate analysis, using OPLS-DA and HCA-heat map was applied to explore the main discriminatory phytoconstituents between organs. Selective virucidal activity of organs extracts against coronavirus (HCoV-229E) was evaluated for the first time using cytopathic effect (CPE) inhibition assay. Correlation coefficient analysis was implemented for detection of potential constituents having virucidal activity. RESULTS UPLC-MS/MS analysis resulted in 87 identified metabolites belonging to different classes. OPLS-DA revealed in-between class discrimination between milk thistle organs proving their significantly different metabolic profiles. The results of CPE assay showed that all tested organ samples exhibited dose dependent inhibitory activity in nanomolar range. Correlation analysis disclosed that caffeic acid-O-hexoside, gadoleic and linolenic acids were the most potentially selective antiviral phytoconstituents. CONCLUSION This study valorizes the importance of different S. marianum organs as wealthy sources of selective and effective antiviral candidates. This approach can be extended to unravel potentially active constituents from complex plant matrices.
Collapse
Affiliation(s)
- Alaa A El-Banna
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Reham S Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| |
Collapse
|
3
|
Siew ZY, Asudas E, Khoo CT, Cho GH, Voon K, Fang CM. Fighting nature with nature: antiviral compounds that target retroviruses. Arch Microbiol 2024; 206:130. [PMID: 38416180 DOI: 10.1007/s00203-024-03846-3] [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: 11/16/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/29/2024]
Abstract
The human immunodeficiency virus (HIV) is a type of lentivirus that targets the human immune system and leads to acquired immunodeficiency syndrome (AIDS) at a later stage. Up to 2021, there are millions still living with HIV and many have lost their lives. To date, many anti-HIV compounds have been discovered in living organisms, especially plants and marine sponges. However, no treatment can offer a complete cure, but only suppressing it with a life-long medication, known as combined antiretroviral therapy (cART) or highly active antiretroviral therapy (HAART) which are often associated with various adverse effects. Also, it takes many years for a discovered compound to be approved for clinical use. Thus, by employing advanced technologies such as automation, conducting systematic screening and testing protocols may boost the discovery and development of potent and curative therapeutics for HIV infection/AIDS. In this review, we aim to summarize the antiretroviral therapies/compounds and their associated drawbacks since the discovery of azidothymidine. Additionally, we aim to provide an updated analysis of the most recent discoveries of promising antiretroviral candidates, along with an exploration of the current limitations within antiretroviral research. Finally, we intend to glean insightful perspectives and propose future research directions in this crucial area of study.
Collapse
Affiliation(s)
- Zhen Yun Siew
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia.
| | - Elishea Asudas
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Chia Ting Khoo
- School of Biosciences, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Gang Hyeon Cho
- School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Kenny Voon
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|
4
|
Inai M, Sagara H, Ueno Y, Ouchi H, Yoshimura F, Asakawa T, Hamashima Y, Kan T. Total Synthesis of (+)-Silybin A. Chem Pharm Bull (Tokyo) 2024; 72:570-573. [PMID: 38910121 DOI: 10.1248/cpb.c24-00276] [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] [Indexed: 06/25/2024]
Abstract
We report the first total synthesis of silybin A (1). Key synthetic steps include the construction of the 1,4-benzodioxane neolignan skeleton, a modified Julia-Kocienski olefination reaction between m-nitrophenyltetrazole sulfone (m-NPT sulfone) 10 and aldehyde 21, the formation of the flavanol lignan skeleton 28 via a quinomethide intermediate under acidic conditions, and stepwise oxidation of the benzylic position of flavanol 29.
Collapse
Affiliation(s)
- Makoto Inai
- School of Pharmaceutical Sciences, University of Shizuoka
| | - Hiroto Sagara
- School of Pharmaceutical Sciences, University of Shizuoka
| | - Yoshinori Ueno
- School of Pharmaceutical Sciences, University of Shizuoka
| | - Hitoshi Ouchi
- School of Pharmaceutical Sciences, University of Shizuoka
| | | | | | | | - Toshiyuki Kan
- School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
5
|
Zarenezhad E, Abdulabbas HT, Kareem AS, Kouhpayeh SA, Barbaresi S, Najafipour S, Mazarzaei A, Sotoudeh M, Ghasemian A. Protective role of flavonoids quercetin and silymarin in the viral-associated inflammatory bowel disease: an updated review. Arch Microbiol 2023; 205:252. [PMID: 37249707 DOI: 10.1007/s00203-023-03590-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/14/2023] [Accepted: 05/18/2023] [Indexed: 05/31/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic recurrent inflammation of the gastrointestinal tract (GIT). IBD patients are susceptible to various infections such as viral infections due to the long-term consumption of immunosuppressive drugs and biologics. The antiviral and IBD protective traits of flavonoids have not been entirely investigated. This study objective included an overview of the protective role of flavonoids quercetin and silymarin in viral-associated IBD. Several viral agents such as cytomegalovirus (CMV), Epstein-Barr virus (EBV), varicella zoster virus (VZV) and enteric viruses can be reactivated and thus develop or exacerbate the IBD conditions or eventually facilitate the disease remission. Flavonoids such as quercetin and silymarin are non-toxic and safe bioactive compounds with remarkable anti-oxidant, anti-inflammatory and anti-viral effects. Mechanisms of anti-inflammatory and antiviral effects of silymarin and quercetin mainly include immune modulation and inhibition of caspase enzymes, viral binding and replication, RNA synthesis, viral proteases and viral assembly. In the nutraceutical sector, natural flavonoids low bioavailability and solubility necessitate the application of delivery systems to enhance their efficacy. This review study provided an updated understanding of the protective role of quercetin and silymarin against viral-associated IBD.
Collapse
Affiliation(s)
- Elham Zarenezhad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Hussein T Abdulabbas
- Department of Medical Microbiology, Medical College, Al Muthanna University, Al Muthanna, Iraq
| | - Ahmed Shayaa Kareem
- Department of Medical Laboratories Techniques, Imam Ja'afar Al-Sadiq University, Al-Muthanna, 66002, Iraq
| | - Seyed Amin Kouhpayeh
- Department of Pharmacology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Silvia Barbaresi
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Sohrab Najafipour
- Department of Microbiology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdulbaset Mazarzaei
- Department of Immunology, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mitra Sotoudeh
- Department of Nutrition, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
6
|
Musazadeh V, Karimi A, Bagheri N, Jafarzadeh J, Sanaie S, Vajdi M, Karimi M, Niazkar HR. The favorable impacts of silibinin polyphenols as adjunctive therapy in reducing the complications of COVID-19: A review of research evidence and underlying mechanisms. Biomed Pharmacother 2022; 154:113593. [PMID: 36027611 PMCID: PMC9393179 DOI: 10.1016/j.biopha.2022.113593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/11/2022] Open
Abstract
The proceeding pandemic of coronavirus disease 2019 is the latest global challenge. Like most other infectious diseases, inflammation, oxidative stress, and immune system dysfunctions play a pivotal role in the pathogenesis of COVID-19. Furthermore, the quest of finding a potential pharmaceutical therapy for preventing and treating COVID-19 is still ongoing. Silymarin, a mixture of flavonolignans extracted from the milk thistle, has exhibited numerous therapeutic benefits. We reviewed the beneficial effects of silymarin on oxidative stress, inflammation, and the immune system, as primary factors involved in the pathogenesis of COVID-19. We searched PubMed/Medline, Web of Science, Scopus, and Science Direct databases up to April 2022 using the relevant keywords. In summary, the current review indicates that silymarin might exert therapeutic effects against COVID-19 by improving the antioxidant system, attenuating inflammatory response and respiratory distress, and enhancing immune system function. Silymarin can also bind to target proteins of SARS-CoV-2, including main protease, spike glycoprotein, and RNA-dependent RNA-polymerase, leading to the inhibition of viral replication. Although multiple lines of evidence suggest the possible promising impacts of silymarin in COVID-19, further clinical trials are encouraged.
Collapse
Affiliation(s)
- Vali Musazadeh
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arash Karimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Nasim Bagheri
- Department of microbiology Islamic Azad University of medical science, Tehran, Iran
| | - Jaber Jafarzadeh
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sarvin Sanaie
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Vajdi
- Student Research Committee, Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozhde Karimi
- Department of Immunology, Faculty ofMedical Sciences ,Tarbiat Modares University
| | - Hamid Reza Niazkar
- Breast Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
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: 61] [Impact Index Per Article: 20.3] [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
|
8
|
Aryan H, Farahani RH, Chamanara M, Elyasi S, Jaafari MR, Haddad M, Sani AT, Ardalan MA, Mosaed R. Evaluation of the efficacy of oral nano-silymarin formulation in hospitalized patients with COVID-19: A double-blind placebo-controlled clinical trial. Phytother Res 2022; 36:3924-3931. [PMID: 35859298 PMCID: PMC9349546 DOI: 10.1002/ptr.7537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/20/2022] [Accepted: 06/12/2022] [Indexed: 11/08/2022]
Abstract
Considering the outbreak pandemic of Coronavirus Disease 2019 (COVID-19), the lack of effective therapeutic strategies for the management of this viral disease, and the increasing evidence on the antiviral potential of silymarin, this study aimed to investigate the effectiveness of silymarin nanomicelles on the symptom's resolution time, laboratory parameters, and liver enzymes in patients with COVID-19. The participants were assigned to the nano-silymarin (n = 25) (receiving SinaLive soft gel, containing 70 mg silymarin as nanomicelles) or placebo groups (n = 25) three times daily for two weeks. Patients' symptoms and laboratory findings were assessed at baseline and during the follow-up period (one week and one month after the beginning of the treatment). No significant differences were observed between the two groups in terms of symptoms resolution time, laboratory parameters, and hospitalization duration (p > 0.05). However, the alanine aminotransferase level decreased significantly in the treatment group, compared to the placebo group (p < 0.001). Concomitant use of dexamethasone and remdesivir with silymarin might make the effects of silymarin on the improvement of patients' condition unclear. Further clinical trials are recommended with diverse dosages and larger sample sizes.
Collapse
Affiliation(s)
- Hossein Aryan
- Department of Anesthesiology and Intensive Care, Aja University of Medical Sciences, Tehran, Iran
| | - Ramin Hamidi Farahani
- Infectious Disease Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, Aja University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Sepideh Elyasi
- Department of Clinical Pharmacy, School of Pharmacy Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboubeh Haddad
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ashraf Tavanaee Sani
- Department of Infectious Diseases and Tropical medicine, Faculty of medicine, Mashhad University of medical sciences, Mashhad, Iran
| | | | - Reza Mosaed
- Internal Medicine Department, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Inai M, Ueno Y, Sagara H, Ouchi H, Yoshimura F, Kan T. Total Synthesis of Isosilybin B. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Makoto Inai
- University of Shizuoka School of Pharmaceutical Sciences 52-1 Yada, Suruga-ku 422-8526 Shizuoka JAPAN
| | - Yoshinori Ueno
- Shizuoka Kenritsu Daigaku Department School of Pharmaceutical Sciences JAPAN
| | - Hiroto Sagara
- Shizuoka Kenritsu Daigaku Department School of Pharmaceutical Sciences JAPAN
| | - Hitoshi Ouchi
- Shizuoka Kenritsu Daigaku Department School of Pharmaceutical Sciences JAPAN
| | - Fumihiko Yoshimura
- Shizuoka Kenritsu Daigaku Department School of Pharmaceutical Sciences JAPAN
| | - Toshiyuki Kan
- Shizuoka Kenritsu Daigaku Department School of Pharmaceutical Sciences JAPAN
| |
Collapse
|
10
|
Baranwal M, Gupta Y, Dey P, Majaw S. Antiinflammatory phytochemicals against virus-induced hyperinflammatory responses: Scope, rationale, application, and limitations. Phytother Res 2021; 35:6148-6169. [PMID: 34816512 DOI: 10.1002/ptr.7222] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/26/2021] [Accepted: 07/03/2021] [Indexed: 12/11/2022]
Abstract
Uncontrolled inflammatory responses or cytokine storm associated with viral infections results in deleterious consequences such as vascular leakage, severe hemorrhage, shock, immune paralysis, multi-organ failure, and even death. With the emerging new viral infections and lack of effective prophylactic vaccines, evidence-based complementary strategies that limit viral infection-mediated hyperinflammatory responses could be a promising approach to limit host tissue injury. The present review emphasizes the potentials of antiinflammatory phytochemicals in limiting hyperinflammatory injury caused by viral infections. The predominant phytochemicals along with their mechanism in limiting hyperimmune and pro-inflammatory responses under viral infection have been reviewed comprehensively. How certain phytochemicals can be effective in limiting hyper-inflammatory response indirectly by favorably modulating gut microbiota and maintaining a functional intestinal barrier has also been presented. Finally, we have discussed improved systemic bioavailability of phytochemicals, efficient delivery strategies, and safety measures for effective antiinflammatory phytotherapies, in addition to emphasizing the requirement of tightly controlled clinical studies to establish the antiinflammatory efficacy of the phytochemicals. Collectively, the review provides a scooping overview on the potentials of bioactive phytochemicals to mitigate pro-inflammatory injury associated with viral infections.
Collapse
Affiliation(s)
- Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Yogita Gupta
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Suktilang Majaw
- Department of Biotechnology & Bioinformatics, North-Eastern Hill University, Shillong, India
| |
Collapse
|
11
|
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: 51] [Impact Index Per Article: 10.2] [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
|
12
|
Antiviral Activities of Silymarin and Derivatives. Molecules 2019; 24:molecules24081552. [PMID: 31010179 PMCID: PMC6514695 DOI: 10.3390/molecules24081552] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/22/2022] Open
Abstract
Silymarin flavonolignans are well-known agents that typically possess antioxidative, anti-inflammatory, and hepatoprotective functions. Recent studies have also documented the antiviral activities of silymarin and its derivatives against several viruses, including the flaviviruses (hepatitis C virus and dengue virus), togaviruses (Chikungunya virus and Mayaro virus), influenza virus, human immunodeficiency virus, and hepatitis B virus. This review will describe some of the latest preclinical and clinical studies detailing the antiviral profiles of silymarin and its derivatives, and discuss their relevance for antiviral drug development.
Collapse
|
13
|
Keyhanmehr N, Motedayyen H, Eskandari N. The Effects of Silymarin and Cyclosporine A on the Proliferation and Cytokine Production of Regulatory T Cells. Immunol Invest 2019; 48:533-548. [DOI: 10.1080/08820139.2019.1571506] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Niloufar Keyhanmehr
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
14
|
Romanucci V, Di Fabio G, Zarrelli A. A New Class of Synthetic Flavonolignan-Like Dimers: Still Few Molecules, but with Attractive Properties. Molecules 2018; 24:E108. [PMID: 30597952 PMCID: PMC6337569 DOI: 10.3390/molecules24010108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
In recent years, there has been increasing interest in dimeric molecules due to reports of their promising therapeutic value in the treatment of numerous diseases (such as cancer, HIV, Alzheimer's and, malaria). Many reports in the literature have highlighted the ability of these molecules to interact not only with specific biologic receptors but also to induce a biological response that more than doubles the results of the corresponding monomeric counterpart. In this regard, flavonolignan dimers or simply bi-flavonolignans are an emerging class of dimeric compounds that unlike bi-flavonoids, which are very widespread in nature, consist of synthetic dimers of some flavonolignans isolated from the milk thistle Silybum marianum [L. Gaertn. (Asteraceae)]. This mini-review will discuss recent developments in the synthesis, characterization and antioxidant activity of new families of flavonolignan dimers, in light of emerging medicinal chemistry strategies.
Collapse
Affiliation(s)
- Valeria Romanucci
- Department of Chemical Sciences, University of Napoli 'Federico II', Via Cintia 4, I-80126 Napoli, Italy.
| | - Giovanni Di Fabio
- Department of Chemical Sciences, University of Napoli 'Federico II', Via Cintia 4, I-80126 Napoli, Italy.
| | - Armando Zarrelli
- Department of Chemical Sciences, University of Napoli 'Federico II', Via Cintia 4, I-80126 Napoli, Italy.
| |
Collapse
|
15
|
Shen X, Liu H, Li G, Deng X, Wang J. Silibinin attenuates Streptococcus suis serotype 2 virulence by targeting suilysin. J Appl Microbiol 2018; 126:435-442. [PMID: 30408277 DOI: 10.1111/jam.14149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/15/2018] [Accepted: 11/02/2018] [Indexed: 12/20/2022]
Abstract
AIMS To determine the antivirulence properties of silibinin against suilysin (SLY), a virulence factor of Streptococcus suis serotype 2 (SS2) that plays an important role in the pathogenesis of S. suis infection and its protective effect against SS2 infection in a mouse model. METHODS AND RESULTS Susceptibility testing, haemolysis assay and Western blot assays were employed to evaluate the performance of silibinin on SLY pore-forming activity. Cytotoxicity assays and mouse infection tests were also performed to determine the efficacy of silibinin against SS2 infection. The results showed that silibinin, a flavonoid with little anti-S. suis activity, was identified to be a potent antagonist of SLY-mediated haemolysis through the inhibition of its oligomerization. Treatment with silibinin reduced S. suis-induced cytotoxicity in macrophages (J774 cells). In addition, S. suis-infected mice that received silibinin showed a lower bacterial burden. CONCLUSIONS Our results demonstrated that silibinin is a promising candidate for the development of antivirulence therapeutic agents to treat S. suis infections. SIGNIFICANCE AND IMPACT OF THE STUDY The antivirulent property of silibinin against SS2 by targeting SLY provides the possibility for the future pharmaceutical application of silibinin to prevent and treat S. suis infection.
Collapse
Affiliation(s)
- X Shen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - H Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - G Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - X Deng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - J Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
16
|
Kyriakopoulos G, Tsaroucha AK, Valsami G, Lambropoulou M, Kostomitsopoulos N, Christodoulou E, Kakazanis Z, Anagnostopoulos C, Tsalikidis C, Simopoulos CE. Silibinin Improves TNF-α and M30 Expression and Histological Parameters in Rat Kidneys After Hepatic Ischemia/Reperfusion. J INVEST SURG 2018; 31:201-209. [PMID: 28418711 DOI: 10.1080/08941939.2017.1308044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Remote kidney damage is a sequel of hepatic ischemia-reperfusion (I/R) injury. Silibinin is the main ingredient of the milk thistle plant seed extract with known antioxidant and hepatoprotective activity. Our study investigates the nephroprotective potential of intravenously administered silibinin, as a lyophilized SLB-hydoxypropyl-beta-cyclodextrin product, in hepatic I/R injury. MATERIAL AND METHODS 63 Wistar rats were divided into three groups: Sham (virtual intervention); Control (45 min ischemia and reperfusion); and Silibinin (200 μL intravenous silibinin administration after 45 min of ischemia). Kidney tissues were collected to determine TNF-α, M30 and histopathological changes at predetermined time intervals. RESULTS Comparing Sham vs. Control groups, proved that hepatic I/R injury increased renal TNF-α and M30 expression. Deterioration was observed in hyperemia/filtration of renal parenchyma and tubules, cortical filtration, tubular necrosis and edema (tissue swelling index). Intravenous silibinin administration and comparison of the Control vs. Silibinin groups showed a statistically significant decrease in TNF-α levels at 240 min following I/R (p < 0.0001), and in M30 at 180 min (p = 0.03) and 240 min (p < 0.0001). Renal parameters have significantly decreased in: hyperemia/filtration of renal parenchyma at 120 min (p = 0.003), 180 min (p = 0.0001) and 240 min (p = 0.0002); hyperemia/filtration of renal tubules at 120 min (p = 0.02), 180 min (p = 0.0001) and 240 min (p = 0.0005); cortical filtration (240 min - p = 0.005); tubular necrosis (240 min - p = 0.021); and edema (240 min - p = 0.001). CONCLUSION Our study confirms that hepatic I/R injury causes remote renal damage while the intravenous administration of silibinin leads to statistically significant nephroprotective action.
Collapse
Affiliation(s)
- Georgios Kyriakopoulos
- a Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine , Democritus University of Thrace , Alexandroupolis , Greece
| | - Alexandra K Tsaroucha
- a Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine , Democritus University of Thrace , Alexandroupolis , Greece
- b 2nd Department of Surgery and Laboratory of Experimental Surgery, Faculty of Medicine , Democritus University of Thrace , Alexandroupolis , Greece
| | - Georgia Valsami
- c School of Health Sciences, Department of Pharmacy , National and Kapodistrian University of Athens , Greece
| | - Maria Lambropoulou
- d Laboratory of Histology-Embryology, Faculty of Medicine , Democritus University of Thrace , Alexandroupolis , Greece
| | - Nikolaos Kostomitsopoulos
- e Department of Experimental Surgery , Bioresearch Foundation of the Academy of Athens , Athens , Greece
| | - Eirini Christodoulou
- c School of Health Sciences, Department of Pharmacy , National and Kapodistrian University of Athens , Greece
| | - Zacharias Kakazanis
- e Department of Experimental Surgery , Bioresearch Foundation of the Academy of Athens , Athens , Greece
| | | | - Christos Tsalikidis
- a Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine , Democritus University of Thrace , Alexandroupolis , Greece
| | - Constantinos E Simopoulos
- a Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine , Democritus University of Thrace , Alexandroupolis , Greece
- b 2nd Department of Surgery and Laboratory of Experimental Surgery, Faculty of Medicine , Democritus University of Thrace , Alexandroupolis , Greece
- e Department of Experimental Surgery , Bioresearch Foundation of the Academy of Athens , Athens , Greece
| |
Collapse
|
17
|
Silibinin phosphodiester glyco-conjugates: Synthesis, redox behaviour and biological investigations. Bioorg Chem 2018; 77:349-359. [PMID: 29421711 DOI: 10.1016/j.bioorg.2018.01.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 01/13/2023]
Abstract
New silibinin phosphodiester glyco-conjugates were synthesized by efficient phosphoramidite chemistry and were fully characterized by 2D-NMR. A wide-ranging study focused on the determination of their pKa and E° values as well as on their radical scavenging activities by different assays (DPPH, ABTS+ and HRSA) was conducted. The new glyco-conjugates are more water-soluble than silibinin, and their radical scavenging activities are higher than those of silibinin. The conjugation therefore improves both the water solubilities and antioxidant activities of the flavonolignan moieties. The serum stability was evaluated under physiological conditions, and the glyco-conjugates degraded with half-lives of 40-70 h, making them useful in pro-drug approaches. We started by treating androgen-dependent prostate cancer (PCa) LNCaP cells and then expanded our studies to androgen-independent PCa PC3 and DU145 cells. In most cases, the new derivatives significantly reduced both total and live cell numbers, albeit at different levels. Anti-HIV activities were evaluated and the glucosamine-phosphate silibinin derivative showed higher activity (IC50 = 73 μM) than silibinin.
Collapse
|
18
|
Kumar V, Ahmad A. Role of MAIT cells in the immunopathogenesis of inflammatory diseases: New players in old game. Int Rev Immunol 2017; 37:90-110. [PMID: 29106304 DOI: 10.1080/08830185.2017.1380199] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Current advances in immunology have led to the identification of a population of novel innate immune T cells, called mucosa-associated invariant T (MAIT) cells. The cells in humans express an invariant TCRα chain (Vα7.2-Jα33) paired with a limited subset of TCRβ chains (Vβ2, 13 and 22), are restricted by the MHC class I (MH1)-related (MR)-1, and recognize molecules that are produced in the bacterial riboflavin (vitamin B2) biosynthetic pathway. They are present in the circulation, liver and at various mucosal sites (i.e. intestine, lungs and female reproductive tract, etc.). They kill host cells infected with bacteria and yeast, and secrete soluble mediators such as TNF-α, IFN-γ, IL-17, etc. The cells regulate immune responses and inflammation associated with a wide spectrum of acute and chronic diseases in humans. Since their discovery in 1993, significant advances have been made in understanding biology of MAIT cells and the potential role of these cells in the pathogenesis of autoimmune, inflammatory and infectious diseases as well as cancer in humans. The purpose of this review is to provide a current state of our knowledge about MAIT cell biology and delineate their role in autoimmune and inflammatory diseases (sterile or caused by infectious agents) and cancer in humans. A better understanding of the role of MAIT cells in human diseases may lead to novel ways of immunotherapies.
Collapse
Affiliation(s)
- Vijay Kumar
- a Department of Paediatrics and Child Care , Children's Health Queensland Clinical unit School of Medicine, Mater Research, Faculty of Medicine and Biomedical Sciences, University of Queensland , ST Lucia, Brisbane , Queensland , Australia
| | - Ali Ahmad
- b Laboratory of Innate Immunity, CHU Ste-Justine/Department of Microbiology , Infectious Diseases & Immunology, University of Montreal , Montreal , Quebec , Canada
| |
Collapse
|
19
|
Lovelace ES, Maurice NJ, Miller HW, Slichter CK, Harrington R, Magaret A, Prlic M, De Rosa S, Polyak SJ. Silymarin suppresses basal and stimulus-induced activation, exhaustion, differentiation, and inflammatory markers in primary human immune cells. PLoS One 2017; 12:e0171139. [PMID: 28158203 PMCID: PMC5291532 DOI: 10.1371/journal.pone.0171139] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/15/2017] [Indexed: 12/13/2022] Open
Abstract
Silymarin (SM), and its flavonolignan components, alter cellular metabolism and inhibit inflammatory status in human liver and T cell lines. In this study, we hypothesized that SM suppresses both acute and chronic immune activation (CIA), including in the context of HIV infection. SM treatment suppressed the expression of T cell activation and exhaustion markers on CD4+ and CD8+ T cells from chronically-infected, HIV-positive subjects. SM also showed a trend towards modifying CD4+ T cell memory subsets from HIV+ subjects. In the HIV-negative setting, SM treatment showed trends towards suppressing pro-inflammatory cytokines from non-activated and pathogen-associated molecular pattern (PAMP)-activated primary human monocytes, and non-activated and cytokine- and T cell receptor (TCR)-activated mucosal-associated invariant T (MAIT) cells. The data suggest that SM elicits broad anti-inflammatory and immunoregulatory activity in primary human immune cells. By using novel compounds to alter cellular inflammatory status, it may be possible to regulate inflammation in both non-disease and disease states.
Collapse
Affiliation(s)
- Erica S. Lovelace
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America
| | - Nicholas J. Maurice
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Hannah W. Miller
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Chloe K. Slichter
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
| | - Robert Harrington
- Division of Allergy and Infectious Disease, University of Washington, Seattle, WA, United States of America
| | - Amalia Magaret
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Martin Prlic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
| | - Stephen De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Stephen J. Polyak
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
20
|
Lovelace ES, Polyak SJ. Natural Products as Tools for Defining How Cellular Metabolism Influences Cellular Immune and Inflammatory Function during Chronic Infection. Viruses 2015; 7:6218-32. [PMID: 26633463 PMCID: PMC4690857 DOI: 10.3390/v7122933] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/13/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022] Open
Abstract
Chronic viral infections like those caused by hepatitis C virus (HCV) and human immunodeficiency virus (HIV) cause disease that establishes an ongoing state of chronic inflammation. While there have been tremendous improvements towards curing HCV with directly acting antiviral agents (DAA) and keeping HIV viral loads below detection with antiretroviral therapy (ART), there is still a need to control inflammation in these diseases. Recent studies indicate that many natural products like curcumin, resveratrol and silymarin alter cellular metabolism and signal transduction pathways via enzymes such as adenosine monophosphate kinase (AMPK) and mechanistic target of rapamycin (mTOR), and these pathways directly influence cellular inflammatory status (such as NF-κB) and immune function. Natural products represent a vast toolkit to dissect and define how cellular metabolism controls cellular immune and inflammatory function.
Collapse
Affiliation(s)
- Erica S Lovelace
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Stephen J Polyak
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA.
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA.
- Department of Global Health, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
21
|
Borgmann K, Ghorpade A. HIV-1, methamphetamine and astrocytes at neuroinflammatory Crossroads. Front Microbiol 2015; 6:1143. [PMID: 26579077 PMCID: PMC4621459 DOI: 10.3389/fmicb.2015.01143] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022] Open
Abstract
As a popular psychostimulant, methamphetamine (METH) use leads to long-lasting, strong euphoric effects. While METH abuse is common in the general population, between 10 and 15% of human immunodeficiency virus-1 (HIV-1) patients report having abused METH. METH exacerbates the severity and onset of HIV-1-associated neurocognitive disorders (HAND) through direct and indirect mechanisms. Repetitive METH use impedes adherence to antiretroviral drug regimens, increasing the likelihood of HIV-1 disease progression toward AIDS. METH exposure also directly affects both innate and adaptive immunity, altering lymphocyte numbers and activity, cytokine signaling, phagocytic function and infiltration through the blood brain barrier. Further, METH triggers the dopamine reward pathway and leads to impaired neuronal activity and direct toxicity. Concurrently, METH and HIV-1 alter the neuroimmune balance and induce neuroinflammation, which modulates a wide range of brain functions including neuronal signaling and activity, glial activation, viral infection, oxidative stress, and excitotoxicity. Pathologically, reactive gliosis is a hallmark of both HIV-1- and METH-associated neuroinflammation. Significant commonality exists in the neurotoxic mechanisms for both METH and HAND; however, the pathways dysregulated in astroglia during METH exposure are less clear. Thus, this review highlights alterations in astrocyte intracellular signaling pathways, gene expression and function during METH and HIV-1 comorbidity, with special emphasis on HAND-associated neuroinflammation. Importantly, this review carefully evaluates interventions targeting astrocytes in HAND and METH as potential novel therapeutic approaches. This comprehensive overview indicates, without a doubt, that during HIV-1 infection and METH abuse, a complex dialog between all neural cells is orchestrated through astrocyte regulated neuroinflammation.
Collapse
Affiliation(s)
- Kathleen Borgmann
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Anuja Ghorpade
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| |
Collapse
|
22
|
Wang Y, Liang WC, Pan WL, Law WK, Hu JS, Ip DTM, Waye MMY, Ng TB, Wan DCC. Silibinin, a novel chemokine receptor type 4 antagonist, inhibits chemokine ligand 12-induced migration in breast cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:1310-1317. [PMID: 25172795 DOI: 10.1016/j.phymed.2014.06.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/15/2014] [Accepted: 06/27/2014] [Indexed: 06/03/2023]
Abstract
PURPOSE C-X-C chemokine receptor type 4 (CXCR4) signaling has been demonstrated to be involved in cancer invasion and migration; therefore, CXCR4 antagonist can serve as an anti-cancer drug by preventing tumor metastasis. This study aimed to identify the CXCR4 antagonists that could reduce and/or inhibit tumor metastasis from natural products. METHODS AND RESULTS According to the molecular docking screening, we reported here silibinin as a novel CXCR4 antagonist. Biochemical characterization showed that silibinin blocked chemokine ligand 12 (CXCL12)-induced CXCR4 internalization by competitive binding to CXCR4, therefore inhibiting downstream intracellular signaling. In human breast cancer cells MDA-MB-231, which expresses high levels of CXCR4, inhibition of CXCL12-induced chemomigration can be found under silibinin treatment. Overexpression of CXCL12 sensitized MDA-MB-231 cells to the inhibition of silibinin, which was abolished by CXCR4 knockdown. The inhibition of silibinin was also observed in MCF-7/CXCR4 cells rather than MCF-7 cells that express low level of CXCR4. CONCLUSIONS Our work demonstrated that silibinin is a novel CXCR4 antagonist that may have potential therapeutic use for prevention of tumor metastasis.
Collapse
Affiliation(s)
- Yan Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Wei-Cheng Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Wen-Liang Pan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Wai-Kit Law
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Jian-Shu Hu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Denis Tsz-Ming Ip
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Mary Miu-Yee Waye
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Tzi-Bun Ng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - David Chi-Cheong Wan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
23
|
Lim R, Morwood CJ, Barker G, Lappas M. Effect of silibinin in reducing inflammatory pathways in in vitro and in vivo models of infection-induced preterm birth. PLoS One 2014; 9:e92505. [PMID: 24647589 PMCID: PMC3960267 DOI: 10.1371/journal.pone.0092505] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/21/2014] [Indexed: 12/21/2022] Open
Abstract
Infection-induced preterm birth is the largest cause of infant death and of neurological disabilities in survivors. Silibinin, from milk thistle, exerts potent anti-inflammatory activities in non-gestational tissues. The aims of this study were to determine the effect of silibinin on pro-inflammatory mediators in (i) human fetal membranes and myometrium treated with bacterial endotoxin lipopolysaccharide (LPS) or the pro-inflammatory cytokine IL-1β, and (ii) in preterm fetal membranes with active infection. The effect of silibinin on infection induced inflammation and brain injury in pregnant mice was also assessed. Fetal membranes and myometrium (tissue explants and primary cells) were treated with 200 μM silibinin in the presence or absence of 10 μg/ml LPS or 1 ng/ml IL-1β. C57BL/6 mice were injected with 70 mg/kg silibinin with or without 50 μg LPS on embryonic day 16. Fetal brains were collected after 6 h. In human fetal membranes, silibinin significantly decreased LPS-stimulated expression of IL-6 and IL-8, COX-2, and prostaglandins PGE2 and PGF2α. In primary amnion and myometrial cells, silibinin also decreased IL-1β-induced MMP-9 expression. Preterm fetal membranes with active infection treated with silibinin showed a decrease in IL-6, IL-8 and MMP-9 expression. Fetal brains from mice treated with silibinin showed a significant decrease in LPS-induced IL-8 and ninjurin, a marker of brain injury. Our study demonstrates that silibinin can reduce infection and inflammation-induced pro-labour mediators in human fetal membranes and myometrium. Excitingly, the in vivo results indicate a protective effect of silibinin on infection-induced brain injury in a mouse model of preterm birth.
Collapse
Affiliation(s)
- Ratana Lim
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Carrington J. Morwood
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Gillian Barker
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Martha Lappas
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
24
|
McClure J, Margineantu DH, Sweet IR, Polyak SJ. Inhibition of HIV by Legalon-SIL is independent of its effect on cellular metabolism. Virology 2014; 449:96-103. [PMID: 24418542 PMCID: PMC3909448 DOI: 10.1016/j.virol.2013.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 09/22/2013] [Accepted: 11/01/2013] [Indexed: 01/18/2023]
Abstract
In this report, we further characterized the effects of silibinin (SbN), derived from milk thistle extract, and Legalon-SIL (SIL), a water-soluble derivative of SbN, on T cell metabolism and HIV infection. We assessed the effects of SbN and SIL on peripheral blood mononuclear cells (PBMC) and CEM-T4 cells in terms of cellular growth, ATP content, metabolism, and HIV infection. SIL and SbN caused a rapid and reversible (upon removal) decrease in cellular ATP levels, which was associated with suppression of mitochondrial respiration and glycolysis. SbN, but not SIL inhibited glucose uptake. Exposure of T cells to SIL (but not SbN or metabolic inhibitors) during virus adsorption blocked HIV infection. Thus, both SbN and SIL rapidly perturb T cell metabolism in vitro, which may account for its anti-inflammatory and anti-proliferative effects that arise with prolonged exposure of cells. However, the metabolic effects are not involved in SIL's unique ability to block HIV entry.
Collapse
Affiliation(s)
- Janela McClure
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States
| | - Daciana H Margineantu
- Department of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Ian R Sweet
- Department of Medicine (Division of Metabolism, Endocrinology, and Nutrition), University of Washington, Seattle, WA, United States
| | - Stephen J Polyak
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States; Department of Global Health, University of Washington, Seattle, WA, United States.
| |
Collapse
|
25
|
Blaising J, Lévy PL, Gondeau C, Phelip C, Varbanov M, Teissier E, Ruggiero F, Polyak SJ, Oberlies NH, Ivanovic T, Boulant S, Pécheur EI. Silibinin inhibits hepatitis C virus entry into hepatocytes by hindering clathrin-dependent trafficking. Cell Microbiol 2013; 15:1866-82. [PMID: 23701235 DOI: 10.1111/cmi.12155] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 05/01/2013] [Accepted: 05/20/2013] [Indexed: 12/17/2022]
Abstract
Hepatitis C virus (HCV) is a global health concern infecting 170 million people worldwide. Previous studies indicate that the extract from milk thistle known as silymarin and its main component silibinin inhibit HCV infection. Here we investigated the mechanism of anti-HCV action of silymarin-derived compounds at the molecular level. By using live-cell confocal imaging, single particle tracking, transmission electron microscopy and biochemical approaches on HCV-infected human hepatoma cells and primary hepatocytes, we show that silibinin potently inhibits HCV infection and hinders HCV entry by slowing down trafficking through clathrin-coated pits and vesicles. Detailed analyses revealed that silibinin altered the formation of both clathrin-coated pits and vesicles in cells and caused abnormal uptake and trafficking of transferrin, a well-known cargo of the clathrin endocytic pathway. Silibinin also inhibited infection by other viruses that enter cells by clathrin-mediated endocytosis including reovirus, vesicular stomatitis and influenza viruses. Our study demonstrates that silibinin inhibits HCV early steps of infection by affecting endosomal trafficking of virions. It provides new insights into the molecular mechanisms of action of silibinin against HCV entry and also suggests that silibinin is a potential broad-spectrum antiviral therapy.
Collapse
Affiliation(s)
- Julie Blaising
- UMR CNRS 5086, IBCP, Lyon, France; UMR Inserm U1052/CNRS 5286, Cancer Research Center of Lyon, University of Lyon, Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Dai JP, Wu LQ, Li R, Zhao XF, Wan QY, Chen XX, Li WZ, Wang GF, Li KS. Identification of 23-(s)-2-amino-3-phenylpropanoyl-silybin as an antiviral agent for influenza A virus infection in vitro and in vivo. Antimicrob Agents Chemother 2013; 57:4433-43. [PMID: 23836164 PMCID: PMC3754338 DOI: 10.1128/aac.00759-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/26/2013] [Indexed: 02/05/2023] Open
Abstract
It has been reported that autophagy is involved in the replication of many viruses. In this study, we screened 89 medicinal plants, using an assay based on the inhibition of the formation of the Atg12-Atg5/Atg16 heterotrimer, an important regulator of autophagy, and selected Silybum marianum L. for further study. An antiviral assay indicated that silybin (S0), the major active compound of S. marianum L., can inhibit influenza A virus (IAV) infection. We later synthesized 5 silybin derivatives (S1 through S5) and found that 23-(S)-2-amino-3-phenylpropanoyl-silybin (S3) had the best activity. When we compared the polarities of the substituent groups, we found that the hydrophobicity of the substituent groups was positively correlated with their activities. We further studied the mechanisms of action of these compounds and determined that S0 and S3 also inhibited both the formation of the Atg12-Atg5/Atg16 heterotrimer and the elevated autophagy induced by IAV infection. In addition, we found that S0 and S3 could inhibit several components induced by IAV infection, including oxidative stress, the activation of extracellular signal-regulated kinase (ERK)/p38 mitogen-activated protein kinase (MAPK) and IκB kinase (IKK) pathways, and the expression of autophagic genes, especially Atg7 and Atg3. All of these components have been reported to be related to the formation of the Atg12-Atg5/Atg16 heterotrimer, which might validate our screening strategy. Finally, we demonstrated that S3 can significantly reduce influenza virus replication and the associated mortality in infected mice. In conclusion, we identified 23-(S)-2-amino-3-phenylpropanoyl-silybin as a promising inhibitor of IAV infection.
Collapse
Affiliation(s)
- Jian-Ping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Li-Qi Wu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Rui Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Xiang-Feng Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Qian-Ying Wan
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Xiao-Xuan Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Wei-Zhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Ge-Fei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Kang-Sheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| |
Collapse
|
27
|
Polyak SJ, Ferenci P, Pawlotsky JM. Hepatoprotective and antiviral functions of silymarin components in hepatitis C virus infection. Hepatology 2013; 57:1262-1271. [PMID: 23213025 PMCID: PMC3594650 DOI: 10.1002/hep.26179] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 09/05/2012] [Indexed: 12/21/2022]
Affiliation(s)
- Stephen J Polyak
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98104, USA.
| | | | | |
Collapse
|
28
|
Qiu LP, Chen KP. Anti-HBV agents derived from botanical origin. Fitoterapia 2012; 84:140-57. [PMID: 23164603 DOI: 10.1016/j.fitote.2012.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 10/29/2012] [Accepted: 11/04/2012] [Indexed: 01/16/2023]
Abstract
There are 350,000 hepatitis B virus (HBV) carriers all over the world. Chronic HBV infection is at a high risk of developing liver cirrhosis and hepatocelluar carcinoma (HCC), and heavily threatened people's health. Two kinds of drugs approved by FDA for anti-HBV therapy are immunomodulators (interferon α, pegylated-interferon α) and nucleos(t)ide analogues (lamivudine, adefovir dipivoxil, entecavir, telbivudine, and tenofovir disoproxil fumarate). These drugs have been proved to be far from being satisfactory due to their low specificity, side effects, and high rate of drug resistance. There is an urgent need to discover and develop novel effective anti-HBV drugs. With vast resources, various structures, diverse biological activities and action mechanisms, as well as abundant clinical experiences, botanical agents become a promising source of finding new anti-HBV drugs. This review summarizes the recent research and development of anti-HBV agents derived from botanical origin on their sources and active components, inhibitory effects and possible toxicities, as well as action targets and mechanisms, and also addresses the advantages and the existing shortcomings in the development of botanical inhibitors. This information may not only broaden the knowledge of anti-HBV therapy, and offer possible alternative or substitutive drugs for CHB patients, but also provides considerable information for developing new safe and effective anti-HBV drugs.
Collapse
Affiliation(s)
- Li-Peng Qiu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | | |
Collapse
|
29
|
Polyak SJ, Oberlies NH, Pécheur EI, Dahari H, Ferenci P, Pawlotsky JM. Silymarin for HCV infection. Antivir Ther 2012; 18:141-147. [PMID: 23011959 PMCID: PMC4076489 DOI: 10.3851/imp2402] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2012] [Indexed: 12/11/2022]
Abstract
Silymarin, an extract of milk thistle seeds, and silymarin-derived compounds have been considered hepatoprotective since the plant was first described in ancient times. Hepatoprotection is defined as several non-mutually exclusive biological activities including antiviral, antioxidant, anti-inflammatory and immunomodulatory functions. Despite clear evidence for silymarin-induced hepatoprotection in cell culture and animal models, evidence for beneficial effects in humans has been equivocal. This review will summarize the current state of knowledge on silymarin in the context of HCV infection. The information was collated from a recent workshop on silibinin in Germany.
Collapse
Affiliation(s)
- Stephen J Polyak
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.
| | | | | | | | | | | |
Collapse
|