1
|
Libin KV, Debnath M, Sisodiya S, Rathod SB, Prajapati PB, Lisina KV, Bhuyan R, Evanjelene VK. Bioefficacy, chromatographic profiling and drug-likeness analysis of flavonoids and terpenoids as potential inhibitors of H1N1 influenza viral proteins. Int J Biol Macromol 2024; 281:136125. [PMID: 39357733 DOI: 10.1016/j.ijbiomac.2024.136125] [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: 01/16/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Considering medicinal plants, natural products present in these plants are the best sources of medications for combating viral infection. The possible drug target against viral H1N1 influenza proteins lead to identification of selected secondary metabolites from potential plants Tinospora cordifolia, Ocimum sanctum, and Piper nigrum. On analysis of in vitro cell based antiviral activity of the selected plant extracts, an indication for a possible lead compound against neuraminidase activity was evident. Potent ligands were selected using drug docking and ADMET analysis, and the screened lead metabolites were ultimately identified as terpenoid (Columbin) and, flavonoid (Cubebin, and Apigenin). Among the selected ligands, the drug binding activity of Cubebin with all the 6 proteins of H1N1 influenza type A virus, HA (4r8w), NA (4qn7), M2 (3lbw), PA (4wsb), PB1 (2znl) and PB2 (3wil), was pronounced. In addition, physicochemical and pharmacokinetic parameters linked to absorption, distribution, metabolism, excretion and toxicity (ADMET) have been evaluated and corroborate with our in vitro results. Molecular dynamics modelling indicated Cubebin can be a potential phytochemical in a drug discovery pipeline for the development of neuraminidase inhibitors. Further studies can provide a possibility for an alternative therapy against Influenza viruses.
Collapse
Affiliation(s)
- K V Libin
- Department of Biosciences and Biotechnology, Banasthali Vidyapith Jaipur, Rajasthan 304802, India
| | - Mousumi Debnath
- Department of Biosciences, Manipal University Jaipur, Rajasthan 303007, India.
| | - Smita Sisodiya
- Department of Biosciences, Manipal University Jaipur, Rajasthan 303007, India
| | - Shravan B Rathod
- Department of Chemistry, Smt. S. M. Panchal Science College, Talod, Gujarat, India
| | - Pravin B Prajapati
- Department of Chemistry, Sheth M. N. Science College, Patan, Gujarat, India
| | - K V Lisina
- Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Rajabrata Bhuyan
- Department of Biosciences and Biotechnology, Banasthali Vidyapith Jaipur, Rajasthan 304802, India
| | | |
Collapse
|
2
|
Wei Y, Liu H, Hu D, He Q, Yao C, Li H, Hu K, Wang J. Recent Advances in Enterovirus A71 Infection and Antiviral Agents. J Transl Med 2024; 104:100298. [PMID: 38008182 DOI: 10.1016/j.labinv.2023.100298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/29/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023] Open
Abstract
Enterovirus A71 (EV-A71) is one of the major causative agents of hand, foot, and mouth disease (HFMD) that majorly affects children. Most of the time, HFMD is a mild disease but can progress to severe complications, such as meningitis, brain stem encephalitis, acute flaccid paralysis, and even death. HFMD caused by EV-A71 has emerged as an acutely infectious disease of highly pathogenic potential in the Asia-Pacific region. In this review, we introduced the properties and life cycle of EV-A71, and the pathogenesis and the pathophysiology of EV-A71 infection, including tissue tropism and host range of virus infection, the diseases caused by the virus, as well as the genes and host cell immune mechanisms of major diseases caused by enterovirus 71 (EV-A71) infection, such as encephalitis and neurologic pulmonary edema. At the same time, clinicopathologic characteristics of EV-A71 infection were introduced. There is currently no specific medication for EV-A71 infection, highlighting the urgency and significance of developing suitable anti-EV-A71 agents. This overview also summarizes the targets of existing anti-EV-A71 agents, including virus entry, translation, polyprotein processing, replication, assembly and release; interferons; interleukins; the mitogen-activated protein kinase, phosphatidylinositol 3-kinase, and protein kinase B signaling pathways; the oxidative stress pathway; the ubiquitin-proteasome system; and so on. Furthermore, it overviews the effects of natural products, monoclonal antibodies, and RNA interference against EV-A71. It also discusses issues limiting the research of antiviral drugs. This review is a systematic and comprehensive summary of the mechanism and pathological characteristics of EV-A71 infection, the latest progress of existing anti-EV-A71 agents. It would provide better understanding and guidance for the research and application of EV-A71 infection and antiviral inhibitors.
Collapse
Affiliation(s)
- Yanhong Wei
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Huihui Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Da Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Qun He
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Chenguang Yao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Hanluo Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Kanghong Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China.
| | - Jun Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Bjørklund G, Lysiuk R, Semenova Y, Lenchyk L, Dub N, Doşa MD, Hangan T. Herbal Substances with Antiviral Effects: Features and Prospects for the Treatment of Viral Diseases with Emphasis on Pro-Inflammatory Cytokines. Curr Med Chem 2024; 31:393-409. [PMID: 36698239 DOI: 10.2174/0929867330666230125121758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 01/26/2023]
Abstract
Viral diseases have a significant impact on human health, and three novel coronaviruses (CoV) have emerged during the 21st century. In this review, we have emphasized the potential of herbal substances with antiviral effects. Our investigation focused on the features and prospects of viral disease treatment, with a particular emphasis on proinflammatory cytokines. We conducted comprehensive searches of various databases, including Science Direct, CABI Direct, Web of Science, PubMed, and Scopus. Cytokine storm mechanisms play a crucial role in inducing a pro-inflammatory response by triggering the expression of cytokines and chemokines. This response leads to the recruitment of leukocytes and promotes antiviral effects, forming the first line of defense against viruses. Numerous studies have investigated the use of herbal medicine candidates as immunomodulators or antivirals. However, cytokine-storm-targeted therapy is recommended for patients with acute respiratory distress syndrome caused by SARS-CoV to survive severe pulmonary failure. Our reviews have demonstrated that herbal formulations could serve as alternative medicines and significantly reduce complicated viral infections. Furthermore, they hold promising potential as specific antiviral agents in experimental animal models.
Collapse
Affiliation(s)
- Geir Bjørklund
- Department of Research, Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Roman Lysiuk
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Yuliya Semenova
- School of Medicine, Nazarbayev University , Astana, Kazakhstan
| | - Larysa Lenchyk
- Department of Research, National University of Pharmacy, Kharkiv, Ukraine
- CONEM Ukraine Pharmacognosy and Natural Product Chemistry Research Group, National University of Pharmacy, Kharkiv, Ukraine
| | - Natalia Dub
- Andrei Krupynskyi Lviv Medical Academy, Lviv, Ukraine
| | | | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| |
Collapse
|
4
|
Gabbianelli R, Shahar E, de Simone G, Rucci C, Bordoni L, Feliziani G, Zhao F, Ferrati M, Maggi F, Spinozzi E, Mahajna J. Plant-Derived Epi-Nutraceuticals as Potential Broad-Spectrum Anti-Viral Agents. Nutrients 2023; 15:4719. [PMID: 38004113 PMCID: PMC10675658 DOI: 10.3390/nu15224719] [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: 10/17/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Although the COVID-19 pandemic appears to be diminishing, the emergence of SARS-CoV-2 variants represents a threat to humans due to their inherent transmissibility, immunological evasion, virulence, and invulnerability to existing therapies. The COVID-19 pandemic affected more than 500 million people and caused over 6 million deaths. Vaccines are essential, but in circumstances in which vaccination is not accessible or in individuals with compromised immune systems, drugs can provide additional protection. Targeting host signaling pathways is recommended due to their genomic stability and resistance barriers. Moreover, targeting host factors allows us to develop compounds that are effective against different viral variants as well as against newly emerging virus strains. In recent years, the globe has experienced climate change, which may contribute to the emergence and spread of infectious diseases through a variety of factors. Warmer temperatures and changing precipitation patterns can increase the geographic range of disease-carrying vectors, increasing the risk of diseases spreading to new areas. Climate change may also affect vector behavior, leading to a longer breeding season and more breeding sites for disease vectors. Climate change may also disrupt ecosystems, bringing humans closer to wildlife that transmits zoonotic diseases. All the above factors may accelerate the emergence of new viral epidemics. Plant-derived products, which have been used in traditional medicine for treating pathological conditions, offer structurally novel therapeutic compounds, including those with anti-viral activity. In addition, plant-derived bioactive substances might serve as the ideal basis for developing sustainable/efficient/cost-effective anti-viral alternatives. Interest in herbal antiviral products has increased. More than 50% of approved drugs originate from herbal sources. Plant-derived compounds offer diverse structures and bioactive molecules that are candidates for new drug development. Combining these therapies with conventional drugs could improve patient outcomes. Epigenetics modifications in the genome can affect gene expression without altering DNA sequences. Host cells can use epigenetic gene regulation as a mechanism to silence incoming viral DNA molecules, while viruses recruit cellular epitranscriptomic (covalent modifications of RNAs) modifiers to increase the translational efficiency and transcript stability of viral transcripts to enhance viral gene expression and replication. Moreover, viruses manipulate host cells' epigenetic machinery to ensure productive viral infections. Environmental factors, such as natural products, may influence epigenetic modifications. In this review, we explore the potential of plant-derived substances as epigenetic modifiers for broad-spectrum anti-viral activity, reviewing their modulation processes and anti-viral effects on DNA and RNA viruses, as well as addressing future research objectives in this rapidly emerging field.
Collapse
Affiliation(s)
- Rosita Gabbianelli
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Ehud Shahar
- Department of Nutrition and Natural Products, Migal—Galilee Research Institute, Kiryat Shmona 11016, Israel;
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 1220800, Israel
| | - Gaia de Simone
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Chiara Rucci
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Laura Bordoni
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Giulia Feliziani
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Fanrui Zhao
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Marta Ferrati
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Filippo Maggi
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Eleonora Spinozzi
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal—Galilee Research Institute, Kiryat Shmona 11016, Israel;
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 1220800, Israel
| |
Collapse
|
5
|
Jadimurthy R, Jagadish S, Nayak SC, Kumar S, Mohan CD, Rangappa KS. Phytochemicals as Invaluable Sources of Potent Antimicrobial Agents to Combat Antibiotic Resistance. Life (Basel) 2023; 13:948. [PMID: 37109477 PMCID: PMC10145550 DOI: 10.3390/life13040948] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/04/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Plants have been used for therapeutic purposes against various human ailments for several centuries. Plant-derived natural compounds have been implemented in clinics against microbial diseases. Unfortunately, the emergence of antimicrobial resistance has significantly reduced the efficacy of existing standard antimicrobials. The World Health Organization (WHO) has declared antimicrobial resistance as one of the top 10 global public health threats facing humanity. Therefore, it is the need of the hour to discover new antimicrobial agents against drug-resistant pathogens. In the present article, we have discussed the importance of plant metabolites in the context of their medicinal applications and elaborated on their mechanism of antimicrobial action against human pathogens. The WHO has categorized some drug-resistant bacteria and fungi as critical and high priority based on the need to develope new drugs, and we have considered the plant metabolites that target these bacteria and fungi. We have also emphasized the role of phytochemicals that target deadly viruses such as COVID-19, Ebola, and dengue. Additionally, we have also elaborated on the synergetic effect of plant-derived compounds with standard antimicrobials against clinically important microbes. Overall, this article provides an overview of the importance of considering phytogenous compounds in the development of antimicrobial compounds as therapeutic agents against drug-resistant microbes.
Collapse
Affiliation(s)
- Ragi Jadimurthy
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | - Swamy Jagadish
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | - Siddaiah Chandra Nayak
- Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore 570006, India;
| | - Sumana Kumar
- Department of Microbiology, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysore 570015, India
| | - Chakrabhavi Dhananjaya Mohan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | | |
Collapse
|
6
|
Abd Aziz MF, Yip CW, Md Nor NS. In Silico and In Vitro Antiviral Activity Evaluation of Prodigiosin from Serratia marcescens Against Enterovirus 71. MALAYSIAN APPLIED BIOLOGY 2022; 51:113-128. [DOI: 10.55230/mabjournal.v51i5.2371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Prodigiosin, a red linear tripyrrole pigment found in Serratia marcescens, is one such naturally occurring compound that has gained wide attention owing to its numerous biological activities, including antibacterial, antifungal, antimalarial, anticancer, and immunosuppressive properties. This study was conducted to evaluate the possible antiviral activity of prodigiosin against Enterovirus 71, a causative agent of hand, foot, and mouth disease (HFMD). Preliminary studies were done in silico by analyzing the interaction of prodigiosin with amino acid residues of five EV71-target proteins. Interaction refinement analysis with FireDock revealed that 2C helicase (-48.01 kcal/moL) has the most negative global energy, followed by capsid (-36.52 kcal/moL), 3C protease (-34.16 kcal/moL), 3D RNA polymerase (-30.93 kcal/moL) and 2A protease (-20.61 kcal/moL). These values are indicative of the interaction strength. Prodigiosin was shown to form chemical bonds with specific amino acid residues in capsid (Gln-30, Asn-223), 2A protease (Trp-33, Trp-142), 2C helicase (Tyr-150, His-151, Gln-169, Ser-212), 3C protease (Glu-50), and 3D RNA polymerase (Ala-239, Tyr-237). To investigate further, prodigiosin was extracted from S. marcescens using a methanolic extraction method. In vitro studies revealed that prodigiosin, with an IC50 value of 0.5112 μg/mL, reduced virus titers by 0.17 log (32.39%) in 30 min and 0.19 log (35.43%) in 60 min. The findings suggest that prodigiosin has antiviral activity with an intermediate inhibitory effect against EV71. As a result of this research, new biological activities of prodigiosin have been identified.
Collapse
|
7
|
Zhou M, Liu Y, Cao J, Dong S, Hou Y, Yu Y, Zhang Q, Zhang Y, Jia X, Zhang B, Xiao G, Li G, Wang W. Bergamottin, a bioactive component of bergamot, inhibits SARS-CoV-2 infection in golden Syrian hamsters. Antiviral Res 2022; 204:105365. [PMID: 35732228 PMCID: PMC9212731 DOI: 10.1016/j.antiviral.2022.105365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 11/02/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has caused an ongoing pandemic, coronavirus disease-2019 (COVID-19), which has become a major global public health event. Antiviral compounds remain the predominant means of treating COVID-19. Here, we reported that bergamottin, a furanocoumarin originally found in bergamot, exhibited inhibitory activity against SARS-CoV-2 in vitro, ex vivo, and in vivo. Bergamottin interfered with multiple stages of virus life cycles, specifically blocking the SARS-CoV-2 spike-mediated membrane fusion and effectively reducing viral RNA synthesis. Oral delivery of bergamottin to golden Syrian hamsters at dosages of both 50 mg/kg and 75 mg/kg reduced the SARS-CoV-2 load in nasal turbinates and lung tissues. Pathological damage caused by viral infection was also ameliorated after bergamottin treatment. Overall, our study provides evidence of bergamottin as a promising natural compound, with broad-spectrum anti-coronavirus activity, that could be further developed in the fight against COVID-19 infection during the current pandemic.
Collapse
Affiliation(s)
- Minmin Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Junyuan Cao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Siqi Dong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuxia Hou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan Yu
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Engineering and Technology Research Centre of Organoid, Guangzhou, 510515, China
| | - Qiuyan Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yueli Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; College of Pharmacy and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300450, China
| | - Xiaoying Jia
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Bo Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Gang Li
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wei Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
8
|
Mehrbod P, Safari H, Mollai Z, Fotouhi F, Mirfakhraei Y, Entezari H, Goodarzi S, Tofighi Z. Potential antiviral effects of some native Iranian medicinal plants extracts and fractions against influenza A virus. BMC Complement Med Ther 2021; 21:246. [PMID: 34598697 PMCID: PMC8485427 DOI: 10.1186/s12906-021-03423-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 09/24/2021] [Indexed: 01/11/2023] Open
Abstract
Background Influenza A virus (IAV) infection is a continual threat to the health of animals and humans globally. Consumption of the conventional drugs has shown several side effects and drug resistance. This study was aimed to screen some Iranian medicinal plants extracts and their fractions against influenza A virus. Methods Glycyrrhiza glabra (rhizome), Myrtus commonis (leaves), Melissa officinalis (leaves), Hypericum perforatum (aerial parts), Tilia platyphyllos (flower), Salix alba (bark), and Camellia sinensis (green and fermented leaves) were extracted with 80% methanol and fractionated with chloroform and methanol, respectively. The cytotoxicity of the compounds were determined by MTT colorimetric assay on MDCK cells. The effective concentrations (EC50) of the compounds were calculated from the MTT results compared to the negative control with no significant effects on cell viability. The effects of EC50 of the compounds on viral surface glycoproteins and viral titer were tested by HI and HA virological assays, respectively and compared with oseltamivir and amantadine. Preliminary phytochemical analysis were done for promising anti-IAV extracts and fractions. Results The most effective samples against IAV titer (P ≤ 0.05) were crude extracts of G. glabra, M. officinalis and S. alba; methanol fractions of M. communis and M. officinalis; and chloroform fractions of M. communis and C. sinensis (fermented) mostly in co- and pre-penetration combined treatments. The potential extracts and fractions were rich in flavonoids, tannins, steroids and triterpenoids. Conclusion The outcomes confirmed a scientific basis for anti-influenza A virus capacity of the extracts and fractions from the selected plants for the first time, and correlated their effects with their phytochemical constituents. It is worth focusing on elucidating pure compounds and identifying their mechanism(s) of action. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03423-x.
Collapse
Affiliation(s)
- Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran, Iran
| | - Hanieh Safari
- Department of Pharmacognosy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Mollai
- Department of Pharmacognosy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fotouhi
- Influenza and Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran, Iran
| | - Yasaman Mirfakhraei
- Department of Pharmacognosy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh Entezari
- Department of Pharmacognosy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Saied Goodarzi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zahra Tofighi
- Department of Pharmacognosy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran. .,Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Musarra-Pizzo M, Pennisi R, Ben-Amor I, Mandalari G, Sciortino MT. Antiviral Activity Exerted by Natural Products against Human Viruses. Viruses 2021; 13:v13050828. [PMID: 34064347 PMCID: PMC8147851 DOI: 10.3390/v13050828] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/01/2021] [Indexed: 12/13/2022] Open
Abstract
Viral infections are responsible for several chronic and acute diseases in both humans and animals. Despite the incredible progress in human medicine, several viral diseases, such as acquired immunodeficiency syndrome, respiratory syndromes, and hepatitis, are still associated with high morbidity and mortality rates in humans. Natural products from plants or other organisms are a rich source of structurally novel chemical compounds including antivirals. Indeed, in traditional medicine, many pathological conditions have been treated using plant-derived medicines. Thus, the identification of novel alternative antiviral agents is of critical importance. In this review, we summarize novel phytochemicals with antiviral activity against human viruses and their potential application in treating or preventing viral disease.
Collapse
Affiliation(s)
- Maria Musarra-Pizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
| | - Rosamaria Pennisi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
- Shenzhen International Institute for Biomedical Research, 1301 Guanguang Rd. 3F Building 1-B, Silver Star Hi-Tech Park Longhua District, Shenzhen 518116, China
| | - Ichrak Ben-Amor
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
- Unit of Biotechnology and Pathologies, Higher Institute of Biotechnology of Sfax, University of Sfax, Sfax 3029, Tunisia
| | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
- Correspondence: (G.M.); (M.T.S.); Tel.: +39-090-6767-5217 (G.M. & M.T.S.)
| | - Maria Teresa Sciortino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
- Correspondence: (G.M.); (M.T.S.); Tel.: +39-090-6767-5217 (G.M. & M.T.S.)
| |
Collapse
|
10
|
Ali SI, Sheikh WM, Rather MA, Venkatesalu V, Muzamil Bashir S, Nabi SU. Medicinal plants: Treasure for antiviral drug discovery. Phytother Res 2021; 35:3447-3483. [PMID: 33590931 PMCID: PMC8013762 DOI: 10.1002/ptr.7039] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/04/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
The pandemic of viral diseases like novel coronavirus (2019-nCoV) prompted the scientific world to examine antiviral bioactive compounds rather than nucleic acid analogous, protease inhibitors, or other toxic synthetic molecules. The emerging viral infections significantly associated with 2019-nCoV have challenged humanity's survival. Further, there is a constant emergence of new resistant viral strains that demand novel antiviral agents with fewer side effects and cell toxicity. Despite significant progress made in immunization and regenerative medicine, numerous viruses still lack prophylactic vaccines and specific antiviral treatments that are so often influenced by the generation of viral escape mutants. Of importance, medicinal herbs offer a wide variety of therapeutic antiviral chemotypes that can inhibit viral replication by preventing viral adsorption, adhering to cell receptors, inhibiting virus penetration in the host cell, and competing for pathways of activation of intracellular signals. The present review will comprehensively summarize the promising antiviral activities of medicinal plants and their bioactive molecules. Furthermore, it will elucidate their mechanism of action and possible implications in the treatment/prevention of viral diseases even when their mechanism of action is not fully understood, which could serve as the base for the future development of novel or complementary antiviral treatments.
Collapse
Affiliation(s)
- Sofi Imtiyaz Ali
- Biochemistry & Molecular Biology Lab, Division of veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Srinagar, India
| | - Wajid Mohammad Sheikh
- Biochemistry & Molecular Biology Lab, Division of veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Srinagar, India
| | - Muzafar Ahmad Rather
- Biochemistry & Molecular Biology Lab, Division of veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Srinagar, India
| | | | - Showkeen Muzamil Bashir
- Biochemistry & Molecular Biology Lab, Division of veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Srinagar, India
| | - Showkat Ul Nabi
- Large Animal Diagnostic Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Srinagar, India
| |
Collapse
|
11
|
Malekmohammad K, Rafieian-Kopaei M, Sardari S, Sewell RDE. Effective Antiviral Medicinal Plants and Biological Compounds Against Central Nervous System Infections: A Mechanistic Review. Curr Drug Discov Technol 2020; 17:469-483. [PMID: 31309894 DOI: 10.2174/1570163816666190715114741] [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: 12/20/2018] [Revised: 03/26/2019] [Accepted: 04/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE Infectious diseases are amongst the leading causes of death in the world and central nervous system infections produced by viruses may either be fatal or generate a wide range of symptoms that affect global human health. Most antiviral plants contain active phytoconstituents such as alkaloids, flavonoids, and polyphenols, some of which play an important antiviral role. Herein, we present a background to viral central nervous system (CNS) infections, followed by a review of medicinal plants and bioactive compounds that are effective against viral pathogens in CNS infections. METHODS A comprehensive literature search was conducted on scientific databases including: PubMed, Scopus, Google Scholar, and Web of Science. The relevant keywords used as search terms were: "myelitis", "encephalitis", "meningitis", "meningoencephalitis", "encephalomyelitis", "central nervous system", "brain", "spinal cord", "infection", "virus", "medicinal plants", and "biological compounds". RESULTS The most significant viruses involved in central nervous system infections are: Herpes Simplex Virus (HSV), Varicella Zoster Virus (VZV), West Nile Virus (WNV), Enterovirus 71 (EV71), Japanese Encephalitis Virus (JEV), and Dengue Virus (DENV). The inhibitory activity of medicinal plants against CNS viruses is mostly active through prevention of viral binding to cell membranes, blocking viral genome replication, prevention of viral protein expression, scavenging reactive Oxygen Species (ROS), and reduction of plaque formation. CONCLUSION Due to the increased resistance of microorganisms (bacteria, viruses, and parasites) to antimicrobial therapies, alternative treatments, especially using plant sources and their bioactive constituents, appear to be more fruitful.
Collapse
Affiliation(s)
- Khojasteh Malekmohammad
- Department of Animal Sciences, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Mahmoud Rafieian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samira Sardari
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Robert D E Sewell
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, Wales, United Kingdom
| |
Collapse
|
12
|
Pooladanda V, Thatikonda S, Godugu C. The current understanding and potential therapeutic options to combat COVID-19. Life Sci 2020; 254:117765. [PMID: 32437797 PMCID: PMC7207108 DOI: 10.1016/j.lfs.2020.117765] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
The ongoing wreaking global outbreak of the novel human beta coronavirus (CoV) pathogen was presumed to be from a seafood wholesale market in Wuhan, China, belongs to the Coronaviridae family in the Nidovirales order. The virus is highly contagious with potential human-human transmission which was named as the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has spread across six continents and emerged as a global pandemic in short span with alarming levels of spread and severity. This virus associated symptoms and infectious respiratory illness is designated as coronavirus disease 19 (COVID-19). The SARS-CoV-2 possesses enveloped club-like spike protein projections with positive-sense large RNA genome and has a unique replication strategy. This virus was believed to have zoonotic origin with genetical identity to bat and pangolin CoV. In the current review, we introduce a general overview about the human CoVs and the associated diseases, the origin, structure, replication and key clinical events that occur in the COVID-19 pathogenicity. Furthermore, we focused on possible therapeutic options such as repurposing drugs including antimalarials, antivirals, antiparasitic drugs, and anti-HIV drugs, as well as monoclonal antibodies, vaccines as potential treatment options. Also we have summarized the latest research progress on the usage of stem cell therapy, human convalescent serum, interferon's, in the treatment of COVID-19.
Collapse
Affiliation(s)
- Venkatesh Pooladanda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
| | - Sowjanya Thatikonda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India.
| |
Collapse
|
13
|
Afify AEMM, El Baroty GS, El Baz FK, Abd El Baky HH, Murad SA. Scenedesmus obliquus: Antioxidant and antiviral activity of proteins hydrolyzed by three enzymes. J Genet Eng Biotechnol 2018; 16:399-408. [PMID: 30733753 PMCID: PMC6353658 DOI: 10.1016/j.jgeb.2018.01.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 11/22/2017] [Accepted: 01/16/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE To obtain protein hydrolysates from fresh water green algae Scenedesmus obliquus by three different enzymes and evaluate its antioxidant and antiviral activity. METHODS Enzymatic hydrolysates of green algae Scenedesmus obliquus protein were prepared by treatment with: 1.2% solution of pepsin, trypsin or papain. Protein was extracted from S. obliquus by three different extraction methods. Protein extracts and hydrolysates were assessed from stained gels following SDS-PAGE of samples. Antioxidant activity of protein hydrolysates was investigated. RESULTS S. obliquus cells and protein extracts were rich in Arg, Lys, Asp, Ala, and His. Protein hydrolyzed by papain (Sd1pa) and protein hydrolyzed by trypsin (Sd2Try) induced highest antioxidant activity based on 1,1-diphenyl-2-picryl-hydrazyl (DPPH) radical-scavenging (41.41% and 40.62%) respectively, and on 2,2'-azinobis 3-ethyl-benzothiazoline-6-sulphonate (ABTS) radical (87.03% and 45.12%) respectively, at 150 µg/ml. The inhibitory effect and mode of action of protein hydrolysates were evaluated against Coxsackie B3 virus (CVB3). Protein hydrolyzed by papain (Sd2pa) and protein hydrolyzed by pepsin (Sd1pep) at 100 µg/ml exhibited antiviral activity (66.2% and 57.6%, respectively), against (CVB3) from all protein hydrolysates. CONCLUSION S. obliquus protein hydrolysates have a potential as antioxidative neutraceutical ingredients and a potential therapeutic agent against CVB3.
Collapse
Affiliation(s)
| | - Gamal S. El Baroty
- Biochemistry Department, Faculty of Agriculture, Cairo University, Cairo, Egypt
| | - Farouk K. El Baz
- Plant Biochemistry Department, National Research Centre, Dokki, Cairo, Egypt
| | | | - Soha A. Murad
- Plant Biochemistry Department, National Research Centre, Dokki, Cairo, Egypt
| |
Collapse
|
14
|
Mehrbod P, Abdalla MA, Njoya EM, Ahmed AS, Fotouhi F, Farahmand B, Gado DA, Tabatabaian M, Fasanmi OG, Eloff JN, McGaw LJ, Fasina FO. South African medicinal plant extracts active against influenza A virus. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:112. [PMID: 29587734 PMCID: PMC5872571 DOI: 10.1186/s12906-018-2184-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 03/22/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Influenza infection remains a major health threat for animals and humans which crucially requires effective antiviral remedies. The usage of herbal medications as readily available alternatives for their compatibility with the body and fewer side effects compared to synthetic chemical treatments has become popular globally. The aim of this study was to investigate and screen in vitro anti-influenza activity of extracts of five South African medicinal plants, namely Tabernaemontana ventricosa, Cussonia spicata, Rapanea melanophloeos, Pittosporum viridiflorum and Clerodendrum glabrum, species which are used traditionally for the treatment of several diseases such as inflammatory and respiratory diseases. METHODS Methanol, ethanol (100% and 30%), acetone, hot and cold water extracts of the powdered plants leaves were obtained by standard methods. The cytotoxicity was determined by the MTT colorimetric assay on MDCK cells. The concentrations below CC50 values were tested for antiviral activity against influenza A virus (IAV) in different combination treatments. The effect of extracts on viral surface glycoproteins and viral titer were tested by HI and HA virological assays, respectively. RESULTS Based on the applied methods, the most effective results against IAV were obtained from Rapanea melanophloeos methanol leaf extract (EC50 = 113.3 μg/ml) and Pittosporum viridiflorum methanol, 100% and 30% ethanol and acetone leaf extracts (EC50 values = 3.6, 3.4, 19.2, 82.3 μg/ml, respectively) in all types of combined treatments especially in pre- and post-penetration combined treatments with highly significant effects against viral titer (P ≤ 0.01). CONCLUSION The outcomes offer for the first time a scientific basis for the use of extracts of Rapanea melanophloeos and Pittosporum viridiflorum against IAV. It is worth focusing on the isolation and identification of effective active compounds and elucidating the mechanism of action from these species. However, Tabernaemontana ventricosa, Cussonia spicata and Clerodendrum glabrum leaf extracts were ineffective in vitro in this study.
Collapse
Affiliation(s)
- Parvaneh Mehrbod
- 0000 0001 2107 2298grid.49697.35Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
- 0000 0000 9562 2611grid.420169.8Influenza and Other Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran, Iran
| | - Muna A. Abdalla
- 0000 0001 2107 2298grid.49697.35Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Pretoria, South Africa
| | - Emmanuel M. Njoya
- 0000 0001 2107 2298grid.49697.35Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Pretoria, South Africa
| | - Aroke S. Ahmed
- 0000 0001 2107 2298grid.49697.35Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Pretoria, South Africa
- grid.463291.bFederal Institute of Industrial Research, Oshodi, Lagos, Nigeria
| | - Fatemeh Fotouhi
- 0000 0000 9562 2611grid.420169.8Influenza and Other Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran, Iran
| | - Behrokh Farahmand
- 0000 0000 9562 2611grid.420169.8Influenza and Other Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran, Iran
| | - Dorcas A. Gado
- 0000 0001 2107 2298grid.49697.35Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Pretoria, South Africa
| | - Mansoureh Tabatabaian
- 0000 0000 9562 2611grid.420169.8Influenza and Other Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran, Iran
| | - Olubunmi G. Fasanmi
- 0000 0001 2107 2298grid.49697.35Department of Production Animal Studies, University of Pretoria, Pretoria, South Africa
- Department of Animal Health, Federal College of Animal Health and Production Technology, Ibadan, Nigeria
| | - Jacobus N. Eloff
- 0000 0001 2107 2298grid.49697.35Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Pretoria, South Africa
| | - Lyndy J. McGaw
- 0000 0001 2107 2298grid.49697.35Phytomedicine Programme, Department of Paraclinical Sciences, University of Pretoria, Pretoria, South Africa
| | - Folorunso O. Fasina
- 0000 0001 2107 2298grid.49697.35Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
- ECTAD, Food and Agriculture Organization of the United Nations (FAO), Block P, Level 3, United Nations Complex, UN Avenue, Gigiri, Nairobi, Kenya
| |
Collapse
|
15
|
Dai JP, Wang QW, Su Y, Gu LM, Deng HX, Chen XX, Li WZ, Li KS. Oxymatrine Inhibits Influenza A Virus Replication and Inflammation via TLR4, p38 MAPK and NF-κB Pathways. Int J Mol Sci 2018; 19:ijms19040965. [PMID: 29570670 PMCID: PMC5979549 DOI: 10.3390/ijms19040965] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/11/2018] [Accepted: 03/13/2018] [Indexed: 02/05/2023] Open
Abstract
Oxymatrine (OMT) is a strong immunosuppressive agent that has been used in the clinic for many years. In the present study, by using plaque inhibition, luciferase reporter plasmids, qRT-PCR, western blotting, and ELISA assays, we have investigated the effect and mechanism of OMT on influenza A virus (IAV) replication and IAV-induced inflammation in vitro and in vivo. The results showed that OMT had excellent anti-IAV activity on eight IAV strains in vitro. OMT could significantly decrease the promoter activity of TLR3, TLR4, TLR7, MyD88, and TRAF6 genes, inhibit IAV-induced activations of Akt, ERK1/2, p38 MAPK, and NF-κB pathways, and suppress the expressions of inflammatory cytokines and MMP-2/-9. Activators of TLR4, p38 MAPK and NF-κB pathways could significantly antagonize the anti-IAV activity of OMT in vitro, including IAV replication and IAV-induced cytopathogenic effect (CPE). Furthermore, OMT could reduce the loss of body weight, significantly increase the survival rate of IAV-infected mice, decrease the lung index, pulmonary inflammation and lung viral titter, and improve pulmonary histopathological changes. In conclusion, OMT possesses anti-IAV and anti-inflammatory activities, the mechanism of action may be linked to its ability to inhibit IAV-induced activations of TLR4, p38 MAPK, and NF-κB pathways.
Collapse
Affiliation(s)
- Jian-Ping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Qian-Wen Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Yun Su
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Li-Ming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Hui-Xiong Deng
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Xiao-Xuan Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Wei-Zhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA.
| | - Kang-Sheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
16
|
Dai J, Gu L, Su Y, Wang Q, Zhao Y, Chen X, Deng H, Li W, Wang G, Li K. Inhibition of curcumin on influenza A virus infection and influenzal pneumonia via oxidative stress, TLR2/4, p38/JNK MAPK and NF-κB pathways. Int Immunopharmacol 2018; 54:177-187. [PMID: 29153953 DOI: 10.1016/j.intimp.2017.11.009] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 10/29/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Oxidative stress, Nrf2-HO-1 and TLR-MAPK/NF-κB signaling pathways have been proved to be involved in influenza A virus (IAV) replication and influenzal pneumonia. In the previous studies, we have performed several high-throughput drug screenings based on the TLR pathways. In the present study, through plaque inhibition test, luciferase reporter assay, TCID50, qRT-PCR, western blotting, ELISA and siRNA assays, we investigated the effect and mechanism of action of curcumin against IAV infection in vitro and in vivo. The results showed that curcumin could directly inactivate IAV, blocked IAV adsorption and inhibited IAV proliferation. As for the underlying mechanisms, we found that curcumin could significantly inhibit IAV-induced oxidative stress, increased Nrf2, HO-1, NQO1, GSTA3 and IFN-β production, and suppressed IAV-induced activation of TLR2/4/7, Akt, p38/JNK MAPK and NF-κB pathways. Suppression of Nrf2 via siRNA significantly abolished the stimulatory effect of curcumin on HO-1, NQO1, GSTA3 and IFN-β production and meanwhile blocked the inhibitory effect of curcumin on IAV M2 production. Oxidant H2O2 and TLR2/4, p38/JNK and NF-κB agonists could significantly antagonize the anti-IAV activity of curcumin in vitro. Additionally, curcumin significantly increased the survival rate of mice, reduced lung index, inflammatory cytokines and lung IAV titer, and finally improved pulmonary histopathological changes after IAV infection. In conclusion, curcumin can directly inactivate IAV, inhibits IAV adsorption and replication; and its inhibition on IAV replication may be via activating Nrf2 signal and inhibiting IAV-induced activation of TLR2/4, p38/JNK MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Jianping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China.
| | - Liming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Yun Su
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Qianwen Wang
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Ying Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Xiaoxua Chen
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Huixiong Deng
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Weizhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, Virginia-Maryland Regional College of Veterinary Medicine, 159 College Park Rd, MD 20742, USA
| | - Gefei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Kangsheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| |
Collapse
|
17
|
Wang QW, Su Y, Sheng JT, Gu LM, Zhao Y, Chen XX, Chen C, Li WZ, Li KS, Dai JP. Anti-influenza A virus activity of rhein through regulating oxidative stress, TLR4, Akt, MAPK, and NF-κB signal pathways. PLoS One 2018; 13:e0191793. [PMID: 29385192 PMCID: PMC5791991 DOI: 10.1371/journal.pone.0191793] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/11/2018] [Indexed: 02/06/2023] Open
Abstract
Rhein, an anthraquinone compound existing in many traditional herbal medicines, has anti-inflammatory, antioxidant, antitumor, antiviral, hepatoprotective, and nephroprotective activities, but its anti-influenza A virus (IAV) activity is ambiguous. In the present study, through plaque inhibition assay, time-of-addition assay, antioxidant assay, qRT-PCR, ELISA, and western blotting assays, we investigated the anti-IAV effect and mechanism of action of rhein in vitro and in vivo. The results showed that rhein could significantly inhibit IAV adsorption and replication, decrease IAV-induced oxidative stress, activations of TLR4, Akt, p38, JNK MAPK, and NF-κB pathways, and production of inflammatory cytokines and matrix metalloproteinases in vitro. Oxidant H2O2 and agonists of TLR4, Akt, p38/JNK and IKK/NF-κB could significantly antagonize the inhibitory effects of rhein on IAV-induced cytopathic effect (CPE) and IAV replication. Through an in vivo test in mice, we also found that rhein could significantly improve the survival rate, lung index, pulmonary cytokines, and pulmonary histopathological changes. Rhein also significantly decreased pulmonary viral load at a high dose. In conclusion, rhein can inhibit IAV adsorption and replication, and the mechanism of action to inhibit IAV replication may be due to its ability to suppress IAV-induced oxidative stress and activations of TLR4, Akt, p38, JNK MAPK, and NF-κB signal pathways.
Collapse
Affiliation(s)
- Qian-Wen Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yun Su
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jiang-Tao Sheng
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Li-Ming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Ying Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiao-Xuan Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Cheng Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Wei-Zhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, United States of America
| | - Kang-Sheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jian-Ping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
- * E-mail:
| |
Collapse
|
18
|
Dai JP, Wang QW, Su Y, Gu LM, Zhao Y, Chen XX, Chen C, Li WZ, Wang GF, Li KS. Emodin Inhibition of Influenza A Virus Replication and Influenza Viral Pneumonia via the Nrf2, TLR4, p38/JNK and NF-kappaB Pathways. Molecules 2017; 22:molecules22101754. [PMID: 29057806 PMCID: PMC6151665 DOI: 10.3390/molecules22101754] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 02/08/2023] Open
Abstract
Lasting activations of toll-like receptors (TLRs), MAPK and NF-κB pathways can support influenza A virus (IAV) infection and promote pneumonia. In this study, we have investigated the effect and mechanism of action of emodin on IAV infection using qRT-PCR, western blotting, ELISA, Nrf2 luciferase reporter, siRNA and plaque inhibition assays. The results showed that emodin could significantly inhibit IAV (ST169, H1N1) replication, reduce IAV-induced expressions of TLR2/3/4/7, MyD88 and TRAF6, decrease IAV-induced phosphorylations of p38/JNK MAPK and nuclear translocation of NF-κB p65. Emodin also activated the Nrf2 pathway, decreased ROS levels, increased GSH levelss and GSH/GSSG ratio, and upregulated the activities of SOD, GR, CAT and GSH-Px after IAV infection. Suppression of Nrf2 via siRNA markedly blocked the inhibitory effects of emodin on IAV-induced activations of TLR4, p38/JNK, and NF-κB pathways and on IAV-induced production of IL-1β, IL-6 and expression of IAV M2 protein. Emodin also dramatically increased the survival rate of mice, reduced lung edema, pulmonary viral titer and inflammatory cytokines, and improved lung histopathological changes. In conclusion, emodin can inhibit IAV replication and influenza viral pneumonia, at least in part, by activating Nrf2 signaling and inhibiting IAV-induced activations of the TLR4, p38/JNK MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Jian-Ping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Qian-Wen Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Yun Su
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Li-Ming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Ying Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Xiao-Xua Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Cheng Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Wei-Zhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, and Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD 20742, USA.
| | - Ge-Fei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| | - Kang-Sheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
19
|
Choi HJ, Song HH, Lee JS, Ko HJ, Song JH. Inhibitory Effects of Norwogonin, Oroxylin A, and Mosloflavone on Enterovirus 71. Biomol Ther (Seoul) 2016; 24:552-8. [PMID: 27257010 PMCID: PMC5012882 DOI: 10.4062/biomolther.2015.200] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/23/2016] [Accepted: 03/03/2016] [Indexed: 12/04/2022] Open
Abstract
Severe complications associated with EV71 infections are a common cause of neonatal death. Lack of effective therapeutic agents for these infections underlines the importance of research for the development of new antiviral compounds. In the present study, the anti-EV71 activity of norwogonin, oroxylin A, and mosloflavone from Scutellaria baicalensis Georgi was evaluated using a cytopathic effect (CPE) reduction method, which demonstrated that all three compounds possessed strong anti-EV71 activity and decreased the formation of visible CPEs. Norwogonin, oroxylin A, and mosloflavone also inhibited virus replication during the initial stage of virus infection, and they inhibited viral VP2 protein expression, thereby inhibiting viral capsid protein synthesis. However, ribavirin has a relatively weaker efficacy compared to the other drugs. Therefore, these findings provide important information that will aid in the utilization of norwogonin, oroxylin A, and mosloflavone for EV71 treatment.
Collapse
Affiliation(s)
- Hwa Jung Choi
- Department of Beauty Science, Kwangju Women's University, Gwangju 62393, Republic of Korea
| | - Hyuk-Hwan Song
- Agency for Korea National Food Cluster (AnFC), Iksan 54538, Republic of Korea
| | - Jae-Sug Lee
- Department of Beauty Science, Kwangju Women's University, Gwangju 62393, Republic of Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jae-Hyoung Song
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
20
|
Kim C, Kang H, Kim DE, Song JH, Choi M, Kang M, Lee K, Kim HS, Shin JS, Jeong H, Jung S, Han SB, Kim JH, Ko HJ, Lee CK, Kim M, Cho S. Antiviral activity of micafungin against enterovirus 71. Virol J 2016; 13:99. [PMID: 27296985 PMCID: PMC4907259 DOI: 10.1186/s12985-016-0557-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/07/2016] [Indexed: 12/30/2022] Open
Abstract
Background Enterovirus 71 (EV71) is a major causative agent of hand-foot-mouth disease (HFMD) and also causes severe neurological complications, leading to fatality in young children. However, no effective therapy is currently available for the treatment of this infection. Methods We identified small-molecule inhibitors of EV71 from a screen of 968 Food and Drug Administration (FDA)-approved drugs, with which clinical application for EV71-associated diseases would be more feasible, using EV71 subgenomic replicon system. Primary hits were extensively evaluated for their antiviral activities in EV71-infected cells. Results We identified micafungin, an echinocandin antifungal drug, as a novel inhibitor of EV71. Micafungin potently inhibits the proliferation of EV71 as well as the replication of EV71 replicon in cells with a low micromolar IC50 (~5 μM). The strong antiviral effect of micafungin on EV71 replicon and the result from time-of-addition experiment demonstrated a targeting of micafungin on virion-independent intracellular process(es) during EV71 infection. Moreover, an extensive analysis excluded the involvement of 2C and 3A proteins, IRES-dependent translation, and also that of polyprotein processing in the antiviral effect of micafungin. Conclusions Our research revealed a new indication of micafungin as an effective inhibitor of EV71, which is the first case reporting antiviral activity of micafungin, an antifungal drug. Electronic supplementary material The online version of this article (doi:10.1186/s12985-016-0557-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chonsaeng Kim
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Hyunju Kang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do, 28116, South Korea.,College of Pharmacy, Chungbuk National University, 1 Chungdae-ro Seowon-gu, Cheongju-si, Chungcheongbuk-do, 28644, South Korea
| | - Dong-Eun Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do, 28116, South Korea.,College of Pharmacy, Chungbuk National University, 1 Chungdae-ro Seowon-gu, Cheongju-si, Chungcheongbuk-do, 28644, South Korea
| | - Jae-Hyoung Song
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, 1 Gangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Miri Choi
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do, 28116, South Korea
| | - Mingu Kang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do, 28116, South Korea
| | - Kyungjin Lee
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Hae Soo Kim
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Jin Soo Shin
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Hyejeong Jeong
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do, 28116, South Korea
| | - Sunhee Jung
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do, 28116, South Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, 1 Chungdae-ro Seowon-gu, Cheongju-si, Chungcheongbuk-do, 28644, South Korea
| | - Jong Heon Kim
- Cancer Cell and Molecular Biology Branch, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10408, South Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, 1 Gangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Chong-Kyo Lee
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Meehyein Kim
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Sungchan Cho
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do, 28116, South Korea. .,Department of Biomolecular Science, Korea University of Science and Technology, 217 Gajeong-ro, Daejeon, 34113, South Korea.
| |
Collapse
|
21
|
Shakiba Y, Rezatofighi SE, Seyyed Nejad SM, Roayaei Ardakani M. Antiviral Activity of Alhagi maurorum Medik’s Methanolic Extract on Foot and Mouth Disease Virus (FMDV) in Cell Cultures. Jundishapur J Nat Pharm Prod 2016. [DOI: 10.17795/jjnpp-30641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Wang M, Tao L, Xu H. Chinese herbal medicines as a source of molecules with anti-enterovirus 71 activity. Chin Med 2016; 11:2. [PMID: 26834824 PMCID: PMC4731985 DOI: 10.1186/s13020-016-0074-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/08/2016] [Indexed: 02/06/2023] Open
Abstract
Enterovirus 71 (EV71) is one of the causative agents of hand, foot, and mouth disease (HFMD), which sometimes leads to severe neurological disease and death in the Asia-Pacific region. In Chinese medicine, HFMD is caused mainly by an accumulation of damp-heat and toxicity in the body. No effective drugs are currently available for the treatment and prevention of EV71 infection. This review summarizes the potential Chinese herbal extracts and isolated compounds with antiviral activity against EV71 and their clinical applications, especially those categorized as heat-clearing and detoxifying.
Collapse
Affiliation(s)
- Mengjie Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China
| | - Ling Tao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China ; Xinxiang Medical University, Jinsui Road 601, Xinxiang, Henan 453003 China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China
| |
Collapse
|
23
|
Kang H, Kim C, Kim DE, Song JH, Choi M, Choi K, Kang M, Lee K, Kim HS, Shin JS, Kim J, Han SB, Lee MY, Lee SU, Lee CK, Kim M, Ko HJ, van Kuppeveld FJM, Cho S. Synergistic antiviral activity of gemcitabine and ribavirin against enteroviruses. Antiviral Res 2015; 124:1-10. [PMID: 26526589 DOI: 10.1016/j.antiviral.2015.10.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/25/2015] [Accepted: 10/10/2015] [Indexed: 12/21/2022]
Abstract
Enteroviruses are major causative agents of various human diseases, and some of them are currently considered to be an enormous threat to public health. However, no effective therapy is currently available for the treatment of these infections. We identified gemcitabine, a nucleoside-analog drug used for cancer treatment, from a screen of bioactive chemicals as a novel inhibitor of coxsackievirus B3 (CVB3) and enterovirus 71 (EV71). Gemcitabine potently inhibited the proliferation of CVB3 and EV71, as well as the replication of CVB3 and EV71 replicons, in cells with a low micromolar IC50 (1-5 μM). Its strong inhibitory effect was also observed in cells infected with human rhinoviruses, demonstrating broad-spectrum antiviral effects on enteroviruses. Mechanistically, an extensive analysis excluded the involvement of 2C, 3A, IRES-dependent translation, and also that of polyprotein processing in the antiviral effects of gemcitabine. Importantly, gemcitabine in combination with ribavirin, an antiviral drug currently being used against a few RNA viruses, exhibited a synergistic antiviral effect on the replication of CVB3 and EV71 replicons. Consequently, our results clearly demonstrate a new indication for gemcitabine as an effective broad-spectrum inhibitor of enteroviruses and strongly suggest a new therapeutic strategy using gemcitabine alone or in combination with ribavirin for the treatment of various diseases associated with enterovirus infection.
Collapse
Affiliation(s)
- Hyunju Kang
- Incurable Diseases Therapeutics Research Center, Korea Research Institute of Bioscience & Biotechnology, Cheongju, South Korea; College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Chonsaeng Kim
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Dong-eun Kim
- Incurable Diseases Therapeutics Research Center, Korea Research Institute of Bioscience & Biotechnology, Cheongju, South Korea; College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Jae-Hyoung Song
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Miri Choi
- Incurable Diseases Therapeutics Research Center, Korea Research Institute of Bioscience & Biotechnology, Cheongju, South Korea; College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Kwangman Choi
- Incurable Diseases Therapeutics Research Center, Korea Research Institute of Bioscience & Biotechnology, Cheongju, South Korea; Department of Medical Science, Soonchunhyang University, Asan, South Korea
| | - Mingu Kang
- Incurable Diseases Therapeutics Research Center, Korea Research Institute of Bioscience & Biotechnology, Cheongju, South Korea
| | - Kyungjin Lee
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Hae Soo Kim
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Jin Soo Shin
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, South Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Mi-Young Lee
- Department of Medical Science, Soonchunhyang University, Asan, South Korea
| | - Su Ui Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, Cheongju, South Korea
| | - Chong-Kyo Lee
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Meehyein Kim
- Virus Research and Testing Center, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Frank J M van Kuppeveld
- Section of Virology, Department Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sungchan Cho
- Incurable Diseases Therapeutics Research Center, Korea Research Institute of Bioscience & Biotechnology, Cheongju, South Korea; Department of Biomolecular Science, Korea University of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
24
|
Wang L, Wang J, Wang L, Ma S, Liu Y. Anti-Enterovirus 71 Agents of Natural Products. Molecules 2015; 20:16320-33. [PMID: 26370955 PMCID: PMC6331931 DOI: 10.3390/molecules200916320] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 08/18/2015] [Accepted: 08/26/2015] [Indexed: 12/25/2022] Open
Abstract
This review, with 42 references, presents the fascinating area of anti-enterovirus 71 natural products over the last three decades for the first time. It covers literature published from 2005–2015 and refers to compounds isolated from biogenic sources. In total, 58 naturally-occurring anti-EV71 compounds are recorded.
Collapse
Affiliation(s)
- Liyan Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China,.
| | - Junfeng Wang
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China.
| | - Lishu Wang
- Jilin Provincial Academy of Chinese Medicine Sciences, Changchun 130021, China.
| | - Shurong Ma
- Endoscopy Center, China-Japan Union Hospital, Jilin University, Changchun 130021, China.
| | - Yonghong Liu
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China.
- South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510301, China.
| |
Collapse
|
25
|
Ngan LTM, Jang MJ, Kwon MJ, Ahn YJ. Antiviral activity and possible mechanism of action of constituents identified in Paeonia lactiflora root toward human rhinoviruses. PLoS One 2015; 10:e0121629. [PMID: 25860871 PMCID: PMC4393083 DOI: 10.1371/journal.pone.0121629] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 02/11/2015] [Indexed: 11/19/2022] Open
Abstract
Human rhinoviruses (HRVs) are responsible for more than half of all cases of the common cold and cost billions of USD annually in medical visits and missed school and work. An assessment was made of the antiviral activities and mechanisms of action of paeonol (PA) and 1,2,3,4,6-penta-O-galloyl-β-D-glucopyranose (PGG) from Paeonia lactiflora root toward HRV-2 and HRV-4 in MRC5 cells using a tetrazolium method and real-time quantitative reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay. Results were compared with those of a reference control ribavirin. Based on 50% inhibitory concentration values, PGG was 13.4 and 18.0 times more active toward HRV-2 (17.89 μM) and HRV-4 (17.33 μM) in MRC5 cells, respectively, than ribavirin. The constituents had relatively high selective index values (3.3->8.5). The 100 μg/mL PA and 20 μg/mL PGG did not interact with the HRV-4 particles. These constituents inhibited HRV-4 infection only when they were added during the virus inoculation (0 h), the adsorption period of HRVs, but not after 1 h or later. Moreover, the RNA replication levels of HRVs were remarkably reduced in the MRC5 cultures treated with these constituents. These findings suggest that PGG and PA may block or reduce the entry of the viruses into the cells to protect the cells from the virus destruction and abate virus replication, which may play an important role in interfering with expressions of rhinovirus receptors (intercellular adhesion molecule-1 and low-density lipoprotein receptor), inflammatory cytokines (interleukin (IL)-6, IL-8, tumor necrosis factor, interferon beta, and IL-1β), and Toll-like receptor, which resulted in diminishing symptoms induced by HRV. Global efforts to reduce the level of synthetic drugs justify further studies on P. lactiflora root-derived materials as potential anti-HRV products or lead molecules for the prevention or treatment of HRV.
Collapse
Affiliation(s)
- Luong Thi My Ngan
- Department of Plant Biotechnology and Biotransformation, Faculty of Biology, Ho Chi Minh City University of Science, Vietnam National University, Ho Chi Minh, Vietnam
| | - Myeong Jin Jang
- Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | - Min Jung Kwon
- Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | - Young Joon Ahn
- Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| |
Collapse
|
26
|
Shang L, Wang Y, Qing J, Shu B, Cao L, Lou Z, Gong P, Sun Y, Yin Z. An adenosine nucleoside analogue NITD008 inhibits EV71 proliferation. Antiviral Res 2014; 112:47-58. [PMID: 25446894 DOI: 10.1016/j.antiviral.2014.10.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/12/2014] [Accepted: 10/14/2014] [Indexed: 02/05/2023]
Abstract
Enterovirus 71 (EV71), one of the major causative agents of Hand-Foot-Mouth Disease (HFMD), causes severe pandemics and hundreds of deaths in the Asia-Pacific region annually and is an enormous public health threat. However, effective therapeutic antiviral drugs against EV71 are rare. Nucleoside analogues have been successfully used in the clinic for the treatment of various viral infections. We evaluated a total of 27 nucleoside analogues and discovered that an adenosine nucleoside analogue NITD008, which has been reported to be an antiviral reagent that specifically inhibits flaviviruses, effectively suppressed the propagation of different strains of EV71 in RD, 293T and Vero cells with a relatively high selectivity index. Triphosphorylated NITD008 (ppp-NITD008) functions as a chain terminator to directly inhibit the RNA-dependent RNA polymerase activity of EV71, and it does not affect the EV71 VPg uridylylation process. A significant synergistic anti-EV71 effect of NITD008 with rupintrivir (AG7088) (a protease inhibitor) was documented, supporting the potential combination therapy of NITD008 with other inhibitors for the treatment of EV71 infections.
Collapse
Affiliation(s)
- Luqing Shang
- College of Pharmacy & State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| | - Yaxin Wang
- National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Jie Qing
- Tsinghua-Peking Center for Life Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Bo Shu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Lin Cao
- College of Pharmacy & State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China; School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zhiyong Lou
- School of Medicine, Tsinghua University, Beijing 100084, China; Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Peng Gong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yuna Sun
- National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China.
| | - Zheng Yin
- College of Pharmacy & State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China.
| |
Collapse
|
27
|
Abstract
Viral infections play an important role in human diseases, and recent outbreaks in the advent of globalization and ease of travel have underscored their prevention as a critical issue in safeguarding public health. Despite the progress made in immunization and drug development, many viruses lack preventive vaccines and efficient antiviral therapies, which are often beset by the generation of viral escape mutants. Thus, identifying novel antiviral drugs is of critical importance and natural products are an excellent source for such discoveries. In this mini-review, we summarize the antiviral effects reported for several natural products and herbal medicines.
Collapse
Affiliation(s)
- Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Chan Hsu
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Ching Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
28
|
Park SW, Kwon MJ, Yoo JY, Choi HJ, Ahn YJ. Antiviral activity and possible mode of action of ellagic acid identified in Lagerstroemia speciosa leaves toward human rhinoviruses. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:171. [PMID: 24885569 PMCID: PMC4052798 DOI: 10.1186/1472-6882-14-171] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/21/2014] [Indexed: 12/02/2022]
Abstract
BACKGROUND Human rhinoviruses (HRVs) are responsible for more than half of all cases of the common cold and cause billions of USD annually in medical visits and school and work absenteeism. An assessment was made of the cytotoxic and antiviral activities and possible mode of action of the tannin ellagic acid from the leaves of Lagerstroemia speciosa toward HeLa cells and three rhinoviruses, HRV-2, -3, and -4. METHODS The antiviral property and mechanism of action of ellagic acid were evaluated using a sulforhodamine B assay and real-time reverse transcription-PCR (RT-PCR) with SYBR Green dye. Results were compared with those of the currently used broad-spectrum antiviral agent, ribavirin. RESULTS As judged by 50% inhibitory concentration values, natural ellagic acid was 1.8, 2.3, and 2.2 times more toxic toward HRV-2 (38 μg/mL), HRV-3 (31 μg/mL), and HRV-4 (29 μg/mL) than ribavirin, respectively. The inhibition rate of preincubation with 50 μg/mL ellagic acid was 17%, whereas continuous presence of ellagic acid during infection led to a significant increase in the inhibition (70%). Treatment with 50 μg/mL ellagic acid considerably suppressed HRV-4 infection only when added just after the virus inoculation (0 h) (87% inhibition), but not before -1 h or after 1 h or later (<20% inhibition). These findings suggest that ellagic acid does not interact with the HRV-4 particles and may directly interact with the human cells in the early stage of HRV infections to protect the cells from the virus destruction. Furthermore, RT-PCR analysis revealed that 50 μg/mL ellagic acid strongly inhibited the RNA replication of HRV-4 in HeLa cells, suggesting that ellagic acid inhibits virus replication by targeting on cellular molecules, rather than virus molecules. CONCLUSIONS Global efforts to reduce the level of antibiotics justify further studies on L. speciosa leaf-derived materials containing ellagic acid as potential anti-HRV products or a lead molecule for the prevention or treatment of HRV infection.
Collapse
Affiliation(s)
- Sang Wook Park
- Interdisciplinary Program in Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 151-921, Republic of Korea
| | - Min Jung Kwon
- Department of Agricultural Biotechnology, Seoul National University, Seoul 151-921, Republic of Korea
| | - Ji Young Yoo
- Department of Agricultural Biotechnology, Seoul National University, Seoul 151-921, Republic of Korea
| | - Hwa-Jung Choi
- Department of Infection Biology, Zoonosis Research Center, Wonkwang University School of Medicine, Iksan 570-749, Jeollabuk-do, Republic of Korea
| | - Young-Joon Ahn
- Department of Agricultural Biotechnology, Seoul National University, Seoul 151-921, Republic of Korea
| |
Collapse
|
29
|
Song JH, Choi HJ, Song HH, Hong EH, Lee BR, Oh SR, Choi K, Yeo SG, Lee YP, Cho S, Ko HJ. Antiviral activity of ginsenosides against coxsackievirus B3, enterovirus 71, and human rhinovirus 3. J Ginseng Res 2014; 38:173-9. [PMID: 25378991 PMCID: PMC4213867 DOI: 10.1016/j.jgr.2014.04.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/09/2014] [Accepted: 04/10/2014] [Indexed: 01/12/2023] Open
Abstract
Background Ginsenosides are the major components responsible for the biochemical and pharmacological actions of ginseng, and have been shown to have various biological activities. In this study, we investigated the antiviral activities of seven ginsenosides [protopanaxatriol (PT) type: Re, Rf, and Rg2; protopanaxadiol (PD) type: Rb1, Rb2, Rc, and Rd)] against coxsackievirus B3 (CVB3), enterovirus 71 (EV71), and human rhinovirus 3 (HRV3). Methods Assays of antiviral activity and cytotoxicity were evaluated by the sulforhodamine B method using the cytopathic effect (CPE) reduction assay. Results The antiviral assays demonstrated that, of the seven ginsenosides, the PT-type ginsenosides (Re, Rf, and Rg2) possess significant antiviral activities against CVB3 and HRV3 at a concentration of 100 μg/mL. Among the PT-type ginsenosides, only ginsenoside Rg2 showed significant anti-EV71 activity with no cytotoxicity to cells at 100 μg/mL. The PD-type ginsenosides (Rb1, Rb2, Rc, and Rd), by contrast, did not show any significant antiviral activity against CVB3, EV71, and HRV3, and exhibited cytotoxic effects to virus-infected cells. Notably, the antiviral efficacies of PT-type ginsenosides were comparable to those of ribavirin, a commonly used antiviral drug. Conclusion Collectively, our findings suggest that the ginsenosides Re, Rf, and Rg2 have the potential to be effective in the treatment of CVB3, EV71, and HRV3 infection.
Collapse
Affiliation(s)
- Jae-Hyoung Song
- College of Pharmacy, Kangwon National University, Chuncheon, Korea
| | - Hwa-Jung Choi
- Department of Beauty Science, Kwangju Women's University, Gwangju, Korea
| | - Hyuk-Hwan Song
- Natural Medicine Research Center, Korea Research Institute Bioscience and Biotechnology, Chungcheongbuk-do, Korea
| | - Eun-Hye Hong
- College of Pharmacy, Kangwon National University, Chuncheon, Korea
| | - Bo-Ra Lee
- College of Pharmacy, Kangwon National University, Chuncheon, Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute Bioscience and Biotechnology, Chungcheongbuk-do, Korea
| | - Kwangman Choi
- Targeted Medicine Research Center, Korea Research Institute Bioscience and Biotechnology, Chungcheongbuk-do, Korea
| | - Sang-Gu Yeo
- Division of Vaccine Research, Center for Infectious Diseases, National Institute of Health, Korea Centers for Diseases Control and Prevention, Chungcheongbuk-do, Korea
| | - Yong-Pyo Lee
- Division of Vaccine Research, Center for Infectious Diseases, National Institute of Health, Korea Centers for Diseases Control and Prevention, Chungcheongbuk-do, Korea
| | - Sungchan Cho
- Targeted Medicine Research Center, Korea Research Institute Bioscience and Biotechnology, Chungcheongbuk-do, Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
30
|
Abstract
This review highlights ten "hot topics" in current antiviral research: (i) new nucleoside derivatives (i.e., PSI-352938) showing high potential as a direct antiviral against hepatitis C virus (HCV); (ii) cyclopropavir, which should be further pursued for treatment of human cytomegalovirus (HCMV) infections; (iii) North-methanocarbathymidine (N-MCT), with a N-locked conformation, showing promising activity against both α- and γ-herpesviruses; (iv) CMX001, an orally bioavailable prodrug of cidofovir with broad-spectrum activity against DNA viruses, including polyoma, adeno, herpes, and pox; (v) favipiravir, which is primarily pursued for the treatment of influenza virus infections, but also inhibits the replication of other RNA viruses, particularly (-)RNA viruses such as arena, bunya, and hanta; (vi) newly emerging antiarenaviral compounds which should be more effective (and less toxic) than the ubiquitously used ribavirin; (vii) antipicornavirus agents in clinical development (pleconaril, BTA-798, and V-073); (viii) natural products receiving increased attention as potential antiviral drugs; (ix) antivirals such as U0126 targeted at specific cellular kinase pathways [i.e., mitogen extracellular kinase (MEK)], showing activity against influenza and other viruses; and (x) two structurally unrelated compounds (i.e., LJ-001 and dUY11) with broad-spectrum activity against virtually all enveloped RNA and DNA viruses.
Collapse
Affiliation(s)
- Erik De Clercq
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, B-3000, Leuven, Belgium.
| |
Collapse
|
31
|
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: 2.1] [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
|
32
|
Chebulagic acid, a hydrolyzable tannin, exhibited antiviral activity in vitro and in vivo against human enterovirus 71. Int J Mol Sci 2013; 14:9618-27. [PMID: 23644889 PMCID: PMC3676802 DOI: 10.3390/ijms14059618] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 04/12/2013] [Accepted: 04/27/2013] [Indexed: 12/17/2022] Open
Abstract
Human enterovirus 71 is one of the major causative agents of hand, foot and mouth disease in children under six years of age. Presently, no vaccines or antiviral drugs have been clinically available to employ against EV71. In this study, we demonstrate that treatment with chebulagic acid reduced the viral cytopathic effect on rhabdomyosarcoma cells with an IC50 of 12.5 μg/mL. The utilization of the chebulagic acid treatment on mice challenged with a lethal dose of enterovirus 71 was able to efficiently reduce mortality and relieve clinical symptoms through the inhibition of viral replication. Chebulagic acid may represent a potential therapeutic agent to control infections to enterovirus 71.
Collapse
|
33
|
Dai JP, Zhao XF, Zeng J, Wan QY, Yang JC, Li WZ, Chen XX, Wang GF, Li KS. Drug screening for autophagy inhibitors based on the dissociation of Beclin1-Bcl2 complex using BiFC technique and mechanism of eugenol on anti-influenza A virus activity. PLoS One 2013; 8:e61026. [PMID: 23613775 PMCID: PMC3628889 DOI: 10.1371/journal.pone.0061026] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 03/05/2013] [Indexed: 02/05/2023] Open
Abstract
Autophagy is involved in many human diseases, such as cancer, cardiovascular disease and virus infection, including human immunodeficiency virus (HIV), hepatitis C virus (HCV), influenza A virus (IAV) and coxsackievirus B3/B4 (CVB3/B4), so a drug screening model targeting autophagy may be very useful for the therapy of these diseases. In our study, we established a drug screening model based on the inhibition of the dissociation of Beclin1-Bcl2 heterodimer, an important negative regulator of autophagy, using bimolecular fluorescence complementation (BiFC) technique for developing novel autophagy inhibitors and anti-IAV agents. From 86 examples of traditional Chinese medicines, we found Syzygium aromaticum L. had the best activity. We then determined the anti-autophagy and anti-IAV activity of eugenol, the major active compound of Syzygium aromaticum L., and explored its mechanism of action. Eugenol could inhibit autophagy and IAV replication, inhibited the activation of ERK, p38MAPK and IKK/NF-κB signal pathways and antagonized the effects of the activators of these pathways. Eugenol also ameliorated the oxidative stress and inhibited the expressions of autophagic genes. We speculated that the mechanism underlying might be that eugenol inhibited the oxidative stress and the activation of ERK1/2, p38MAPK and IKK/NF-κB pathways, subsequently inhibited the dissociation of Beclin1-Bcl2 heterodimer and autophagy, and finally impaired IAV replication. These results might conversely display the reasonableness of the design of our screening model. In conclusion, we have established a drug screening model for developing novel autophagy inhibitor, and find eugenol as a promising inhibitor for autophagy and IAV infection.
Collapse
Affiliation(s)
- Jian-Ping Dai
- 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
| | - Jun Zeng
- 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
| | - Jia-Cai Yang
- 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, United States of America
| | - Xiao-Xuan Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Ge-Fei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
- * E-mail: (G-FW); (K-SL)
| | - Kang-Sheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
- * E-mail: (G-FW); (K-SL)
| |
Collapse
|
34
|
Shang L, Xu M, Yin Z. Antiviral drug discovery for the treatment of enterovirus 71 infections. Antiviral Res 2012; 97:183-94. [PMID: 23261847 DOI: 10.1016/j.antiviral.2012.12.005] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 12/05/2012] [Accepted: 12/06/2012] [Indexed: 12/17/2022]
Abstract
Enterovirus 71 (EV71) is a small, positive-sense, single-stranded RNA virus in the genus Enterovirus, family Picornavirus. It causes hand, foot and mouth disease in infants and children, which in a small percentage of cases progresses to central nervous system infection, ranging from aseptic meningitis to fatal encephalitis. Sporadic cases of EV71 infection occur throughout the world, but large epidemics have occurred recently in Southeast Asia and China. There are currently no approved vaccines or antiviral therapies for the prevention or treatment of EV71 infection. This paper reviews efforts to develop antiviral therapies against EV71.
Collapse
Affiliation(s)
- Luqing Shang
- College of Pharmacy, Nankai University, Tianjin, PR China
| | | | | |
Collapse
|
35
|
Yang Y, Xiu J, Zhang L, Qin C, Liu J. Antiviral activity of punicalagin toward human enterovirus 71 in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2012; 20:67-70. [PMID: 23146421 DOI: 10.1016/j.phymed.2012.08.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/09/2012] [Accepted: 08/23/2012] [Indexed: 06/01/2023]
Abstract
Human enterovirus 71 is one of the major causative agents of hand, foot and mouth disease in children and has caused mortalities in large-scale outbreaks in the Asia-Pacific region in recent years. No vaccine or antiviral therapy is available currently in the clinic. In this work, we investigated the antiviral effect of punicalagin on enterovirus 71 both in vitro and in vivo. The results showed that punicalagin reduced the viral cytopathic effect on rhabdomyosarcoma cells with an IC₅₀) value of 15 μg/ml. Moreover, punicalagin treatment of mice challenged with a lethal dose of enterovirus 71 resulted in a reduction of mortality and relieved clinical symptoms by inhibiting viral replication. Our work suggested that punicalagin have the potential for further development as antiviral agents against enterovirus 71.
Collapse
Affiliation(s)
- Yajun Yang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, CAMS & Comparative Medicine Centre, PUMC, Beijing 100021, China
| | | | | | | | | |
Collapse
|
36
|
Abstract
In recent years, significant progress has been achieved for the development of novel anti-viral drugs. These newly developed drugs belong to three groups of compounds, nucleoside analogues, thymidine kinase-dependent nucleotide analogues and specific viral enzyme inhibitors. It has been found that the natural products, like plant-derived compounds (phytochemicals) as well as traditional medicines, like traditional Chinese medicines (TCM), Ayurvedic medicines and so on, are the important sources for potential and novel anti-viral drugs. In this chapter, the history of natural products as antiviral drugs, the approaches to discover potential lead compounds, and the anti-viral properties of phytochemicals with different action mechanisms are discussed. The key conclusion is that natural products are most important sources for novel anti-viral drugs.
Collapse
|
37
|
Dai JP, Li WZ, Zhao XF, Wang GF, Yang JC, Zhang L, Chen XX, Xu YX, Li KS. A drug screening method based on the autophagy pathway and studies of the mechanism of evodiamine against influenza A virus. PLoS One 2012; 7:e42706. [PMID: 22900043 PMCID: PMC3416798 DOI: 10.1371/journal.pone.0042706] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 07/10/2012] [Indexed: 02/05/2023] Open
Abstract
In this research, we have established a drug screening method based on the autophagy signal pathway using the bimolecular fluorescence complementation-fluorescence resonance energy transfer (BiFC-FRET) technique to develop novel anti-influenza A virus (IAV) drugs. We selected Evodia rutaecarpa Benth out of 83 examples of traditional Chinese medicine and explored the mechanisms of evodiamine, the major active component of Evodia rutaecarpa Benth, on anti-IAV activity. Our results showed that evodiamine could significantly inhibit IAV replication, as determined by a plaque inhibition assay, an IAV vRNA promoter luciferase reporter assay and the Sulforhodamine B method using cytopathic effect (CPE) reduction. Additionally, evodiamine could significantly inhibit the accumulation of LC3-II and p62, and the dot-like aggregation of EGFP-LC3. This compound also inhibited the formation of the Atg5-Atg12/Atg16 heterotrimer, the expressions of Atg5, Atg7 and Atg12, and the cytokine release of TNF-α, IL-1β, IL-6 and IL-8 after IAV infection. Evodiamine inhibited IAV-induced autophagy was also dependent on its action on the AMPK/TSC2/mTOR signal pathway. In conclusion, we have established a new drug screening method, and selected evodiamine as a promising anti-IAV compound.
Collapse
Affiliation(s)
- Jian-Ping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Wei-Zhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, United States of America
| | - Xiang-Feng Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Ge-Fei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jia-Cai Yang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Lin Zhang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiao-Xuan Chen
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yan-Xuan Xu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
| | - Kang-Sheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong, China
- * E-mail:
| |
Collapse
|
38
|
Bagla VP, McGaw LJ, Eloff JN. The antiviral activity of six South African plants traditionally used against infections in ethnoveterinary medicine. Vet Microbiol 2011; 155:198-206. [PMID: 21982126 DOI: 10.1016/j.vetmic.2011.09.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 09/06/2011] [Accepted: 09/12/2011] [Indexed: 11/18/2022]
Abstract
Viral infections remain a major threat to humans and animals and there is a crucial need for new antiviral agents especially with the development of resistant viruses. The hexane, dichloromethane, acetone and methanol extracts of six plant species selected for their traditional use against infections were tested for in vitro antiviral activity against canine distemper virus (CDV), canine parainfluenza virus-2 (CPIV-2), feline herpesvirus-1 (FHV-1) and lumpy skin disease virus (LSDV). All extracts were tested for their cytotoxicity using a colorimetric tetrazolium-based (MTT) assay and were tested for antiviral efficacy at concentrations below CC(50) values on the various cell types used in this study. The antiviral activity of extracts was tested using virucidal and attachment assays. In the virucidal assay, extracts were incubated with virus prior to infection. The most potent inhibition was observed with the acetone and methanol extracts of Podocarpus henkelii against CDV and LSDV, which inhibited replication of the viruses by >75% at 3μg/ml with selectivity index (SI) values ranging between 12 and 45. Excellent activity was also found with the hexane extracts of Plumbago zeylanica and Carissa edulis against CDV, with the extracts reducing viral-induced CPE by 50% and 75% respectively. The hexane extract of C. edulis had moderate activity against FHV-1 with EC(50)<70μg/ml and SI value <2. Only the acetone extract of P. henkelii moderately inhibited replication of LSD virus in the attachment assay, with low activity in other extracts. Of the four extracts with significant antiviral activity, two were prepared from P. henkelii. Therefore, future work will focus on isolating and characterizing the substance(s) responsible for bioactivity in extracts of this species.
Collapse
Affiliation(s)
- Victor P Bagla
- Phytomedicine Programme, Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110, South Africa
| | | | | |
Collapse
|
39
|
Yi L, Lu J, Kung HF, He ML. The virology and developments toward control of human enterovirus 71. Crit Rev Microbiol 2011; 37:313-27. [PMID: 21651436 DOI: 10.3109/1040841x.2011.580723] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Enterovirus 71 (EV71), a member of the Enterovirus genus in the Picornaviridae family, was first recognized as a dermotrophic virus that usually cause mild, self-limiting hand-foot-and-mouth disease (HFMD). However, EV71 infection can sometimes induce a variety of severe neurological complications and even death. Current large outbreaks of EV71 make this virus being a major public health issue. Intense effort has been made to address its underlying pathogenesis and to develop effective means for combating EV71 infections. Here, we aimed to provide an overview of cellular mechanisms underlying EV71 infection and to assess potential agents for prevention and treatment of EV71 infections.
Collapse
Affiliation(s)
- Lina Yi
- Stanley Ho Center for Emerging Infectious Diseases, School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
40
|
Rollinger JM, Schmidtke M. The human rhinovirus: human-pathological impact, mechanisms of antirhinoviral agents, and strategies for their discovery. Med Res Rev 2011; 31:42-92. [PMID: 19714577 PMCID: PMC7168442 DOI: 10.1002/med.20176] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
As the major etiological agent of the common cold, human rhinoviruses (HRV) cause millions of lost working and school days annually. Moreover, clinical studies proved an association between harmless upper respiratory tract infections and more severe diseases e.g. sinusitis, asthma, and chronic obstructive pulmonary disease. Both the medicinal and socio-economic impact of HRV infections and the lack of antiviral drugs substantiate the need for intensive antiviral research. A common structural feature of the approximately 100 HRV serotypes is the icosahedrally shaped capsid formed by 60 identical copies of viral capsid proteins VP1-4. The capsid protects the single-stranded, positive sense RNA genome of about 7,400 bases in length. Both structural as well as nonstructural proteins produced during the viral life cycle have been identified as potential targets for blocking viral replication at the step of attachment, entry, uncoating, RNA and protein synthesis by synthetic or natural compounds. Moreover, interferon and phytoceuticals were shown to protect host cells. Most of the known inhibitors of HRV replication were discovered as a result of empirical or semi-empirical screening in cell culture. Structure-activity relationship studies are used for hit optimization and lead structure discovery. The increasing structural insight and molecular understanding of viral proteins on the one hand and the advent of innovative computer-assisted technologies on the other hand have facilitated a rationalized access for the discovery of small chemical entities with antirhinoviral (anti-HRV) activity. This review will (i) summarize existing structural knowledge about HRV, (ii) focus on mechanisms of anti-HRV agents from synthetic and natural origin, and (iii) demonstrate strategies for efficient lead structure discovery.
Collapse
Affiliation(s)
- Judith M Rollinger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 52c, A-6020 Innsbruck, Austria.
| | | |
Collapse
|
41
|
Yi L, He Y, Chen Y, Kung HF, He ML. Potent inhibition of human enterovirus 71 replication by type I interferon subtypes. Antivir Ther 2011; 16:51-8. [DOI: 10.3851/imp1720] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
42
|
Wu KX, Ng MML, Chu JJH. Developments towards antiviral therapies against enterovirus 71. Drug Discov Today 2010; 15:1041-51. [PMID: 20974282 PMCID: PMC7108380 DOI: 10.1016/j.drudis.2010.10.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 09/20/2010] [Accepted: 10/18/2010] [Indexed: 12/21/2022]
Abstract
Enterovirus 71 (EV71) has emerged as a clinically important neurotropic virus that can cause acute flaccid paralysis and encephalitis, leading to cardiopulmonary failure and death. Recurring outbreaks of EV71 have been reported in several countries. The current lack of approved anti-EV71 therapy has prompted intense research into antiviral development. Several strategies--ranging from target-based chemical design to compound library screenings--have been employed, while others revisited compound series generated from antiviral developments against poliovirus and human rhinoviruses. These efforts have given rise to a diversity of antiviral candidates that include small molecules and non-conventional nucleic-acid-based strategies. This review aims to highlight candidates with potential for further clinical development based on their putative modes of action.
Collapse
Affiliation(s)
- Kan X Wu
- Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System, 5 Science Drive 2, National University of Singapore, Singapore
| | | | | |
Collapse
|
43
|
Choi HJ, Song JH, Lim CH, Baek SH, Kwon DH. Anti-human rhinovirus activity of raoulic acid from Raoulia australis. J Med Food 2010; 13:326-8. [PMID: 20412019 DOI: 10.1089/jmf.2009.1149] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human rhinoviruses (HRVs), members of the Picornaviridae family, are composed of over 100 different virus serotypes. Until now there is no recorded clinically effective antiviral chemotherapeutic agent for treatment of diseases caused by HRVs. Our previous study of raoulic acid tested against serotype human rhinoviruses showed anti-HRV2 (species A) and -3 (species B) activities. In this study, raoulic acid was found to possess broad-spectrum antiviral activity against six HRVs with a 50% inhibition concentration of less than 9.5 microg/mL through inhibition of the cellular absorption of the HRV particles. Furthermore, the effect of raoulic acid on resistance of HRV5 exhibited to pleconaril was more pronounced than the effect on HRV1b, -6, -14, -15, and -40. However, ribavirin did possess weak antiviral activity against HRVs. Collectively, the results demonstrate that raoulic acid is a novel therapeutic candidate for two different groups of human rhinovirus.
Collapse
Affiliation(s)
- Hwa Jung Choi
- Department of Clinic Laboratory Science, Daejeon Health Sciences College, Daejeon, Republic of Korea
| | | | | | | | | |
Collapse
|