1
|
Xiong Y, Tao K, Li T, Zhou Y, Zhang Z, Ou W, Wang Z, Wang S, Hou Y, Cao P, Ji J. Both chebulagic acid and punicalagin inhibit respiratory syncytial virus entry via multi-targeting glycoprotein and fusion protein. J Virol 2024; 98:e0153624. [PMID: 39508604 DOI: 10.1128/jvi.01536-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections, with no currently available small-molecule drugs that are both safe and effective. A major obstacle in antiviral drug development is the rapid emergence of drug-resistant viral strains. Targeting multiple viral compounds may help mitigate the development of resistance. Herein, we conducted a drug screening using the Antiviral Traditional Chinese Medicine Active Compound Library, aiming to identify compounds that simultaneously target the RSV fusion (F) protein, glycoprotein (G), and the host heparan sulfate proteoglycans (HSPGs). From this screening, 10 candidate compounds were identified for their ability to interact with all three targets. Among these 10 candidates, chebulagic acid (CHLA) and punicalagin (PUG) demonstrated the most potent inhibition of RSV replication. In vitro dose-response assays confirmed the antiviral efficacy of CHLA (IC50: 0.07864 µM) and PUG (IC50: 0.08065 µM). Further experiments revealed both CHLA and PUG disrupt RSV attachment and membrane fusion by targeting the RSV-F and G proteins, rather than HSPG. Notably, CHLA and PUG were found to bind to the CX3C motif of the RSV-G protein, with docking assays predicting their binding sites at cysteines 176 and 182. Additionally, CHLA enhanced the conformational stability of the RSV-F protein before fusion. In an in vivo study, both CHLA and PUG were shown to alleviate RSV-induced pulmonary pathology by reducing viral titers, mitigating lung injury, and suppressing the inflammatory responses in the lungs. Our findings suggest that CHLA and PUG hold potential as therapeutic agents for RSV infection.IMPORTANCEA significant challenge in developing anti-respiratory syncytial virus (RSV) agents is the rapid emergence of resistant viral strains. Designing drugs that target multiple viral components can effectively reduce the likelihood of developing resistant strains. In this study, we screened compounds from the Antiviral Traditional Chinese Medicine Active Compound Library, aiming to simultaneously targe the RSV fusion (F) protein, glycoprotein (G), and host heparan sulfate proteoglycans (HSPGs). Our findings revealed that chebulagic acid (CHLA) and punicalagin (PUG) significantly inhibited RSV replication both in vitro and in vivo and interacted with all three targets. Both CHLA and PUG were able to disrupt RSV attachment and membrane fusion. Mechanistically, CHLA and PUG were found to bind to the CX3C motif of the RSV-G protein, with CHLA also enhancing the conformational stability of the RSV-F protein before fusion. In conclusion, our study suggests that CHLA and PUG hold promise as therapeutic agents against RSV infection.
Collapse
Affiliation(s)
- Yingcai Xiong
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Keyu Tao
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tao Li
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yinghui Zhou
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaowei Zhang
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiying Ou
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhao Wang
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shouchuan Wang
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Peng Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Medical Innovation Center, Afliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
Geng R, Yin D, Liu Y, Lv H, Zhou X, Bao C, Gong L, Shao H, Qian K, Chen H, Qin A. Punicalagin Inhibits African Swine Fever Virus Replication by Targeting Early Viral Stages and Modulating Inflammatory Pathways. Vet Sci 2024; 11:440. [PMID: 39330819 PMCID: PMC11435760 DOI: 10.3390/vetsci11090440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
African swine fever (ASF), caused by the African swine fever virus (ASFV), has resulted in significant losses in the global pig industry. Considering the absence of effective vaccines, developing drugs against ASFV may be a crucial strategy for its prevention and control in the future. In this study, punicalagin, a polyphenolic substance extracted from pomegranate peel, was found to significantly inhibit ASFV replication in MA-104, PK-15, WSL, and 3D4/21 cells by screening an antiviral compound library containing 536 compounds. Time-of-addition studies demonstrated that punicalagin acted on early viral replication stages, impinging on viral attachment and internalization. Meanwhile, punicalagin could directly inactivate the virus according to virucidal assay. RT-qPCR and Western blot results indicated that punicalagin modulated the NF-κB/STAT3/NLRP3 inflammasome signaling pathway and reduced the levels of inflammatory mediators induced by ASFV. In conclusion, this study reveals the anti-ASFV activity of punicalagin and the mechanism of action, which may have great potential for developing effective drugs against ASFV.
Collapse
Affiliation(s)
- Renhao Geng
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Dan Yin
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Yingnan Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Hui Lv
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiaoyu Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Chunhui Bao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Lang Gong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China
| | - Hongxia Shao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Kun Qian
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Hongjun Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Aijian Qin
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
3
|
Li C, Li Y, Huang X, Li S, Sangji K, Gu R. Traditional Tibetan medicine: therapeutic potential in lung diseases. Front Pharmacol 2024; 15:1365911. [PMID: 38567353 PMCID: PMC10986185 DOI: 10.3389/fphar.2024.1365911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Lung diseases have become a major threat to human health worldwide. Despite advances in treatment and intervention in recent years, effective drugs are still lacking for many lung diseases. As a traditional natural medicine, Tibetan medicine has had a long history of medicinal use in ethnic minority areas, and from ancient times to the present, it has a good effect on the treatment of lung diseases and has attracted more and more attention. In this review, a total of 586 Tibetan medicines were compiled through literature research of 25 classical works on Tibetan medicine, drug standards, and some Chinese and English databases. Among them, 33 Tibetan medicines have been studied to show their effectiveness in treating lung diseases. To investigate the uses of these Tibetan medicines in greater depth, we have reviewed the ethnomedicinal, phytochemical and pharmacological properties of the four commonly used Tibetan medicines for lung diseases (rhodiola, gentian, sea buckthorn, liexiang dujuan) and the five most frequently used Tibetan medicines (safflower, licorice, sandalwood, costus, myrobalan). It is expected to provide some reference for the development of new drugs of lung diseases in the future.
Collapse
Affiliation(s)
- Canlin Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xi Huang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Si Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kangzhuo Sangji
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Gu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
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
|
5
|
Sultan MT, Anwar MJ, Imran M, Khalil I, Saeed F, Neelum S, Alsagaby SA, Al Abdulmonem W, Abdelgawad MA, Hussain M, El-Ghorab AH, Umar M, Al Jbawi E. Phytochemical profile and pro-healthy properties of Terminalia chebula: A comprehensive review. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2023. [DOI: 10.1080/10942912.2023.2166951] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
| | | | - Muhammad Imran
- Department of Food Science and Technology, University of Narowal-Pakistan, Narowal, Pakistan
| | - Ijaz Khalil
- Institute of Food and Nutrition, Bahauddin Zakariya University, Multan, Pakistan
| | - Farhan Saeed
- Department of Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Shahzadi Neelum
- Department of Biochemistry, Hamdard University, Karachi, Pakistan
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Muzzamal Hussain
- Department of Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ahmed H. El-Ghorab
- Department of Chemistry, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Maryam Umar
- Department of Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | | |
Collapse
|
6
|
Thakur M, Singh M, Kumar S, Dwivedi VP, Dakal TC, Yadav V. A Reappraisal of the Antiviral Properties of and Immune Regulation through Dietary Phytochemicals. ACS Pharmacol Transl Sci 2023; 6:1600-1615. [PMID: 37974620 PMCID: PMC10644413 DOI: 10.1021/acsptsci.3c00178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Indexed: 11/19/2023]
Abstract
In the present era of the COVID-19 pandemic, viral infections remain a major cause of morbidity and mortality worldwide. In this day and age, viral infections are rampant and spreading rapidly. Among the most aggressive viral infections are ebola, AIDS (acquired immunodeficiency syndrome), influenza, and SARS (severe acute respiratory syndrome). Even though there are few treatment options for viral diseases, most of the antiviral therapies are ineffective owing to frequent mutations, the development of more aggressive strains, drug resistance, and possible side effects. Traditionally, herbal remedies have been used by healers, including for dietary and medicinal purposes. Many clinical and scientific studies have demonstrated the therapeutic potential of plant-derived natural compounds. Because of unsafe practices like blood transfusions and organ transplants from infected patients, medical supply contamination. Our antiviral therapies cannot achieve sterile immunity, and we have yet to find a cure for these pernicious infections. Herbs have been shown to improve therapeutic efficacy against a wide variety of viral diseases because of their high concentration of immunomodulatory phytochemicals (both immunoinhibitory and anti-inflammatory). Combined with biotechnology, this folk medicine system can lead to the development of novel antiviral drugs and therapies. In this Review, we will summarize some selected bioactive compounds with probable mechanisms of their antiviral actions, focusing on the immunological axis of these compounds.
Collapse
Affiliation(s)
- Mony Thakur
- Department
of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Mona Singh
- Department
of Obstetrics and Gynaecology, Medical College
of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Sandeep Kumar
- Division
of Cell Biology and Immunology, Council
of Scientific and Industrial Research - Institute of Microbial Technology, Chandigarh 160036, India
| | - Ved Prakash Dwivedi
- International
Centre for Genetic Engineering and Biotechnology, ICGEB Campus, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Tikam Chand Dakal
- Genome
and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001, India
| | - Vinod Yadav
- Department
of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| |
Collapse
|
7
|
Liu CH, Kuo YT, Lin CJ, Lin LT. Involvement of cell surface glycosaminoglycans in chebulagic acid's and punicalagin's antiviral activities against Coxsackievirus A16 infection. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155047. [PMID: 37690230 DOI: 10.1016/j.phymed.2023.155047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/11/2023] [Accepted: 08/24/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Coxsackievirus A16 (CVA16) is responsible for several recent outbreaks of Hand, Foot, and Mouth Disease in the Asia-Pacific region, and there are currently no vaccines or specific treatments available. We have previously identified two tannins, chebulagic acid (CHLA) and punicalagin (PUG), as efficient entry inhibitors against multiple viruses known to engage cell surface glycosaminoglycans (GAGs). Interestingly, these two phytochemicals could also block enterovirus infection by directly inactivating CVA16 virions, which were recently reported to utilize GAGs to mediate its entry. PURPOSE The aim of this study is to evaluate the involvement of GAGs in the anti-CVA16 activities of CHLA and PUG. METHODS To explore a potential mechanistic link, the role of GAGs in promoting CVA16 entry was first confirmed by treating human rhabdomyosarcoma (RD) cells with soluble heparin or GAG lyases including heparinase and chondroitinase. We then performed a combination treatment of CHLA or PUG with the GAG interaction inhibitors to assess whether CHLA's and PUG's anti-CVA16 activities were related to GAG competition. Molecular docking and surface plasmon resonance (SPR) were conducted to analyze the interactions between CHLA, PUG, and CVA16 capsid. Lastly, CRISPR/Cas9 knockout (KO) of the Exostosin glycosyltransferase 1 (EXT1) gene, which encodes a transmembrane glycosyltransferase involved in heparan sulfate biosynthesis, was used to validate the importance of GAGs in CHLA's and PUG's antiviral effects. RESULTS Intriguingly, combining GAG inhibition via heparin/GAG lyases treatments with CHLA and PUG revealed that their inhibitory activities against CVA16 infection were overlapping. Further molecular docking analysis indicated that the predicted binding sites of CHLA and PUG on the CVA16 capsid are in proximity to the putative residues recognized for GAG interaction, thus pointing to potential interference with the CVA16-GAG association. SPR analysis also confirmed the direct binding of CHLA and PUG to CVA16 capsid. Finally, RD cells with EXT1 KO decreased CHLA's and PUG's antiviral effect on CVA16 infection. CONCLUSION Altogether, our results suggest that CHLA and PUG bind to CVA16 capsid and prevent the virus' interaction with heparan sulfate and chondroitin sulfate for its entry. This study provides mechanistic insight into the antiviral activity of CHLA and PUG against CVA16, which may be helpful for the development of antiviral strategies against the enterovirus.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Yu-Ting Kuo
- Department of Medical Imaging, Chi Mei Medical Center, Tainan 710, Taiwan
| | - Chien-Ju Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
8
|
Ribelato EV, Wouk J, Celestino GG, Rodrigues BCD, Darido MLG, Barboza MGL, Botura TJ, de Oliveira MC, de Andrade FG, Lonni AASG, de Mello JCP, da Rocha SPD, Faccin-Galhardi LC. Topical formulations containing Trichilia catigua extract as therapeutic options for a genital and an acyclovir-resistant strain of herpes recurrent infection. Braz J Microbiol 2023; 54:1501-1511. [PMID: 37338788 PMCID: PMC10485181 DOI: 10.1007/s42770-023-01027-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/30/2023] [Indexed: 06/21/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) infect, respectively, 67% and 13% of the world population, most commonly causing mild symptoms, such as blisters/ulcers. However, severe conditions such as keratitis, encephalitis, and systemic infections may occur, generally associated with the patient's immunological condition. Although Acyclovir® (ACV) and its analogs are the reference drugs for herpetic infections, the number of ACV-resistant HSV infections is growing exponentially. Therefore, new natural products' bioactive compounds have been studied to develop novel effective anti-herpetics. Trichilia catigua is a plant widely used in traditional medicine, including the treatment of skin diseases and sexual infections. In our study, 16 extracts from the bark of T. catigua, obtained with different solvents and their combinations, were evaluated against HSV-1 AR and HSV-2, respectively, ACV resistance and genital strains in vitro. The extracts with the highest selectivity index were used to prepare new topical anti-herpetic formulations and confirmed in vivo. Two new topical formulations were suggested to treat cutaneous and genital herpetic recurrent lesions. The cytotoxicity and antiviral activity were tested using the MTT method. The cytotoxic (CC50) and inhibitory (IC50) concentrations of 50% and the selectivity index (SI: CC50/IC50) were determined. Tc12, Tc13, and Tc16 were added to the formulations. Infected BALB/c mice were treated for 8 days, and the severity of the herpetic lesions was analyzed daily. All CEs showed a CC50 value ranging from 143 to 400 µg/mL, except for Tc3 and Tc10. Tc12, Tc13, and Tc16 showed the best SI in the 0 h, virucidal, and adsorption inhibition assays. In the in vivo test against HSV-1 AR, the infected animals treated with creams were statistically different from the infected non-treated animals and similar to ACV-treated mice. In HSV-2-infected genitalia, similar effects were found for Tc13 and Tc16 gels. The present study demonstrated that extracts from the bark of T. catigua, traditionally used in folk medicine, are a valuable source of active compounds with anti-herpetic activity. The extracts showed a virucidal mechanism of action and prevented the initial stages of viral replication. The cutaneous and genital infections were strongly inhibited by the Tc12, Tc13, and Tc16 extracts. New topical therapeutic alternatives using Trichilia catigua extracts are suggested for patients infected with ACV-resistant strains of HSV.
Collapse
Affiliation(s)
- Elisa Vicente Ribelato
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Jéssica Wouk
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Gabriela Gomes Celestino
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Bianca Cerqueira Dias Rodrigues
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Maria Laura Goussain Darido
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Mario Gabriel Lopes Barboza
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Tatiana Jabor Botura
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Maylla Cardoso de Oliveira
- Departamento de Histologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Fábio Goulart de Andrade
- Departamento de Histologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Audrey A S G Lonni
- Departamento de Ciências Farmacêuticas-Centro de Ciências da Saúde, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - João Carlos Palazzo de Mello
- Laboratório de Biologia Farmacêutica, Palafito, Departamento de Farmácia, Universidade Estadual de Maringá (UEM), Maringá, Paraná, Brazil
| | - Sérgio Paulo Dejato da Rocha
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil
| | - Lígia C Faccin-Galhardi
- Laboratório de Virologia, Departamento de Microbiologia-Centro de Ciências Biológicas, Universidade Estadual de Londrina (UEL), Londrina, Paraná, Brazil.
| |
Collapse
|
9
|
Pennisi R, Trischitta P, Tamburello MP, Barreca D, Mandalari G, Sciortino MT. Mechanistic Understanding of the Antiviral Properties of Pistachios and Zeaxanthin against HSV-1. Viruses 2023; 15:1651. [PMID: 37631995 PMCID: PMC10459438 DOI: 10.3390/v15081651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
The search for alternative clinical treatments to fight resistance and find alternative antiviral treatments for the herpes simplex virus (HSV) is of great interest. Plants are rich sources of novel antiviral, pharmacologically active agents that provide several advantages, including reduced side effects, less resistance, low toxicity, and different mechanisms of action. In the present work, the antiviral activity of Californian natural raw (NRRE) and roasted unsalted (RURE) pistachio polyphenols-rich extracts was evaluated against HSV-1 using VERO cells. Two different extraction methods, with or without n-hexane, were used. Results showed that n-hexane-extracted NRRE and RURE exerted an antiviral effect against HSV-1, blocking virus binding on the cell surface, affecting viral DNA synthesis as well as accumulation of ICP0, UL42, and Us11 viral proteins. Additionally, the identification and quantification of phenolic compounds by RP-HPLC-DAD confirmed that extraction with n-hexane exclusively accumulated tocopherols, carotenoids, and xanthophylls. Amongst these, zeaxanthin exhibited strong antiviral activity against HSV-1 (CC50: 16.1 µM, EC50 4.08 µM, SI 3.96), affecting both the viral attachment and penetration and viral DNA synthesis. Zeaxanthin is a dietary carotenoid that accumulates in the retina as a macular pigment. The use of pistachio extracts and derivates should be encouraged for the topical treatment of ocular herpetic infections.
Collapse
Affiliation(s)
- Rosamaria Pennisi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.); (D.B.); (M.T.S.)
| | - Paola Trischitta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.); (D.B.); (M.T.S.)
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Maria Pia Tamburello
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.); (D.B.); (M.T.S.)
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.); (D.B.); (M.T.S.)
| | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.); (D.B.); (M.T.S.)
| | - Maria Teresa Sciortino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (P.T.); (M.P.T.); (D.B.); (M.T.S.)
| |
Collapse
|
10
|
Elebeedy D, Ghanem A, Aly SH, Ali MA, Faraag AHI, El-Ashrey MK, salem AM, Hassab MAE, Maksoud AIAE. Synergistic antiviral activity of Lactobacillus acidophilus and Glycyrrhiza glabra against Herpes Simplex-1 Virus (HSV-1) and Vesicular Stomatitis Virus (VSV): experimental and In Silico insights. BMC Microbiol 2023; 23:173. [PMID: 37391715 PMCID: PMC10311774 DOI: 10.1186/s12866-023-02911-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/25/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND The emergence of different viral infections calls for the development of new, effective, and safe antiviral drugs. Glycyrrhiza glabra is a well-known herbal remedy possessing antiviral properties. OBJECTIVE The objective of our research was to evaluate the effectiveness of a newly developed combination of the probiotics Lactobacillus acidophilus and G. glabra root extract against two viral models, namely the DNA virus Herpes simplex virus-1 (HSV-1) and the RNA virus Vesicular Stomatitis Virus (VSV), with regards to their antiviral properties. METHODOLOGY To examine the antiviral impacts of various treatments, we employed the MTT assay and real-time PCR methodology. RESULTS The findings of our study indicate that the co-administration of L. acidophilus and G. glabra resulted in a significant improvement in the survival rate of Vero cells, while also leading to a reduction in the titers of Herpes Simplex Virus Type 1 (HSV-1) and Vesicular Stomatitis Virus (VSV) in comparison to cells that were not treated. Additionally, an investigation was conducted on glycyrrhizin, the primary constituent of G. glabra extract, utilizing molecular docking techniques. The results indicated that glycyrrhizin exhibited a greater binding energy score for HSV-1 polymerase (- 22.45 kcal/mol) and VSV nucleocapsid (- 19.77 kcal/mol) in comparison to the cocrystallized ligand (- 13.31 and - 11.44 kcal/mol, respectively). CONCLUSIONS The combination of L. acidophilus and G. glabra extract can be used to develop a new, natural antiviral agent that is safe and effective.
Collapse
Affiliation(s)
- Dalia Elebeedy
- Department of Pharmaceutical Biotechnology Faculty of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo, Badr City, 11829 Cairo Egypt
| | - Shaza H. Aly
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo, Badr City, Cairo, 11829 Egypt
| | - Mohamed A. Ali
- School of Biotechnology, Badr University in Cairo, Badr City, 11829 Cairo Egypt
| | - Ahmed H. I. Faraag
- School of Biotechnology, Badr University in Cairo, Badr City, 11829 Cairo Egypt
- Botany and Microbiology Department, Faculty of Science, Helwan University, Ain Helwan, Cairo, 11795 Egypt
| | - Mohamed K. El-Ashrey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman Inter-National University, Ras Sudr, Egypt
| | - Aya M. salem
- Faculty of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Mahmoud A. El Hassab
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman Inter-National University, Ras Sudr, Egypt
| | - Ahmed I. Abd El Maksoud
- Industrial Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Monufia, Egypt
| |
Collapse
|
11
|
Tzen JTC. Strictinin: A Key Ingredient of Tea. Molecules 2023; 28:molecules28093961. [PMID: 37175375 PMCID: PMC10180463 DOI: 10.3390/molecules28093961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/26/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023] Open
Abstract
Strictinin is a relatively tiny ellagitannin, which is found in many plants as a minor constituent. Catechins are known as the major constituents in the young leaves of most tea plants, while strictinin was found as a major constituent in the Pu'er tea plant. In some Pu'er tea varieties, strictinin was identified as the most abundant phenolic compound rather than catechins. In the past decade, strictinin was demonstrated to possess several functional activities, including antiviral, antibacterial, anti-obesity, laxative, anticaries, anti-allergic, antipsoriatic, antihyperuricemia, antidiabetic, and anticancer effects. These functional activities were in accordance with the therapeutic effects empirically perceived for Pu'er tea. Evidently, strictinin is the key ingredient in Pu'er tea that acts as a herbal medicine. In functionally-based applications, an instant powder of Pu'er tea infusion was formulated as an active raw material to be supplemented in food, cosmetics, and beverages; a new type of tea named Bitter Citrus Tzen Tea was developed by combining three teas empirically consumed to expel the cold, and new edible oral care products were designed for caries prevention by supplementation with Pu'er tea extract. More functional activities and practical applications of strictinin are scientifically anticipated in follow-up research.
Collapse
Affiliation(s)
- Jason T C Tzen
- Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung 402, Taiwan
| |
Collapse
|
12
|
Alexova R, Alexandrova S, Dragomanova S, Kalfin R, Solak A, Mehan S, Petralia MC, Fagone P, Mangano K, Nicoletti F, Tancheva L. Anti-COVID-19 Potential of Ellagic Acid and Polyphenols of Punica granatum L. Molecules 2023; 28:molecules28093772. [PMID: 37175181 PMCID: PMC10180134 DOI: 10.3390/molecules28093772] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Pomegranate (Punica granatum L.) is a rich source of polyphenols, including ellagitannins and ellagic acid. The plant is used in traditional medicine, and its purified components can provide anti-inflammatory and antioxidant activity and support of host defenses during viral infection and recovery from disease. Current data show that pomegranate polyphenol extract and its ellagitannin components and metabolites exert their beneficial effects by controlling immune cell infiltration, regulating the cytokine secretion and reactive oxygen and nitrogen species production, and by modulating the activity of the NFκB pathway. In vitro, pomegranate extracts and ellagitannins interact with and inhibit the infectivity of a range of viruses, including SARS-CoV-2. In silico docking studies show that ellagitannins bind to several SARS-CoV-2 and human proteins, including a number of proteases. This warrants further exploration of polyphenol-viral and polyphenol-host interactions in in vitro and in vivo studies. Pomegranate extracts, ellagitannins and ellagic acid are promising agents to target the SARS-CoV-2 virus and to restrict the host inflammatory response to viral infections, as well as to supplement the depleted host antioxidant levels during the stage of recovery from COVID-19.
Collapse
Affiliation(s)
- Ralitza Alexova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University-Sofia, Zdrave Str. 2, 1431 Sofia, Bulgaria
| | - Simona Alexandrova
- Department of Biological Effects of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 23, 1113 Sofia, Bulgaria
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University, Marin Drinov Str. 55, 9002 Varna, Bulgaria
| | - Reni Kalfin
- Department of Biological Effects of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 23, 1113 Sofia, Bulgaria
- Department of Healthcare, South-West University "Neofit Rilski", Ivan Mihailov Str. 66, 2700 Blagoevgrad, Bulgaria
| | - Ayten Solak
- Institute of Cryobiology and Food Technologies, Cherni Vrah Blvd. 5, 1407 Sofia, Bulgaria
| | - Sidharth Mehan
- Department of Pharmacology, Division of Neuroscience, ISF College of Pharmacy, Moga 142001, India
| | - Maria Cristina Petralia
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Lyubka Tancheva
- Department of Biological Effects of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 23, 1113 Sofia, Bulgaria
| |
Collapse
|
13
|
Yin W, Ding L. Nanocarrier-based drug delivery system in herpes simplex virus treatment. Future Virol 2023. [DOI: 10.2217/fvl-2022-0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Herpes simplex virus (HSV) is a highly contagious DNA virus that affects the majority of people worldwide. HSV establishes a latent infection in the ganglia, where it can reactivate, leading to recurrent disease. Currently, there are many experimental vaccines against HSV, but none have been used to treat herpes infections. At the same time, the therapeutic effect of existing anti-HSV drugs is limited. Nanocarriers, which deliver drugs to specific targets, have been used in different diseases, including viral infections. Nanocarriers could be designed to encapsulate drugs and directly target infected cells. This review will describe in detail the use of nanocarriers for targeted therapy of HSV infection.
Collapse
Affiliation(s)
- Wei Yin
- Department of Radiology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science & Technology, Xianning, China
| | - Liqiong Ding
- Department of Pharmaceutics, School of Pharmacy, Hubei University of Science & Technology, Xianning, China
| |
Collapse
|
14
|
Zhang K, Wang L, Peng J, Sangji K, Luo Y, Zeng Y, Zeweng Y, Fan G. Traditional Tibetan medicine to fight against COVID-19: Basic theory and therapeutic drugs. Front Pharmacol 2023; 14:1098253. [PMID: 36874035 PMCID: PMC9978713 DOI: 10.3389/fphar.2023.1098253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
The Coronavirus Diseases 2019 (COVID-19) has been rapidly spreading globally and has caused severe harm to the health of people and a substantial social burden. In response to this situation, experts around the world have considered various treatments, including the use of traditional medicine. Traditional Tibetan medicine (TTM), one of the traditional medicines in China, has played an important role in the treatment of infectious diseases in history. It has formed a solid theoretical foundation and accumulated rich experience in the treatment of infectious diseases. In this review, we provide a comprehensive introduction to the basic theory, treatment strategies, and commonly used drugs of TTM for the treatment of COVID-19. In addition, the efficacies and potential mechanisms of these TTM drugs against COVID-19 are discussed based on available experimental data. This review may provide important information for the basic research, clinical application and drug development of traditional medicines for the treatment of COVID-19 or other infectious diseases. More pharmacological studies are needed to reveal the therapeutic mechanisms and active ingredients of TTM drugs in the treatment of COVID-19.
Collapse
Affiliation(s)
- Kun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijie Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayan Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kangzhuo Sangji
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuting Luo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yujiao Zeng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongzhong Zeweng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gang Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Sivarajan R, Oberwinkler H, Roll V, König EM, Steinke M, Bodem J. A defined anthocyanin mixture sourced from bilberry and black currant inhibits Measles virus and various herpesviruses. BMC Complement Med Ther 2022; 22:181. [PMID: 35804339 PMCID: PMC9264518 DOI: 10.1186/s12906-022-03661-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Anthocyanin-containing plant extracts and carotenoids, such as astaxanthin, have been well-known for their antiviral and anti-inflammatory activity, respectively. We hypothesised that a mixture of Ribes nigrum L. (Grossulariaceae) (common name black currant (BC)) and Vaccinium myrtillus L. (Ericaceae) (common name bilberry (BL)) extracts (BC/BL) with standardised anthocyanin content as well as single plant extracts interfered with the replication of Measles virus and Herpesviruses in vitro.
Methods
We treated cell cultures with BC/BL or defined single plant extracts, purified anthocyanins and astaxanthin in different concentrations and subsequently infected the cultures with the Measles virus (wild-type or vaccine strain Edmonston), Herpesvirus 1 or 8, or murine Cytomegalovirus. Then, we analysed the number of infected cells and viral infectivity and compared the data to non-treated controls.
Results
The BC/BL extract inhibited wild-type Measles virus replication, syncytia formation and cell-to-cell spread. This suppression was dependent on the wild-type virus-receptor-interaction since the Measles vaccine strain was unaffected by BC/BL treatment. Furthermore, the evidence was provided that the delphinidin-3-rutinoside chloride, a component of BC/BL, and purified astaxanthin, were effective anti-Measles virus compounds. Human Herpesvirus 1 and murine Cytomegalovirus replication was inhibited by BC/BL, single bilberry or black currant extracts, and the BC/BL component delphinidin-3-glucoside chloride. Additionally, we observed that BC/BL seemed to act synergistically with aciclovir. Moreover, BC/BL, the single bilberry and black currant extracts, and the BC/BL components delphinidin-3-glucoside chloride, cyanidin-3-glucoside, delphinidin-3-rutinoside chloride, and petunidin-3-galactoside inhibited human Herpesvirus 8 replication.
Conclusions
Our data indicate that Measles viruses and Herpesviruses are differentially susceptible to a specific BC/BL mixture, single plant extracts, purified anthocyanins and astaxanthin. These compounds might be used in the prevention of viral diseases and in addition to direct-acting antivirals, such as aciclovir.
Collapse
|
16
|
EL-Aguel A, Pennisi R, Smeriglio A, Kallel I, Tamburello MP, D’Arrigo M, Barreca D, Gargouri A, Trombetta D, Mandalari G, Sciortino MT. Punica granatum Peel and Leaf Extracts as Promising Strategies for HSV-1 Treatment. Viruses 2022; 14:v14122639. [PMID: 36560643 PMCID: PMC9782130 DOI: 10.3390/v14122639] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Punica granatum is a rich source of bioactive compounds which exhibit various biological effects. In this study, pomegranate peel and leaf ethanolic crude extracts (PPE and PLE, respectively) were phytochemically characterized and screened for antioxidant, antimicrobial and antiviral activity. LC-PDA-ESI-MS analysis led to the identification of different compounds, including ellagitannins, flavonoids and phenolic acids. The low IC50 values, obtained by DPPH and FRAP assays, showed a noticeable antioxidant effect of PPE and PLE comparable to the reference standards. Both crude extracts and their main compounds (gallic acid, ellagic acid and punicalagin) were not toxic on Vero cells and exhibited a remarkable inhibitory effect on herpes simplex type 1 (HSV-1) viral plaques formation. Specifically, PPE inhibited HSV-1 adsorption to the cell surface more than PLE. Indeed, the viral DNA accumulation, the transcription of viral genes and the expression of viral proteins were significantly affected by PPE treatment. Amongst the compounds, punicalagin, which is abundant in PPE crude extract, inhibited HSV-1 replication, reducing viral DNA and transcripts accumulation, as well as proteins of all three phases of the viral replication cascade. In contrast, no antibacterial activity was detected. In conclusion, our findings indicate that Punica granatum peel and leaf extracts, especially punicalagin, could be a promising therapeutic candidate against HSV-1.
Collapse
Affiliation(s)
- Asma EL-Aguel
- Research Laboratory Toxicology-Environmental Microbiology and Health (LR17ES06), Faculty of Sciences of Sfax, P.O. Box 1171, Sfax 3000, Tunisia
| | - Rosamaria Pennisi
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
- Correspondence: (R.P.); (G.M.)
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Imen Kallel
- Research Laboratory Toxicology-Environmental Microbiology and Health (LR17ES06), Faculty of Sciences of Sfax, P.O. Box 1171, Sfax 3000, Tunisia
| | - Maria Pia Tamburello
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Manuela D’Arrigo
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Ahmed Gargouri
- Research Laboratory Toxicology-Environmental Microbiology and Health (LR17ES06), Faculty of Sciences of Sfax, P.O. Box 1171, Sfax 3000, Tunisia
| | - Domenico Trombetta
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
- Correspondence: (R.P.); (G.M.)
| | - Maria Teresa Sciortino
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
17
|
Tomaszewska E, Ranoszek-Soliwoda K, Bednarczyk K, Lech A, Janicka M, Chodkowski M, Psarski M, Celichowski G, Krzyzowska M, Grobelny J. Anti-HSV Activity of Metallic Nanoparticles Functionalized with Sulfonates vs. Polyphenols. Int J Mol Sci 2022; 23:13104. [PMID: 36361890 PMCID: PMC9657688 DOI: 10.3390/ijms232113104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 10/15/2023] Open
Abstract
Metallic nanoparticles exhibit broad-spectrum activity against bacteria, fungi, and viruses. The antiviral activity of nanoparticles results from the multivalent interactions of nanoparticles with viral surface components, which result from the nanometer size of the material and the presence of functional compounds adsorbed on the nanomaterial surface. A critical step in the virus infection process is docking and entry of the virus into the host cell. This stage of the infection can be influenced by functional nanomaterials that exhibit high affinity to the virus surface and hence can disrupt the infection process. The affinity of the virus to the nanomaterial surface can be tuned by the specific surface functionalization of the nanomaterial. The main purpose of this work was to determine the influence of the ligand type present on nanomaterial on the antiviral properties against herpes simplex virus type 1 and 2. We investigated the metallic nanoparticles (gold and silver) with different sizes (5 nm and 30 nm), coated either with polyphenol (tannic acid) or sulfonates (ligands with terminated sulfonate groups). We found that the antiviral activity of nano-conjugates depends significantly on the ligand type present on the nanoparticle surface.
Collapse
Affiliation(s)
- Emilia Tomaszewska
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Katarzyna Ranoszek-Soliwoda
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Katarzyna Bednarczyk
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Agnieszka Lech
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Martyna Janicka
- Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Kozielska 4 St., 01-063 Warsaw, Poland
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Marcin Chodkowski
- Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Kozielska 4 St., 01-063 Warsaw, Poland
| | - Maciej Psarski
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Grzegorz Celichowski
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Malgorzata Krzyzowska
- Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Kozielska 4 St., 01-063 Warsaw, Poland
| | - Jarosław Grobelny
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| |
Collapse
|
18
|
Lishchynskyi O, Shymborska Y, Stetsyshyn Y, Raczkowska J, Skirtach AG, Peretiatko T, Budkowski A. Passive antifouling and active self-disinfecting antiviral surfaces. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2022; 446:137048. [PMID: 35601363 PMCID: PMC9113772 DOI: 10.1016/j.cej.2022.137048] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/01/2022] [Accepted: 05/15/2022] [Indexed: 05/15/2023]
Abstract
Viruses pose a serious threat to human health and society in general, as virus infections are one of the main causes of morbidity and mortality. Till May 2022, over 513 million people around the world have been confirmed to be infected and more than 6.2 million have died due to SARS-CoV-2. Although the COVID-19 pandemic will be defeated in the near future, we are likely to face new viral threats in the coming years. One of the important instruments to protect from viruses are antiviral surfaces, which are essentially capable of limiting their spread. The formulation of the concept of antiviral surfaces is relatively new. In general, five types of mechanism directed against virus spread can be proposed for antiviral surfaces; involving: direct and indirect actions, receptor inactivation, photothermal effect, and antifouling behavior. All antiviral surfaces can be classified into two main types - passive and active. Passive antiviral surfaces are based on superhydrophobic coatings that are able to repel virus contaminated droplets. In turn, viruses can become biologically inert (e.g., blocked or destroyed) upon contact with active antiviral surfaces, as they contain antiviral agents: metal atoms, synthetic or natural polymers, and small molecules. The functionality of antiviral surfaces can be significantly improved with additional properties, such as temperature- or pH-responsivity, multifunctionality, non-specific action on different virus types, long-term application, high antiviral efficiency and self-cleaning.
Collapse
Affiliation(s)
- Ostap Lishchynskyi
- Lviv Polytechnic National University, St. George's Square 2, 79013 Lviv, Ukraine
- Department of Biotechnology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Yana Shymborska
- Lviv Polytechnic National University, St. George's Square 2, 79013 Lviv, Ukraine
- Smoluchowski Institute of Physics, Jagiellonian University, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Yurij Stetsyshyn
- Lviv Polytechnic National University, St. George's Square 2, 79013 Lviv, Ukraine
- Smoluchowski Institute of Physics, Jagiellonian University, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Joanna Raczkowska
- Smoluchowski Institute of Physics, Jagiellonian University, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Andre G Skirtach
- Department of Biotechnology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Taras Peretiatko
- Ivan Franko National University of Lviv, Universytetska 1, 79000 Lviv, Ukraine
| | - Andrzej Budkowski
- Smoluchowski Institute of Physics, Jagiellonian University, Łojasiewicza 11, 30-348, Kraków, Poland
| |
Collapse
|
19
|
Semwal P, Painuli S, Jamloki A, Rauf A, Rahman MM, Olatunde A, Hemeg HA, Abu-Izneid T, Naz S, Punia Bangar S, Lorenzo JM, Simal-Gandara J. Himalayan Wild Fruits as a Strong Source of Nutraceuticals, Therapeutics, Food and Nutrition Security. FOOD REVIEWS INTERNATIONAL 2022. [DOI: 10.1080/87559129.2022.2121407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Prabhakar Semwal
- Department of Life Sciences, Graphic Era Deemed to be University, Dehradun, India
| | - Sakshi Painuli
- Uttarakhand Council for Biotechnology, Premnagar Dehradun, India
| | - Abhishek Jamloki
- High Altitude Plant Physiology Research Centre (HAPPRC), H.N.B. Garhwal University, Srinagar, India
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Khyber, Pakhtunkhwa, Pakistan
| | - Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Ahmed Olatunde
- Department of Medical Biochemistry, Abubakar Tafawa Balewa University, Bauchi, Nigeria
| | - Hassan A. Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, Al-Medinah Al-Monawara, Saudi Arabia
| | - Tareq Abu-Izneid
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, Al Ain Campus, Abu Dhabi, United Arab Emirates
| | - Saima Naz
- Department of Biotechnology, Bacha Khan University Charsadda, Khyber, Pakhtunkhwa, Pakistan
| | - Sneh Punia Bangar
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, South Carolina, USA
| | - Jose M. Lorenzo
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, South Carolina, USA
- Centro Tecnológico de la Carne de Galicia, Ourense, Spain
- Área de Tecnología de los Alimentos, Facultad de Ciencias de Ourense, Universidade de Vigo, Ourense, Spain
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Analytical Chemistry and Food Science Department, Faculty of Science, Universidade de Vigo, Ourense, Spain
| |
Collapse
|
20
|
The Activity of Chelidonium majus L. Latex and Its Components on HPV Reveal Insights into the Antiviral Molecular Mechanism. Int J Mol Sci 2022; 23:ijms23169241. [PMID: 36012505 PMCID: PMC9409487 DOI: 10.3390/ijms23169241] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Yellow-orange latex of Chelidonium majus L. has been used in folk medicine as a therapeutic agent against warts and other visible symptoms of human papillomavirus (HPV) infections for centuries. The observed antiviral and antitumor properties of C. majus latex are often attributed to alkaloids contained therein, but recent studies indicate that latex proteins may also play an important role in its pharmacological activities. Therefore, the aim of the study was to investigate the effect of the crude C. majus latex and its protein and alkaloid-rich fractions on different stages of the HPV replication cycle. The results showed that the latex components, such as alkaloids and proteins, decrease HPV infectivity and inhibit the expression of viral oncogenes (E6, E7) on mRNA and protein levels. However, the crude latex and its fractions do not affect the stability of structural proteins in HPV pseudovirions and they do not inhibit the virus from attaching to the cell surface. In addition, the protein fraction causes increased TNFα secretion, which may indicate the induction of an inflammatory response. These findings indicate that the antiviral properties of C. majus latex arise both from alkaloids and proteins contained therein, acting on different stages of the viral replication cycle.
Collapse
|
21
|
Studies on the antiviral activity of chebulinic acid against dengue and chikungunya viruses and in silico investigation of its mechanism of inhibition. Sci Rep 2022; 12:10397. [PMID: 35729191 PMCID: PMC9213501 DOI: 10.1038/s41598-022-13923-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/30/2022] [Indexed: 11/08/2022] Open
Abstract
Chebulinic acid (CA), originally isolated from the flower extract of the plant Terminalia chebula, has been shown to inhibit infection of herpes simplex virus-2 (HSV-2), suggestively by inhibiting the host entry step of viral infection. Like HSV-2, the dengue virus (DENV) and chikungunya virus (CHIKV) also use receptor glycosaminoglycans (GAG) to gain host entry, therefore, the activity of CA was tested against these viruses. Co-treatment of 8 µM CA with DENV-2 caused 2 log decrease in the virus titer (4.0 log10FFU/mL) at 120 h post infection, compared to virus control (5.95 log10FFU/mL). In contrast, no inhibitory effect of CA was observed against CHIKV infection under any condition. The mechanism of action of CA was investigated in silico by employing DENV-2 and CHIKV envelope glycoproteins. During docking, CA demonstrated equivalent binding at multiple sites on DENV-2 envelope protein, including GAG binding site, which have previously been reported to play a crucial role in host attachment and fusion, indicating blocking of these sites. However, CA did not show binding to the GAG binding site on envelope protein-2 of CHIKV. The in vitro and in silico findings suggest that CA possesses the ability to inhibit DENV-2 infection at the entry stage of its infection cycle and may be developed as a potential therapeutic agent against it.
Collapse
|
22
|
Tale of Viruses in Male Infertility. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1358:275-323. [PMID: 35641875 DOI: 10.1007/978-3-030-89340-8_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Male infertility is a condition where the males either become sterile or critically infertile. The World Health Organisation assessed that approximately 9% of the couple have fertility issues where the contribution of the male partner was estimated to be 50%. There are several factors that can amalgamate to give rise to male infertility. Among them are lifestyle factors, genetic factors and as well as several environmental factors. The causes of male infertility may be acquired, congenital or sometimes idiopathic. All these factors adversely affect the spermatogenesis process as well as they impart serious threats to male genital organs thus resulting in infertility. Viruses are submicroscopic pathogenic agents that rely on host for their replication and survival. They enter the host cell, hijack the host cell machinery to aid their own replication and exit the cell for a new round of infection. With the growing abundance of different types of viruses and the havoc they have stirred in the form of pandemics, it is very essential to decipher their route of entry inside the human body and understand their diverse functional roles in order to combat them. In this chapter, we will review how viruses invade the male genital system thus in turn leading to detrimental consequence on male fertility. We will discuss the tropism of various viruses in the male genital organs and explore their sexual transmissibility. This chapter will summarise the functional and mechanistic approaches employed by the viruses in inducing oxidative stress inside spermatozoa thus leading to male infertility. Moreover, we will also highlight the various antiviral therapies that have been studied so far in order to ameliorate viral infection in order to combat the harmful consequences leading to male infertility.
Collapse
|
23
|
Al-Harrasi A, Behl T, Upadhyay T, Chigurupati S, Bhatt S, Sehgal A, Bhatia S, Singh S, Sharma N, Vijayabalan S, Palanimuthu VR, Das S, Kaur R, Aleya L, Bungau S. Targeting natural products against SARS-CoV-2. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:42404-42432. [PMID: 35362883 PMCID: PMC8972763 DOI: 10.1007/s11356-022-19770-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 03/13/2022] [Indexed: 06/01/2023]
Abstract
The human coronavirus disease (COVID-19) pandemic is caused by a novel coronavirus; the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Natural products, secondary metabolites show positive leads with antiviral and immunotherapy treatments using genomic studies in silico docking. In addition, it includes the action of a mechanism targeting the SARS-CoV-2. In this literature, we aimed to evaluate the antiviral movement of the NT-VRL-1 unique terpene definition to Human coronavirus (HCoV-229E). The effects of 19 hydrolysable tannins on the SARS-CoV-2 were therefore theoretically reviewed and analyzed utilising the molecular operating surroundings for their C-Like protease 3CLpro catalytic dyad residues Angiotensin converting enzyme-2 (MOE 09). Pedunculagin, tercatan, and castalin were detected as interacting strongly with SARS-receptor Cov-2's binding site and catalytic dyad (Cys145 and His41). SARS-CoV-2 methods of subunit S1 (ACE2) inhibit the interaction of the receiver with the s-protein once a drug molecule is coupled to the s-protein and prevent it from infecting the target cells in alkaloids. Our review strongly demonstrates the evidence that natural compounds and their derivatives can be used against the human coronavirus and serves as an area of research for future perspective.
Collapse
Affiliation(s)
- Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, Birkat Al Mawz, Oman
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Tanuj Upadhyay
- Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh, India
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Shvetank Bhatt
- Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Birkat Al Mawz, Oman
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shantini Vijayabalan
- Faculty of Health and Medical Sciences, School of Pharmacy, Taylor's University, Subang Jaya, Kuala Lumpur, Malaysia
| | - Vasanth Raj Palanimuthu
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu, India
| | - Suprava Das
- Department of Pharmacology, Faculty of Medicine, AIMST University, Semeling, Bedong, Kedah, Malaysia
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
24
|
Majewska A, Mlynarczyk-Bonikowska B. 40 Years after the Registration of Acyclovir: Do We Need New Anti-Herpetic Drugs? Int J Mol Sci 2022; 23:ijms23073431. [PMID: 35408788 PMCID: PMC8998721 DOI: 10.3390/ijms23073431] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 01/17/2023] Open
Abstract
Herpes simplex virus types 1 and 2 HSV1 and 2, namely varicella-zoster VZV and cytomegalovirus CMV, are among the most common pathogens worldwide. They remain in the host body for life. The course of infection with these viruses is often asymptomatic or mild and self-limiting, but in immunocompromised patients, such as solid organ or bone marrow transplant recipients, the course can be very severe or even life-threatening. Unfortunately, in the latter group, the highest percentage of infections with strains resistant to routinely used drugs is observed. On the other hand, frequent recurrences of genital herpes can be a problem even in people with normal immunity. Genital herpes also increases the risk of acquiring sexually transmitted diseases, including HIV infection and, if present in pregnant women, poses a risk to the fetus and newborn. Even more frequently than herpes simplex, congenital infections can be caused by cytomegalovirus. We present the most important anti-herpesviral agents, the mechanisms of resistance to these drugs, and the associated mutations in the viral genome. Special emphasis was placed on newly introduced drugs such as maribavir and brincidofovir. We also briefly discuss the most promising substances in preclinical testing as well as immunotherapy options and vaccines currently in use and under investigation.
Collapse
Affiliation(s)
- Anna Majewska
- Department of Medical Microbiology, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw, Poland;
| | - Beata Mlynarczyk-Bonikowska
- Department of Dermatology, Immunodermatology and Venereology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
- Correspondence: ; Tel.: +48-225021313
| |
Collapse
|
25
|
Ghosh U, Sayef Ahammed K, Mishra S, Bhaumik A. The Emerging Roles of Silver Nanoparticles to Target Viral Life Cycle and Detect Viral Pathogens. Chem Asian J 2022; 17:e202101149. [PMID: 35020270 PMCID: PMC9011828 DOI: 10.1002/asia.202101149] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/05/2022] [Indexed: 11/26/2022]
Abstract
Along the line of recent vaccine advancements, new antiviral therapeutics are compelling to combat viral infection-related public health crises. Several properties of silver nanoparticles (AgNPs) such as low level of cytotoxicity, ease of tunability of the AgNPs in the ultra-small nanoscale size and shape through different convenient bottom-up chemistry approaches, high penetration of the composite with drug formulations into host cells has made AgNPs, a promising candidate for developing antivirals. In this review, we have highlighted the recent advancements in the AgNPs based nano-formulations to target cellular mechanisms of viral propagation, immune modulation of the host, and the ability to synergistically enhance the activity of existing antiviral drugs. On the other hand, we have discussed the recent advancements on AgNPs based detection of viral pathogens from clinical samples using inherent physicochemical properties. This article will provide an overview of our current knowledge on AgNPs based formulations that has promising potential for developing a counteractive strategy against emerging and existing viruses.
Collapse
Affiliation(s)
- Ujjyani Ghosh
- Cancer & Inflammatory Disorder DivisionCSIR-Indian Institute of Chemical BiologyJadavpur, Kolkata700032India
- Present address: The University of UtahSalt Lake CityUT84112USA
| | - Khondakar Sayef Ahammed
- Cancer & Inflammatory Disorder DivisionCSIR-Indian Institute of Chemical BiologyJadavpur, Kolkata700032India
- Present address: The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical SciencesHoustonTX77030USA
| | - Snehasis Mishra
- Cancer & Inflammatory Disorder DivisionCSIR-Indian Institute of Chemical BiologyJadavpur, Kolkata700032India
| | - Asim Bhaumik
- School of Materials SciencesIndian Association for the Cultivation of ScienceJadavpur, Kolkata700 032India
| |
Collapse
|
26
|
Effect of denture liners surface modification with Equisetum giganteum and Punica granatum on Candida albicans biofilm inhibition. Ther Deliv 2022; 13:157-166. [PMID: 35195016 DOI: 10.4155/tde-2021-0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: This study investigated the effect of denture liners surface modification with Equisetum giganteum (EG) and Punica granatum (PG) on Candida albicans biofilm inhibition supposing its usage as a sustained-release therapeutical delivery system for Candida-associated denture stomatitis. Materials & methods: C. albicans biofilm (SC5314 or ATCC 90028) was formed on soft liners superficially modified by a primer mixed to drugs at minimum inhibitory concentrations (0.100 g for EG and PG or 0.016 g for nystatin per ml of primer). After 24 h, 7 or 14 days, antibiofilm activity was evaluated by colony-forming unit counts. Results: Not all groups were equi-efficient to nystatin after 24 h and 7 days. After 14 days, EG and PG efficacies were not different from nystatin (almost 100% inhibition). Conclusion: The proposed protocol presents a promising option to allopathic drugs for Candida-associated denture stomatitis treatment.
Collapse
|
27
|
Patil VS, Harish DR, Vetrivel U, Roy S, Deshpande SH, Hegde HV. Hepatitis C Virus NS3/4A Inhibition and Host Immunomodulation by Tannins from Terminalia chebula: A Structural Perspective. Molecules 2022; 27:molecules27031076. [PMID: 35164341 PMCID: PMC8839135 DOI: 10.3390/molecules27031076] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/15/2022] [Accepted: 01/22/2022] [Indexed: 01/27/2023] Open
Abstract
Terminalia chebula Retz. forms a key component of traditional folk medicine and is also reported to possess antihepatitis C virus (HCV) and immunomodulatory activities. However, information on the intermolecular interactions of phytochemicals from this plant with HCV and human proteins are yet to be established. Thus, by this current study, we investigated the HCV NS3/4A inhibitory and host immune-modulatory activity of phytocompounds from T. chebula through in silico strategies involving network pharmacology and structural bioinformatics techniques. To start with, the phytochemical dataset of T. chebula was curated from biological databases and the published literature. Further, the target ability of the phytocompounds was predicted using BindingDB for both HCV NS3/4A and other probable host targets involved in the immune system. Further, the identified targets were docked to the phytochemical dataset using AutoDock Vina executed through the POAP pipeline. The resultant docked complexes with significant binding energy were subjected to 50 ns molecular dynamics (MD) simulation in order to infer the stability of complex formation. During network pharmacology analysis, the gene set pathway enrichment of host targets was performed using the STRING and Reactome pathway databases. Further, the biological network among compounds, proteins, and pathways was constructed using Cytoscape 3.6.1. Furthermore, the druglikeness, side effects, and toxicity of the phytocompounds were also predicted using the MolSoft, ADVERpred, and PreADMET methods, respectively. Out of 41 selected compounds, 10 were predicted to target HCV NS3/4A and also to possess druglike and nontoxic properties. Among these 10 molecules, Chebulagic acid and 1,2,3,4,6-Pentagalloyl glucose exhibited potent HCV NS3/4A inhibitory activity, as these scored a lowest binding energy (BE) of −8.6 kcal/mol and −7.7 kcal/mol with 11 and 20 intermolecular interactions with active site residues, respectively. These findings are highly comparable with Asunaprevir (known inhibitor of HCV NS3/4A), which scored a BE of −7.4 kcal/mol with 20 key intermolecular interactions. MD studies also strongly suggest that chebulagic acid and 1,2,3,4,6-Pentagalloyl glucose as promising leads, as these molecules showed stable binding during 50 ns of production run. Further, the gene set enrichment and network analysis of 18 protein targets prioritized 10 compounds and were predicted to potentially modulate the host immune system, hemostasis, cytokine levels, interleukins signaling pathways, and platelet aggregation. On overall analysis, this present study predicts that tannins from T. chebula have a potential HCV NS3/4A inhibitory and host immune-modulatory activity. However, further experimental studies are required to confirm the efficacies.
Collapse
Affiliation(s)
- Vishal S. Patil
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India; (V.S.P.); (U.V.); (S.H.D.); (H.V.H.)
| | - Darasaguppe R. Harish
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India; (V.S.P.); (U.V.); (S.H.D.); (H.V.H.)
- Correspondence: (D.R.H.); (S.R.)
| | - Umashankar Vetrivel
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India; (V.S.P.); (U.V.); (S.H.D.); (H.V.H.)
- ICMR-National Institute for Research in Tuberculosis, Chetpet, Chennai 600031, India
| | - Subarna Roy
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India; (V.S.P.); (U.V.); (S.H.D.); (H.V.H.)
- Correspondence: (D.R.H.); (S.R.)
| | - Sanjay H. Deshpande
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India; (V.S.P.); (U.V.); (S.H.D.); (H.V.H.)
- Regional Centre for Biotechnology, NCR-Biotech Science Cluster, Faridabad 121001, India
| | - Harsha V. Hegde
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi 590010, India; (V.S.P.); (U.V.); (S.H.D.); (H.V.H.)
| |
Collapse
|
28
|
Lin H, Wang Q, Niu Y, Gu L, Hu L, Li C, Zhao G. Antifungal and Anti-inflammatory Effect of Punicalagin on Murine Aspergillus fumigatus Keratitis. Curr Eye Res 2021; 47:517-524. [PMID: 34797193 DOI: 10.1080/02713683.2021.2008982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE This study aimed to investigate the anti-inflammatory effect and antifungal effect of punicalagin in murine fungal keratitis. METHODS We used in vitro and in vivo protocols to assess the anti-inflammatory effect and antifungal effect of punicalagin. In vitro, time kill and mycelial stain were done. In vivo, murine fungal keratitis was established and treated with PBS or PUN. Clinical scores were taken on days 1, 3, and 5 post infection. The mRNA and protein levels of inflammatory factors were detected by RT-PCR and Western blot, and the number and location of macrophages were analyzed by flow cytometry and immunofluorescence. Also, fungal plate counting was used to assess the antifungal effect. The DCFH-DA fluorescence probe detected the ROS level. RESULTS In vitro, PUN showed activity against A.fumigatus. (A.F.), with MIC90 values of 250 μg/ml, and significantly reduced A.F. biofilm formation (p < .001). In vivo, the mouse fungal keratitis model after punicalagin treatment exhibited less disease, lower clinical scores (p < .05), lower reduced macrophage infiltrate (p < .001), and fungal load (p < .001) than those treated with PBS. Treatment with punicalagin also reduced the mRNA expression and protein level of pro-inflammatory factors. At the cellular level, PUN significantly reduced the mRNA expression of inflammatory factors and ROS production caused by the stimulation of mycelia in RAW264.7 (p < .001). CONCLUSIONS The results show that punicalagin is beneficial in the treatment of murine fungal keratitis. The mechanism of its anti-inflammatory effect was synthetical, including antifungal activity, an inhibitory effect of proinflammatory factor and macrophages, and anti-oxidation.
Collapse
Affiliation(s)
- Hao Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yawen Niu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liting Hu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Drug-Screening Strategies for Inhibition of Virus-Induced Neuronal Cell Death. Viruses 2021; 13:v13112317. [PMID: 34835123 PMCID: PMC8619239 DOI: 10.3390/v13112317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023] Open
Abstract
A number of viruses, including Herpes Simplex Virus (HSV), West Nile Virus (WNV), La Crosse Virus (LACV), Zika virus (ZIKV) and Tick-borne encephalitis virus (TBEV), have the ability to gain access to the central nervous system (CNS) and cause severe neurological disease or death. Although encephalitis cases caused by these viruses are generally rare, there are relatively few treatment options available for patients with viral encephalitis other than palliative care. Many of these viruses directly infect neurons and can cause neuronal death. Thus, there is the need for the identification of useful therapeutic compounds that can inhibit virus replication in neurons or inhibit virus-induced neuronal cell death. In this paper, we describe the methodology to test compounds for their ability to inhibit virus-induced neuronal cell death. These protocols include the isolation and culturing of primary neurons; the culturing of neuroblastoma and neuronal stem cell lines; infection of these cells with viruses; treatment of these cells with selected drugs; measuring virus-induced cell death using MTT or XTT reagents; analysis of virus production from these cells; as well as the basic understanding in mode of action. We further show direct evidence of the effectiveness of these protocols by utilizing them to test the effectiveness of the polyphenol drug, Rottlerin, at inhibiting Zika virus infection and death of neuronal cell lines.
Collapse
|
30
|
Ojha D, Winkler CW, Leung JM, Woods TA, Chen CZ, Nair V, Taylor K, Yeh CD, Tawa GJ, Larson CL, Zheng W, Haigh CL, Peterson KE. Rottlerin inhibits La Crosse virus-induced encephalitis in mice and blocks release of replicating virus from the Golgi body in neurons. Nat Microbiol 2021; 6:1398-1409. [PMID: 34675384 DOI: 10.1038/s41564-021-00968-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 08/25/2021] [Indexed: 11/09/2022]
Abstract
La Crosse virus (LACV) is a mosquito-borne orthobunyavirus that causes approximately 60 to 80 hospitalized pediatric encephalitis cases in the United States yearly. The primary treatment for most viral encephalitis, including LACV, is palliative care, and specific antiviral therapeutics are needed. We screened the National Center for Advancing Translational Sciences library of 3,833 FDA-approved and bioactive small molecules for the ability to inhibit LACV-induced death in SH-SY5Y neuronal cells. The top three hits from the initial screen were validated by examining their ability to inhibit virus-induced cell death in multiple neuronal cell lines. Rottlerin consistently reduced LACV-induced death by 50% in multiple human and mouse neuronal cell lines with an effective concentration of 0.16-0.69 µg ml-1 depending on cell line. Rottlerin was effective up to 12 hours post-infection in vitro and inhibited virus particle trafficking from the Golgi apparatus to trans-Golgi vesicles. In human inducible pluripotent stem cell-derived cerebral organoids, rottlerin reduced virus production by one log and cell death by 35% compared with dimethyl sulfoxide-treated controls. Administration of rottlerin in mice by intraperitoneal or intracranial routes starting at 3 days post-infection decreased disease development by 30-50%. Furthermore, rottlerin also inhibited virus replication of other pathogenic California serogroup orthobunyaviruses (Jamestown Canyon and Tahyna virus) in neuronal cell lines.
Collapse
Affiliation(s)
- Durbadal Ojha
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Clayton W Winkler
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jacqueline M Leung
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Tyson A Woods
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Catherine Z Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Vinod Nair
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Katherine Taylor
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Charles D Yeh
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Gregory J Tawa
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Charles L Larson
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Cathryn L Haigh
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Karin E Peterson
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
31
|
Singh R, Goel S, Bourgeade P, Aleya L, Tewari D. Ayurveda Rasayana as antivirals and immunomodulators: potential applications in COVID-19. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:55925-55951. [PMID: 34491498 PMCID: PMC8422837 DOI: 10.1007/s11356-021-16280-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/27/2021] [Indexed: 05/08/2023]
Abstract
Coronavirus disease (COVID-19) has been declared as a pandemic by the World Health Organization with rapid spread across 216 countries. COVID-19 pandemic has left its imprints on various health systems globally and caused immense social and economic disruptions. The scientific community across the globe is in a quest for digging the effective treatment for COVID-19 and exploring potential leads from traditional systems of healthcare across the world too. Ayurveda (Indian traditional system of medicine) has a comprehensive aspect of immunity through Rasayana which is a rejuvenation therapy. Here we attempt to generate the potential leads based on the classical text from Ayurveda in general and Rasayana in particular to develop effective antiviral and/or immunomodulator for potential or adjunct therapy in SARS-CoV-2. The Rasayana acts not only by resisting body to restrain or withstand the strength, severity or progression of a disease but also by promoting power of the body to prevent the manifestation of a disease. These Rasayana herbs are common in practice as immunomodulator, antiviral and protectives. The studies on Rasayana can provide an insight into the future course of research for the plausible development of effective management of COVID-19 by the utilization and development of various traditional systems of healthcare. Keeping in view the current pandemic situation, there is an urgent need of developing potential medicines. This study proposes certain prominent medicinal plants which may be further studied for drug development process and also in clinical setup under repurposing of these herbs.
Collapse
Affiliation(s)
- Rajeshwari Singh
- Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Janakpuri, New Delhi, 110058, India
| | - Sumeet Goel
- Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Government of India, Janakpuri, New Delhi, 110058, India
| | - Pascale Bourgeade
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030, Besançon Cedex, France
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030, Besançon Cedex, France.
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| |
Collapse
|
32
|
The Methanolic Extract of Perilla frutescens Robustly Restricts Ebola Virus Glycoprotein-Mediated Entry. Viruses 2021; 13:v13091793. [PMID: 34578374 PMCID: PMC8473196 DOI: 10.3390/v13091793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/25/2021] [Accepted: 09/04/2021] [Indexed: 11/17/2022] Open
Abstract
Ebola virus (EBOV), one of the most infectious human viruses and a leading cause of viral hemorrhagic fever, imposes a potential public health threat with several recent outbreaks. Despite the difficulties associated with working with this pathogen in biosafety level-4 containment, a protective vaccine and antiviral therapeutic were recently approved. However, the high mortality rate of EBOV infection underscores the necessity to continuously identify novel antiviral strategies to help expand the scope of prophylaxis/therapeutic management against future outbreaks. This includes identifying antiviral agents that target EBOV entry, which could improve the management of EBOV infection. Herein, using EBOV glycoprotein (GP)-pseudotyped particles, we screened a panel of natural medicinal extracts, and identified the methanolic extract of Perilla frutescens (PFME) as a robust inhibitor of EBOV entry. We show that PFME dose-dependently impeded EBOV GP-mediated infection at non-cytotoxic concentrations, and exerted the most significant antiviral activity when both the extract and the pseudoparticles are concurrently present on the host cells. Specifically, we demonstrate that PFME could block viral attachment and neutralize the cell-free viral particles. Our results, therefore, identified PFME as a potent inhibitor of EBOV entry, which merits further evaluation for development as a therapeutic strategy against EBOV infection.
Collapse
|
33
|
Salles TS, Meneses MDF, Caldas LA, Sá-Guimarães TE, de Oliveira DM, Ventura JA, Azevedo RC, Kuster RM, Soares MR, Ferreira DF. Virucidal and antiviral activities of pomegranate (Punica granatum) extract against the mosquito-borne Mayaro virus. Parasit Vectors 2021; 14:443. [PMID: 34479605 PMCID: PMC8414858 DOI: 10.1186/s13071-021-04955-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The arthropod-borne Mayaro virus (MAYV) causes "Mayaro fever," a disease of medical significance, primarily affecting individuals in permanent contact with forested areas in tropical South America. Recently, MAYV has attracted attention due to its likely urbanization. There are currently no licensed drugs against most mosquito-transmitted viruses. Punica granatum (pomegranate) fruits cultivated in Brazil have been subjected to phytochemical investigation for the identification and isolation of antiviral compounds. In the present study, we explored the antiviral activity of pomegranate extracts in Vero cells infected with Mayaro virus. METHODS The ethanol extract and punicalagin of pomegranate were extracted solely from the shell and purified by chromatographic fractionation, and were chemically identified using spectroscopic techniques. The cytotoxicity of the purified compounds was measured by the dye uptake assay, while their antiviral activity was evaluated by a virus yield inhibition assay. RESULTS Pomegranate ethanol extract (CC50 = 588.9, IC50 = 12.3) and a fraction containing punicalagin as major compound (CC50 = 441.5, IC50 = 28.2) were shown to have antiviral activity (SI 49 and 16, respectively) against Mayaro virus, an alphavirus. Immunofluorescence analysis showed the virucidal effect of pomegranate extract, and transmission electron microscopy (TEM) revealed damage in viral particles treated with this extract. CONCLUSIONS The P. granatum extract is a promising source of antiviral compounds against the alphavirus MAYV and represents an excellent candidate for future studies with other enveloped RNA viruses.
Collapse
Affiliation(s)
- Tiago Souza Salles
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. .,Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | - Lucio Ayres Caldas
- National Institute of Science and Technology for Structural Biology and Bioimaging, INBEB, Rio de Janeiro, RJ, Brazil.,Laboratory of Cellular Ultrastructure Hertha Meyer, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Danielle M de Oliveira
- Natural Products Research Institute, IPPN, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - José A Ventura
- Capixaba Institute of Research, Technical Assistance and Rural Extension, Espirito Santo, Vitoria, Brazil
| | - Renata Campos Azevedo
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo M Kuster
- Natural Products Research Institute, IPPN, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Chemistry Department, Federal University of Espírito Santo, Vitoria, Espirito Santo, Brazil
| | - Márcia Regina Soares
- Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Davis Fernandes Ferreira
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Structural Biology and Bioimaging, INBEB, Rio de Janeiro, RJ, Brazil.,Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
34
|
Kuo YT, Liu CH, Wong SH, Pan YC, Lin LT. Small molecules baicalein and cinnamaldehyde are potentiators of measles virus-induced breast cancer oncolysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 89:153611. [PMID: 34144429 DOI: 10.1016/j.phymed.2021.153611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 05/06/2021] [Accepted: 05/21/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Although the breast cancer mortality has slowed down from 2008 to 2017, breast cancer incidence rate continues to rise and thus, new and/or improved treatments are highly needed. Among them, oncolytic virotherapy which has the ability of facilitating the antitumor adaptive immunity, appears as a promising anticancer therapy. Oncolytic measles virus (MV) is particularly suitable for targeting breast cancer due to the upregulation of MV's receptor nectin-4. Nonetheless, with limited clinical success currently, ways of boosting MV-induced breast cancer oncolysis are therefore necessary. Oncolytic virotherapy alone and combined with chemotherapeutic drugs are two strategic areas with intensive development for the search of anticancer drugs. Considering that baicalein (BAI) and cinnamaldehyde (CIN) have demonstrated antitumor properties against multiple cancers including breast cancer, they could be good partners for MV-based oncolytic virotherapy. PURPOSE To assess the in vitro effect of BAI and CIN with MV and assess their combination effects. METHODS We examined the combinatorial cytotoxic effect of oncolytic MV and BAI or CIN on MCF-7 breast cancer cells. Potential anti-MV activities of the phytochemicals were first investigated in vitro to determine the optimal combination model. Synergism of MV and BAI or CIN was then evaluated in vitro by calculating the combination indices. Finally, cell cycle analysis and apoptosis assays were performed to confirm the mechanism of synergism. RESULTS Overall, the viral sensitization combination modality using oncolytic MV to first infect MCF-7 breast cancer cells followed by drug treatment with BAI or CIN was found to produce significantly enhanced tumor killing. Further mechanistic studies showed that the combinations 'MV-BAI' and 'MV-CIN' display synergistic anti-breast cancer effect, mediated by elevated apoptosis. CONCLUSION We demonstrated, for the first time, effective combination of oncolytic MV with BAI or CIN that could be further explored and potentially developed into novel therapeutic strategies targeting nectin-4-marked breast cancer cells.
Collapse
Affiliation(s)
- Yu-Ting Kuo
- Department of Medical Imaging, Chi Mei Medical Center, Tainan 71004, Taiwan.
| | - Ching-Hsuan Liu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | - Shu Hui Wong
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu-Chi Pan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Liang-Tzung Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
35
|
Antiviral Active Compounds Derived from Natural Sources against Herpes Simplex Viruses. Viruses 2021; 13:v13071386. [PMID: 34372592 PMCID: PMC8310208 DOI: 10.3390/v13071386] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Herpes simplex viruses (HSV) are ubiquitously distributed with a seroprevalence ranging up to 95% in the adult population. Refractory viral infections with herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) represent a major global health issue. In particular, the increasing occurrence of resistance to conventional antiviral drugs make the therapy of such infections even more challenging. For instance, the frequent and long-term use of acyclovir and other nucleoside analogues targeting the viral DNA-polymerase enhance the development of resistant viruses. Particularly, the incidental increase of those strains in immunocompromised patients is alarming and represent a major health concern. Alternative treatment concepts are clearly needed. Natural products such as herbal medicines showed antiherpetic activity in vitro and in vivo and proved to be an excellent source for the discovery and isolation of novel antivirals. By this means, numerous plant-derived compounds with antiviral or antimicrobial activity could be isolated. Natural medicines and their ingredients are well-tolerated and could be a good alternative for treating herpes simplex virus infections. This review provides an overview of the recent status of natural sources such as plants, bacteria, fungi, and their ingredients with antiviral activity against herpes simplex viruses. Furthermore, we highlight the most potent herbal medicines and ingredients as promising candidates for clinical investigation and give an overview about the most important drug classes along with their potential antiviral mechanisms. The content of this review is based on articles that were published between 1996 and 2021.
Collapse
|
36
|
Evidence-based traditional Siddha formulations for prophylaxis and management of respiratory symptoms in COVID-19 pandemic-a review. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2021; 35:102056. [PMID: 34122672 PMCID: PMC8180453 DOI: 10.1016/j.bcab.2021.102056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/21/2022]
Abstract
The recent outbreak of COVID-19 is attributed to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This viral disease is rapidly spreading across the globe, including India. The mainstay in managing the disease is supportive care, nutrition, and preventing further progression in the absence of proven antiviral drugs. Currently two vaccines Covishield and Covaxin are administered in India. Long-term plans of developing most reliable mRNA-based vaccines are also underway for the future method of prophylaxis. The Siddha system of medicine's holistic approach emphasizes lifestyle modification, prophylactic interventions, and dietary management to boost the host immunity and treatment with herbal medicines and higher-order medicines as the case may be. In this review, a brief outline of the disease COVID-19, Coronavirus, evidence-based traditional Siddha interventions for respiratory ailments and immune boosters highlighting the relevant published research on individual herbs are dealt, which pave way for further research on drug repurposing for COVID-19. Historical evidence on the prevention and treatment of infections especially antivirals in Siddha classics is studied.
Collapse
|
37
|
Karimi A, Moradi MT, Rabiei M, Alidadi S. In vitro anti-adenoviral activities of ethanol extract, fractions, and main phenolic compounds of pomegranate ( Punica granatum L.) peel. Antivir Chem Chemother 2021; 28:2040206620916571. [PMID: 32306749 PMCID: PMC7169357 DOI: 10.1177/2040206620916571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background Adenovirus causes a number of diseases in human, and can cause serious infection in severely immunosuppressed individuals. Despite the seriousness of adenovirus infection, there is no definitely approved anti-adenoviral therapy. Many studies have shown that compounds derived from medicinal plants have antiviral activity. Therefore, this study evaluated in vitro anti-adenoviral activity of ethanol extract, fractions, and main phenolic compounds of pomegranate peel. Methods The ethanol extract of pomegranate peel was prepared with maceration method and fractionated by consecutive liquid/liquid partition. The cytotoxic and anti-adenovirus activities of the extract, fractions, and main phenolic compounds (ellagic acid, punicalagin and gallic acid) were evaluated on Hep-2 cell line using MTT assay. Inhibitory effect on adsorption and post-adsorption phases of the virus replication cycle was also evaluated. Results Pomegranate peel extract had a desirable effect against adenovirus with IC50 of 5.77 µg/mL and selectivity index of 49.9. Among the fractions and compounds, the n-butanol fraction and gallic acid had the highest anti-adenoviral activity with IC50 of 2.16 µg/mL and 4.67 µM and selectivity indices of 122.5 and 10.5, respectively. The crude extract, n-butanol fraction and gallic acid inhibited the virus replication in post-adsorption phase (p < 0.01). Conclusion Pomegranate peel extract, especially its n-butanol fraction, could serve as a new promising anti-adenovirus agent due to high inhibitory effect against adenovirus replication. The effect of the n-butanol fraction may be related to the synergistic effect or other compounds of this fraction. Further understanding of the bioassay guided isolation of natural compounds of this fraction seems essential.
Collapse
Affiliation(s)
- Ali Karimi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad-Taghi Moradi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Rabiei
- Department of Pathobiology, Infectious Disease and Public Health, School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, Australia
| | - Somayeh Alidadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
| |
Collapse
|
38
|
Mondal J, Das Mahapatra A, Mandal KC, Chattopadhyay D. An extract of Stephania hernandifolia, an ethnomedicinal plant, inhibits herpes simplex virus 1 entry. Arch Virol 2021; 166:2187-2198. [PMID: 34041610 DOI: 10.1007/s00705-021-05093-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/22/2021] [Indexed: 11/25/2022]
Abstract
Stephania hernandifolia (Nimukho), an ethnomedicinal herb from rural Bengal, has been used traditionally for the management of nerve, skin, urinary, and digestive ailments. Here, we attempted to confirm the antiviral potential of aqueous, methanol, and chloroform extracts of S. hernandifolia against herpes simplex virus type 1 (HSV-1), the causative agent of orolabial herpes in humans, and decipher its underlying mechanism of action. The bioactive extract was standardized and characterized by gas chromatography-mass spectroscopy, while cytotoxicity and antiviral activity were evaluated by MTT and plaque reduction assay, respectively. Two HSV strains, HSV-1F and the clinical isolate VU-09, were inhibited by the chloroform extract (CE) with a median effective concentration (EC50) of 4.32 and 4.50 µg/ml respectively, with a selectivity index (SI) of 11. Time-of-addition assays showed that pre-treatment of virus-infected cells with the CE and its removal before infection reduced the number of plaques without lasting toxicity to the cell, indicating that the CE affected the early stage in the viral life cycle. The number of plaques was also reduced by direct inactivation of virions and by the addition of CE for a short time following attachment of virions. These results together suggest that modification of either the virion surface or the cell surface by the CE inhibits virus entry into the host cell.
Collapse
Affiliation(s)
- Joy Mondal
- ICMR-NICED Virus Unit, ID and BG Hospital, GB-4, First Floor, 57 Dr. Suresh C Banerjee Road, Beliaghata, Kolkata, 700010, India
- Department of Microbiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Ananya Das Mahapatra
- ICMR-NICED Virus Unit, ID and BG Hospital, GB-4, First Floor, 57 Dr. Suresh C Banerjee Road, Beliaghata, Kolkata, 700010, India
| | - Keshab C Mandal
- Department of Microbiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Debprasad Chattopadhyay
- ICMR-NICED Virus Unit, ID and BG Hospital, GB-4, First Floor, 57 Dr. Suresh C Banerjee Road, Beliaghata, Kolkata, 700010, India.
- ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi, 590010, India.
| |
Collapse
|
39
|
Tito A, Colantuono A, Pirone L, Pedone E, Intartaglia D, Giamundo G, Conte I, Vitaglione P, Apone F. Pomegranate Peel Extract as an Inhibitor of SARS-CoV-2 Spike Binding to Human ACE2 Receptor ( in vitro): A Promising Source of Novel Antiviral Drugs. Front Chem 2021; 9:638187. [PMID: 33996744 PMCID: PMC8114579 DOI: 10.3389/fchem.2021.638187] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/25/2021] [Indexed: 01/08/2023] Open
Abstract
Plant extracts are rich in bioactive compounds, such as polyphenols, sesquiterpenes, and triterpenes, which potentially have antiviral activities. As a consequence of the coronavirus disease 2019 pandemic, caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus, thousands of scientists have been working tirelessly trying to understand the biology of this new virus and the disease pathophysiology, with the main goal of discovering effective preventive treatments and therapeutic agents. Plant-derived secondary metabolites may play key roles in preventing and counteracting the rapid spread of SARS-CoV-2 infections by inhibiting the activity of several viral proteins, in particular those involved in the virus entry into the host cells and its replication. Using in vitro approaches, we investigated the role of a pomegranate peel extract (PPE) in attenuating the interaction between the SARS-CoV-2 Spike glycoprotein and the human angiotensin-converting enzyme 2 receptor, and on the activity of the virus 3CL protease. Although further studies will be determinant to assess the efficacy of this extract in vivo, our results opened new promising opportunities to employ natural extracts for the development of effective and innovative therapies in the fight against SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Luciano Pirone
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Emilia Pedone
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | | | - Giuliana Giamundo
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Paola Vitaglione
- Department of Agricultural Science, University of Naples Federico II, Portici, Italy
| | - Fabio Apone
- Arterra Bioscience SPA, Naples, Italy
- Vitalab Srl, Naples, Italy
| |
Collapse
|
40
|
Aamir K, Sugumar V, Khan HU, Looi CY, Juneja R, Waqas M, Arya A. Non-toxic nature of chebulinic acid on biochemical, hematological and histopathological analysis in normal Sprague Dawley rats. Toxicol Res 2021; 38:159-174. [PMID: 35419271 PMCID: PMC8960548 DOI: 10.1007/s43188-021-00092-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/04/2021] [Accepted: 02/04/2021] [Indexed: 01/20/2023] Open
Abstract
Chebulinic acid (CA) is an ellagitannins isolated from the dried fruits of Terminalia chebula with diverse pharmacological activities. The present study focused on the acute toxicity of CA in normal Sprague Dawley (SD) rats. CA was administered via oral gavage to different groups in 300 and 2000 mg/kg body weight and vehicle respectively. All the animals were monitored carefully for any physiological or behavioral changes for 14 days. On day 15th animals were euthanized and blood was collected for hematological and biochemical analysis. Different tissues were collected for histopathological study using four different staining techniques (hematoxylin and eosin, Masson's trichrome, periodic acid Schiff and picro sirius red) to observe any pathological alterations. The results highlighted no morbidity and mortality after oral ingestion of CA (300 and 2000 mg/kg). Food and water consumption, body weight, relative organ weight, hematological and biochemical parameters were normal without any gross pathological lesions in harvested tissues. The outcome of the current study supported safety of CA even at high dose. However, further detailed study is required on experimentally disease model to unfold its therapeutic potential in laboratory animals.
Collapse
|
41
|
Discovery of chebulagic acid and punicalagin as novel allosteric inhibitors of SARS-CoV-2 3CL pro. Antiviral Res 2021; 190:105075. [PMID: 33872675 PMCID: PMC8052511 DOI: 10.1016/j.antiviral.2021.105075] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/16/2022]
Abstract
The emerging SARS-CoV-2 infection is the cause of the global COVID-19 pandemic. To date, there are limited therapeutic options available to fight this disease. Here we examined the inhibitory abilities of two broad-spectrum antiviral natural products chebulagic acid (CHLA) and punicalagin (PUG) against SARS-CoV-2 viral replication. Both CHLA and PUG reduced virus-induced plaque formation in Vero-E6 monolayer at noncytotoxic concentrations, by targeting the enzymatic activity of viral 3-chymotrypsin-like cysteine protease (3CLpro) as allosteric regulators. Our study demonstrates the potential use of CHLA and PUG as novel COVID-19 therapies.
Collapse
|
42
|
Anand AV, Balamuralikrishnan B, Kaviya M, Bharathi K, Parithathvi A, Arun M, Senthilkumar N, Velayuthaprabhu S, Saradhadevi M, Al-Dhabi NA, Arasu MV, Yatoo MI, Tiwari R, Dhama K. Medicinal Plants, Phytochemicals, and Herbs to Combat Viral Pathogens Including SARS-CoV-2. Molecules 2021; 26:1775. [PMID: 33809963 PMCID: PMC8004635 DOI: 10.3390/molecules26061775] [Citation(s) in RCA: 45] [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: 02/17/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome corona virus-2 (SARS-CoV-2), is the most important health issue, internationally. With no specific and effective antiviral therapy for COVID-19, new or repurposed antiviral are urgently needed. Phytochemicals pose a ray of hope for human health during this pandemic, and a great deal of research is concentrated on it. Phytochemicals have been used as antiviral agents against several viruses since they could inhibit several viruses via different mechanisms of direct inhibition either at the viral entry point or the replication stages and via immunomodulation potentials. Recent evidence also suggests that some plants and its components have shown promising antiviral properties against SARS-CoV-2. This review summarizes certain phytochemical agents along with their mode of actions and potential antiviral activities against important viral pathogens. A special focus has been given on medicinal plants and their extracts as well as herbs which have shown promising results to combat SARS-CoV-2 infection and can be useful in treating patients with COVID-19 as alternatives for treatment under phytotherapy approaches during this devastating pandemic situation.
Collapse
Affiliation(s)
- Arumugam Vijaya Anand
- Medical Genetics and Epigenetics Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, India; (M.K.); (K.B.); (A.P.)
| | | | - Mohandass Kaviya
- Medical Genetics and Epigenetics Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, India; (M.K.); (K.B.); (A.P.)
| | - Kathirvel Bharathi
- Medical Genetics and Epigenetics Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, India; (M.K.); (K.B.); (A.P.)
| | - Aluru Parithathvi
- Medical Genetics and Epigenetics Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, India; (M.K.); (K.B.); (A.P.)
| | - Meyyazhagan Arun
- Department of Life Sciences, CHRIST (Deemed to be University), Bengaluru 560029, India;
| | - Nachiappan Senthilkumar
- Institute of Forest Genetics and Tree Breeding (IFGTB), Forest Campus, Cowley Brown Road, RS Puram, Coimbatore 641002, India;
| | | | | | - Naif Abdullah Al-Dhabi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (N.A.A.-D.); (M.V.A.)
| | - Mariadhas Valan Arasu
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (N.A.A.-D.); (M.V.A.)
- Xavier Research Foundation, St. Xavier’s College, Palayamkottai, Thirunelveli 627002, India
| | - Mohammad Iqbal Yatoo
- Faculty of Veterinary Sciences and Animal Husbandry, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Srinagar 190006, India;
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India;
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India
| |
Collapse
|
43
|
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: 10.0] [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
|
44
|
Elkousy RH, Said ZNA, Abd El-Baseer MA, Abu El Wafa SA. Antiviral activity of castor oil plant (Ricinus communis) leaf extracts. JOURNAL OF ETHNOPHARMACOLOGY 2021; 271:113878. [PMID: 33515683 DOI: 10.1016/j.jep.2021.113878] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ricinus communis L., commonly known as castor oil plant, is a precious traditional medicine with a history of thousands of years in the world. Castor oil plant has high traditional and medicinal values for treating liver infections, stomach ache, flatulence, constipation, inflammation, warts, colic, enteritis, fever, headache, and as a counter irritant. Its diverse phytochemicals have a wide range of valuable medicinal activities including hepatoprotective, anti-nociceptive, antioxidant, antiulcer, anticancer, anti-inflammatory, central analgesic, antidiabetic, antimicrobial, antiviral, and wound healing activity. AIM OF THE WORK To provide a complete characterization of the composition of Ricinus communis leaves using ultra-performance liquid chromatography coupled with hybrid triple time-of-flight mass spectrometry (UPLC-Triple TOF-MS/MS) and different chromatographic techniques and to evaluate its antiviral potential using three mechanisms against three common viruses. MATERIALS AND METHODS R. communis leaves were extracted with 70% methanol and further partitioned with solvents of increasing polarities: petroleum ether, dichloromethane (CH2Cl2), ethyl acetate, and n-butanol. The CH2Cl2 and n-butanol fractions were subjected to repeated chromatographic separation to isolate the phytochemicals, and their structures were elucidated using nuclear magnetic resonance spectroscopy. UPLC-Triple TOF-MS/MS was performed to determine the different phytochemicals in the ethyl acetate fraction. The antiviral activity of the extracts was investigated using the maximum nontoxic concentration of each against the challenge dose of the virus (CDV) and 1/10 and 1/100 dilutions of the CDV for Coxsackie B virus type 4 (COXB4), herpes simplex virus type 1 (HSV1), and hepatitis A virus (HAV) using Vero cell cultures that were treated according to three protocols to test for anti-replicative, protective, and anti-infective antiviral activity. Cell viability was evaluated using the MTT colorimetric assay and each experiment is repeated three times independently of each other. RESULTS R. communis leaves possessed antiviral activity. Evaluation of the anti-replicative activity showed that all extracts possessed high anti-replicative activity against HAV especially methanol and methylene chloride fractions and moderate activity against COXB4; butanol > methylene chloride and ethyl acetate > methanol. All extracts showed protective activity against HAV, especially butanol extract, while methanol extracts showed higher non-significant antiviral protective activity against HSV1 vs Acyclovir. Almost no anti-infective effects were recorded for any extract against the studied viruses. CONCLUSION The discriminatory effect against each virus by different mechanisms suggests the presence of different chemical compounds. The alkaloid and phenolic derivatives of the extracts of R. communis leaves may help develop a drug to prevent or treat common viral infections. Further investigations are recommended to define the bioactive antiviral properties of R. communis leaves.
Collapse
Affiliation(s)
- Rawah H Elkousy
- Department of Pharmacognosy, Faculty of Pharmacy (for Girls), Al-Azhar University, P.O. Box 11651, Nasr City, Cairo, Egypt
| | - Zeinab N A Said
- Department of Microbiology, Faculty of Medicine (for Girls), Al-Azhar University, P.O. Box 11754, Nasr City, Cairo, Egypt
| | - Mohamed A Abd El-Baseer
- Department of Microbiology, Faculty of Science (for Boys), Al-Azhar University, P.O. Box 13129, Nasr City, Cairo, Egypt
| | - Salwa A Abu El Wafa
- Department of Pharmacognosy, Faculty of Pharmacy (for Girls), Al-Azhar University, P.O. Box 11651, Nasr City, Cairo, Egypt.
| |
Collapse
|
45
|
Liu CH, Wong SH, Tai CJ, Tai CJ, Pan YC, Hsu HY, Richardson CD, Lin LT. Ursolic Acid and Its Nanoparticles Are Potentiators of Oncolytic Measles Virotherapy against Breast Cancer Cells. Cancers (Basel) 2021; 13:cancers13010136. [PMID: 33406633 PMCID: PMC7795983 DOI: 10.3390/cancers13010136] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Despite the advancing treatments, female breast cancer is one of the most common cancers and a leading cause of cancer deaths in women. To help broaden the therapeutic spectrum of breast cancer, we identified the natural compound ursolic acid (UA) as a potentiator that enhances the oncolytic activity of measles virus (MV) against breast cancer cells through the induction of apoptosis. In addition, to increase clinical applicability, we further generated UA nanoparticles that achieved improved solubility. UA nanoparticles similarly synergized with MV in killing breast cancer cells by triggering apoptosis, and this synergistic anticancer effect was also observed in various breast cancer cell types. This study demonstrates for the first time that UA and its nanoparticles enhance MV’s oncolytic activity in breast cancer cells, suggesting that such combinations may be worth further exploring as an anticancer strategy against breast cancer. Abstract Oncolytic viruses (OVs) and phytochemical ursolic acid (UA) are two efficacious therapeutic candidates in development against breast cancer, the deadliest women’s cancer worldwide. However, as single agents, OVs and UA have limited clinical efficacies. As a common strategy of enhancing monotherapeutic anticancer efficacy, we explored the combinatorial chemovirotherapeutic approach of combining oncolytic measles virus (MV), which targets the breast tumor marker Nectin-4, and the anticancer UA against breast adenocarcinoma. Our findings revealed that in vitro co-treatment with UA synergistically potentiated the killing of human breast cancer cells by oncolytic MV, without UA interfering the various steps of the viral infection. Mechanistic studies revealed that the synergistic outcome from the combined treatment was mediated through UA’s potentiation of apoptotic killing by MV. To circumvent UA’s poor solubility and bioavailability and strengthen its clinical applicability, we further developed UA nanoparticles (UA-NP) by nanoemulsification. Compared to the non-formulated UA, UA-NP exhibited improved drug dissolution property and similarly synergized with oncolytic MV in inducing apoptotic breast cancer cell death. This oncolytic potentiation was partly attributed to the enhanced autophagic flux induced by the UA-NP and MV combined treatment. Finally, the synergistic effect from the UA-NP and MV combination was also observed in BT-474 and MDA-MB-468 breast cancer cells. Our study thus highlights the potential value of oncolytic MV and UA-based chemovirotherapy for further development as a treatment strategy against breast cancer, and the feasibility of employing nanoformulation to enhance UA’s applicability.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Shu Hui Wong
- International M.Sc. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Chen-Jei Tai
- Department of Traditional Chinese Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Cheng-Jeng Tai
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Chi Pan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien 970, Taiwan;
| | - Christopher D. Richardson
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Department of Pediatrics and Canadian Center for Vaccinology, Izaak Walton Killam Health Centre, Halifax, NS B3K 6R8, Canada
| | - Liang-Tzung Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Correspondence: ; Tel.: +886-2-2736-1661 (ext. 3911)
| |
Collapse
|
46
|
Loaiza-Cano V, Monsalve-Escudero LM, Filho CDSMB, Martinez-Gutierrez M, de Sousa DP. Antiviral Role of Phenolic Compounds against Dengue Virus: A Review. Biomolecules 2020; 11:biom11010011. [PMID: 33374457 PMCID: PMC7823413 DOI: 10.3390/biom11010011] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Phenolic compounds have been related to multiple biological activities, and the antiviral effect of these compounds has been demonstrated in several viral models of public health concern. In this review, we show the antiviral role of phenolic compounds against dengue virus (DENV), the most widespread arbovirus globally that, after its re-emergence, has caused multiple epidemic outbreaks, especially in the last two years. Twenty phenolic compounds with anti-DENV activity are discussed, including the multiple mechanisms of action, such as those directed against viral particles or viral proteins, host proteins or pathways related to the productive replication viral cycle and the spread of the infection.
Collapse
Affiliation(s)
- Vanessa Loaiza-Cano
- Grupo de Investigacion en Ciencias Animales-GRICA, Universidad Cooperativa de Colombia, 680003 Bucaramanga, Colombia; (V.L.-C.); (L.M.M.-E.)
| | - Laura Milena Monsalve-Escudero
- Grupo de Investigacion en Ciencias Animales-GRICA, Universidad Cooperativa de Colombia, 680003 Bucaramanga, Colombia; (V.L.-C.); (L.M.M.-E.)
| | | | - Marlen Martinez-Gutierrez
- Grupo de Investigacion en Ciencias Animales-GRICA, Universidad Cooperativa de Colombia, 680003 Bucaramanga, Colombia; (V.L.-C.); (L.M.M.-E.)
- Correspondence: (M.M.-G.); (D.P.d.S.); Tel.: +57-310-543-8583 (M.M.-G.); +55-833-216-7347 (D.P.d.S.)
| | - Damião Pergentino de Sousa
- Department of Pharmaceutical Sciences, Federal University of Paraíba, CEP 58051-970 João Pessoa, PB, Brazil;
- Correspondence: (M.M.-G.); (D.P.d.S.); Tel.: +57-310-543-8583 (M.M.-G.); +55-833-216-7347 (D.P.d.S.)
| |
Collapse
|
47
|
Luo Z, Kuang XP, Zhou QQ, Yan CY, Li W, Gong HB, Kurihara H, Li WX, Li YF, He RR. Inhibitory effects of baicalein against herpes simplex virus type 1. Acta Pharm Sin B 2020; 10:2323-2338. [PMID: 33354504 PMCID: PMC7745058 DOI: 10.1016/j.apsb.2020.06.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/10/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a ubiquitous and widespread human pathogen, which gives rise to a range of diseases, including cold sores, corneal blindness, and encephalitis. Currently, the use of nucleoside analogs, such as acyclovir and penciclovir, in treating HSV-1 infection often presents limitation due to their side effects and low efficacy for drug-resistance strains. Therefore, new anti-herpetic drugs and strategies should be urgently developed. Here, we reported that baicalein, a naturally derived compound widely used in Asian countries, strongly inhibited HSV-1 replication in several models. Baicalein was effective against the replication of both HSV-1/F and HSV-1/Blue (an acyclovir-resistant strain) in vitro. In the ocular inoculation mice model, baicalein markedly reduced in vivo HSV-1/F replication, receded inflammatory storm and attenuated histological changes in the cornea. Consistently, baicalein was found to reduce the mortality of mice, viral loads both in nose and trigeminal ganglia in HSV-1 intranasal infection model. Moreover, an ex vivo HSV-1-EGFP infection model established in isolated murine epidermal sheets confirmed that baicalein suppressed HSV-1 replication. Further investigations unraveled that dual mechanisms, inactivating viral particles and inhibiting IκB kinase beta (IKK-β) phosphorylation, were involved in the anti-HSV-1 effect of baicalein. Collectively, our findings identified baicalein as a promising therapy candidate against the infection of HSV-1, especially acyclovir-resistant strain. Baicalein is highly effective against HSV-1infection ex vivo and in vivo. Inactivation of viral particles and suppression of NF-κB activation were involved in the anti-viral effect of baicalein. Hence, our work offers experimental basis for baicalein as a potential drug in treating HSV-1 associated diseases.
Collapse
Key Words
- Anti-HSV-1
- Baicalein
- CC50, 50% cytotoxic concentration
- DCFH-DA, 2′,7′-dichlorofluorescin diacetate
- EC50, 50% effective concentration
- GB, glycoprotein B
- HSV-1 infection
- HSV-1, herpes simplex virus types 1
- ICP, infected cell polypeptide
- IKK-β phosphorylation
- IKK-β, IκB kinase beta
- IL-1β, interleukin 1 beta
- IL-6, interleukin 6
- IκB-α, inhibitor of NF-κB alpha
- LPS, lipopolysaccharides
- MOI, multiplicity of infection
- NAC, N-acetyl-l-cysteine
- NF-κB activation
- NF-κB, nuclear factor kappa-B
- PFU, plaque-forming units
- PGA1, prostaglandin A1
- ROS, reactive oxygen species
- SI, selectivity index
- TG, trigeminal ganglia
- TNF-α, tumor necrosis factor alpha
- Viral inactivation
- dpi, days post-infection
- p-IKK-β, phosphorylated-IKK beta
- p-IκB-α, phosphorylated-IκB alpha
Collapse
|
48
|
Kuo YT, Liu CH, Li JW, Lin CJ, Jassey A, Wu HN, Perng GC, Yen MH, Lin LT. Identification of the phytobioactive Polygonum cuspidatum as an antiviral source for restricting dengue virus entry. Sci Rep 2020; 10:16378. [PMID: 33009425 PMCID: PMC7532532 DOI: 10.1038/s41598-020-71849-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/18/2020] [Indexed: 11/09/2022] Open
Abstract
Dengue virus (DENV) is a mosquito-borne pathogen that is becoming a serious global threat, owing to its rising incidence in inter-tropical regions that yield over 50 million annual infections. There are currently no approved antiviral agents for the management of dengue, and recent shortcomings in its immunization called for immediate action to develop effective drugs with prophylactic ability to better manage its infection. In an attempt to discover novel antiviral sources, we identified the medicinal herb Polygonum cuspidatum (PC) as a bioactive botanical material against DENV infectivity. Specifically, the methanolic extract from PC rhizomes (PCME) potently inhibited DENV infection without causing significant cytotoxicity. Further examination on the viral life cycle demonstrated that PCME particularly targeted the initial stages of DENV infection, while pre- and post-infection treatments had no effect. More importantly, the PCME could efficiently inactivate DENV free virus particles and block the viral attachment and entry/fusion events without apparently influencing viral replication, egress, and cell-to-cell spread. The antiviral effect of PCME was also recapitulated in infection analysis using DENV pseudoparticles displaying viral structural proteins that mediate DENV particle entry. Besides, PCME treatment also inhibited direct DENV entry into several cell types relevant to its infection and reduced viral infectivity of other members of the Flaviviridae family, including the hepatitis C virus (HCV) and Zika virus (ZIKV). Due to its potency against DENV entry, we suggest that the phytobioactive extract from PC is an excellent starting point as an antiviral source material for further development of therapeutic strategies in the prophylactic management of DENV infection.
Collapse
Affiliation(s)
- Yu-Ting Kuo
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan
| | - Ching-Hsuan Liu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jin-Wei Li
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Ju Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Alagie Jassey
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Huey-Nan Wu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Guey Chuen Perng
- Department of Microbiology and Immunology & Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Diseases and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Hong Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Liang-Tzung Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
49
|
Khalifa I, Zhu W, Mohammed HHH, Dutta K, Li C. Tannins inhibit SARS-CoV-2 through binding with catalytic dyad residues of 3CL pro: An in silico approach with 19 structural different hydrolysable tannins. J Food Biochem 2020; 44:e13432. [PMID: 32783247 PMCID: PMC7435556 DOI: 10.1111/jfbc.13432] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 01/09/2023]
Abstract
Coronavirus epidemic 2019 (COVID-19), instigated by SARS-CoV-2 virus, is recently raising worldwide and inspiring global health worries. The main 3-chymotrypsin-like cysteine protease (3CLPro) enzyme of SARS-CoV-2, which operates its replication, could be used as a medication discovery point. We therefore theoretically studied and docked the effects of 19 hydrolysable tannins on SARS-CoV-2 by assembling with the catalytic dyad residues of its 3CLpro using molecular operating environment (MOE 09). Results discovered that pedunculagin, tercatain, and castalin intensely interacted with the receptor binding site and catalytic dyad (Cys145 and His41) of SARS-CoV-2. Our analyses estimated that the top three hits might serve as potential inhibitor of SARS-CoV-2 leading molecules for additional optimization and drug development process to combat COVID-19. This study unleashed that tannins with specific structure could be utilized as natural inhibitors against COVID-19. PRACTICAL APPLICATIONS: The 3CLPro controls SARS-CoV-2 copying and manages its life series, which was targeted in case of SARS-CoV and MERS-CoV coronavirus. About 19 hydrolysable tannins were computed against 3CLpro of SARS-CoV-2. Pedunculagin, tercatain, and castalin interacted with Cys145 and His41 of SARS-CoV-2-3CLpro. Pedunculagin-SARS-CoV-2-3CLpro remain stable, with no obvious fluctuations. We predicted that the understandings gained in the current research may evidence valued for discovering and unindustrialized innovative natural inhibitors for COVID-19 in the nearby future.
Collapse
Affiliation(s)
- Ibrahim Khalifa
- Food Technology Department, Faculty of AgricultureBenha UniversityBenhaEgypt
| | - Wei Zhu
- College of Food Science and Technology, Key Laboratory of Environment Correlative Food Science, Ministry of EducationHuazhong Agricultural UniversityWuhanChina
| | - Hammad Hamed Hammad Mohammed
- College of Food Science and Technology, Key Laboratory of Environment Correlative Food Science, Ministry of EducationHuazhong Agricultural UniversityWuhanChina
- Ministry of Agriculture and ForestryNational Food Research CentreKhartoum NorthSudan
| | - Kunal Dutta
- Microbiology and Immunology Laboratory, Department of Human Physiology with Community HealthVidyasagar UniversityMidnaporeIndia
| | - Chunmei Li
- College of Food Science and Technology, Key Laboratory of Environment Correlative Food Science, Ministry of EducationHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
50
|
Li P, Du R, Chen Z, Wang Y, Zhan P, Liu X, Kang D, Chen Z, Zhao X, Wang L, Rong L, Cui Q. Punicalagin is a neuraminidase inhibitor of influenza viruses. J Med Virol 2020; 93:3465-3472. [PMID: 32827314 DOI: 10.1002/jmv.26449] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/17/2020] [Indexed: 02/03/2023]
Abstract
Influenza A virus (IAV) causes great morbidity and mortality worldwide every year. However, there are only a limited number of drugs clinically available against IAV infection. Further, emergence of drug-resistant strains can render those drugs ineffective. Thus there is an unmet medical need to develop new anti-influenza agents. In this study, we show that punicalagin from plants possesses strong anti-influenza activity with a low micromolar IC50 value in tissue culture. Using a battery of bioassays such as single-cycle replication assay, neuraminidase (NA) inhibition assay, and virus yield reduction assay, we demonstrate that the primary mechanism of action (MOA) of punicalagin is the NA-mediated viral release. Moreover, punicalagin can inhibit replication of different strains of influenza A and B viruses, including oseltamivir-resistant virus (NA/H274Y), indicating that punicalagin is a broad spectrum antiviral against both IAV and IBV. Further, although punicalagin targets NA like oseltamivir, it has a different MOA. These results suggest that punicalagin is an influenza NA inhibitor that may be further developed as a novel antiviral against influenza viruses.
Collapse
Affiliation(s)
- Ping Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruikun Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China.,Research Center, College of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zinuo Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanyan Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhaoyu Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiujuan Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Qinghua Cui
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China.,Research Center, College of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|