1
|
Wanas AS, Radwan MM, Marzouk AA, Elkaeed EB, Alsfouk BA, Mostafa AE, Eissa IH, Metwaly AM, ElSohly MA. Isolation and in silico investigation of cannflavins from Cannabis sativa leaves as potential anti-SARS-CoV-2 agents targeting the Papain-Like Protease. Nat Prod Res 2025; 39:1081-1094. [PMID: 38100380 DOI: 10.1080/14786419.2023.2294111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/28/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
This study aimed to isolate and identify three prenylflavonoids (cannflavin A, B, and C) from Cannabis sativa leaves using different chromatographic techniques. The potential of the isolated compounds against SARS-CoV-2 was suggested through several in silico analysis. Structural similarity studies against nine co-crystallized ligands of SARS-CoV-2's proteins indicated the similarities of the isolated cannflavins with the SARS-CoV-2 Papain-Like Protease (PLP) ligand, Y95. Then, flexible allignment study confirmed this similarity. Docking experiments showed successful binding of all cannflavins within the active pocket of PLP, with energies comparable to Y95. Among them, cannflavin A demonstrated the most similar binding mode, while cannflavin C exhibited the best energy. Molecular dynamics (MD) simulations and MM-GPSA confirmed the accurate binding of cannflavin A to the PLP. In silico ADMET studies indicated favourable drug-like properties for all three compounds, suggesting their potential as anti-SARS-CoV-2 agents. Further In vitro and In vivo investigations are necessary to validate these findings and establish their efficacy and safety profiles.
Collapse
Affiliation(s)
- Amira S Wanas
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Mississippi, USA
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Mohamed M Radwan
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Mississippi, USA
| | - Adel A Marzouk
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Mississippi, USA
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Bshra A Alsfouk
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ahmad E Mostafa
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed M Metwaly
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
- Biopharmaceutical Products Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
| | - Mahmoud A ElSohly
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Mississippi, USA
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University, Mississippi, USA
| |
Collapse
|
2
|
Kim EA, Kang N, Heo JH, Park A, Heo SY, Kim HS, Heo SJ. Antiviral Activity of Ecklonia cava Extracts and Dieckol Against Zika Virus. Int J Mol Sci 2024; 25:13694. [PMID: 39769456 PMCID: PMC11728237 DOI: 10.3390/ijms252413694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
Ecklonia cava and its major compound dieckol, both natural marine products, possess antioxidant, anti-inflammatory, and metabolic-regulating effects. Zika virus (ZIKV), an arbovirus from the Flaviviridae family, is transmitted by mosquitoes and causes serious illnesses in humans. This study aimed to evaluate the anti-ZIKV potential of Ecklonia cava and dieckol. The antiviral activity of Ecklonia cava extract (ECE), prepared with 80% ethanol, was assessed in ZIKV-infected Vero E6 cells through MTT assay, plaque assay, and quantitative polymerase chain reaction (qPCR), demonstrating no cytotoxicity and a significant reduction in viral titers and ZIKV mRNA levels. In addition, ECE decreased the expression of tumor necrosis factor-α and interferon-induced protein with tetratricopeptide repeats in the ZIKV-infected cells. Dieckol, the primary active compound in ECE, exhibited potent anti-ZIKV activity, with a half maximal inhibitory concentration (IC50), value of 4.8 µM. In silico molecular docking analysis revealed that dieckol forms stable complexes with key ZIKV proteins, including the envelope, NS2B/NS3, and RNA-dependent RNA polymerase (RdRp) protein, exhibiting high binding energies of -438.09 kcal/mol, -1040.51 kcal/mol, and -1043.40 kcal/mol, respectively. Overall, our findings suggest that ECE and dieckol are promising candidates for the development of anti-ZIKV agents.
Collapse
Affiliation(s)
- Eun-A Kim
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea; (E.-A.K.); (N.K.); (J.-H.H.); (A.P.); (S.-Y.H.)
| | - Nalae Kang
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea; (E.-A.K.); (N.K.); (J.-H.H.); (A.P.); (S.-Y.H.)
| | - Jun-Ho Heo
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea; (E.-A.K.); (N.K.); (J.-H.H.); (A.P.); (S.-Y.H.)
| | - Areumi Park
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea; (E.-A.K.); (N.K.); (J.-H.H.); (A.P.); (S.-Y.H.)
| | - Seong-Yeong Heo
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea; (E.-A.K.); (N.K.); (J.-H.H.); (A.P.); (S.-Y.H.)
| | - Hyun-Soo Kim
- Department of Seafood Science and Technology, The Institute of Marine Industry, Gyeongsang National University, Tongyeong 53064, Republic of Korea;
| | - Soo-Jin Heo
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea; (E.-A.K.); (N.K.); (J.-H.H.); (A.P.); (S.-Y.H.)
- Department of Marine Biology, University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
3
|
Abdel-Kader MS, Radwan MM, Metwaly AM, Eissa IH, Hazekamp A, ElSohly MA. Chemistry and Biological Activities of Cannflavins of the Cannabis Plant. Cannabis Cannabinoid Res 2023; 8:974-985. [PMID: 37756221 PMCID: PMC10714118 DOI: 10.1089/can.2023.0128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023] Open
Abstract
Background: Throughout history, Cannabis has had a significant influence on human life as one of the earliest plants cultivated by humans. The plant was a source of fibers used by the oldest known civilizations. Cannabis was also used medicinally in China, India, and ancient Egypt. Delta-9-tetrahydrocannabinol (Δ9-THC), the main psychoactive compound in the plant was identified in 1964 followed by more than 125 cannabinoids. More than 30 flavonoids were isolated from the plant including the characteristic flavonoids called cannflavins, which are prenylated or geranylated flavones. Material and Methods: In this review, the methods of extraction, isolation, identification, biosynthesis, chemical synthesis, analysis and pharmacological activity of these flavonoids are described. Results: The biosynthetic routes of the cannflavins from phenylalanine and malonyl CoA as well as the microbial biotransformation are also discussed. Details of the chemical synthesis are illustrated as an alternative to the isolation from the plant materials along with other possible sources of obtaining cannflavins. Detailed methods discussing the analysis of flavonoids in cannabis are presented, including the techniques used for separation and detection. Finally, the various biological activities of cannflavins are reviewed along with the available molecular docking studies. Conclusion: Despite the low level of cannflavins in cannabis hamper their development as naturally derived products, efforts need to be put in place to develop high yield synthetic or biosynthetic protocols for their production in order for their development as pharmaceutical products.
Collapse
Affiliation(s)
- Maged S. Abdel-Kader
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mohamed M. Radwan
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Mississippi, USA
| | - Ahmed M. Metwaly
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ibrahim H. Eissa
- Department of Pharmaceutical Medicinal Chemistry & Drug Design, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | | | - Mahmoud A. ElSohly
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Mississippi, USA
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, Mississippi, USA
| |
Collapse
|
4
|
Mohanty SS, Sahoo CR, Paidesetty SK, Padhy RN. Role of phytocompounds as the potential anti-viral agent: an overview. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2311-2329. [PMID: 37160482 PMCID: PMC10169142 DOI: 10.1007/s00210-023-02517-2] [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: 02/04/2023] [Accepted: 04/28/2023] [Indexed: 05/11/2023]
Abstract
Viral diseases are the most notorious infective agent(s) causing morbidity and mortality in every nook and corner for ages; viruses are active in host cells, and specific anti-virus medicines' developments remain uncanny. In this century of the biological era, human viruses act predominantly as versatile spreaders. The infection of the present COVID-19 virus is up in the air; blithely, the integument of medicinal chemistry approaches, particularly bioactive derived phytocompounds could be helpful to control those human viruses, recognized in the last 100 years. Indeed, natural products are being used for various therapeutic purposes. The major bioactive phytocompounds are chemically containing coumarin, thiosulfonate, steroid, polysaccharide, tannin, lignin, proanthocyanidin, terpene, quinone, saponin, flavonoid, alkaloid, and polyphenol, that are documented for inhibitory action against several viral infections. Mostly, about 20-30% of plants from tropical or temperate regions are known to have some antiviral activity. This comprehensive analysis of bioactive-derived phytocompounds would represent a significant impact and might be helpful for antiviral research and the current state of viral treatments.
Collapse
Affiliation(s)
- Swati Sucharita Mohanty
- Department of Medical Oncology, IMS & Sum Hospital, Siksha ‘O’ Anusandhan Deemed to Be University, Bhubaneswar, 751003 Odisha India
| | - Chita Ranjan Sahoo
- Central Research Laboratory, IMS & Sum Hospital, Siksha ‘O’ Anusandhan Deemed to Be University, Bhubaneswar, 751003 Odisha India
- Present Address: Department of Health Research, Ministry of Health & Family Welfare, Govt. of India, ICMR-Regional Medical Research Centre, 751023 Bhubaneswar, India
| | - Sudhir Kumar Paidesetty
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Siksha ‘O’ Anusandhan Deemed to Be University, Bhubaneswar, 751003 Odisha India
| | - Rabindra Nath Padhy
- Central Research Laboratory, IMS & Sum Hospital, Siksha ‘O’ Anusandhan Deemed to Be University, Bhubaneswar, 751003 Odisha India
| |
Collapse
|
5
|
Rababi D, Nag A. Evaluation of therapeutic potentials of selected phytochemicals against Nipah virus, a multi-dimensional in silico study. 3 Biotech 2023; 13:174. [PMID: 37180429 PMCID: PMC10170460 DOI: 10.1007/s13205-023-03595-y] [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: 02/11/2023] [Accepted: 04/26/2023] [Indexed: 05/16/2023] Open
Abstract
The current study attempted to evaluate the potential of fifty-three (53) natural compounds as Nipah virus attachment glycoprotein (NiV G) inhibitors through in silico molecular docking study. Pharmacophore alignment of the four (4) selected compounds (Naringin, Mulberrofuran B, Rutin and Quercetin 3-galactoside) through Principal Component Analysis (PCA) revealed that common pharmacophores, namely four H bond acceptors, one H bond donor and two aromatic groups were responsible for the residual interaction with the target protein. Out of these four compounds, Naringin was found to have the highest inhibitory potential ( - 9.19 kcal mol-1) against the target protein NiV G, when compared to the control drug, Ribavirin ( - 6.95 kcal mol-1). The molecular dynamic simulation revealed that Naringin could make a stable complex with the target protein in the near-native physiological condition. Finally, MM-PBSA (Molecular Mechanics-Poisson-Boltzmann Solvent-Accessible Surface Area) analysis in agreement with our molecular docking result, showed that Naringin ( - 218.664 kJ mol-1) could strongly bind with the target protein NiV G than the control drug Ribavirin ( - 83.812 kJ mol-1). Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03595-y.
Collapse
Affiliation(s)
- Deblina Rababi
- Department of Life Sciences, Bangalore Central Campus, CHRIST (Deemed to be University), Bangalore, India
| | - Anish Nag
- Department of Life Sciences, Bangalore Central Campus, CHRIST (Deemed to be University), Bangalore, India
| |
Collapse
|
6
|
Pereira RS, Santos FCP, Campana PRV, Costa VV, de Pádua RM, Souza DG, Teixeira MM, Braga FC. Natural Products and Derivatives as Potential Zika virus Inhibitors: A Comprehensive Review. Viruses 2023; 15:v15051211. [PMID: 37243296 DOI: 10.3390/v15051211] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/30/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Zika virus (ZIKV) is an arbovirus whose infection in humans can lead to severe outcomes. This article reviews studies reporting the anti-ZIKV activity of natural products (NPs) and derivatives published from 1997 to 2022, which were carried out with NPs obtained from plants (82.4%) or semisynthetic/synthetic derivatives, fungi (3.1%), bacteria (7.6%), animals (1.2%) and marine organisms (1.9%) along with miscellaneous compounds (3.8%). Classes of NPs reported to present anti-ZIKV activity include polyphenols, triterpenes, alkaloids, and steroids, among others. The highest values of the selectivity index, the ratio between cytotoxicity and antiviral activity (SI = CC50/EC50), were reported for epigallocatechin gallate (SI ≥ 25,000) and anisomycin (SI ≥ 11,900) obtained from Streptomyces bacteria, dolastane (SI = 1246) isolated from the marine seaweed Canistrocarpus cervicorni, and the flavonol myricetin (SI ≥ 862). NPs mostly act at the stages of viral adsorption and internalization in addition to presenting virucidal effect. The data demonstrate the potential of NPs for developing new anti-ZIKV agents and highlight the lack of studies addressing their molecular mechanisms of action and pre-clinical studies of efficacy and safety in animal models. To the best of our knowledge, none of the active compounds has been submitted to clinical studies.
Collapse
Affiliation(s)
- Rosângela Santos Pereira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Françoise Camila Pereira Santos
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | | | - Vivian Vasconcelos Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Rodrigo Maia de Pádua
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Daniele G Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Fernão Castro Braga
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
7
|
Sadeer NB, El Kalamouni C, Khalid A, Abdalla AN, Zengin G, Khoa Bao LV, Mahomoodally MF. Secondary metabolites as potential drug candidates against Zika virus, an emerging looming human threat: Current landscape, molecular mechanism and challenges ahead. J Infect Public Health 2023; 16:754-770. [PMID: 36958171 DOI: 10.1016/j.jiph.2023.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/12/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Nature has given us yet another wild card in the form of Zika virus (ZIKV). It was found in 1947, but has only recently become an important public health risk, predominantly to pregnant women and their unborn offspring. Currently, no specific therapeutic agent exists for ZIKV and treatment is mainly supportive. Natural products (NPs) can serve as a major source of potent antiviral drugs. To create this review, a comprehensive search was conducted from different databases (PubMed, ScienceDirect, Google scholar). A statistical analysis on the number of publications related to NPs and ZIKV was conducted to analyse the trend in research covering the period 1980-2020. From the data collated in this review, a number of NPs have been found to be inhibitive towards different stages of the ZIKV lifecycle in in vitro studies. For instance, baicalin, (-)-epigallocatechin gallate, curcumin, nanchangmycin, gossypol, cephaeline, emetine, resveratrol, berberine, amongst others, can prevent viral entry by attacking ZIKV E protein. Compounds luteolin, myricetin, astragalin, rutin, (-)-epigallocatechin gallate, carnosine, pedalitin, amongst others, inhibited NS2B-NS3 protease activity which consequently hamper replication. Interestingly, a few NPs had the ability to arrest both viral entry and replication, namely baicalin, (-)-epigallocatechin gallate, curcumin, cephaeline, emetine, and resveratrol. To the best of our knowledge, we obtained only one in vivo study conducted on emetine and results showed that it decreased the levels of circulating ZIKV by approximately 10-fold. Our understanding on NPs exhibiting anti-ZIKV effects in in vivo testing as well as clinical trials is limited. Our trend analysis showed that interest in searching for a cure or prevention against Zika in NPs is negligible and there are no publications yet covering the clinical evaluation. NPs with anti-ZIKV property can a winning strategy in controlling the bio-burden of an epidemic or pandemic. We therefore opine that in the future, more research should be devoted to ZIKV. This review attempts to provide baseline data and roadmap to pursuit detailed investigations for developing potent and novel therapeutic agents to prevent and cure ZIKV infection.
Collapse
Affiliation(s)
- Nabeelah Bibi Sadeer
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Mauritius
| | - Chaker El Kalamouni
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France.
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan 45142, Saudi Arabia; Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Center for Research, P. O. Box 2404, Khartoum, the Republic of the Sudan
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University, Campus, 42250 Konya, Turkey
| | - Le Van Khoa Bao
- Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam; School of Engineering & Technology, Duy Tan University, Da Nang, Viet Nam.
| | - Mohamad Fawzi Mahomoodally
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Mauritius; Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Chennai 600077, India
| |
Collapse
|
8
|
Sadeer NB, Haddad JG, Ezzat MO, Desprès P, Abdallah HH, Zengin G, Alshamrani IM, Barnawi J, Khalid A, Abdalla AN, Le Van B, El Kalamouni C, Mahomoodally MF. Rhizophora mucronata Lam., a halophyte from Mauritius Island, inhibits the entry of Zika virus in human cells (A549)- an in vitro and in silico analysis. J Biomol Struct Dyn 2023; 41:12599-12609. [PMID: 36648248 DOI: 10.1080/07391102.2023.2167115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023]
Abstract
The recent appearance of Zika virus (ZIKV) in Brazil should serve as a wake-up call to international authorities, as it poses a threat to global public health. In the present study, we investigated whether a mangrove plant, Rhizophora mucronata Lam. (R. mucronata) collected in Mauritius, possesses anti-ZIKV activity at the non-cytotoxic doses. ZIKVMC-MR766NIID (ZIKVGFP) was used for assessing anti ZIKV activity. In silico docking (Autodock 4) and molecular simulation were performed on collected data. Using a recombinant ZIKV expressing reporter green fluorescent protein(GFP) protein, we discovered that fruit and root methanolic, decocted fruit and root extracts were effective inhibitors of ZIKV infection in human epithelial A549 cells at negligible cytotoxicity. The mechanisms by which such extracts prevented ZIKV infection are linked to the inability of the virus to attach to the host cell surface. The outcomes of this study were supported by the docking calculations in which some of the dominant compounds have shown high binding affinity against ZIKV. The scientific data gathered in this study might pave the way for the future development of possible R. mucronata inhibitors to combat ZIKV.fCommunicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nabeelah Bibi Sadeer
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Mauritius
| | - Juliano G Haddad
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, La Réunion, France
| | - Mohammed Oday Ezzat
- Department of Chemistry, College of Education for Women, University of Anbar, Ramadi, Anbar, Iraq
| | - Philippe Desprès
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, La Réunion, France
| | - Hassan H Abdallah
- Chemistry Department, College of Education, Salahaddin University-erbil, erbil, Iraq
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Campus, Konya, Turkey
| | | | - Jameel Barnawi
- Department of Medical Lab Technology, Prince Fahd Bin Sultan Research chair, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan, Saudi Arabia
- Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Center for Research, Khartoum, Sudan
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Bao Le Van
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- Faculty of Natural Sciences, Duy Tan University, Da Nang, Vietnam
| | - Chaker El Kalamouni
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, La Réunion, France
| | - Mohamad Fawzi Mahomoodally
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Mauritius
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Chennai, India
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen), North West University, Potchefstroom, South Africa
| |
Collapse
|
9
|
Zothantluanga JH, Umar AK, Lalhlenmawia H, Vinayagam S, Borthakur MS, Patowary L, Tayeng D. Computational screening of phytochemicals for anti-parasitic drug discovery. PHYTOCHEMISTRY, COMPUTATIONAL TOOLS AND DATABASES IN DRUG DISCOVERY 2023:257-283. [DOI: 10.1016/b978-0-323-90593-0.00005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
10
|
Muthuraj PG, Krishnamoorthy C, Anderson-Berry A, Hanson C, Natarajan SK. Novel Therapeutic Nutrients Molecules That Protect against Zika Virus Infection with a Special Note on Palmitoleate. Nutrients 2022; 15:124. [PMID: 36615782 PMCID: PMC9823984 DOI: 10.3390/nu15010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/11/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Zika virus (ZIKV) is a Flavivirus from the Flaviviridae family and a positive-sense single strand RNA virus. ZIKV infection can cause a mild infection to the mother but can be vertically transmitted to the developing fetus, causing congenital anomalies. The prevalence of ZIKV infections was relatively insignificant with sporadic outbreaks in the Asian and African continents until 2006. However, recent epidemic in the Caribbean showed significant increased incidence of Congenital Zika Syndrome. ZIKV infection results in placental pathology which plays a crucial role in disease transmission from mother to fetus. Currently, there is no Food and Drug Administration (FDA) approved vaccine or therapeutic drug against ZIKV. This review article summarizes the recent advances on ZIKV transmission and diagnosis and reviews nutraceuticals which can protect against the ZIKV infection. Further, we have reviewed recent advances related to the novel therapeutic nutrient molecules that have been shown to possess activity against Zika virus infected cells. We also review the mechanism of ZIKV-induced endoplasmic reticulum and apoptosis and the protective role of palmitoleate (nutrient molecule) against ZIKV-induced ER stress and apoptosis in the placental trophoblasts.
Collapse
Affiliation(s)
- Philma Glora Muthuraj
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Chandan Krishnamoorthy
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Ann Anderson-Berry
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Corrine Hanson
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Medical Nutrition Education, College of Allied Health Profession, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Medical Nutrition Education, College of Allied Health Profession, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
11
|
Sharma D, Sharma N, Manchanda N, Prasad SK, Sharma PC, Thakur VK, Rahman MM, Dhobi M. Bioactivity and In Silico Studies of Isoquinoline and Related Alkaloids as Promising Antiviral Agents: An Insight. Biomolecules 2022; 13:17. [PMID: 36671402 PMCID: PMC9856122 DOI: 10.3390/biom13010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Viruses are widely recognized as the primary cause of infectious diseases around the world. The ongoing global pandemic due to the emergence of SARS-CoV-2 further added fuel to the fire. The development of therapeutics becomes very difficult as viruses can mutate their genome to become more complex and resistant. Medicinal plants and phytocompounds could be alternative options. Isoquinoline and their related alkaloids are naturally occurring compounds that interfere with multiple pathways including nuclear factor-κB, mitogen-activated protein kinase/extracellular-signal-regulated kinase, and inhibition of Ca2+-mediated fusion. These pathways play a crucial role in viral replication. Thus, the major goal of this study is to comprehend the function of various isoquinoline and related alkaloids in viral infections by examining their potential mechanisms of action, structure-activity relationships (SAR), in silico (particularly for SARS-CoV-2), in vitro and in vivo studies. The current advancements in isoquinoline and related alkaloids as discussed in the present review could facilitate an in-depth understanding of their role in the drug discovery process.
Collapse
Affiliation(s)
- Divya Sharma
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| | - Neetika Sharma
- Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| | - Namish Manchanda
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| | - Satyendra K. Prasad
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
| | - Prabodh Chander Sharma
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Centre, Scotland’s Rural College (SRUC), Kings Buildings, 11 West Mains Road, Edinburgh EH9 3JG, UK
- School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, India
| | - M. Mukhlesur Rahman
- Pharmaceutical and Natural Products Chemistry, School of Health, Sports and Bioscience, University of East London, Stratford Campus, London E15 4LZ, UK
| | - Mahaveer Dhobi
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, New Delhi 110017, India
| |
Collapse
|
12
|
Ramiharimanana FD, Haddad JG, Andrianavalonirina MA, Apel C, Olivon F, Diotel N, Desprès P, Ramanandraibe VV, El Kalamouni C. Antiviral Effect of Stenocline ericoides DC. and Stenocline inuloides DC., Two Flavonoid-Rich Endemic Plants from Madagascar, against Dengue and Zika Viruses. Pharmaceuticals (Basel) 2022; 15:ph15121500. [PMID: 36558951 PMCID: PMC9787939 DOI: 10.3390/ph15121500] [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: 11/04/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Dengue and Zika viruses are identified as the most medically important arthropod-borne viral pathogens. Over the past 20 years, the global dengue incidence has dramatically increased with epidemics of severe dengue where the case fatality rate can reach up to 20% in untreated patients. The association between Zika virus infection and severe congenital anomalies was first reported in 2015. Today no specific antiviral therapies are available for dengue and Zika virus infections, accentuating the need of adapted antiviral strategies based on medicinal plant drug discovery. Plants are a potential source of antiviral phytocompounds which act primarily by blocking virus entry in the host-cell. In the present study, we evaluated whether crude extracts from Stenocline ericoides DC. and Stenocline inuloides DC., two endemic plants from Madagascar, may have antiviral effects against dengue and Zika viruses. We showed that S. ericoides has virucidal action whereas S. inuloides inhibits the early steps of virus infection with a non-cytotoxic effect in human cells. The administration of S. ericoides and S. inuloides extracts in zebrafish had no effect on the behavior of animals at the active doses against dengue and Zika viruses, suggesting the absence of adverse effects at these doses. LC-HRMS2 and molecular networking analyses revealed the richness of these two plants in polyphenols and flavonoid with the presence of clusters of phytocompounds specific to each Stenocline species. Consequently, S. ericoides and S. inuloides represent potential sources for natural and safe antiviral phytocompounds against flaviviruses of medical concern.
Collapse
Affiliation(s)
- Fenia D. Ramiharimanana
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Université de la Réunion, 94791 Sainte Clotilde, France
- International Associated Laboratory, University of Antananarivo-Lyon 1, Antananarivo P.O. Box 906, Madagascar
| | - Juliano G. Haddad
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Université de la Réunion, 94791 Sainte Clotilde, France
| | | | - Cécile Apel
- Institut de Chimie des Substances Naturelles, CNRS, University of Paris-Saclay, UPR 2301, 91198 Gif-sur-Yvette, France
| | | | - Nicolas Diotel
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM, UMR 1188, 97491 Sainte Clotilde, France
| | - Philippe Desprès
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Université de la Réunion, 94791 Sainte Clotilde, France
| | | | - Chaker El Kalamouni
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Université de la Réunion, 94791 Sainte Clotilde, France
- Correspondence:
| |
Collapse
|
13
|
Yilmaz H, Gultekin Subasi B, Celebioglu HU, Ozdal T, Capanoglu E. Chemistry of Protein-Phenolic Interactions Toward the Microbiota and Microbial Infections. Front Nutr 2022; 9:914118. [PMID: 35845785 PMCID: PMC9284217 DOI: 10.3389/fnut.2022.914118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Along with health concerns, interest in plants as food and bioactive phytochemical sources has been increased in the last few decades. Phytochemicals as secondary plant metabolites have been the subject of many studies in different fields. Breakthrough for research interest on this topic is re-juvenilized with rising relevance in this global pandemics' era. The recent COVID-19 pandemic attracted the attention of people to viral infections and molecular mechanisms behind these infections. Thus, the core of the present review is the interaction of plant phytochemicals with proteins as these interactions can affect the functions of co-existing proteins, especially focusing on microbial proteins. To the best of our knowledge, there is no work covering the protein-phenolic interactions based on their effects on microbiota and microbial infections. The present review collects and defines the recent data, representing the interactions of phenolic compounds -primarily flavonoids and phenolic acids- with various proteins and explores how these molecular-level interactions account for the human health directly and/or indirectly, such as increased antioxidant properties and antimicrobial capabilities. Furthermore, it provides an insight about the further biological activities of interacted protein-phenolic structure from an antiviral activity perspective. The research on the protein-phenolic interaction mechanisms is of great value for guiding how to take advantage of synergistic effects of proteins and polyphenolics for future medical and nutritive approaches and related technologies.
Collapse
Affiliation(s)
- Hilal Yilmaz
- Department of Biotechnology, Faculty of Science, Bartin University, Bartin, Turkey
| | - Busra Gultekin Subasi
- Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
- Hafik Kamer Ornek MYO, Sivas Cumhuriyet University, Sivas, Turkey
| | | | - Tugba Ozdal
- Department of Food Engineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, Turkey
| | - Esra Capanoglu
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Istanbul, Turkey
- *Correspondence: Esra Capanoglu
| |
Collapse
|
14
|
Roshni J, Vaishali R, Ganesh KS, Dharani N, Alzahrani KJ, Banjer HJ, Alghamdi AH, Theyab A, Ahmed SS, Patil S. Multi-target potential of Indian phytochemicals against SARS-CoV-2: A docking, molecular dynamics and MM-GBSA approach extended to Omicron B.1.1.529. J Infect Public Health 2022; 15:662-669. [PMID: 35617830 PMCID: PMC9101941 DOI: 10.1016/j.jiph.2022.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/24/2022] [Accepted: 05/04/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND SARS-CoV-2, an emerged strain of corona virus family became almost serious health concern worldwide. Despite vaccines availability, reports suggest the occurrence of SARS-CoV-2 infection even in a vaccinated population. With frequent evolution and expected multiple COVID-19 waves, improved preventive, diagnostic, and treatment measures are required. In recent times, phytochemicals have gained attention due to their therapeutic characteristics and are suggested as alternative and complementary treatments for infectious diseases. This present study aimed to identify potential inhibitors against reported protein targets of SARS-CoV-2. METHODOLOGY We computationally investigated potential SARS-CoV-2 protein targets from the literature and collected druggable phytochemicals from Indian Medicinal Plants, Phytochemistry and Therapeutics (IMPPAT) database. Further, we implemented a systematic workflow of molecular docking, dynamic simulations and generalized born surface area free-energy calculations (MM-GBSA). RESULTS Extensive literature search and assessment of 1508 articles identifies 13 potential SARS-CoV-2 protein targets. We screened 501 druggable phytochemicals with proven biological activities. Analysis of 6513(501 *13) docked phytochemicals complex, 26 were efficient against SARS-CoV-2. Amongst, 4,8-dihydroxysesamin and arboreal from Gmelina arborea were ranked potential against most of the targets with binding energy ranging between - 10.7 to - 8.2 kcal/mol. Additionally, comparative docking with known drugs such as arbidol (-6.6 to -5.1 kcal/mol), favipiravir (-5.5 to -4.5 kcal/mol), hydroxychloroquine (-6.5 to -5.1 kcal/mol), and remedesivir (-8.0 to -5.3 kcal/mol) revealed equal/less affinity than 4,8-dihydroxysesamin and arboreal. Interestingly, the nucleocapsid target was found commonly inhibited by 4,8-dihydroxysesamin and arboreal. Molecular dynamic simulation and Molecular mechanics generalized born surface area (MM-GBSA)calculations reflect that both the compounds possess high inhibiting potential against SARS-CoV-2 including the recently emerged Omicron variant (B.1.1.529). CONCLUSION Overall our study imparts the usage of phytochemicals as antiviral agents for SARS-CoV-2 infection. Additional in vitro and in vivo testing of these phytochemicals is required to confirm their potency.
Collapse
Affiliation(s)
- Jency Roshni
- Department of Medical Biotechnology, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam 603103, India.
| | - R Vaishali
- Department of Medical Biotechnology, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam 603103, India.
| | - K S Ganesh
- Department of Medical Biotechnology, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam 603103, India.
| | - N Dharani
- Department of Medical Biotechnology, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam 603103, India.
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Hamsa Jameel Banjer
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Ali H Alghamdi
- Department of Biology, Faculty of Science, Albaha University, Albaha 7738-65799, Saudi Arabia.
| | - Abdulrahman Theyab
- College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh, 11533, Saudi Arabia; Department of Laboratory Medicine, Security Forces Hospital, Mecca, Saudi Arabia.
| | - Shiek Ssj Ahmed
- Multi-omics and Drug Discovery Lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education(CARE), Kelambakkam 603103, India.
| | - Shankargouda Patil
- Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Jazan 45412, Saudi Arabia.
| |
Collapse
|
15
|
Tamkutė L, Haddad JG, Diotel N, Desprès P, Venskutonis PR, El Kalamouni C. Cranberry Pomace Extract Exerts Antiviral Activity against Zika and Dengue Virus at Safe Doses for Adult Zebrafish. Viruses 2022; 14:1101. [PMID: 35632841 PMCID: PMC9147401 DOI: 10.3390/v14051101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023] Open
Abstract
Mosquito-borne dengue virus (DENV) and zika virus (ZIKV) infections constitute a global health emergency. Antivirals directly targeting the virus infectious cycle are still needed to prevent dengue hemorrhagic fever and congenital zika syndrome. In the present study, we demonstrated that Cranberry Pomace (CP) extract, a polyphenol-rich agrifood byproduct recovered following cranberry juice extraction, blocks DENV and ZIKV infection in human Huh7.5 and A549 cell lines, respectively, in non-cytotoxic concentrations. Our virological assays identified CP extract as a potential inhibitor of virus entry into the host-cell by acting directly on viral particles, thus preventing their attachment to the cell surface. At effective antiviral doses, CP extract proved safe and tolerable in a zebrafish model. In conclusion, polyphenol-rich agrifood byproducts such as berry extracts are a promising source of safe and naturally derived nutraceutical antivirals that target medically important pathogens.
Collapse
Affiliation(s)
- Laura Tamkutė
- Unité Mixte Processus Infectieux En Milieu Insulaire Tropical, Université De La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (L.T.); (J.G.H.); (P.D.)
- Department of Food Science and Technology, Kaunas University of Technology, Radvilenu, pl. 19, LT-50254 Kaunas, Lithuania;
| | - Juliano G. Haddad
- Unité Mixte Processus Infectieux En Milieu Insulaire Tropical, Université De La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (L.T.); (J.G.H.); (P.D.)
| | - Nicolas Diotel
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM, UMR 1188, 97490 Saint-Denis de La Réunion, France;
| | - Philippe Desprès
- Unité Mixte Processus Infectieux En Milieu Insulaire Tropical, Université De La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (L.T.); (J.G.H.); (P.D.)
| | - Petras Rimantas Venskutonis
- Department of Food Science and Technology, Kaunas University of Technology, Radvilenu, pl. 19, LT-50254 Kaunas, Lithuania;
| | - Chaker El Kalamouni
- Unité Mixte Processus Infectieux En Milieu Insulaire Tropical, Université De La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (L.T.); (J.G.H.); (P.D.)
| |
Collapse
|
16
|
Fischer TR, Meidner L, Schwickert M, Weber M, Zimmermann RA, Kersten C, Schirmeister T, Helm M. Chemical biology and medicinal chemistry of RNA methyltransferases. Nucleic Acids Res 2022; 50:4216-4245. [PMID: 35412633 PMCID: PMC9071492 DOI: 10.1093/nar/gkac224] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/17/2022] [Accepted: 04/08/2022] [Indexed: 12/24/2022] Open
Abstract
RNA methyltransferases (MTases) are ubiquitous enzymes whose hitherto low profile in medicinal chemistry, contrasts with the surging interest in RNA methylation, the arguably most important aspect of the new field of epitranscriptomics. As MTases become validated as drug targets in all major fields of biomedicine, the development of small molecule compounds as tools and inhibitors is picking up considerable momentum, in academia as well as in biotech. Here we discuss the development of small molecules for two related aspects of chemical biology. Firstly, derivates of the ubiquitous cofactor S-adenosyl-l-methionine (SAM) are being developed as bioconjugation tools for targeted transfer of functional groups and labels to increasingly visible targets. Secondly, SAM-derived compounds are being investigated for their ability to act as inhibitors of RNA MTases. Drug development is moving from derivatives of cosubstrates towards higher generation compounds that may address allosteric sites in addition to the catalytic centre. Progress in assay development and screening techniques from medicinal chemistry have led to recent breakthroughs, e.g. in addressing human enzymes targeted for their role in cancer. Spurred by the current pandemic, new inhibitors against coronaviral MTases have emerged at a spectacular rate, including a repurposed drug which is now in clinical trial.
Collapse
Affiliation(s)
- Tim R Fischer
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Laurenz Meidner
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Marvin Schwickert
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Marlies Weber
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Robert A Zimmermann
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Christian Kersten
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Mark Helm
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| |
Collapse
|
17
|
Sundar S, Piramanayagam S, Natarajan J. A review on structural genomics approach applied for drug discovery against three vector-borne viral diseases: Dengue, Chikungunya and Zika. Virus Genes 2022; 58:151-171. [PMID: 35394596 DOI: 10.1007/s11262-022-01898-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/22/2022] [Indexed: 12/22/2022]
Abstract
Structural genomics involves the advent of three-dimensional structures of the genome encoded proteins through various techniques available. Numerous structural genomics research groups have been developed across the globe and they contribute enormously to the identification of three-dimensional structures of various proteins. In this review, we have discussed the applications of the structural genomics approach towards the discovery of potential lead-like molecules against the genomic drug targets of three vector-borne diseases, namely, Dengue, Chikungunya and Zika. Currently, all these three diseases are associated with the most important global public health problems and significant economic burden in tropical countries. Structural genomics has accelerated the identification of novel drug targets and inhibitors for the treatment of these diseases. We start with the current development status of the drug targets and antiviral drugs against these three diseases and conclude by describing challenges that need to be addressed to overcome the shortcomings in the process of drug discovery.
Collapse
Affiliation(s)
- Shobana Sundar
- Computational Biology Lab, Department of Bioinformatics, Bharathiar University, Coimbatore, India
| | | | - Jeyakumar Natarajan
- Data Mining and Text Mining Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
18
|
Bibi Sadeer N, Haddad JG, Oday Ezzat M, Desprès P, Abdallah HH, Zengin G, Uysal A, El Kalamouni C, Gallo M, Montesano D, Mahomoodally MF. Bruguiera gymnorhiza (L.) Lam. at the Forefront of Pharma to Confront Zika Virus and Microbial Infections-An In Vitro and In Silico Perspective. Molecules 2021; 26:5768. [PMID: 34641314 PMCID: PMC8510246 DOI: 10.3390/molecules26195768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023] Open
Abstract
The recent emergence of Zika virus (ZIKV) in Brazil and the increasing resistance developed by pathogenic bacteria to nearly all existing antibiotics should be taken as a wakeup call for the international authority as this represents a risk for global public health. The lack of antiviral drugs and effective antibiotics on the market triggers the need to search for safe therapeutics from medicinal plants to fight viral and microbial infections. In the present study, we investigated whether a mangrove plant, Bruguiera gymnorhiza (L.) Lam. (B. gymnorhiza) collected in Mauritius, possesses antimicrobial and antibiotic potentiating abilities and exerts anti-ZIKV activity at non-cytotoxic doses. Microorganisms Escherichia coli ATCC 25922, Pseudomonas aeruginosa ATCC 27853, Klebsiella pneumoniae ATCC 70603, methicillin-resistant Staphylococcus aureus ATCC 43300 (MRSA), Salmonella enteritidis ATCC 13076, Sarcina lutea ATCC 9341, Proteus mirabilis ATCC 25933, Bacillus cereus ATCC 11778 and Candida albicans ATCC 26555 were used to evaluate the antimicrobial properties. Ciprofloxacin, chloramphenicol and streptomycin antibiotics were used for assessing antibiotic potentiating activity. ZIKVMC-MR766NIID (ZIKVGFP) was used for assessing anti-ZIKV activity. In silico docking (Autodock 4) and ADME (SwissADME) analyses were performed on collected data. Antimicrobial results revealed that Bruguiera twig ethyl acetate (BTE) was the most potent extract inhibiting the growth of all nine microbes tested, with minimum inhibitory concentrations ranging from 0.19-0.39 mg/mL. BTE showed partial synergy effects against MRSA and Pseudomonas aeruginosa when applied in combination with streptomycin and ciprofloxacin, respectively. By using a recombinant ZIKV-expressing reporter GFP protein, we identified both Bruguiera root aqueous and Bruguiera fruit aqueous extracts as potent inhibitors of ZIKV infection in human epithelial A549 cells. The mechanisms by which such extracts prevented ZIKV infection are linked to the inability of the virus to bind to the host cell surface. In silico docking showed that ZIKV E protein, which is involved in cell receptor binding, could be a target for cryptochlorogenic acid, a chemical compound identified in B. gymnorhiza. From ADME results, cryptochlorogenic acid is predicted to be not orally bioavailable because it is too polar. Scientific data collected in this present work can open a new avenue for the development of potential inhibitors from B. gymnorhiza to fight ZIKV and microbial infections in the future.
Collapse
Affiliation(s)
- Nabeelah Bibi Sadeer
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit 80837, Mauritius;
| | - Juliano G. Haddad
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France; (J.G.H.); (P.D.); (C.E.K.)
| | - Mohammed Oday Ezzat
- Department of Chemistry, College of Education for Women, University of Anbar, Ramadi 31001, Iraq;
| | - Philippe Desprès
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France; (J.G.H.); (P.D.); (C.E.K.)
| | - Hassan H. Abdallah
- Chemistry Department, College of Education, Salahaddin University-Erbil, Erbil 44001, Iraq;
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Campus, 42130 Konya, Turkey;
| | - Ahmet Uysal
- Department of Medicinal Laboratory, Vocational School of Health Services, Selcuk University, 42130 Konya, Turkey;
| | - Chaker El Kalamouni
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France; (J.G.H.); (P.D.); (C.E.K.)
| | - Monica Gallo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, via Pansini 5, 80131 Naples, Italy;
| | - Domenico Montesano
- Department of Pharmacy, University of Naples Federico II, via D. Montesano 49, 80131 Naples, Italy
| | - Mohamad Fawzi Mahomoodally
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit 80837, Mauritius;
| |
Collapse
|
19
|
Dharmashekara C, Pradeep S, Prasad SK, Jain AS, Syed A, Prasad KS, Patil SS, Beelagi MS, Srinivasa C, Shivamallu C. Virtual screening of potential phyto-candidates as therapeutic leads against SARS-CoV-2 infection. ENVIRONMENTAL CHALLENGES (AMSTERDAM, NETHERLANDS) 2021; 4:100136. [PMID: 38620722 PMCID: PMC8110638 DOI: 10.1016/j.envc.2021.100136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 05/08/2023]
Abstract
The outbreak of novel coronavirus strain (Covid-19) with a high pandemic threat has predict grave public health and economic concerns. This virus, originating from the Wuhan region in China has spread worldwide affecting millions with no registered persuasive targeted therapy. In this paper, we analyze the three important proteins encoded by the virus, envelope protein 5 × 29, RNA binding nucleocapsid protein 1SSK, and spike glycoprotein 6ACD, for an effective virion accumulation, and remdesivir was the first drug approved by the FDA and EMA for the treatment of COVID-19 cases that require hospitalization, there is still much controversy about its efficacy and also an alternative for novel phytochemicals, deoxynojirimycin, trigoneoside IB, and octanoic acid. The in-silico evaluations were conducted using the PyRx virtual screening tools which lead to the target based on high binding affinity. Trigoneoside IB, derived from Trigonella foenum-graecum (Fenugreek), showed the highest binding affinity and stable interaction with the amino acid residues present in active sites of Covid-19 proteins. Meanwhile, the other two compounds derived from Morus alba (Mulberry) and Morinda citrifolia (Noni), as well as the anti-HIV remdesivir drug exhibited good binding affinity and favorable ADME properties. Thereby offering scope for validation of the new therapeutic components for their in vitro and in vivo efficacy against the Covid-19 proteins.
Collapse
Affiliation(s)
- Chandan Dharmashekara
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka 570015, India
| | - Sushma Pradeep
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka 570015, India
| | - Shashanka K Prasad
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka 570015, India
| | - Anisha S Jain
- Department of Microbiology and Tissue Culture, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka 570015, India
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Kollur Shiva Prasad
- Department of Sciences, Amrita School of Arts and Sciences, Amrita Vishwa Vidyapeetham, Mysuru campus, Karnataka, India
| | - Sharanagouda S Patil
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Yelahanka, Bengaluru 560064, India
| | - Mallikarjun S Beelagi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka 570015, India
| | - Chandrashekar Srinivasa
- Department of Studies in Biotechnology, Davangere University, Davangere 577007, Karnataka, India
| | - Chandan Shivamallu
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka 570015, India
| |
Collapse
|
20
|
Insights on Dengue and Zika NS5 RNA-dependent RNA polymerase (RdRp) inhibitors. Eur J Med Chem 2021; 224:113698. [PMID: 34274831 DOI: 10.1016/j.ejmech.2021.113698] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/20/2022]
Abstract
Over recent years, many outbreaks caused by (re)emerging RNA viruses have been reported worldwide, including life-threatening Flaviviruses, such as Dengue (DENV) and Zika (ZIKV). Currently, there is only one licensed vaccine against Dengue, Dengvaxia®. However, its administration is not recommended for children under nine years. Still, there are no specific inhibitors available to treat these infectious diseases. Among the flaviviral proteins, NS5 RNA-dependent RNA polymerase (RdRp) is a metalloenzyme essential for viral replication, suggesting that it is a promising macromolecular target since it has no human homolog. Nowadays, several NS5 RdRp inhibitors have been reported, while none inhibitors are currently in clinical development. In this context, this review constitutes a comprehensive work focused on RdRp inhibitors from natural, synthetic, and even repurposing sources. Furthermore, their main aspects associated with the structure-activity relationship (SAR), proposed mechanisms of action, computational studies, and other topics will be discussed in detail.
Collapse
|
21
|
Bautista J, Yu S, Tian L. Flavonoids in Cannabis sativa: Biosynthesis, Bioactivities, and Biotechnology. ACS OMEGA 2021; 6:5119-5123. [PMID: 33681553 PMCID: PMC7931196 DOI: 10.1021/acsomega.1c00318] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/11/2021] [Indexed: 05/02/2023]
Abstract
Although Cannabis sativa synthesizes a wide range of phytochemicals, much attention has been primarily given to two phytocannabinoids, Δ9-tetrahydocannabinol (THC) and cannabidiol (CBD), due to their distinctive activities in humans. These bioactivities can be further enhanced through the interaction of THC and CBD with other phytocannabinoids or non-phytocannabinoid chemicals, such as terpenes and flavonoids, a phenomenon that is termed the entourage effect. Flavonoid metabolism in C. sativa and the entourage effect are currently understudied. This mini-review examines recent advances in the biosynthesis and bioactivities of cannflavins, which are prenylated (C5) and geranylated (C10) flavones that are relatively unique to C. sativa. We also discuss the rapidly developing omics tools that enable discoveries in flavonoid metabolism in C. sativa and manipulation of flavonoid production through biotechnology. These advances set the stage for interrogating the health benefits of C. sativa flavonoids, deciphering the contribution of flavonoids to the entourage effect, and developing drugs.
Collapse
Affiliation(s)
| | | | - Li Tian
- . Telephone: +1 530 7520940. Fax: +1 530 7529659
| |
Collapse
|
22
|
Erridge S, Mangal N, Salazar O, Pacchetti B, Sodergren MH. Cannflavins - From plant to patient: A scoping review. Fitoterapia 2020; 146:104712. [PMID: 32858172 DOI: 10.1016/j.fitote.2020.104712] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Cannflavins are a group of prenylflavonoids derived from Cannabis sativa L.. Cannflavin A (CFL-A), B (CFL-B) and C (CFL-C) have been heralded for their anti-inflammatory properties in pre-clinical evaluations. This scoping review aims to synthesise the evidence base on cannflavins to provide an overview of the current research landscape to inform research strategies to aid clinical translation. METHODS A scoping review was conducted of EMBASE, MEDLINE, Pubmed, CENTRAL and Google Scholar databases up to 26th February 2020. All studies describing original research on cannflavins and their isomers were included for review. RESULTS 26 full text articles were included. CFL-A and CFL-B demonstrated potent anti-inflammatory activity via inhibition of 12-o-tetradecanoylphorbol 13-acetate induced PGE2 release (CFL-A half maximal inhibitory concentration (IC50): 0.7 μM; CFL-B IC50: 0.7 μM) and microsomal prostaglandin E synthase-1 (CFL-A IC50: 1.8 μM; CFL-B IC50: 3.7 μM). Outcomes were also described in preclinical models of anti-oxidation (CFL-A), anti-parasitic activity (CFL-A, CFL-C), neuroprotection (CFL-A) and cancer (Isocannflavin B, a CFL-B isomer). In-silico screening identified that CFL-A has binding affinity with viral proteins that warrant further investigation. CONCLUSIONS Cannflavins demonstrate a number of promising therapeutic properties, most notably as an anti-inflammatory agent. Low yields of extraction however have previously limited research to small pre-clinical investigations. Identification of cannflavin-rich chemovars, novel extraction techniques and recent identification of a biosynthetic pathway will hopefully allow research to be scaled appropriately. In order to fully evaluate the therapeutic properties of cannflavins focused research now needs to be embedded within institutions with a track-record of clinical translation.
Collapse
Affiliation(s)
- Simon Erridge
- Department of Surgery and Cancer, Imperial College London, UK
| | - Nagina Mangal
- Department of Surgery and Cancer, Imperial College London, UK
| | - Oliver Salazar
- Department of Surgery and Cancer, Imperial College London, UK
| | | | - Mikael H Sodergren
- Department of Surgery and Cancer, Imperial College London, UK; Emmac Life Sciences, London, UK.
| |
Collapse
|
23
|
Haddad JG, Grauzdytė D, Koishi AC, Viranaicken W, Venskutonis PR, Nunes Duarte dos Santos C, Desprès P, Diotel N, El Kalamouni C. The Geraniin-Rich Extract from Reunion Island Endemic Medicinal Plant Phyllanthus phillyreifolius Inhibits Zika and Dengue Virus Infection at Non-Toxic Effect Doses in Zebrafish. Molecules 2020; 25:molecules25102316. [PMID: 32429073 PMCID: PMC7287739 DOI: 10.3390/molecules25102316] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
The mosquito-borne viruses dengue (DENV) and Zika (ZIKV) viruses are two medically important pathogens in tropical and subtropical regions of the world. There is an urgent need of therapeutics against DENV and ZIKV, and medicinal plants are considered as a promising source of antiviral bioactive metabolites. In the present study, we evaluated the ability of Phyllanthus phillyreifolius, an endemic medicinal plant from Reunion Island, to prevent DENV and ZIKV infection in human cells. At non-cytotoxic concentration in vitro, incubation of infected A549 cells with a P. phillyreifolius extract or its major active phytochemical geraniin resulted in a dramatic reduction of virus progeny production for ZIKV as well as four serotypes of DENV. Virological assays showed that P. phillyreifolius extract-mediated virus inhibition relates to a blockade in internalization of virus particles into the host cell. Infectivity studies on ZIKV showed that both P. phillyreifolius and geraniin cause a loss of infectivity of the viral particles. Using a zebrafish model, we demonstrated that administration of P. phillyreifolius and geraniin has no effect on zebrafish locomotor activity while no morbidity nor mortality was observed up to 5 days post-inoculation. Thus, P. phillyreifolius could act as an important source of plant metabolite geraniin which is a promising antiviral compound in the fight against DENV and ZIKV.
Collapse
Affiliation(s)
- Juliano G. Haddad
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (J.G.H.); (W.V.); (P.D.)
| | - Dovilė Grauzdytė
- Department of Food Science and Technology, Kaunas University of Technology, Radvilėnų pl. 19, Kaunas LT-50254, Lithuania; (D.G.); (P.R.V.)
| | - Andrea Cristine Koishi
- Laboratorio de Virologia Molecular, Instituto Carlos Chagas, ICC/FIOCRUZ/PR, Curitiba 81350-010, Brazil; (A.C.K.); (C.N.D.d.S.)
| | - Wildriss Viranaicken
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (J.G.H.); (W.V.); (P.D.)
| | - Petras Rimantas Venskutonis
- Department of Food Science and Technology, Kaunas University of Technology, Radvilėnų pl. 19, Kaunas LT-50254, Lithuania; (D.G.); (P.R.V.)
| | - Claudia Nunes Duarte dos Santos
- Laboratorio de Virologia Molecular, Instituto Carlos Chagas, ICC/FIOCRUZ/PR, Curitiba 81350-010, Brazil; (A.C.K.); (C.N.D.d.S.)
| | - Philippe Desprès
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (J.G.H.); (W.V.); (P.D.)
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97490 Saint-Denis de La Réunion, France;
| | - Chaker El Kalamouni
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (J.G.H.); (W.V.); (P.D.)
- Correspondence: ; Tel.: +33-262-938822
| |
Collapse
|
24
|
Natural Product Medicines for Honey Bees: Perspective and Protocols. INSECTS 2019; 10:insects10100356. [PMID: 31635365 PMCID: PMC6835950 DOI: 10.3390/insects10100356] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/15/2022]
Abstract
The western honey bee remains the most important pollinator for agricultural crops. Disease and stressors threaten honey bee populations and productivity during winter- and summertime, creating costs for beekeepers and negative impacts on agriculture. To combat diseases and improve overall bee health, researchers are constantly developing honey bee medicines using the tools of microbiology, molecular biology and chemistry. Below, we present a manifesto alongside standardized protocols that outline the development and a systematic approach to test natural products as ‘bee medicines’. These will be accomplished in both artificial rearing conditions and in colonies situated in the field. Output will be scored by gene expression data of host immunity, bee survivorship, reduction in pathogen titers, and more subjective merits of the compound in question. Natural products, some of which are already encountered by bees in the form of plant resins and nectar compounds, provide promising low-cost candidates for safe prophylaxis or treatment of bee diseases.
Collapse
|
25
|
Ayapana triplinervis Essential Oil and Its Main Component Thymohydroquinone Dimethyl Ether Inhibit Zika Virus at Doses Devoid of Toxicity in Zebrafish. Molecules 2019; 24:molecules24193447. [PMID: 31547527 PMCID: PMC6804133 DOI: 10.3390/molecules24193447] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 01/17/2023] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne virus of medical concern. ZIKV infection may represent a serious disease, causing neonatal microcephaly and neurological disorders. Nowadays, there is no approved antiviral against ZIKV. Several indigenous or endemic medicinal plants from Mascarene archipelago in Indian Ocean have been found able to inhibit ZIKV infection. The purpose of our study was to determine whether essential oil (EO) from Reunion Island medicinal plant Ayapana triplinervis, whose thymohydroquinone dimethyl ether (THQ) is the main component has the potential to prevent ZIKV infection in human cells. Virological assays were performed on human epithelial A549 cells infected with either GFP reporter ZIKV or epidemic viral strain. Zebrafish assay was employed to evaluate the acute toxicity of THQ in vivo. We showed that both EO and THQ inhibit ZIKV infection in human cells with IC50 values of 38 and 45 µg/mL, respectively. At the noncytotoxic concentrations, EO and THQ reduced virus progeny production by 3-log. Time-of-drug-addition assays revealed that THQ could act as viral entry inhibitor. At the antiviral effective concentration, THQ injection in zebrafish does not lead to any signs of stress and does not impact fish survival, demonstrating the absence of acute toxicity for THQ. From our data, we propose that THQ is a new potent antiviral phytocompound against ZIKV, supporting the potential use of medicinal plants from Reunion Island as a source of natural and safe antiviral substances against medically important mosquito-borne viruses.
Collapse
|
26
|
Chesnut M, Muñoz LS, Harris G, Freeman D, Gama L, Pardo CA, Pamies D. In vitro and in silico Models to Study Mosquito-Borne Flavivirus Neuropathogenesis, Prevention, and Treatment. Front Cell Infect Microbiol 2019; 9:223. [PMID: 31338335 PMCID: PMC6629778 DOI: 10.3389/fcimb.2019.00223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/11/2019] [Indexed: 01/07/2023] Open
Abstract
Mosquito-borne flaviviruses can cause disease in the nervous system, resulting in a significant burden of morbidity and mortality. Disease models are necessary to understand neuropathogenesis and identify potential therapeutics and vaccines. Non-human primates have been used extensively but present major challenges. Advances have also been made toward the development of humanized mouse models, but these models still do not fully represent human pathophysiology. Recent developments in stem cell technology and cell culture techniques have allowed the development of more physiologically relevant human cell-based models. In silico modeling has also allowed researchers to identify and predict transmission patterns and discover potential vaccine and therapeutic candidates. This review summarizes the research on in vitro and in silico models used to study three mosquito-borne flaviviruses that cause neurological disease in humans: West Nile, Dengue, and Zika. We also propose a roadmap for 21st century research on mosquito-borne flavivirus neuropathogenesis, prevention, and treatment.
Collapse
Affiliation(s)
- Megan Chesnut
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Laura S. Muñoz
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Neuroviruses Emerging in the Americas Study, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Georgina Harris
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Dana Freeman
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Carlos A. Pardo
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Neuroviruses Emerging in the Americas Study, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David Pamies
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
27
|
Volatile Secondary Metabolites with Potent Antidiabetic Activity from the Roots of Prangos pabularia Lindl.—Computational and Experimental Investigations. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9112362] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
(1) Background: Almost 500 million people worldwide are suffering from diabetes. Since ancient times, humans have used medicinal plants for the treatment of diabetes. Medicinal plants continue to serve as natural sources for the discovery of antidiabetic compounds. Prangos pabularia Lindl. is a widely distributed herb with large reserves in Tajikistan. Its roots and fruits have been used in Tajik traditional medicine. To our best knowledge, there are no previously published reports concerning the antidiabetic activity and the chemical composition of the essential oil obtained from roots of P. pabularia. (2) Methods: The volatile secondary metabolites were obtained by hydrodistillation from the underground parts of P. pabularia growing wild in Tajikistan and were analyzed by gas chromatography (GC) and gas chromatography-mass spectrometry (GC-MS). Protein tyrosine phosphatase 1B (PTP-1B) inhibition assay and molecular docking analysis were carried out to evaluate the potential antidiabetic activity of the P. pabularia essential oil. (3) Results: The main constituents of the volatile oil of P. pabularia were 5-pentylcyclohexa-1,3-diene (44.6%), menthone (12.6%), 1-tridecyne (10.9%), and osthole (6.0%). PTP-1B inhibition assay of the essential oil and osthole resulted in significant inhibitory activity with an IC50 value of 0.06 ± 0.01 and 0.93 ± 0.1 μg/mL. Molecular docking analysis suggests volatile compounds such as osthole inhibit PTP-1B, and the results are also in agreement with experimental investigations. (4) Conclusions: Volatile secondary metabolites and the pure isolated compound (osthole) from the roots of P. pabularia exhibited potent antidiabetic activity, twenty-five and nearly two times more than the positive control (3-(3,5-dibromo-4-hydroxybenzoyl)-2-ethylbenzofuran-6-sulfonic acid-(4-(thiazol-2-ylsulfamyl)-phenyl)-amide)) with an IC50 value of 1.46 ± 0.4 μg/mL, respectively.
Collapse
|
28
|
Doratoxylon apetalum, an Indigenous Medicinal Plant from Mascarene Islands, Is a Potent Inhibitor of Zika and Dengue Virus Infection in Human Cells. Int J Mol Sci 2019; 20:ijms20102382. [PMID: 31091703 PMCID: PMC6567149 DOI: 10.3390/ijms20102382] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 02/06/2023] Open
Abstract
Zika virus (ZIKV) and Dengue virus (DENV) are mosquito-borne viruses of the Flavivirus genus that could cause congenital microcephaly and hemorrhage, respectively, in humans, and thus present a risk to global public health. A preventive vaccine against ZIKV remains unavailable, and no specific antiviral drugs against ZIKV and DENV are licensed. Medicinal plants may be a source of natural antiviral drugs which mostly target viral entry. In this study, we evaluate the antiviral activity of Doratoxylum apetalum, an indigenous medicinal plant from the Mascarene Islands, against ZIKV and DENV infection. Our data indicated that D. apetalum exhibited potent antiviral activity against a contemporary epidemic strain of ZIKV and clinical isolates of four DENV serotypes at non-cytotoxic concentrations in human cells. Time-of-drug-addition assays revealed that D. apetalum extract acts on ZIKV entry by preventing the internalisation of virus particles into the host cells. Our data suggest that D. apetalum-mediated ZIKV inhibition relates to virus particle inactivation. We suggest that D. apetalum could be a promising natural source for the development of potential antivirals against medically important flaviviruses.
Collapse
|
29
|
The Polyphenol-Rich Extract from Psiloxylon mauritianum, an Endemic Medicinal Plant from Reunion Island, Inhibits the Early Stages of Dengue and Zika Virus Infection. Int J Mol Sci 2019; 20:ijms20081860. [PMID: 30991717 PMCID: PMC6515236 DOI: 10.3390/ijms20081860] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 02/06/2023] Open
Abstract
The recent emergence and re-emergence of viral infections transmitted by vectors, such as the Zika virus (ZIKV) and Dengue virus (DENV), is a cause for international concern. These highly pathogenic arboviruses represent a serious health burden in tropical and subtropical areas of the world. Despite the high morbidity and mortality associated with these viral infections, antiviral therapies are missing. Medicinal plants have been widely used to treat various infectious diseases since millenaries. Several compounds extracted from plants exhibit potent effects against viruses in vitro, calling for further investigations regarding their efficacy as antiviral drugs. Here, we demonstrate that an extract from Psiloxylon mauritianum, an endemic medicinal plant from Reunion Island, inhibits the infection of ZIKV in vitro without exhibiting cytotoxic effects. The extract was active against different ZIKV African and Asian strains, including an epidemic one. Time-of-drug-addition assays revealed that the P. mauritianum extract interfered with the attachment of the viral particles to the host cells. Importantly, the P. mauritianum extract was also able to prevent the infection of human cells by four dengue virus serotypes. Due to its potency and ability to target ZIKV and DENV particles, P. mauritianum may be of value for identifying and characterizing antiviral compounds to fight medically-important flaviviruses.
Collapse
|
30
|
Discovery and Computational Analyses of Novel Small Molecule Zika Virus Inhibitors. Molecules 2019; 24:molecules24081465. [PMID: 31013906 PMCID: PMC6514826 DOI: 10.3390/molecules24081465] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/04/2019] [Accepted: 04/12/2019] [Indexed: 01/19/2023] Open
Abstract
Zika virus (ZIKV), one of the flaviviruses, has attracted worldwide attention since its large epidemics around Brazil. Association of ZIKV infection with microcephaly and neurological problems such as Guillain–Barré syndrome has prompted intensive pathological investigations. However, there is still a long way to go on the discovery of effective anti-ZIKV therapeutics. In this study, an in silico screening of the National Cancer Institute (NCI) diversity set based on ZIKV NS3 helicase was performed using a molecular docking approach. Selected compounds with drug-like properties were subjected to cell-based antiviral assays resulting in the identification of two novel lead compounds (named Compounds 1 and 2). They inhibited ZIKV infection with IC50 values at the micro-molar level (8.5 μM and 15.2 μM, respectively). Binding mode analysis, absolute binding free energy calculation, and structure–activity relationship studies of these two compounds revealed their possible interactions with ZIKV NS3 helicase, suggesting a mechanistic basis for further optimization. These two novel small molecules may represent new leads for the development of inhibitory drugs against ZIKV.
Collapse
|
31
|
Zou J, Shi PY. Strategies for Zika drug discovery. Curr Opin Virol 2019; 35:19-26. [PMID: 30852345 DOI: 10.1016/j.coviro.2019.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 12/15/2022]
Abstract
Zika virus (ZIKV) can cause devastating congenital syndrome in fetuses from pregnant women and autoimmune disorder Guillain-Barré syndrome in adults. No clinically approved vaccine or drug is currently available for ZIKV. This unmet medical need has motivated a global effort to develop countermeasures. Several promising ZIKV vaccine candidates have already entered clinical trials. In contrast, antiviral development of ZIKV is lagging behind. Here, we review the overall strategies for ZIKV drug discovery, including (i) repurposing of clinically approved drugs, (ii) viral replication-based phenotypic screening for inhibitors, and (iii) targeted drug discovery of viral proteins. Along with vaccines, the development of antiviral treatment will provide a complementary means to control ZIKV infections.
Collapse
Affiliation(s)
- Jing Zou
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA; Department of Phamarcology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
32
|
Gorshkov K, Shiryaev SA, Fertel S, Lin YW, Huang CT, Pinto A, Farhy C, Strongin AY, Zheng W, Terskikh AV. Zika Virus: Origins, Pathological Action, and Treatment Strategies. Front Microbiol 2019; 9:3252. [PMID: 30666246 PMCID: PMC6330993 DOI: 10.3389/fmicb.2018.03252] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/14/2018] [Indexed: 01/05/2023] Open
Abstract
The Zika virus (ZIKV) global epidemic prompted the World Health Organization to declare it a 2016 Public Health Emergency of International Concern. The overwhelming experience over the past several years teaches us that ZIKV and the associated neurological complications represent a long-term world-wide challenge to public health. Although the number of ZIKV cases in the Western Hemisphere has dropped since 2016, the need for basic research and anti-ZIKV drug development remains strong. Re-emerging viruses like ZIKV are an ever-present threat in the 21st century where fast transcontinental travel lends itself to viral epidemics. Here, we first present the origin story for ZIKV and review the rapid progress researchers have made toward understanding of the ZIKV pathology and in the design, re-purposing, and testing–particularly in vivo–drug candidates for ZIKV prophylaxis and therapy ZIKV. Quite remarkably, a short, but intensive, drug-repurposing effort has already resulted in several readily available FDA-approved drugs that are capable of effectively combating the virus in infected adult mouse models and, most importantly, in both preventing maternal-fetal transmission and severe microcephaly in newborns in pregnant mouse models.
Collapse
Affiliation(s)
- Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Sergey A Shiryaev
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Sophie Fertel
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Yi-Wen Lin
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Chun-Teng Huang
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Antonella Pinto
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Chen Farhy
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Alex Y Strongin
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Alexey V Terskikh
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| |
Collapse
|
33
|
Mottin M, Borba JVVB, Melo-Filho CC, Neves BJ, Muratov E, Torres PHM, Braga RC, Perryman A, Ekins S, Andrade CH. Computational drug discovery for the Zika virus. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000001002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
| | | | | | - Bruno Junior Neves
- Federal University of Goiás, Brazil; University Center of Anápolis, Brazil
| | - Eugene Muratov
- University of North Carolin, USA; Odessa National Polytechnic University, Ukraine
| | | | | | | | | | | |
Collapse
|
34
|
Alves MP, Vielle NJ, Thiel V, Pfaender S. Research Models and Tools for the Identification of Antivirals and Therapeutics against Zika Virus Infection. Viruses 2018; 10:v10110593. [PMID: 30380760 PMCID: PMC6265910 DOI: 10.3390/v10110593] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/24/2018] [Accepted: 10/26/2018] [Indexed: 12/13/2022] Open
Abstract
Zika virus recently re-emerged and caused global outbreaks mainly in Central Africa, Southeast Asia, the Pacific Islands and in Central and South America. Even though there is a declining trend, the virus continues to spread throughout different geographical regions of the world. Since its re-emergence in 2015, massive advances have been made regarding our understanding of clinical manifestations, epidemiology, genetic diversity, genomic structure and potential therapeutic intervention strategies. Nevertheless, treatment remains a challenge as there is no licensed effective therapy available. This review focuses on the recent advances regarding research models, as well as available experimental tools that can be used for the identification and characterization of potential antiviral targets and therapeutic intervention strategies.
Collapse
Affiliation(s)
- Marco P Alves
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| | - Nathalie J Vielle
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland.
| | - Volker Thiel
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| | - Stephanie Pfaender
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| |
Collapse
|
35
|
Sinigaglia A, Riccetti S, Trevisan M, Barzon L. In silico approaches to Zika virus drug discovery. Expert Opin Drug Discov 2018; 13:825-835. [PMID: 30160181 DOI: 10.1080/17460441.2018.1515909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION After the WHO declared Zika virus (ZIKV) as a public health emergency of international concern, intense research for the development of vaccines and drugs has been undertaken, leading to the development of several candidates. Areas covered: This review discusses the developments achieved so far by computational methods in the discovery of candidate compounds targeting ZIKV proteins, i.e. the envelope and capsid structural proteins, the NS3 helicase/protease, and the NS5 methyltransferase/RNA-dependent RNA polymerase. Expert opinion: Research for effective drugs against ZIKV is still in a very early discovery phase. Notwithstanding the intense efforts for the development of new drugs and the identification of several promising candidates by using different approaches, including computational methods, so far only a few candidates have been experimentally tested. An important caveat of anti-flavivirus drug development is represented by the difficult of reproducing the in vivo microenvironment of the replication complex, which may lead to discrepancies between in vitro results and experimental evaluation in vivo. Moreover, anti-ZIKV drugs have the additional requirement of an excellent safety profile in pregnancy and ability to diffuse to different tissues, including the central nervous system, the testis, and the placenta.
Collapse
Affiliation(s)
| | - Silvia Riccetti
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| | - Marta Trevisan
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| | - Luisa Barzon
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| |
Collapse
|
36
|
Arora N, Banerjee AK, Narasu ML. Zika outbreak aftermath: status, progress, concerns and new insights. Future Virol 2018. [DOI: 10.2217/fvl-2018-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Zika, a neurotrophic virus belonging to Flaviviridae family of viruses and transmitted by vector mosquitoes of Aedes species, took the world by storm during its recent outbreak. Its spread to newer territories, unprecedented pace of transmission, lack of existing therapeutic agents and vaccines and an empty drug pipeline raised an alarm. Uncertainty about full spectrum of diseases and its long-term consequences, newly discovered modes of transmission and controversies over vector status of mosquito species like Culex quinquefasciatus led to layers of complexity and presented new hurdles and challenges in Zika virus research. This review summarizes the progress and updates of efforts, concerns, financial burden and available resources in light of newly acquired knowledge in Zika virus research.
Collapse
Affiliation(s)
- Neelima Arora
- Centre for Biotechnology, Institute of Science & Technology (Autonomous), Jawaharlal Nehru Technological University-Hyderabad, Kukatpally, Hyderabad 500085, Telangana, India
| | - Amit K Banerjee
- Biology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
| | - Mangamoori L Narasu
- Centre for Biotechnology, Institute of Science & Technology (Autonomous), Jawaharlal Nehru Technological University-Hyderabad, Kukatpally, Hyderabad 500085, Telangana, India
| |
Collapse
|
37
|
Clain E, Sinigaglia L, Koishi AC, Gorgette O, Gadea G, Viranaicken W, Krejbich-Trotot P, Mavingui P, Desprès P, Nunes Duarte Dos Santos C, Guiraud P, Jouvenet N, El Kalamouni C. Extract from Aphloia theiformis, an edible indigenous plant from Reunion Island, impairs Zika virus attachment to the host cell surface. Sci Rep 2018; 8:10856. [PMID: 30022045 PMCID: PMC6052117 DOI: 10.1038/s41598-018-29183-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/06/2018] [Indexed: 01/05/2023] Open
Abstract
The mosquito-borne Zika virus (ZIKV) belongs to the flavivirus genus of the Flaviviridae family. Contemporary epidemic strains of ZIKV are associated with congenital malformations in infants, including microcephaly, as well as Guillain-Barré syndrome in adults. A risk of human-to-human transmission of ZIKV is also well documented. A worldwide research effort has been undertaken to identify safe and effective strategies to prevent or treat ZIKV infection. We show here that extract from Aphloia theiformis, an edible endemic plant from Indian Ocean islands, exerts a potent antiviral effect against ZIKV strains of African and Asian lineages, including epidemic strains. The antiviral effect of A. theiformis extract was extended to clinical isolates of dengue virus (DENV) of the four serotypes in human hepatocytes. A. theiformis inhibited virus entry in host cells by acting directly on viral particles, thus impairing their attachment to the cell surface. Electron microscopic observations revealed that organization of ZIKV particles was severely affected by A. theiformis. We propose a model of antiviral action for A. theiformis against flaviviruses that highlights the potential of medicinal plants as promising sources of naturally-derived antiviral compounds to prevent ZIKV and DENV infections.
Collapse
Affiliation(s)
- Elodie Clain
- Université de La Réunion, UM134 Processus Infectieux Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490, Sainte, Clotilde, France
| | - Laura Sinigaglia
- UMR CNRS 3569, Viral Genomics and Vaccination Unit, Pasteur Institute, 75724, Paris, France
| | - Andrea Cristine Koishi
- Laboratorio de Virologia Molecular, Instituto Carlos Chagas, ICC/FIOCRUZ/PR, Curitiba, Parana, Brazil
| | - Olivier Gorgette
- Ultrastructural BioImaging (UTechsUBI), Pasteur Institute, 75724, Paris, France
| | - Gilles Gadea
- Université de La Réunion, UM134 Processus Infectieux Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490, Sainte, Clotilde, France
| | - Wildriss Viranaicken
- Université de La Réunion, UM134 Processus Infectieux Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490, Sainte, Clotilde, France
| | - Pascale Krejbich-Trotot
- Université de La Réunion, UM134 Processus Infectieux Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490, Sainte, Clotilde, France
| | - Patrick Mavingui
- Université de La Réunion, UM134 Processus Infectieux Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490, Sainte, Clotilde, France
| | - Philippe Desprès
- Université de La Réunion, UM134 Processus Infectieux Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490, Sainte, Clotilde, France
| | | | - Pascale Guiraud
- Université de La Réunion, UM134 Processus Infectieux Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490, Sainte, Clotilde, France
| | - Nolwenn Jouvenet
- UMR CNRS 3569, Viral Genomics and Vaccination Unit, Pasteur Institute, 75724, Paris, France
| | - Chaker El Kalamouni
- Université de La Réunion, UM134 Processus Infectieux Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490, Sainte, Clotilde, France.
| |
Collapse
|
38
|
Robinson CL, Chong ACN, Ashbrook AW, Jeng G, Jin J, Chen H, Tang EI, Martin LA, Kim RS, Kenyon RM, Do E, Luna JM, Saeed M, Zeltser L, Ralph H, Dudley VL, Goldstein M, Rice CM, Cheng CY, Seandel M, Chen S. Male germ cells support long-term propagation of Zika virus. Nat Commun 2018; 9:2090. [PMID: 29844387 PMCID: PMC5974187 DOI: 10.1038/s41467-018-04444-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/26/2018] [Indexed: 01/22/2023] Open
Abstract
Evidence of male-to-female sexual transmission of Zika virus (ZIKV) and viral RNA in semen and sperm months after infection supports a potential role for testicular cells in ZIKV propagation. Here, we demonstrate that germ cells (GCs) are most susceptible to ZIKV. We found that only GCs infected by ZIKV, but not those infected by dengue virus and yellow fever virus, produce high levels of infectious virus. This observation coincides with decreased expression of interferon-stimulated gene Ifi44l in ZIKV-infected GCs, and overexpression of Ifi44l results in reduced ZIKV production. Using primary human testicular tissue, we demonstrate that human GCs are also permissive for ZIKV infection and production. Finally, we identified berberine chloride as a potent inhibitor of ZIKV infection in both murine and human testes. Together, these studies identify a potential cellular source for propagation of ZIKV in testes and a candidate drug for preventing sexual transmission of ZIKV.
Collapse
Affiliation(s)
- Christopher L Robinson
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Angie C N Chong
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Alison W Ashbrook
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY, 10065, USA
| | - Ginnie Jeng
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Julia Jin
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Haiqi Chen
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Laura A Martin
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Rosa S Kim
- Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Reyn M Kenyon
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Eileen Do
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Joseph M Luna
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY, 10065, USA
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY, 10065, USA
| | - Lori Zeltser
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, 10032, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Harold Ralph
- Weill Cornell Medical College-Microscopy and Image Analysis Core Facility, 1300 York Avenue, New York, NY, 10065, USA
| | - Vanessa L Dudley
- Institute of Reproductive Medicine at Weill Cornell Medicine, Weill Cornell Medicine-New York Presbyterian Hospital, New York, NY, 10065, USA
| | - Marc Goldstein
- Department of Urology and Institute for Reproductive Medicine, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY, 10065, USA
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY, 10065, USA.
| | - Marco Seandel
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA. .,Department of Biochemistry, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
39
|
Frangulosid as a novel hepatitis B virus DNA polymerase inhibitor: a virtual screening study. In Silico Pharmacol 2018; 6:10. [PMID: 30607323 DOI: 10.1007/s40203-018-0047-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/17/2018] [Indexed: 12/17/2022] Open
Abstract
Hepatitis B virus (HBV) infects more than 400 million humans Worldwide. Currently, development of new anti-HBV agents is focused on inhibiting of HBV DNA polymerase activity. The natural components of medicinal plant have a broad spectrum of biological activities with therapeutic properties which can be exploited in various steps of drug discovery. Currently, in silico analyses have been introduced as alternative or supplements methods for drug discovery. This study was planned to in silico screening novel HBV DNA polymerase inhibitor(s) from R. palmatum, R. coreanus and S. officinalis. For this purpose, a set of dominant phytochemicals from mentioned plants were retrieved from PubChem database and primary screening was performed with molecular docking method using iGemdock 2.1 software. SwissADME and MedChem Designer 3.0 were used to calculate the drug-likeness parameters of the ligands. Furthermore, the genotoxicity of the studied ligands was predicted using Toxtree 2.6.6 software. Final analysis of screened compounds was done using Autodock 4 software. Result confirmed that Frangulosid and Lindleyin acid have most and least efficacy in HBV DNA polymerase inhibition with the inhibition constant of 2.97 and 53.83 µM, respectively. Results also showed that, the amino acids, involved in interaction, were different for each compound. In this regards, results revealed that the main amino acids residues of the receptor, involved in interaction with Quercetin-3-glucuronide, Frangulosid and Lindleyin separately, located in 420-424, 606-615 and 512-542 spectra, respectively. In conclusion, Frangulosid can be considered as a good candidate for more investigation of its anti-HBV activity.
Collapse
|
40
|
da Silva S, Oliveira Silva Martins D, Jardim ACG. A Review of the Ongoing Research on Zika Virus Treatment. Viruses 2018; 10:E255. [PMID: 29758005 PMCID: PMC5977248 DOI: 10.3390/v10050255] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 12/17/2022] Open
Abstract
The Zika fever is an arboviral disease resulting from the infection with Zika virus (ZIKV). The virus is transmitted to humans by the bite of Aedes mosquitos, mainly Aedes aegypti and Aedes albopictus. ZIKV has been detected for decades in African and Asian regions and, since 2007, has spread to other continents; among them, infections are most reported in the Americas. This can be explained by the presence of vectors in highly populated and tropical regions where people are susceptible to contamination. ZIKV has been considered by the World Health Organization a serious public health problem because of the increasing number of cases of congenital malformation and neurological disorders related to its infection, such as microcephaly, Guillain⁻Barré syndrome, meningoencephalitis, and myelitis. There is no vaccine or specific antiviral against ZIKV. The infection is best prevented by avoiding mosquito bite, and the treatment of infected patients is palliative. In this context, the search for efficient antivirals is necessary but remains challenging. Here, we aim to review the molecules that have been described to interfere with ZIKV life cycle and discuss their potential use in ZIKV therapy.
Collapse
Affiliation(s)
- Suely da Silva
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG 38405-302, Brazil.
- Genomics Study Laboratory, São Paulo State University, IBILCE, São José do Rio Preto, SP 15054-000, Brazil.
| | - Daniel Oliveira Silva Martins
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG 38405-302, Brazil.
- Genomics Study Laboratory, São Paulo State University, IBILCE, São José do Rio Preto, SP 15054-000, Brazil.
| | - Ana Carolina Gomes Jardim
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG 38405-302, Brazil.
- Genomics Study Laboratory, São Paulo State University, IBILCE, São José do Rio Preto, SP 15054-000, Brazil.
| |
Collapse
|
41
|
Devillers J. Repurposing drugs for use against Zika virus infection. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2018; 29:103-115. [PMID: 29299939 DOI: 10.1080/1062936x.2017.1411642] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/25/2017] [Indexed: 06/07/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus for which there are no vaccines or specific therapeutics. To find drugs active on the virus is a complex, expensive and time-consuming process. The prospect of drug repurposing, which consists of finding new indications for existing drugs, is an interesting alternative to expedite drug development for specific diseases. In theory, drug repurposing is also able to respond much more rapidly to a crisis than a classical drug discovery process. Consequently, the methodology is attractive for vector-borne diseases that can emerge or re-emerge worldwide with the risk to become pandemic quickly. Different drugs, showing various structures, have been repurposed to be used against ZIKV infection. They are reviewed in this study and the conditions for their potential use in practice are discussed.
Collapse
|
42
|
Discovery of potential Zika virus RNA polymerase inhibitors by docking-based virtual screening. Comput Biol Chem 2017; 71:144-151. [DOI: 10.1016/j.compbiolchem.2017.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/14/2017] [Accepted: 10/18/2017] [Indexed: 11/20/2022]
|
43
|
de Carvalho OV, Félix DM, de Mendonça LR, de Araújo CMCS, de Oliveira Franca RF, Cordeiro MT, Silva Júnior A, Pena LJ. The thiopurine nucleoside analogue 6-methylmercaptopurine riboside (6MMPr) effectively blocks Zika virus replication. Int J Antimicrob Agents 2017; 50:718-725. [PMID: 28803932 DOI: 10.1016/j.ijantimicag.2017.08.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 07/05/2017] [Accepted: 08/05/2017] [Indexed: 10/19/2022]
Abstract
Since the emergence of Zika virus (ZIKV) in Brazil in 2015, 48 countries and territories in the Americas have confirmed autochthonous cases of disease caused by the virus. ZIKV-associated neurological manifestations and congenital defects make the development of safe and effective antivirals against ZIKV of utmost importance. Here we evaluated the antiviral activity of 6-methylmercaptopurine riboside (6MMPr), a thiopurine nucleoside analogue derived from the prodrug azathioprine, against the epidemic ZIKV strain circulating in Brazil. In all of the assays, an epithelial (Vero) and a human neuronal (SH-SY5Y) cell line were used to evaluate the cytotoxicity and effective concentrations of 6MMPr against ZIKV. Levels of ZIKV-RNA, viral infectious titre and the percentage of infected cells in the presence or absence of 6MMPr were used to determine antiviral efficacy. 6MMPr decreased ZIKV production by >99% in both cell lines in a dose- and time-dependent manner. Interestingly, 6MMPr was 1.6 times less toxic to SH-SY5Y cells compared with Vero cells, presenting a 50% cytotoxic concentrations (CC50) of 460.3 µM and 291 µM, respectively. The selectivity index of 6MMPr for Vero and SH-SY5Y cells was 11.9 and 22.7, respectively, highlighting the safety profile of the drug to neuronal cells. Taken together, these results identify, for the first time, the thiopurine nucleoside analogue 6MMPr as a promising antiviral candidate against ZIKV that warrants further in vivo evaluation.
Collapse
Affiliation(s)
- Otavio Valério de Carvalho
- Department of Virology, Oswaldo Cruz Foundation (Fiocruz), Avenida Professor Moraes Rego, Recife, Pernambuco, Brazil
| | - Daniele Mendes Félix
- Department of Virology, Oswaldo Cruz Foundation (Fiocruz), Avenida Professor Moraes Rego, Recife, Pernambuco, Brazil
| | - Leila Rodrigues de Mendonça
- Department of Virology, Oswaldo Cruz Foundation (Fiocruz), Avenida Professor Moraes Rego, Recife, Pernambuco, Brazil
| | | | | | - Marli Tenório Cordeiro
- Department of Virology, Oswaldo Cruz Foundation (Fiocruz), Avenida Professor Moraes Rego, Recife, Pernambuco, Brazil
| | - Abelardo Silva Júnior
- Department of Veterinary Medicine, Federal University of Viçosa (UFV), Av. PH Rolfs, s/n, Viçosa, Minas Gerais 36570-900, Brazil.
| | - Lindomar José Pena
- Department of Virology, Oswaldo Cruz Foundation (Fiocruz), Avenida Professor Moraes Rego, Recife, Pernambuco, Brazil.
| |
Collapse
|
44
|
Onawole AT, Sulaiman KO, Adegoke RO, Kolapo TU. Identification of potential inhibitors against the Zika virus using consensus scoring. J Mol Graph Model 2017; 73:54-61. [PMID: 28236744 DOI: 10.1016/j.jmgm.2017.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 01/23/2017] [Indexed: 10/20/2022]
Abstract
The Zika virus (ZIKV) is a life threatening pathogen of zoonotic importance with prevalence in some parts of Africa and America. Unfortunately, there is yet to be a single approved vaccine or antiviral drug to treat the diseases and deformations being caused by the Zika virus infection. In this study, about 36 million compounds from MCULE database were virtually screened against a real matured ZIKV protein using a consensus scoring method to get improved hit rates. The consensus scoring method combined the result from the 25 top ranked molecules from both MCULE and Drug Score eXtended (DSX) docking programs which led to the selection of two hit compounds. The inhibition constant (Ki) values of 0.08 and 0.30μm were obtained for the two selected compounds MCULE-8830369631-0-1 and MCULE-9236850811-0-1 respectively, to remark them as hit compounds. The molecular interactions of the two selected hit compounds with the amino acids (ALA 48, ILE 49, ILE 468 and LEU 472) present in the ZIKV protein indicated that they both have similar binding modes. The result of the computationally predicted physicochemical properties including ADMET for the selected compounds showed their great potential in becoming lead compounds upon optimization and thus could be used in treating the Zika virus diseases.
Collapse
Affiliation(s)
- Abdulmujeeb T Onawole
- Department of Chemistry, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia
| | - Kazeem O Sulaiman
- Department of Chemistry, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia.
| | - Rukayat O Adegoke
- Department of Pure and Applied Biology, Ladoke Akintola University of Technology, P.M.B. 4000 Ogbomoso, Nigeria
| | - Temitope U Kolapo
- Department of Veterinary Parasitology and Entomology, Faculty of Veterinary Medicine, University of Ilorin, P.M.B. 1515 Ilorin, Nigeria
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to present what is known about the Zika virus (ZIKV) at the time of writing this review. The viral structure and its phylogeny, as well as the limitations of current available techniques used for diagnosis, are discussed. RECENT FINDINGS Crystallography and cryo-electron microscopy of the whole ZIKV, or a few of its proteins, are confirming its overall antigenic relatedness to other flaviviruses. Sequencing has revealed its dynamic genetic variation and has placed the Western cluster of Zika isolates within the Asian phylogenic tree. Genetic codon mutations, although highly prevalent, do not usually translate into modifications at amino acid or proteomic levels, revealing conserved enzymatic functions that could potentially be addressed therapeutically. Clinical characterization of ZIKV infection is complicated because of symptoms similar to dengue and chikungunya. Diagnosis requires specialized laboratories with costly reagents and highly trained personnel. Although commercial labs are now offering ZIKV diagnostic tests, most of them are not fully tested in comparison with standard molecular techniques standardized at CDC and local health departments. We are still in desperate need of simpler diagnostic tests that better discriminate ZIKV from coendemic arboviruses. SUMMARY The area of better Zika diagnostic assays is a rapidly developing field with the public attention directed to this epidemic. Academic interest in this topic is driving fast disclosure of information in peer-reviewed journals and grey papers via web-based forums. We expect in the near future that new promising strategies for improved Zika diagnostics will translate into preventive and therapeutic tools.
Collapse
|
46
|
Munjal A, Khandia R, Dhama K, Sachan S, Karthik K, Tiwari R, Malik YS, Kumar D, Singh RK, Iqbal HMN, Joshi SK. Advances in Developing Therapies to Combat Zika Virus: Current Knowledge and Future Perspectives. Front Microbiol 2017; 8:1469. [PMID: 28824594 PMCID: PMC5541032 DOI: 10.3389/fmicb.2017.01469] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/20/2017] [Indexed: 02/05/2023] Open
Abstract
Zika virus (ZIKV) remained largely quiescent for nearly six decades after its first appearance in 1947. ZIKV reappeared after 2007, resulting in a declaration of an international "public health emergency" in 2016 by the World Health Organization (WHO). Until this time, ZIKV was considered to induce only mild illness, but it has now been established as the cause of severe clinical manifestations, including fetal anomalies, neurological problems, and autoimmune disorders. Infection during pregnancy can cause congenital brain abnormalities, including microcephaly and neurological degeneration, and in other cases, Guillain-Barré syndrome, making infections with ZIKV a substantial public health concern. Genomic and molecular investigations are underway to investigate ZIKV pathology and its recent enhanced pathogenicity, as well as to design safe and potent vaccines, drugs, and therapeutics. This review describes progress in the design and development of various anti-ZIKV therapeutics, including drugs targeting virus entry into cells and the helicase protein, nucleosides, inhibitors of NS3 protein, small molecules, methyltransferase inhibitors, interferons, repurposed drugs, drugs designed with the aid of computers, neutralizing antibodies, convalescent serum, antibodies that limit antibody-dependent enhancement, and herbal medicines. Additionally, covalent inhibitors of viral protein expression and anti-Toll-like receptor molecules are discussed. To counter ZIKV-associated disease, we need to make rapid progress in developing novel therapies that work effectually to inhibit ZIKV.
Collapse
Affiliation(s)
- Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah UniversityBhopal, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah UniversityBhopal, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research InstituteBareilly, India
- *Correspondence: Kuldeep Dhama,
| | - Swati Sachan
- Immunology Section, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences UniversityChennai, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan SansthanMathura, India
| | - Yashpal S. Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Raj K. Singh
- ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Hafiz M. N. Iqbal
- School of Engineering and Science, Tecnologico de Monterrey, Campus MonterreyMonterrey, Mexico
| | - Sunil K. Joshi
- Cellular Immunology Lab, Frank Reidy Research Center of Bioelectrics, Old Dominion University, NorfolkVA, United States
| |
Collapse
|
47
|
Liu J, Kanetake S, Wu YH, Tam C, Cheng LW, Land KM, Friedman M. Antiprotozoal Effects of the Tomato Tetrasaccharide Glycoalkaloid Tomatine and the Aglycone Tomatidine on Mucosal Trichomonads. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:8806-8810. [PMID: 27934291 DOI: 10.1021/acs.jafc.6b04030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The present study investigated the inhibitory effects of the commercial tetrasaccharide tomato glycoalkaloid tomatine and the aglycone tomatidine on three mucosal pathogenic protozoa that are reported to infect humans, cattle, and cats, respectively: Trichomonas vaginalis strain G3, Tritrichomonas foetus strain D1, and Tritrichomonas foetus strain C1. A preliminary screen showed that tomatine at 100 μM concentration completely inhibited the growth of all three trichomonads. In contrast, the inhibition of all three pathogens by tomatidine was much lower, suggesting the involvement of the lycotetraose carbohydrate side chain in the mechanism of inhibition. Midpoints of concentration-response sigmoid plots of tomatine on the three strains correspond to IC50 values, the concentration that inhibits 50% of growth of the pathogenic protozoa. The concentration data were used to calculate the IC50 values for G3, D1, and C1 of 7.9, 1.9, and 2.2 μM, respectively. The results show an approximately 4-fold variation from the lowest to the highest value (lowest activity). Although the inhibition by tomatine was not as effective as that of the medicinal drug metronidazole, the relatively low IC50 values for both T. vaginalis and T. foetus indicated tomatine as a possible natural alternative therapeutic for trichomoniasis in humans and food-producing (cattle and pigs) and domestic (cats) animals. Because tomatine has the potential to serve as a new antiprotozoan functional (medical) food, the distribution of this glycoalkaloid in tomatoes and suggestions for further research are discussed.
Collapse
Affiliation(s)
- Jenny Liu
- Department of Biological Sciences, University of the Pacific , Stockton, California 95211, United States
| | - Sierra Kanetake
- Department of Biological Sciences, University of the Pacific , Stockton, California 95211, United States
| | - Yun-Hsuan Wu
- Department of Biological Sciences, University of the Pacific , Stockton, California 95211, United States
| | - Christina Tam
- Foodborne Toxin Detection and Prevention, Agricultural Research Service, United States Department of Agriculture , Albany, California 94556, United States
| | - Luisa W Cheng
- Foodborne Toxin Detection and Prevention, Agricultural Research Service, United States Department of Agriculture , Albany, California 94556, United States
| | - Kirkwood M Land
- Department of Biological Sciences, University of the Pacific , Stockton, California 95211, United States
| | - Mendel Friedman
- Healthy Processed Foods Research, Agricultural Research Service, United States Department of Agriculture , Albany, California 94556, United States
| |
Collapse
|