1
|
Karunakaran K, Salam AAA, Mudgal PP. Exploiting the chikungunya virus capsid protein: a focused target for antiviral therapeutic development. Arch Virol 2025; 170:141. [PMID: 40423856 DOI: 10.1007/s00705-025-06325-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/04/2025] [Indexed: 05/28/2025]
Abstract
Chikungunya disease is spread by the bite of infected Aedes mosquitoes. It is considered a neglected tropical disease that has the potential to cause sporadic epidemics in naive populations. Despite the substantial investment in research, there are no approved antiviral treatments for chikungunya. Several screening approaches have been used to identify potential antiviral molecules that target the whole virus, viral proteins, and viral-host interactions, often in conjunction with computational studies. The genome of chikungunya virus (CHIKV) encodes four nonstructural and five structural proteins. The capsid protein (CP) is a small structural protein with enzymatic activity. Owing to its critical role in different stages of the viral life cycle, the CP can be targeted at multiple stages, thereby impeding viral multiplication. There is evidence suggesting that the CP may be a promising target for drug development, and this has led to the discovery of various inhibitors through diverse in vitro and in silico analyses. Both cell-based and cell-free assays have been widely used to identify and evaluate CHIKV CP inhibitors. Computer-based studies targeting CHIKV proteins, including CP, have identified several lead compounds, which are being further evaluated in various in vitro systems. No review has been published on the CHIKV CP, and papers have focused on drug development and the targeting of viral proteins and associated factors. In this review, we summarize the research that has been conducted on the CHIKV CP, including structural studies, antiviral research, and prospects for the use of the CP as an antiviral target.
Collapse
Affiliation(s)
- Kavitha Karunakaran
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal, India
| | | | - Piya Paul Mudgal
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
2
|
Cao J, Zhang H, Zhang J, Wang J, Li C, Ma J, Ye Z, Zheng Y, Liu H, Xiao G, Dai W, Zhang L. Screening of botanical drugs reveals the potential anti-human adenovirus activity of berberine. Antiviral Res 2025; 237:106105. [PMID: 39956272 DOI: 10.1016/j.antiviral.2025.106105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Human adenovirus (HAdV) is a significant viral pathogen that causes severe acute respiratory infections (SARIs) in children and immunocompromised patients. Currently, no specific treatment options are available for HAdV infections. This study used a green fluorescence protein-based, high-throughput screening (HTS) assay on a botanical drug library containing 3697 botanical compounds to identify agents that could inhibit HAdV. Four compounds with anti-HAdV-C5 activity in the low-micromolar range were identified and inhibited other wild-type HAdVs known to cause SARIs. Among these compounds, 13-methylberberine chloride presented the highest select index values. Berberine is a commercially available natural product-derived isoquinoline alkaloid with multiple pharmacological effects and is widely used in Asian countries. We systematically evaluated the anti-HAdV activity of six berberine-derived compounds in vitro and performed a time-of-drug-addition assay to explore their antiviral modes of action. Mechanistic studies revealed that berberine and its analogs inhibit HAdV replication by downregulating the MAPK signaling pathway, particularly ERK activation, which is crucial for viral replication and progeny production. Our findings suggest that berberine is a promising candidate for the development of anti-HAdV therapies.
Collapse
Affiliation(s)
- Junyuan Cao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China
| | - Hao Zhang
- Hubei Jiangxia Laboratory, Wuhan, 430200, China; HuaWu Biotech Wuhan, 430200, China
| | - Jixiang Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinlin Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chen Li
- Hubei Jiangxia Laboratory, Wuhan, 430200, China; HuaWu Biotech Wuhan, 430200, China
| | - Jin Ma
- Hubei Jiangxia Laboratory, Wuhan, 430200, China; HuaWu Biotech Wuhan, 430200, China
| | - Zhengyu Ye
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yunting Zheng
- Hubei Jiangxia Laboratory, Wuhan, 430200, China; HuaWu Biotech Wuhan, 430200, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China.
| | - Wenhao Dai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China.
| |
Collapse
|
3
|
Nehul S, Rani R, Walia P, Panda PK, Singh S, Chattopadhyay S, Kumar P, Tomar S. Repurposing Efavirenz, the HIV Antiretroviral Drug for Chikungunya Virus Infection. ACS Infect Dis 2025; 11:963-976. [DOI: https:/doi.org/10.1021/acsinfecdis.4c00992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Affiliation(s)
- Sanketkumar Nehul
- Department of Biosciences and Bioengineering
- Indian Institute of Technology
| | - Ruchi Rani
- Department of Biosciences and Bioengineering
- Indian Institute of Technology
| | - Priyanshu Walia
- Department of Biosciences and Bioengineering
- Indian Institute of Technology
| | | | - Sharad Singh
- Institute of Life Sciences (ILS)
- KIIT School of Biotechnology
- KIIT University
| | | | - Pravindra Kumar
- Department of Biosciences and Bioengineering
- Indian Institute of Technology
| | - Shailly Tomar
- Department of Biosciences and Bioengineering
- Indian Institute of Technology
| |
Collapse
|
4
|
Nehul S, Rani R, Walia P, Panda PK, Singh S, Chattopadhyay S, Kumar P, Tomar S. Repurposing Efavirenz, the HIV Antiretroviral Drug for Chikungunya Virus Infection. ACS Infect Dis 2025; 11:963-976. [PMID: 40051136 DOI: 10.1021/acsinfecdis.4c00992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Chikungunya virus (CHIKV) has frequently recurred in recent decades, causing outbreaks worldwide in tropical and subtropical regions. The re-emergence of CHIKV poses a substantial risk to human health, as no efficacious drugs are currently available to curb new outbreaks. Here, the anti-CHIKV activity of efavirenz was investigated by in vitro cell culture-based antiviral assays in different relevant cell lines. Efavirenz is a non-nucleoside reverse transcriptase inhibitor (NNRTI) used for the treatment of acquired immunodeficiency syndrome (AIDS), and it has good oral bioavailability, long half-life, and affordable low cost. This study demonstrated dose-dependent robust anti-CHIKV activity of efavirenz at low micromolar concentration in two different cell lines with 50% effective concentration (EC50) of 1.1 to 1.3 μM. Interestingly, efavirenz also inhibited the replication of Sindbis virus (SINV) at a low micromolar range indicating potential broad anti-alphavirus activity. Time of addition assay, direct transfection of virus replicon RNA, and minus-sense-specific reverse transcription polymerase chain reaction (RT-PCR) elucidated that efavirenz hinders the viral replication at an early stage after the virus entry by inhibiting the viral RNA synthesis. Further, the binding affinity of efavirenz toward purified capsid protein (CP) was observed, suggesting that CP could be one of the antiviral targets for efavirenz in addition to viral or host proteins involved in viral RNA replication. Finally, the in vivo efficacy of efavirenz was assessed in a murine model and a decrease in CHIKV viral load was observed. In summary, the present study underscores the potential of repurposing efavirenz for antiviral therapy against CHIKV to curb future viral outbreaks.
Collapse
Affiliation(s)
- Sanketkumar Nehul
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Ruchi Rani
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Priyanshu Walia
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Prasan Kumar Panda
- Department of medicine, All India Institute of Medical Sciences (AIIMS), Rishikesh 249203, India
| | - Sharad Singh
- Institute of Life Sciences (ILS), Bhubaneswar, Odisha 751023, India
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, Odisha 751024, India
| | | | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| |
Collapse
|
5
|
Khan A, Zakirullah, Wahab S, Hong ST. Advances in antiviral strategies targeting mosquito-borne viruses: cellular, viral, and immune-related approaches. Virol J 2025; 22:26. [PMID: 39905499 PMCID: PMC11792744 DOI: 10.1186/s12985-025-02622-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025] Open
Abstract
Mosquito-borne viruses (MBVs) are a major global health threat, causing significant morbidity and mortality. MBVs belong to several distinct viral families, each with unique characteristics. The primary families include Flaviviridae (e.g., Dengue, Zika, West Nile, Yellow Fever, Japanese Encephalitis), transmitted predominantly by Aedes and Culex mosquitoes; Togaviridae, which consists of the genus Alphavirus (e.g., Chikungunya, Eastern and Western Equine Encephalitis viruses), also transmitted by Aedes and Culex; Bunyaviridae (recently reorganized), containing viruses like Rift Valley Fever and Oropouche virus, transmitted by mosquitoes and sometimes sandflies; and Reoviridae, which includes the genus Orbivirus (e.g., West Nile and Bluetongue viruses), primarily affecting animals and transmitted by mosquitoes and sandflies. Despite extensive research, effective antiviral treatments for MBVs remain scarce, and current therapies mainly provide symptomatic relief and supportive care. This review examines the viral components and cellular and immune factors involved in the life cycle of MBVs. It also highlights recent advances in antiviral strategies targeting host factors such as lipid metabolism, ion channels, and proteasomes, as well as viral targets like NS2B-NS3 proteases and nonstructural proteins. Additionally, it explores immunomodulatory therapies to enhance antiviral responses and emphasizes the potential of drug repurposing, bioinformatics, artificial intelligence, and deep learning in identifying novel antiviral candidates. Continued research is crucial in mitigating MBVs' impact and preventing future outbreaks.
Collapse
Affiliation(s)
- Ayyaz Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, 54907, South Korea
| | - Zakirullah
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shahid Wahab
- Department of Agriculture, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, 54907, South Korea.
| |
Collapse
|
6
|
Qu J, Schinkel M, Chiggiato L, Rosendo Machado S, Overheul GJ, Miesen P, van Rij RP. The Hsf1-sHsp cascade has pan-antiviral activity in mosquito cells. Commun Biol 2025; 8:123. [PMID: 39863754 PMCID: PMC11762766 DOI: 10.1038/s42003-024-07435-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Aedes mosquitoes transmit pathogenic arthropod-borne (arbo) viruses, putting nearly half the world's population at risk. Blocking virus replication in mosquitoes is a promising approach to prevent arbovirus transmission, the development of which requires in-depth knowledge of virus-host interactions and mosquito immunity. By integrating multi-omics data, we find that heat shock factor 1 (Hsf1) regulates eight small heat shock protein (sHsp) genes within one topologically associated domain in the genome of the Aedes aegypti mosquito. This Hsf1-sHsp cascade acts as an early response against chikungunya virus infection and shows pan-antiviral activity against chikungunya, Sindbis, and dengue virus as well as the insect-specific Agua Salud alphavirus in Ae. aegypti cells and against chikungunya virus and O'nyong-nyong virus in Aedes albopictus and Anopheles gambiae cells, respectively. Our comprehensive in vitro data suggest that Hsf1 could serve as a promising target for the development of novel intervention strategies to limit arbovirus transmission by mosquitoes.
Collapse
Affiliation(s)
- Jieqiong Qu
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michelle Schinkel
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lisa Chiggiato
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Samara Rosendo Machado
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gijs J Overheul
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pascal Miesen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
7
|
Dansana J, Purohit P, Panda M, Meher BR. Recent advances in phytocompounds as potential Chikungunya virus non-structural protein 2 protease antagonists: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156359. [PMID: 39756312 DOI: 10.1016/j.phymed.2024.156359] [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: 07/09/2024] [Revised: 12/17/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND The mosquito-borne pathogenic alphavirus known as Chikungunya virus (CHIKV) is becoming a greater hazard to public health, which causes thousands of cases annually in both rural and urban areas of many different nations throughout the world. Finding and creating new leads for the CHIKV virus is crucial because there are currently no effective medications or vaccinations against it. The non-structural protein 2 (nsP2) protease has emerged as a promising target for therapeutic intervention due to its crucial role in viral replication. PURPOSE This systematic review aims to evaluate recent advances in natural products as inhibitors of the CHIKV nsP2 protease, summarizing current research, identifying promising compounds, and highlighting gaps in the existing knowledge. STUDY DESIGN A comprehensive literature search was conducted between January 2006, and June 2024 using databases including PubMed, Scopus, Science Direct, and Google Scholar. Search terms included CHIKV, nsP2 protease, antivirals, natural products, phytochemicals, and inhibitors. Studies were selected based on predefined inclusion and exclusion criteria, focusing on original research articles examining natural products as inhibitors of CHIKV nsP2 protease. METHODS Relevant studies were screened, and data were extracted regarding the source of natural compounds, methods of extraction, chemical structures, mechanisms of action, potency, and efficacy in inhibiting nsP2 protease or CHIKV replication. RESULTS The review included 40 studies, revealing a variety of natural products and their derivatives with inhibitory effects on CHIKV nsP2 protease. Several compounds demonstrated promising inhibitory activity with EC50 values in the micromolar range. Mechanistic studies revealed diverse modes of action, including inhibition of protease activity or interference with viral replication processes. CONCLUSION Natural products have gained attention for their diverse chemical structures and bioactivities, offering a rich source of compounds with antiviral potential. We summarize the current knowledge on natural products derived from various sources including flavonoids, alkaloids, terpenoids, polyphenols, and some derivative compounds that have demonstrated inhibitory effects against CHIKV through different mechanisms of action. Overall, this systematic review underscores the importance of exploring natural products as promising candidates for the development of effective therapeutics against Chikungunya fever, particularly through targeting the nsP2 protease.
Collapse
Affiliation(s)
- Jarmani Dansana
- Computational Biology and Bioinformatics Laboratory, PG Department of Botany, Berhampur University, Berhampur, Odisha, 760007, India
| | - Priyanka Purohit
- Computational Biology and Bioinformatics Laboratory, PG Department of Botany, Berhampur University, Berhampur, Odisha, 760007, India
| | - Madhusmita Panda
- Computational Biology and Bioinformatics Laboratory, PG Department of Botany, Berhampur University, Berhampur, Odisha, 760007, India
| | - Biswa Ranjan Meher
- Computational Biology and Bioinformatics Laboratory, PG Department of Botany, Berhampur University, Berhampur, Odisha, 760007, India.
| |
Collapse
|
8
|
Yamashita A, Kasai H, Maekawa S, Tanaka T, Akaike Y, Ryo A, Enomoto N, Moriishi K. Berberine promotes K 48-linked polyubiquitination of HNF4α, leading to the inhibition of HBV replication. Antiviral Res 2024; 232:106027. [PMID: 39489302 DOI: 10.1016/j.antiviral.2024.106027] [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: 08/07/2024] [Revised: 10/17/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
The current antiviral agents for the treatment of chronic infection with hepatitis B virus (HBV) do not completely remove covalently closed circular DNA (cccDNA) and integrated viral DNA fragments from patients. Berberine is an isoquinoline alkaloid extracted from various plants and has been reported to inhibit the replication of various types of DNA. In this study, we tested the effects of berberine and its derivatives on HBV infection. Berberine inhibited viral core promoter activity at the highest level among the compounds tested and suppressed HBV production and cccDNA synthesis in primary human hepatocytes and HBV-infected HepG2-NTCP cells at an EC50 value of 3.6 μM and a CC50 value of over 240.0 μM. Compared with other viral promoter activities, berberine treatment potently downregulated core promoter activity and reduced protein levels, but not RNA levels, of hepatic nuclear factor 4α (HNF4α), which primarily enhances enhancer II/core promoter activity. Furthermore, berberine treatment enhanced K48-linked, but not K63-linked, polyubiquitination and subsequent proteasome-dependent degradation of HNF4α. These results suggest that berberine enhances the polyubiquitination- and proteasome-dependent degradation of HNF4α and then inhibits HBV replication via the suppression of core promoter activity. The development of antiviral agents based on berberine may contribute to the amelioration of HBV-related disorders, regardless of the presence of residual cccDNA or integrated viral DNA fragments.
Collapse
Affiliation(s)
- Atsuya Yamashita
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Hirotake Kasai
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Shinya Maekawa
- The First Department of Internal Medicine, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Tomohisa Tanaka
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan; Division of Hepatitis Virology, Institute for Genetic Medicine, Hokkaido University, Hokkaido, 060-0808, Japan
| | - Yasunori Akaike
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Akihide Ryo
- Department of Virology III, National Institute for Infectious Diseases, Tokyo, 208-0011, Japan
| | - Nobuyuki Enomoto
- The First Department of Internal Medicine, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Kohji Moriishi
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan; Division of Hepatitis Virology, Institute for Genetic Medicine, Hokkaido University, Hokkaido, 060-0808, Japan; Center for Life Science Research, University of Yamanashi, Yamanashi, 409-3898, Japan.
| |
Collapse
|
9
|
Jiang Z, Merits A, Qin Y, Xing G, Zhang L, Chen J, Wang N, Varjak M, Zhai X, Li D, Song W, Su S. Attenuated Getah virus confers protection against multiple arthritogenic alphaviruses. PLoS Pathog 2024; 20:e1012700. [PMID: 39556619 PMCID: PMC11630583 DOI: 10.1371/journal.ppat.1012700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/10/2024] [Accepted: 10/27/2024] [Indexed: 11/20/2024] Open
Abstract
Alphaviruses are important arthropod-transmitted pathogens of humans and livestock. Getah virus (GETV) is an arthritogenic alphavirus that causes disease in horses and piglets; it also poses a potential threat to humans. A live attenuated vaccine candidate named GETV-3ΔS2-CM1, harbouring a deletion in nonstructural protein 3 and substitutions in the capsid protein, is genetically stable and exhibits robust immunogenicity. It was shown to confer passive protection to piglets born to immunized sows. In mice, a single dose of GETV-3ΔS2-CM1 protected against infection with different strains of GETV, Semliki Forest virus, Ross River virus, o'nyong'nyong virus, chikungunya virus, and Barmah Forest virus. Chimaeras based on the GETV-3ΔS2-CM1 backbone maintained both the attenuated phenotype and high immunogenicity. The safety, efficacy, and ability to induce protection against multiple alphaviruses highlights the potential of GETV-3ΔS2-CM1 and chimaeras using this backbone as promising vaccine candidates. By contributing simultaneously to the wellbeing of animals and humans, our universal next generation vaccine strategy helps to achieve "One Health" goals.
Collapse
Affiliation(s)
- Zhiwen Jiang
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Andres Merits
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Ying Qin
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Gang Xing
- MOA Key Laboratory of Animal Virology, Zhejiang University, Hangzhou, China
| | - Letian Zhang
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jie Chen
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ningning Wang
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Margus Varjak
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Xiaofeng Zhai
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Dongyan Li
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wanjie Song
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shuo Su
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
10
|
Khamaru S, Mukherjee T, Tung KS, Kumar PS, Bandyopadhyay S, Mahish C, Chattopadhyay S, Chattopadhyay S. Chikungunya virus infection inhibits B16 melanoma-induced immunosuppression of T cells and macrophages mediated by interleukin 10. Microb Pathog 2024; 197:107022. [PMID: 39419458 DOI: 10.1016/j.micpath.2024.107022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
Immunosuppression in cancer poses challenges for immunotherapy and highlights the vulnerability of immunocompromised patients to viral infections. This study explored how Chikungunya virus (CHIKV) infection potentially inhibits B16-F10 melanoma-induced immunosuppressive effects on T cells and RAW 264.7 macrophages. We found high expression of CHIKV entry genes in melanoma and other cancers, with B16-F10 cells demonstrating greater susceptibility to CHIKV infection than non-tumorigenic cells. Interestingly, the CHIKV-infected B16-F10 cell culture supernatant (B16-F10-CS) reversed the immunosuppressive effects of uninfected B16-F10-CS on T cells. This reversal was characterised by decreased STAT3 activation and increased MAPK activation in T cells, an effect amplified by interleukin 10 (IL-10) receptor blockade. In RAW 264.7 cells, B16-F10-CS enhanced CHIKV infectivity without triggering activation. However, blocking the IL-10 receptor (IL-10R) in RAW 264.7 reduced CHIKV infection. CHIKV infection and IL-10R blockade synergistically inhibited B16-F10-CS-mediated polarisation of RAW 264.7 cells towards immunosuppressive macrophage. Our findings suggest that CHIKV modulates cancer-induced immunosuppression through IL-10-dependent pathways, providing new insights into viral-cancer interactions. This research may contribute to developing novel antiviral immunotherapies and virotherapies beneficial for cancer patients and immunocompromised individuals.
Collapse
Affiliation(s)
- Somlata Khamaru
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - Tathagata Mukherjee
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India; Institute of Life Sciences, Bhubaneswar, India
| | - Kshyama Subhadarsini Tung
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - P Sanjai Kumar
- Institute of Life Sciences, Bhubaneswar, India; Division of Neonatology and Newborn Nursery, University of Wisconsin, Madison, USA
| | - Saumya Bandyopadhyay
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India; Johns Hopkins University School of Medicine, Department of Biological Chemistry, 725 North Wolfe Street, Baltimore, Maryland, USA
| | - Chandan Mahish
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | | | - Subhasis Chattopadhyay
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India.
| |
Collapse
|
11
|
Altay Benetti A, Thwin MT, Suhaimi A, Liang RST, Ng LFP, Lum FM, Benetti C. Development of Proniosome Gel Formulation for CHIKV Infection. Pharmaceutics 2024; 16:994. [PMID: 39204339 PMCID: PMC11360264 DOI: 10.3390/pharmaceutics16080994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Given the increasing aging population and the rising prevalence of musculoskeletal diseases due to obesity and injury, urgent research is needed to formulate new treatment alternatives, as current options remain inadequate. Viruses can exacerbate arthritis and worsen symptoms in patients with pre-existing osteoarthritis. Over the past decade, the chikungunya virus (CHIKV) has emerged as a significant public health concern, especially in Asia and South America. Exploring natural products, such as berberine, has shown promise due to its anticatabolic, antioxidative, and anti-inflammatory effects. However, berberine's low stability and bioavailability limit its efficacy. We hypothesized that encapsulating berberine into a proniosome gel, known for its ease of preparation and stability, could enhance its bioavailability and efficacy when applied topically, potentially treating CHIKV infection. Our investigation focused on how varying berberine loads and selected excipients in the proniosome gel influenced its physical properties, stability, and skin permeability. We also examined the biological half-life of berberine in plasma upon topical administration in mice to assess the potential for controlled and sustained drug release. Additionally, we analyzed the antioxidant stress activity and cell viability of HaCaT keratinocytes and developed a lipopolysaccharide-stimulated cell culture model to evaluate anti-inflammatory effects using pro-inflammatory cytokines. Overall, the research aims to transform the treatment landscape for arthritis by leveraging berberine's therapeutic potential.
Collapse
Affiliation(s)
- Ayça Altay Benetti
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (M.T.T.); (R.S.T.L.)
| | - Ma Thinzar Thwin
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (M.T.T.); (R.S.T.L.)
| | - Ahmad Suhaimi
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.S.); (L.F.-P.N.)
| | - Ryan Sia Tze Liang
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (M.T.T.); (R.S.T.L.)
| | - Lisa Fong-Poh Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.S.); (L.F.-P.N.)
| | - Fok-Moon Lum
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.S.); (L.F.-P.N.)
| | - Camillo Benetti
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (M.T.T.); (R.S.T.L.)
| |
Collapse
|
12
|
Jiang Y, Han C, Gong H, Chen J, Tang B, Yang M, Qin Q, Wei S. Berberine inhibits SGIV replication by suppressing inflammatory response and oxidative stress. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109522. [PMID: 38548190 DOI: 10.1016/j.fsi.2024.109522] [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: 01/09/2024] [Revised: 02/21/2024] [Accepted: 03/21/2024] [Indexed: 04/09/2024]
Abstract
Singapore grouper iridovirus (SGIV) is one of the major infectious diseases responsible for high mortality and huge economic losses in the grouper aquaculture industry. Berberine (BBR), a naturally occurring plant alkaloid, is a phytochemical having a variety of biological properties, such as antiviral, antioxidant, and anti-inflammatory effects. In this work, we used an in vitro model based on Western blot, ROS fluorescence probe, and real-time quantitative PCR (qRT-PCR) to examine the antiviral qualities of BBR against SGIV. The outcomes demonstrated that varying BBR concentrations could significantly inhibit the replication of SGIV. In addition, BBR greatly inhibited the production of genes associated with pro-inflammatory cytokines in SGIV-infected or SGIV-uninfected GS cells based on qRT-PCR data. Subsequent investigations demonstrated that BBR suppressed the expression of the promoter activity of NF-κB and NF-κB-p65 protein. Additionally, BBR reduced the phosphorylation of ERK 1/2, JNK, and p38. Furthermore, BBR also inhibits SGIV-induced ROS production by upregulating the expression of antioxidant-related genes. In conclusion, BBR is a viable therapy option for SGIV infection due to its antiviral properties.
Collapse
Affiliation(s)
- Yunxiang Jiang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Chengzong Han
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Hannan Gong
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Jiatao Chen
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Biao Tang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Min Yang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, 511457, China
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, 511457, China.
| | - Shina Wei
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, 511457, China.
| |
Collapse
|
13
|
Arman M, Alam S, Maruf RA, Shams Z, Islam MN. Molecular modeling of some commercially available antiviral drugs and their derivatives against SARS-CoV-2 infection. NARRA J 2024; 4:e319. [PMID: 38798846 PMCID: PMC11125382 DOI: 10.52225/narra.v4i1.319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/07/2024] [Indexed: 05/29/2024]
Abstract
Numerous prior studies have identified therapeutic targets that could effectively combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, including the angiotensin-converting enzyme 2 (ACE2) receptor, RNA-dependent RNA polymerase (RdRp), and Main protease (Mpro). In parallel, antiviral compounds like abacavir, acyclovir, adefovir, amantadine, amprenavir, darunavir, didanosine, oseltamivir, penciclovir, and tenofovir are under investigation for their potential in drug repurposing to address this infection. The aim of the study was to determine the effect of modifying the functional groups of the aforementioned antivirals in silico. Using the genetic optimization for ligand docking algorithm on software Maestro (version 11.1), the modified antivirals were docked onto ACE2 receptor, RdRp, and Mpro. Using QuickProp (Maestro v11.1), PASS (prediction of activity spectra for the substances), and altogether with SwissADME, the ADMET (absorption, distribution, metabolism, excretion, and toxicity) of the modified antivirals, as well as their bioavailability and the predicted activity spectra, were determined. Discovery studio software was used to undertake post-docking analysis. Among the 10 antivirals, N(CH3)2 derivative of darunavir, N(CH3)2 derivative of amprenavir and NCH3 derivative of darunavir exhibited best binding affinities with ACE2 receptor (docking scores: -10.333, -9.527 and -9.695 kJ/mol, respectively). Moreover, NCH3 derivative of abacavir (-6.506 kJ/mol), NO2 derivative of didanosine (-6.877 kJ/mol), NCH3 derivative of darunavir (-7.618 kJ/mol) exerted promising affinity to Mpro. In conclusion, the results of the in silico screenings can serve as a useful information for future experimental works.
Collapse
Affiliation(s)
- Mohammad Arman
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Safaet Alam
- Pharmaceutical Sciences Research Division, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Rifat A. Maruf
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Ziaus Shams
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Mohammad N. Islam
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
| |
Collapse
|
14
|
Valipour M, Zakeri Khatir Z, Abdollahi E, Ayati A. Recent Applications of Protoberberines as Privileged Starting Materials for the Development of Novel Broad-Spectrum Antiviral Agents: A Concise Review (2017-2023). ACS Pharmacol Transl Sci 2024; 7:48-71. [PMID: 38230282 PMCID: PMC10789142 DOI: 10.1021/acsptsci.3c00292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024]
Abstract
Berberine is a well-known phytochemical with significant antiviral activity against a wide range of viruses. Due to having a unique backbone consisting of four interconnected rings, it can be used as a platform for the design and development of novel semisynthetic antiviral agents. The question here is whether novel broad-spectrum antiviral drugs with enhanced activity and toxicity potential can be obtained by attempting to modify the structure of this privileged lead compound. The present study aims to review the results of recent studies in which berberine and its close analogues (protoberberine alkaloids) have been used as starting materials for the production of new semisynthetic antiviral structures. For this purpose, relevant studies published in high-quality journals indexed in databases such as Scopus, Web of Science, PubMed, etc. in the time frame of 2017 to 2023 were collected. Our selection criterion in the current review focuses on the studies in which protoberberines were used as starting materials for the production of semisynthetic agents with antiviral activity during the indicated time period. Correspondingly, studies were identified in which semisynthetic derivatives with significant inhibitory activity against a wide range of viruses including human immunodeficiency virus (HIV), enterovirus 71 (EV71), zika virus (ZIKV), influenza A/B, cytomegalovirus (CMV), respiratory syncytial virus (RSV), and coxsackieviruses were designed and synthesized. Our conclusion is that, despite the introduction of diverse semisynthetic derivatives of berberine with improved activity profiles compared to the parent natural leads, sufficient derivatization has not been done yet and more studies are needed.
Collapse
Affiliation(s)
- Mehdi Valipour
- Razi
Drug Research Center, Iran University of
Medical Sciences, Tehran 1449614535, Iran
| | - Zahra Zakeri Khatir
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 6964114483, Iran
- Student
Research Committee, Mazandaran University
of Medical Sciences, Sari 6964114483, Iran
| | - Elaheh Abdollahi
- Department
of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Adileh Ayati
- Department
of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Sciences
Research Center, Tehran University of Medical
Sciences, Tehran 1416753955, Iran
| |
Collapse
|
15
|
de Mattos Oliveira L, Araújo JSC, de Andrade KVF, Guerrero Moureau ATG, Dos Santos Junior MC. Compounds from Natural Products Candidates to Drug for Chikungunya Virus Infection: A Systematic Review. Curr Drug Targets 2024; 25:635-648. [PMID: 38847165 DOI: 10.2174/0113894501304256240524052446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 09/21/2024]
Abstract
INTRODUCTION Chikungunya fever is a disease caused by infection with the Chikungunya virus, transmitted by Aedes aegypti and Aedes albopictus mosquitoes. Despite its self-limited character, more than 60% of patients have chronic recurrent arthralgia with debilitating pain that lasts for years. AIM The objective of this review was to gather and analyze evidence from the literature on potential therapeutic strategies with molecules from natural products for the treatment of Chikungunya fever. METHODS A search was performed for clinical trials, observational studies, in vitro or in vivo, without restriction of the year of publication or language in electronic databases (Medline/PubMed, EMBASE, Google Scholar, The Cochrane Library, LILACS (BVS), clinical trial registries (Clinical Trials.gov), digital libraries from CAPES theses and dissertations (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior, Brazil) and conference abstracts. A quality assessment of the selected studies was performed using the SYRCLE, RoB2 and SciRAP tools. RESULTS 42 studies were included, which showed molecules with potential antiviral pharmacological activity or with activity in reducing the joint complications caused by CHIKV infection. CONCLUSIONS Among the molecules found in the survey of references, regarding the class of secondary metabolites, flavonoids stood out and for this reason, the molecules may be promising candidates for future clinical trials. Overall, evidence from in vitro studies was of acceptable quality; in vivo and intervention studies showed a high risk of bias, which is a limitation of these studies.
Collapse
Affiliation(s)
- Larissa de Mattos Oliveira
- Programa de Pós-Graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Av. Transnordestina, s/n - Feira de Santana, Novo Horizonte - BA, 44036-900, Brazil
| | - Janay Stefany Carneiro Araújo
- Programa de Pós-Graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Av. Transnordestina, s/n - Feira de Santana, Novo Horizonte - BA, 44036-900, Brazil
| | - Kaio Vinicius Freitas de Andrade
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual de Feira de Santana, Av. Transnordestina, s/n - Feira de Santana, Novo Horizonte - BA, 44036-900, Brazil
| | | | - Manoelito Coelho Dos Santos Junior
- Programa de Pós-Graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Av. Transnordestina, s/n - Feira de Santana, Novo Horizonte - BA, 44036-900, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual de Feira de Santana, Av. Transnordestina, s/n - Feira de Santana, Novo Horizonte - BA, 44036-900, Brazil
| |
Collapse
|
16
|
Cao V, Loeanurit N, Hengphasatporn K, Hairani R, Wacharachaisurapol N, Prompila N, Wittayalertpanya S, Shigeta Y, Khotavivattana T, Chavasiri W, Boonyasuppayakorn S. The 8-bromobaicalein alleviated chikungunya-induced musculoskeletal inflammation and reduced the viral load in healthy adult mice. Emerg Microbes Infect 2023; 12:2270074. [PMID: 37842770 PMCID: PMC10653753 DOI: 10.1080/22221751.2023.2270074] [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] [Received: 06/04/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
Chikungunya virus is a re-emerging arbovirus that has caused epidemic outbreaks in recent decades. Patients in older age groups with high viral load and severe immunologic response during acute infection are likely to develop chronic arthritis and severe joint pain. Currently, no antiviral drug is available. Previous studies suggested that a flavone derivative, 8-bromobaicalein, was a potential dengue and Zika replication inhibitor in a cell-based system targeting flaviviral polymerase. Here we characterized that 8-bromobaicalein inhibited chikungunya virus replication with EC50 of 0.49 ± 0.11 µM in Vero cells. The molecular target predicted at viral nsP1 methyltransferase using molecular binding and fragment molecular orbital calculation. Additionally, oral administration of 250 mg/kg twice daily treatment alleviated chikungunya-induced musculoskeletal inflammation and reduced viral load in healthy adult mice. Pharmacokinetic analysis indicated that the 250 mg/kg administration maintained the compound level above EC99.9 for 12 h. Therefore, 8-bromobaicalein should be a potential candidate for further development as a pan-arboviral drug.
Collapse
Affiliation(s)
- Van Cao
- Center of Excellence in Applied Medical Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Interdisciplinary Program in Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand
- DaNang University of Medical Technology and Pharmacy, DaNang, Vietnam
| | - Naphat Loeanurit
- Center of Excellence in Applied Medical Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Rita Hairani
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Noppadol Wacharachaisurapol
- Clinical Pharmakokinetics and Pharmacogenomics Research Unit, Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nantaporn Prompila
- Chula Pharmacokinetic Research Center, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supeecha Wittayalertpanya
- Clinical Pharmakokinetics and Pharmacogenomics Research Unit, Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Chula Pharmacokinetic Research Center, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yasuteru Shigeta
- Center for Computational Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tanatorn Khotavivattana
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Warinthorn Chavasiri
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Siwaporn Boonyasuppayakorn
- Center of Excellence in Applied Medical Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Vaccine Research and Development, Chulalongkorn University (Chula-VRC), Bangkok, Thailand
| |
Collapse
|
17
|
Gabbianelli R, Shahar E, de Simone G, Rucci C, Bordoni L, Feliziani G, Zhao F, Ferrati M, Maggi F, Spinozzi E, Mahajna J. Plant-Derived Epi-Nutraceuticals as Potential Broad-Spectrum Anti-Viral Agents. Nutrients 2023; 15:4719. [PMID: 38004113 PMCID: PMC10675658 DOI: 10.3390/nu15224719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Although the COVID-19 pandemic appears to be diminishing, the emergence of SARS-CoV-2 variants represents a threat to humans due to their inherent transmissibility, immunological evasion, virulence, and invulnerability to existing therapies. The COVID-19 pandemic affected more than 500 million people and caused over 6 million deaths. Vaccines are essential, but in circumstances in which vaccination is not accessible or in individuals with compromised immune systems, drugs can provide additional protection. Targeting host signaling pathways is recommended due to their genomic stability and resistance barriers. Moreover, targeting host factors allows us to develop compounds that are effective against different viral variants as well as against newly emerging virus strains. In recent years, the globe has experienced climate change, which may contribute to the emergence and spread of infectious diseases through a variety of factors. Warmer temperatures and changing precipitation patterns can increase the geographic range of disease-carrying vectors, increasing the risk of diseases spreading to new areas. Climate change may also affect vector behavior, leading to a longer breeding season and more breeding sites for disease vectors. Climate change may also disrupt ecosystems, bringing humans closer to wildlife that transmits zoonotic diseases. All the above factors may accelerate the emergence of new viral epidemics. Plant-derived products, which have been used in traditional medicine for treating pathological conditions, offer structurally novel therapeutic compounds, including those with anti-viral activity. In addition, plant-derived bioactive substances might serve as the ideal basis for developing sustainable/efficient/cost-effective anti-viral alternatives. Interest in herbal antiviral products has increased. More than 50% of approved drugs originate from herbal sources. Plant-derived compounds offer diverse structures and bioactive molecules that are candidates for new drug development. Combining these therapies with conventional drugs could improve patient outcomes. Epigenetics modifications in the genome can affect gene expression without altering DNA sequences. Host cells can use epigenetic gene regulation as a mechanism to silence incoming viral DNA molecules, while viruses recruit cellular epitranscriptomic (covalent modifications of RNAs) modifiers to increase the translational efficiency and transcript stability of viral transcripts to enhance viral gene expression and replication. Moreover, viruses manipulate host cells' epigenetic machinery to ensure productive viral infections. Environmental factors, such as natural products, may influence epigenetic modifications. In this review, we explore the potential of plant-derived substances as epigenetic modifiers for broad-spectrum anti-viral activity, reviewing their modulation processes and anti-viral effects on DNA and RNA viruses, as well as addressing future research objectives in this rapidly emerging field.
Collapse
Affiliation(s)
- Rosita Gabbianelli
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Ehud Shahar
- Department of Nutrition and Natural Products, Migal—Galilee Research Institute, Kiryat Shmona 11016, Israel;
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 1220800, Israel
| | - Gaia de Simone
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Chiara Rucci
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Laura Bordoni
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Giulia Feliziani
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Fanrui Zhao
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Marta Ferrati
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Filippo Maggi
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Eleonora Spinozzi
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal—Galilee Research Institute, Kiryat Shmona 11016, Israel;
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 1220800, Israel
| |
Collapse
|
18
|
Feng JH, Chen K, Shen SY, Luo YF, Liu XH, Chen X, Gao W, Tong YR. The composition, pharmacological effects, related mechanisms and drug delivery of alkaloids from Corydalis yanhusuo. Biomed Pharmacother 2023; 167:115511. [PMID: 37729733 DOI: 10.1016/j.biopha.2023.115511] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023] Open
Abstract
Corydalis yanhusuo W. T. Wang, also known as yanhusuo, yuanhu, yanhu and xuanhu, is one of the herb components of many Chinese Traditional Medicine prescriptions such as Jin Ling Zi San and Yuanhu-Zhitong priscription. C. yanhusuo was traditionally used to relieve pain and motivate blood and Qi circulation. Now there has been growing interest in pharmacological effects of alkaloids, the main bioactive components of C. yanhusuo. Eighty-four alkaloids isolated from C. yanhusuo are its important bioactive components and can be characterized into protoberberine alkaloids, aporphine alkaloids, opiate alkaloids and others and proper extraction or co-administration methods modulate their contents and efficacy. Alkaloids from C. yanhusuo have various pharmacological effects on the nervous system, cardiovascular system, cancer and others through multiple molecular mechanisms such as modulating neurotransmitters, ion channels, gut microbiota, HPA axis and signaling pathways and are potential treatments for many diseases. Plenty of novel drug delivery methods such as autologous red blood cells, self-microemulsifying drug delivery systems, nanoparticles and others have also been investigated to better exert the effects of alkaloids from C. yanhusuo. This review summarized the alkaloid components of C. yanhusuo, their pharmacological effects and mechanisms, and methods of drug delivery to lay a foundation for future investigations.
Collapse
Affiliation(s)
- Jia-Hua Feng
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Kang Chen
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Si-Yu Shen
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Yun-Feng Luo
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Xi-Hong Liu
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Xin Chen
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wei Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Yu-Ru Tong
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
19
|
He Y, Pan Z, Liu Y, Jiang L, Peng H, Zhao P, Qi Z, Liu Y, Tang H. Identification of tyrphostin AG879 and A9 inhibiting replication of chikungunya virus by screening of a kinase inhibitor library. Virology 2023; 588:109900. [PMID: 37832343 DOI: 10.1016/j.virol.2023.109900] [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: 06/16/2023] [Revised: 09/16/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023]
Abstract
Chikungunya virus (CHIKV) is a globally public health threat. There are currently no medications available to treat CHIKV infection. High-throughput screening of 419 kinase inhibitors was performed based on the cytopathic effect method, and six kinase inhibitors with reduced cytopathic effects, including tyrphostin AG879 (AG879), tyrphostin 9 (A9), sorafenib, sorafenib tosylate, regorafenib, and TAK-632, were identified. The anti-CHIKV activities of two receptor tyrosine kinase inhibitors, AG879 and A9, that have not been previously reported, were selected for further evaluation. The results indicated that 50% cytotoxic concentration (CC50) of AG879 and A9 in Vero cells were greater than 30 μM and 6.50 μM, respectively and 50% effective concentration (EC50) were 0.84 μM and 0.36 μM, respectively. The time-of-addition and time-of-removal assays illustrated that both AG879 and A9 function in the middle stage of CHIKV life cycle. Further, AG879 and A9 do not affect viral attachment; however, they inhibit viral RNA replication, and exhibit antiviral activity against CHIKV Eastern/Central/South African and Asian strains, Ross River virus and Sindbis virus in vitro.
Collapse
Affiliation(s)
- Yanhua He
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China
| | - Zhendong Pan
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China
| | - Yan Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China
| | - Liangliang Jiang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China
| | - Haoran Peng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China
| | - Yangang Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China.
| | - Hailin Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, PR China.
| |
Collapse
|
20
|
Raman K, Rajagopal K, Swaminathan G, Jupudi S, Dhama K, Barua R, Emran TB, Osman H, Khandaker MU. A Critical Review on the Potency of Phytoconstituents in the Management of COVID-19. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2023; 17:1320-1340. [DOI: 10.22207/jpam.17.3.38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024] Open
Abstract
Natural products and their derivatives have traditionally been used as a source of therapeutic agents. Their beneficial properties are due to large varieties in their chemical structures and biochemical actions. The discovery of natural products such as phytoconstituents have crucial role in the development of less toxic and more effective drugs. Phytoconstituents have shown to be beneficial in treating viral diseases such as the previous chikungunya virus, hepatitis C virus, SARS, and MERS viral diseases. Flavonoids, alkaloids, terpenoids, and other group of compounds combat against COVID-19 in several ways like by protease inhibition, spike protein inhibition, Nrf2 inhibition. The accumulation of NRF2 inhibits the development of the SARS-CoV-2 virus and stimulates anti-inflammatory action. The present review highlights the therapeutic importance of compounds isolated from medicinal plants and/or herbs, such as crude extracts of Curcumin I-III, Leptodactylone, Ginsenoside-Rb1, Lycorine, Reserpine, Saikosaponin B2, Cepharanthine, Withanoside V, Gingerol, Piperanine, chromans, flavonoids, Amentoflavone etc. against SARS-CoV-2. Natural products are typically safe, stable, and dependable source for finding drugs to control the current pandemic. Antiviral secondary metabolites many medicinal plants have given ingredients that were isolated. The selected plants based phytoconstituents may potentially be used against viruses’ development on anti-SARS-CoV-2 to offer a reference point in this field.
Collapse
|
21
|
Sandenon Seteyen AL, Guiraud P, Gasque P, Girard-Valenciennes E, Sélambarom J. In Vitro Analyses of the Multifocal Effects of Natural Alkaloids Berberine, Matrine, and Tabersonine against the O'nyong-nyong Arthritogenic Alphavirus Infection and Inflammation. Pharmaceuticals (Basel) 2023; 16:1125. [PMID: 37631040 PMCID: PMC10459185 DOI: 10.3390/ph16081125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
O'nyong-nyong virus (ONNV) is a member of the reemerging arthritogenic alphaviruses that cause chronic debilitating polyarthralgia and/or polyarthritis via their tropism for the musculoskeletal system. Thus, the discovery of dual antiviral and anti-inflammatory drugs is a great challenge in this field. We investigated the effects of the common plant-derived alkaloids berberine (isoquinoline), matrine (quinolizidine), and tabersonine (indole) at a non-toxic concentration (10 μM) on a human fibroblast cell line (HS633T) infected by ONNV (MOI 1). Using qRT-PCR analyses, we measured the RNA levels of the gene coding for the viral proteins and for the host cell immune factors. These alkaloids demonstrated multifocal effects by the inhibition of viral replication, as well as the regulation of the type-I interferon antiviral signaling pathway and the inflammatory mediators and pathways. Berberine and tabersonine proved to be the more valuable compounds. The results supported the proposal that these common alkaloids may be useful scaffolds for drug discovery against arthritogenic alphavirus infection.
Collapse
Affiliation(s)
- Anne-Laure Sandenon Seteyen
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France; (A.-L.S.S.); (P.G.)
| | - Pascale Guiraud
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France; (A.-L.S.S.); (P.G.)
| | - Philippe Gasque
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France; (A.-L.S.S.); (P.G.)
- Centre Hospitalier Universitaire de La Réunion, Laboratoire d’Immunologie Clinique et Expérimentale de la Zone Océan Indien (LICE-OI), Pôle de Biologie, 97400 Saint-Denis, France
| | - Emmanuelle Girard-Valenciennes
- Laboratoire de Chimie et de Biotechnologie des Produits Naturels (CHEMBIOPRO), Université de La Réunion, 97400 Saint-Denis, France
| | - Jimmy Sélambarom
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France; (A.-L.S.S.); (P.G.)
| |
Collapse
|
22
|
de Jesús López Medina Y, Tamayo-Molina YS, Valdés-López JF, Urcuqui-Inchima S. Protective Effects of Caffeine on Chikungunya and Zika Virus Infections: An in Vitro and in Silico Study. Chem Biodivers 2023; 20:e202300192. [PMID: 37489706 DOI: 10.1002/cbdv.202300192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 07/26/2023]
Abstract
Infection by viruses Chikungunya (CHIKV) and Zika (ZIKV) continue to be serious problems in tropical and subtropical areas of the world. Here, we evaluated the antiviral and virucidal activity of caffeine against CHIKV and ZIKV in Vero, A549, and Huh-7 cell lines. Results showed that caffeine displays antiviral properties against both viruses. By pre-and post-infection treatment, caffeine significantly inhibited CHIKV and ZIKV replication in a dose-dependent manner. Furthermore, caffeine showed a virucidal effect against ZIKV. Molecular docking suggests the possible binding of caffeine with envelope protein and RNA-dependent RNA polymerase of CHIKV and ZIKV. This is the first study that showed an antiviral effect of caffeine against CHIKV and ZIKV. Although further studies are needed to better understand the mechanism of caffeine-mediated repression of viral replication, caffeine appears to be a promising compound that could be used for in vivo studies, perhaps in synergy with other compounds present in daily beverages.
Collapse
Affiliation(s)
| | | | - Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| |
Collapse
|
23
|
Sagna A, Nair RVR, Hulyalkar N, Rajasekharan S, Nair VTG, Sivakumar KC, Suja SR, Baby S, Sreekumar E. Ethyl palmitate, an anti-chikungunya virus principle from Sauropus androgynus, a medicinal plant used to alleviate fever in ethnomedicine. JOURNAL OF ETHNOPHARMACOLOGY 2023; 309:116366. [PMID: 36914036 DOI: 10.1016/j.jep.2023.116366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/21/2023] [Accepted: 03/05/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sauropus androgynus is a medicinal shrub used for the treatment of fever in ethnomedical traditions in various Southeast Asian countries. AIM OF THE STUDY This study was aimed to identify antiviral principles from S. androgynus against Chikungunya virus (CHIKV), a major mosquito-borne pathogen that re-emerged in the last decade, and to unravel their mechanism of action. MATERIALS AND METHODS Hydroalcoholic extract of S. androgynus leaves was screened for anti-CHIKV activity using cytopathic effect (CPE) reduction assay. The extract was subjected to activity guided isolation and the resultant pure molecule was characterized by GC-MS, Co-GC and Co-HPTLC. The isolated molecule was further evaluated for its effect by plaque reduction assay, Western blot and immunofluorescence assays. In silico docking with CHIKV envelope proteins and molecular dynamics simulation (MD) analyses were used to elucidate its possible mechanism of action. RESULTS S. androgynus hydroalcoholic extract showed promising anti-CHIKV activity and its active component, obtained by activity guided isolation, was identified as ethyl palmitate (EP), a fatty acid ester. At 1 μg/mL, EP led to 100% inhibition of CPE and a significant 3 log10 reduction in CHIKV replication in Vero cells at 48 h post-infection. EP was highly potent with an EC50 of 0.0019 μg/mL (0.0068 μM) and a very high selectivity index. EP treatment significantly reduced viral protein expression, and time of addition studies revealed that it acts at the stage of viral entry. A strong binding to the viral envelope protein E1 homotrimer during entry, thus preventing viral fusion, was identified as a possible mechanism by which EP imparts its antiviral effect. CONCLUSIONS S. androgynus contains EP as a potent antiviral principle against CHIKV. This justifies the use of the plant against febrile infections, possibly caused by viruses, in various ethnomedical systems. Our results also prompt more studies on fatty acids and their derivatives against viral diseases.
Collapse
Affiliation(s)
- A Sagna
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute (JNTBGRI), Pacha-Palode, Thiruvananthapuram, 695562, Kerala, India.
| | - Reshma V R Nair
- Phytochemistry and Phytopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute (JNTBGRI), Pacha-Palode, Thiruvananthapuram, 695562, Kerala, India.
| | - Neha Hulyalkar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud P.O., Thiruvananthapuram, 695014, Kerala, India.
| | - S Rajasekharan
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute (JNTBGRI), Pacha-Palode, Thiruvananthapuram, 695562, Kerala, India.
| | - Vinodkumar T G Nair
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute (JNTBGRI), Pacha-Palode, Thiruvananthapuram, 695562, Kerala, India.
| | - K C Sivakumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud P.O., Thiruvananthapuram, 695014, Kerala, India.
| | - S R Suja
- Ethnomedicine and Ethnopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute (JNTBGRI), Pacha-Palode, Thiruvananthapuram, 695562, Kerala, India.
| | - Sabulal Baby
- Phytochemistry and Phytopharmacology Division, Jawaharlal Nehru Tropical Botanic Garden and Research Institute (JNTBGRI), Pacha-Palode, Thiruvananthapuram, 695562, Kerala, India.
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud P.O., Thiruvananthapuram, 695014, Kerala, India; Institute of Advanced Virology (IAV), Bio360 Life Sciences Park, Thonnakkal P.O, Thiruvananthapuram, 695317, Kerala, India.
| |
Collapse
|
24
|
Omidvar Tehrani S, Ghasemzadeh Rahbardar M, Shoorgashti K, Dehghan Nayeri MJ, Mohammadpour AH, Hosseinzadeh H. Evaluation of berberine pellet effect on clinical recovery time in COVID-19 outpatients: A pilot clinical trial. AVICENNA JOURNAL OF PHYTOMEDICINE 2023; 13:265-279. [PMID: 37654997 PMCID: PMC10465880 DOI: 10.22038/ajp.2022.21539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 10/08/2022] [Indexed: 09/02/2023]
Abstract
Objective Severe disease onset of COVID-19 may result in alveolar injury and respiratory failure. Apoptosis and inflammation are the main causes of respiratory distress syndrome. Berberine is used in medicine as an analgesic, anti-asthmatic, anti-inflammatory, and antiviral. In the current investigation, the effect of berberine on COVID-19 outpatients was studied. Materials and Methods The present clinical trial was performed on 40 outpatients who were randomly assigned to berberine (300 mg, TID, 2 weeks) (n=19) or placebo groups (n=21). Both groups received standard therapy and they were monitored on days 3, 7, and 14 after the beginning of the therapy for clinical symptoms' improvement, quantitative CRP, lymphopenia, CBC, and SpO2. The severity and frequency of these symptoms and the level of the parameters were statistically compared between the two groups. Results On days (0, 3, 7, and 14, there was no significant difference between the berberine and placebo groups in the improvement of clinical symptoms (cough, shortness of breath, nausea, loss of smell and taste, diarrhea, dizziness, sore throat, stomachache, body aches, and body temperature), quantitative CRP, lymphopenia, WBC, neutrophils, platelets, or SpO2. Conclusion Berberine (300 mg, TID, two weeks) is ineffective in treating COVID-19. More research with a larger sample size is needed to investigate different berberine dosages in other pharmaceutical formulations.
Collapse
Affiliation(s)
| | | | - Kamran Shoorgashti
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir Hooshang Mohammadpour
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
25
|
Jadimurthy R, Jagadish S, Nayak SC, Kumar S, Mohan CD, Rangappa KS. Phytochemicals as Invaluable Sources of Potent Antimicrobial Agents to Combat Antibiotic Resistance. Life (Basel) 2023; 13:948. [PMID: 37109477 PMCID: PMC10145550 DOI: 10.3390/life13040948] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/04/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Plants have been used for therapeutic purposes against various human ailments for several centuries. Plant-derived natural compounds have been implemented in clinics against microbial diseases. Unfortunately, the emergence of antimicrobial resistance has significantly reduced the efficacy of existing standard antimicrobials. The World Health Organization (WHO) has declared antimicrobial resistance as one of the top 10 global public health threats facing humanity. Therefore, it is the need of the hour to discover new antimicrobial agents against drug-resistant pathogens. In the present article, we have discussed the importance of plant metabolites in the context of their medicinal applications and elaborated on their mechanism of antimicrobial action against human pathogens. The WHO has categorized some drug-resistant bacteria and fungi as critical and high priority based on the need to develope new drugs, and we have considered the plant metabolites that target these bacteria and fungi. We have also emphasized the role of phytochemicals that target deadly viruses such as COVID-19, Ebola, and dengue. Additionally, we have also elaborated on the synergetic effect of plant-derived compounds with standard antimicrobials against clinically important microbes. Overall, this article provides an overview of the importance of considering phytogenous compounds in the development of antimicrobial compounds as therapeutic agents against drug-resistant microbes.
Collapse
Affiliation(s)
- Ragi Jadimurthy
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | - Swamy Jagadish
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | - Siddaiah Chandra Nayak
- Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore 570006, India;
| | - Sumana Kumar
- Department of Microbiology, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysore 570015, India
| | - Chakrabhavi Dhananjaya Mohan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; (R.J.); (S.J.)
| | | |
Collapse
|
26
|
Puhl AC, Fernandes RS, Godoy AS, Gil LHVG, Oliva G, Ekins S. The protein disulfide isomerase inhibitor 3-methyltoxoflavin inhibits Chikungunya virus. Bioorg Med Chem 2023; 83:117239. [PMID: 36940609 PMCID: PMC10150329 DOI: 10.1016/j.bmc.2023.117239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Chikungunya virus (CHIKV) is the etiological agent of chikungunya fever, a (re)emerging arbovirus infection, that causes severe and often persistent arthritis, as well as representing a serious health concern worldwide for which no antivirals are currently available. Despite efforts over the last decade to identify and optimize new inhibitors or to reposition existing drugs, no compound has progressed to clinical trials for CHIKV and current prophylaxis is based on vector control, which has shown limited success in containing the virus. Our efforts to rectify this situation were initiated by screening 36 compounds using a replicon system and ultimately identified the natural product derivative 3-methyltoxoflavin with activity against CHIKV using a cell-based assay (EC50 200 nM, SI = 17 in Huh-7 cells). We have additionally screened 3-methyltoxoflavin against a panel of 17 viruses and showed that it only additionally demonstrated inhibition of the yellow fever virus (EC50 370 nM, SI = 3.2 in Huh-7 cells). We have also showed that 3-methyltoxoflavin has excellent in vitro human and mouse microsomal metabolic stability, good solubility and high Caco-2 permeability and it is not likely to be a P-glycoprotein substrate. In summary, we demonstrate that 3-methyltoxoflavin has activity against CHIKV, good in vitro absorption, distribution, metabolism and excretion (ADME) properties as well as good calculated physicochemical properties and may represent a valuable starting point for future optimization to develop inhibitors for this and other related viruses.
Collapse
Affiliation(s)
- Ana C. Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Rafaela S. Fernandes
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 - Jardim Santa Angelina, Sao Carlos, SP, 13563-120, Brazil
| | - Andre S. Godoy
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 - Jardim Santa Angelina, Sao Carlos, SP, 13563-120, Brazil
| | - Laura H. V. G. Gil
- Department of Virology, Oswaldo Cruz Foundation, Aggeu Magalhães Institute, Av. Prof. Moraes Rego, s/n - Cidade Universitaria, Recife, PE, 50670-420, Brazil
| | - Glaucius Oliva
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 - Jardim Santa Angelina, Sao Carlos, SP, 13563-120, Brazil
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| |
Collapse
|
27
|
Ponticelli M, Bellone ML, Parisi V, Iannuzzi A, Braca A, de Tommasi N, Russo D, Sileo A, Quaranta P, Freer G, Pistello M, Milella L. Specialized metabolites from plants as a source of new multi-target antiviral drugs: a systematic review. PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2023; 22:1-79. [PMID: 37359711 PMCID: PMC10008214 DOI: 10.1007/s11101-023-09855-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/30/2023] [Indexed: 06/28/2023]
Abstract
Viral infections have always been the main global health challenge, as several potentially lethal viruses, including the hepatitis virus, herpes virus, and influenza virus, have affected human health for decades. Unfortunately, most licensed antiviral drugs are characterized by many adverse reactions and, in the long-term therapy, also develop viral resistance; for these reasons, researchers have focused their attention on investigating potential antiviral molecules from plants. Natural resources indeed offer a variety of specialized therapeutic metabolites that have been demonstrated to inhibit viral entry into the host cells and replication through the regulation of viral absorption, cell receptor binding, and competition for the activation of intracellular signaling pathways. Many active phytochemicals, including flavonoids, lignans, terpenoids, coumarins, saponins, alkaloids, etc., have been identified as potential candidates for preventing and treating viral infections. Using a systematic approach, this review summarises the knowledge obtained to date on the in vivo antiviral activity of specialized metabolites extracted from plant matrices by focusing on their mechanism of action.
Collapse
Affiliation(s)
- Maria Ponticelli
- Department of Science, University of Basilicata, Viale Dell’ateneo Lucano 10, 85100 Potenza, Italy
| | - Maria Laura Bellone
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
- Ph.D. Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Valentina Parisi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
- Ph.D. Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Annamaria Iannuzzi
- Department of Pharmacy, University of Pisa, Via Bonanno 33, 56126 Pisa, Italy
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandra Braca
- Department of Pharmacy, University of Pisa, Via Bonanno 33, 56126 Pisa, Italy
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Nunziatina de Tommasi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Daniela Russo
- Department of Science, University of Basilicata, Viale Dell’ateneo Lucano 10, 85100 Potenza, Italy
| | - Annalisa Sileo
- Department of Science, University of Basilicata, Viale Dell’ateneo Lucano 10, 85100 Potenza, Italy
| | | | - Giulia Freer
- Virology Unit, Pisa University Hospital, Pisa, Italy
| | | | - Luigi Milella
- Department of Science, University of Basilicata, Viale Dell’ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
28
|
Skidmore AM, Bradfute SB. The life cycle of the alphaviruses: From an antiviral perspective. Antiviral Res 2023; 209:105476. [PMID: 36436722 PMCID: PMC9840710 DOI: 10.1016/j.antiviral.2022.105476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
The alphaviruses are a widely distributed group of positive-sense, single stranded, RNA viruses. These viruses are largely arthropod-borne and can be found on all populated continents. These viruses cause significant human disease, and recently have begun to spread into new populations, such as the expansion of Chikungunya virus into southern Europe and the Caribbean, where it has established itself as endemic. The study of alphaviruses is an active and expanding field, due to their impacts on human health, their effects on agriculture, and the threat that some pose as potential agents of biological warfare and terrorism. In this systematic review we will summarize both historic knowledge in the field as well as recently published data that has potential to shift current theories in how alphaviruses are able to function. This review is comprehensive, covering all parts of the alphaviral life cycle as well as a brief overview of their pathology and the current state of research in regards to vaccines and therapeutics for alphaviral disease.
Collapse
Affiliation(s)
- Andrew M Skidmore
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, IDTC Room 3245, Albuquerque, NM, 87131, USA.
| | - Steven B Bradfute
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, IDTC Room 3330A, Albuquerque, NM, 87131, USA.
| |
Collapse
|
29
|
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
|
30
|
Babalghith AO, Al-kuraishy HM, Al-Gareeb AI, De Waard M, Al-Hamash SM, Jean-Marc S, Negm WA, Batiha GES. The role of berberine in Covid-19: potential adjunct therapy. Inflammopharmacology 2022; 30:2003-2016. [PMID: 36183284 PMCID: PMC9526677 DOI: 10.1007/s10787-022-01080-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/09/2022] [Indexed: 12/16/2022]
Abstract
Coronavirus disease 2019 (Covid-19) is a global diastrophic disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Covid-19 leads to inflammatory, immunological, and oxidative changes, by which SARS-CoV-2 leads to endothelial dysfunction (ED), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and multi-organ failure (MOF). Despite evidence illustrating that some drugs and vaccines effectively manage and prevent Covid-19, complementary herbal medicines are urgently needed to control this pandemic disease. One of the most used herbal medicines is berberine (BBR), which has anti-inflammatory, antioxidant, antiviral, and immune-regulatory effects; thus, BBR may be a prospective candidate against SARS-CoV-2 infection. This review found that BBR has anti-SARS-CoV-2 effects with mitigation of associated inflammatory changes. BBR also reduces the risk of ALI/ARDS in Covid-19 patients by inhibiting the release of pro-inflammatory cytokines and inflammatory signaling pathways. In conclusion, BBR has potent anti-inflammatory, antioxidant, and antiviral effects. Therefore, it can be utilized as a possible anti-SARS-CoV-2 agent. BBR inhibits the proliferation of SARS-CoV-2 and attenuates the associated inflammatory disorders linked by the activation of inflammatory signaling pathways. Indeed, BBR can alleviate ALI/ARDS in patients with severe Covid-19. In this sense, clinical trials and prospective studies are suggested to illustrate the potential role of BBR in treating Covid-19.
Collapse
Affiliation(s)
- Ahmad O. Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Mecca, Kingdom of Saudi Arabia
| | - Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120 Saint-Egrève, France
- L’institut du Thorax, INSERM, CNRS, UNIV NANTES, 44007 Nantes, France
- LabEx « Ion Channels, Science and Therapeutics», Université de Nice Sophia-Antipolis, 06560 Valbonne, France
| | - Sadiq Mohammed Al-Hamash
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Sabatier Jean-Marc
- Faculté des sciences médicales et paramédicales, Aix-Marseille Université, Institut de Neurophysiopathologie (INP), CNRS UMR, 7051, 27 Bd Jean Moulin, 13005 Marseille, France
| | - Walaa A. Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527 Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Al Beheira Egypt
| |
Collapse
|
31
|
Cui Y, Zhang L, Hu D, Yang Y. Berberine Inhibits Herpes Simplex Virus 1 Replication in HEK293T Cells. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7137401. [PMID: 36276998 PMCID: PMC9586773 DOI: 10.1155/2022/7137401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/29/2022] [Indexed: 11/25/2022]
Abstract
Berberine exhibits polytrophic medicinal roles in various diseases and is safe and effective. However, its role and the underlying mechanism in the replication of herpes simplex virus 1 (HSV-1) remain unreported. This research aimed to determine the functional mechanisms of berberine on HSV-1 infection. We determined the CC50 (405.11 ± 15.67 μM) and IC50 (45.6 ± 6.84 μM) of berberine on HEK293T cells infected with HSV-1. Berberine inhibited the transcription and translation of HSV-1 activity-related genes (gD, ICP-4, ICP-5, and ICP-8) in HSV-1-infected HEK293T cells dose-dependently. Berberine also inhibited the phosphorylation of MAPK proteins (JNK and p38) and inflammatory responses induced by HSV-1 infection in HEK293T cells dose-dependently. In conclusion, berberine attenuates HSV-1 replication through its activity, infective ability, and inflammatory response. Our research indicated that berberine may be a candidate drug for HSV-1 infection.
Collapse
Affiliation(s)
- Yujuan Cui
- School of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, China
- Beijing Yanqing Center for Diseases Prevention and Control, Beijing 102100, China
| | - Liangjun Zhang
- Beijing Yanqing Center for Diseases Prevention and Control, Beijing 102100, China
| | - Dandong Hu
- School of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, China
- Beijing Yanqing Center for Diseases Prevention and Control, Beijing 102100, China
- Beijing Yanqing Market Supervision Inspection and Testing Monitoring Center, Beijing 102100, China
| | - Yingli Yang
- School of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, China
| |
Collapse
|
32
|
Basavappa MG, Ferretti M, Dittmar M, Stoute J, Sullivan MC, Whig K, Shen H, Liu KF, Schultz DC, Beiting DP, Lynch KW, Henao-Mejia J, Cherry S. The lncRNA ALPHA specifically targets chikungunya virus to control infection. Mol Cell 2022; 82:3729-3744.e10. [PMID: 36167073 PMCID: PMC10464526 DOI: 10.1016/j.molcel.2022.08.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 07/06/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022]
Abstract
Arthropod-borne viruses, including the alphavirus chikungunya virus (CHIKV), cause acute disease in millions of people and utilize potent mechanisms to antagonize and circumvent innate immune pathways including the type I interferon (IFN) pathway. In response, hosts have evolved antiviral counterdefense strategies that remain incompletely understood. Recent studies have found that long noncoding RNAs (lncRNAs) regulate classical innate immune pathways; how lncRNAs contribute to additional antiviral counterdefenses remains unclear. Using high-throughput genetic screening, we identified a cytoplasmic antiviral lncRNA that we named antiviral lncRNA prohibiting human alphaviruses (ALPHA), which is transcriptionally induced by alphaviruses and functions independently of IFN to inhibit the replication of CHIKV and its closest relative, O'nyong'nyong virus (ONNV), but not other viruses. Furthermore, we showed that ALPHA interacts with CHIKV genomic RNA and restrains viral RNA replication. Together, our findings reveal that ALPHA and potentially other lncRNAs can mediate non-canonical antiviral immune responses against specific viruses.
Collapse
Affiliation(s)
- Megha G Basavappa
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max Ferretti
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark Dittmar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julian Stoute
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Megan C Sullivan
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kanupriya Whig
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA; High-Throughput Screening Core, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hui Shen
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathy Fange Liu
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David C Schultz
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA; High-Throughput Screening Core, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Beiting
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristen W Lynch
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jorge Henao-Mejia
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA; High-Throughput Screening Core, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Muema JM, Bargul JL, Obonyo MA, Njeru SN, Matoke-Muhia D, Mutunga JM. Contemporary exploitation of natural products for arthropod-borne pathogen transmission-blocking interventions. Parasit Vectors 2022; 15:298. [PMID: 36002857 PMCID: PMC9404607 DOI: 10.1186/s13071-022-05367-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
An integrated approach to innovatively counter the transmission of various arthropod-borne diseases to humans would benefit from strategies that sustainably limit onward passage of infective life cycle stages of pathogens and parasites to the insect vectors and vice versa. Aiming to accelerate the impetus towards a disease-free world amid the challenges posed by climate change, discovery, mindful exploitation and integration of active natural products in design of pathogen transmission-blocking interventions is of high priority. Herein, we provide a review of natural compounds endowed with blockade potential against transmissible forms of human pathogens reported in the last 2 decades from 2000 to 2021. Finally, we propose various translational strategies that can exploit these pathogen transmission-blocking natural products into design of novel and sustainable disease control interventions. In summary, tapping these compounds will potentially aid in integrated combat mission to reduce disease transmission trends.
Collapse
Affiliation(s)
- Jackson M Muema
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000, Nairobi, 00200, Kenya.
| | - Joel L Bargul
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000, Nairobi, 00200, Kenya.,International Centre of Insect Physiology and Ecology (Icipe), P.O. Box 30772, Nairobi, 00100, Kenya
| | - Meshack A Obonyo
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Egerton, 20115, Kenya
| | - Sospeter N Njeru
- Centre for Traditional Medicine and Drug Research (CTMDR), Kenya Medical Research Institute (KEMRI), P.O. Box 54840, Nairobi, 00200, Kenya
| | - Damaris Matoke-Muhia
- Centre for Biotechnology Research Development (CBRD), Kenya Medical Research Institute (KEMRI), P.O. Box 54840, Nairobi, 00200, Kenya
| | - James M Mutunga
- Department of Biological Sciences, Mount Kenya University (MKU), P.O. Box 54, Thika, 01000, Kenya.,School of Engineering Design, Technology and Professional Programs, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
34
|
Sandenon Seteyen AL, Girard-Valenciennes E, Septembre-Malaterre A, Gasque P, Guiraud P, Sélambarom J. Anti-Alphaviral Alkaloids: Focus on Some Isoquinolines, Indoles and Quinolizidines. Molecules 2022; 27:molecules27165080. [PMID: 36014321 PMCID: PMC9416297 DOI: 10.3390/molecules27165080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
The discovery and the development of safe and efficient therapeutics against arthritogenic alphaviruses (e.g., chikungunya virus) remain a continuous challenge. Alkaloids are structurally diverse and naturally occurring compounds in plants, with a wide range of biological activities including beneficial effects against prominent pathogenic viruses and inflammation. In this short review, we discuss the effects of some alkaloids of three biologically relevant structural classes (isoquinolines, indoles and quinolizidines). Based on various experimental models (viral infections and chronic diseases), we highlight the immunomodulatory effects of these alkaloids. The data established the capacity of these alkaloids to interfere in host antiviral and inflammatory responses through key components (antiviral interferon response, ROS production, inflammatory signaling pathways and pro- and anti-inflammatory cytokines production) also involved in alphavirus infection and resulting inflammation. Thus, these data may provide a convincing perspective of research for the use of alkaloids as immunomodulators against arthritogenic alphavirus infection and induced inflammation.
Collapse
Affiliation(s)
- Anne-Laure Sandenon Seteyen
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
| | - Emmanuelle Girard-Valenciennes
- Laboratoire de Chimie et de Biotechnologie des Produits Naturels (CHEMBIOPRO), Université de La Réunion, 97400 Saint-Denis, France
| | - Axelle Septembre-Malaterre
- Centre Hospitalier Universitaire de La Réunion, Laboratoire d’Immunologie Clinique et Expérimentale de la Zone Océan Indien (LICE-OI), Pôle de Biologie, 97400 Saint-Denis, France
| | - Philippe Gasque
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
- Centre Hospitalier Universitaire de La Réunion, Laboratoire d’Immunologie Clinique et Expérimentale de la Zone Océan Indien (LICE-OI), Pôle de Biologie, 97400 Saint-Denis, France
| | - Pascale Guiraud
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
| | - Jimmy Sélambarom
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
- Correspondence:
| |
Collapse
|
35
|
Freitas TR, Novais RM, Santos IA, Martins DOS, Danuello A, da Silva Bolzani V, Jardim ACG, Pivatto M. In vitro antiviral activity of piperidine alkaloids from Senna spectabilis flowers on Chikungunya virus infection. Pharmacol Rep 2022; 74:752-758. [PMID: 35882766 DOI: 10.1007/s43440-022-00381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chikungunya fever is an endemic disease caused by the Chikungunya virus (CHIKV). To date there is no antiviral treatment against this infection or licensed vaccine to prevent it. Our study aims to evaluate whether (-)-cassine (1) and (-)-spectaline (2), the main alkaloids of Senna spectabilis, display anti-CHIKV activity. Both compounds have been described to be biologically active against neglected tropical diseases, including malaria, leishmaniasis, and schistosomiasis, which emphasizes that these molecules could be repurposed for chikungunya fever treatment. METHODS The structures of the isolated compounds 1 and 2 were identified by NMR and HRESIMS analyses, and their antiviral activity against CHIKV was assessed by a dose-response assay employing BHK-21 cells and CHIKV-nanoluc, a recombinant virus carrying the nanoluciferase gene reporter. RESULTS Compound 1 presented CC50 of 126.5 µM and EC50 of 14.9 µM, while compound 2 presented CC50 of 91.9 µM and EC50 of 8.3 µM. The calculated selectivity index (SI) was 8.5 for 1 and 11.3 for 2. CONCLUSION The data presented herein show that compounds 1 and 2 have potential for being repurposed as anti-CHIKV drug. Our promising in vitro results encourage further in vitro and in vivo assays. This is the first description of the antiviral activity of compounds 1 and 2 against CHIKV infection, which can impact the development of antiviral drug candidates against chikungunya fever, which sometimes can be debilitating.
Collapse
Affiliation(s)
- Thamires Rodrigues Freitas
- Núcleo de Pesquisa em Compostos Bioativos (NPCBio), Instituto de Química, Universidade Federal de Uberlândia, Uberlândia, MG, 38400-902, Brazil
| | - Raul Marques Novais
- Núcleo de Pesquisa em Compostos Bioativos (NPCBio), Instituto de Química, Universidade Federal de Uberlândia, Uberlândia, MG, 38400-902, Brazil
| | - Igor Andrade Santos
- Laboratório de Pesquisa em Antivirais, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia, MG, 38405-317, Uberlândia, Brazil
| | - Daniel Oliveira Silva Martins
- Laboratório de Pesquisa em Antivirais, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia, MG, 38405-317, Uberlândia, Brazil.,Universidade Estadual Paulista "Júlio de Mesquita Filho", São José Do Rio Preto, SP, 15054-000, Brazil
| | - Amanda Danuello
- Núcleo de Pesquisa em Compostos Bioativos (NPCBio), Instituto de Química, Universidade Federal de Uberlândia, Uberlândia, MG, 38400-902, Brazil
| | - Vanderlan da Silva Bolzani
- Núcleo de Bioensaios, Biossíntese e Ecofisiologia de Produtos Naturais (NuBBE), Departamento de Química Orgânica, Instituto de Química, Universidade Estadual Paulista, P.O. Box 355, Araraquara, SP, 14801-970, Brazil
| | - Ana Carolina Gomes Jardim
- Laboratório de Pesquisa em Antivirais, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia, MG, 38405-317, Uberlândia, Brazil. .,Universidade Estadual Paulista "Júlio de Mesquita Filho", São José Do Rio Preto, SP, 15054-000, Brazil.
| | - Marcos Pivatto
- Núcleo de Pesquisa em Compostos Bioativos (NPCBio), Instituto de Química, Universidade Federal de Uberlândia, Uberlândia, MG, 38400-902, Brazil.
| |
Collapse
|
36
|
MBZM-N-IBT, a Novel Small Molecule, Restricts Chikungunya Virus Infection by Targeting nsP2 Protease Activity In Vitro, In Vivo, and Ex Vivo. Antimicrob Agents Chemother 2022; 66:e0046322. [PMID: 35766508 PMCID: PMC9295557 DOI: 10.1128/aac.00463-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The increase in disease incidences and persistent Chikungunya virus (CHIKV)-induced arthritis have been a huge burden on public health globally. In the absence of specific antivirals or vaccines, it is essential to continue efforts to develop effective anti-CHIKV strategies. Our previous study showing the in vitro anti-CHIKV potential of a novel molecule 1-[(2-methylbenzimidazol-1-yl) methyl]-2-oxo-indolin-3-ylidene] amino] thiourea (MBZM-N-IBT) encouraged us to further validate its efficacy. Here, the effect of MBZM-N-IBT was evaluated in vitro in RAW 264.7 cells, in vivo in C57BL/6 mice, and ex vivo in human peripheral blood mononuclear cells (hPBMCs). The study demonstrated that CHIKV infection was efficiently abrogated in RAW 264.7 cells (IC50 = 22.34 μM) with significant inhibition in viral proteins. The inhibition was effective in the postentry step, and MBZM-N-IBT predominately interfered in the early stages of CHIKV life cycle. It was further supported when the protease activity of CHIKV-nsP2 was hindered by the compound. Moreover, it diminished the CHIKV-induced inflammatory responses in vitro through significant downregulation of all the major mitogen-activated protein kinases (MAPKs), NF-κB, cyclooxygenase (COX)-2, and cytokines. Furthermore, MBZM-N-IBT restricted CHIKV infection and inflammation in vivo, leading to reduced clinical scores and complete survival of C57BL/6 mice. Additionally, it has been noticed that the CHIKV infection was reduced remarkably in hPBMC-derived monocyte-macrophage populations ex vivo by the compound. In conclusion, it can be suggested that this novel compound MBZM-N-IBT has been demonstrated to be a potential anti-CHIKV molecule in vitro, in vivo, and ex vivo and fulfilled all the criteria to investigate further for successful treatment of CHIKV infection.
Collapse
|
37
|
Li XW, Lu YY, Zhang SY, Sai NN, Fan YY, Cheng Y, Liu QS. Mechanism of Neural Regeneration Induced by Natural Product LY01 in the 5×FAD Mouse Model of Alzheimer's Disease. Front Pharmacol 2022; 13:926123. [PMID: 35814256 PMCID: PMC9258960 DOI: 10.3389/fphar.2022.926123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/31/2022] [Indexed: 12/28/2022] Open
Abstract
Background: A sharp decline in neural regeneration in patients with Alzheimer's disease (AD) exacerbates the decline of cognition and memory. It is of great significance to screen for innovative drugs that promote endogenous neural regeneration. Cytisine N-methylene-(5,7,4'-trihydroxy)-isoflavone (LY01) is a new compound isolated from the Chinese herbal medicine Sophora alopecuroides with both isoflavone and alkaloid characteristic structures. Its pharmacological effects are worth studying. Objective: This study was designed to determine whether LY01 delays the cognitive and memory decline in the early stage of AD and whether this effect of LY01 is related to promoting neural regeneration. Methods: Eight-week-old 5×Familial Alzheimer's Disease (5×FAD) mice were used as disease models of early AD. Three doses of LY01 administered in two courses (2 and 5 weeks) of treatment were tested. Cognition, memory, and anxiety-like behaviors in mice were evaluated by the Morris water maze, fear conditioning, and open field experiments. Regeneration of neurons in the mouse hippocampus was observed using immunofluorescence staining. The effect of LY01 on cell regeneration was also demonstrated using a series of tests on primary cultured neurons, astrocytes, and neural stem cells (NSCs). In addition, flow cytometry and transcriptome sequencing were carried out to preliminarily explored the mechanisms. Results: We found that LY01 reduced the decline of cognition and memory in the early stage of 5×FAD mice. This effect was related to the proliferation of astrocytes, the proliferation and migration of NSCs, and increases in the number of new cells and neural precursor cells in the dentate gyrus area of 5×FAD mice. This phenomenon could be observed both in 2-week-old female and 5-week-old male LY01-treated 5×FAD mice. The neuronal regeneration induced by LY01 was related to the regulation of the extracellular matrix and associated receptors, and effects on the S phase of the cell cycle. Conclusion: LY01 increases the proliferation of NSCs and astrocytes and the number of neural precursor cells in the hippocampus, resulting in neural regeneration in 5×FAD mice by acting on the extracellular matrix and associated receptors and regulating the S phase of the cell cycle. This provides a new idea for the early intervention and treatment of AD.
Collapse
Affiliation(s)
- Xiao-Wan Li
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yang-Yang Lu
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, Minzu University of China, Beijing, China
| | - Shu-Yao Zhang
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, Minzu University of China, Beijing, China
| | - Ning-Ning Sai
- University Hospital, Tianjin Normal University, Tianjin, China
| | - Yu-Yan Fan
- Traditional Chinese Medicine Department, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Cheng
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- Institute of National Security, Minzu University of China, Beijing, China
| | - Qing-Shan Liu
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, Minzu University of China, Beijing, China
| |
Collapse
|
38
|
The Antifungal Itraconazole Is a Potent Inhibitor of Chikungunya Virus Replication. Viruses 2022; 14:v14071351. [PMID: 35891332 PMCID: PMC9317443 DOI: 10.3390/v14071351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Chikungunya virus (CHIKV) is the causative agent of chikungunya fever, a disabling disease that can cause long-term severe arthritis. Since the last large CHIKV outbreak in 2015, the reemergence of the virus represents a serious public health concern. The morbidity associated with viral infection emphasizes the need for the development of specific anti-CHIKV drugs. Herein, we describe the development and characterization of a CHIKV reporter replicon cell line and its use in replicon-based screenings. We tested 960 compounds from MMV/DNDi Open Box libraries and identified four candidates with interesting antiviral activities, which were confirmed in viral infection assays employing CHIKV-nanoluc and BHK-21 cells. The most noteworthy compound identified was itraconazole (ITZ), an orally available, safe, and cheap antifungal, that showed high selectivity indexes of >312 and >294 in both replicon-based and viral infection assays, respectively. The antiviral activity of this molecule has been described against positive-sense single stranded RNA viruses (+ssRNA) and was related to cholesterol metabolism that could affect the formation of the replication organelles. Although its precise mechanism of action against CHIKV still needs to be elucidated, our results demonstrate that ITZ is a potent inhibitor of the viral replication that could be repurposed as a broad-spectrum antiviral.
Collapse
|
39
|
Al-Harrasi A, Behl T, Upadhyay T, Chigurupati S, Bhatt S, Sehgal A, Bhatia S, Singh S, Sharma N, Vijayabalan S, Palanimuthu VR, Das S, Kaur R, Aleya L, Bungau S. Targeting natural products against SARS-CoV-2. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:42404-42432. [PMID: 35362883 PMCID: PMC8972763 DOI: 10.1007/s11356-022-19770-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 03/13/2022] [Indexed: 06/01/2023]
Abstract
The human coronavirus disease (COVID-19) pandemic is caused by a novel coronavirus; the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Natural products, secondary metabolites show positive leads with antiviral and immunotherapy treatments using genomic studies in silico docking. In addition, it includes the action of a mechanism targeting the SARS-CoV-2. In this literature, we aimed to evaluate the antiviral movement of the NT-VRL-1 unique terpene definition to Human coronavirus (HCoV-229E). The effects of 19 hydrolysable tannins on the SARS-CoV-2 were therefore theoretically reviewed and analyzed utilising the molecular operating surroundings for their C-Like protease 3CLpro catalytic dyad residues Angiotensin converting enzyme-2 (MOE 09). Pedunculagin, tercatan, and castalin were detected as interacting strongly with SARS-receptor Cov-2's binding site and catalytic dyad (Cys145 and His41). SARS-CoV-2 methods of subunit S1 (ACE2) inhibit the interaction of the receiver with the s-protein once a drug molecule is coupled to the s-protein and prevent it from infecting the target cells in alkaloids. Our review strongly demonstrates the evidence that natural compounds and their derivatives can be used against the human coronavirus and serves as an area of research for future perspective.
Collapse
Affiliation(s)
- Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, Birkat Al Mawz, Oman
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Tanuj Upadhyay
- Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh, India
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Shvetank Bhatt
- Amity Institute of Pharmacy, Amity University, Gwalior, Madhya Pradesh, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Birkat Al Mawz, Oman
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shantini Vijayabalan
- Faculty of Health and Medical Sciences, School of Pharmacy, Taylor's University, Subang Jaya, Kuala Lumpur, Malaysia
| | - Vasanth Raj Palanimuthu
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu, India
| | - Suprava Das
- Department of Pharmacology, Faculty of Medicine, AIMST University, Semeling, Bedong, Kedah, Malaysia
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
40
|
Choudhury D, Jala A, Murty US, Borkar RM, Banerjee S. In Vitro and In Vivo Evaluations of Berberine-Loaded Microparticles Filled In-House 3D Printed Hollow Capsular Device for Improved Oral Bioavailability. AAPS PharmSciTech 2022; 23:89. [PMID: 35296955 PMCID: PMC8926385 DOI: 10.1208/s12249-022-02241-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/19/2022] [Indexed: 11/30/2022] Open
Abstract
The low oral bioavailability, short biological half-life, high dose, and frequent dosing of berberine (BBR) contribute to its restricted clinical use despite its extensive pharmacological activity. Thus, the objective of this study was to formulate sustained-release microparticles (MPs) using a pH-independent release polymer and to evaluate their potential to improve the oral bioavailability of BBR. BBR loaded MPs were prepared using the emulsion crosslinking method and evaluated for particle size, circularity, morphology, entrapment efficiency, solid-state analysis, swelling index, and in vitro BBR release study fitted with different models of release kinetics. The MPs exhibited desired particle sizes ranges between 11.09-11.62 μm and were almost spherical in shape, as confirmed by the circularity value and micrographic images. A loss of BBR crystallinity was observed after encapsulation in MPs, as evident from various solid-state analyses. The final optimized batch (F3) showed highest % BBR entrapment efficiency value of 81.63% ± 4.9. The in vitro BBR release performance in both acidic and alkaline media showed the desired sustained release behavior from the crosslinked MPs, where the maximum BBR release was observed at alkaline pH, which is in accordance with the swelling study data. In the in vivo study, the oral absorption profiles of BBR from both pristine and MPs formats were investigated using in-house prototyped 3D printed hollow capsules as a unit dose carrier. In vivo data showed sustained and prolonged absorption behavior of BBR from MPs compared to their pristine counterparts, which resulted in a cumulative increment of relative oral bioavailability to mitigate the aforementioned issues related to BBR. Graphical Abstract.
Collapse
Affiliation(s)
- Dinesh Choudhury
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER)-Guwahati, Changsari, Assam, India
- National Centre for Pharmacoengineering, NIPER-Guwahati, Changsari, Assam, India
| | - Aishwarya Jala
- Department of Pharmaceutical analysis, NIPER-Guwahati, Changsari, Assam, India
| | | | - Roshan M Borkar
- Department of Pharmaceutical analysis, NIPER-Guwahati, Changsari, Assam, India
| | - Subham Banerjee
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER)-Guwahati, Changsari, Assam, India.
- National Centre for Pharmacoengineering, NIPER-Guwahati, Changsari, Assam, India.
| |
Collapse
|
41
|
Sadeer NB, Zengin G, Mahomoodally MF. Biotechnological applications of mangrove plants and their isolated compounds in medicine-a mechanistic overview. Crit Rev Biotechnol 2022; 43:393-414. [PMID: 35285350 DOI: 10.1080/07388551.2022.2033682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mangrove plants, also known as halophytes, are ecologically important plants that grow in various tropical and subtropical intertidal regions. Owing to the extreme abiotic and biotic stressful conditions they thrive in, these plants produce unique compounds with promising pharmacological propensities. Mangroves are inhabited by an astronomical number of fungal communities which produce a diverse array of extracellular degradative enzymes, namely: amylase, cellulase, xylanase, pectinase, cholesterol oxidase, etc. Such enzymes can be isolated from the mangrove fungi and harnessed for different biotechnological applications, for example, as replacements for chemical catalysts. Mangrove microbes attract considerable attention as they shelter the largest group of marine microorganisms that are resistant to extreme conditions and can produce novel biogenic substances. Vaccines developed from mangrove microbes may promise a safe future by developing effective immunization procedures with a minimum of economic burden. Interestingly, mangroves offer an exciting opportunity for synthesizing nanoparticles in a greener way as these plants are naturally rich in phytochemicals. Rhizophora mucronata Lam., Avicennia officinalis L. and Excoecaria agallocha L. are capable of synthesizing nanoparticles which have evolved recently as an alternative in various industries and are used for their biomedical application. Besides, the phytoconstituents isolated from mangrove plants, such as: gallic acid, galactose, lupeol, catechins, carotenoids, etc., were explored for various biological activities. These compounds are used in the pharmaceutical and nutraceutical industries to produce antimicrobial, antioxidant, anticancer, antidiabetic, and other therapeutic agents. The present review provides information on the biotechnological potentials of mangrove plants and their bioactive compounds as a new source of novel drugs, enzymes, nanoparticles and therapeutically important microbial pigments. Thus, this review forms a base of support and hasten the urgent research on biomedical applications of mangroves.
Collapse
Affiliation(s)
- Nabeelah Bibi Sadeer
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Mauritius
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Mohamad Fawzi Mahomoodally
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Mauritius
| |
Collapse
|
42
|
Abstract
Abstract
Viruses completely rely on the energy and metabolic systems of host cells for life activities. Viral infections usually lead to cytopathic effects and host diseases. To date, there are still no specific clinical vaccines or drugs against most viral infections. Therefore, understanding the molecular and cellular mechanisms of viral infections is of great significance to prevent and treat viral diseases. A variety of viral infections are related to the p38 MAPK signalling pathway, and p38 is an important host factor in virus-infected cells. Here, we introduce the different signalling pathways of p38 activation and then summarise how different viruses induce p38 phosphorylation. Finally, we provide a general summary of the effect of p38 activation on virus replication. Our review provides integrated data on p38 activation and viral infections and describes the potential application of targeting p38 as an antiviral strategy.
Collapse
|
43
|
Kumar B, Misra A, Singh SP, Dhar YV, Rawat P, Chattopadhyay D, Barik SK, Srivastava S. In-silico efficacy of potential phytomolecules from Ayurvedic herbs as an adjuvant therapy in management of COVID-19. J Food Drug Anal 2021; 29:559-580. [PMID: 35649148 PMCID: PMC9931022 DOI: 10.38212/2224-6614.3380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 05/17/2021] [Accepted: 08/26/2021] [Indexed: 11/18/2022] Open
Abstract
The recent COVID-19 outbreak caused by SARS-CoV-2 virus has sparked a new spectrum of investigations, research and studies in multifarious directions. Efforts are being made around the world for discovery of effective vaccines/drugs against COVID-19. In this context, Ayurveda, an alternative traditional system of medicine in India may work as an adjuvant therapy in compromised patients. We selected 40 herbal leads on the basis of their traditional applications. The phytomolecules from these leads were further screened through in-silico molecular docking against two main targets of SARS-CoV-2 i.e. the spike protein (S; structural protein) and the main protease (MPRO; non-structural protein). Out of the selected 40, 12 phytomolecules were able to block or stabilize the major functional sites of the main protease and spike protein. Among these, Ginsenoside, Glycyrrhizic acid, Hespiridin and Tribulosin exhibited high binding energy with both main protease and spike protein. Etoposide showed good binding energy only with Spike protein and Teniposide had high binding energy only with main protease. The above phytocompounds showed promising binding efficiency with target proteins indicating their possible applications against SARS-CoV-2. However, these findings need to be validated through in vitro and in vivo experiments with above mentioned potential molecules as candidate drugs for the management of COVID-19. In addition, there is an opportunity for the development of formulations through different permutations and combinations of these phytomolecules to harness their synergistic potential.
Collapse
Affiliation(s)
- Bhanu Kumar
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, U.P. 226001,
India
| | - Ankita Misra
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, U.P. 226001,
India
| | - Satyendra Pratap Singh
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, U.P. 226001,
India
| | - Yogeshwar Vikram Dhar
- Bioinformatics Division, CSIR-National Botanical Research Institute, Lucknow, U.P. 226001,
India
| | - Poonam Rawat
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, U.P. 226001,
India
| | | | - Saroj Kanta Barik
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, U.P. 226001,
India
| | - Sharad Srivastava
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, U.P. 226001,
India
| |
Collapse
|
44
|
Rodriguez-Rodriguez BA, Noval MG, Kaczmarek ME, Jang KK, Thannickal SA, Cifuentes Kottkamp A, Brown RS, Kielian M, Cadwell K, Stapleford KA. Atovaquone and Berberine Chloride Reduce SARS-CoV-2 Replication In Vitro. Viruses 2021; 13:v13122437. [PMID: 34960706 PMCID: PMC8706021 DOI: 10.3390/v13122437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 01/05/2023] Open
Abstract
Epidemic RNA viruses seem to arise year after year leading to countless infections and devastating disease. SARS-CoV-2 is the most recent of these viruses, but there will undoubtedly be more to come. While effective SARS-CoV-2 vaccines are being deployed, one approach that is still missing is effective antivirals that can be used at the onset of infections and therefore prevent pandemics. Here, we screened FDA-approved compounds against SARS-CoV-2. We found that atovaquone, a pyrimidine biosynthesis inhibitor, is able to reduce SARS-CoV-2 infection in human lung cells. In addition, we found that berberine chloride, a plant-based compound used in holistic medicine, was able to inhibit SARS-CoV-2 infection in cells through direct interaction with the virion. Taken together, these studies highlight potential avenues of antiviral development to block emerging viruses. Such proactive approaches, conducted well before the next pandemic, will be essential to have drugs ready for when the next emerging virus hits.
Collapse
Affiliation(s)
- Bruno A. Rodriguez-Rodriguez
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; (B.A.R.-R.); (M.G.N.); (M.E.K.); (K.K.J.); (S.A.T.); (K.C.)
| | - Maria G. Noval
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; (B.A.R.-R.); (M.G.N.); (M.E.K.); (K.K.J.); (S.A.T.); (K.C.)
| | - Maria E. Kaczmarek
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; (B.A.R.-R.); (M.G.N.); (M.E.K.); (K.K.J.); (S.A.T.); (K.C.)
| | - Kyung Ku Jang
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; (B.A.R.-R.); (M.G.N.); (M.E.K.); (K.K.J.); (S.A.T.); (K.C.)
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sara A. Thannickal
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; (B.A.R.-R.); (M.G.N.); (M.E.K.); (K.K.J.); (S.A.T.); (K.C.)
| | | | - Rebecca S. Brown
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (R.S.B.); (M.K.)
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (R.S.B.); (M.K.)
| | - Ken Cadwell
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; (B.A.R.-R.); (M.G.N.); (M.E.K.); (K.K.J.); (S.A.T.); (K.C.)
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; (B.A.R.-R.); (M.G.N.); (M.E.K.); (K.K.J.); (S.A.T.); (K.C.)
- Correspondence:
| |
Collapse
|
45
|
Joshi T, Bhat S, Pundir H, Chandra S. Identification of Berbamine, Oxyacanthine and Rutin from Berberis asiatica as anti-SARS-CoV-2 compounds: An in silico study. J Mol Graph Model 2021; 109:108028. [PMID: 34649146 PMCID: PMC8504924 DOI: 10.1016/j.jmgm.2021.108028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/31/2021] [Accepted: 09/05/2021] [Indexed: 12/12/2022]
Abstract
Owing to the shortage of specific medicines, the global pandemic of COVID-19 caused by SARS-CoV-2 has been the greatest challenge for the science community. Researchers from all over the world developed some drugs which failed to completely suppress the contiguous disease. SARS-CoV-2 main protease (Mpro), an important component in viral pathogenesis, is considered as a prospective drug target to stop SARS-CoV-2 infection. Since identification of phytochemicals with anti-Mpro activity has been carried out to develop the potential drugs against SARS-CoV-2. Therefore, the present study was conducted to screen phytochemicals of Berberis asiatica for anti-SARS-CoV-2 activity. Through text mining, thirty phytochemicals were reported from B. asiatica, of which, three phytochemicals (Berbamine, Oxyacanthine, and Rutin) show high affinity with the SARS-CoV-2 Mpro and exhibited favorable intermolecular interactions with the catalytic residues (His41 and Cys145) and other essential residues. The molecular dynamics simulation showed that Mpro-phytochemical complexes are more stable, less fluctuating, more compact, and moderately extended than the Mpro-X77 (Reference) complex. The number of H-bonds and MMPBSA results also demonstrates that Berbamine, Oxyacanthine, and Rutin are potent Mpro inhibitors having free energy of -20.79, -33.35, and -31.12 kcal mol-1 respectively. The toxicity risk prediction supports all phytochemicals for drug-like and non-toxic nature. From the result, we propose that binding of these phytochemicals could hamper the function of Mpro. This work suggests that selected phytochemicals could be used as novel anti-COVID-19 drug candidates, and might act as novel compounds for in vitro and in vivo study.
Collapse
Affiliation(s)
- Tanuja Joshi
- Computational Biology & Biotechnology Laboratory, Department of Botany, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Sunaullah Bhat
- Department of Zoology, Kumaun University, S.S.J Campus, Almora, 263601, Nainital, Uttarakhand, India
| | - Hemlata Pundir
- Department of Botany, D.S.B Campus, Kumaun University, Nainital, 263002, Uttarakhand, India
| | - Subhash Chandra
- Computational Biology & Biotechnology Laboratory, Department of Botany, Soban Singh Jeena University, Almora, Uttarakhand, India.
| |
Collapse
|
46
|
Telmisartan restricts Chikungunya virus infection in vitro and in vivo through the AT1/PPAR-γ/MAPKs pathways. Antimicrob Agents Chemother 2021; 66:e0148921. [PMID: 34748384 DOI: 10.1128/aac.01489-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chikungunya virus (CHIKV) has re-emerged as a global public health threat. The inflammatory pathways of RAS and PPAR-γ are usually involved in viral infections. Thus, Telmisartan (TM) with known capacity to block AT1 receptor and activate PPAR-γ, was investigated against CHIKV. The anti-CHIKV effect of TM was investigated in vitro (Vero, RAW 264.7 cells and hPBMCs) and in vivo (C57BL/6 mice). TM was found to abrogate CHIKV infection efficiently (IC50 of 15.34-20.89μM in the Vero and RAW 264.7 cells respectively). Viral RNA and proteins were reduced remarkably. Additionally, TM interfered in the early and late stages of CHIKV life cycle with efficacy in both pre and post-treatment assay. Moreover, the agonist of AT1 receptor and antagonist of PPAR-γ increased CHIKV infection suggesting TM's anti-viral potential by modulating host factors. Besides, reduced activation of all major MAPKs, NF-κB (p65) and cytokines by TM through the inflammatory axis supported the fact that the anti-CHIKV efficacy of TM is partly mediated through the AT1/PPAR-γ/MAPKs pathways. Interestingly, at the human equivalent dose, TM abrogated CHIKV infection and inflammation significantly leading to reduced clinical score and complete survival of C57BL/6 mice. Additionally, TM reduced infection in hPBMC derived monocyte-macrophage populations in vitro. Hence, TM was found to reduce CHIKV infection by targeting both viral and host factors. Considering its safety and in vivo efficacy, it can be a suitable candidate in future for repurposing against CHIKV.
Collapse
|
47
|
Bengue M, Pintong AR, Liegeois F, Nougairède A, Hamel R, Pompon J, de Lamballerie X, Roques P, Choumet V, Missé D. Favipiravir Inhibits Mayaro Virus Infection in Mice. Viruses 2021; 13:v13112213. [PMID: 34835018 PMCID: PMC8622800 DOI: 10.3390/v13112213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
Mayaro virus (MAYV) is an emergent alphavirus that causes MAYV fever. It is often associated with debilitating symptoms, particularly arthralgia and myalgia. MAYV infection is becoming a considerable health issue that, unfortunately, lacks a specific antiviral treatment. Favipiravir, a broad-spectrum antiviral drug, has recently been shown to exert anti-MAYV activity in vitro. In the present study, the potential of Favipiravir to inhibit MAYV replication in an in vivo model was evaluated. Immunocompetent mice were orally administrated 300 mg/kg/dose of Favipiravir at pre-, concurrent-, or post-MAYV infection. The results showed a significant reduction in infectious viral particles and viral RNA transcripts in the tissues and blood of the pre- and concurrently treated infected mice. A significant reduction in the presence of both viral RNA transcript and infectious viral particles in the tissue and blood of pre- and concurrently treated infected mice was observed. By contrast, Favipiravir treatment post-MAYV infection did not result in a reduction in viral replication. Interestingly, Favipiravir strongly decreased the blood levels of the liver disease markers aspartate- and alanine aminotransferase in the pre- and concurrently treated MAYV-infected mice. Taken together, these results suggest that Favipiravir is a potent antiviral drug when administered in a timely manner.
Collapse
Affiliation(s)
- Michèle Bengue
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34394 Montpellier, France; (M.B.); (A.-r.P.); (F.L.); (R.H.); (J.P.)
| | - Ai-rada Pintong
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34394 Montpellier, France; (M.B.); (A.-r.P.); (F.L.); (R.H.); (J.P.)
| | - Florian Liegeois
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34394 Montpellier, France; (M.B.); (A.-r.P.); (F.L.); (R.H.); (J.P.)
| | - Antoine Nougairède
- Unité des Virus Emergents (UVE), Institut de Recherche pour le Développement 190, IHU Méditerranée Infection, Institut National de la Santé et de la Recherche Médicale 1207, Aix Marseille Université, 13005 Marseille, France; (A.N.); (X.d.L.)
| | - Rodolphe Hamel
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34394 Montpellier, France; (M.B.); (A.-r.P.); (F.L.); (R.H.); (J.P.)
| | - Julien Pompon
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34394 Montpellier, France; (M.B.); (A.-r.P.); (F.L.); (R.H.); (J.P.)
| | - Xavier de Lamballerie
- Unité des Virus Emergents (UVE), Institut de Recherche pour le Développement 190, IHU Méditerranée Infection, Institut National de la Santé et de la Recherche Médicale 1207, Aix Marseille Université, 13005 Marseille, France; (A.N.); (X.d.L.)
| | - Pierre Roques
- Unité de Virologie, Institut Pasteur de Guinée, Conakry BP4416, Guinea;
- Immunologie des Maladies Virales Auto-Immunes, Hématologiques et Bactériennes (IMVA-HB), Infectious Disease Models and Innovative Therapies (IDMIT): Commissariat a l’Energie Atomique et aux Energies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale U1184, Université Paris Saclay, 92265 Paris, France
| | - Valérie Choumet
- Unité Environnement et Risques Infectieux Groupe Arbovirus, Institut Pasteur, Université de Paris, 75724 Paris, France
- Correspondence: (V.C.); (D.M.)
| | - Dorothée Missé
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34394 Montpellier, France; (M.B.); (A.-r.P.); (F.L.); (R.H.); (J.P.)
- Correspondence: (V.C.); (D.M.)
| |
Collapse
|
48
|
Barboza CM, Pimenta DC, Vigerelli H, de Cássia Rodrigues da Silva A, Garcia JG, Zamudio RM, Castilho JG, Montanha JA, Roehe PM, de Carvalho Ruthner Batista HB. In vitro effects of bufotenine against RNA and DNA viruses. Braz J Microbiol 2021; 52:2475-2482. [PMID: 34562234 PMCID: PMC8475449 DOI: 10.1007/s42770-021-00612-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 09/19/2021] [Indexed: 12/16/2022] Open
Abstract
Bufotenine, an alkaloid that can be found in plant extracts and skin secretions of amphibians, is reported to have potential antiviral activity. The present study evaluated the antiviral activity of bufotenine against different genetic lineages of rabies virus (RABV, a single-stranded, negative-sense RNA virus), canine coronavirus (CCoV, a positive-sense RNA virus) and two double-stranded DNA viruses (two strains of herpes simplex virus type 1/HSV-1 [KOS and the acyclovir-resistant HSV-1 strain 29R] and canine adenovirus 2, CAV-2). The maximal non-toxic bufotenine concentrations in Vero and BHK-21 cells were determined by MTT assays. The antiviral activity of bufotenine against each virus was assessed by examination of reductions in infectious virus titres and plaque assays. All experiments were performed with and without bufotenine, and the results were compared. Bufotenine demonstrated significant RABV inhibitory activity. No antiviral action was observed against CCoV, CAV-2 or HSV-1. These findings indicate that the antiviral activity of bufotenine is somewhat linked to the particular infectious dose used and the genetic lineage of the virus, although the mechanisms of its effects remain undetermined.
Collapse
Affiliation(s)
- Camila Mosca Barboza
- Instituto Pasteur, Av. Paulista, São Paulo, 393, Brazil.
- Universidade Federal Do ABC, Santo André, Brazil.
| | | | - Hugo Vigerelli
- Laboratório de Genética, Instituto Butantan, São Paulo, SP, Brazil
| | | | - Jaíne Gonçalves Garcia
- Instituto Pasteur, Av. Paulista, São Paulo, 393, Brazil
- Universidade Federal Do ABC, Santo André, Brazil
| | - Raphaela Mello Zamudio
- Instituto Pasteur, Av. Paulista, São Paulo, 393, Brazil
- Universidade Federal Do ABC, Santo André, Brazil
| | | | - Jarbas Alves Montanha
- Departamento de Produção E Matéria-Prima - Faculdade de Farmácia da Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Paulo Michel Roehe
- Instituto de Ciências Básicas da Saúde/Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | | |
Collapse
|
49
|
Ratanakomol T, Roytrakul S, Wikan N, Smith DR. Berberine Inhibits Dengue Virus through Dual Mechanisms. Molecules 2021; 26:5501. [PMID: 34576974 PMCID: PMC8470584 DOI: 10.3390/molecules26185501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022] Open
Abstract
Mosquito transmitted viruses, particularly those of the genus Flavivirus, are a significant healthcare burden worldwide, especially in tropical and sub-tropical areas. However, effective medicines for these viral infections remains lacking. Berberine (BBR) is an alkaloid found in some plants used in traditional medicines in Southeast Asia and elsewhere, and BBR has been shown to possess anti-viral activities. During a screen for potential application to mosquito transmitted viruses, BBR was shown to have virucidal activity against dengue virus (DENV; IC50 42.87 µM) as well as against Zika virus (IC50 11.42 µM) and chikungunya virus (IC50 14.21 µM). BBR was shown to have cellular effects that lead to an increase in cellular DENV E protein without a concomitant effect on DENV nonstructural proteins, suggesting an effect on viral particle formation or egress. While BBR was shown to have an effect of ERK1/2 activation this did not result in defects in viral egress mechanisms. The primary effect of BBR on viral production was likely to be through BBR acting through AMPK activation and disruption of lipid metabolism. Combined these results suggest that BBR has a dual effect on DENV infection, and BBR may have the potential for development as an anti-DENV antiviral.
Collapse
Affiliation(s)
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Rangsit 12120, Thailand;
| | - Nitwara Wikan
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand;
| | - Duncan R. Smith
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand;
| |
Collapse
|
50
|
Battisti V, Urban E, Langer T. Antivirals against the Chikungunya Virus. Viruses 2021; 13:1307. [PMID: 34372513 PMCID: PMC8310245 DOI: 10.3390/v13071307] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/20/2023] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that has re-emerged in recent decades, causing large-scale epidemics in many parts of the world. CHIKV infection leads to a febrile disease known as chikungunya fever (CHIKF), which is characterised by severe joint pain and myalgia. As many patients develop a painful chronic stage and neither antiviral drugs nor vaccines are available, the development of a potent CHIKV inhibiting drug is crucial for CHIKF treatment. A comprehensive summary of current antiviral research and development of small-molecule inhibitor against CHIKV is presented in this review. We highlight different approaches used for the identification of such compounds and further discuss the identification and application of promising viral and host targets.
Collapse
Affiliation(s)
| | | | - Thierry Langer
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Vienna, A-1090 Vienna, Austria; (V.B.); (E.U.)
| |
Collapse
|