101
|
Heravi MM, Nazari A. Samarium(ii) iodide-mediated reactions applied to natural product total synthesis. RSC Adv 2022; 12:9944-9994. [PMID: 35424959 PMCID: PMC8965710 DOI: 10.1039/d1ra08163b] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/12/2022] [Indexed: 12/22/2022] Open
Abstract
Natural product synthesis remains a field in which new synthetic methods and reagents are continually being evaluated. Due to the demanding structures and complex functionality of many natural products, only powerful and selective methods and reagents will be highlighted in this proceeding. Since its introduction by Henri Kagan, samarium(ii) iodide (SmI2, Kagan's reagent) has found increasing use in chemical synthesis. Over the years, many reviews have been published on the application of SmI2 in numerous reductive coupling procedures as well as in natural product total synthesis. This review highlights recent advances in SmI2-mediated synthetic strategies, as applied in the total synthesis of natural products since 2004.
Collapse
Affiliation(s)
- Majid M Heravi
- Department of Chemistry, School of Science, Alzahra University PO Box 1993891176 Vanak Tehran Iran +98 21 88041344 +98 21 88044051
| | - Azadeh Nazari
- Department of Chemistry, School of Science, Alzahra University PO Box 1993891176 Vanak Tehran Iran +98 21 88041344 +98 21 88044051
| |
Collapse
|
102
|
To EE, Erlich JR, Liong F, Liong S, Luong R, Oseghale O, Miles MA, Papagianis PC, Quinn KM, Bozinovski S, Vlahos R, Brooks RD, O’Leary JJ, Brooks DA, Selemidis S. Therapeutic Targeting of Endosome and Mitochondrial Reactive Oxygen Species Protects Mice From Influenza Virus Morbidity. Front Pharmacol 2022; 13:870156. [PMID: 35401240 PMCID: PMC8984148 DOI: 10.3389/fphar.2022.870156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/02/2022] [Indexed: 11/29/2022] Open
Abstract
There is an urgent need to develop effective therapeutic strategies including immunomodulators to combat influenza A virus (IAV) infection. Influenza A viruses increase ROS production, which suppress anti-viral responses and contribute to pathological inflammation and morbidity. Two major cellular sites of ROS production are endosomes via the NOX2-oxidase enzyme and the electron transport chain in mitochondria. Here we examined the effect of administration of Cgp91ds-TAT, an endosome-targeted NOX2 oxidase inhibitor, in combination with mitoTEMPO, a mitochondrial ROS scavenger and compared it to monotherapy treatment during an established IAV infection. Mice were infected with IAV (Hkx31 strain; 104PFU/mouse) and 24 h post infection were treated with Cgp91ds-TAT (0.2 mg/kg), mitoTEMPO (100 μg) or with a combination of these inhibitors [Cgp91ds-TAT (0.2 mg/kg)/mitoTEMPO (100 μg)] intranasally every day for up to 2 days post infection (pi). Mice were euthanized on Days 3 or 6 post infection for analyses of disease severity. A combination of Cgp91ds-TAT and mitoTEMPO treatment was more effective than the ROS inhibitors alone at reducing airway and neutrophilic inflammation, bodyweight loss, lung oedema and improved the lung pathology with a reduction in alveolitis following IAV infection. Dual ROS inhibition also caused a significant elevation in Type I IFN expression at the early phase of infection (day 3 pi), however, this response was suppressed at the later phase of infection (day 6 pi). Furthermore, combined treatment with Cgp91ds-TAT and mitoTEMPO resulted in an increase in IAV-specific CD8+ T cells in the lungs. In conclusion, this study demonstrates that the reduction of ROS production in two major subcellular sites, i.e. endosomes and mitochondria, by intranasal delivery of a combination of Cgp91ds-TAT and mitoTEMPO, suppresses the severity of influenza infection and highlights a novel immunomodulatory approach for IAV disease management.
Collapse
Affiliation(s)
- Eunice E. To
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- F.M Kirby Neurobiology Centre, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jonathan R. Erlich
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Felicia Liong
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Stella Liong
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Raymond Luong
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Osezua Oseghale
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Mark A. Miles
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Paris C. Papagianis
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Kylie M. Quinn
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Robert D. Brooks
- Cancer Research Institute and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - John J. O’Leary
- Sir Patrick Dun’s Laboratory, Central Pathology Laboratory, Department of Histopathology Trinity College Dublin, Dublin, Ireland
- Molecular Pathology Laboratory, Coombe Women and Infants’ University Hospital, Dublin, Ireland
| | - Doug A. Brooks
- Cancer Research Institute and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
- Molecular Pathology Laboratory, Coombe Women and Infants’ University Hospital, Dublin, Ireland
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- *Correspondence: Stavros Selemidis,
| |
Collapse
|
103
|
Wolf JJ, Saba JD, Hahm B. Analyzing Opposing Interactions Between Sphingosine 1-Phosphate Lyase and Influenza A Virus. DNA Cell Biol 2022; 41:331-335. [PMID: 35325556 PMCID: PMC9063141 DOI: 10.1089/dna.2022.0071] [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] [Indexed: 11/12/2022] Open
Abstract
Sphingosine 1-phosphate lyase (SPL) is a critical component of sphingosine 1-phosphate (S1P) metabolism. SPL has been associated with several crucial cellular functions due to its role in S1P metabolism, but its role in viral infections is poorly understood. Studies show that SPL has an antiviral function against influenza A virus (IAV) by interacting with IKKɛ, promoting the type I interferon (IFN) innate immune response to IAV infection. However, a more recent study has revealed that IAV NS1 protein hampers this by triggering ubiquitination and subsequent degradation of SPL, which reduces the type I IFN innate immune response. In this study, we describe SPL, the type I IFN response, and known interactions between SPL and IAV.
Collapse
Affiliation(s)
- Jennifer J. Wolf
- Department of Surgery and Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Julie D. Saba
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Bumsuk Hahm
- Department of Surgery and Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
- Address correspondence to: Bumsuk Hahm, PhD, Departments of Surgery and Molecular Microbiology and Immunology, University of Missouri, 1 Hospital Drive, Columbia, MO 65212, USA
| |
Collapse
|
104
|
Rocaglates as Antivirals: Comparing the Effects on Viral Resistance, Anti-Coronaviral Activity, RNA-Clamping on eIF4A and Immune Cell Toxicity. Viruses 2022; 14:v14030519. [PMID: 35336926 PMCID: PMC8950828 DOI: 10.3390/v14030519] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
Rocaglates are potent broad-spectrum antiviral compounds with a promising safety profile. They inhibit viral protein synthesis for different RNA viruses by clamping the 5′-UTRs of mRNAs onto the surface of the RNA helicase eIF4A. Apart from the natural rocaglate silvestrol, synthetic rocaglates like zotatifin or CR-1-31-B have been developed. Here, we compared the effects of rocaglates on viral 5′-UTR-mediated reporter gene expression and binding to an eIF4A-polypurine complex. Furthermore, we analyzed the cytotoxicity of rocaglates on several human immune cells and compared their antiviral activities in coronavirus-infected cells. Finally, the potential for developing viral resistance was evaluated by passaging human coronavirus 229E (HCoV-229E) in the presence of increasing concentrations of rocaglates in MRC-5 cells. Importantly, no decrease in rocaglate-sensitivity was observed, suggesting that virus escape mutants are unlikely to emerge if the host factor eIF4A is targeted. In summary, all three rocaglates are promising antivirals with differences in cytotoxicity against human immune cells, RNA-clamping efficiency, and antiviral activity. In detail, zotatifin showed reduced RNA-clamping efficiency and antiviral activity compared to silvestrol and CR-1-31-B, but was less cytotoxic for immune cells. Our results underline the potential of rocaglates as broad-spectrum antivirals with no indications for the emergence of escape mutations in HCoV-229E.
Collapse
|
105
|
Strategies for fighting pandemic virus infections: Integration of virology and drug delivery. J Control Release 2022; 343:361-378. [PMID: 35122872 PMCID: PMC8810279 DOI: 10.1016/j.jconrel.2022.01.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Respiratory viruses have sometimes resulted in worldwide pandemics, with the influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) being major participants. Long-term efforts have made it possible to control the influenza virus, but seasonal influenza continues to take many lives each year, and a pandemic influenza virus sometimes emerges. Although vaccines for coronavirus disease 2019 (COVID-19) have been developed, we are not yet able to coexist with the SARS-CoV-2. To overcome such viruses, it is necessary to obtain knowledge about international surveillance systems, virology, ecology and to determine that immune responses are effective. The information must then be transferred to drugs. Delivery systems would be expected to contribute to the rational development of drugs. In this review, virologist and drug delivery system (DDS) researchers discuss drug delivery strategies, especially the use of lipid-based nanocarriers, for fighting to respiratory virus infections.
Collapse
|
106
|
Secondary Structure of Influenza A Virus Genomic Segment 8 RNA Folded in a Cellular Environment. Int J Mol Sci 2022; 23:ijms23052452. [PMID: 35269600 PMCID: PMC8910647 DOI: 10.3390/ijms23052452] [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: 12/06/2021] [Revised: 02/05/2022] [Accepted: 02/17/2022] [Indexed: 11/17/2022] Open
Abstract
Influenza A virus (IAV) is a member of the single-stranded RNA (ssRNA) family of viruses. The most recent global pandemic caused by the SARS-CoV-2 virus has shown the major threat that RNA viruses can pose to humanity. In comparison, influenza has an even higher pandemic potential as a result of its high rate of mutations within its relatively short (<13 kbp) genome, as well as its capability to undergo genetic reassortment. In light of this threat, and the fact that RNA structure is connected to a broad range of known biological functions, deeper investigation of viral RNA (vRNA) structures is of high interest. Here, for the first time, we propose a secondary structure for segment 8 vRNA (vRNA8) of A/California/04/2009 (H1N1) formed in the presence of cellular and viral components. This structure shows similarities with prior in vitro experiments. Additionally, we determined the location of several well-defined, conserved structural motifs of vRNA8 within IAV strains with possible functionality. These RNA motifs appear to fold independently of regional nucleoprotein (NP)-binding affinity, but a low or uneven distribution of NP in each motif region is noted. This research also highlights several accessible sites for oligonucleotide tools and small molecules in vRNA8 in a cellular environment that might be a target for influenza A virus inhibition on the RNA level.
Collapse
|
107
|
p21 restricts influenza A virus by perturbing the viral polymerase complex and upregulating type I interferon signaling. PLoS Pathog 2022; 18:e1010295. [PMID: 35180274 PMCID: PMC8920271 DOI: 10.1371/journal.ppat.1010295] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/14/2022] [Accepted: 01/20/2022] [Indexed: 11/19/2022] Open
Abstract
Many cellular genes and networks induced in human lung epithelial cells infected with the influenza virus remain uncharacterized. Here, we find that p21 levels are elevated in response to influenza A virus (IAV) infection, which is independent of p53. Silencing, pharmacological inhibition or deletion of p21 promotes virus replication in vitro and in vivo, indicating that p21 is an influenza restriction factor. Mechanistically, p21 binds to the C-terminus of IAV polymerase subunit PA and competes with PB1 to limit IAV polymerase activity. Besides, p21 promotes IRF3 activation by blocking K48-linked ubiquitination degradation of HO-1 to enhance type I interferons expression. Furthermore, a synthetic p21 peptide (amino acids 36 to 43) significantly inhibits IAV replication in vitro and in vivo. Collectively, our findings reveal that p21 restricts IAV by perturbing the viral polymerase complex and activating the host innate immune response, which may aid the design of desperately needed new antiviral therapeutics. Influenza A virus (IAV) poses a continuous threat to public health and economic stability. The ribonucleoprotein (RNP) of IAV is responsible for the transcription and replication of the viral RNA. These processes require interplay between host factors and RNP components. Here, we report that p21 can be activated by IAV infection and is controlled by a p53-independent pathway. We demonstrate that p21 directly binds to the viral polymerase acidic protein and limits IAV polymerase activity through disrupting the formation of the ribonucleoprotein complex. Additionally, p21 activation promotes IRF3 activation by blocking K48-linked polyubiquitination degradation of HO-1, thereby activating the type I interferon pathway. We further identify an 8-amino-acid peptide of p21 as the minimum motif that effectively inhibits IAV replication and presents therapeutic efficacy both in vitro and in vivo. Thus, our studies not only identify p21 as an antiviral protein, but also provide mechanistic insight to facilitate drug development.
Collapse
|
108
|
Chan RWY, Tao KP, Ye J, Lui KKY, Yang X, Ma C, Chan PKS. Inhibition of Influenza Virus Replication by Oseltamivir Derivatives. Pathogens 2022; 11:pathogens11020237. [PMID: 35215179 PMCID: PMC8879189 DOI: 10.3390/pathogens11020237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 02/05/2023] Open
Abstract
Characterized by the high morbidity and mortality and seasonal surge, the influenza virus (IV) remains a major public health challenge. Oseltamivir is commonly used as a first-line antiviral. As a neuraminidase inhibitor, it attenuates the penetration of viruses through the mucus on the respiratory tract and inhibits the release of virus progeny from infected cells. However, over the years, oseltamivir-resistant strains have been detected in the IV surveillance programs. Therefore, new antivirals that circumvent the resistant strains would be of great importance. In this study, two novel secondary amine derivatives of oseltamivir CUHK326 (6f) and CUHK392 (10i), which bear heteroaryl groups of M2-S31 proton channel inhibitors, were designed, synthesized and subjected to biological evaluation using plaque assay. Influenza A virus (A/Oklahoma/447/2008, H1N1), influenza B viruses (B/HongKong/CUHK33261/2012), an oseltamivir-resistant influenza A virus (A/HongKong/CUHK71923/2009, H1N1) and an oseltamivir-resistant influenza B virus (B/HongKong/CUHK33280/2012) were included in the antiviral effect assessment compared to oseltamivir carboxylate (OC). Both novel compounds significantly reduced the plaque size of seasonal IV A and B, and performed similarly to OC at their corresponding half-maximal inhibitory concentration (IC50). CUHK392 (10i) functioned more effectively than CUHK326 (6f). More importantly, these compounds showed an inhibitory effect on the oseltamivir-resistant strain under 10 nM with selective index (SI) of >200.
Collapse
Affiliation(s)
- Renee W. Y. Chan
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.P.T.); (K.K.Y.L.)
- Laboratory for Paediatric Respiratory Research, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK-UMCU Joint Research Laboratory of Respiratory Virus & Immunobiology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
- Correspondence: (R.W.Y.C.); (P.K.S.C.)
| | - Kin P. Tao
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.P.T.); (K.K.Y.L.)
- Laboratory for Paediatric Respiratory Research, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK-UMCU Joint Research Laboratory of Respiratory Virus & Immunobiology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiqing Ye
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China; (J.Y.); (C.M.)
| | - Kevin K. Y. Lui
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (K.P.T.); (K.K.Y.L.)
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao Yang
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Cong Ma
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China; (J.Y.); (C.M.)
| | - Paul K. S. Chan
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China;
- Correspondence: (R.W.Y.C.); (P.K.S.C.)
| |
Collapse
|
109
|
Glycomic Analysis Reveals That Sialyltransferase Inhibition Is Involved in the Antiviral Effects of Arbidol. J Virol 2022; 96:e0214121. [PMID: 35044216 PMCID: PMC8941891 DOI: 10.1128/jvi.02141-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Due to the high mutation rate of influenza virus and the rapid increase of drug resistance, it is imperative to discover host-targeting antiviral agents with broad-spectrum antiviral activity. Considering the discrepancy between the urgent demand of antiviral drugs during an influenza pandemic and the long-term process of drug discovery and development, it is feasible to explore host-based antiviral agents and strategies from antiviral drugs on the market. In the current study, the antiviral mechanism of arbidol (ARB), a broad-spectrum antiviral drug with potent activity at early stages of viral replication, was investigated from the aspect of hemagglutinin (HA) receptors of host cells. N-glycans that act as the potential binding receptors of HA on 16-human bronchial epithelial (16-HBE) cells were comprehensively profiled for the first time by using an in-depth glycomic approach based on TiO2-PGC chip-Q-TOF MS. Their relative levels upon the treatment of ARB and virus were carefully examined by employing an ultra-high sensitive qualitative method based on Chip LC-QQQ MS, showing that ARB treatment led to significant and extensive decrease of sialic acid (SA)-linked N-glycans (SA receptors), and thereby impaired the virus utilization on SA receptors for rolling and entry. The SA-decreasing effect of ARB was demonstrated to result from its inhibitory effect on sialyltransferases (ST), ST3GAL4 and ST6GAL1 of 16-HBE cells. Silence of STs, natural ST inhibitors, as well as sialidase treatment of 16-HBE cells, resulted in similar potent antiviral activity, whereas ST-inducing agent led to the diminished antiviral effect of ARB. These observations collectively suggesting the involvement of ST inhibition in the antiviral effect of ARB. IMPORTANCE This study revealed, for the first time, that ST inhibition and the resulted destruction of SA receptors of host cells may be an underlying mechanism for the antiviral activity of ARB. ST inhibition has been proposed as a novel host-targeting antiviral approach recently and several compounds are currently under exploration. ARB is the first antiviral drug on the market that was found to possess ST inhibiting function. This will provide crucial evidence for the clinical usages of ARB, such as in combination with neuraminidase (NA) inhibitors to exert optimized antiviral effect, etc. More importantly, as an agent that can inhibit the expression of STs, ARB can serve as a novel lead compound for the discovery and development of host-targeting antiviral drugs.
Collapse
|
110
|
Aledavood E, Selmi B, Estarellas C, Masetti M, Luque FJ. From Acid Activation Mechanisms of Proton Conduction to Design of Inhibitors of the M2 Proton Channel of Influenza A Virus. Front Mol Biosci 2022; 8:796229. [PMID: 35096969 PMCID: PMC8795881 DOI: 10.3389/fmolb.2021.796229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/24/2021] [Indexed: 11/26/2022] Open
Abstract
With an estimated 1 billion people affected across the globe, influenza is one of the most serious health concerns worldwide. Therapeutic treatments have encompassed a number of key functional viral proteins, mainly focused on the M2 proton channel and neuraminidase. This review highlights the efforts spent in targeting the M2 proton channel, which mediates the proton transport toward the interior of the viral particle as a preliminary step leading to the release of the fusion peptide in hemagglutinin and the fusion of the viral and endosomal membranes. Besides the structural and mechanistic aspects of the M2 proton channel, attention is paid to the challenges posed by the development of efficient small molecule inhibitors and the evolution toward novel ligands and scaffolds motivated by the emergence of resistant strains.
Collapse
Affiliation(s)
- Elnaz Aledavood
- Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Institut de Biomedicina and Institut de Química Teòrica i Computacional, University of Barcelona, Barcelona, Spain
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum – Università di Bologna, Bologna, Italy
| | - Beatrice Selmi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum – Università di Bologna, Bologna, Italy
| | - Carolina Estarellas
- Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Institut de Biomedicina and Institut de Química Teòrica i Computacional, University of Barcelona, Barcelona, Spain
- *Correspondence: Carolina Estarellas, ; Matteo Masetti, ; F. Javier Luque,
| | - Matteo Masetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum – Università di Bologna, Bologna, Italy
- *Correspondence: Carolina Estarellas, ; Matteo Masetti, ; F. Javier Luque,
| | - F. Javier Luque
- Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Institut de Biomedicina and Institut de Química Teòrica i Computacional, University of Barcelona, Barcelona, Spain
- *Correspondence: Carolina Estarellas, ; Matteo Masetti, ; F. Javier Luque,
| |
Collapse
|
111
|
Ahmed F, Husain M. Human N-Alpha-Acetyltransferase 60 Promotes Influenza A Virus Infection by Dampening the Interferon Alpha Signaling. Front Immunol 2022; 12:771792. [PMID: 35095845 PMCID: PMC8790067 DOI: 10.3389/fimmu.2021.771792] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/20/2021] [Indexed: 11/20/2022] Open
Abstract
N-alpha-acetyltransferase 60 (NAA60) is the most recently discovered N-terminal acetyltransferase and found only in multicellular eukaryotes. NAA60 localizes to the Golgi complex and is one of the only two N-terminal acetyltransferases known to localize to an organelle. Furthermore, NAA60 possesses a unique ability of catalyzing the acetylation of membrane-anchored proteins at the N-terminus and histones at the lysine side chains. Herein, we demonstrate that NAA60 exhibits proviral properties during influenza A virus (IAV) infection by interfering with the interferon (IFN) α signaling. We found that the depletion and overexpression of NAA60 reduced and enhanced, respectively, the IAV growth in a cell type- and IAV strain-independent manner. Mechanistically, the IAV-induced expression of IFNα was increased and decreased in NAA60-depleted and -overexpressing cells, respectively. Furthermore, the depletion of NAA60 enhanced the level of phosphorylated STAT1 transcription factor as well as the expression of several IFN-stimulated genes (ISGs) such as MX1, CH25H, IFITM3, ISG15 and viperin in infected cells. Whereas the overexpression of NAA60 produced opposite results. Finally, similar results were obtained when the NAA60-depleted cells were treated with purified IFNα. These findings, in conjunction with our recent findings where N-terminal acetylation of many host proteins increased in response to the IAV infection, indicate an important role of N-terminal acetylation during IAV replication.
Collapse
|
112
|
From high-throughput to therapeutic: host-directed interventions against influenza viruses. Curr Opin Virol 2022; 53:101198. [PMID: 35030353 PMCID: PMC9086133 DOI: 10.1016/j.coviro.2021.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Influenza viruses are simultaneously supported and antagonized by factors within the host cell. This close relationship is the theoretical basis for future antivirals that target the host rather than the virus itself, a concept termed host-directed therapeutics. Genetic screening has led to the identification of host factors capable of modulating influenza virus infections, and these factors represent candidate targets for host-directed antiviral strategies. Despite advances in understanding host targets, however, there are currently no host-directed interventions for influenza viruses in clinical use. In this brief review, we discuss some host factors identified in knockout/knockdown and overexpression screens that could potentially be targeted as host-directed influenza intervention strategies. We further comment on the feasibility of changing gene expression in the respiratory tract with RNA delivery vectors and transient CRISPR-mediated gene targeting.
Collapse
|
113
|
Jeyaram RA, Radha CA. Investigation on the Binding Properties of N1 Neuraminidase of H5N1 Influenza Virus in Complex with Fluorinated Sialic Acid Analog Compounds—a Study by Molecular Docking and Molecular Dynamics Simulations. BRAZILIAN JOURNAL OF PHYSICS 2022; 52:21. [PMCID: PMC8656140 DOI: 10.1007/s13538-021-01009-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/12/2021] [Indexed: 03/19/2024]
Abstract
Worldwide, only two types of antiviral inhibitors (M2 ion channel protein inhibitor and Neuraminidase inhibitors) are approved to treat the influenza viral infection. But the mutation of amino acid sequence in the viral membrane proteins creates the viral resistance to existing antiviral drugs or inhibitors. So the corresponding antiviral drugs have to be reformulated to match these antigenic variations. Fluorination on the carbon–based molecule significantly enriches its biological properties. Hence this study is motivated to design the fluorinated sialic acid (SIA) analog inhibitors for the neuraminidase of H5N1 influenza A virus by substituting fluorine atom at different hydroxyls (O2, O4, O7, O8, and O9) of sialic acid. 100 ns molecular dynamics simulations are carried out for each protein–ligand complex system. NAMD pair interaction energy and MM–PBSA binding free energy calculations predict two possible binding modes for N1–SIA_F2, N1–SIA_F4, and N1–SIA_F7 complexes and single binding mode for N1–SIA_F8 and N1–SIA_F9 complexes. RMSD, RMSF, and hydrogen bonding analyses are used to understand the conformational flexibility and structural stability of each complex system. It has been concluded that the fluorinated sialic acid drug candidates SIA_F2 and SIA_F7 have better inhibiting potency against the N1 neuraminidase of H5N1 influenza virus.
Collapse
Affiliation(s)
- R. A. Jeyaram
- Research Laboratory of Molecular Biophysics, Department of Physics, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu India
| | - C. Anu Radha
- Research Laboratory of Molecular Biophysics, Department of Physics, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu India
| |
Collapse
|
114
|
Xu XY, Wang DY, Li YP, Deyrup ST, Zhang HJ. Plant-derived lignans as potential antiviral agents: a systematic review. PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2022; 21:239-289. [PMID: 34093097 PMCID: PMC8165688 DOI: 10.1007/s11101-021-09758-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/20/2021] [Indexed: 05/04/2023]
Abstract
Medicinal plants are one of the most important sources of antiviral agents and lead compounds. Lignans are a large class of natural compounds comprising two phenyl propane units. Many of them have demonstrated biological activities, and some of them have even been developed as therapeutic drugs. In this review, 630 lignans, including those obtained from medicinal plants and their chemical derivatives, were systematically reviewed for their antiviral activity and mechanism of action. The compounds discussed herein were published in articles between 1998 and 2020. The articles were identified using both database searches (e.g., Web of Science, Pub Med and Scifinder) using key words such as: antiviral activity, antiviral effects, lignans, HBV, HCV, HIV, HPV, HSV, JEV, SARS-CoV, RSV and influenza A virus, and directed searches of scholarly publisher's websites including ACS, Elsevier, Springer, Thieme, and Wiley. The compounds were classified on their structural characteristics as 1) arylnaphthalene lignans, 2) aryltetralin lignans, 3) dibenzylbutyrolactone lignans, 4) dibenzylbutane lignans, 5) tetrahydrofuranoid and tetrahydrofurofuranoid lignans, 6) benzofuran lignans, 7) neolignans, 8) dibenzocyclooctadiene lignans and homolignans, and 9) norlignans and other lignoids. Details on isolation and antiviral activities of the most active compounds within each class of lignan are discussed in detail, as are studies of synthetic lignans that provide structure-activity relationship information.
Collapse
Affiliation(s)
- Xin-Ya Xu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, P. R. China
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200 P. R. China
| | - Dong-Ying Wang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, P. R. China
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, 450001 P. R. China
| | - Yi-Ping Li
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 P. R. China
| | - Stephen T. Deyrup
- Department of Chemistry and Biochemistry, Siena College, Loudonville, NY 12211 USA
| | - Hong-Jie Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, P. R. China
| |
Collapse
|
115
|
Abstract
The ongoing SARS-CoV-2 pandemic poses a severe global threat to public health, as do influenza viruses and other coronaviruses. Here, we present chimpanzee adenovirus 68 (AdC68)-based vaccines designed to universally target coronaviruses and influenza. Our design is centered on an immunogen generated by fusing the SARS-CoV-2 receptor-binding domain (RBD) to the conserved stalk of H7N9 hemagglutinin (HA). Remarkably, the constructed vaccine effectively induced both SARS-CoV-2-targeting antibodies and anti-influenza antibodies in mice, consequently affording protection from lethal SARS-CoV-2 and H7N9 challenges as well as effective H3N2 control. We propose our AdC68-vectored coronavirus-influenza vaccine as a universal approach toward curbing respiratory virus-causing pandemics. IMPORTANCE The COVID-19 pandemic exemplifies the severe public health threats of respiratory virus infection and influenza A viruses. The currently envisioned strategy for the prevention of respiratory virus-causing diseases requires the comprehensive administration of vaccines tailored for individual viruses. Here, we present an alternative strategy by designing chimpanzee adenovirus 68-based vaccines which target both the SARS-CoV-2 receptor-binding-domain and the conserved stalk of influenza hemagglutinin. When tested in mice, this strategy attained potent neutralizing antibodies against wild-type SARS-CoV-2 and its emerging variants, enabling an effective protection against lethal SARS-CoV-2 challenge. Notably, it also provided complete protection from lethal H7N9 challenge and efficient control of H3N2-induced morbidity. Our study opens a new avenue to universally curb respiratory virus infection by vaccination.
Collapse
|
116
|
Kwon EB, Oh YC, Hwang YH, Li W, Park SM, Kong R, Kim YS, Choi JG. A Herbal Mixture Formula of OCD20015-V009 Prophylactic Administration to Enhance Interferon-Mediated Antiviral Activity Against Influenza A Virus. Front Pharmacol 2021; 12:764297. [PMID: 34899320 PMCID: PMC8651992 DOI: 10.3389/fphar.2021.764297] [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: 08/25/2021] [Accepted: 10/29/2021] [Indexed: 11/15/2022] Open
Abstract
OCD20015-V009 is an herbal mix of water-extracted Ginseng Radix, Poria (Hoelen), Rehmanniae Radix, Adenophorae Radix, Platycodi Radix, Crataegii Fructus, and Astragali Radix. In this study, its in vitro and in vivo antiviral activity and mechanisms against the influenza A virus were evaluated using a GFP-tagged influenza A virus (A/PR/8/34-GFP) to infect murine macrophages. We found that OCD20015-V009 pre-treatment substantially reduced A/PR/8/34-GFP replication. Also, OCD20015-V009 pre-treatment increased the phosphorylation of type-I IFN-related proteins TBK-1 and STAT1 and the secretion of pro-inflammatory cytokines TNF-α and IL-6 by murine macrophages. Moreover, OCD20015-V009 prophylactic administration increased IFN-stimulated genes-related 15, 20, and 56 and IFN-β mRNA in vitro. Thus, OCD20015-V009 likely modulates murine innate immune response via macrophages. This finding is potentially useful for developing prophylactics or therapeutics against the influenza A virus. Furthermore, pre-treatment with OCD20015-V009 decreased the mortality of the mice exposed to A/PR/8/34-GFP by 20% compared to that in the untreated animals. Thus, OCD20015-V009 stimulates the antiviral response in murine macrophages and mice to viral infections. Additionally, we identified chlorogenic acid and ginsenoside Rd as the antiviral components in OCD20015-V009. Further investigations are needed to elucidate the protective effects of active components of OCD20015-V009 against influenza A viruses.
Collapse
Affiliation(s)
- Eun-Bin Kwon
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, South Korea
| | - You-Chang Oh
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, South Korea
| | - Youn-Hwan Hwang
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Wei Li
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, South Korea
| | | | | | - Young Soo Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, South Korea
| | - Jang-Gi Choi
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, South Korea
| |
Collapse
|
117
|
Bifunctional Inhibitors of Influenza Virus Neuraminidase: Molecular Design of a Sulfonamide Linker. Int J Mol Sci 2021; 22:ijms222313112. [PMID: 34884917 PMCID: PMC8657994 DOI: 10.3390/ijms222313112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022] Open
Abstract
The growing resistance of the influenza virus to widely used competitive neuraminidase inhibitors occupying the active site of the enzyme requires the development of bifunctional compounds that can simultaneously interact with other regulatory sites on the protein surface. When developing such an inhibitor and combining structural fragments that could be located in the sialic acid cavity of the active site and the adjacent 430-cavity, it is necessary to select a suitable linker not only for connecting the fragments, but also to ensure effective interactions with the unique arginine triad Arg118-Arg292-Arg371 of neuraminidase. Using molecular modeling, we have demonstrated the usefulness of the sulfonamide group in the linker design and the potential advantage of this functional group over other isosteric analogues.
Collapse
|
118
|
Tani N, Kawai N, Chong Y, Bando T, Iwaki N, Kashiwagi S, Ikematsu H. Susceptibility of epidemic viruses to neuraminidase inhibitors and treatment-emergent resistance in the Japanese 2019-20 influenza season. J Infect 2021; 84:151-157. [PMID: 34861350 DOI: 10.1016/j.jinf.2021.11.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 11/04/2021] [Accepted: 11/25/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVES To investigate the susceptibility of epidemic influenza viruses to neuraminidase inhibitors (NAIs) and the emergence of resistant viruses after treatment, a prospective, observational study was done in the 2019-20 Japanese influenza season. METHODS Influenza viruses were isolated before and twice after treatment, the first at day 5 and the second at day 10. The 50% inhibitory concentrations (IC50s) to oseltamivir, zanamivir, peramivir, and laninamivir were measured and compared with those of 2010-11 to 2018-19 seasons. NA amino acid sequences were analyzed by next generation sequencing (NGS). RESULTS The IC50 geometric means of the NAIs for 128 A(H1N1)pdm09, 2 A(H3N2), and 33 B were comparable to those of the previous seasons. Only 2 (1.6%) A(H1N1)pdm09 with significantly high IC50 to oseltamivir were found pretreatment. No A(H3N2) or B had resistance. Treatment-emergent oseltamivir resistance was found in 2 among 33 oseltamivir-treated A(H1N1)pdm09, only at the second follow-up. The NGS indicated a rapid increase in the proportion of H275Y to wild type, from 0% to almost 100% between days 5 and 10. CONCLUSIONS These results suggest the continued effectiveness of these NAIs for epidemic influenza in Japan. Treatment-emergent resistant H275Y A(H1N1)pdm09 viruses were detected after oseltamivir treatment, rapidly replacing the wild type.
Collapse
Affiliation(s)
- Naoki Tani
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan.
| | - Naoki Kawai
- Japan Physicians Association, Tokyo Medical Association Building 3F, 2-5 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Yong Chong
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Takuma Bando
- Japan Physicians Association, Tokyo Medical Association Building 3F, 2-5 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Norio Iwaki
- Japan Physicians Association, Tokyo Medical Association Building 3F, 2-5 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Seizaburo Kashiwagi
- Japan Physicians Association, Tokyo Medical Association Building 3F, 2-5 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Hideyuki Ikematsu
- Japan Physicians Association, Tokyo Medical Association Building 3F, 2-5 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan; Ricerca Clinica Co., 2-18-30 Hakataekihigashi, Hakata-ku, Fukuoka 812-0013, Japan
| |
Collapse
|
119
|
Almutairi F, Sarr D, Tucker SL, Fantone K, Lee JK, Rada B. RGS10 Reduces Lethal Influenza Infection and Associated Lung Inflammation in Mice. Front Immunol 2021; 12:772288. [PMID: 34912341 PMCID: PMC8667315 DOI: 10.3389/fimmu.2021.772288] [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: 09/07/2021] [Accepted: 11/10/2021] [Indexed: 01/05/2023] Open
Abstract
Seasonal influenza epidemics represent a significant global health threat. The exacerbated immune response triggered by respiratory influenza virus infection causes severe pulmonary damage and contributes to substantial morbidity and mortality. Regulator of G-protein signaling 10 (RGS10) belongs to the RGS protein family that act as GTPase activating proteins for heterotrimeric G proteins to terminate signaling pathways downstream of G protein-coupled receptors. While RGS10 is highly expressed in immune cells, in particular monocytes and macrophages, where it has strong anti-inflammatory effects, its physiological role in the respiratory immune system has not been explored yet. Here, we show that Rgs10 negatively modulates lung immune and inflammatory responses associated with severe influenza H1N1 virus respiratory infection in a mouse model. In response to influenza A virus challenge, mice lacking RGS10 experience enhanced weight loss and lung viral titers, higher mortality and significantly faster disease onset. Deficiency of Rgs10 upregulates the levels of several proinflammatory cytokines and chemokines and increases myeloid leukocyte accumulation in the infected lung, markedly neutrophils, monocytes, and inflammatory monocytes, which is associated with more pronounced lung damage. Consistent with this, influenza-infected Rgs10-deficent lungs contain more neutrophil extracellular traps and exhibit higher neutrophil elastase activities than wild-type lungs. Overall, these findings propose a novel, in vivo role for RGS10 in the respiratory immune system controlling myeloid leukocyte infiltration, viral clearance and associated clinical symptoms following lethal influenza challenge. RGS10 also holds promise as a new, potential therapeutic target for respiratory infections.
Collapse
Affiliation(s)
- Faris Almutairi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, United States
| | - Demba Sarr
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Samantha L. Tucker
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Kayla Fantone
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Jae-Kyung Lee
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
120
|
Yeo JY, Gan SKE. Peering into Avian Influenza A(H5N8) for a Framework towards Pandemic Preparedness. Viruses 2021; 13:2276. [PMID: 34835082 PMCID: PMC8622263 DOI: 10.3390/v13112276] [Citation(s) in RCA: 4] [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: 09/27/2021] [Revised: 10/20/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
2014 marked the first emergence of avian influenza A(H5N8) in Jeonbuk Province, South Korea, which then quickly spread worldwide. In the midst of the 2020-2021 H5N8 outbreak, it spread to domestic poultry and wild waterfowl shorebirds, leading to the first human infection in Astrakhan Oblast, Russia. Despite being clinically asymptomatic and without direct human-to-human transmission, the World Health Organization stressed the need for continued risk assessment given the nature of Influenza to reassort and generate novel strains. Given its promiscuity and easy cross to humans, the urgency to understand the mechanisms of possible species jumping to avert disastrous pandemics is increasing. Addressing the epidemiology of H5N8, its mechanisms of species jumping and its implications, mutational and reassortment libraries can potentially be built, allowing them to be tested on various models complemented with deep-sequencing and automation. With knowledge on mutational patterns, cellular pathways, drug resistance mechanisms and effects of host proteins, we can be better prepared against H5N8 and other influenza A viruses.
Collapse
Affiliation(s)
- Joshua Yi Yeo
- Antibody & Product Development Lab, EDDC-BII, Agency for Science, Technology and Research (A*STAR), Singapore 138672, Singapore;
| | - Samuel Ken-En Gan
- Antibody & Product Development Lab, EDDC-BII, Agency for Science, Technology and Research (A*STAR), Singapore 138672, Singapore;
- APD SKEG Pte Ltd., Singapore 439444, Singapore
| |
Collapse
|
121
|
Fakhri S, Mohammadi Pour P, Piri S, Farzaei MH, Echeverría J. Modulating Neurological Complications of Emerging Infectious Diseases: Mechanistic Approaches to Candidate Phytochemicals. Front Pharmacol 2021; 12:742146. [PMID: 34764869 PMCID: PMC8576094 DOI: 10.3389/fphar.2021.742146] [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: 07/15/2021] [Accepted: 09/23/2021] [Indexed: 12/02/2022] Open
Abstract
Growing studies are revealing the critical manifestations of influenza, dengue virus (DENV) infection, Zika virus (ZIKV) disease, and Ebola virus disease (EVD) as emerging infectious diseases. However, their corresponding mechanisms of major complications headed for neuronal dysfunction are not entirely understood. From the mechanistic point of view, inflammatory/oxidative mediators are activated during emerging infectious diseases towards less cell migration, neurogenesis impairment, and neuronal death. Accordingly, the virus life cycle and associated enzymes, as well as host receptors, cytokine storm, and multiple signaling mediators, are the leading players of emerging infectious diseases. Consequently, chemokines, interleukins, interferons, carbohydrate molecules, toll-like receptors (TLRs), and tyrosine kinases are leading orchestrates of peripheral and central complications which are in near interconnections. Some of the resulting neuronal manifestations have attracted much attention, including inflammatory polyneuropathy, encephalopathy, meningitis, myelitis, stroke, Guillain-Barré syndrome (GBS), radiculomyelitis, meningoencephalitis, memory loss, headaches, cranial nerve abnormalities, tremor, and seizure. The complex pathophysiological mechanism behind the aforementioned complications urges the need for finding multi-target agents with higher efficacy and lower side effects. In recent decades, the natural kingdom has been highlighted as promising neuroprotective natural products in modulating several dysregulated signaling pathways/mediators. The present study provides neuronal manifestations of some emerging infectious diseases and underlying pathophysiological mechanisms. Besides, a mechanistic-based strategy is developed to introduce candidate natural products as promising multi-target agents in combating major dysregulated pathways towards neuroprotection in influenza, DENV infection, ZIKV disease, and EVD.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pardis Mohammadi Pour
- Department of Pharmacognosy, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sana Piri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
122
|
Molecular Docking and Virtual Screening of an Influenza Virus Inhibitor That Disrupts Protein-Protein Interactions. Viruses 2021; 13:v13112229. [PMID: 34835035 PMCID: PMC8620322 DOI: 10.3390/v13112229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 10/31/2021] [Indexed: 02/02/2023] Open
Abstract
Influenza is an acute respiratory infection caused by the influenza virus, but few drugs are available for its treatment. Consequently, researchers have been engaged in efforts to discover new antiviral mechanisms that can lay the foundation for novel anti-influenza drugs. The viral RNA-dependent RNA polymerase (RdRp) is an enzyme that plays an indispensable role in the viral infection process, which is directly linked to the survival of the virus. Methods of inhibiting PB1-PB2 (basic polymerase 1-basic polymerase 2) interactions, which are a key part of RdRp enzyme activity, are integral in the design of novel antiviral drugs, a specific PB1-PB2 interactions inhibitor has not been reported. We have screened Enamine's database and conducted a parallel screening of multiple docking schemes, followed by simulations of molecular dynamics to determine the structure of a stable ligand-PB1 complex. We also calculated the free energy of binding between the screened compounds and PB1 protein. Ultimately, we screened and identified a potential PB1-PB2 inhibitor using the ADMET prediction model.
Collapse
|
123
|
Hunter J, Arentz S, Goldenberg J, Yang G, Beardsley J, Myers SP, Mertz D, Leeder S. Zinc for the prevention or treatment of acute viral respiratory tract infections in adults: a rapid systematic review and meta-analysis of randomised controlled trials. BMJ Open 2021; 11:e047474. [PMID: 34728441 PMCID: PMC8578211 DOI: 10.1136/bmjopen-2020-047474] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 09/08/2021] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To evaluate the benefits and risks of zinc formulations compared with controls for prevention or treatment of acute viral respiratory tract infections (RTIs) in adults. METHOD Seventeen English and Chinese databases were searched in April/May 2020 for randomised controlled trials (RCTs), and from April/May 2020 to August 2020 for SARS-CoV-2 RCTs. Cochrane rapid review methods were applied. Quality appraisals used the Risk of Bias 2.0 and Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach. RESULTS Twenty-eight RCTs with 5446 participants were identified. None were specific to SARS-CoV-2. Compared with placebo, oral or intranasal zinc prevented 5 RTIs per 100 person-months (95% CI 1 to 8, numbers needed to treat (NNT)=20, moderate-certainty/quality). Sublingual zinc did not prevent clinical colds following human rhinovirus inoculations (relative risk, RR 0.96, 95% CI 0.77 to 1.21, moderate-certainty/quality). On average, symptoms resolved 2 days earlier with sublingual or intranasal zinc compared with placebo (95% CI 0.61 to 3.50, very low-certainty/quality) and 19 more adults per 100 were likely to remain symptomatic on day 7 without zinc (95% CI 2 to 38, NNT=5, low-certainty/quality). There were clinically significant reductions in day 3 symptom severity scores (mean difference, MD -1.20 points, 95% CI -0.66 to -1.74, low-certainty/quality), but not average daily symptom severity scores (standardised MD -0.15, 95% CI -0.43 to 0.13, low-certainty/quality). Non-serious adverse events (AEs) (eg, nausea, mouth/nasal irritation) were higher (RR 1.41, 95% CI 1.17 to 1.69, NNHarm=7, moderate-certainty/quality). Compared with active controls, there were no differences in illness duration or AEs (low-certainty/quality). No serious AEs were reported in the 25 RCTs that monitored them (low-certainty/quality). CONCLUSIONS In adult populations unlikely to be zinc deficient, there was some evidence suggesting zinc might prevent RTIs symptoms and shorten duration. Non-serious AEs may limit tolerability for some. The comparative efficacy/effectiveness of different zinc formulations and doses were unclear. The GRADE-certainty/quality of the evidence was limited by a high risk of bias, small sample sizes and/or heterogeneity. Further research, including SARS-CoV-2 clinical trials is warranted. PROSPERO REGISTRATION NUMBER CRD42020182044.
Collapse
Affiliation(s)
- Jennifer Hunter
- NICM Health Research Institute, Western Sydney University, Penrith, New South Wales, Australia
| | - Susan Arentz
- NICM Health Research Institute, Western Sydney University, Penrith, New South Wales, Australia
| | - Joshua Goldenberg
- Helfgott Research Institute, National University of Natural Medicine, Portland, Oregon, USA
| | - Guoyan Yang
- NICM Health Research Institute, Western Sydney University, Penrith, New South Wales, Australia
| | | | - Stephen P Myers
- NICM Health Research Institute, Western Sydney University, Penrith, New South Wales, Australia
- National Centre for Naturopathic Medicine, Southern Cross University, Lismore, New South Wales, Australia
| | - Dominik Mertz
- Division of Infectious Diseases, Department of Medicine, Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Stephen Leeder
- The Menzies Centre for Health Policy, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
124
|
Orr-Burks N, Murray J, Todd KV, Bakre A, Tripp RA. MicroRNAs affect GPCR and Ion channel genes needed for influenza replication. J Gen Virol 2021; 102:001691. [PMID: 34787540 PMCID: PMC8742985 DOI: 10.1099/jgv.0.001691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/03/2021] [Indexed: 11/18/2022] Open
Abstract
Influenza virus causes seasonal epidemics and sporadic pandemics resulting in morbidity, mortality, and economic losses worldwide. Understanding how to regulate influenza virus replication is important for developing vaccine and therapeutic strategies. Identifying microRNAs (miRs) that affect host genes used by influenza virus for replication can support an antiviral strategy. In this study, G-protein coupled receptor (GPCR) and ion channel (IC) host genes in human alveolar epithelial (A549) cells used by influenza virus for replication (Orr-Burks et al., 2021) were examined as miR target genes following A/CA/04/09- or B/Yamagata/16/1988 replication. Thirty-three miRs were predicted to target GPCR or IC genes and their miR mimics were evaluated for their ability to decrease influenza virus replication. Paired miR inhibitors were used as an ancillary measure to confirm or not the antiviral effects of a miR mimic. Fifteen miRs lowered influenza virus replication and four miRs were found to reduce replication irrespective of virus strain and type differences. These findings provide evidence for novel miR disease intervention strategies for influenza viruses.
Collapse
Affiliation(s)
- Nichole Orr-Burks
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Jackelyn Murray
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Kyle V. Todd
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Abhijeet Bakre
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
125
|
Aroso RT, Schaberle FA, Arnaut LG, Pereira MM. Photodynamic disinfection and its role in controlling infectious diseases. Photochem Photobiol Sci 2021; 20:1497-1545. [PMID: 34705261 PMCID: PMC8548867 DOI: 10.1007/s43630-021-00102-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/03/2021] [Indexed: 12/23/2022]
Abstract
Photodynamic therapy is witnessing a revival of its origins as a response to the rise of multi-drug resistant infections and the shortage of new classes of antibiotics. Photodynamic disinfection (PDDI) of microorganisms is making progresses in preclinical models and in clinical cases, and the perception of its role in the clinical armamentarium for the management of infectious diseases is changing. We review the positioning of PDDI from the perspective of its ability to respond to clinical needs. Emphasis is placed on the pipeline of photosensitizers that proved effective to inactivate biofilms, showed efficacy in animal models of infectious diseases or reached clinical trials. Novel opportunities resulting from the COVID-19 pandemic are briefly discussed. The molecular features of promising photosensitizers are emphasized and contrasted with those of photosensitizers used in the treatment of solid tumors. The development of photosensitizers has been accompanied by the fabrication of a variety of affordable and customizable light sources. We critically discuss the combination between photosensitizer and light source properties that may leverage PDDI and expand its applications to wider markets. The success of PDDI in the management of infectious diseases will ultimately depend on the efficacy of photosensitizers, affordability of the light sources, simplicity of the procedures, and availability of fast and efficient treatments.
Collapse
Affiliation(s)
- Rafael T Aroso
- Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Fábio A Schaberle
- Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Luís G Arnaut
- Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal.
| | - Mariette M Pereira
- Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal.
| |
Collapse
|
126
|
New C, Lee ZY, Tan KS, Wong AHP, Wang DY, Tran T. Tetraspanins: Host Factors in Viral Infections. Int J Mol Sci 2021; 22:11609. [PMID: 34769038 PMCID: PMC8583825 DOI: 10.3390/ijms222111609] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 12/17/2022] Open
Abstract
Tetraspanins are transmembrane glycoproteins that have been shown increasing interest as host factors in infectious diseases. In particular, they were implicated in the pathogenesis of both non-enveloped (human papillomavirus (HPV)) and enveloped (human immunodeficiency virus (HIV), Zika, influenza A virus, (IAV), and coronavirus) viruses through multiple stages of infection, from the initial cell membrane attachment to the syncytium formation and viral particle release. However, the mechanisms by which different tetraspanins mediate their effects vary. This review aimed to compare and contrast the role of tetraspanins in the life cycles of HPV, HIV, Zika, IAV, and coronavirus viruses, which cause the most significant health and economic burdens to society. In doing so, a better understanding of the relative contribution of tetraspanins in virus infection will allow for a more targeted approach in the treatment of these diseases.
Collapse
Affiliation(s)
- ChihSheng New
- Infectious Disease Translational Research Program, National University of Singapore, Singapore 119228, Singapore; (C.N.); (Z.-Y.L.); (K.S.T.); (A.H.-P.W.)
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Zhao-Yong Lee
- Infectious Disease Translational Research Program, National University of Singapore, Singapore 119228, Singapore; (C.N.); (Z.-Y.L.); (K.S.T.); (A.H.-P.W.)
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Kai Sen Tan
- Infectious Disease Translational Research Program, National University of Singapore, Singapore 119228, Singapore; (C.N.); (Z.-Y.L.); (K.S.T.); (A.H.-P.W.)
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 119228, Singapore
| | - Amanda Huee-Ping Wong
- Infectious Disease Translational Research Program, National University of Singapore, Singapore 119228, Singapore; (C.N.); (Z.-Y.L.); (K.S.T.); (A.H.-P.W.)
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - De Yun Wang
- Infectious Disease Translational Research Program, National University of Singapore, Singapore 119228, Singapore; (C.N.); (Z.-Y.L.); (K.S.T.); (A.H.-P.W.)
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Thai Tran
- Infectious Disease Translational Research Program, National University of Singapore, Singapore 119228, Singapore; (C.N.); (Z.-Y.L.); (K.S.T.); (A.H.-P.W.)
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|
127
|
Evidence that two instead of one defective interfering RNA in influenza A virus-derived defective interfering particles (DIPs) does not enhance antiviral activity. Sci Rep 2021; 11:20477. [PMID: 34650149 PMCID: PMC8516915 DOI: 10.1038/s41598-021-99691-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/23/2021] [Indexed: 11/08/2022] Open
Abstract
Influenza A virus (IAV) infection constitutes a significant health threat. Defective interfering particles (DIPs) can arise during IAV infection and inhibit spread of wild type (WT) IAV. DIPs harbor defective RNA segments, termed DI RNAs, that usually contain internal deletions and interfere with replication of WT viral RNA segments. Here, we asked whether DIPs harboring two instead of one DI RNA exert increased antiviral activity. For this, we focused on DI RNAs derived from segments 1 and 3, which encode the polymerase subunits PB2 and PA, respectively. We demonstrate the successful production of DIPs harboring deletions in segments 1 and/or 3, using cell lines that co-express PB2 and PA. Further, we demonstrate that DIPs harboring two instead of one DI RNA do not exhibit increased ability to inhibit replication of a WT RNA segment. Similarly, the presence of two DI RNAs did not augment the induction of the interferon-stimulated gene MxA and the inhibition of IAV infection. Collectively, our findings suggest that the presence of multiple DI RNAs derived from genomic segments encoding polymerase subunits might not result in increased antiviral activity.
Collapse
|
128
|
Latha K, Jamison KF, Watford WT. Tpl2 Ablation Leads to Hypercytokinemia and Excessive Cellular Infiltration to the Lungs During Late Stages of Influenza Infection. Front Immunol 2021; 12:738490. [PMID: 34691044 PMCID: PMC8529111 DOI: 10.3389/fimmu.2021.738490] [Citation(s) in RCA: 4] [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: 07/08/2021] [Accepted: 09/07/2021] [Indexed: 01/22/2023] Open
Abstract
Tumor progression locus 2 (Tpl2) is a serine-threonine kinase known to promote inflammation in response to various pathogen-associated molecular patterns (PAMPs), inflammatory cytokines and G-protein-coupled receptors and consequently aids in host resistance to pathogens. We have recently shown that Tpl2-/- mice succumb to infection with a low-pathogenicity strain of influenza (x31, H3N2) by an unknown mechanism. In this study, we sought to characterize the cytokine and immune cell profile of influenza-infected Tpl2-/- mice to gain insight into its host protective effects. Although Tpl2-/- mice display modestly impaired viral control, no virus was observed in the lungs of Tpl2-/- mice on the day of peak morbidity and mortality suggesting that morbidity is not due to virus cytopathic effects but rather to an overactive antiviral immune response. Indeed, increased levels of interferon-β (IFN-β), the IFN-inducible monocyte chemoattractant protein-1 (MCP-1, CCL2), Macrophage inflammatory protein 1 alpha (MIP-1α; CCL3), MIP-1β (CCL4), RANTES (CCL5), IP-10 (CXCL10) and Interferon-γ (IFN-γ) was observed in the lungs of influenza-infected Tpl2-/- mice at 7 days post infection (dpi). Elevated cytokine and chemokines were accompanied by increased infiltration of the lungs with inflammatory monocytes and neutrophils. Additionally, we noted that increased IFN-β correlated with increased CCL2, CXCL1 and nitric oxide synthase (NOS2) expression in the lungs, which has been associated with severe influenza infections. Bone marrow chimeras with Tpl2 ablation localized to radioresistant cells confirmed that Tpl2 functions, at least in part, within radioresistant cells to limit pro-inflammatory response to viral infection. Collectively, this study suggests that Tpl2 tempers inflammation during influenza infection by constraining the production of interferons and chemokines which are known to promote the recruitment of detrimental inflammatory monocytes and neutrophils.
Collapse
Affiliation(s)
- Krishna Latha
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| | - Katelyn F. Jamison
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Wendy T. Watford
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
129
|
Hossain MG, Akter S, Dhole P, Saha S, Kazi T, Majbauddin A, Islam MS. Analysis of the Genetic Diversity Associated With the Drug Resistance and Pathogenicity of Influenza A Virus Isolated in Bangladesh From 2002 to 2019. Front Microbiol 2021; 12:735305. [PMID: 34603265 PMCID: PMC8484749 DOI: 10.3389/fmicb.2021.735305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/18/2021] [Indexed: 11/16/2022] Open
Abstract
The subtype prevalence, drug resistance- and pathogenicity-associated mutations, and the distribution of the influenza A virus (IAV) isolates identified in Bangladesh from 2002 to 2019 were analyzed using bioinformatic tools. A total of 30 IAV subtypes have been identified in humans (4), avian species (29), and environment (5) in Bangladesh. The predominant subtypes in human and avian species are H1N1/H3N2 and H5N1/H9N2, respectively. However, the subtypes H5N1/H9N2 infecting humans and H3N2/H1N1 infecting avian species have also been identified. Among the avian species, the maximum number of subtypes (27) have been identified in ducks. A 3.56% of the isolates showed neuraminidase inhibitor (NAI) resistance with a prevalence of 8.50, 1.33, and 2.67% in avian species, humans, and the environment, respectively, the following mutations were detected: V116A, I117V, D198N, I223R, S247N, H275Y, and N295S. Prevalence of adamantane-resistant IAVs was 100, 50, and 30.54% in humans, the environment, and avian species, respectively, the subtypes H3N2, H1N1, H9N2, and H5N2 were highly prevalent, with the subtype H5N1 showing a comparatively lower prevalence. Important PB2 mutations such D9N, K526R, A588V, A588I, G590S, Q591R, E627K, K702R, and S714R were identified. A wide range of IAV subtypes have been identified in Bangladesh with a diversified genetic variation in the NA, M2, and PB2 proteins providing drug resistance and enhanced pathogenicity. This study provides a detailed analysis of the subtypes, and the host range of the IAV isolates and the genetic variations related to their proteins, which may aid in the prevention, treatment, and control of IAV infections in Bangladesh, and would serve as a basis for future investigations.
Collapse
Affiliation(s)
- Md Golzar Hossain
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Sharmin Akter
- Department of Physiology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Priya Dhole
- Department of Biology, The Pennsylvania State University, Pennsylvania, PA, United States
| | - Sukumar Saha
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Taheruzzaman Kazi
- Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Abir Majbauddin
- Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Md Sayeedul Islam
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| |
Collapse
|
130
|
Alghanem AF, Aldhahri FA, Althemery AU. Impact of empirical oseltamivir discontinuation in hospitalized patients with community-acquired pneumonia after confirmed negative for influenza. Saudi Med J 2021; 42:1072-1077. [PMID: 34611001 PMCID: PMC9129246 DOI: 10.15537/smj.2021.42.10.20210324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/25/2021] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVES To study the effect of appropriate oseltamivir discontinuation in patients hospitalized with pneumonia, after they tested negative for influenza. METHODS A retrospective study was conducted at King Abdulaziz Medical City, Riyadh, Saudi Arabia. Patients admitted with a diagnosis of community-acquired pneumonia and started on empirical oseltamivir were included. The duration of stay at the hospital and readmission rates were identified. Additionally, we studied factors that led healthcare providers to continue patients on oseltamivir therapy despite testing negative for influenza. RESULTS A total of 210 patients were studied. The rate of empirical oseltamivir appropriate discontinuation was 31% (66 patients). No significant difference was noted between the 2 groups in the length of hospital stay (p=0.46). There was no significant difference in terms of 30-day (OR=0.67, 95% CI [0.28-1.59]), 60-day (OR=1.14, 95% CI [0.47, 2.78]), and 90-day readmission rates (OR=1.35, 95% CI [0.35-5.27]). After adjusting for other variables, admission to the intensive care unit was independently associated with appropriate discontinuation compared with patients admitted to general wards. CONCLUSIONS This study showed that appropriate discontinuation of empirical antiviral therapy is safe, effective, and has no impact on the length of stay and readmission rates.
Collapse
Affiliation(s)
- Ashjan F. Alghanem
- From the Pharmaceutical Care Services, Ministry of the National Guard – Health Affairs (Ashjan and Fahad), King Abdullah International Medical Research Center, and King Saud bin Abdulaziz University for Health Sciences, Riyadh; and from the Clinical Pharmacy Department, College of Pharmacy (Abdullah), Prince Sattam bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia.
| | - Fahad A. Aldhahri
- From the Pharmaceutical Care Services, Ministry of the National Guard – Health Affairs (Ashjan and Fahad), King Abdullah International Medical Research Center, and King Saud bin Abdulaziz University for Health Sciences, Riyadh; and from the Clinical Pharmacy Department, College of Pharmacy (Abdullah), Prince Sattam bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia.
| | - Abdullah U. Althemery
- From the Pharmaceutical Care Services, Ministry of the National Guard – Health Affairs (Ashjan and Fahad), King Abdullah International Medical Research Center, and King Saud bin Abdulaziz University for Health Sciences, Riyadh; and from the Clinical Pharmacy Department, College of Pharmacy (Abdullah), Prince Sattam bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia.
| |
Collapse
|
131
|
Review of Influenza Virus Vaccines: The Qualitative Nature of Immune Responses to Infection and Vaccination Is a Critical Consideration. Vaccines (Basel) 2021; 9:vaccines9090979. [PMID: 34579216 PMCID: PMC8471734 DOI: 10.3390/vaccines9090979] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/28/2021] [Accepted: 08/30/2021] [Indexed: 01/06/2023] Open
Abstract
Influenza viruses have affected the world for over a century, causing multiple pandemics. Throughout the years, many prophylactic vaccines have been developed for influenza; however, these viruses are still a global issue and take many lives. In this paper, we review influenza viruses, associated immunological mechanisms, current influenza vaccine platforms, and influenza infection, in the context of immunocompromised populations. This review focuses on the qualitative nature of immune responses against influenza viruses, with an emphasis on trained immunity and an assessment of the characteristics of the host–pathogen that compromise the effectiveness of immunization. We also highlight innovative immunological concepts that are important considerations for the development of the next generation of vaccines against influenza viruses.
Collapse
|
132
|
Aroso RT, Piccirillo G, Arnaut ZA, Gonzalez AC, Rodrigues FM, Pereira MM. Photodynamic inactivation of influenza virus as a potential alternative for the control of respiratory tract infections. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2021. [DOI: 10.1016/j.jpap.2021.100043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
133
|
Hiscox JA, Khoo SH, Stewart JP, Owen A. Shutting the gate before the horse has bolted: is it time for a conversation about SARS-CoV-2 and antiviral drug resistance? J Antimicrob Chemother 2021; 76:2230-2233. [PMID: 34142123 PMCID: PMC8361339 DOI: 10.1093/jac/dkab189] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
This article provides a brief overview of drug resistance to antiviral therapy as well as known and emergent variability in key SARS-CoV-2 viral sequences. The purpose is to stimulate deliberation about the need to consider drug resistance prior to widespread roll-out of antivirals for SARS-CoV-2. Many existing candidate agents have mechanisms of action involving drug targets likely to be critical for future drug development. Resistance emerged quickly with monotherapies deployed for other pulmonary viruses such as influenza virus, and in HIV mutations in key drug targets compromised efficacy of multiple drugs within a class. The potential for drug resistance in SARS-CoV-2 has not yet been rigorously debated or assessed, and we call for more academic and industry research on this potentially important future threat prior to widespread roll-out of monotherapies for COVID-19 treatment and prevention.
Collapse
Affiliation(s)
- Julian A. Hiscox
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Infectious Diseases Horizontal Technology Centre (ID HTC), A*STAR, Singapore
| | - Saye H. Khoo
- Department of Pharmacology and Therapeutics, Materials Innovation Factory, University of Liverpool, Liverpool, L7 3NY, UK
| | - James P. Stewart
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Andrew Owen
- Department of Pharmacology and Therapeutics, Materials Innovation Factory, University of Liverpool, Liverpool, L7 3NY, UK
- Centre of Excellence in Long acting Therapeutics (CELT), University of Liverpool, Liverpool, L69 3BX, UK
| |
Collapse
|
134
|
Dawre S, Maru S. Human respiratory viral infections: Current status and future prospects of nanotechnology-based approaches for prophylaxis and treatment. Life Sci 2021; 278:119561. [PMID: 33915132 PMCID: PMC8074533 DOI: 10.1016/j.lfs.2021.119561] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/09/2021] [Accepted: 04/18/2021] [Indexed: 12/23/2022]
Abstract
Respiratory viral infections are major cause of highly mortal pandemics. They are impacting socioeconomic development and healthcare system globally. These emerging deadly respiratory viruses develop newer survival strategies to live inside host cells and tricking the immune system of host. Currently, medical facilities, therapies and research -development teams of every country kneel down before novel corona virus (SARS-CoV-2) which claimed ~2,828,629 lives till date. Thus, there is urgent requirement of novel treatment strategies to combat against these emerging respiratory viral infections. Nanocarriers come under the umbrella of nanotechnology and offer numerous benefits compared to traditional dosage forms. Further, unique physicochemical properties (size, shape and surface charge) of nanocarriers provide additional advantage for targeted delivery. This review discusses in detail about the respiratory viruses, their transmission mode and cell invasion pathways, survival strategies, available therapies, and nanocarriers for the delivery of therapeutics. Further, the role of nanocarriers in the development of treatment therapy against SARS-CoV-2 is also overviewed.
Collapse
Affiliation(s)
- Shilpa Dawre
- Department of Pharmaceutics, School of Pharmacy &, Technology Management, SVKM's NMIMS, Babulde Banks of Tapi River, Mumbai-Agra Road, Shirpur, Maharashtra 425405, India.
| | - Saurabh Maru
- School of Pharmacy and Technology Management, SVKM's NMIMS, Babulde Banks of Tapi River, Mumbai-Agra Road, Shirpur, Maharashtra 425405, India.
| |
Collapse
|
135
|
Sasidharan S, Gosu V, Shin D, Nath S, Tripathi T, Saudagar P. Therapeutic p28 peptide targets essential H1N1 influenza virus proteins: insights from docking and molecular dynamics simulations. Mol Divers 2021; 25:1929-1943. [PMID: 33575983 PMCID: PMC7877518 DOI: 10.1007/s11030-021-10193-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/28/2021] [Indexed: 10/28/2022]
Abstract
The H1N1 influenza virus causes a severe disease that affects the human respiratory tract leading to millions of deaths every year. At present, certain vaccines and few drugs are used to control the virus during seasonal outbreaks. However, high mutation rates and genetic reassortment make it challenging to prevent and mitigate outbreaks, leading to pandemics. Thus, alternate therapies are required for its management and control. Here, we report that a bacterial protein, azurin, and its peptide derivatives p18 and p28 target critical proteins of the influenza virus in an effective manner. The molecular docking studies show that the p28 peptide could target C-PB1, NS1-ED, PB2-CBD, PB2-RBD, NP, and PA proteins. These complexes were further subjected to the simulation of molecular dynamics and binding free energy calculations. The data indicate that p28 has an unusually high affinity and forms stable complexes with the viral proteins C-PB1, PB2-CBD, PB2-RBD, and NP. We suggest that the azurin derivative p28 peptide can act as an anti-influenza agent as it can bind to multiple targets and neutralize the virus. Additional experimental studies need to be conducted to evaluate its safety and efficacy as an anti-H1N1 molecule.
Collapse
Affiliation(s)
- Santanu Sasidharan
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Vijayakumar Gosu
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Donghyun Shin
- The Animal Molecular Genetics and Breeding Center, Jeonbuk National University, Jeonju, 54896, Republic of Korea
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Subhradip Nath
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India.
| |
Collapse
|
136
|
Bowles WHD, Gloster TM. Sialidase and Sialyltransferase Inhibitors: Targeting Pathogenicity and Disease. Front Mol Biosci 2021; 8:705133. [PMID: 34395532 PMCID: PMC8358268 DOI: 10.3389/fmolb.2021.705133] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
Sialidases (SAs) and sialyltransferases (STs), the enzymes responsible for removing and adding sialic acid to other glycans, play essential roles in viruses, bacteria, parasites, and humans. Sialic acid is often the terminal sugar on glycans protruding from the cell surface in humans and is an important component for recognition and cell function. Pathogens have evolved to exploit this and use sialic acid to either “cloak” themselves, ensuring they remain undetected, or as a mechanism to enable release of virus progeny. The development of inhibitors against SAs and STs therefore provides the opportunity to target a range of diseases. Inhibitors targeting viral, bacterial, or parasitic enzymes can directly target their pathogenicity in humans. Excellent examples of this can be found with the anti-influenza drugs Zanamivir (Relenza™, GlaxoSmithKline) and Oseltamivir (Tamiflu™, Roche and Gilead), which have been used in the clinic for over two decades. However, the development of resistance against these drugs means there is an ongoing need for novel potent and specific inhibitors. Humans possess 20 STs and four SAs that play essential roles in cellular function, but have also been implicated in cancer progression, as glycans on many cancer cells are found to be hyper-sialylated. Whilst much remains unknown about how STs function in relation to disease, it is clear that specific inhibitors of them can serve both as tools to gain a better understanding of their activity and form the basis for development of anti-cancer drugs. Here we review the recent developments in the design of SA and ST inhibitors against pathogens and humans.
Collapse
Affiliation(s)
- William H D Bowles
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, St Andrews, United Kingdom
| | - Tracey M Gloster
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
137
|
Pedrina J, Stambas J. Targeting the Host Response: Can We Manipulate Extracellular Matrix Metalloproteinase Activity to Improve Influenza Virus Infection Outcomes? Front Mol Biosci 2021; 8:703456. [PMID: 34291090 PMCID: PMC8287203 DOI: 10.3389/fmolb.2021.703456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/08/2021] [Indexed: 11/25/2022] Open
Abstract
Each year, hundreds of thousands of individuals succumb to influenza virus infection and its associated complications. Several preventative and therapeutic options may be applied in order to preserve life. These traditional approaches include administration of seasonal influenza vaccines, pharmacological interventions in the form of antiviral drug therapy and supportive clinical approaches including mechanical ventilation and extracorporeal membrane oxygenation. While these measures have shown varying degrees of success, antiviral therapies and vaccination are constrained due to ongoing antigenic drift. Moreover, clinical approaches can also be associated with complications and drawbacks. These factors have led to the exploration and development of more sophisticated and nuanced therapeutic approaches involving host proteins. Advances in immunotherapy in the cancer field or administration of steroids following virus infection have highlighted the therapeutic potential of targeting host immune responses. We have now reached a point where we can consider the contribution of other “non-traditional” host components such as the extracellular matrix in immunity. Herein, we will review current, established therapeutic interventions and consider novel therapeutic approaches involving the extracellular matrix.
Collapse
Affiliation(s)
- Jess Pedrina
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - John Stambas
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
138
|
Rawangkan A, Kengkla K, Kanchanasurakit S, Duangjai A, Saokaew S. Anti-Influenza with Green Tea Catechins: A Systematic Review and Meta-Analysis. Molecules 2021; 26:molecules26134014. [PMID: 34209247 PMCID: PMC8272076 DOI: 10.3390/molecules26134014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 12/14/2022] Open
Abstract
Influenza is one of the most serious respiratory viral infections worldwide. Although several studies have reported that green tea catechins (GTCs) might prevent influenza virus infection, this remains controversial. We performed a systematic review and meta-analysis of eight studies with 5048 participants that examined the effect of GTC administration on influenza prevention. In a random-effects meta-analysis of five RCTs, 884 participants treated with GTCs showed statistically significant effects on the prevention of influenza infection compared to the control group (risk ratio (RR) 0.67, 95% CIs 0.51–0.89, p = 0.005) without evidence of heterogeneity (I2 = 0%, p = 0.629). Similarly, in three cohort studies with 2223 participants treated with GTCs, there were also statistically significant effects (RR 0.52, 95% CIs 0.35–0.77, p = 0.001) with very low evidence of heterogeneity (I2 = 3%, p = 0.358). Additionally, the overall effect in the subgroup analysis of gargling and orally ingested items (taking capsules and drinking) showed a pooled RR of 0.62 (95% CIs 0.49–0.77, p = 0.003) without heterogeneity (I2 = 0%, p = 0.554). There were no obvious publication biases (Egger’s test (p = 0.138) and Begg’s test (p = 0.103)). Our analysis suggests that green tea consumption is effective in the prophylaxis of influenza infections. To confirm the findings before implementation, longitudinal clinical trials with specific doses of green tea consumption are warranted.
Collapse
Affiliation(s)
- Anchalee Rawangkan
- School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (A.R.); (A.D.)
- Unit of Excellence on Clinical Outcomes Research and IntegratioN (UNICORN), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Kirati Kengkla
- Division of Pharmacy Practice, Department of Pharmaceutical Care, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand; (K.K.); (S.K.)
- Center of Health Outcomes Research and Therapeutic Safety (Cohorts), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Unit of Excellence on Herbal Medicine, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Sukrit Kanchanasurakit
- Division of Pharmacy Practice, Department of Pharmaceutical Care, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand; (K.K.); (S.K.)
- Center of Health Outcomes Research and Therapeutic Safety (Cohorts), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Unit of Excellence on Herbal Medicine, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Division of Pharmaceutical Care, Department of Pharmacy, Phrae Hospital, Phrae 54000, Thailand
| | - Acharaporn Duangjai
- School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (A.R.); (A.D.)
- Unit of Excellence on Clinical Outcomes Research and IntegratioN (UNICORN), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Center of Health Outcomes Research and Therapeutic Safety (Cohorts), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Surasak Saokaew
- Unit of Excellence on Clinical Outcomes Research and IntegratioN (UNICORN), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Division of Pharmacy Practice, Department of Pharmaceutical Care, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand; (K.K.); (S.K.)
- Center of Health Outcomes Research and Therapeutic Safety (Cohorts), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Unit of Excellence on Herbal Medicine, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Division of Pharmaceutical Care, Department of Pharmacy, Phrae Hospital, Phrae 54000, Thailand
- Biofunctional Molecule Exploratory Research Group, Biomedicine Research Advancement Centre, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia
- Correspondence: ; Tel.: +66 (0)5446 6666; Fax: +66 (0)5446 6661
| |
Collapse
|
139
|
Leidner F, Kurt Yilmaz N, Schiffer CA. Deciphering Antifungal Drug Resistance in Pneumocystis jirovecii DHFR with Molecular Dynamics and Machine Learning. J Chem Inf Model 2021; 61:2537-2541. [PMID: 34138546 PMCID: PMC9109225 DOI: 10.1021/acs.jcim.1c00403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Drug resistance impacts the effectiveness of many new therapeutics. Mutations in the therapeutic target confer resistance; however, deciphering which mutations, often remote from the enzyme active site, drive resistance is challenging. In a series of Pneumocystis jirovecii dihydrofolate reductase variants, we elucidate which interactions are key bellwethers to confer resistance to trimethoprim using homology modeling, molecular dynamics, and machine learning. Six molecular features involving mainly residues that did not vary were the best indicators of resistance.
Collapse
Affiliation(s)
- Florian Leidner
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Celia A. Schiffer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
140
|
Hu Z, Lin J, Chen J, Cai T, Xia L, Liu Y, Song X, He Z. Overview of Viral Pneumonia Associated With Influenza Virus, Respiratory Syncytial Virus, and Coronavirus, and Therapeutics Based on Natural Products of Medicinal Plants. Front Pharmacol 2021; 12:630834. [PMID: 34234668 PMCID: PMC8256264 DOI: 10.3389/fphar.2021.630834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/19/2021] [Indexed: 01/29/2023] Open
Abstract
Viral pneumonia has been a serious threat to global health, especially now we have dramatic challenges such as the COVID-19 pandemic. Approximately six million cases of community-acquired pneumonia occur every year, and over 20% of which need hospital admission. Influenza virus, respiratory virus, and coronavirus are the noteworthy causative agents to be investigated based on recent clinical research. Currently, anaphylactic reaction and inflammation induced by antiviral immunity can be incriminated as causative factors for clinicopathological symptoms of viral pneumonia. In this article, we illustrate the structure and related infection mechanisms of these viruses and the current status of antiviral therapies. Owing to a set of antiviral regiments with unsatisfactory clinical effects resulting from side effects, genetic mutation, and growing incidence of resistance, much attention has been paid on medicinal plants as a natural source of antiviral agents. Previous research mainly referred to herbal medicines and plant extracts with curative effects on viral infection models of influenza virus, respiratory virus, and coronavirus. This review summarizes the results of antiviral activities of various medicinal plants and their isolated substances, exclusively focusing on natural products for the treatment of the three types of pathogens that elicit pneumonia. Furthermore, we have introduced several useful screening tools to develop antiviral lead compounds.
Collapse
Affiliation(s)
- Ziwei Hu
- School of Basic Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jinhong Lin
- School of Basic Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jintao Chen
- School of Basic Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Tengxi Cai
- School of Basic Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Lixin Xia
- School of Basic Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Ying Liu
- School of Basic Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Xun Song
- School of Basic Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhendan He
- School of Basic Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China.,College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| |
Collapse
|
141
|
Guo R, Zhao M, Liu H, Su R, Mao Q, Gong L, Cao X, Hao Y. Uncovering the pharmacological mechanisms of Xijiao Dihuang decoction combined with Yinqiao powder in treating influenza viral pneumonia by an integrative pharmacology strategy. Biomed Pharmacother 2021; 141:111676. [PMID: 34126353 DOI: 10.1016/j.biopha.2021.111676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 12/12/2022] Open
Abstract
Xijiao Dihuang decoction combined with Yinqiao powder (XDD-YQP) is a classical combination formula; however, its therapeutic effects in treating influenza viral pneumonia and the pharmacological mechanisms remain unclear. The therapeutic effect of XDD-YQP in influenza viral pneumonia was evaluated in mice. Subsequently, an everted gut sac model coupled with UPLC/Q-TOF MS were used to screen and identify the active compounds of XDD-YQP. Furthermore, network pharmacological analysis was adopted to probe the mechanisms of the active compounds. Lastly, we verified the targets predicted from network pharmacological analysis by differential bioinformatics analysis. Animal experiments showed that XDD-YQP has a therapeutic effect on influenza viral pneumonia. Moreover, 113 active compounds were identified from intestinal absorbed solutions of XDD-YQP. Using network pharmacological analysis, 90 major targets were selected as critical in the treatment of influenza viral pneumonia through 12 relevant pathways. Importantly, the MAPK signaling pathway was found to be closely associated with the other 11 pathways. Moreover, seven key targets, EGFR, FOS, MAPK1, MAP2K1, HRAS, NRAS, and RELA, which are common targets in the MAPK signaling pathway, were investigated. These seven key targets were identified as differentially expressed genes (DEGs) between influenza virus-infected and uninfected individuals. Hence, the seven key targets in the MAPK signaling pathway may play a vital role in the treatment of influenza viral pneumonia with XDD-YQP. This research may offer an integrative pharmacology strategy to clarify the pharmacological mechanisms of traditional Chinese medicines. The results provide a theoretical basis for a broader clinical application of XDD-YQP.
Collapse
Affiliation(s)
- Rui Guo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mengfan Zhao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rina Su
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qin Mao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Leilei Gong
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Xu Cao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Hao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
142
|
Wolf JJ, Xia C, Studstill CJ, Ngo H, Brody SL, Anderson PE, Hahm B. Influenza A virus NS1 induces degradation of sphingosine 1-phosphate lyase to obstruct the host innate immune response. Virology 2021; 558:67-75. [PMID: 33730651 PMCID: PMC8109848 DOI: 10.1016/j.virol.2021.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/11/2021] [Accepted: 02/17/2021] [Indexed: 12/14/2022]
Abstract
The type I interferon (IFN)-mediated innate immune response is one of the central obstacles influenza A virus (IAV) must overcome in order to successfully replicate within the host. We have previously shown that sphingosine 1-phosphate (S1P) lyase (SPL) enhances IKKϵ-mediated type I IFN responses. Here, we demonstrate that the nonstructural protein 1 (NS1) of IAV counteracts the SPL-mediated antiviral response by inducing degradation of SPL. SPL was ubiquitinated and downregulated upon IAV infection or NS1 expression, whereas NS1-deficient IAV failed to elicit SPL ubiquitination or downregulation. Transiently overexpressed SPL increased phosphorylation of IKKϵ, resulting in enhanced expression of type I IFNs. However, this induction was markedly inhibited by IAV NS1. Collectively, this study reveals a novel strategy employed by IAV to subvert the type I IFN response, providing new insights into the interplay between IAV and host innate immunity.
Collapse
Affiliation(s)
- Jennifer J Wolf
- Department of Surgery, University of Missouri, Columbia, MO, 65212, USA; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Chuan Xia
- Department of Surgery, University of Missouri, Columbia, MO, 65212, USA; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA; Present Address: State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Caleb J Studstill
- Department of Surgery, University of Missouri, Columbia, MO, 65212, USA; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Hanh Ngo
- Department of Surgery, University of Missouri, Columbia, MO, 65212, USA; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Steven L Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Paul E Anderson
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, MO, 65212, USA
| | - Bumsuk Hahm
- Department of Surgery, University of Missouri, Columbia, MO, 65212, USA; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
143
|
Ma R, Ma RQ, Chen B, Wang LY, Fan XY. Compound Cocktail Inhibits Influenza Viral Pneumonia via Phospholipase Cγ1 Phosphorylation-Related Necroptosis and Partial Autophagy in Natural Killer Cells. PLANTA MEDICA 2021; 87:538-549. [PMID: 33545719 DOI: 10.1055/a-1353-6672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Influenza viral infections are prone to global outbreaks and cause pneumonia in affected populations. High morbidity and mortality caused by pneumonia occur during an influenza pandemic. Antivirals or control of inflammation is the primary means of influenza treatment. A compound cocktail composed of arctiin, daidzein, glycyrrhizic acid, and liquiritin inhibited mouse pneumonia resulting from a PR8 viral infection and caused a weight gain after oral administration. Natural killer cell activating receptors, both Ly49D and Ly49H in the lungs, were increased in the treatment in mice. In H3N2 virus-infected natural killer-92MI cells, the cocktail treatment had different effects on phosphorylation sites of phospholipase Cγ1 (PLCγ1) and killed infected cells through necroptosis or late apoptosis, in which RIP3 was increased and both caspase-3 and phosphorylated-JNK in the cells were downregulated. Acid phosphatase activity in viral-infected natural killer-92MI cells was induced by the compound cocktail treatment, which could be related to the p62 decrease in natural killer-92MI cells. In addition, an autophagic flux induction was observed in alveolar basal epithelial cells (A549). Protein p65, but not phosphorylated-p65, was significantly decreased by the treatment. Our results indicate that the compound cocktail strengthened the phosphorylation of PLCγ1-related necroptosis and partial autophagy in natural killer cells, which could yield an inhibitory effect on viral pneumonia in influenza.
Collapse
Affiliation(s)
- Rong Ma
- Clinical Research Institute of Integrative Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui-Qing Ma
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Bei Chen
- Department of Clinical Pharmacy, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Li-Yu Wang
- Oncology Department, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
144
|
Zhao H, Jiang S, Ye Z, Zhu H, Hu B, Meng P, Hu Y, Zhang H, Wang K, Wang J, Tian Y. Discovery of hydrazide-containing oseltamivir analogues as potent inhibitors of influenza A neuraminidase. Eur J Med Chem 2021; 221:113567. [PMID: 34082224 DOI: 10.1016/j.ejmech.2021.113567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 01/13/2023]
Abstract
Neuraminidase (NA) inhibitors play a prime role in treating influenza. However, a variety of viruses containing mutant NAs have developed severe drug resistance towards NA inhibitors, so it is of crucial significance to solve this problem. Encouraged by urea-containing compound 12 disclosed by our lab, we designed a series of oseltamivir derivatives bearing hydrazide fragment for targeting the 150 cavity. Among the synthesized compounds, compound 17a showed 8.77-fold, 4.12-fold, 203-fold and 6.23-fold more potent activity than oseltamivir carboxylate against NAs from H5N1, H1N1, H5N1-H274Y, H1N1-H274Y, respectively. Meanwhile, the best compound 17a exhibited satisfactory metabolic stability in vitro. This study offers an important reference for the structural optimization of oseltamivir aiming at potent inhibition against H274Y mutant of NAs.
Collapse
Affiliation(s)
- Hongqian Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Siyuan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Zhifan Ye
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Hongxi Zhu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Baichun Hu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Peipei Meng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Huicong Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China.
| | - Kuanglei Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China.
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.
| | - Yongshou Tian
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China.
| |
Collapse
|
145
|
Shyr ZA, Cheng YS, Lo DC, Zheng W. Drug combination therapy for emerging viral diseases. Drug Discov Today 2021; 26:2367-2376. [PMID: 34023496 PMCID: PMC8139175 DOI: 10.1016/j.drudis.2021.05.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/14/2021] [Accepted: 05/16/2021] [Indexed: 12/17/2022]
Abstract
Effective therapeutics to combat emerging viral infections are an unmet need. Historically, treatments for chronic viral infections with single drugs have not been successful, as exemplified by human immunodeficiency virus (HIV) and hepatitis C virus (HCV) infections. Combination therapy for these diseases has led to improved clinical outcomes with dramatic reductions in viral load, morbidity, and mortality. Drug combinations can enhance therapeutic efficacy through additive, and ideally synergistic, effects for emerging and re-emerging viruses, such as influenza, severe acute respiratory syndrome-coronavirus (SARS-CoV), Middle East respiratory syndrome (MERS)-CoV, Ebola, Zika, and SARS-coronavirus 2 (CoV-2). Although novel drug development through traditional pipelines remains a priority, in the interim, effective synergistic drug candidates could be rapidly identified by drug-repurposing screens, facilitating accelerated paths to clinical testing and potential emergency use authorizations.
Collapse
Affiliation(s)
- Zeenat A Shyr
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| | - Yu-Shan Cheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Donald C Lo
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
146
|
Lin CY, Yang ZS, Wang WH, Urbina AN, Lin YT, Huang JC, Liu FT, Wang SF. The Antiviral Role of Galectins toward Influenza A Virus Infection-An Alternative Strategy for Influenza Therapy. Pharmaceuticals (Basel) 2021; 14:490. [PMID: 34065500 PMCID: PMC8160607 DOI: 10.3390/ph14050490] [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: 04/13/2021] [Revised: 05/19/2021] [Accepted: 05/19/2021] [Indexed: 12/27/2022] Open
Abstract
Animal lectins are proteins with carbohydrate recognition activity. Galectins, the β-galactoside binding lectins, are expressed in various cells and have been reported to regulate several immunological and physiological responses. Recently, some galectins have been reported to regulate some viral infections, including influenza A virus (IAV); however, the mechanism is still not fully understood. Thus, we aim to review systemically the roles of galectins in their antiviral functions against IAVs. The PRISMA guidelines were used to select the eligible articles. Results indicated that only Galectin-1, Galectin-3, and Galectin-9 were reported to play a regulatory role in IAV infection. These regulatory effects occur extracellularly, through their carbohydrate recognition domain (CRD) interacting with glycans expressed on the virus surface, as well as endogenously, in a cell-cell interaction manner. The inhibition effects induced by galectins on IAV infection were through blocking virus-host receptors interaction, activation of NLRP-3 inflammasome, augment expression of antiviral genes and related cytokines, as well as stimulation of Tim-3 related signaling to enhance virus-specific T cells and humoral immune response. Combined, this study concludes that currently, only three galectins have reported antiviral capabilities against IAV infection, thereby having the potential to be applied as an alternative anti-influenza therapeutic strategy.
Collapse
Affiliation(s)
- Chih-Yen Lin
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-Y.L.); (Z.-S.Y.); (W.-H.W.); (A.N.U.)
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Zih-Syuan Yang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-Y.L.); (Z.-S.Y.); (W.-H.W.); (A.N.U.)
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Wen-Hung Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-Y.L.); (Z.-S.Y.); (W.-H.W.); (A.N.U.)
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Aspiro Nayim Urbina
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-Y.L.); (Z.-S.Y.); (W.-H.W.); (A.N.U.)
| | - Yu-Ting Lin
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Jason C. Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112304, Taiwan;
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan;
| | - Sheng-Fan Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-Y.L.); (Z.-S.Y.); (W.-H.W.); (A.N.U.)
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
147
|
K SD, Puranik R, N S, K K, Fathima F, K R A, Joseph A, J A, Arunkumar G, Mudgal PP. Structure-based identification of small molecules against influenza A virus endonuclease: an in silico and in vitro approach. Pathog Dis 2021; 78:5866476. [PMID: 32614388 DOI: 10.1093/femspd/ftaa032] [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: 04/02/2020] [Accepted: 06/30/2020] [Indexed: 11/14/2022] Open
Abstract
Influenza viruses are known to cause acute respiratory illness, sometimes leading to high mortality rates. Though there are approved influenza antivirals available, their efficacy has reduced over time, due to the drug resistance crisis. There is a perpetual need for newer and better drugs. Drug screening based on the interaction dynamics with different viral target proteins has been a preferred approach in the antiviral drug discovery process. In this study, the FDA approved drug database was virtually screened with the help of Schrödinger software, to select small molecules exhibiting best interactions with the influenza A virus endonuclease protein. A detailed cytotoxicity profiling was carried out for the two selected compounds, cefepime and dolutegravir, followed by in vitro anti-influenza screening using plaque reduction assay. Cefepime showed no cytotoxicity up to 200 μM, while dolutegravir was non-toxic up to 100 μM in Madin-Darby canine kidney cells. The compounds did not show any reduction in viral plaque numbers indicating no anti-influenza activity. An inefficiency in the translation of the molecular interactions into antiviral activity does not necessarily mean that the molecules were inactive. Nevertheless, testing the molecules for endonuclease inhibition per se can be considered a worthwhile approach.
Collapse
Affiliation(s)
- Sai Disha K
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Rashmi Puranik
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Sudheesh N
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Kavitha K
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Fajeelath Fathima
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Anu K R
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Anitha J
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - G Arunkumar
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Piya Paul Mudgal
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| |
Collapse
|
148
|
Yang ZS, Lin CY, Huang SW, Wang WH, Urbina AN, Tseng SP, Lu PL, Chen YH, Wang SF. Regulatory roles of galectins on influenza A virus and their potential as a therapeutic strategy. Biomed Pharmacother 2021; 139:111713. [PMID: 34243634 DOI: 10.1016/j.biopha.2021.111713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/06/2021] [Indexed: 11/19/2022] Open
Abstract
Galectins, are β-galactoside binding lectins expressed in numerous cells and are known to regulate various immune responses and cellular physiological functions. Galectins have been reported to participate in the regulation of several viral infections via carbohydrate‑dependent/independent manner. Galectins have displayed various regulatory functions on viral infection, however, the detailed mechanism remains unclear. More recently, some members of galectins have been reported to regulate influenza A virus (IAV) infection. In this review, we aim to analyze and summarize current findings regarding the role of galectins in IAV infection and their antiviral potential therapeutic application in the treatment of IAVs. The eligible articles were selected according to the PRISMA guidelines. Results indicate that Galectin-1(Gal-1), Galectin-3(Gal-3) and Galectin-9 (Gal-9) were found as the predominant galectins reported to participate in the regulation of IAVs infection. The inhibitory regulation of IAVs by these galectins occurred mainly through extracellular binding to glycosylated envelope proteins, further blocking the interaction between influenza envelope and sialic acid receptor, interacting with ligands or receptors on immune cells to trigger immunol or cellular response against IAVs, and endogenously interacting cellular components in the cytoplasm to activate inflammasome and autophagy. This study offers information regarding the multiple roles of galectins observed in IAVs infection and suggest that galectins has the potential to be used as therapeutic agents for IAVs.
Collapse
Affiliation(s)
- Zih-Syuan Yang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chih-Yen Lin
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Szu-Wei Huang
- Model Development Section, Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wen-Hung Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Aspiro Nayim Urbina
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Sung-Pin Tseng
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Po-Liang Lu
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yen-Hsu Chen
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Sheng-Fan Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
149
|
Tonk M, Růžek D, Vilcinskas A. Compelling Evidence for the Activity of Antiviral Peptides against SARS-CoV-2. Viruses 2021; 13:v13050912. [PMID: 34069206 PMCID: PMC8156787 DOI: 10.3390/v13050912] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022] Open
Abstract
Multiple outbreaks of epidemic and pandemic viral diseases have occurred in the last 20 years, including those caused by Ebola virus, Zika virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The emergence or re-emergence of such diseases has revealed the deficiency in our pipeline for the discovery and development of antiviral drugs. One promising solution is the extensive library of antimicrobial peptides (AMPs) produced by all eukaryotic organisms. AMPs are widely known for their activity against bacteria, but many possess additional antifungal, antiparasitic, insecticidal, anticancer, or antiviral activities. AMPs could therefore be suitable as leads for the development of new peptide-based antiviral drugs. Sixty therapeutic peptides had been approved by the end of 2018, with at least another 150 in preclinical or clinical development. Peptides undergoing clinical trials include analogs, mimetics, and natural AMPs. The advantages of AMPs include novel mechanisms of action that hinder the evolution of resistance, low molecular weight, low toxicity toward human cells but high specificity and efficacy, the latter enhanced by the optimization of AMP sequences. In this opinion article, we summarize the evidence supporting the efficacy of antiviral AMPs and discuss their potential to treat emerging viral diseases including COVID-19.
Collapse
Affiliation(s)
- Miray Tonk
- Institute for Insect Biotechnology, Justus Liebig University of Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany;
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
| | - Daniel Růžek
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic;
- Biology Centre of the Czech Academy of Sciences, Institute of Parasitology, Branisovska 31, 37005 Ceske Budejovice, Czech Republic
| | - Andreas Vilcinskas
- Institute for Insect Biotechnology, Justus Liebig University of Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany;
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Ohlebergsweg 12, 35392 Giessen, Germany
- Correspondence:
| |
Collapse
|
150
|
Zonker B, Becker J, Hrdina R. Synthesis of noradamantane derivatives by ring-contraction of the adamantane framework. Org Biomol Chem 2021; 19:4027-4031. [PMID: 33978046 DOI: 10.1039/d1ob00471a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We describe a triflic acid promoted cascade reaction of adamantane derivatives consisting of a decarboxylation of N-methyl protected cyclic carbamates and a subsequent intramolecular nucleophilic 1,2-alkyl shift to generate ring contracted iminium triflates. This reaction expands the family of similar transformations, such as Wagner-Meerwein-, Demjanov-Tiffeneau-, Meinwald- or (semi-)pinacol-rearrangement. It allows the preparation of noradamantane derivatives in a few steps, starting from simple hydroxy-substituted adamantane precursors.
Collapse
Affiliation(s)
- Benjamin Zonker
- Institute of Organic Chemistry, Justus-Liebig University, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Jonathan Becker
- Institute of Inorganic and Analytical Chemistry, Justus-Liebig University, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Radim Hrdina
- Department of Organic Chemistry, Faculty of Science, Charles University in Prague, Hlavova 8, 128 40 Praha, Czech Republic.
| |
Collapse
|