1
|
Prashar N, Mohammed SB, Raja NS, Mohideen HS. Rerouting therapeutic peptides and unlocking their potential against SARS-CoV2. 3 Biotech 2025; 15:116. [PMID: 40191455 PMCID: PMC11971104 DOI: 10.1007/s13205-025-04270-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
The COVID-19 pandemic highlighted the potential of peptide-based therapies as an alternative to traditional pharmaceutical treatments for SARS-CoV-2 and its variants. Our review explores the role of therapeutic peptides in modulating immune responses, inhibiting viral entry, and disrupting replication. Despite challenges such as stability, bioavailability, and the rapid mutation of the virus, ongoing research and clinical trials show that peptide-based treatments are increasingly becoming integral to future viral outbreak responses. Advancements in computational modelling methods in combination with artificial intelligence will enable mass screening of therapeutic peptides and thereby, comprehending a peptide repurposing strategy similar to the small molecule repurposing. These findings suggest that peptide-based therapies play a critical and promising role in future pandemic preparedness and outbreak management.
Collapse
Affiliation(s)
- Namrata Prashar
- Bioinformatics and Entomoinformatics Lab, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Chennai, Tamil Nadu 603203 India
| | - Saharuddin Bin Mohammed
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - N. S. Raja
- Deparmtent of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Chennai, Tamil Nadu 603203 India
| | - Habeeb Shaik Mohideen
- Bioinformatics and Entomoinformatics Lab, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Chennai, Tamil Nadu 603203 India
| |
Collapse
|
2
|
Sardar A, Bhowmick S, Kamble M, Dewangan N, Hazra B, Mallick AI, Tarafdar PK. De Novo Design of Lipopeptide-Based β-Sheet-Like Self-Assemblies: A Strategy to Develop Fusion Inhibitors as Broad-Spectrum Antivirals. Chemistry 2025; 31:e202403039. [PMID: 39716967 DOI: 10.1002/chem.202403039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024]
Abstract
The recent surge in emerging viral infections warrants the design of broad-spectrum antivirals. We aim to develop a lead molecule that targets a common biochemical feature of many enveloped viruses, membrane fusion. To achieve the broad-spectrum ability, instead of targeting the fusion machinery, we plan to modulate the physicochemical properties of the host and viral membranes to block fusion. The approach is based on the Coronin-1 protein of Mycobacterium, which presumably inhibits the phagosome-lysosome fusion, and a unique Trp-Asp (WD) sequence is placed at the distorted β-meander motif. We designed a WD-based branched lipopeptide (Myr-D(WD)2) that supported the intermolecular interactions to create a β-sheet-like supramolecular assembly at the membrane surface. TEM and confocal fluorescence experiments also suggest that the lipopeptide self-assembled at the bilayer interface and modulated the interfacial order and the water penetration. We demonstrated that the supramolecular organization of Myr-D(WD)2 could block artificial membrane fusion completely and restrict pH-dependent influenza virus (H1N1, H9N2), and pH-independent mouse hepatitis virus, human coronavirus (HCoV-OC43) infections. The present study provided an evidence-based broad-spectrum antiviral potential of a designed self-assembled lipopeptide.
Collapse
Affiliation(s)
- Avijit Sardar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Sucharita Bhowmick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Mithila Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Nikesh Dewangan
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Bibhas Hazra
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Amirul I Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| | - Pradip K Tarafdar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741 246, India
| |
Collapse
|
3
|
Liang QZ, Ji CM, Wang B, Chen W, Cong F, Huang Y, Huang YW. Deltacoronavirus HKU11, HKU13, PDCoV (HKU15) and HKU17 spike pseudoviruses enter avian DF-1 cells via clathrin-mediated endocytosis in a Rab5-, Rab7- and pH-dependent manner. Vet Res 2025; 56:15. [PMID: 39825424 PMCID: PMC11740469 DOI: 10.1186/s13567-024-01442-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/14/2024] [Indexed: 01/20/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV), also known as HKU15, is a swine enteropathogenic virus that is believed to have originated in birds. PDCoV belongs to the genus Deltacoronavirus (DCoV), the members of which have mostly been identified in diverse avian species. We recently reported that chicken or porcine aminopeptidase N (APN), the major cellular receptor for PDCoV, can mediate cellular entry via three pseudotyped retroviruses displaying spike proteins from three avian DCoVs (HKU11, HKU13, and HKU17). In the present work, to better understand how avian-origin CoVs may be transmitted to pigs, we investigated the unknown DCoV entry pathway in avian cells. We show that clathrin-mediated endocytosis is involved in the entry of these DCoV pseudoviruses into chicken-origin DF-1 cells. Pseudovirus entry was suppressed by means of pharmacological inhibitors, dominant-negative mutants, and siRNAs targeting various cellular proteins and signalling molecules, suggesting that PDCoV and avian DCoV pseudovirus entry into DF-1 cells depends on clathrin, dynamin-2, cathepsins and a low-pH environment but is independent of caveolae and macropinocytosis. Furthermore, we found that DCoV pseudovirus entry was linked to Rab5- and Rab7-dependent pathways. This is the first report demonstrating that these DCoVs utilize clathrin-mediated endocytosis pathways to enter avian-origin cells, providing new insights into interspecies transmission of DCoVs.
Collapse
Affiliation(s)
- Qi-Zhang Liang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Chun-Miao Ji
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Bin Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Wei Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Feng Cong
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China.
| | - Yu Huang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China.
| | - Yao-Wei Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China.
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Mousseau G, Préault N, Souquere S, Bireau C, Cassonnet P, Bacquin A, Beck L, Pierron G, Jacob Y, Dupressoir A, Heidmann T. Sodium-dependent phosphate transporter PiT1/SLC20A1 as the receptor for the endogenous retroviral envelope syncytin-B involved in mouse placenta formation. J Virol 2024; 98:e0091524. [PMID: 39287391 PMCID: PMC11495048 DOI: 10.1128/jvi.00915-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
Syncytins are envelope genes of retroviral origin that play a critical role in the formation of a syncytial structure at the fetomaternal interface via their fusogenic activity. The mouse placenta is unique among placental mammals since the fetomaternal interface comprises two syncytiotrophoblast layers (ST-I and ST-II) instead of one observed in all other hemochorial placentae. Each layer specifically expresses a distinct mouse syncytin, namely syncytin-A (SynA) for ST-I and syncytin-B (SynB) for ST-II, which have been shown to be essential to placentogenesis and embryonic development. The cellular receptor for SynA has been identified as the membrane protein LY6E and is not the receptor for SynB. Here, by combining a cell-cell fusion assay with the screening of a human ORFeome-derived expression library, we identified the transmembrane multipass sodium-dependent phosphate transporter 1 PiT1/SLC20A1 as the receptor for SynB. Transfection of cells with the cloned receptor, but not the closely related PiT2/SLC20A2, leads to their fusion with cells expressing SynB, with no cross-reactive fusion activity with SynA. The interaction between the two partners was further demonstrated by immunoprecipitation. PiT1/PiT2 chimera and truncation experiments identified the PiT1 N-terminus as the major determinant for SynB-mediated fusion. RT-qPCR analysis of PiT1 expression on a panel of mouse adult and fetal tissues revealed a concomitant increase of PiT1 and SynB specifically in the developing placenta. Finally, electron microscopy analysis of the placenta of PiT1 null embryo before they die (E11.5) disclosed default of ST-II formation with lack of syncytialization, as previously observed in cognate SynB null placenta, and consistent with the present identification of PiT1 as the SynB partner.IMPORTANCESyncytins are envelope genes of endogenous retroviruses, coopted for a physiological function in placentation. They are fusogenic proteins that mediate cell-cell fusion by interacting with receptors present on the partner cells. Here, by devising an in vitro fusion assay that enables the screening of an ORFeome-derived expression library, we identified the long-sought receptor for syncytin-B (SynB), a mouse syncytin responsible for syncytiotrophoblast formation at the fetomaternal interface of the mouse placenta. This protein - PiT1/SLC20A1 - is a multipass transmembrane protein, also known as the receptor for a series of infectious retroviruses. Its profile of expression is consistent with a role in both ancestral endogenization of a SynB founder retrovirus and present-day mouse placenta formation, with evidence-in PiT1 knockout mice-of unfused cells at the level of the cognate placental syncytiotrophoblast layer.
Collapse
Affiliation(s)
| | - Noémie Préault
- Unité Physiologie et Pathologie Moléculaires des Rétrovirus Endogènes et Infectieux, CNRS UMR 9196, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Sylvie Souquere
- AMMICA UMS-3655, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Caroline Bireau
- Unité Physiologie et Pathologie Moléculaires des Rétrovirus Endogènes et Infectieux, CNRS UMR 9196, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Patricia Cassonnet
- Unité de Génétique Moléculaire des Virus à ARN, Département Virologie, Institut Pasteur, Paris, France
- UMR3569, Centre National de la Recherche Scientifique, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | - Laurent Beck
- Nantes Université, CNRS, Inserm, l'Institut du Thorax, Nantes, France
| | - Gérard Pierron
- Unité Physiologie et Pathologie Moléculaires des Rétrovirus Endogènes et Infectieux, CNRS UMR 9196, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Yves Jacob
- Unité de Génétique Moléculaire des Virus à ARN, Département Virologie, Institut Pasteur, Paris, France
- UMR3569, Centre National de la Recherche Scientifique, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute (DFCI), Boston, Massachusetts, USA
| | - Anne Dupressoir
- Unité Physiologie et Pathologie Moléculaires des Rétrovirus Endogènes et Infectieux, CNRS UMR 9196, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Thierry Heidmann
- Viroxis, Institut Gustave Roussy, Villejuif, France
- Unité Physiologie et Pathologie Moléculaires des Rétrovirus Endogènes et Infectieux, CNRS UMR 9196, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
5
|
Shekunov EV, Efimova SS, Kever LV, Ishmanov TF, Ostroumova OS. Lipid Selectivity of Membrane Action of the Fragments of Fusion Peptides of Marburg and Ebola Viruses. Int J Mol Sci 2024; 25:9901. [PMID: 39337389 PMCID: PMC11432738 DOI: 10.3390/ijms25189901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The life cycle of Ebola and Marburg viruses includes a step of the virion envelope fusion with the cell membrane. Here, we analyzed whether the fusion of liposome membranes under the action of fragments of fusion peptides of Ebola and Marburg viruses depends on the composition of lipid vesicles. A fluorescence assay and electron microscopy were used to quantify the fusogenic activity of the virus fusion peptides and to identify the lipid determinants affecting membrane merging. Differential scanning calorimetry of lipid phase transitions revealed alterations in the physical properties of the lipid matrix produced by virus fusion peptides. Additionally, we found that plant polyphenols, quercetin, and myricetin inhibited vesicle fusion induced by the Marburg virus fusion peptide.
Collapse
Affiliation(s)
- Egor V Shekunov
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Svetlana S Efimova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Lyudmila V Kever
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Tagir F Ishmanov
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Olga S Ostroumova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| |
Collapse
|
6
|
McGraw A, Hillmer G, Medehincu SM, Hikichi Y, Gagliardi S, Narayan K, Tibebe H, Marquez D, Mei Bose L, Keating A, Izumi C, Peese K, Joshi S, Krystal M, DeCicco-Skinner KL, Freed EO, Sardo L, Izumi T. Exploring HIV-1 Maturation: A New Frontier in Antiviral Development. Viruses 2024; 16:1423. [PMID: 39339899 PMCID: PMC11437483 DOI: 10.3390/v16091423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
HIV-1 virion maturation is an essential step in the viral replication cycle to produce infectious virus particles. Gag and Gag-Pol polyproteins are assembled at the plasma membrane of the virus-producer cells and bud from it to the extracellular compartment. The newly released progeny virions are initially immature and noninfectious. However, once the Gag polyprotein is cleaved by the viral protease in progeny virions, the mature capsid proteins assemble to form the fullerene core. This core, harboring two copies of viral genomic RNA, transforms the virion morphology into infectious virus particles. This morphological transformation is referred to as maturation. Virion maturation influences the distribution of the Env glycoprotein on the virion surface and induces conformational changes necessary for the subsequent interaction with the CD4 receptor. Several host factors, including proteins like cyclophilin A, metabolites such as IP6, and lipid rafts containing sphingomyelins, have been demonstrated to have an influence on virion maturation. This review article delves into the processes of virus maturation and Env glycoprotein recruitment, with an emphasis on the role of host cell factors and environmental conditions. Additionally, we discuss microscopic technologies for assessing virion maturation and the development of current antivirals specifically targeting this critical step in viral replication, offering long-acting therapeutic options.
Collapse
Affiliation(s)
- Aidan McGraw
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Grace Hillmer
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Stefania M. Medehincu
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Yuta Hikichi
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MS 21702, USA; (Y.H.); (E.O.F.)
| | - Sophia Gagliardi
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Kedhar Narayan
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Hasset Tibebe
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Dacia Marquez
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Lilia Mei Bose
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Adleigh Keating
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Coco Izumi
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Kevin Peese
- ViiV Healthcare, 36 E. Industrial Road, Branford, CT 06405, USA; (K.P.) (S.J.); (M.K.)
| | - Samit Joshi
- ViiV Healthcare, 36 E. Industrial Road, Branford, CT 06405, USA; (K.P.) (S.J.); (M.K.)
| | - Mark Krystal
- ViiV Healthcare, 36 E. Industrial Road, Branford, CT 06405, USA; (K.P.) (S.J.); (M.K.)
| | - Kathleen L. DeCicco-Skinner
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
| | - Eric O. Freed
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MS 21702, USA; (Y.H.); (E.O.F.)
| | - Luca Sardo
- ViiV Healthcare, 36 E. Industrial Road, Branford, CT 06405, USA; (K.P.) (S.J.); (M.K.)
| | - Taisuke Izumi
- Department Biology, College of Arts and Sciences, American University, Washington, DC 20016, USA; (A.M.); (G.H.); (S.M.M.); (S.G.); (K.N.); (H.T.); (D.M.); (L.M.B.); (A.K.); (C.I.); (K.L.D.-S.)
- District of Columbia Center for AIDS Research, Washington, DC 20052, USA
| |
Collapse
|
7
|
Santisteban Celis IC, Matoba N. Lectibodies as antivirals. Antiviral Res 2024; 227:105901. [PMID: 38734211 DOI: 10.1016/j.antiviral.2024.105901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Growing concerns regarding the emergence of highly transmissible viral diseases highlight the urgent need to expand the repertoire of antiviral therapeutics. For this reason, new strategies for neutralizing and inhibiting these viruses are necessary. A promising approach involves targeting the glycans present on the surfaces of enveloped viruses. Lectins, known for their ability to recognize specific carbohydrate molecules, offer the potential for glycan-targeted antiviral strategies. Indeed, numerous studies have reported the antiviral effects of various lectins of both endogenous and exogenous origins. However, many lectins in their natural forms, are not suitable for use as antiviral therapeutics due to toxicity, other unfavorable pharmacological effects, and/or unreliable manufacturing sources. Therefore, improvements are crucial for employing lectins as effective antiviral therapeutics. A novel approach to enhance lectins' suitability as pharmaceuticals could be the generation of recombinant lectin-Fc fusion proteins, termed "lectibodies." In this review, we discuss the scientific rationale behind lectin-based antiviral strategies and explore how lectibodies could facilitate the development of new antiviral therapeutics. We will also share our perspective on the potential of these molecules to transcend their potential use as antiviral agents.
Collapse
Affiliation(s)
- Ian Carlosalberto Santisteban Celis
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA; Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville School of Medicine, Louisville, KY, USA
| | - Nobuyuki Matoba
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA; Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville School of Medicine, Louisville, KY, USA; UofL Health - Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
8
|
Shi Q, Zhao R, Chen L, Liu T, Di T, Zhang C, Zhang Z, Wang F, Han Z, Sun J, Liu S. Newcastle disease virus activates diverse signaling pathways via Src to facilitate virus entry into host macrophages. J Virol 2024; 98:e0191523. [PMID: 38334327 PMCID: PMC10949470 DOI: 10.1128/jvi.01915-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/27/2023] [Indexed: 02/10/2024] Open
Abstract
As an intrinsic cellular mechanism responsible for the internalization of extracellular ligands and membrane components, caveolae-mediated endocytosis (CavME) is also exploited by certain pathogens for endocytic entry [e.g., Newcastle disease virus (NDV) of paramyxovirus]. However, the molecular mechanisms of NDV-induced CavME remain poorly understood. Herein, we demonstrate that sialic acid-containing gangliosides, rather than glycoproteins, were utilized by NDV as receptors to initiate the endocytic entry of NDV into HD11 cells. The binding of NDV to gangliosides induced the activation of a non-receptor tyrosine kinase, Src, leading to the phosphorylation of caveolin-1 (Cav1) and dynamin-2 (Dyn2), which contributed to the endocytic entry of NDV. Moreover, an inoculation of cells with NDV-induced actin cytoskeletal rearrangement through Src to facilitate NDV entry via endocytosis and direct fusion with the plasma membrane. Subsequently, unique members of the Rho GTPases family, RhoA and Cdc42, were activated by NDV in a Src-dependent manner. Further analyses revealed that RhoA and Cdc42 regulated the activities of specific effectors, cofilin and myosin regulatory light chain 2, responsible for actin cytoskeleton rearrangement, through diverse intracellular signaling cascades. Taken together, our results suggest that an inoculation of NDV-induced Src-mediated cellular activation by binding to ganglioside receptors. This process orchestrated NDV endocytic entry by modulating the activities of caveolae-associated Cav1 and Dyn2, as well as specific Rho GTPases and downstream effectors. IMPORTANCE In general, it is known that the paramyxovirus gains access to host cells through direct penetration at the plasma membrane; however, emerging evidence suggests more complex entry mechanisms for paramyxoviruses. The endocytic entry of Newcastle disease virus (NDV), a representative member of the paramyxovirus family, into multiple types of cells has been recently reported. Herein, we demonstrate the binding of NDV to induce ganglioside-activated Src signaling, which is responsible for the endocytic entry of NDV through caveolae-mediated endocytosis. This process involved Src-dependent activation of the caveolae-associated Cav1 and Dyn2, as well as specific Rho GTPase and downstream effectors, thereby orchestrating the endocytic entry process of NDV. Our findings uncover a novel molecular mechanism of endocytic entry of NDV into host cells and provide novel insight into paramyxovirus mechanisms of entry.
Collapse
Affiliation(s)
- Qiankai Shi
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ran Zhao
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Linna Chen
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tianyi Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tao Di
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chunwei Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhiying Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Fangfang Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zongxi Han
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Junfeng Sun
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shengwang Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
9
|
Yaacoub C, Wehbe R, Roufayel R, Fajloun Z, Coutard B. Bee Venom and Its Two Main Components-Melittin and Phospholipase A2-As Promising Antiviral Drug Candidates. Pathogens 2023; 12:1354. [PMID: 38003818 PMCID: PMC10674158 DOI: 10.3390/pathogens12111354] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/08/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Viruses are known to infect most types of organisms. In humans, they can cause several diseases that range from mild to severe. Although many antiviral therapies have been developed, viral infections continue to be a leading cause of morbidity and mortality worldwide. Therefore, the discovery of new and effective antiviral agents is desperately needed. Animal venoms are a rich source of bioactive molecules found in natural goods that have been used since ancient times in alternative medicine to treat a variety of human diseases. Recently, and with the onset of the COVID-19 pandemic, scientists have regained their interest in the possible use of natural products, such as bee venom (BV), as a potential antiviral agent to treat viral infections. BV is known to exert many therapeutic activities such as anti-proliferative, anti-bacterial, and anti-inflammatory effects. However, there is limited discussion of the antiviral activity of BV in the literature. Therefore, this review aims to highlight the antiviral properties of BV and its two primary constituents, melittin (MEL) and phospholipase A2 (PLA2), against a variety of enveloped and non-enveloped viruses. Finally, the innovative strategies used to reduce the toxicity of BV and its two compounds for the development of new antiviral treatments are also considered.
Collapse
Affiliation(s)
- Carole Yaacoub
- Unité des Virus Emergents, Aix-Marseille University, IRD 190-Inserm 1207, IHU Méditerranée Infection, 13005 Marseille, France;
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and Its Applications, Doctoral School for Sciences and Technology, Lebanese University, Tripoli 1300, Lebanon;
| | - Rim Wehbe
- Biology Department, Faculty of Arts and Sciences, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Rabih Roufayel
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait;
| | - Ziad Fajloun
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and Its Applications, Doctoral School for Sciences and Technology, Lebanese University, Tripoli 1300, Lebanon;
- Faculty of Sciences III, Department of Biology, Michel Slayman Tripoli Campus, Lebanese University, Tripoli 1352, Lebanon
| | - Bruno Coutard
- Unité des Virus Emergents, Aix-Marseille University, IRD 190-Inserm 1207, IHU Méditerranée Infection, 13005 Marseille, France;
| |
Collapse
|
10
|
Letoha A, Hudák A, Letoha T. Exploring the Syndecan-Mediated Cellular Internalization of the SARS-CoV-2 Omicron Variant. Int J Mol Sci 2023; 24:14140. [PMID: 37762442 PMCID: PMC10531417 DOI: 10.3390/ijms241814140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
SARS-CoV-2 variants evolve to rely more on heparan sulfate (HS) for viral attachment and subsequent infection. In our earlier work, we demonstrated that the Delta variant's spike protein binds more strongly to HS compared to WT SARS-CoV-2, leading to enhanced cell internalization via syndecans (SDCs), a family of transmembrane HS proteoglycans (HSPGs) facilitating the cellular entry of the original strain. Using our previously established ACE2- or SDC-overexpressing cellular models, we now compare the ACE2- and SDC-dependent cellular uptake of heat-inactivated WT SARS-CoV-2 with the Delta and Omicron variants. Internalization studies with inactivated virus particles showed that ACE2 overexpression could not compensate for the loss of HS in Omicron's internalization, suggesting that this variant primarily uses HSPGs to enter cells. Although SDCs increased the internalization of all three viruses, subtle differences could be detected between their SDC isoform preferences. The Delta variant particularly benefitted from SDC1, 2, and 4 overexpression for cellular entry, while SDC4 had the most prominent effect on Omicron internalization. The SDC4 knockdown (KD) in Calu-3 cells reduced the cellular uptake of all three viruses, but the inhibition was the most pronounced for Omicron. The polyanionic heparin also hindered the cellular internalization of all three viruses with a dominant inhibitory effect on Omicron. Omicron's predominant HSPG affinity, combined with its preference for the universally expressed SDC4, might account for its efficient transmission yet reduced pathogenicity.
Collapse
Affiliation(s)
- Annamária Letoha
- Department of Medicine, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | | | | |
Collapse
|
11
|
Targeting Human Proteins for Antiviral Drug Discovery and Repurposing Efforts: A Focus on Protein Kinases. Viruses 2023; 15:v15020568. [PMID: 36851782 PMCID: PMC9966946 DOI: 10.3390/v15020568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Despite the great technological and medical advances in fighting viral diseases, new therapies for most of them are still lacking, and existing antivirals suffer from major limitations regarding drug resistance and a limited spectrum of activity. In fact, most approved antivirals are directly acting antiviral (DAA) drugs, which interfere with viral proteins and confer great selectivity towards their viral targets but suffer from resistance and limited spectrum. Nowadays, host-targeted antivirals (HTAs) are on the rise, in the drug discovery and development pipelines, in academia and in the pharmaceutical industry. These drugs target host proteins involved in the virus life cycle and are considered promising alternatives to DAAs due to their broader spectrum and lower potential for resistance. Herein, we discuss an important class of HTAs that modulate signal transduction pathways by targeting host kinases. Kinases are considered key enzymes that control virus-host interactions. We also provide a synopsis of the antiviral drug discovery and development pipeline detailing antiviral kinase targets, drug types, therapeutic classes for repurposed drugs, and top developing organizations. Furthermore, we detail the drug design and repurposing considerations, as well as the limitations and challenges, for kinase-targeted antivirals, including the choice of the binding sites, physicochemical properties, and drug combinations.
Collapse
|
12
|
Smith J, Alfieri JM, Anthony N, Arensburger P, Athrey GN, Balacco J, Balic A, Bardou P, Barela P, Bigot Y, Blackmon H, Borodin PM, Carroll R, Casono MC, Charles M, Cheng H, Chiodi M, Cigan L, Coghill LM, Crooijmans R, Das N, Davey S, Davidian A, Degalez F, Dekkers JM, Derks M, Diack AB, Djikeng A, Drechsler Y, Dyomin A, Fedrigo O, Fiddaman SR, Formenti G, Frantz LA, Fulton JE, Gaginskaya E, Galkina S, Gallardo RA, Geibel J, Gheyas AA, Godinez CJP, Goodell A, Graves JA, Griffin DK, Haase B, Han JL, Hanotte O, Henderson LJ, Hou ZC, Howe K, Huynh L, Ilatsia E, Jarvis ED, Johnson SM, Kaufman J, Kelly T, Kemp S, Kern C, Keroack JH, Klopp C, Lagarrigue S, Lamont SJ, Lange M, Lanke A, Larkin DM, Larson G, Layos JKN, Lebrasseur O, Malinovskaya LP, Martin RJ, Martin Cerezo ML, Mason AS, McCarthy FM, McGrew MJ, Mountcastle J, Muhonja CK, Muir W, Muret K, Murphy TD, Ng'ang'a I, Nishibori M, O'Connor RE, Ogugo M, Okimoto R, Ouko O, Patel HR, Perini F, Pigozzi MI, Potter KC, Price PD, Reimer C, Rice ES, Rocos N, Rogers TF, Saelao P, Schauer J, Schnabel RD, Schneider VA, Simianer H, Smith A, et alSmith J, Alfieri JM, Anthony N, Arensburger P, Athrey GN, Balacco J, Balic A, Bardou P, Barela P, Bigot Y, Blackmon H, Borodin PM, Carroll R, Casono MC, Charles M, Cheng H, Chiodi M, Cigan L, Coghill LM, Crooijmans R, Das N, Davey S, Davidian A, Degalez F, Dekkers JM, Derks M, Diack AB, Djikeng A, Drechsler Y, Dyomin A, Fedrigo O, Fiddaman SR, Formenti G, Frantz LA, Fulton JE, Gaginskaya E, Galkina S, Gallardo RA, Geibel J, Gheyas AA, Godinez CJP, Goodell A, Graves JA, Griffin DK, Haase B, Han JL, Hanotte O, Henderson LJ, Hou ZC, Howe K, Huynh L, Ilatsia E, Jarvis ED, Johnson SM, Kaufman J, Kelly T, Kemp S, Kern C, Keroack JH, Klopp C, Lagarrigue S, Lamont SJ, Lange M, Lanke A, Larkin DM, Larson G, Layos JKN, Lebrasseur O, Malinovskaya LP, Martin RJ, Martin Cerezo ML, Mason AS, McCarthy FM, McGrew MJ, Mountcastle J, Muhonja CK, Muir W, Muret K, Murphy TD, Ng'ang'a I, Nishibori M, O'Connor RE, Ogugo M, Okimoto R, Ouko O, Patel HR, Perini F, Pigozzi MI, Potter KC, Price PD, Reimer C, Rice ES, Rocos N, Rogers TF, Saelao P, Schauer J, Schnabel RD, Schneider VA, Simianer H, Smith A, Stevens MP, Stiers K, Tiambo CK, Tixier-Boichard M, Torgasheva AA, Tracey A, Tregaskes CA, Vervelde L, Wang Y, Warren WC, Waters PD, Webb D, Weigend S, Wolc A, Wright AE, Wright D, Wu Z, Yamagata M, Yang C, Yin ZT, Young MC, Zhang G, Zhao B, Zhou H. Fourth Report on Chicken Genes and Chromosomes 2022. Cytogenet Genome Res 2023; 162:405-528. [PMID: 36716736 PMCID: PMC11835228 DOI: 10.1159/000529376] [Show More Authors] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/22/2023] [Indexed: 02/01/2023] Open
Affiliation(s)
- Jacqueline Smith
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - James M. Alfieri
- Interdisciplinary Program in Ecology and Evolutionary Biology, Texas A&M University, College Station, Texas, USA
- Department of Biology, Texas A&M University, College Station, Texas, USA
- Department of Poultry Science, Texas A&M University, College Station, Texas, USA
| | | | - Peter Arensburger
- Biological Sciences Department, California State Polytechnic University, Pomona, California, USA
| | - Giridhar N. Athrey
- Interdisciplinary Program in Ecology and Evolutionary Biology, Texas A&M University, College Station, Texas, USA
- Department of Poultry Science, Texas A&M University, College Station, Texas, USA
| | | | - Adam Balic
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Philippe Bardou
- Université de Toulouse, INRAE, ENVT, GenPhySE, Sigenae, Castanet Tolosan, France
| | | | - Yves Bigot
- PRC, UMR INRAE 0085, CNRS 7247, Centre INRAE Val de Loire, Nouzilly, France
| | - Heath Blackmon
- Interdisciplinary Program in Ecology and Evolutionary Biology, Texas A&M University, College Station, Texas, USA
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Pavel M. Borodin
- Department of Molecular Genetics, Cell Biology and Bioinformatics, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Rachel Carroll
- Department of Animal Sciences, Data Science and Informatics Institute, University of Missouri, Columbia, Missouri, USA
| | | | - Mathieu Charles
- University Paris-Saclay, INRAE, AgroParisTech, GABI, Sigenae, Jouy-en-Josas, France
| | - Hans Cheng
- USDA, ARS, USNPRC, Avian Disease and Oncology Laboratory, East Lansing, Michigan, USA
| | | | | | - Lyndon M. Coghill
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
| | - Richard Crooijmans
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
| | | | - Sean Davey
- University of Arizona, Tucson, Arizona, USA
| | - Asya Davidian
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Fabien Degalez
- Centre for Tropical Livestock Genetics and Health (CTLGH) − ILRI, Nairobi, Kenya
| | - Jack M. Dekkers
- Department of Animal Science, University of California, Davis, California, USA
- INRAE, MIAT UR875, Sigenae, Castanet Tolosan, France
| | - Martijn Derks
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Abigail B. Diack
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Appolinaire Djikeng
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | | | - Alexander Dyomin
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | | | | | | | - Laurent A.F. Frantz
- Queen Mary University of London, Bethnal Green, London, UK
- Palaeogenomics Group, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Janet E. Fulton
- Hy-Line International, Research and Development, Dallas Center, Iowa, USA
| | - Elena Gaginskaya
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Svetlana Galkina
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Rodrigo A. Gallardo
- School of Veterinary Medicine, University of California, Davis, California, USA
- Department of Animal Science, University of California, Davis, California, USA
| | - Johannes Geibel
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
- Center for Integrated Breeding Research, University of Göttingen, Göttingen, Germany
| | - Almas A. Gheyas
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Cyrill John P. Godinez
- Department of Animal Science, College of Agriculture and Food Science, Visayas State University, Baybay City, Philippines
| | | | - Jennifer A.M. Graves
- Department of Environment and Genetics, La Trobe University, Melbourne, Victoria, Australia
- Institute for Applied Ecology, University of Canberra, Canberra, Australian Capital Territory, Australia
| | | | | | - Jian-Lin Han
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| | - Olivier Hanotte
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
- Cells, Organisms and Molecular Genetics, School of Life Sciences, University of Nottingham, Nottingham, UK
- Centre for Tropical Livestock Genetics and Health, The Roslin Institute, Edinburgh, UK
| | - Lindsay J. Henderson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Zhuo-Cheng Hou
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | | | - Lan Huynh
- Institute for Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Evans Ilatsia
- Dairy Research Institute, Kenya Agricultural and Livestock Organization, Naivasha, Kenya
| | | | | | - Jim Kaufman
- Institute for Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Terra Kelly
- School of Veterinary Medicine, University of California, Davis, California, USA
- Department of Animal Science, University of California, Davis, California, USA
| | - Steve Kemp
- INRAE, INSTITUT AGRO, PEGASE UMR 1348, Saint-Gilles, France
| | - Colin Kern
- Feed the Future Innovation Lab for Genomics to Improve Poultry, University of California, Davis, California, USA
| | | | - Christophe Klopp
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Sandrine Lagarrigue
- Centre for Tropical Livestock Genetics and Health (CTLGH) − ILRI, Nairobi, Kenya
| | - Susan J. Lamont
- Department of Animal Science, University of California, Davis, California, USA
- INRAE, MIAT UR875, Sigenae, Castanet Tolosan, France
| | - Margaret Lange
- Centre for Tropical Livestock Genetics and Health (CTLGH) − The Roslin Institute, Edinburgh, UK
| | - Anika Lanke
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, California, USA
| | - Denis M. Larkin
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, UK
| | - Greger Larson
- The Palaeogenomics and Bio-Archaeology Research Network, Research Laboratory for Archaeology and History of Art, The University of Oxford, Oxford, UK
| | - John King N. Layos
- College of Agriculture and Forestry, Capiz State University, Mambusao, Philippines
| | - Ophélie Lebrasseur
- Centre d'Anthropobiologie et de Génomique de Toulouse (CAGT), CNRS UMR 5288, Université Toulouse III Paul Sabatier, Toulouse, France
- Instituto Nacional de Antropología y Pensamiento Latinoamericano, Ciudad Autónoma de Buenos Aires, Argentina
| | - Lyubov P. Malinovskaya
- Department of Cytology and Genetics, Novosibirsk State University, Novosibirsk, Russian Federation
| | - Rebecca J. Martin
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | | | | | | | - Michael J. McGrew
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | | | - Christine Kamidi Muhonja
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
- Centre for Tropical Livestock Genetics and Health (CTLGH) − ILRI, Nairobi, Kenya
| | - William Muir
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Kévin Muret
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, Evry, France
| | - Terence D. Murphy
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Masahide Nishibori
- Laboratory of Animal Genetics, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | | | - Moses Ogugo
- Centre for Tropical Livestock Genetics and Health (CTLGH) − ILRI, Nairobi, Kenya
| | - Ron Okimoto
- Cobb-Vantress, Siloam Springs, Arkansas, USA
| | - Ochieng Ouko
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
| | - Hardip R. Patel
- The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Francesco Perini
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
- Department of Agricultural, Food and Environmental Sciences, University of Perugia, Perugia, Italy
| | - María Ines Pigozzi
- INBIOMED (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Peter D. Price
- Ecology and Evolutionary Biology, School of Biosciences, University of Sheffield, Sheffield, UK
| | - Christian Reimer
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Edward S. Rice
- Department of Animal Sciences, Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Nicolas Rocos
- USDA, ARS, USNPRC, Avian Disease and Oncology Laboratory, East Lansing, Michigan, USA
| | - Thea F. Rogers
- Department of Molecular Evolution and Development, University of Vienna, Vienna, Austria
| | - Perot Saelao
- Department of Animal Science, University of California, Davis, California, USA
- Veterinary Pest Genetics Research Unit, USDA, Kerrville, Texas, USA
| | - Jens Schauer
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Robert D. Schnabel
- Department of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Valerie A. Schneider
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Henner Simianer
- Center for Integrated Breeding Research, University of Göttingen, Göttingen, Germany
| | - Adrian Smith
- Department of Zoology, University of Oxford, Oxford, UK
| | - Mark P. Stevens
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Kyle Stiers
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
| | | | | | - Anna A. Torgasheva
- Department of Molecular Genetics, Cell Biology and Bioinformatics, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Alan Tracey
- University Paris-Saclay, INRAE, AgroParisTech, GABI, Sigenae, Jouy-en-Josas, France
| | - Clive A. Tregaskes
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Lonneke Vervelde
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Ying Wang
- Department of Animal Science, University of California, Davis, California, USA
| | - Wesley C. Warren
- Department of Animal Sciences, Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- Department of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Paul D. Waters
- School of Biotechnology and Biomolecular Science, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - David Webb
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Steffen Weigend
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
- Center for Integrated Breeding Research, University of Göttingen, Göttingen, Germany
| | - Anna Wolc
- INRAE, MIAT UR875, Sigenae, Castanet Tolosan, France
- Hy-Line International, Research and Development, Dallas Center, Iowa, USA
| | - Alison E. Wright
- Ecology and Evolutionary Biology, School of Biosciences, University of Sheffield, Sheffield, UK
| | - Dominic Wright
- AVIAN Behavioural Genomics and Physiology, IFM Biology, Linköping University, Linköping, Sweden
| | - Zhou Wu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Masahito Yamagata
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
| | | | - Zhong-Tao Yin
- Department of Animal Sciences, Data Science and Informatics Institute, University of Missouri, Columbia, Missouri, USA
| | | | - Guojie Zhang
- Center for Evolutionary and Organismal Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bingru Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, California, USA
| |
Collapse
|
13
|
Matveeva O, Nechipurenko Y, Lagutkin D, Yegorov YE, Kzhyshkowska J. SARS-CoV-2 infection of phagocytic immune cells and COVID-19 pathology: Antibody-dependent as well as independent cell entry. Front Immunol 2022; 13:1050478. [PMID: 36532011 PMCID: PMC9751203 DOI: 10.3389/fimmu.2022.1050478] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
Our review summarizes the evidence that COVID-19 can be complicated by SARS-CoV-2 infection of immune cells. This evidence is widespread and accumulating at an increasing rate. Research teams from around the world, studying primary and established cell cultures, animal models, and analyzing autopsy material from COVID-19 deceased patients, are seeing the same thing, namely that some immune cells are infected or capable of being infected with the virus. Human cells most vulnerable to infection include both professional phagocytes, such as monocytes, macrophages, and dendritic cells, as well as nonprofessional phagocytes, such as B-cells. Convincing evidence has accumulated to suggest that the virus can infect monocytes and macrophages, while data on infection of dendritic cells and B-cells are still scarce. Viral infection of immune cells can occur directly through cell receptors, but it can also be mediated or enhanced by antibodies through the Fc gamma receptors of phagocytic cells. Antibody-dependent enhancement (ADE) most likely occurs during the primary encounter with the pathogen through the first COVID-19 infection rather than during the second encounter, which is characteristic of ADE caused by other viruses. Highly fucosylated antibodies of vaccinees seems to be incapable of causing ADE, whereas afucosylated antibodies of persons with acute primary infection or convalescents are capable. SARS-CoV-2 entry into immune cells can lead to an abortive infection followed by host cell pyroptosis, and a massive inflammatory cascade. This scenario has the most experimental evidence. Other scenarios are also possible, for which the evidence base is not yet as extensive, namely productive infection of immune cells or trans-infection of other non-immune permissive cells. The chance of a latent infection cannot be ruled out either.
Collapse
Affiliation(s)
- Olga Matveeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Denis Lagutkin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Center of Phthisiopulmonology and Infectious Diseases under the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yegor E. Yegorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- German Red Cross Blood Service Baden-Württemberg – Hessen, Mannheim, Germany
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| |
Collapse
|
14
|
Wang Y, Zhang Y, Zuo W, Bo Z, Zhang C, Zhang X, Wu Y. Avian Reovirus σB Interacts with Caveolin-1 in Lipid Rafts during Dynamin-Dependent Caveolae-Mediated Endocytosis. Viruses 2022; 14:v14102201. [PMID: 36298756 PMCID: PMC9608613 DOI: 10.3390/v14102201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022] Open
Abstract
Caveolin-1 (Cav-1) is the basic component of caveolae, a specialized form of lipid raft that plays an essential role in endocytic viral entry. However, the evidence of direct involvement of caveolae and Cav-1 in avian reovirus (ARV) entry remains insufficient. In this study, the membrane lipid rafts were isolated as detergent-resistant microdomains (DRMs) by sucrose gradient centrifugation, and the capsid protein σB of ARV was found to associate with Cav-1 in DRMs fractions. Additionally, the interaction between ARV σB protein and Cav-1 was demonstrated by immunofluorescence co-localization and co-immunoprecipitation assays. Furthermore, we found that the internalization of ARV is sensitive to caveolae and dynamin inhibitors, while it is insensitive to clathrin inhibitors. In conclusion, these results indicate that the ARV σB protein interacts with Cav-1 during dynamin-dependent caveolae-mediated endocytosis for the entry of ARV.
Collapse
Affiliation(s)
- Yuyang Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Testing Center, Yangzhou University, Yangzhou 225009, China
| | - Yangyang Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Wei Zuo
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zongyi Bo
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Chengcheng Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiaorong Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yantao Wu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
15
|
Mejía-Méndez JL, Vazquez-Duhalt R, Hernández LR, Sánchez-Arreola E, Bach H. Virus-like Particles: Fundamentals and Biomedical Applications. Int J Mol Sci 2022; 23:8579. [PMID: 35955711 PMCID: PMC9369363 DOI: 10.3390/ijms23158579] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
Nanotechnology is a fast-evolving field focused on fabricating nanoscale objects for industrial, cosmetic, and therapeutic applications. Virus-like particles (VLPs) are self-assembled nanoparticles whose intrinsic properties, such as heterogeneity, and highly ordered structural organization are exploited to prepare vaccines; imaging agents; construct nanobioreactors; cancer treatment approaches; or deliver drugs, genes, and enzymes. However, depending upon the intrinsic features of the native virus from which they are produced, the therapeutic performance of VLPs can vary. This review compiles the recent scientific literature about the fundamentals of VLPs with biomedical applications. We consulted different databases to present a general scenario about viruses and how VLPs are produced in eukaryotic and prokaryotic cell lines to entrap therapeutic cargo. Moreover, the structural classification, morphology, and methods to functionalize the surface of VLPs are discussed. Finally, different characterization techniques required to examine the size, charge, aggregation, and composition of VLPs are described.
Collapse
Affiliation(s)
- Jorge L. Mejía-Méndez
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología UNAM, Km 107 Carretera Tijuana-Ensenada, Ensenada 22860, Baja California, Mexico;
| | - Luis R. Hernández
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Eugenio Sánchez-Arreola
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| |
Collapse
|
16
|
Silva PG, Branco PTBS, Soares RRG, Mesquita JR, Sousa SIV. SARS-CoV-2 air sampling: A systematic review on the methodologies for detection and infectivity. INDOOR AIR 2022; 32:e13083. [PMID: 36040285 PMCID: PMC9538005 DOI: 10.1111/ina.13083] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
This systematic review aims to present an overview of the current aerosol sampling methods (and equipment) being used to investigate the presence of SARS-CoV-2 in the air, along with the main parameters reported in the studies that are essential to analyze the advantages and disadvantages of each method and perspectives for future research regarding this mode of transmission. A systematic literature review was performed on PubMed/MEDLINE, Web of Science, and Scopus to assess the current air sampling methodologies being applied to SARS-CoV-2. Most of the studies took place in indoor environments and healthcare settings and included air and environmental sampling. The collection mechanisms used were impinger, cyclone, impactor, filters, water-based condensation, and passive sampling. Most of the reviewed studies used RT-PCR to test the presence of SARS-CoV-2 RNA in the collected samples. SARS-CoV-2 RNA was detected with all collection mechanisms. From the studies detecting the presence of SARS-CoV-2 RNA, fourteen assessed infectivity. Five studies detected viable viruses using impactor, water-based condensation, and cyclone collection mechanisms. There is a need for a standardized protocol for sampling SARS-CoV-2 in air, which should also account for other influencing parameters, including air exchange ratio in the room sampled, relative humidity, temperature, and lighting conditions.
Collapse
Affiliation(s)
- Priscilla G. Silva
- Laboratory for Integrative and Translational Research in Population Health (ITR)PortoPortugal
- School of Medicine and Biomedical Sciences (ICBAS)University of PortoPortoPortugal
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of EngineeringUniversity of PortoPortoPortugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of EngineeringUniversity of PortoPortoPortugal
- Epidemiology Research Unit (EPI Unit), Institute of Public HealthUniversity of PortoPortoPortugal
| | - Pedro T. B. S. Branco
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of EngineeringUniversity of PortoPortoPortugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of EngineeringUniversity of PortoPortoPortugal
| | - Ruben R. G. Soares
- Department of Biochemistry and Biophysics, Science for Life LaboratoryStockholm UniversitySolnaSweden
- Division of Nanobiotechnology, Department of Protein Science, Science for Life LaboratoryKTH Royal Institute of TechnologySolnaSweden
| | - João R. Mesquita
- Laboratory for Integrative and Translational Research in Population Health (ITR)PortoPortugal
- Epidemiology Research Unit (EPI Unit), Institute of Public HealthUniversity of PortoPortoPortugal
| | - Sofia I. V. Sousa
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of EngineeringUniversity of PortoPortoPortugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of EngineeringUniversity of PortoPortoPortugal
| |
Collapse
|
17
|
Nabi-Afjadi M, Heydari M, Zalpoor H, Arman I, Sadoughi A, Sahami P, Aghazadeh S. Lectins and lectibodies: potential promising antiviral agents. Cell Mol Biol Lett 2022; 27:37. [PMID: 35562647 PMCID: PMC9100318 DOI: 10.1186/s11658-022-00338-4] [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: 02/09/2022] [Accepted: 04/21/2022] [Indexed: 12/30/2022] Open
Abstract
In nature, lectins are widely dispersed proteins that selectively recognize and bind to carbohydrates and glycoconjugates via reversible bonds at specific binding sites. Many viral diseases have been treated with lectins due to their wide range of structures, specificity for carbohydrates, and ability to bind carbohydrates. Through hemagglutination assays, these proteins can be detected interacting with various carbohydrates on the surface of cells and viral envelopes. This review discusses the most robust lectins and their rationally engineered versions, such as lectibodies, as antiviral proteins. Fusion of lectin and antibody’s crystallizable fragment (Fc) of immunoglobulin G (IgG) produces a molecule called a “lectibody” that can act as a carbohydrate-targeting antibody. Lectibodies can not only bind to the surface glycoproteins via their lectins and neutralize and clear viruses or infected cells by viruses but also perform Fc-mediated antibody effector functions. These functions include complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), and antibody-dependent cell-mediated phagocytosis (ADCP). In addition to entering host cells, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein S1 binds to angiotensin-converting enzyme 2 (ACE2) and downregulates it and type I interferons in a way that may lead to lung disease. The SARS-CoV-2 spike protein S1 and human immunodeficiency virus (HIV) envelope are heavily glycosylated, which could make them a major target for developing vaccines, diagnostic tests, and therapeutic drugs. Lectibodies can lead to neutralization and clearance of viruses and cells infected by viruses by binding to glycans located on the envelope surface (e.g., the heavily glycosylated SARS-CoV-2 spike protein).
Collapse
Affiliation(s)
- Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Morteza Heydari
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 13145-1384, Iran
| | - Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,American Association of Kidney Patients, Tampa, FL, USA
| | - Ibrahim Arman
- Department of Molecular Biology and Genetics, Faculty of Sciences and Arts, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Arezoo Sadoughi
- Department of Immunology, International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Parisa Sahami
- Medical Biology Research Center, Health Technologies Institute, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | - Safiyeh Aghazadeh
- Division of Biochemistry, Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, 5756151818, Iran.
| |
Collapse
|
18
|
Channel catfish virus entry into host cells via clathrin-mediated endocytosis. Virus Res 2022; 315:198794. [DOI: 10.1016/j.virusres.2022.198794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/19/2022]
|
19
|
Griffiths WJ, Wang Y. Cholesterol metabolism: from lipidomics to immunology. J Lipid Res 2022; 63:100165. [PMID: 34953867 PMCID: PMC8953665 DOI: 10.1016/j.jlr.2021.100165] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022] Open
Abstract
Oxysterols, the oxidized forms of cholesterol or of its precursors, are formed in the first steps of cholesterol metabolism. Oxysterols have interested chemists, biologists, and physicians for many decades, but their exact biological relevance in vivo, other than as intermediates in bile acid biosynthesis, has long been debated. However, in the first quarter of this century, a role for side-chain oxysterols and their C-7 oxidized metabolites has been convincingly established in the immune system. 25-Hydroxycholesterol has been shown to be synthesized by macrophages in response to the activation of Toll-like receptors and to offer protection against microbial pathogens, whereas 7α,25-dihydroxycholesterol has been shown to act as a chemoattractant to lymphocytes expressing the G protein-coupled receptor Epstein-Barr virus-induced gene 2 and to be important in coordinating the action of B cells, T cells, and dendritic cells in secondary lymphoid tissue. There is a growing body of evidence that not only these two oxysterols but also many of their isomers are of importance to the proper function of the immune system. Here, we review recent findings related to the roles of oxysterols in immunology.
Collapse
Affiliation(s)
| | - Yuqin Wang
- Swansea University Medical School, Swansea, Wales, United Kingdom.
| |
Collapse
|
20
|
Zhang F, Wu LB, Yu Q, Wang MJ, Zeng XL, Wei XT, Wu ZJ, Cai RL, Hu L. Neurotropic Viruses as a Tool for Neural Circuit-Tracing. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421040176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Amanzougaghene N, Tajeri S, Yalaoui S, Lorthiois A, Soulard V, Gego A, Rametti A, Risco-Castillo V, Moreno A, Tefit M, van Gemert GJ, Sauerwein RW, Vaillant JC, Ravassard P, Pérignon JL, Froissard P, Mazier D, Franetich JF. The Host Protein Aquaporin-9 is Required for Efficient Plasmodium falciparum Sporozoite Entry into Human Hepatocytes. Front Cell Infect Microbiol 2021; 11:704662. [PMID: 34268141 PMCID: PMC8276244 DOI: 10.3389/fcimb.2021.704662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocyte invasion by Plasmodium sporozoites represents a promising target for innovative antimalarial therapy, but the molecular events mediating this process are still largely uncharacterized. We previously showed that Plasmodium falciparum sporozoite entry into hepatocytes strictly requires CD81. However, CD81-overexpressing human hepatoma cells remain refractory to P. falciparum infection, suggesting the existence of additional host factors necessary for sporozoite entry. Here, through differential transcriptomic analysis of human hepatocytes and hepatoma HepG2-CD81 cells, the transmembrane protein Aquaporin-9 (AQP9) was found to be among the most downregulated genes in hepatoma cells. RNA silencing showed that sporozoite invasion of hepatocytes requires AQP9 expression. AQP9 overexpression in hepatocytes increased their permissiveness to P. falciparum. Moreover, chemical disruption with the AQP9 inhibitor phloretin markedly inhibited hepatocyte infection. Our findings identify AQP9 as a novel host factor required for P. falciparum sporozoite hepatocyte-entry and indicate that AQP9 could be a potential therapeutic target.
Collapse
Affiliation(s)
- Nadia Amanzougaghene
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Shahin Tajeri
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Samir Yalaoui
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Audrey Lorthiois
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Valérie Soulard
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Audrey Gego
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Armelle Rametti
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | | | - Alicia Moreno
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Maurel Tefit
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, MMB-NCMLS, Nijmegen, Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, MMB-NCMLS, Nijmegen, Netherlands
| | - Jean-Christophe Vaillant
- AP-HP, Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
| | - Philippe Ravassard
- CR-ICM - LGN CNRS UMR-7991, IFR des Neurosciences, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jean-Louis Pérignon
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Patrick Froissard
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Dominique Mazier
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Jean-François Franetich
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| |
Collapse
|
22
|
Newcastle Disease Virus Entry into Chicken Macrophages via a pH-Dependent, Dynamin and Caveola-Mediated Endocytic Pathway That Requires Rab5. J Virol 2021; 95:e0228820. [PMID: 33762417 DOI: 10.1128/jvi.02288-20] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The cellular entry pathways and the mechanisms of Newcastle disease virus (NDV) entry into cells are poorly characterized. In this study, we demonstrated that chicken interferon-induced transmembrane protein 1 (chIFITM1), which is located in the early endosomes, could limit the replication of NDV in chicken macrophage cell line HD11, suggesting the endocytic entry of NDV into chicken macrophages. Then, we presented a systematic study about the entry mechanism of NDV into chicken macrophages. First, we demonstrated that a low-pH condition and dynamin were required during NDV entry. However, NDV entry into chicken macrophages was independent of clathrin-mediated endocytosis. We also found that NDV entry was dependent on membrane cholesterol. The NDV entry and replication were significantly reduced by nystatin and phorbol 12-myristate 13-acetate treatment, overexpression of dominant-negative (DN) caveolin-1, or knockdown of caveolin-1, suggesting that NDV entry depends on caveola-mediated endocytosis. However, macropinocytosis did not play a role in NDV entry into chicken macrophages. In addition, we found that Rab5, rather than Rab7, was involved in the entry and traffic of NDV. The colocalization of NDV with Rab5 and early endosome suggested that NDV virion was transported to early endosomes in a Rab5-dependent manner after internalization. Of particular note, the caveola-mediated endocytosis was also utilized by NDV to enter primary chicken macrophages. Moreover, NDV entered different cell types using different pathways. Collectively, our findings demonstrate for the first time that NDV virion enters chicken macrophages via a pH-dependent, dynamin and caveola-mediated endocytosis pathway and that Rab5 is involved in the traffic and location of NDV. IMPORTANCE Although the pathogenesis of Newcastle disease virus (NDV) has been extensively studied, the detailed mechanism of NDV entry into host cells is largely unknown. Macrophages are the first-line defenders of host defense against infection of pathogens. Chicken macrophages are considered one of the main types of target cells during NDV infection. Here, we comprehensively investigated the entry mechanism of NDV in chicken macrophages. This is the first report to demonstrate that NDV enters chicken macrophages via a pH-dependent, dynamin and caveola-mediated endocytosis pathway that requires Rab5. The result is important for our understanding of the entry of NDV in chicken macrophages, which will further advance the knowledge of NDV pathogenesis and provide useful clues for the development of novel preventive or therapeutic strategies against NDV infection. In addition, this information will contribute to our further understanding of pathogenesis with regard to other members of the Avulavirus genus in the Paramyxoviridae family.
Collapse
|
23
|
Nascimento da Silva LC, Mendonça JSP, de Oliveira WF, Batista KLR, Zagmignan A, Viana IFT, Dos Santos Correia MT. Exploring lectin-glycan interactions to combat COVID-19: Lessons acquired from other enveloped viruses. Glycobiology 2021; 31:358-371. [PMID: 33094324 PMCID: PMC7665446 DOI: 10.1093/glycob/cwaa099] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/30/2020] [Accepted: 09/26/2020] [Indexed: 01/08/2023] Open
Abstract
The emergence of a new human coronavirus (SARS-CoV-2) has imposed great pressure on the health system worldwide. The presence of glycoproteins on the viral envelope opens a wide range of possibilities for application of lectins to address some urgent problems involved in this pandemic. In this work, we discuss the potential contributions of lectins from non-mammalian sources in the development of several fields associated with viral infections, most notably COVID-19. We review the literature on the use of non-mammalian lectins as a therapeutic approach against members of the Coronaviridae family, including recent advances in strategies of protein engineering to improve their efficacy. The applications of lectins as adjuvants for antiviral vaccines are also discussed. Finally, we present some emerging strategies employing lectins for the development of biosensors, microarrays, immunoassays and tools for purification of viruses from whole blood. Altogether, the data compiled in this review highlights the importance of structural studies aiming to improve our knowledge about the basis of glycan recognition by lectins and its repercussions in several fields, providing potential solutions for complex aspects that are emerging from different health challenges.
Collapse
Affiliation(s)
- Luís Cláudio Nascimento da Silva
- Programa de Pós-graduação em Biologia Microbiana, Laboratório de Patogenicidade Bacteriana, Universidade CEUMA, São Luís 65075-120, Brazil.,Programa de Pós-graduação em Biodiversidade e Biotecnologia da Amazônia Legal, Laboratório de Patogenicidade Bacteriana, Universidade CEUMA, São Luís 65075-120, Brazil
| | - Juliana Silva Pereira Mendonça
- Programa de Pós-graduação em Biologia Microbiana, Laboratório de Patogenicidade Bacteriana, Universidade CEUMA, São Luís 65075-120, Brazil
| | - Weslley Felix de Oliveira
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife 50.670-901, Brazil
| | - Karla Lílian Rodrigues Batista
- Programa de Pós-graduação em Biodiversidade e Biotecnologia da Amazônia Legal, Laboratório de Patogenicidade Bacteriana, Universidade CEUMA, São Luís 65075-120, Brazil
| | - Adrielle Zagmignan
- Programa de Pós-graduação em Biodiversidade e Biotecnologia da Amazônia Legal, Laboratório de Patogenicidade Bacteriana, Universidade CEUMA, São Luís 65075-120, Brazil
| | | | | |
Collapse
|
24
|
Sardar A, Lahiri A, Kamble M, Mallick AI, Tarafdar PK. Translation of Mycobacterium Survival Strategy to Develop a Lipo‐peptide based Fusion Inhibitor**. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202013848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Avijit Sardar
- Department of Chemical Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| | - Aritraa Lahiri
- Department of Biological Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| | - Mithila Kamble
- Department of Biological Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| | - Amirul I. Mallick
- Department of Biological Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| | - Pradip K. Tarafdar
- Department of Chemical Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| |
Collapse
|
25
|
Sardar A, Lahiri A, Kamble M, Mallick AI, Tarafdar PK. Translation of Mycobacterium Survival Strategy to Develop a Lipo-peptide based Fusion Inhibitor*. Angew Chem Int Ed Engl 2021; 60:6101-6106. [PMID: 33241871 PMCID: PMC7753697 DOI: 10.1002/anie.202013848] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Indexed: 12/16/2022]
Abstract
The entry of enveloped virus requires the fusion of viral and host cell membranes. An effective fusion inhibitor aiming at impeding such membrane fusion may emerge as a broad-spectrum antiviral agent against a wide range of viral infections. Mycobacterium survives inside the phagosome by inhibiting phagosome-lysosome fusion with the help of a coat protein coronin 1. Structural analysis of coronin 1 and other WD40-repeat protein suggest that the trp-asp (WD) sequence is placed at distorted β-meander motif (more exposed) in coronin 1. The unique structural feature of coronin 1 was explored to identify a simple lipo-peptide sequence (myr-WD), which effectively inhibits membrane fusion by modulating the interfacial order, water penetration, and surface potential. The mycobacterium inspired lipo-dipeptide was successfully tested to combat type 1 influenza virus (H1N1) and murine coronavirus infections as a potential broad-spectrum antiviral agent.
Collapse
Affiliation(s)
- Avijit Sardar
- Department of Chemical SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| | - Aritraa Lahiri
- Department of Biological SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| | - Mithila Kamble
- Department of Biological SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| | - Amirul I. Mallick
- Department of Biological SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| | - Pradip K. Tarafdar
- Department of Chemical SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| |
Collapse
|
26
|
Lay Mendoza MF, Acciani MD, Levit CN, Santa Maria C, Brindley MA. Monitoring Viral Entry in Real-Time Using a Luciferase Recombinant Vesicular Stomatitis Virus Producing SARS-CoV-2, EBOV, LASV, CHIKV, and VSV Glycoproteins. Viruses 2020; 12:E1457. [PMID: 33348746 PMCID: PMC7766484 DOI: 10.3390/v12121457] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 01/06/2023] Open
Abstract
Viral entry is the first stage in the virus replication cycle and, for enveloped viruses, is mediated by virally encoded glycoproteins. Viral glycoproteins have different receptor affinities and triggering mechanisms. We employed vesicular stomatitis virus (VSV), a BSL-2 enveloped virus that can incorporate non-native glycoproteins, to examine the entry efficiencies of diverse viral glycoproteins. To compare the glycoprotein-mediated entry efficiencies of VSV glycoprotein (G), Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S), Ebola (EBOV) glycoprotein (GP), Lassa (LASV) GP, and Chikungunya (CHIKV) envelope (E) protein, we produced recombinant VSV (rVSV) viruses that produce the five glycoproteins. The rVSV virions encoded a nano luciferase (NLucP) reporter gene fused to a destabilization domain (PEST), which we used in combination with the live-cell substrate EndurazineTM to monitor viral entry kinetics in real time. Our data indicate that rVSV particles with glycoproteins that require more post-internalization priming typically demonstrate delayed entry in comparison to VSV G. In addition to determining the time required for each virus to complete entry, we also used our system to evaluate viral cell surface receptor preferences, monitor fusion, and elucidate endocytosis mechanisms. This system can be rapidly employed to examine diverse viral glycoproteins and their entry requirements.
Collapse
Affiliation(s)
- Maria Fernanda Lay Mendoza
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (M.F.L.M.); (M.D.A.); (C.N.L.); (C.S.M.)
| | - Marissa Danielle Acciani
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (M.F.L.M.); (M.D.A.); (C.N.L.); (C.S.M.)
| | - Courtney Nina Levit
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (M.F.L.M.); (M.D.A.); (C.N.L.); (C.S.M.)
| | - Christopher Santa Maria
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (M.F.L.M.); (M.D.A.); (C.N.L.); (C.S.M.)
| | - Melinda Ann Brindley
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (M.F.L.M.); (M.D.A.); (C.N.L.); (C.S.M.)
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
27
|
Affiliation(s)
- Jyoti Verma
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Naidu Subbarao
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | | |
Collapse
|
28
|
Selective EMC subunits act as molecular tethers of intracellular organelles exploited during viral entry. Nat Commun 2020; 11:1127. [PMID: 32111841 PMCID: PMC7048770 DOI: 10.1038/s41467-020-14967-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 02/10/2020] [Indexed: 12/22/2022] Open
Abstract
Although viruses must navigate the complex host endomembrane system to infect cells, the strategies used to achieve this is unclear. During entry, polyomavirus SV40 is sorted from the late endosome (LE) to the endoplasmic reticulum (ER) to cause infection, yet how this is accomplished remains enigmatic. Here we find that EMC4 and EMC7, two ER membrane protein complex (EMC) subunits, support SV40 infection by promoting LE-to-ER targeting of the virus. They do this by engaging LE-associated Rab7, presumably to stabilize contact between the LE and ER. These EMC subunits also bind to the ER-resident fusion machinery component syntaxin18, which is required for SV40-arrival to the ER. Our data suggest that EMC4 and EMC7 act as molecular tethers, inter-connecting two intracellular compartments to enable efficient transport of a virus between these compartments. As LE-to-ER transport of cellular cargos is unclear, our results have broad implications for illuminating inter-organelle cargo transport. The endoplasmic reticulum membrane protein complex (EMC) is known to play a role in SV40 viral infection but precise mechanisms are unclear. Here, the authors report that the EMC acts as tether of late endosome–endoplasmic reticulum interorganellar membrane contact sites to promote SV40 viral infection.
Collapse
|
29
|
Huo Y, Yu Y, Liu Q, Liu D, Zhang M, Liang J, Chen X, Zhang L, Fang R. Rice stripe virus hitchhikes the vector insect vitellogenin ligand-receptor pathway for ovary entry. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180312. [PMID: 30967014 DOI: 10.1098/rstb.2018.0312] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It is known that plant arboviruses infect insect vector cells by endocytosis; however, the cellular receptors that mediate endocytosis have not been well defined. In our recently published work and this study, by clarifying the vertical transmission mechanism of Rice stripe virus (RSV) in Laodelphax striatellus, we provide a novel paradigm for how arboviruses enter insect germ-line cells. Instead of direct interaction with a viral receptor, the virus binds to a secreted ligand protein, hitchhiking the ligand-receptor pathway to achieve cell entry. Vitellogenin (Vg) is an indispensable protein for embryo development that is synthesized extra-ovarially and taken up by germ-line cells through Vg receptor (VgR)-mediated endocytosis. After revealing that RSV invades L. striatellus ovary by a specific molecular interaction with the insect Vg in haemolymph, this study addressed VgR's function in mediating the RSV invasion of the germarium nurse cells, further confirming the ligand's receptor-mediated viral cell-invasion mechanism. Understanding the viral ovary-entry pathways in vectors will help to find suitable measures to block the trans-generation transmission of the viruses. This article is part of the theme issue 'Biotic signalling sheds light on smart pest management'.
Collapse
Affiliation(s)
- Yan Huo
- 1 State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences , Beijing 100101 , People's Republic of China.,2 National Plant Gene Research Center , Beijing 100101 , People's Republic of China
| | - Yuanling Yu
- 1 State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences , Beijing 100101 , People's Republic of China.,2 National Plant Gene Research Center , Beijing 100101 , People's Republic of China.,3 University of the Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Qing Liu
- 1 State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences , Beijing 100101 , People's Republic of China.,2 National Plant Gene Research Center , Beijing 100101 , People's Republic of China.,3 University of the Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Da Liu
- 1 State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences , Beijing 100101 , People's Republic of China.,2 National Plant Gene Research Center , Beijing 100101 , People's Republic of China.,3 University of the Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Mengting Zhang
- 1 State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences , Beijing 100101 , People's Republic of China.,2 National Plant Gene Research Center , Beijing 100101 , People's Republic of China.,3 University of the Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Jingnan Liang
- 4 Public Technology Service Center, Institute of Microbiology , Beijing 100101, People's Republic of China
| | - Xiaoying Chen
- 1 State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences , Beijing 100101 , People's Republic of China.,2 National Plant Gene Research Center , Beijing 100101 , People's Republic of China
| | - Lili Zhang
- 1 State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences , Beijing 100101 , People's Republic of China.,2 National Plant Gene Research Center , Beijing 100101 , People's Republic of China
| | - Rongxiang Fang
- 1 State Key Laboratory of Plant Genomics, Institute of Microbiology, Chinese Academy of Sciences , Beijing 100101 , People's Republic of China.,2 National Plant Gene Research Center , Beijing 100101 , People's Republic of China
| |
Collapse
|
30
|
Reverse Engineering Provides Insights on the Evolution of Subgroups A to E Avian Sarcoma and Leukosis Virus Receptor Specificity. Viruses 2019; 11:v11060497. [PMID: 31151254 PMCID: PMC6630264 DOI: 10.3390/v11060497] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/31/2022] Open
Abstract
The initial step of retrovirus entry—the interaction between the virus envelope glycoprotein trimer and a cellular receptor—is complex, involving multiple, noncontiguous determinants in both proteins that specify receptor choice, binding affinity and the ability to trigger conformational changes in the viral glycoproteins. Despite the complexity of this interaction, retroviruses have the ability to evolve the structure of their envelope glycoproteins to use a different cellular protein as receptors. The highly homologous subgroup A to E Avian Sarcoma and Leukosis Virus (ASLV) glycoproteins belong to the group of class 1 viral fusion proteins with a two-step triggering mechanism that allows experimental access to intermediate structures during the fusion process. We and others have taken advantage of replication-competent ASLVs and exploited genetic selection strategies to force the ASLVs to naturally evolve and acquire envelope glycoprotein mutations to escape the pressure on virus entry and still yield a functional replicating virus. This approach allows for the simultaneous selection of multiple mutations in multiple functional domains of the envelope glycoprotein that may be required to yield a functional virus. Here, we review the ASLV family and experimental system and the reverse engineering approaches used to understand the evolution of ASLV receptor usage.
Collapse
|
31
|
Yang B, Qi X, Guo H, Jia P, Chen S, Chen Z, Wang T, Wang J, Xue Q. Peste des Petits Ruminants Virus Enters Caprine Endometrial Epithelial Cells via the Caveolae-Mediated Endocytosis Pathway. Front Microbiol 2018; 9:210. [PMID: 29497407 PMCID: PMC5818419 DOI: 10.3389/fmicb.2018.00210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/30/2018] [Indexed: 12/11/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) causes an acute and highly contagious disease of sheep and goats and has spread with alarming speed around the world. The pathology of Peste des petits ruminants is linked to retrogressive changes and necrotic lesions in lymphoid tissues and epithelial cells. However, the process of PPRV entry into host epithelial cells remains largely unknown. Here, we performed a comprehensive study of the entry mechanism of PPRV into caprine endometrial epithelial cells (EECs). We clearly demonstrated that PPRV internalization was inhibited by chloroquine and ammonium chloride, which elevate the pH of various organelles. However, PPRV entry was not affected by chlorpromazine and knockdown of the clathrin heavy chain in EECs. In addition, we found that the internalization of PPRV was dependent on dynamin and membrane cholesterol and was suppressed by silencing of caveolin-1. Macropinocytosis did not play a role, but phosphatidylinositol 3-kinase (PI3K) was required for PPRV internalization. Cell type and receptor-dependent differences indicated that PPRV entry into caprine fetal fibroblast cells (FFCs) occurred via a different route. Taken together, our findings demonstrate that PPRV enters EECs through a cholesterol-dependent caveolae-mediated uptake mechanism that is pH-dependent and requires dynamin and PI3K but is independent of clathrin. This potentially provides insight into the entry mechanisms of other morbilliviruses.
Collapse
Affiliation(s)
- Bo Yang
- China Institute of Veterinary Drug Control, Beijing, China.,College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Hui Guo
- China Institute of Veterinary Drug Control, Beijing, China
| | - Peilong Jia
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Shuying Chen
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Zhijie Chen
- China Institute of Veterinary Drug Control, Beijing, China.,College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Ting Wang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, China
| |
Collapse
|
32
|
Retroviral envelope proteins: Involvement in neuropathogenesis. J Neurol Sci 2017; 380:151-163. [DOI: 10.1016/j.jns.2017.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/23/2017] [Accepted: 07/18/2017] [Indexed: 02/07/2023]
|
33
|
A Cell Fusion-Based Screening Method Identifies Glycosylphosphatidylinositol-Anchored Protein Ly6e as the Receptor for Mouse Endogenous Retroviral Envelope Syncytin-A. J Virol 2017; 91:JVI.00832-17. [PMID: 28679758 DOI: 10.1128/jvi.00832-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/28/2017] [Indexed: 11/20/2022] Open
Abstract
Syncytin genes are envelope genes of retroviral origin that have been exapted for a role in placentation. They are involved in the formation of a syncytial structure (the syncytiotrophoblast) at the fetomaternal interface via their fusogenic activity. The mouse placenta is unique among placental mammals since the fetomaternal interface comprises two syncytiotrophoblast layers (ST-I and ST-II) instead of one, as observed in humans and all other hemochorial placentae. Each layer specifically expresses a distinct mouse syncytin, namely, syncytin-A (SynA) for ST-I and syncytin-B (SynB) for ST-II, which have been shown to be essential to placentogenesis and embryo survival. Their cognate cellular receptors, which are necessary to mediate cell-cell fusion and syncytiotrophoblast formation, are still unknown. By devising a sensitive method that combines a cell-cell fusion assay with the screening of a mouse cDNA library, we succeeded in identifying the glycosylphosphatidylinositol (GPI)-anchored membrane protein lymphocyte antigen 6E (Ly6e) as a candidate receptor for SynA. Transfection of cells with the cloned receptor led to their fusion to cells expressing SynA, with no cross-reactive fusion activity with SynB. Knocking down Ly6e greatly reduced SynA-induced cell fusion, thus suggesting that Ly6e is the sole receptor for SynA in vivo Interaction of SynA with Ly6e was further demonstrated by a competition assay using the soluble ectodomain of Ly6e. Finally, reverse transcription-quantitative PCR (RT-qPCR) analysis of Ly6e expression on a representative panel of mouse tissues shows that it is significantly expressed in the mouse placenta together with SynA.IMPORTANCE Syncytin genes are envelope genes of endogenous retroviruses, co-opted for a physiological function in placentation. Syncytins are fusogenic proteins that mediate cell-cell fusion by interacting with receptors present on the partner cells. Here, by devising a sensitive in vitro fusion assay that enables the high-throughput screening of normalized cDNA libraries, we identified the long-sought receptor for syncytin-A (SynA), a mouse syncytin responsible for syncytiotrophoblast formation at the maternofetal interface of the mouse placenta. This protein, Ly6e (lymphocyte antigen 6E), is a GPI-anchored membrane protein, and small interfering RNA (siRNA) experiments targeting its deletion as well as a decoy assay using a recombinant soluble receptor show that Ly6e is the necessary and sufficient partner of SynA. Its profile of expression is consistent with a role in both ancestral endogenization of a SynA founder retrovirus and present-day placenta formation. This study provides a powerful general method to identify genes involved in cell-cell fusion processes.
Collapse
|
34
|
Bagchi P, Inoue T, Tsai B. EMC1-dependent stabilization drives membrane penetration of a partially destabilized non-enveloped virus. eLife 2016; 5. [PMID: 28012275 PMCID: PMC5224922 DOI: 10.7554/elife.21470] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/23/2016] [Indexed: 11/13/2022] Open
Abstract
Destabilization of a non-enveloped virus generates a membrane transport-competent viral particle. Here we probe polyomavirus SV40 endoplasmic reticulum (ER)-to-cytosol membrane transport, a decisive infection step where destabilization initiates this non-enveloped virus for membrane penetration. We find that a member of the ER membrane protein complex (EMC) called EMC1 promotes SV40 ER membrane transport and infection. Surprisingly, EMC1 does so by using its predicted transmembrane residue D961 to bind to and stabilize the membrane-embedded partially destabilized SV40, thereby preventing premature viral disassembly. EMC1-dependent stabilization enables SV40 to engage a cytosolic extraction complex that ejects the virus into the cytosol. Thus EMC1 acts as a molecular chaperone, bracing the destabilized SV40 in a transport-competent state. Our findings reveal the novel principle that coordinated destabilization-stabilization drives membrane transport of a non-enveloped virus. DOI:http://dx.doi.org/10.7554/eLife.21470.001
Collapse
Affiliation(s)
- Parikshit Bagchi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Takamasa Inoue
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
35
|
Ahani R, Roohvand F, Cohan RA, Etemadzadeh MH, Mohajel N, Behdani M, Shahosseini Z, Madani N, Azadmanesh K. Sindbis Virus-Pseudotyped Lentiviral Vectors Carrying VEGFR2-Specific Nanobody for Potential Transductional Targeting of Tumor Vasculature. Mol Biotechnol 2016; 58:738-747. [PMID: 27647452 DOI: 10.1007/s12033-016-9973-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction of selectivity/specificity into viral-based gene delivery systems, such as lentiviral vectors (LVs), is crucial in their systemic administration for cancer gene therapy. The pivotal role of tumor-associated endothelial cells (TAECs) in tumor angiogenesis and overexpression of vascular endothelial growth factor receptor-2 (VEGFR2 or KDR) in TAECs makes them a potent target in cancer treatment. Herein, we report the development of VEGFR2-targeted LVs pseudotyped with chimeric sindbis virus E2 glycoprotein (cSVE2s). For this purpose, either sequence of a VEGFR2-specific nanobody or its natural ligand (VEGF121) was inserted into the binding site of sindbis virus E2 glycoprotein. In silico modeling data suggested that the inserted targeting motifs were exposed in the context of cSVE2s. Western blot analysis of LVs indicated the incorporation of cSVE2s into viral particles. Capture ELISA demonstrated the specificity/functionality of the incorporated cSVE2s. Transduction of 293/KDR (expressing VEGFR2) or 293T cells (negative control) by constructed LVs followed by fluorescent microscopy and flow cytometric analyses indicated selective transduction of 293/KDR cells (30 %) by both targeting motifs compared to 293T control cells (1-2 %). These results implied similar targeting properties of VEGFR2-specific nanobody compared to the VEGF121 and indicated the potential for transductional targeting of tumor vasculature by the nanobody displaying LVs.
Collapse
Affiliation(s)
- Roshank Ahani
- Department of Virology, Pasteur Institute of Iran, 69 Pasteur Avenue, Kargar Avenue, Tehran, 1316943551, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, 69 Pasteur Avenue, Kargar Avenue, Tehran, 1316943551, Iran.
| | - Reza Ahangari Cohan
- New Technologies Research Group, Department of Pilot Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Nasir Mohajel
- Department of Virology, Pasteur Institute of Iran, 69 Pasteur Avenue, Kargar Avenue, Tehran, 1316943551, Iran
| | - Mahdi Behdani
- Biotechnology Research Center, Venom & Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Shahosseini
- Department of Virology, Pasteur Institute of Iran, 69 Pasteur Avenue, Kargar Avenue, Tehran, 1316943551, Iran
| | - Navid Madani
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Kayhan Azadmanesh
- Department of Virology, Pasteur Institute of Iran, 69 Pasteur Avenue, Kargar Avenue, Tehran, 1316943551, Iran.
| |
Collapse
|
36
|
Liu Q, Zhou YH, Ye F, Yang ZQ. Antivirals for Respiratory Viral Infections: Problems and Prospects. Semin Respir Crit Care Med 2016; 37:640-6. [PMID: 27486742 PMCID: PMC7171711 DOI: 10.1055/s-0036-1584803] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the past two decades, several newly emerging and reemerging viral respiratory pathogens including several influenza viruses (avian influenza and pandemic influenza), severe acute respiratory syndrome coronavirus (SARS-CoV), and Middle East respiratory syndrome coronavirus (MERS-CoV), have continued to challenge medical and public health systems. Thereafter, the development of cost-effective, broad-spectrum antiviral agents is the urgent mission of both virologists and pharmacologists. Current antiviral developments have focused targets on viral entry, replication, release, and intercellular pathways essential for viral life cycle. Here, we review the current literature on challenges and prospects in the development of these antivirals.
Collapse
Affiliation(s)
- Qiang Liu
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang, China
| | - Yuan-Hong Zhou
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang, China
| | - Feng Ye
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang, China
| | - Zhan-Qiu Yang
- State Key Laboratory of Virology, Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan, China
| |
Collapse
|
37
|
Markosyan RM, Miao C, Zheng YM, Melikyan GB, Liu SL, Cohen FS. Induction of Cell-Cell Fusion by Ebola Virus Glycoprotein: Low pH Is Not a Trigger. PLoS Pathog 2016; 12:e1005373. [PMID: 26730950 PMCID: PMC4711667 DOI: 10.1371/journal.ppat.1005373] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 12/08/2015] [Indexed: 12/11/2022] Open
Abstract
Ebola virus (EBOV) is a highly pathogenic filovirus that causes hemorrhagic fever in humans and animals. Currently, how EBOV fuses its envelope membrane within an endosomal membrane to cause infection is poorly understood. We successfully measure cell-cell fusion mediated by the EBOV fusion protein, GP, assayed by the transfer of both cytoplasmic and membrane dyes. A small molecule fusion inhibitor, a neutralizing antibody, as well as mutations in EBOV GP known to reduce viral infection, all greatly reduce fusion. By monitoring redistribution of small aqueous dyes between cells and by electrical capacitance measurements, we discovered that EBOV GP-mediated fusion pores do not readily enlarge—a marked difference from the behavior of other viral fusion proteins. EBOV GP must be cleaved by late endosome-resident cathepsins B or L in order to become fusion-competent. Cleavage of cell surface-expressed GP appears to occur in endosomes, as evidenced by the fusion block imposed by cathepsin inhibitors, agents that raise endosomal pH, or an inhibitor of anterograde trafficking. Treating effector cells with a recombinant soluble cathepsin B or thermolysin, which cleaves GP into an active form, increases the extent of fusion, suggesting that a fraction of surface-expressed GP is not cleaved. Whereas the rate of fusion is increased by a brief exposure to acidic pH, fusion does occur at neutral pH. Importantly, the extent of fusion is independent of external pH in experiments in which cathepsin activity is blocked and EBOV GP is cleaved by thermolysin. These results imply that low pH promotes fusion through the well-known pH-dependent activity of cathepsins; fusion induced by cleaved EBOV GP is a process that is fundamentally independent of pH. The cell-cell fusion system has revealed some previously unappreciated features of EBOV entry, which could not be readily elucidated in the context of endosomal entry. The devastation and transmissibility of Ebola virus (EBOV) are well known. However, the manner in which EBOV enters host cells through endosomal membrane remains elusive. Here, we have developed a convenient experimental system to mimic EBOV fusion in endosomes: cells expressing the fusion protein of EBOV, GP, on their surface are fused to target cells. This system exhibits the known key properties of EBOV fusion. We show that the pH-dependence of EBOV fusion is caused by the pH-dependence of cathepsins, proteases known to cleave EBOV GP into a fusion-competent form. We demonstrate that the fusion activity of this cleaved form is independent of pH. We further show that the enlargement of the fusion pore created by EBOV GP is unusually slow in reaching sizes necessary to pass EBOV’s genome—this is atypical of virally created fusion pores. This cell-cell fusion system should provide a useful platform for developing drugs against EBOV infection.
Collapse
Affiliation(s)
- Ruben M. Markosyan
- Rush University Medical Center, Department of Molecular Biophysics and Physiology, Chicago, Illinois, United States of America
| | - Chunhui Miao
- University of Missouri School of Medicine, Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, Columbia, Missouri, United States of America
| | - Yi-Min Zheng
- University of Missouri School of Medicine, Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, Columbia, Missouri, United States of America
| | - Gregory B. Melikyan
- Emory University Medical School, Department of Pediatrics, Infectious Diseases, Atlanta, Georgia, United States of America
| | - Shan-Lu Liu
- University of Missouri School of Medicine, Bond Life Sciences Center, Department of Molecular Microbiology and Immunology, Columbia, Missouri, United States of America
- * E-mail: (SLL); (FSC)
| | - Fredric S. Cohen
- Rush University Medical Center, Department of Molecular Biophysics and Physiology, Chicago, Illinois, United States of America
- * E-mail: (SLL); (FSC)
| |
Collapse
|
38
|
Curcumin and Boswellia serrata gum resin extract inhibit chikungunya and vesicular stomatitis virus infections in vitro. Antiviral Res 2015; 125:51-7. [PMID: 26611396 DOI: 10.1016/j.antiviral.2015.11.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/11/2015] [Accepted: 11/14/2015] [Indexed: 01/01/2023]
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes chikungunya fever and has infected millions of people mainly in developing countries. The associated disease is characterized by rash, high fever, and severe arthritis that can persist for years. CHIKV has adapted to Aedes albopictus, which also inhabits temperate regions including Europe and the United States of America. CHIKV has recently caused large outbreaks in Latin America. No treatment or licensed CHIKV vaccine exists. Traditional medicines are known to have anti-viral effects; therefore, we examined whether curcumin or Boswellia serrata gum resin extract have antiviral activity against CHIKV. Both compounds blocked entry of CHIKV Env-pseudotyped lentiviral vectors and inhibited CHIKV infection in vitro. In addition, vesicular stomatitis virus vector particles and viral infections were also inhibited to the same extent, indicating a broad antiviral activity. Although the bioavailability of these compounds is rather poor, they might be used as a lead structure to develop more effective antiviral drugs or might be used topically to prevent CHIKV spread in the skin after mosquito bites.
Collapse
|
39
|
Fogel GB, Lamers SL, Liu ES, Salemi M, McGrath MS. Identification of dual-tropic HIV-1 using evolved neural networks. Biosystems 2015; 137:12-9. [PMID: 26419858 PMCID: PMC4921197 DOI: 10.1016/j.biosystems.2015.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 09/24/2015] [Accepted: 09/26/2015] [Indexed: 02/07/2023]
Abstract
Blocking the binding of the envelope HIV-1 protein to immune cells is a popular concept for development of anti-HIV therapeutics. R5 HIV-1 binds CCR5, X4 HIV-1 binds CXCR4, and dual-tropic HIV-1 can bind either coreceptor for cellular entry. R5 viruses are associated with early infection and over time can evolve to X4 viruses that are associated with immune failure. Dual-tropic HIV-1 is less studied; however, it represents functional antigenic intermediates during the transition of R5 to X4 viruses. Viral tropism is linked partly to the HIV-1 envelope V3 domain, where the amino acid sequence helps dictate the receptor a particular virus will target; however, using V3 sequence information to identify dual-tropic HIV-1 isolates has remained difficult. Our goal in this study was to elucidate features of dual-tropic HIV-1 isolates that assist in the biological understanding of dual-tropism and develop an approach for their detection. Over 1559 HIV-1 subtype B sequences with known tropisms were analyzed. Each sequence was represented by 73 structural, biochemical and regional features. These features were provided to an evolved neural network classifier and evaluated using balanced and unbalanced data sets. The study resolved R5X4 viruses from R5 with an accuracy of 81.8% and from X4 with an accuracy of 78.8%. The approach also identified a set of V3 features (hydrophobicity, structural and polarity) that are associated with tropism transitions. The ability to distinguish R5X4 isolates will improve computational tropism decisions for R5 vs. X4 and assist in HIV-1 research and drug development efforts.
Collapse
Affiliation(s)
- Gary B Fogel
- Natural Selection, Inc., San Diego, CA 92121, United States
| | | | - Enoch S Liu
- Natural Selection, Inc., San Diego, CA 92121, United States
| | - Marco Salemi
- University of Florida, Department of Pathology and Laboratory Medicine, Gainesville, FL 32610, United States
| | - Michael S McGrath
- University of California at San Francisco, Department of Laboratory Medicine and The AIDS and Cancer Specimen Resource, San Francisco, CA 94143, United States
| |
Collapse
|
40
|
Abstract
Effective antivirals have been developed against specific viruses, such as HIV, Hepatitis C virus and influenza virus. This 'one bug-one drug' approach to antiviral drug development can be successful, but it may be inadequate for responding to an increasing diversity of viruses that cause significant diseases in humans. The majority of viral pathogens that cause emerging and re-emerging infectious diseases are membrane-enveloped viruses, which require the fusion of viral and cell membranes for virus entry. Therefore, antivirals that target the membrane fusion process represent new paradigms for broad-spectrum antiviral discovery. In this Review, we discuss the mechanisms responsible for the fusion between virus and cell membranes and explore how broad-spectrum antivirals target this process to prevent virus entry.
Collapse
Affiliation(s)
- Frederic Vigant
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, New York 10029, USA
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, New York 10029, USA
| |
Collapse
|
41
|
Büttner S, Koch B, Dolnik O, Eickmann M, Freiwald T, Rudolf S, Engel J, Becker S, Ronco C, Geiger H. Extracorporeal Virus Elimination for the Treatment of Severe Ebola Virus Disease - First Experience with Lectin Affinity Plasmapheresis. Blood Purif 2015; 38:286-91. [DOI: 10.1159/000375229] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 11/19/2022]
|
42
|
Cosset FL, Dreux M. HCV transmission by hepatic exosomes establishes a productive infection. J Hepatol 2014; 60:674-5. [PMID: 24512825 DOI: 10.1016/j.jhep.2013.10.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/09/2013] [Accepted: 10/10/2013] [Indexed: 12/04/2022]
Affiliation(s)
- François-Loïc Cosset
- CIRI - International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France; Inserm, U1111, Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France; CNRS, UMR5308, Lyon, France; LabEx Ecofect, Université de Lyon, Lyon, France.
| | - Marlène Dreux
- CIRI - International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France; Inserm, U1111, Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France; CNRS, UMR5308, Lyon, France; LabEx Ecofect, Université de Lyon, Lyon, France.
| |
Collapse
|
43
|
Abstract
Viruses are important pathogens of the nervous system. Here we describe the basic properties of viruses and the principles of virus classification, evolution, structure, and replication, with a focus on neurotropic viruses that are important neuropathogens of humans. These properties then provide the background for introductions to pathogenesis of viral diseases of the nervous system, host immune responses to virus infection, and the diagnosis and treatment of virus infections of the nervous system.
Collapse
Affiliation(s)
- Philip E Pellett
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Subhash Mitra
- Division of Infectious Diseases, Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI, USA; Division of Infectious Diseases, Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Thomas C Holland
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
44
|
Chenine AL, Wieczorek L, Sanders-Buell E, Wesberry M, Towle T, Pillis DM, Molnar S, McLinden R, Edmonds T, Hirsch I, O’Connell R, McCutchan FE, Montefiori DC, Ochsenbauer C, Kappes JC, Kim JH, Polonis VR, Tovanabutra S. Impact of HIV-1 backbone on neutralization sensitivity: neutralization profiles of heterologous envelope glycoproteins expressed in native subtype C and CRF01_AE backbone. PLoS One 2013; 8:e76104. [PMID: 24312165 PMCID: PMC3843658 DOI: 10.1371/journal.pone.0076104] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/20/2013] [Indexed: 01/24/2023] Open
Abstract
Standardized assays to assess vaccine and antiviral drug efficacy are critical for the development of protective HIV-1 vaccines and drugs. These immune assays will be advanced by the development of standardized viral stocks, such as HIV-1 infectious molecular clones (IMC), that i) express a reporter gene, ii) are representative of globally diverse subtypes and iii) are engineered to easily exchange envelope (env) genes for expression of sequences of interest. Thus far, a subtype B IMC backbone expressing Renilla luciferase (LucR), and into which the ectodomain of heterologous env coding sequences can be expressed has been successfully developed but as execution of HIV-1 vaccine efficacy trials shifts increasingly to non-subtype B epidemics (Southern African and Southeast Asia), non-subtype B HIV-1 reagents are needed to support vaccine development. Here we describe two IMCs derived from subtypes C and CRF01_AE HIV-1 primary isolates expressing LucR (IMC.LucR) that were engineered to express heterologous gp160 Envs. 18 constructs expressing various subtypes C and CRF01_AE Envs, mostly acute, in subtype-matched and -unmatched HIV backbones were tested for functionality and neutralization sensitivity. Our results suggest a possible effect of non-env HIV-1 genes on the interaction of Env and neutralizing antibodies and highlight the need to generate a library of IMCs representative of the HIV-1 subtype spectrum to be used as standardized neutralization assay reagents for assessing HIV-1 vaccine efficacy.
Collapse
Affiliation(s)
- Agnès-Laurence Chenine
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
- * E-mail:
| | - Lindsay Wieczorek
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
| | - Eric Sanders-Buell
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
| | - Maggie Wesberry
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
| | - Teresa Towle
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
| | - Devin M. Pillis
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
| | - Sebastian Molnar
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
| | - Robert McLinden
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
| | - Tara Edmonds
- University of Alabama, Birmingham, Birmingham, Alabama, United States of America
| | - Ivan Hirsch
- Inserm UMR891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - Robert O’Connell
- Military HIV Research Program, Silver Spring, Maryland, United States of America
- Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | | | | | | | - John C. Kappes
- University of Alabama, Birmingham, Birmingham, Alabama, United States of America
| | - Jerome H. Kim
- Military HIV Research Program, Silver Spring, Maryland, United States of America
- Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Victoria R. Polonis
- Military HIV Research Program, Silver Spring, Maryland, United States of America
- Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Sodsai Tovanabutra
- The Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
- Military HIV Research Program, Silver Spring, Maryland, United States of America
| |
Collapse
|
45
|
Peptide-lipid interactions: experiments and applications. Int J Mol Sci 2013; 14:18758-89. [PMID: 24036440 PMCID: PMC3794806 DOI: 10.3390/ijms140918758] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/27/2013] [Accepted: 08/28/2013] [Indexed: 02/06/2023] Open
Abstract
The interactions between peptides and lipids are of fundamental importance in the functioning of numerous membrane-mediated cellular processes including antimicrobial peptide action, hormone-receptor interactions, drug bioavailability across the blood-brain barrier and viral fusion processes. Moreover, a major goal of modern biotechnology is obtaining new potent pharmaceutical agents whose biological action is dependent on the binding of peptides to lipid-bilayers. Several issues need to be addressed such as secondary structure, orientation, oligomerization and localization inside the membrane. At the same time, the structural effects which the peptides cause on the lipid bilayer are important for the interactions and need to be elucidated. The structural characterization of membrane active peptides in membranes is a harsh experimental challenge. It is in fact accepted that no single experimental technique can give a complete structural picture of the interaction, but rather a combination of different techniques is necessary.
Collapse
|
46
|
Rider MA, Zou J, Vanlandingham D, Nuckols JT, Higgs S, Zhang Q, Lacey M, Kim J, Wang G, Hong YS. Quantitative proteomic analysis of the Anopheles gambiae (Diptera: Culicidae) midgut infected with o'nyong-nyong virus. JOURNAL OF MEDICAL ENTOMOLOGY 2013; 50:1077-1088. [PMID: 24180113 DOI: 10.1603/me12155] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Alphaviruses are arthropod-borne pathogens that infect a range of hosts. In humans and other mammals, alphavirus infection can cause severe disease. In mosquito hosts, however, there are generally few symptoms. Little is known about the cellular responses of mosquitoes that allow them to cope with infection. In this investigation, a six-plex tandem mass tagging proteomic approach was used to study protein accumulation changes in the midgut of Anopheles gambiae (Giles) (Diptera: Culicidae) mosquitoes infected with o'nyong-nyong virus (Togaviridae, Alphavirus). Five hundred thirty-six nonredundant proteins were identified. Twenty-two were found in significantly different quantities in infected midguts compared with controls. Of interest, analysis revealed molecular pathways possibly targeted by virus proteins, such as those involving TAF4 and DNA polymerase phi proteins. Also identified was an FK506-binding protein. FK506-binding protein orthologs have been described as conserved host resistance factors, which suppress dengue and West Nile virus infection in human HeLa cells. This investigation constitutes the first study of the midgut-specific proteome of An. gambiae in relation to alphavirus infection. Our findings offer insight into mosquito immunity, including factors that possibly contribute to the different pathological outcomes observed in vertebrate and insect hosts.
Collapse
Affiliation(s)
- Mark A Rider
- Department of Tropical Medicine, Tulane University, 1430 Tulane Ave, SL-17, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sánchez-Felipe L, Villar E, Muñoz-Barroso I. Entry of Newcastle Disease Virus into the host cell: role of acidic pH and endocytosis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:300-9. [PMID: 23994097 PMCID: PMC7094467 DOI: 10.1016/j.bbamem.2013.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 08/02/2013] [Accepted: 08/13/2013] [Indexed: 12/24/2022]
Abstract
Most paramyxoviruses enter the cell by direct fusion of the viral envelope with the plasma membrane. Our previous studies have shown the colocalization of Newcastle Disease Virus (NDV) with the early endosome marker EEA1 and the inhibition of NDV fusion by the caveolin-phosphorylating drug phorbol 12-myristate 13-acetate (PMA) prompted us to propose that NDV enters the cells via endocytosis. Here we show that the virus-cell fusion and cell-cell fusion promoted by NDV-F are increased by about 30% after brief exposure to low pH in HeLa and ELL-0 cells but not in NDV receptor- deficient cell lines such as GM95 or Lec1. After a brief low-pH exposure, the percentage of NDV fusion at 29 °C was similar to that at 37 °C without acid-pH stimulation, meaning that acid pH would decrease the energetic barrier to enhance fusion. Furthermore, preincubation of cells with the protein kinase C inhibitor bisindolylmaleimide led to the inhibition of about 30% of NDV infectivity, suggesting that a population of virus enters cells through receptor-mediated endocytosis. Moreover, the involvement of the GTPase dynamin in NDV entry is shown as its specific inhibitor, dynasore, also impaired NDV fusion and infectivity. Optimal infection of the host cells was significantly affected by drugs that inhibit endosomal acidification such as concanamycin A, monensin and chloroquine. These results support our hypothesis that entry of NDV into ELL-0 and HeLa cells occurs through the plasma membrane as well as by dynamin- low pH- and receptor- dependent endocytosis. A pulse of low-pH enhanced NDV fusion and infectivity in a cell-dependent manner. NDV infectivity was impaired by a protein kinase C inhibitor. A specific inhibitor of the GTPase dynamin impaired NDV fusion and infectivity. Inhibition of endosomal acidification inhibited NDV fusion and infectivity. NDV may enter by dynamin-acid- and receptor-dependent endocytosis.
Collapse
Affiliation(s)
- Lorena Sánchez-Felipe
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab. 106/108, Plaza Doctores de la Reina s/n, 37007 Salamanca, Spain
| | | | | |
Collapse
|
48
|
Petrareanu C, Macovei A, Sokolowska I, Woods AG, Lazar C, Radu GL, Darie CC, Branza-Nichita N. Comparative proteomics reveals novel components at the plasma membrane of differentiated HepaRG cells and different distribution in hepatocyte- and biliary-like cells. PLoS One 2013; 8:e71859. [PMID: 23977166 PMCID: PMC3748114 DOI: 10.1371/journal.pone.0071859] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 07/04/2013] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus (HBV) is a human pathogen causing severe liver disease and eventually death. Despite important progress in deciphering HBV internalization, the early virus-cell interactions leading to infection are not known. HepaRG is a human bipotent liver cell line bearing the unique ability to differentiate towards a mixture of hepatocyte- and biliary-like cells. In addition to expressing metabolic functions normally found in liver, differentiated HepaRG cells support HBV infection in vitro, thus resembling cultured primary hepatocytes more than other hepatoma cells. Therefore, extensive characterization of the plasma membrane proteome from HepaRG cells would allow the identification of new cellular factors potentially involved in infection. Here we analyzed the plasma membranes of non-differentiated and differentiated HepaRG cells using nanoliquid chromatography-tandem mass spectrometry to identify the differences between the proteomes and the changes that lead to differentiation of these cells. We followed up on differentially-regulated proteins in hepatocytes- and biliary-like cells, focusing on Cathepsins D and K, Cyclophilin A, Annexin 1/A1, PDI and PDI A4/ERp72. Major differences between the two proteomes were found, including differentially regulated proteins, protein-protein interactions and intracellular localizations following differentiation. The results advance our current understanding of HepaRG differentiation and the unique properties of these cells.
Collapse
Affiliation(s)
- Catalina Petrareanu
- Department of Viral Glycoproteins, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
- Department of Analytical Chemistry and Enviromental Engineering, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, Bucharest, Romania
| | - Alina Macovei
- Department of Viral Glycoproteins, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Izabela Sokolowska
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York, United States of America
| | - Alisa G. Woods
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York, United States of America
| | - Catalin Lazar
- Department of Viral Glycoproteins, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Gabriel L. Radu
- Department of Analytical Chemistry and Enviromental Engineering, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, Bucharest, Romania
| | - Costel C. Darie
- Biochemistry and Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York, United States of America
| | - Norica Branza-Nichita
- Department of Viral Glycoproteins, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
- * E-mail:
| |
Collapse
|
49
|
Lavialle C, Cornelis G, Dupressoir A, Esnault C, Heidmann O, Vernochet C, Heidmann T. Paleovirology of 'syncytins', retroviral env genes exapted for a role in placentation. Philos Trans R Soc Lond B Biol Sci 2013; 368:20120507. [PMID: 23938756 DOI: 10.1098/rstb.2012.0507] [Citation(s) in RCA: 271] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The development of the emerging field of 'paleovirology' allows biologists to reconstruct the evolutionary history of fossil endogenous retroviral sequences integrated within the genome of living organisms and has led to the retrieval of conserved, ancient retroviral genes 'exapted' by ancestral hosts to fulfil essential physiological roles, syncytin genes being undoubtedly among the most remarkable examples of such a phenomenon. Indeed, syncytins are 'new' genes encoding proteins derived from the envelope protein of endogenous retroviral elements that have been captured and domesticated on multiple occasions and independently in diverse mammalian species, through a process of convergent evolution. Knockout of syncytin genes in mice provided evidence for their absolute requirement for placenta development and embryo survival, via formation by cell-cell fusion of syncytial cell layers at the fetal-maternal interface. These genes of exogenous origin, acquired 'by chance' and yet still 'necessary' to carry out a basic function in placental mammals, may have been pivotal in the emergence of mammalian ancestors with a placenta from egg-laying animals via the capture of a founding retroviral env gene, subsequently replaced in the diverse mammalian lineages by new env-derived syncytin genes, each providing its host with a positive selective advantage.
Collapse
Affiliation(s)
- Christian Lavialle
- UMR 8122, Unité des Rétrovirus Endogènes et Éléments Rétroïdes des Eucaryotes Supérieurs, CNRS, Institut Gustave Roussy, , 94805 Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Qin ZL, Ju HP, Liu Y, Gao TT, Wang WB, Aurelian L, Zhao P, Qi ZT. Fetal bovine serum inhibits hepatitis C virus attachment to host cells. J Virol Methods 2013; 193:261-9. [PMID: 23845899 DOI: 10.1016/j.jviromet.2013.06.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 06/08/2013] [Accepted: 06/14/2013] [Indexed: 12/22/2022]
Abstract
Fetal bovine serum (FBS), used normally as a basic cell culture supplement, inhibits influenza virus growth. However, the role of FBS in the regulation of hepatitis C virus (HCV) infection has not been studied extensively and remains largely unclear. We adopted the established cell-cultured HCV (HCVcc) isolated from the JFH-1 strain and two sets of solutions (cDMEM7.4 and cDMEM6.8; RHMNB6.8 and RHMN6.8) to investigate the effect of FBS on HCV infection. Our data indicate that FBS blocks HCV infection in a dose-dependent manner. The infectivity of HCV diluted in the RHMNB solution was more susceptible to the addition of FBS than that diluted in the cDMEM solution. In addition, FBS-mediated blocking of HCV infection occurred at the step of virus attachment to the target cells, suggesting that FBS contains factors that interfere with the early steps in HCV infection.
Collapse
Affiliation(s)
- Zhao-ling Qin
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | | | | | |
Collapse
|