1
|
Zhang Z, Xing J, Tang X, Sheng X, Chi H, Zhan W. Nectin1 is a pivotal host factor involved in attachment and entry of red-spotted grouper nervous necrosis virus in the early stages of the viral life cycle. J Virol 2024; 98:e0090124. [PMID: 39194240 PMCID: PMC11406929 DOI: 10.1128/jvi.00901-24] [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: 05/23/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Nervous necrosis virus (NNV) is a highly neurotropic virus that poses a persistent threat to the survival of multiple fish species. However, its inimitable neuropathogenesis remains largely elusive. To rummage potential partners germane to the nervous system, we investigated the interaction between red-spotted grouper NNV (RGNNV) and grouper brain by immunoprecipitation coupled with mass spectrometry and discerned Nectin1 as a novel host factor subtly involved in viral early invasion events. Nectin1 was abundant in neural tissues and implicated in the inception of tunnel nanotubes triggered by RGNNV. Its overexpression not only dramatically potentiated the replication dynamics of RGNNV in susceptible cells, but also empowered non-sensitive cells to expeditiously capture free virions within 2 min. This potency was impervious to low temperatures but was dose-dependently suppressed by soluble protein or specific antibody of Nectin1 ectodomain, indicating Nectin1 as an attachment receptor for RGNNV. Mechanistically, efficient hijacking of virions by Nectin1 strictly depended on intricate linkages to different modules of viral capsid protein, especially the direct binding between the IgC1 loop and P-domain. More strikingly, despite abortive proliferation in Nectin1-reconstructed CHSE-214 cells, a non-sensitive cell, RGNNV could gain access to the intracellular compartment by capitalizing on Nectin1, thereby inducing canonical cytoplasmic vacuolation. Altogether, our findings delineate a candidate entrance for RGNNV infiltration into the nervous system, which may shed unprecedented insights into the exploration and elucidation of RGNNV pathogenesis.IMPORTANCENervous necrosis virus (NNV) is one of the most virulent pathogens in the aquaculture industry, which inflicts catastrophic damage to ecology, environment, and economy annually around the world. Nevertheless, its idiosyncratic invasion and latency mechanisms pose enormous hardships to epidemic prevention and control. In this study, deploying grouper brain as a natural screening library, a single-transmembrane glycoprotein, Nectin1, was first identified as an emergent functional receptor for red-spotted grouper NNV (RGNNV) that widely allocated in nervous tissues and directly interacted with viral capsid protein through distinct Ig-like loops to bridge virus-host crosstalk, apprehend free virions, and concomitantly propel viral entry. Our findings illuminate the critical role of Nectin1 in RGNNV attachment and entry and provide a potential target for future clinical intervention strategies in the therapeutic race against RGNNV.
Collapse
Affiliation(s)
- Zhiqi Zhang
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
2
|
Lecoultre M, Walker PR, El Helali A. Oncolytic virus and tumor-associated macrophage interactions in cancer immunotherapy. Clin Exp Med 2024; 24:202. [PMID: 39196415 PMCID: PMC11358230 DOI: 10.1007/s10238-024-01443-8] [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: 05/30/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024]
Abstract
Oncolytic viruses (OV) are a promising strategy in cancer immunotherapy. Their capacity to promote anti-tumoral immunity locally raises hope that cancers unresponsive to current immunotherapy approaches could be tackled more efficiently. In this context, tumor-associated macrophages (TAM) must be considered because of their pivotal role in cancer immunity. Even though TAM tend to inhibit anti-tumoral responses, their ability to secrete pro-inflammatory cytokines and phagocytose cancer cells can be harnessed to promote therapeutic cancer immunity. OVs have the potential to promote TAM pro-inflammatory functions that favor anti-tumoral immunity. But in parallel, TAM pro-inflammatory functions induce OV clearance in the tumor, thereby limiting OV efficacy and highlighting that the interaction between OV and TAM is a double edge sword. Moreover, engineered OVs were recently developed to modulate specific TAM functions such as phagocytic activity. The potential of circulating monocytes to deliver OV into the tumor after intravenous administration is also emerging. In this review, we will present the interaction between OV and TAM, the potential of engineered OV to modulate specific TAM functions, and the promising role of circulating monocytes in OV delivery to the tumor.
Collapse
Affiliation(s)
- Marc Lecoultre
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, China
- Division of General Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Paul R Walker
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Immunobiology of Brain Tumours Laboratory, Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland
| | - Aya El Helali
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, China.
| |
Collapse
|
3
|
Elste J, Saini A, Mejia-Alvarez R, Mejía A, Millán-Pacheco C, Swanson-Mungerson M, Tiwari V. Significance of Artificial Intelligence in the Study of Virus-Host Cell Interactions. Biomolecules 2024; 14:911. [PMID: 39199298 PMCID: PMC11352483 DOI: 10.3390/biom14080911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
A highly critical event in a virus's life cycle is successfully entering a given host. This process begins when a viral glycoprotein interacts with a target cell receptor, which provides the molecular basis for target virus-host cell interactions for novel drug discovery. Over the years, extensive research has been carried out in the field of virus-host cell interaction, generating a massive number of genetic and molecular data sources. These datasets are an asset for predicting virus-host interactions at the molecular level using machine learning (ML), a subset of artificial intelligence (AI). In this direction, ML tools are now being applied to recognize patterns in these massive datasets to predict critical interactions between virus and host cells at the protein-protein and protein-sugar levels, as well as to perform transcriptional and translational analysis. On the other end, deep learning (DL) algorithms-a subfield of ML-can extract high-level features from very large datasets to recognize the hidden patterns within genomic sequences and images to develop models for rapid drug discovery predictions that address pathogenic viruses displaying heightened affinity for receptor docking and enhanced cell entry. ML and DL are pivotal forces, driving innovation with their ability to perform analysis of enormous datasets in a highly efficient, cost-effective, accurate, and high-throughput manner. This review focuses on the complexity of virus-host cell interactions at the molecular level in light of the current advances of ML and AI in viral pathogenesis to improve new treatments and prevention strategies.
Collapse
Affiliation(s)
- James Elste
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (M.S.-M.)
| | - Akash Saini
- Hinsdale Central High School, 5500 S Grant St, Hinsdale, IL 60521, USA;
| | - Rafael Mejia-Alvarez
- Department of Physiology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA;
| | - Armando Mejía
- Departamento de Biotechnology, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico 09340, Mexico;
| | - Cesar Millán-Pacheco
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad No. 1001, Col Chamilpa, Cuernavaca 62209, Mexico;
| | - Michelle Swanson-Mungerson
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (M.S.-M.)
| | - Vaibhav Tiwari
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (M.S.-M.)
| |
Collapse
|
4
|
Luo S, Liang J, Yang G, Lu J, Chen J. The laminin receptor is a receptor for Micropterus salmoides rhabdovirus. J Virol 2024; 98:e0069724. [PMID: 38916400 PMCID: PMC11265286 DOI: 10.1128/jvi.00697-24] [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: 04/17/2024] [Accepted: 06/01/2024] [Indexed: 06/26/2024] Open
Abstract
Micropterus salmoides rhabdovirus (MSRV) is an important pathogen of largemouth bass. Despite extensive research, the functional receptors of MSRV remained unknown. This study identified the host protein, laminin receptor (LamR), as a cellular receptor facilitating MSRV entry into host cells. Our results demonstrated that LamR directly interacts with MSRV G protein, playing a pivotal role in the attachment and internalization processes of MSRV. Knockdown of LamR with siRNA, blocking cells with LamR antibody, or incubating MSRV virions with soluble LamR protein significantly reduced MSRV entry. Notably, we found that LamR mediated MSRV entry via clathrin-mediated endocytosis. Additionally, our findings revealed that MSRV G and LamR were internalized into cells and co-localized in the early and late endosomes. These findings highlight the significance of LamR as a cellular receptor facilitating MSRV binding and entry into target cells through interaction with the MSRV G protein. IMPORTANCE Despite the serious epidemic caused by Micropterus salmoides rhabdovirus (MSRV) in largemouth bass, the precise mechanism by which it invades host cells remains unclear. Here, we determined that laminin receptor (LamR) is a novel target of MSRV, that interacts with its G protein and is involved in viral attachment and internalization, transporting with MSRV together in early and late endosomes. This is the first report demonstrating that LamR is a cellular receptor in the MSRV life cycle, thus contributing new insights into host-pathogen interactions.
Collapse
Affiliation(s)
- Sheng Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, China
| | - Jiahui Liang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, China
| | - Guanjun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, China
| | - Jianfei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, China
| |
Collapse
|
5
|
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
|
6
|
Katz M, Diskin R. The underlying mechanisms of arenaviral entry through matriglycan. Front Mol Biosci 2024; 11:1371551. [PMID: 38516183 PMCID: PMC10955480 DOI: 10.3389/fmolb.2024.1371551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
Matriglycan, a recently characterized linear polysaccharide, is composed of alternating xylose and glucuronic acid subunits bound to the ubiquitously expressed protein α-dystroglycan (α-DG). Pathogenic arenaviruses, like the Lassa virus (LASV), hijack this long linear polysaccharide to gain cellular entry. Until recently, it was unclear through what mechanisms LASV engages its matriglycan receptor to initiate infection. Additionally, how matriglycan is synthesized onto α-DG by the Golgi-resident glycosyltransferase LARGE1 remained enigmatic. Recent structural data for LARGE1 and for the LASV spike complex informs us about the synthesis of matriglycan as well as its usage as an entry receptor by arenaviruses. In this review, we discuss structural insights into the system of matriglycan generation and eventual recognition by pathogenic viruses. We also highlight the unique usage of matriglycan as a high-affinity host receptor compared with other polysaccharides that decorate cells.
Collapse
Affiliation(s)
| | - Ron Diskin
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
7
|
Xia Z, Long D, Hong X, Lan Y, Xie L. Tissue expression of porcine transient receptor potential mucolipin protein channels and their differential responses to porcine reproductive and respiratory syndrome virus infection in vitro. J Vet Res 2024; 68:45-53. [PMID: 38525220 PMCID: PMC10960329 DOI: 10.2478/jvetres-2024-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction Porcine reproductive and respiratory syndrome virus (PRRSV) infection results in a serious disease, posing a huge economic threat to the global swine industry. The transient receptor potential mucolipin proteins (TRPMLs) have been shown to be strongly associated with virus infection and other physiological processes in humans, but their tissue distribution and responses to PRRSV in pigs remain unknown. Material and Methods Quantitative reverse-transcription PCR analysis was undertaken to determine the optimal primer for TRPML expression detection and for quantifying that expression individually in different pig tissue samples. Meat Animal Research Center 145 (MARC-145) monkey kidney cells and the TRPML-specific activator mucolipin synthetic agonist 1 (ML-SA1) were used to reveal the relationship between TRPML and PRRSV-2 infection. Results The best primers for each TRPML gene used in a fluorescence-based quantitative method were identified and TRPML1 was found to be highly expressed in the kidneys and liver of pigs, while TRPML2 and TRPML3 were observed to be primarily expressed in the kidney and spleen tissues. The expression of TRPML2 was upregulated with the rise in PRRSV-2 infection titre but not the expression of TRPML1 or TRPML3, and ML-SA1 inhibited PRRSV-2 in a dose-dependent manner. Conclusion Our research revealed the gene expression of TRPMLs in pigs and identified that TRPML channels may act as key host factors against PRRSV infection, which could lead to new targets for the prevention and treatment of pig infectious diseases.
Collapse
Affiliation(s)
- Zhiqiang Xia
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian463000, China
- Zhumadian Huazhong Chia Tai Co., Ltd., Zhumadian463000, China
- Henan Topfond Pharmaceutical Company Limited, Zhumadian463000, China
| | - Denggao Long
- Sixteenth Middle School of Yiyang City, Yiyang413064, Hunan Province, China
| | - Xinyi Hong
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian463000, China
| | - Ying Lan
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian463000, China
| | - Lixia Xie
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian463000, China
| |
Collapse
|
8
|
Korosec CS, Wahl LM, Heffernan JM. Within-host evolution of SARS-CoV-2: how often are de novo mutations transmitted from symptomatic infections? Virus Evol 2024; 10:veae006. [PMID: 38425472 PMCID: PMC10904108 DOI: 10.1093/ve/veae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/20/2023] [Accepted: 01/12/2024] [Indexed: 03/02/2024] Open
Abstract
Despite a relatively low mutation rate, the large number of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections has allowed for substantial genetic change, leading to a multitude of emerging variants. Using a recently determined mutation rate (per site replication), as well as within-host parameter estimates for symptomatic SARS-CoV-2 infection, we apply a stochastic transmission-bottleneck model to describe the survival probability of de novo SARS-CoV-2 mutations as a function of bottleneck size and selection coefficient. For narrow bottlenecks, we find that mutations affecting per-target-cell attachment rate (with phenotypes associated with fusogenicity and ACE2 binding) have similar transmission probabilities to mutations affecting viral load clearance (with phenotypes associated with humoral evasion). We further find that mutations affecting the eclipse rate (with phenotypes associated with reorganization of cellular metabolic processes and synthesis of viral budding precursor material) are highly favoured relative to all other traits examined. We find that mutations leading to reduced removal rates of infected cells (with phenotypes associated with innate immune evasion) have limited transmission advantage relative to mutations leading to humoral evasion. Predicted transmission probabilities, however, for mutations affecting innate immune evasion are more consistent with the range of clinically estimated household transmission probabilities for de novo mutations. This result suggests that although mutations affecting humoral evasion are more easily transmitted when they occur, mutations affecting innate immune evasion may occur more readily. We examine our predictions in the context of a number of previously characterized mutations in circulating strains of SARS-CoV-2. Our work offers both a null model for SARS-CoV-2 mutation rates and predicts which aspects of viral life history are most likely to successfully evolve, despite low mutation rates and repeated transmission bottlenecks.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON M3J 1P3, Canada
| | - Lindi M Wahl
- Applied Mathematics, Western University, 1151 Richmond St, London, ON N6A 5B7, Canada
| | - Jane M Heffernan
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
9
|
Uckeley ZM, Duboeuf M, Gu Y, Erny A, Mazelier M, Lüchtenborg C, Winter SL, Schad P, Mathieu C, Koch J, Boulant S, Chlanda P, Maisse C, Brügger B, Lozach PY. Glucosylceramide in bunyavirus particles is essential for virus binding to host cells. Cell Mol Life Sci 2024; 81:71. [PMID: 38300320 PMCID: PMC10834583 DOI: 10.1007/s00018-023-05103-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 02/02/2024]
Abstract
Hexosylceramides (HexCer) are implicated in the infection process of various pathogens. However, the molecular and cellular functions of HexCer in infectious cycles are poorly understood. Investigating the enveloped virus Uukuniemi (UUKV), a bunyavirus of the Phenuiviridae family, we performed a lipidomic analysis with mass spectrometry and determined the lipidome of both infected cells and derived virions. We found that UUKV alters the processing of HexCer to glycosphingolipids (GSL) in infected cells. The infection resulted in the overexpression of glucosylceramide (GlcCer) synthase (UGCG) and the specific accumulation of GlcCer and its subsequent incorporation into viral progeny. UUKV and several pathogenic bunyaviruses relied on GlcCer in the viral envelope for binding to various host cell types. Overall, our results indicate that GlcCer is a structural determinant of virions crucial for bunyavirus infectivity. This study also highlights the importance of glycolipids on virions in facilitating interactions with host cell receptors and infectious entry of enveloped viruses.
Collapse
Affiliation(s)
- Zina M Uckeley
- Center for Integrative Infectious Diseases Research (CIID), University Hospital Heidelberg, 69120, Heidelberg, Germany
- Cluster of Excellence, CellNetworks, 69120, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Department for Molecular Genetics and Microbiology, University of Florida, Gainesville, USA
| | - Maëva Duboeuf
- Université Claude Bernard Lyon 1, INRAE, EPHE, IVPC UMR754, Team iWays, 69007, Lyon, France
| | - Yu Gu
- Université Claude Bernard Lyon 1, INRAE, EPHE, IVPC UMR754, Team iWays, 69007, Lyon, France
| | - Alexandra Erny
- Université Claude Bernard Lyon 1, INRAE, EPHE, IVPC UMR754, Team iWays, 69007, Lyon, France
| | - Magalie Mazelier
- Center for Integrative Infectious Diseases Research (CIID), University Hospital Heidelberg, 69120, Heidelberg, Germany
- Cluster of Excellence, CellNetworks, 69120, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | | | - Sophie L Winter
- Center for Integrative Infectious Diseases Research (CIID), University Hospital Heidelberg, 69120, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Schaller Research Groups, Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - Paulina Schad
- Center for Integrative Infectious Diseases Research (CIID), University Hospital Heidelberg, 69120, Heidelberg, Germany
- Cluster of Excellence, CellNetworks, 69120, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Cyrille Mathieu
- CIRI (Centre International de Recherche en Infectiologie), Team Neuro-Invasion, TROpism and VIRal Encephalitis, INSERM U1111, CNRS UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Jana Koch
- Center for Integrative Infectious Diseases Research (CIID), University Hospital Heidelberg, 69120, Heidelberg, Germany
- Cluster of Excellence, CellNetworks, 69120, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Université Claude Bernard Lyon 1, INRAE, EPHE, IVPC UMR754, Team iWays, 69007, Lyon, France
| | - Steeve Boulant
- Department for Molecular Genetics and Microbiology, University of Florida, Gainesville, USA
| | - Petr Chlanda
- Center for Integrative Infectious Diseases Research (CIID), University Hospital Heidelberg, 69120, Heidelberg, Germany
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Schaller Research Groups, Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carine Maisse
- Université Claude Bernard Lyon 1, INRAE, EPHE, IVPC UMR754, Team iWays, 69007, Lyon, France
| | - Britta Brügger
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Pierre-Yves Lozach
- Center for Integrative Infectious Diseases Research (CIID), University Hospital Heidelberg, 69120, Heidelberg, Germany.
- Cluster of Excellence, CellNetworks, 69120, Heidelberg, Germany.
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120, Heidelberg, Germany.
- Université Claude Bernard Lyon 1, INRAE, EPHE, IVPC UMR754, Team iWays, 69007, Lyon, France.
| |
Collapse
|
10
|
Rubio A, de Toro M, Pérez-Pulido AJ. The most exposed regions of SARS-CoV-2 structural proteins are subject to strong positive selection and gene overlap may locally modify this behavior. mSystems 2024; 9:e0071323. [PMID: 38095866 PMCID: PMC10804949 DOI: 10.1128/msystems.00713-23] [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: 07/10/2023] [Accepted: 11/10/2023] [Indexed: 12/22/2023] Open
Abstract
The SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) pandemic that emerged in 2019 has been an unprecedented event in international science, as it has been possible to sequence millions of genomes, tracking their evolution very closely. This has enabled various types of secondary analyses of these genomes, including the measurement of their sequence selection pressure. In this work, we have been able to measure the selective pressure of all the described SARS-CoV-2 genes, even analyzed by sequence regions, and we show how this type of analysis allows us to separate the genes between those subject to positive selection (usually those that code for surface proteins or those exposed to the host immune system) and those subject to negative selection because they require greater conservation of their structure and function. We have also seen that when another gene with an overlapping reading frame appears within a gene sequence, the overlapping sequence between the two genes evolves under a stronger purifying selection than the average of the non-overlapping regions of the main gene. We propose this type of analysis as a useful tool for locating and analyzing all the genes of a viral genome when an adequate number of sequences are available.IMPORTANCEWe have analyzed the selection pressure of all severe acute respiratory syndrome coronavirus 2 genes by means of the nonsynonymous (Ka) to synonymous (Ks) substitution rate. We found that protein-coding genes are exposed to strong positive selection, especially in the regions of interaction with other molecules (host receptor and genome of the virus itself). However, overlapping coding regions are more protected and show negative selection. This suggests that this measure could be used to study viral gene function as well as overlapping genes.
Collapse
Affiliation(s)
- Alejandro Rubio
- Faculty of Experimental Sciences, Genetics Area, Andalusian Centre for Developmental Biology (CABD, UPO-CSIC-JA), University Pablo de Olavide, Sevilla, Spain
| | - Maria de Toro
- Genomics and Bioinformatics Core Facility, Center for Biomedical Research of La Rioja, Logroño, Spain
| | - Antonio J. Pérez-Pulido
- Faculty of Experimental Sciences, Genetics Area, Andalusian Centre for Developmental Biology (CABD, UPO-CSIC-JA), University Pablo de Olavide, Sevilla, Spain
| |
Collapse
|
11
|
Stanifer ML, Karst SM, Boulant S. Regionalization of the antiviral response in the gastrointestinal tract to provide spatially controlled host/pathogen interactions. mBio 2023; 14:e0279122. [PMID: 37260237 PMCID: PMC10470817 DOI: 10.1128/mbio.02791-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/12/2023] [Indexed: 06/02/2023] Open
Abstract
As the largest mucosal surface, the gastrointestinal (GI) tract plays a key role in protecting the host against pathogen infections. It is a first line of defense against enteric viruses and must act to control infection while remaining tolerant to the high commensal bacteria load found within the GI tract. The GI tract can be divided into six main sections (stomach, duodenum, jejunum, ileum, colon, and rectum), and enteric pathogens have evolved to infect distinct parts of the GI tract. The intestinal epithelial cells (IECs) lining the GI tract are immune competent and can counteract these infections through their intrinsic immune response. Type I and type III interferons (IFNs) are antiviral cytokines that play a key role in protecting IECs against viruses with the type III IFN being the most important. Recent work has shown that IECs derived from the different sections of the GI tract display a unique expression of pattern recognition receptors used to fight pathogen infections. Additionally, it was also shown that these cells show a section-specific response to enteric viruses. This mini-review will discuss the molecular strategies used by IECs to detect and combat enteric viruses highlighting the differences existing along the entero-caudal axis of the GI tract. We will provide a perspective on how these spatially controlled mechanisms may influence virus tropism and discuss how the intestinal micro-environment may further shape the response of IECs to virus infections.
Collapse
Affiliation(s)
- Megan L. Stanifer
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Stephanie M. Karst
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Steeve Boulant
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
12
|
Al Mashud MA, Kumer A, Mukerjee N, Chandro A, Maitra S, Chakma U, Dey A, Akash S, Alexiou A, Khan AA, Alanazi AM, Ghosh A, Chen KT, Sharma R. Mechanistic inhibition of Monkeypox and Marburg virus infection by O-rhamnosides and Kaempferol-o-rhamnosides derivatives: a new-fangled computational approach. Front Cell Infect Microbiol 2023; 13:1188763. [PMID: 37293201 PMCID: PMC10245557 DOI: 10.3389/fcimb.2023.1188763] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/26/2023] [Indexed: 06/10/2023] Open
Abstract
The increasing incidence of Monkeypox virus (Mpox) and Marburg virus (MARV) infections worldwide presents a significant challenge to global health, as limited treatment options are currently available. This study investigates the potential of several O-rhamnosides and Kaempferol-O-rhamnosides as Mpox and MARV inhibitors using molecular modeling methods, including ADMET, molecular docking, and molecular dynamics/MD simulation. The effectiveness of these compounds against the viruses was assessed using the Prediction of Activity Spectra for Substances (PASS) prediction. The study's primary focus is molecular docking prediction, which demonstrated that ligands (L07, L08, and L09) bind to Mpox (PDB ID: 4QWO) and MARV (PDB ID: 4OR8) with binding affinities ranging from -8.00 kcal/mol to -9.5 kcal/mol. HOMO-LUMO based quantum calculations were employed to determine the HOMO-LUMO gap of frontier molecular orbitals (FMOs) and to estimate chemical potential, electronegativity, hardness, and softness. Drug similarity and ADMET prediction assessments of pharmacokinetic properties revealed that the compounds were likely non-carcinogenic, non-hepatotoxic, and rapidly soluble. Molecular dynamic (MD) modeling was used to identify the most favorable docked complexes involving bioactive chemicals. MD simulations indicate that varying types of kaempferol-O-rhamnoside are necessary for successful docking validation and maintaining the stability of the docked complex. These findings could facilitate the discovery of novel therapeutic agents for treating illnesses caused by the Mpox and MARV viruses.
Collapse
Affiliation(s)
- Md. Abdullah Al Mashud
- Biophysics and Biomedicine Research Lab, Department of Electrical & Electronic Engineering, Islamic University, Kushtia, Bangladesh
| | - Ajoy Kumer
- Laboratory of Computational Research for Drug Design and Material Science, Department of Chemistry, European University of Bangladesh, Dhaka, Bangladesh
| | - Nobendu Mukerjee
- Department of Microbiology, West Bengal State University, West Bengal, Kolkata, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Habersham, NSW, Australia
| | - Akhel Chandro
- Department of Poultry Science, Faculty of Animal Science & Veterinary Medicine, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| | - Swastika Maitra
- Department of Microbiology, Adamas University, West Bengal, Kolkata, India
| | - Unesco Chakma
- Laboratory of Computational Research for Drug Design and Material Science, Department of Chemistry, European University of Bangladesh, Dhaka, Bangladesh
- School of Electronic Science and Engineering, Southeast University, Nanjing, China
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Shopnil Akash
- Department of Pharmacy, Daffodil International University, Sukrabad, Dhaka, Bangladesh
| | - Athanasiosis Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Habersham, NSW, Australia
- Department of Neuroscience, AFNP Med, Wien, Austria
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Amer M. Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Arabinda Ghosh
- Microbiology Division, Department of Botany, Gauhati University, Assam, India
| | - Kow-Tong Chen
- Department of Occupational Medicine, Tainan Municipal Hospital (managed by Show Chwan Medical Care Corporation), Tainan, Taiwan
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
13
|
Viruses Binding to Host Receptors Interacts with Autophagy. Int J Mol Sci 2023; 24:ijms24043423. [PMID: 36834833 PMCID: PMC9968160 DOI: 10.3390/ijms24043423] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/19/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Viruses must cross the plasma membrane to infect cells, making them eager to overcome this barrier in order to replicate in hosts. They bind to cell surface receptors as the first step of initiating entry. Viruses can use several surface molecules that allow them to evade defense mechanisms. Various mechanisms are stimulated to defend against viruses upon their entry into cells. Autophagy, one of the defense systems, degrades cellular components to maintain homeostasis. The presence of viruses in the cytosol regulates autophagy; however, the mechanisms by which viral binding to receptors regulates autophagy have not yet been fully established. This review discusses recent findings on autophagy induced by interactions between viruses and receptors. It provides novel perspectives on the mechanism of autophagy as regulated by viruses.
Collapse
|
14
|
SARS-CoV-2 Spike Protein Induces Hemagglutination: Implications for COVID-19 Morbidities and Therapeutics and for Vaccine Adverse Effects. Int J Mol Sci 2022; 23:ijms232415480. [PMID: 36555121 PMCID: PMC9779393 DOI: 10.3390/ijms232415480] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Experimental findings for SARS-CoV-2 related to the glycan biochemistry of coronaviruses indicate that attachments from spike protein to glycoconjugates on the surfaces of red blood cells (RBCs), other blood cells and endothelial cells are key to the infectivity and morbidity of COVID-19. To provide further insight into these glycan attachments and their potential clinical relevance, the classic hemagglutination (HA) assay was applied using spike protein from the Wuhan, Alpha, Delta and Omicron B.1.1.529 lineages of SARS-CoV-2 mixed with human RBCs. The electrostatic potential of the central region of spike protein from these four lineages was studied through molecular modeling simulations. Inhibition of spike protein-induced HA was tested using the macrocyclic lactone ivermectin (IVM), which is indicated to bind strongly to SARS-CoV-2 spike protein glycan sites. The results of these experiments were, first, that spike protein from these four lineages of SARS-CoV-2 induced HA. Omicron induced HA at a significantly lower threshold concentration of spike protein than the three prior lineages and was much more electropositive on its central spike protein region. IVM blocked HA when added to RBCs prior to spike protein and reversed HA when added afterward. These results validate and extend prior findings on the role of glycan bindings of viral spike protein in COVID-19. They furthermore suggest therapeutic options using competitive glycan-binding agents such as IVM and may help elucidate rare serious adverse effects (AEs) associated with COVID-19 mRNA vaccines, which use spike protein as the generated antigen.
Collapse
|
15
|
Mehmood K, Wilczek MP, DuShane JK, Parent MT, Mayberry CL, Wallace JN, Levasseur FL, Fong TM, Hess ST, Maginnis MS. Dynamics and Patterning of 5-Hydroxytryptamine 2 Subtype Receptors in JC Polyomavirus Entry. Viruses 2022; 14:2597. [PMID: 36560603 PMCID: PMC9782046 DOI: 10.3390/v14122597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The organization and dynamics of plasma membrane receptors are a critical link in virus-receptor interactions, which finetune signaling efficiency and determine cellular responses during infection. Characterizing the mechanisms responsible for the active rearrangement and clustering of receptors may aid in developing novel strategies for the therapeutic treatment of viruses. Virus-receptor interactions are poorly understood at the nanoscale, yet they present an attractive target for the design of drugs and for the illumination of viral infection and pathogenesis. This study utilizes super-resolution microscopy and related techniques, which surpass traditional microscopy resolution limitations, to provide both a spatial and temporal assessment of the interactions of human JC polyomavirus (JCPyV) with 5-hydroxytrypamine 2 receptors (5-HT2Rs) subtypes during viral entry. JCPyV causes asymptomatic kidney infection in the majority of the population and can cause fatal brain disease, and progressive multifocal leukoencephalopathy (PML), in immunocompromised individuals. Using Fluorescence Photoactivation Localization Microscopy (FPALM), the colocalization of JCPyV with 5-HT2 receptor subtypes (5-HT2A, 5-HT2B, and 5-HT2C) during viral attachment and viral entry was analyzed. JCPyV was found to significantly enhance the clustering of 5-HT2 receptors during entry. Cluster analysis of infected cells reveals changes in 5-HT2 receptor cluster attributes, and radial distribution function (RDF) analyses suggest a significant increase in the aggregation of JCPyV particles colocalized with 5-HT2 receptor clusters in JCPyV-infected samples. These findings provide novel insights into receptor patterning during viral entry and highlight improved technologies for the future development of therapies for JCPyV infection as well as therapies for diseases involving 5-HT2 receptors.
Collapse
Affiliation(s)
- Kashif Mehmood
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Michael P. Wilczek
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Jeanne K. DuShane
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Matthew T. Parent
- Department of Physics & Astronomy, The University of Maine, Orono, ME 04469, USA
| | - Colleen L. Mayberry
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Jaqulin N. Wallace
- Department of Physics & Astronomy, The University of Maine, Orono, ME 04469, USA
| | - Francois L. Levasseur
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Tristan M. Fong
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
| | - Samuel T. Hess
- Department of Physics & Astronomy, The University of Maine, Orono, ME 04469, USA
- Graduate School of Biomedical Science and Engineering, The University of Maine, Orono, ME 04469, USA
| | - Melissa S. Maginnis
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469, USA
- Graduate School of Biomedical Science and Engineering, The University of Maine, Orono, ME 04469, USA
| |
Collapse
|
16
|
Deffieu MS, Clément CMH, Dorobantu CM, Partiot E, Bare Y, Faklaris O, Rivière B, Ayala-Nunez NV, Baumert TF, Rondé P, Mély Y, Lucansky V, Gaudin R. Occludin stalls HCV particle dynamics apart from hepatocyte tight junctions, promoting virion internalization. Hepatology 2022; 76:1164-1179. [PMID: 35388524 DOI: 10.1002/hep.32514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/28/2022] [Accepted: 04/03/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Numerous HCV entry factors have been identified, and yet information regarding their spatiotemporal dynamics is still limited. Specifically, one of the main entry factors of HCV is occludin (OCLN), a protein clustered at tight junctions (TJs), away from the HCV landing site. Thus, whether HCV particles slide toward TJs or, conversely, OCLN is recruited away from TJs remain debated. APPROACH AND RESULTS Here, we generated CRISPR/CRISPR-associated protein 9 edited Huh7.5.1 cells expressing endogenous levels of enhanced green fluorescent protein/OCLN and showed that incoming HCV particles recruit OCLN outside TJs, independently of claudin 1 (CLDN1) expression, another important HCV entry factor located at TJs. Using ex vivo organotypic culture of hepatic slices obtained from human liver explants, a physiologically relevant model that preserves the overall tissue architecture, we confirmed that HCV associates with OCLN away from TJs. Furthermore, we showed, by live cell imaging, that increased OCLN recruitment beneath HCV particles correlated with lower HCV motility. To decipher the mechanism underlying virus slow-down upon OCLN recruitment, we performed CRISPR knockout (KO) of CLDN1, an HCV entry factor proposed to act upstream of OCLN. Although CLDN1 KO potently inhibits HCV infection, OCLN kept accumulating underneath the particle, indicating that OCLN recruitment is CLDN1 independent. Moreover, inhibition of the phosphorylation of Ezrin, a protein involved in HCV entry that links receptors to the actin cytoskeleton, increased OCLN accumulation and correlated with more efficient HCV internalization. CONCLUSIONS Together, our data provide robust evidence that HCV particles interact with OCLN away from TJs and shed mechanistic insights regarding the manipulation of transmembrane receptor localization by extracellular virus particles.
Collapse
Affiliation(s)
- Maika S Deffieu
- Institut de Recherche en infectiologie de Montpellier (IRIM), CNRS, Montpellier, France.,Université de Montpellier, Montpellier, France
| | - Camille M H Clément
- Institut de Recherche en infectiologie de Montpellier (IRIM), CNRS, Montpellier, France.,Université de Montpellier, Montpellier, France.,Université de Strasbourg, Strasbourg, France.,INSERM, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Cristina M Dorobantu
- Université de Strasbourg, Strasbourg, France.,INSERM, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Emma Partiot
- Institut de Recherche en infectiologie de Montpellier (IRIM), CNRS, Montpellier, France.,Université de Montpellier, Montpellier, France
| | - Yonis Bare
- Institut de Recherche en infectiologie de Montpellier (IRIM), CNRS, Montpellier, France.,Université de Montpellier, Montpellier, France
| | - Orestis Faklaris
- BCM, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Benjamin Rivière
- CHU Montpellier, Laboratoire d'Anatomie et Cytologie Pathologiques-CRB, Montpellier, France
| | - Nilda Vanesa Ayala-Nunez
- Institut de Recherche en infectiologie de Montpellier (IRIM), CNRS, Montpellier, France.,Université de Montpellier, Montpellier, France
| | - Thomas F Baumert
- Université de Strasbourg, Strasbourg, France.,INSERM, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Philippe Rondé
- Université de Strasbourg, Strasbourg, France.,UMR 7021 CNRS, Laboratoire de Bioimagerie et Pathologies, Université de Strasbourg, Faculté de pharmacie, Illkirch, France
| | - Yves Mély
- Université de Strasbourg, Strasbourg, France.,UMR 7021 CNRS, Laboratoire de Bioimagerie et Pathologies, Université de Strasbourg, Faculté de pharmacie, Illkirch, France
| | - Vincent Lucansky
- Université de Strasbourg, Strasbourg, France.,INSERM, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Raphael Gaudin
- Institut de Recherche en infectiologie de Montpellier (IRIM), CNRS, Montpellier, France.,Université de Montpellier, Montpellier, France
| |
Collapse
|
17
|
Lam A, Yuan DS, Ahmed SH, Rawle RJ. Viral Size Modulates Sendai Virus Binding to Cholesterol-Stabilized Receptor Nanoclusters. J Phys Chem B 2022; 126:6802-6810. [PMID: 36001793 PMCID: PMC9484459 DOI: 10.1021/acs.jpcb.2c03830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/12/2022] [Indexed: 11/29/2022]
Abstract
Binding to the host membrane is the initial infection step for animal viruses. Sendai virus (SeV), the model respirovirus studied here, utilizes sialic-acid-conjugated glycoproteins and glycolipids as receptors for binding. In a previous report studying single virus binding to supported lipid bilayers (SLBs), we found a puzzling mechanistic difference between the binding of SeV and influenza A virus (strain X31, IAVX31). Both viruses use similar receptors and exhibit similar cooperative binding behavior, but whereas IAVX31 binding was altered by SLB cholesterol concentration, which can stabilize receptor nanoclusters, SeV was not. Here, we propose that differences in viral size distributions can explain this discrepancy; viral size could alter the number of virus-receptor interactions in the contact area and, therefore, the sensitivity to receptor nanoclusters. To test this, we compared the dependence of SeV binding on SLB cholesterol concentration between size-filtered and unfiltered SeV. At high receptor density, the unfiltered virus showed little dependence, but the size-filtered virus exhibited a linear cholesterol dependence, similar to IAVX31. However, at low receptor densities, the unfiltered virus did exhibit a cholesterol dependence, indicating that receptor nanoclusters enhance viral binding only when the number of potential virus-receptor interactions is small enough. We also studied the influence of viral size and receptor nanoclusters on viral mobility following binding. Whereas differences in viral size greatly influenced mobility, the effect of receptor nanoclusters on mobility was small. Together, our results highlight the mechanistic salience of both the distribution of viral sizes and the lateral distribution of receptors in a viral infection.
Collapse
Affiliation(s)
- Amy Lam
- Department of Chemistry, Williams
College, Williamstown, Massachusetts01267, United States
| | - Daniel S. Yuan
- Department of Chemistry, Williams
College, Williamstown, Massachusetts01267, United States
| | - Samir H. Ahmed
- Department of Chemistry, Williams
College, Williamstown, Massachusetts01267, United States
| | - Robert J. Rawle
- Department of Chemistry, Williams
College, Williamstown, Massachusetts01267, United States
| |
Collapse
|
18
|
Guerreiro SFC, Ferreira CAM, Valente JFA, Patrício TMF, Alves NMF, Dias JR. Electrospun-Based Membranes as a Key Tool to Prevent Respiratory Infections. Polymers (Basel) 2022; 14:3787. [PMID: 36145931 PMCID: PMC9504510 DOI: 10.3390/polym14183787] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 11/24/2022] Open
Abstract
The use of electrospun meshes has been proposed as highly efficient protective equipment to prevent respiratory infections. Those infections can result from the activity of micro-organisms and other small dust particles, such as those resulting from air pollution, that impair the respiratory tract, induce cellular damage and compromise breathing capacity. Therefore, electrospun meshes can contribute to promoting air-breathing quality and controlling the spread of such epidemic-disrupting agents due to their intrinsic characteristics, namely, low pore size, and high porosity and surface area. In this review, the mechanisms behind the pathogenesis of several stressors of the respiratory system are covered as well as the strategies adopted to inhibit their action. The main goal is to discuss the performance of antimicrobial electrospun nanofibers by comparing the results already reported in the literature. Further, the main aspects of the certification of filtering systems are highlighted, and the expected technology developments in the industry are also discussed.
Collapse
Affiliation(s)
- Sara F. C. Guerreiro
- Centre for Rapid and Sustainable Product Development (CDRSP), Instituto Politécnico de Leiria, 2030-028 Marinha Grande, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Medical Physics Department, Portuguese Institute of Oncology (IPO-Porto), 4200-072 Porto, Portugal
| | - Carolina A. M. Ferreira
- Centre for Rapid and Sustainable Product Development (CDRSP), Instituto Politécnico de Leiria, 2030-028 Marinha Grande, Portugal
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA), Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- Marine and Environmental Sciences Centre (MARE), ESTM, Instituto Politécnico de Leiria, 2050-641 Peniche, Portugal
| | - Joana F. A. Valente
- Centre for Rapid and Sustainable Product Development (CDRSP), Instituto Politécnico de Leiria, 2030-028 Marinha Grande, Portugal
| | - Tatiana M. F. Patrício
- Centre for Rapid and Sustainable Product Development (CDRSP), Instituto Politécnico de Leiria, 2030-028 Marinha Grande, Portugal
| | - Nuno M. F. Alves
- Centre for Rapid and Sustainable Product Development (CDRSP), Instituto Politécnico de Leiria, 2030-028 Marinha Grande, Portugal
| | - Juliana R. Dias
- Centre for Rapid and Sustainable Product Development (CDRSP), Instituto Politécnico de Leiria, 2030-028 Marinha Grande, Portugal
| |
Collapse
|
19
|
Guarracino DA, Iannaccone J, Cabrera A, Kancharla S. Harnessing the Therapeutic Potential and Biological Activity of Antiviral Peptides. Chembiochem 2022; 23:e202200415. [PMID: 36075015 DOI: 10.1002/cbic.202200415] [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: 07/21/2022] [Revised: 09/07/2022] [Indexed: 11/09/2022]
Abstract
Peptides are ideal candidates for the development of antiviral therapeutics due to their specificity, chemical diversity and potential for highly potent, safe, molecular interventions. By restricting conformational freedom and flexibility, cyclic peptides frequently increase peptide stability. Viral targets are often very challenging as their evasive strategies for infectivity can preclude standard therapies. In recent years, several peptides from natural sources mitigated an array of viral infections. In parallel, short peptides derived from key viral proteins, modified with chemical groups such as lipids and cell-penetrating sequences, led to highly effective antiviral inhibitor designs. These strategies have been further developed during the recent COVID-19 pandemic caused by the novel coronavirus SARS-CoV-2. Several anti-SARS-CoV-2 peptides are gaining ground in pre-clinical development. Overall, peptides are strong contenders for lead compounds against many life-threatening viruses and may prove to be the key to future efforts revealing viral mechanisms of action and alleviating their effects.
Collapse
Affiliation(s)
| | | | | | - Sneha Kancharla
- The College of New Jersey School of Science, Chemistry, UNITED STATES
| |
Collapse
|
20
|
Abidine Y, Liu L, Wallén O, Trybala E, Olofsson S, Bergström T, Bally M. Cellular Chondroitin Sulfate and the Mucin-like Domain of Viral Glycoprotein C Promote Diffusion of Herpes Simplex Virus 1 While Heparan Sulfate Restricts Mobility. Viruses 2022; 14:v14081836. [PMID: 36016458 PMCID: PMC9412521 DOI: 10.3390/v14081836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 01/26/2023] Open
Abstract
The diffusion of viruses at the cell membrane is essential to reach a suitable entry site and initiate subsequent internalization. Although many viruses take advantage of glycosaminoglycans (GAG) to bind to the cell surface, little is known about the dynamics of the virus–GAG interactions. Here, single-particle tracking of the initial interaction of individual herpes simplex virus 1 (HSV-1) virions reveals a heterogeneous diffusive behavior, regulated by cell-surface GAGs with two main diffusion types: confined and normal free. This study reports that different GAGs can have competing influences in mediating diffusion on the cells used here: chondroitin sulfate (CS) enhances free diffusion but hinders virus attachment to cell surfaces, while heparan sulfate (HS) promotes virus confinement and increases entry efficiency. In addition, the role that the viral mucin-like domains (MLD) of the HSV-1 glycoprotein C plays in facilitating the diffusion of the virus and accelerating virus penetration into cells is demonstrated. Together, our results shed new light on the mechanisms of GAG-regulated virus diffusion at the cell surface for optimal internalization. These findings may be extendable to other GAG-binding viruses.
Collapse
Affiliation(s)
- Yara Abidine
- Department of Clinical Microbiology, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, SE-90185 Umeå, Sweden
| | - Lifeng Liu
- Department of Clinical Microbiology, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, SE-90185 Umeå, Sweden
| | - Oskar Wallén
- Department of Clinical Microbiology, Umeå University, SE-90185 Umeå, Sweden
| | - Edward Trybala
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, SE-41346 Göteborg, Sweden
| | - Sigvard Olofsson
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, SE-41346 Göteborg, Sweden
| | - Tomas Bergström
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, SE-41346 Göteborg, Sweden
| | - Marta Bally
- Department of Clinical Microbiology, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, SE-90185 Umeå, Sweden
- Correspondence:
| |
Collapse
|
21
|
Acosta-Gutiérrez S, Matias D, Avila-Olias M, Gouveia VM, Scarpa E, Forth J, Contini C, Duro-Castano A, Rizzello L, Battaglia G. A Multiscale Study of Phosphorylcholine Driven Cellular Phenotypic Targeting. ACS CENTRAL SCIENCE 2022; 8:891-904. [PMID: 35912343 PMCID: PMC9335915 DOI: 10.1021/acscentsci.2c00146] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Phenotypic targeting requires the ability of the drug delivery system to discriminate over cell populations expressing a particular receptor combination. Such selectivity control can be achieved using multiplexed-multivalent carriers often decorated with multiple ligands. Here, we demonstrate that the promiscuity of a single ligand can be leveraged to create multiplexed-multivalent carriers achieving phenotypic targeting. We show how the cellular uptake of poly(2-(methacryloyloxy)ethyl phosphorylcholine)-poly(2-(diisopropylamino)ethyl methacry-late) (PMPC-PDPA) polymersomes varies depending on the receptor expression among different cells. We investigate the PMPC-PDPA polymersome insertion at the single chain/receptor level using all-atom molecular modeling. We propose a theoretical statistical mechanics-based model for polymersome-cell association that explicitly considers the interaction of the polymersome with the cell glycocalyx shedding light on its effect on the polymersome binding. We validate our model experimentally and show that the binding energy is a nonlinear function, allowing us to tune the interaction by varying the radius and degree of polymerization. Finally, we show that PMPC-PDPA polymersomes can be used to target monocytes in vivo due to their promiscuous interaction with SRB1, CD36, and CD81.
Collapse
Affiliation(s)
- Silvia Acosta-Gutiérrez
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
- Institute
for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Diana Matias
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
| | - Milagros Avila-Olias
- Department
of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, United Kingdom
| | - Virginia M. Gouveia
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
- SomaServe
Ltd U.K., Babraham Research Campus, Cambridge, CB22 3AT, United
Kingdom
| | - Edoardo Scarpa
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
- Department
of Pharmaceutical Sciences, University of
Milan, 20133 Milan, Italy
- INGM,
Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica
Invernizzi”, 20122 Milan, Italy
| | - Joe Forth
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
| | - Claudia Contini
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, W12 0BZ, United Kingdom
| | - Aroa Duro-Castano
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
| | - Loris Rizzello
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
- Department
of Pharmaceutical Sciences, University of
Milan, 20133 Milan, Italy
- INGM,
Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica
Invernizzi”, 20122 Milan, Italy
- Institute
for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Giuseppe Battaglia
- Department
of Chemistry and Institute for the Physics of Living Systems, University
College London, London, WC1H 0AJ, United Kingdom
- Institute
for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Catalan
Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
22
|
Cai J, Zhang BD, Li YQ, Zhu WF, Akihisa T, Kikuchi T, Xu J, Liu WY, Feng F, Zhang J. Cardiac glycosides from the roots of Streblus asper Lour. with activity against Epstein-Barr virus lytic replication. Bioorg Chem 2022; 127:106004. [PMID: 35843015 DOI: 10.1016/j.bioorg.2022.106004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 11/15/2022]
Abstract
Cardiac glycosides (CGs) show potential broad-spectrum antiviral activity by targeting cellular host proteins. Herein are reported the isolation of five new (1-5) and eight known (7-13) CGs from the roots of Streblus asper Lour. Of these compounds 1 and 7 exhibited inhibitory action against EBV early antigen (EA) expression, with half-maximal effective concentration values (EC50) being less than 60 nM, and they also showed selectivity, with selectivity index (SI) values being 56.80 and 103.17, respectively. Preliminary structure activity relationships indicated that the C-10 substituent, C-5 hydroxy groups, and C-3 sugar unit play essential roles in the mediation of the inhibitory activity of CGs against EBV. Further enzyme experiments demonstrated that these compounds might inhibit ion pump function and thereby change the intracellular signal transduction pathway by binding to Na+/K+-ATPase, as validated by simulated molecular docking. This study is the first report that CGs can effectively limit EBV lytic replication, and the observations made in this study may be of value for lead compound development.
Collapse
Affiliation(s)
- Jing Cai
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Bo-Dou Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yu-Qi Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wan-Fang Zhu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Toshihiro Akihisa
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China; Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Takashi Kikuchi
- Faculty of Pharmaceutical Sciences, Toho University, Chiba 274-8510, Japan
| | - Jian Xu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wen-Yuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Feng Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China; Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China
| | - Jie Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China; Jiangsu Food and Pharmaceutical Science College, Huaian 223003, China.
| |
Collapse
|
23
|
Han H, Wang Y, Xu S, Han C, Qin Q, Wei S. High-density lipoproteins negatively regulate innate immunity and facilitate red-spotted grouper nervous necrosis virus entry via scavenger receptor B type 1. Int J Biol Macromol 2022; 215:424-433. [PMID: 35752331 DOI: 10.1016/j.ijbiomac.2022.06.126] [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: 03/29/2022] [Revised: 05/24/2022] [Accepted: 06/17/2022] [Indexed: 11/05/2022]
Abstract
Lipid metabolism plays an important role in viral infections, and it can directly or indirectly affect various stages of viral infection in cells. As an important component of lipid metabolism, high-density lipoprotein (HDL) plays crucial roles in inflammation, immunity, and viral infections. Scavenger receptor B type 1 (SR-B1), a receptor of HDL, cannot be ignored in the regulation of lipid metabolism. Here, we investigate, for the first time, the role of Epinephelus coioides SR-B1 (Ec-SR-B1) in red-spotted grouper nervous necrosis virus (RGNNV) infection. Our results indicate that Ec-SR-B1 could promote RGNNV infection. We also demonstrate that Ec-SR-B1 could facilitate viral entry and interact with capsid protein (CP) of RGNNV. As the natural ligand of SR-B1, HDL significantly increased RGNNV entry in a dose-dependent manner. However, we observed no effect of HDL on Ec-SR-B1 expression. The results of the micro-scale thermophoresis assay did not reveal an association between HDL and CP, suggesting that RGNNV does not enter target cells by using HDL as a ligand to bind to its receptor. In addition, block lipid transport-1, a compound that inhibits HDL-mediated cholesterol transfer, reduced the HDL-induced enhancement of RGNNV infection, indicating a role for lipid transfer in facilitating RGNNV entry. Furthermore, HDL inhibited the expression of pro-inflammatory factors and antiviral genes in a dose-dependent manner. These findings suggest that the HDL-induced enhancement of RGNNV entry involves the complex interplay between Ec-SR-B1, HDL, and RGNNV, as well as the regulation of innate antiviral responses by HDL. In summary, we highlight the crucial role of HDL in RGNNV entry, identify a possible molecular connection between RGNNV and lipoprotein metabolism, and indicate the role of Ec-SR-B1 in RGNNV infection.
Collapse
Affiliation(s)
- Honglin Han
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Yuexuan Wang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Suifeng Xu
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Chengzong Han
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), 528478, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, China.
| | - Shina Wei
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
24
|
Statistics of antibody binding to the spike protein explain the dependence of COVID 19 infection risk on antibody concentration and affinity. Sci Rep 2022; 12:9379. [PMID: 35672372 PMCID: PMC9172616 DOI: 10.1038/s41598-022-13748-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
The increase of COVID-19 breakthrough infection risk with time since vaccination has a clear relationship to the decrease of antibody concentration with time. The empirically-observed dependence on blood IgG anti-receptor binding domain antibody concentration of SARS-CoV-2 vaccine efficacy against infection has a rational explanation in the statistics of binding of antibody to spike proteins on the virus surface, leading to blocking of binding to the receptor: namely that the probability of infection is the probability that a critical number of the spike proteins protruding from the virus are unblocked. The model is consistent with the observed antibody concentrations required to induce immunity and with the observed dependence of vaccine efficacy on antibody concentration and thus is a useful tool in the development of models to relate, for an individual person, risk of infection given measured antibody concentration. It can be used to relate population breakthrough infection risk to the distribution across the population of antibody concentration, and its variation with time.
Collapse
|
25
|
Bhide K, Mochnáčová E, Tkáčová Z, Petroušková P, Kulkarni A, Bhide M. Signaling events evoked by domain III of envelop glycoprotein of tick-borne encephalitis virus and West Nile virus in human brain microvascular endothelial cells. Sci Rep 2022; 12:8863. [PMID: 35614140 PMCID: PMC9133079 DOI: 10.1038/s41598-022-13043-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022] Open
Abstract
Tick-borne encephalitis virus and West Nile virus can cross the blood–brain barrier via hematogenous route. The attachment of a virion to the cells of a neurovascular unit, which is mediated by domain III of glycoprotein E, initiates a series of events that may aid viral entry. Thus, we sought to uncover the post-attachment biological events elicited in brain microvascular endothelial cells by domain III. RNA sequencing of cells treated with DIII of TBEV and WNV showed significant alteration in the expression of 309 and 1076 genes, respectively. Pathway analysis revealed activation of the TAM receptor pathway. Several genes that regulate tight-junction integrity were also activated, including pro-inflammatory cytokines and chemokines, cell-adhesion molecules, claudins, and matrix metalloprotease (mainly ADAM17). Results also indicate activation of a pro-apoptotic pathway. TLR2 was upregulated in both cases, but MyD88 was not. In the case of TBEV DIII, a MyD88 independent pathway was activated. Furthermore, both cases showed dramatic dysregulation of IFN and IFN-induced genes. Results strongly suggest that the virus contact to the cell surface emanates a series of events namely viral attachment and diffusion, breakdown of tight junctions, induction of virus uptake, apoptosis, reorganization of the extracellular-matrix, and activation of the innate immune system.
Collapse
Affiliation(s)
- Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic
| | - Evelína Mochnáčová
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic
| | - Zuzana Tkáčová
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic
| | - Patrícia Petroušková
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic
| | - Amod Kulkarni
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic.,Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic. .,Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovak Republic.
| |
Collapse
|
26
|
Petitjean SJL, Chen W, Koehler M, Jimmidi R, Yang J, Mohammed D, Juniku B, Stanifer ML, Boulant S, Vincent SP, Alsteens D. Multivalent 9-O-Acetylated-sialic acid glycoclusters as potent inhibitors for SARS-CoV-2 infection. Nat Commun 2022; 13:2564. [PMID: 35538121 PMCID: PMC9091252 DOI: 10.1038/s41467-022-30313-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/25/2022] [Indexed: 01/08/2023] Open
Abstract
The recent emergence of highly transmissible SARS-CoV-2 variants illustrates the urgent need to better understand the molecular details of the virus binding to its host cell and to develop anti-viral strategies. While many studies focused on the role of the angiotensin-converting enzyme 2 receptor in the infection, others suggest the important role of cell attachment factors such as glycans. Here, we use atomic force microscopy to study these early binding events with the focus on the role of sialic acids (SA). We show that SARS-CoV-2 binds specifically to 9-O-acetylated-SA with a moderate affinity, supporting its role as an attachment factor during virus landing to cell host surfaces. For therapeutic purposes and based on this finding, we have designed novel blocking molecules with various topologies and carrying a controlled number of SA residues, enhancing affinity through a multivalent effect. Inhibition assays show that the AcSA-derived glycoclusters are potent inhibitors of cell binding and infectivity, offering new perspectives in the treatment of SARS-CoV-2 infection. Cell surface attachment factors, such as glycans, play an important role in viral infection. Here, Petitjean et al. show that SARS-CoV-2 specifically binds to 9-Oacetylated sialic acid and have designed novel inhibitors based on multivalent derivatives.
Collapse
Affiliation(s)
- Simon J L Petitjean
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Wenzhang Chen
- Laboratory of Bio-Organic Chemistry (NARILIS), UNamur, Namur, Belgium
| | - Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Ravikumar Jimmidi
- Laboratory of Bio-Organic Chemistry (NARILIS), UNamur, Namur, Belgium
| | - Jinsung Yang
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Danahe Mohammed
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Blinera Juniku
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Megan L Stanifer
- Dept. of Infectious Diseases, Medical Faculty, Center for Integrative Infectious Diseases Research (CIID), University of Heidelberg, 69120, Heidelberg, Germany.,Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, USA
| | - Steeve Boulant
- Dept. of Infectious Diseases, Medical Faculty, Center for Integrative Infectious Diseases Research (CIID), University of Heidelberg, 69120, Heidelberg, Germany.,Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, USA
| | | | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium. .,Walloon Excellence in Life sciences and Biotechnology (WELBIO), Wavre, Belgium.
| |
Collapse
|
27
|
Kianpour M, Akbarian M, Uversky VN. Nanoparticles for Coronavirus Control. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1602. [PMID: 35564311 PMCID: PMC9104235 DOI: 10.3390/nano12091602] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 01/18/2023]
Abstract
More than 2 years have passed since the SARS-CoV-2 outbreak began, and many challenges that existed at the beginning of this pandemic have been solved. Some countries have been able to overcome this global challenge by relying on vaccines against the virus, and vaccination has begun in many countries. Many of the proposed vaccines have nanoparticles as carriers, and there are different nano-based diagnostic approaches for rapid detection of the virus. In this review article, we briefly examine the biology of SARS-CoV-2, including the structure of the virus and what makes it pathogenic, as well as describe biotechnological methods of vaccine production, and types of the available and published nano-based ideas for overcoming the virus pandemic. Among these issues, various physical and chemical properties of nanoparticles are discussed to evaluate the optimal conditions for the production of the nano-mediated vaccines. At the end, challenges facing the international community and biotechnological answers for future viral attacks are reviewed.
Collapse
Affiliation(s)
- Maryam Kianpour
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Mohsen Akbarian
- Department of Chemistry, National Cheng Kung University, Tainan 701, Taiwan
| | - Vladimir N. Uversky
- Department of Molecular Medicine and Health Byrd Alzheimer’s Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center ‘‘Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences’’, 142290 Pushchino, Moscow Region, Russia
| |
Collapse
|
28
|
Ruiz PAS, Ziebert F, Kulić IM. Physics of self-rolling viruses. Phys Rev E 2022; 105:054411. [PMID: 35706307 DOI: 10.1103/physreve.105.054411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/29/2022] [Indexed: 06/15/2023]
Abstract
Viruses are right at the interface of inanimate matter and life. However, recent experiments [Sakai et al., J. Virol. 92, e01522-17 (2018)0022-538X10.1128/JVI.01522-17] have shown that some influenza strains can actively roll on glycan-covered surfaces. In a previous letter [Ziebert and Kulić, Phys. Rev. Lett. 126, 218101 (2021)0031-900710.1103/PhysRevLett.126.218101] we suggested this to be a form of viral surface metabolism: a collection of spike proteins that attach to and cut the glycans act as a self-organized mechano-chemical motor. Here we study in more depth the physics of the emergent self-rolling states. We give scaling arguments how the motion arises, substantiated by a detailed analytical theory that yields the full torque-angular velocity relation of the self-organized motor. Stochastic Gillespie simulations are used to validate the theory and to quantify stochastic effects like virus detachment and reversals of its direction. Finally, we also cross-check several approximations made previously and show that the proposed mechanism is very robust. All these results point together to the statistical inevitability of viral rolling in the presence of enzymatic activity.
Collapse
Affiliation(s)
- Pedro A Soria Ruiz
- Institute for Theoretical Physics, Heidelberg University, D-69120 Heidelberg, Germany
| | - Falko Ziebert
- Institute for Theoretical Physics, Heidelberg University, D-69120 Heidelberg, Germany
- BioQuant, Heidelberg University, D-69120 Heidelberg, Germany
| | - Igor M Kulić
- Institut Charles Sadron UPR22-CNRS, F-67034 Strasbourg, France
- Institute Theory of Polymers, Leibniz-Institute of Polymer Research, D-01069 Dresden, Germany
| |
Collapse
|
29
|
Morzy D, Bastings M. Significance of Receptor Mobility in Multivalent Binding on Lipid Membranes. Angew Chem Int Ed Engl 2022; 61:e202114167. [PMID: 34982497 PMCID: PMC9303963 DOI: 10.1002/anie.202114167] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/07/2021] [Indexed: 01/16/2023]
Abstract
Numerous key biological processes rely on the concept of multivalency, where ligands achieve stable binding only upon engaging multiple receptors. These processes, like viral entry or immune synapse formation, occur on the diffusive cellular membrane. One crucial, yet underexplored aspect of multivalent binding is the mobility of coupled receptors. Here, we discuss the consequences of mobility in multivalent processes from four perspectives: (I) The facilitation of receptor recruitment by the multivalent ligand due to their diffusivity prior to binding. (II) The effects of receptor preassembly, which allows their local accumulation. (III) The consequences of changes in mobility upon the formation of receptor/ligand complex. (IV) The changes in the diffusivity of lipid environment surrounding engaged receptors. We demonstrate how understanding mobility is essential for fully unravelling the principles of multivalent membrane processes, leading to further development in studies on receptor interactions, and guide the design of new generations of multivalent ligands.
Collapse
Affiliation(s)
- Diana Morzy
- Programmable Biomaterials Laboratory, Institute of Materials, School of Engineering, École Polytechnique Fédérale de Lausanne, Route Cantonale, 1015, Lausanne, Switzerland
| | - Maartje Bastings
- Programmable Biomaterials Laboratory, Institute of Materials, School of Engineering, École Polytechnique Fédérale de Lausanne, Route Cantonale, 1015, Lausanne, Switzerland
| |
Collapse
|
30
|
Multiomics Analysis of Endocytosis upon HBV Infection and Identification of SCAMP1 as a Novel Host Restriction Factor against HBV Replication. Int J Mol Sci 2022; 23:ijms23042211. [PMID: 35216324 PMCID: PMC8874515 DOI: 10.3390/ijms23042211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023] Open
Abstract
Hepatitis B virus (HBV) infection remains a major global health problem and the primary cause of cirrhosis and hepatocellular carcinoma (HCC). HBV intrusion into host cells is prompted by virus–receptor interactions in clathrin-mediated endocytosis. Here, we report a comprehensive view of the cellular endocytosis-associated transcriptome, proteome and ubiquitylome upon HBV infection. In this study, we quantified 273 genes in the transcriptome and 190 endocytosis-associated proteins in the proteome by performing multi-omics analysis. We further identified 221 Lys sites in 77 endocytosis-associated ubiquitinated proteins. A weak negative correlation was observed among endocytosis-associated transcriptome, proteome and ubiquitylome. We found 33 common differentially expressed genes (DEGs), differentially expressed proteins (DEPs), and Kub-sites. Notably, we reported the HBV-induced ubiquitination change of secretory carrier membrane protein (SCAMP1) for the first time, differentially expressed across all three omics data sets. Overexpression of SCAMP1 efficiently inhibited HBV RNAs/pgRNA and secreted viral proteins, whereas knockdown of SCAMP1 significantly increased viral production. Mechanistically, the EnhI/XP, SP1, and SP2 promoters were inhibited by SCAMP1, which accounts for HBV X and S mRNA inhibition. Overall, our study unveils the previously unknown role of SCAMP1 in viral replication and HBV pathogenesis and provides cumulative and novel information for a better understanding of endocytosis in response to HBV infection.
Collapse
|
31
|
Morzy D, Bastings M. Significance of Receptor Mobility in Multivalent Binding on Lipid Membranes. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202114167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Diana Morzy
- Programmable Biomaterials Laboratory Institute of Materials School of Engineering École Polytechnique Fédérale de Lausanne Route Cantonale 1015 Lausanne Switzerland
| | - Maartje Bastings
- Programmable Biomaterials Laboratory Institute of Materials School of Engineering École Polytechnique Fédérale de Lausanne Route Cantonale 1015 Lausanne Switzerland
| |
Collapse
|
32
|
Dong B, Tang N, Guan Y, Qu G, Miao L, Han W, Shen Z. Type and Abundance of Sialic Acid Receptors on Host Cell Membrane Affect Infectivity and Viral Titer of Different Strains of Newcastle Disease Virus. J Virol Methods 2022; 302:114488. [DOI: 10.1016/j.jviromet.2022.114488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 11/24/2022]
|
33
|
Liu C, Wu K, Gao H, Li J, Xu X. Current Strategies and Potential Prospects for Nanoparticle-Mediated Treatment of Diabetic Nephropathy. Diabetes Metab Syndr Obes 2022; 15:2653-2673. [PMID: 36068795 PMCID: PMC9441178 DOI: 10.2147/dmso.s380550] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/20/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetic nephropathy (DN), a severe microvascular complication of diabetes mellitus (DM), is the most common form of chronic kidney disease (CKD) and a leading cause of renal failure in end-stage renal disease. No currently available treatment can achieve complete cure. Traditional treatments have many limitations, such as painful subcutaneous insulin injections, nephrotoxicity and hepatotoxicity with oral medication, and poor patient compliance with continual medication intake. Given the known drawbacks, recent research has suggested that nanoparticle-based drug delivery platforms as therapeutics may provide a promising strategy for treating debilitating diseases such as DN in the future. This administration method provides multiple advantages, such as delivering the loaded drug to the precise target of action and enabling early prevention of CKD progression. This article discusses the development of the main currently used nanoplatforms, such as liposomes, polymeric NPs, and inorganic NPs, as well as the prospects and drawbacks of nanoplatform application in the treatment of CKD.
Collapse
Affiliation(s)
- Chunkang Liu
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Huan Gao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Jianyang Li
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Xiaohua Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
- Correspondence: Xiaohua Xu, Email
| |
Collapse
|
34
|
Pfafenrot C, Schneider T, Müller C, Hung LH, Schreiner S, Ziebuhr J, Bindereif A. Inhibition of SARS-CoV-2 coronavirus proliferation by designer antisense-circRNAs. Nucleic Acids Res 2021; 49:12502-12516. [PMID: 34850109 PMCID: PMC8643703 DOI: 10.1093/nar/gkab1096] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 01/22/2023] Open
Abstract
Circular RNAs (circRNAs) are noncoding RNAs that exist in all eukaryotes investigated and are derived from back-splicing of certain pre-mRNA exons. Here, we report the application of artificial circRNAs designed to act as antisense-RNAs. We systematically tested a series of antisense-circRNAs targeted to the SARS-CoV-2 genome RNA, in particular its structurally conserved 5'-untranslated region. Functional assays with both reporter transfections as well as with SARS-CoV-2 infections revealed that specific segments of the SARS-CoV-2 5'-untranslated region can be efficiently accessed by specific antisense-circRNAs, resulting in up to 90% reduction of virus proliferation in cell culture, and with a durability of at least 48 h. Presenting the antisense sequence within a circRNA clearly proved more efficient than in the corresponding linear configuration and is superior to modified antisense oligonucleotides. The activity of the antisense-circRNA is surprisingly robust towards point mutations in the target sequence. This strategy opens up novel applications for designer circRNAs and promising therapeutic strategies in molecular medicine.
Collapse
Affiliation(s)
- Christina Pfafenrot
- Institute of Biochemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Tim Schneider
- Institute of Biochemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Christin Müller
- Institute of Medical Virology, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Lee-Hsueh Hung
- Institute of Biochemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Silke Schreiner
- Institute of Biochemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - John Ziebuhr
- Institute of Medical Virology, Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Albrecht Bindereif
- Institute of Biochemistry, Justus Liebig University of Giessen, 35392 Giessen, Germany
| |
Collapse
|
35
|
Rubio-Sánchez R, Fabrini G, Cicuta P, Di Michele L. Amphiphilic DNA nanostructures for bottom-up synthetic biology. Chem Commun (Camb) 2021; 57:12725-12740. [PMID: 34750602 PMCID: PMC8631003 DOI: 10.1039/d1cc04311k] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/28/2021] [Indexed: 12/28/2022]
Abstract
DNA nanotechnology enables the construction of sophisticated biomimetic nanomachines that are increasingly central to the growing efforts of creating complex cell-like entities from the bottom-up. DNA nanostructures have been proposed as both structural and functional elements of these artificial cells, and in many instances are decorated with hydrophobic moieties to enable interfacing with synthetic lipid bilayers or regulating bulk self-organisation. In this feature article we review recent efforts to design biomimetic membrane-anchored DNA nanostructures capable of imparting complex functionalities to cell-like objects, such as regulated adhesion, tissue formation, communication and transport. We then discuss the ability of hydrophobic modifications to enable the self-assembly of DNA-based nanostructured frameworks with prescribed morphology and functionality, and explore the relevance of these novel materials for artificial cell science and beyond. Finally, we comment on the yet mostly unexpressed potential of amphiphilic DNA-nanotechnology as a complete toolbox for bottom-up synthetic biology - a figurative and literal scaffold upon which the next generation of synthetic cells could be built.
Collapse
Affiliation(s)
- Roger Rubio-Sánchez
- Biological and Soft Systems, Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK.
- fabriCELL, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Giacomo Fabrini
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
- fabriCELL, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Pietro Cicuta
- Biological and Soft Systems, Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK.
| | - Lorenzo Di Michele
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
- Biological and Soft Systems, Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK.
- fabriCELL, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| |
Collapse
|
36
|
Involvement of adaptor proteins in clathrin-mediated endocytosis of virus entry. Microb Pathog 2021; 161:105278. [PMID: 34740810 DOI: 10.1016/j.micpath.2021.105278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
The first step in the initiation of effective viral infection is breaking through the cytomembrane to enter the cell. Clathrin-mediated endocytosis is a key vesicular trafficking process in which a variety of cargo molecules are transported from the outside to the inside of the cell. This process is hijacked by numerous families of enveloped or non-enveloped viruses, which use it to enter host cells, followed by trafficking to their replicating sites. Various adaptor proteins that assist in cargo selection, coat assembly, and clathrin-coated bud maturation are important in this process. Research data documented on the involvement of adaptor proteins, such as AP-2, Eps-15, Epsin1, and AP180/CALM, in the invasion of viruses via the clathrin-mediated endocytosis have provided novel insights into understanding the viral life cycle and have led to the development of novel therapeutics. Here, we summarize the latest discoveries on the role of these adaptor proteins in clathrin-mediated endocytosis of virus entry and also discuss the future trends in this field.
Collapse
|
37
|
Xu X, Zhang QY, Chu XY, Quan Y, Lv BM, Zhang HY. Facilitating Antiviral Drug Discovery Using Genetic and Evolutionary Knowledge. Viruses 2021; 13:v13112117. [PMID: 34834924 PMCID: PMC8626054 DOI: 10.3390/v13112117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022] Open
Abstract
Over the course of human history, billions of people worldwide have been infected by various viruses. Despite rapid progress in the development of biomedical techniques, it is still a significant challenge to find promising new antiviral targets and drugs. In the past, antiviral drugs mainly targeted viral proteins when they were used as part of treatment strategies. Since the virus mutation rate is much faster than that of the host, such drugs feature drug resistance and narrow-spectrum antiviral problems. Therefore, the targeting of host molecules has gradually become an important area of research for the development of antiviral drugs. In recent years, rapid advances in high-throughput sequencing techniques have enabled numerous genetic studies (such as genome-wide association studies (GWAS), clustered regularly interspersed short palindromic repeats (CRISPR) screening, etc.) for human diseases, providing valuable genetic and evolutionary resources. Furthermore, it has been revealed that successful drug targets exhibit similar genetic and evolutionary features, which are of great value in identifying promising drug targets and discovering new drugs. Considering these developments, in this article the authors propose a host-targeted antiviral drug discovery strategy based on knowledge of genetics and evolution. We first comprehensively summarized the genetic, subcellular location, and evolutionary features of the human genes that have been successfully used as antiviral targets. Next, the summarized features were used to screen novel druggable antiviral targets and to find potential antiviral drugs, in an attempt to promote the discovery of new antiviral drugs.
Collapse
Affiliation(s)
| | - Qing-Ye Zhang
- Correspondence: (Q.-Y.Z.); (H.-Y.Z.); Tel.: +86-27-8728-0877 (H.-Y.Z.)
| | | | | | | | - Hong-Yu Zhang
- Correspondence: (Q.-Y.Z.); (H.-Y.Z.); Tel.: +86-27-8728-0877 (H.-Y.Z.)
| |
Collapse
|
38
|
Olsén E, Jõemetsa S, González A, Joyce P, Zhdanov VP, Midtvedt D, Höök F. Diffusion of Lipid Nanovesicles Bound to a Lipid Membrane Is Associated with the Partial-Slip Boundary Condition. NANO LETTERS 2021; 21:8503-8509. [PMID: 34403260 PMCID: PMC8517973 DOI: 10.1021/acs.nanolett.1c02092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
During diffusion of nanoparticles bound to a cellular membrane by ligand-receptor pairs, the distance to the laterally mobile interface is sufficiently short for their motion to depend not only on the membrane-mediated diffusivity of the tethers but also in a not yet fully understood manner on nanoparticle size and interfacial hydrodynamics. By quantifying diffusivity, velocity, and size of individual membrane-bound liposomes subjected to a hydrodynamic shear flow, we have successfully separated the diffusivity contributions from particle size and number of tethers. The obtained diffusion-size relations for synthetic and extracellular lipid vesicles are not well-described by the conventional no-slip boundary condition, suggesting partial slip as well as a significant diffusivity dependence on the distance to the lipid bilayer. These insights, extending the understanding of diffusion of biological nanoparticles at lipid bilayers, are of relevance for processes such as cellular uptake of viruses and lipid nanoparticles or labeling of cell-membrane-residing molecules.
Collapse
Affiliation(s)
- Erik Olsén
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Silver Jõemetsa
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Adrián González
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Paul Joyce
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
- UniSA:
Clinical and Health Sciences, University
of South Australia, 5000 Adelaide, Australia
| | - Vladimir P. Zhdanov
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
- Boreskov
Institute of Catalysis, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Daniel Midtvedt
- Department
of Physics, University of Gothenburg, SE-41296 Göteborg, Sweden
| | - Fredrik Höök
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| |
Collapse
|
39
|
Cellular Organelles Involved in Hepatitis E Virus Infection. Pathogens 2021; 10:pathogens10091206. [PMID: 34578238 PMCID: PMC8469867 DOI: 10.3390/pathogens10091206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatitis E virus (HEV), a major cause of acute hepatitis worldwide, infects approximately 20 million individuals annually. HEV can infect a wide range of mammalian and avian species, and cause frequent zoonotic spillover, increasingly raising public health concerns. To establish a successful infection, HEV needs to usurp host machineries to accomplish its life cycle from initial attachment to egress. However, relatively little is known about the HEV life cycle, especially the functional role(s) of cellular organelles and their associated proteins at different stages of HEV infection. Here, we summarize current knowledge regarding the relation of HEV with the different cell organelles during HEV infection. Furthermore, we discuss the underlying mechanisms by which HEV infection is precisely regulated in infected cells and the modification of host cell organelles and their associated proteins upon HEV infection.
Collapse
|
40
|
Shafat Z, Hamza A, Islam A, Al-Dosari MS, Parvez MK, Parveen S. Structural exploration of Y-domain reveals its essentiality in HEV pathogenesis. Protein Expr Purif 2021; 187:105947. [PMID: 34314826 DOI: 10.1016/j.pep.2021.105947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/08/2021] [Accepted: 07/21/2021] [Indexed: 12/19/2022]
Abstract
Hepatitis E virus (HEV) is a major causative agent of hepatitis E infections across the globe. Although the essentiality of HEV nonstructural polyprotein (pORF1) putative Y-domain (Yd) has been established in viral pathogenesis, its structural-functional role remains elusive. The current research discusses the novel exploration on Yd protein expression, purification, biophysical characterization and structure-based docking analysis. The codon optimized synthetic gene and optimized expression parameters i.e., 5 h induction with 0.25 mM IPTG at 37 °C, resulted in efficient production of Yd protein (∼40 kDa) in E. coli BL21(DE3) cells. Majority of the recombinant Yd (rYd) protein expressed as inclusion bodies was solubilized in 0.5% N-lauroylsarcosine and purified using Ni-NTA chromatography. Circular dichroism (CD) and UV visible absorption spectroscopic studies on Yd revealed both secondary and tertiary structure stability in alkaline range (pH 8.0-10.0), suggesting correlation with its physiological activity. Thus, loss in structure at low pH perhaps play crucial role in cytoplasmic-membrane interaction. The biophysical data were in good agreement with in-silico structural analyses, which suggested mixed α/β fold, non-random and basic nature of Yd protein. Furthermore, due to Yd protein essentiality in HEV replication and pathogenesis, it was considered as a template for docking and drug-likeness analyses. The 3D modeling of Yd protein and structure-based screening and drug-likeness of inhibitory compounds, including established antiviral drugs led to the identification of top nine promising candidates. Nonetheless, in vitro studies on the predicted interaction of Yd with intracellular-membrane towards establishing replication-complexes as well as validations of the proposed therapeutic agents are warranted.
Collapse
Affiliation(s)
- Zoya Shafat
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Abu Hamza
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Mohammed S Al-Dosari
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad K Parvez
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Shama Parveen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
41
|
SARS-CoV-2 Cellular Entry Is Independent of the ACE2 Cytoplasmic Domain Signaling. Cells 2021; 10:cells10071814. [PMID: 34359983 PMCID: PMC8304749 DOI: 10.3390/cells10071814] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/10/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022] Open
Abstract
Recently emerged severe acute respiratory syndrome coronavirus (SARS-CoV)-1 and -2 initiate virus infection by binding of their spike glycoprotein with the cell-surface receptor angiotensin-converting enzyme 2 (ACE2) and enter into the host cells mainly via the clathrin-mediated endocytosis pathway. However, the internalization process post attachment with the receptor is not clear for both SARS-CoV-1 and -2. Understanding the cellular factor/s or pathways used by these CoVs for internalization might provide insights into viral pathogenesis, transmission, and development of novel therapeutics. Here, we demonstrated that the cytoplasmic tail of ACE2 is not essential for the entry of SARS-CoV-1 and -2 by using bioinformatics, mutational, confocal imaging, and pseudotyped SARS-CoVs infection studies. ACE2 cytoplasmic domain (cytACE2) contains a conserved internalization motif and eight putative phosphorylation sites. Complete cytoplasmic domain deleted ACE2 (∆cytACE2) was properly synthesized and presented on the surface of HEK293T and BHK21 cells like wtACE2. The SARS-CoVs S1 or RBD of spike protein binds and colocalizes with the receptors followed by internalization into the host cells. Moreover, pseudotyped SARS-CoVs entered into wtACE2- and ∆cytACE2-transfected cells but not into dipeptidyl peptidase 4 (DPP4)-expressing cells. Their entry was significantly inhibited by treatment with dynasore, a dynamin inhibitor, and NH4Cl, an endosomal acidification inhibitor. Furthermore, SARS-CoV antibodies and the soluble form of ACE2-treated pseudotyped SARS-CoVs were unable to enter the wtACE2 and ∆cytACE2-expressing cells. Altogether, our data show that ACE2 cytoplasmic domain signaling is not essential for the entry of SARS-CoV-1 and -2 and that SARS-CoVs entry might be mediated via known/unknown host factor/s.
Collapse
|
42
|
Prabhakara C, Godbole R, Sil P, Jahnavi S, Gulzar SEJ, van Zanten TS, Sheth D, Subhash N, Chandra A, Shivaraj A, Panikulam P, U I, Nuthakki VK, Puthiyapurayil TP, Ahmed R, Najar AH, Lingamallu SM, Das S, Mahajan B, Vemula P, Bharate SB, Singh PP, Vishwakarma R, Guha A, Sundaramurthy V, Mayor S. Strategies to target SARS-CoV-2 entry and infection using dual mechanisms of inhibition by acidification inhibitors. PLoS Pathog 2021; 17:e1009706. [PMID: 34252168 PMCID: PMC8297935 DOI: 10.1371/journal.ppat.1009706] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/22/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
Many viruses utilize the host endo-lysosomal network for infection. Tracing the endocytic itinerary of SARS-CoV-2 can provide insights into viral trafficking and aid in designing new therapeutic strategies. Here, we demonstrate that the receptor binding domain (RBD) of SARS-CoV-2 spike protein is internalized via the pH-dependent CLIC/GEEC (CG) endocytic pathway in human gastric-adenocarcinoma (AGS) cells expressing undetectable levels of ACE2. Ectopic expression of ACE2 (AGS-ACE2) results in RBD traffic via both CG and clathrin-mediated endocytosis. Endosomal acidification inhibitors like BafilomycinA1 and NH4Cl, which inhibit the CG pathway, reduce the uptake of RBD and impede Spike-pseudoviral infection in both AGS and AGS-ACE2 cells. The inhibition by BafilomycinA1 was found to be distinct from Chloroquine which neither affects RBD uptake nor alters endosomal pH, yet attenuates Spike-pseudovirus entry. By screening a subset of FDA-approved inhibitors for functionality similar to BafilomycinA1, we identified Niclosamide as a SARS-CoV-2 entry inhibitor. Further validation using a clinical isolate of SARS-CoV-2 in AGS-ACE2 and Vero cells confirmed its antiviral effect. We propose that Niclosamide, and other drugs which neutralize endosomal pH as well as inhibit the endocytic uptake, could provide broader applicability in subverting infection of viruses entering host cells via a pH-dependent endocytic pathway.
Collapse
Affiliation(s)
| | - Rashmi Godbole
- National Centre for Biological Sciences (TIFR), Bengaluru, India
- University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, India
| | - Parijat Sil
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Sowmya Jahnavi
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Shah-e-Jahan Gulzar
- National Centre for Biological Sciences (TIFR), Bengaluru, India
- SASTRA University, Thanjavur, India
| | | | - Dhruv Sheth
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Neeraja Subhash
- National Centre for Biological Sciences (TIFR), Bengaluru, India
- SASTRA University, Thanjavur, India
| | - Anchal Chandra
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | | | | | - Ibrahim U
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | | | | | - Riyaz Ahmed
- CSIR—Indian Institute of Integrative Medicine, Jammu, India
| | | | - Sai Manoz Lingamallu
- Institute for Stem Cell Science and Regenerative Medicine (inSTEM), Bengaluru, India
- Manipal Academy of Higher Education (MAHE), Madhav Nagar, Manipal, Karnataka, India
| | - Snigdhadev Das
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | | | - Praveen Vemula
- Institute for Stem Cell Science and Regenerative Medicine (inSTEM), Bengaluru, India
| | | | | | | | - Arjun Guha
- Institute for Stem Cell Science and Regenerative Medicine (inSTEM), Bengaluru, India
| | | | - Satyajit Mayor
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| |
Collapse
|
43
|
Chan Y, Ng SW, Singh SK, Gulati M, Gupta G, Chaudhary SK, Hing GB, Collet T, MacLoughlin R, Löbenberg R, Oliver BG, Chellappan DK, Dua K. Revolutionizing polymer-based nanoparticle-linked vaccines for targeting respiratory viruses: A perspective. Life Sci 2021; 280:119744. [PMID: 34174324 PMCID: PMC8223024 DOI: 10.1016/j.lfs.2021.119744] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022]
Abstract
Viral respiratory tract infections have significantly impacted global health as well as socio-economic growth. Respiratory viruses such as the influenza virus, respiratory syncytial virus (RSV), and the recent SARS-CoV-2 infection (COVID-19) typically infect the upper respiratory tract by entry through the respiratory mucosa before reaching the lower respiratory tract, resulting in respiratory disease. Generally, vaccination is the primary method in preventing virus pathogenicity and it has been shown to remarkably reduce the burden of various infectious diseases. Nevertheless, the efficacy of conventional vaccines may be hindered by certain limitations, prompting the need to develop novel vaccine delivery vehicles to immunize against various strains of respiratory viruses and to mitigate the risk of a pandemic. In this review, we provide an insight into how polymer-based nanoparticles can be integrated with the development of vaccines to effectively enhance immune responses for combating viral respiratory tract infections.
Collapse
Affiliation(s)
- Yinghan Chan
- School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sin Wi Ng
- School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, India
| | - Sushil Kumar Chaudhary
- Faculty of Pharmacy, DIT University, Mussoorie-Diversion Road, Makkawala, Dehradun 248 009, Uttarakhand, India
| | - Goh Bey Hing
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Trudi Collet
- Innovative Medicines Group, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Dangan, H91 HE94 Galway, Ireland; School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Raimar Löbenberg
- University of Alberta, Faculty of Pharmacy and Pharmaceutical Sciences, Edmonton, AB T6G 2N8, Canada
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| | - Kamal Dua
- University of Alberta, Faculty of Pharmacy and Pharmaceutical Sciences, Edmonton, AB T6G 2N8, Canada; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia.
| |
Collapse
|
44
|
Shahryari A, Burtscher I, Nazari Z, Lickert H. Engineering Gene Therapy: Advances and Barriers. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alireza Shahryari
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- School of Medicine Department of Human Genetics Technical University of Munich Klinikum Rechts der Isar 81675 München Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
- Stem Cell Research Center Golestan University of Medical Sciences Gorgan 49341‐74515 Iran
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
| | - Zahra Nazari
- Department of Biology School of Basic Sciences Golestan University Gorgan 49361‐79142 Iran
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- School of Medicine Department of Human Genetics Technical University of Munich Klinikum Rechts der Isar 81675 München Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
| |
Collapse
|
45
|
Orthobunyaviruses: From Virus Binding to Penetration into Mammalian Host Cells. Viruses 2021; 13:v13050872. [PMID: 34068494 PMCID: PMC8151349 DOI: 10.3390/v13050872] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 12/04/2022] Open
Abstract
With over 80 members worldwide, Orthobunyavirus is the largest genus in the Peribunyaviridae family. Orthobunyaviruses (OBVs) are arthropod-borne viruses that are structurally simple, with a trisegmented, negative-sense RNA genome and only four structural proteins. OBVs are potential agents of emerging and re-emerging diseases and overall represent a global threat to both public and veterinary health. The focus of this review is on the very first steps of OBV infection in mammalian hosts, from virus binding to penetration and release of the viral genome into the cytosol. Here, we address the most current knowledge and advances regarding OBV receptors, endocytosis, and fusion.
Collapse
|
46
|
Devnath P, Masud H. Nipah virus: a potential pandemic agent in the context of the current severe acute respiratory syndrome coronavirus 2 pandemic. New Microbes New Infect 2021; 41:100873. [PMID: 33758670 PMCID: PMC7972828 DOI: 10.1016/j.nmni.2021.100873] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 02/23/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
For centuries, zoonotic diseases have been responsible for various outbreaks resulting in the deaths of millions of people. The best example of this is the current coronavirus disease 2019 (COVID-19) pandemic. Like severe acute respiratory syndrome coronavirus, Nipah virus is another deadly virus which has caused several outbreaks in the last few years. Though it causes a low number of infections, disease severity results in a higher death rate. In the context of the recent COVID-19 pandemic, we speculate that many countries will be unable to deal with the sudden onset of such a viral outbreak. Thus, further research and attention to the virus are needed to address future outbreaks.
Collapse
Affiliation(s)
- P. Devnath
- Department of Microbiology, Faculty of Sciences, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - H.M.A.A. Masud
- Department of Microbiology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| |
Collapse
|
47
|
Souza E Souza KFC, Moraes BPT, Paixão ICNDP, Burth P, Silva AR, Gonçalves-de-Albuquerque CF. Na +/K +-ATPase as a Target of Cardiac Glycosides for the Treatment of SARS-CoV-2 Infection. Front Pharmacol 2021; 12:624704. [PMID: 33935717 PMCID: PMC8085498 DOI: 10.3389/fphar.2021.624704] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), identified for the first time in Wuhan, China, causes coronavirus disease 2019 (COVID-19), which moved from epidemic status to becoming a pandemic. Since its discovery in December 2019, there have been countless cases of mortality and morbidity due to this virus. Several compounds such as chloroquine, hydroxychloroquine, lopinavir-ritonavir, and remdesivir have been tested as potential therapies; however, no effective treatment is currently recommended by regulatory agencies. Some studies on respiratory non-enveloped viruses such as adenoviruses and rhinovirus and some respiratory enveloped viruses including human respiratory syncytial viruses, influenza A, parainfluenza, SARS-CoV, and SARS-CoV-2 have shown the antiviral activity of cardiac glycosides, correlating their effect with Na+/K+-ATPase (NKA) modulation. Cardiac glycosides are secondary metabolites used to treat patients with cardiac insufficiency because they are the most potent inotropic agents. The effects of cardiac glycosides on NKA are dependent on cell type, exposure time, and drug concentration. They may also cause blockage of Na+ and K+ ionic transport or trigger signaling pathways. The antiviral activity of cardiac glycosides is related to cell signaling activation through NKA inhibition. Nuclear factor kappa B (NFκB) seems to be an essential transcription factor for SARS-CoV-2 infection. NFκB inhibition by cardiac glycosides interferes directly with SARS-CoV-2 yield and inflammatory cytokine production. Interestingly, the antiviral effect of cardiac glycosides is associated with tyrosine kinase (Src) activation, and NFκB appears to be regulated by Src. Src is one of the main signaling targets of the NKA α-subunit, modulating other signaling factors that may also impair viral infection. These data suggest that Src-NFκB signaling modulated by NKA plays a crucial role in the inhibition of SARS-CoV-2 infection. Herein, we discuss the antiviral effects of cardiac glycosides on different respiratory viruses, SARS-CoV-2 pathology, cell signaling pathways, and NKA as a possible molecular target for the treatment of COVID-19.
Collapse
Affiliation(s)
- Kauê Francisco Corrêa Souza E Souza
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Laboratório de Imunofarmacologia, Departamento de Bioquímica, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bianca Portugal Tavares Moraes
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Programa de Pós-Graduação Em Neurociências (PPGNEURO), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Izabel Christina Nunes de Palmer Paixão
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Ciências e Biotecnologia (PPBI), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Neûrologia/Neurociências, Hospital Antônio Pedro Universidade Federal Fluminense, Niterói, Brazil
| | - Patrícia Burth
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Ciências e Biotecnologia (PPBI), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Programa de Pós-Graduação Em Neurociências (PPGNEURO), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Departamento de Bioquímica, Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Programa de Pós-Graduação Em Neurociências (PPGNEURO), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Ciências e Biotecnologia (PPBI), Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.,Programa de Pós-Graduação Em Biologia Celular e Molecular (PPGBMC), Universidade Federal Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
48
|
Pérez-Pulido AJ, Asencio-Cortés G, Brokate-Llanos AM, Brea-Calvo G, Rodríguez-Griñolo R, Garzón A, Muñoz MJ. Serial co-expression analysis of host factors from SARS-CoV viruses highly converges with former high-throughput screenings and proposes key regulators. Brief Bioinform 2021; 22:1038-1052. [PMID: 33458747 PMCID: PMC7929451 DOI: 10.1093/bib/bbaa419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/11/2020] [Accepted: 12/19/2020] [Indexed: 11/14/2022] Open
Abstract
The current genomics era is bringing an unprecedented growth in the amount of gene expression data, only comparable to the exponential growth of sequences in databases during the last decades. This data allow the design of secondary analyses that take advantage of this information to create new knowledge. One of these feasible analyses is the evaluation of the expression level for a gene through a series of different conditions or cell types. Based on this idea, we have developed Automatic and Serial Analysis of CO-expression, which performs expression profiles for a given gene along hundreds of heterogeneous and normalized transcriptomics experiments and discover other genes that show either a similar or an inverse behavior. It might help to discover co-regulated genes, and common transcriptional regulators in any biological model. The present severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic is an opportunity to test this novel approach due to the wealth of data that are being generated, which could be used for validating results. Thus, we have identified 35 host factors in the literature putatively involved in the infectious cycle of SARS-CoV viruses and searched for genes tightly co-expressed with them. We have found 1899 co-expressed genes whose assigned functions are strongly related to viral cycles. Moreover, this set of genes heavily overlaps with those identified by former laboratory high-throughput screenings (with P-value near 0). Our results reveal a series of common regulators, involved in immune and inflammatory responses that might be key virus targets to induce the coordinated expression of SARS-CoV-2 host factors.
Collapse
Affiliation(s)
- Antonio J Pérez-Pulido
- Centro Andaluz de Biologia del Desarrollo (CABD, UPO-CSIC-JA). Facultad de Ciencias Experimentales (Área de Genética), Universidad Pablo de Olavide, 41013, Sevilla, Spain
| | | | - Ana M Brokate-Llanos
- Centro Andaluz de Biologia del Desarrollo (CABD, UPO-CSIC-JA). Facultad de Ciencias Experimentales (Área de Genética), Universidad Pablo de Olavide, 41013, Sevilla, Spain
| | - Gloria Brea-Calvo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, 28000, Madrid, Spain
| | - Rosario Rodríguez-Griñolo
- Dpto. de Economía, Métodos Cuantitativos e Historia Económica. Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Andrés Garzón
- Centro Andaluz de Biologia del Desarrollo (CABD, UPO-CSIC-JA). Facultad de Ciencias Experimentales (Área de Genética), Universidad Pablo de Olavide, 41013, Sevilla, Spain
| | - Manuel J Muñoz
- Centro Andaluz de Biologia del Desarrollo (CABD, UPO-CSIC-JA). Facultad de Ciencias Experimentales (Área de Genética), Universidad Pablo de Olavide, 41013, Sevilla, Spain
| |
Collapse
|
49
|
Kryvenko V, Vadász I. Molecular mechanisms of Na,K-ATPase dysregulation driving alveolar epithelial barrier failure in severe COVID-19. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1186-L1193. [PMID: 33689516 PMCID: PMC8238442 DOI: 10.1152/ajplung.00056.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A significant number of patients with coronavirus disease 2019 (COVID-19) develop acute respiratory distress syndrome (ARDS) that is associated with a poor outcome. The molecular mechanisms driving failure of the alveolar barrier upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection remain incompletely understood. The Na,K-ATPase is an adhesion molecule and a plasma membrane transporter that is critically required for proper alveolar epithelial function by both promoting barrier integrity and resolution of excess alveolar fluid, thus enabling appropriate gas exchange. However, numerous SARS-CoV-2-mediated and COVID-19-related signals directly or indirectly impair the function of the Na,K-ATPase, thereby potentially contributing to disease progression. In this Perspective, we highlight some of the putative mechanisms of SARS-CoV-2-driven dysfunction of the Na,K-ATPase, focusing on expression, maturation, and trafficking of the transporter. A therapeutic mean to selectively inhibit the maladaptive signals that impair the Na,K-ATPase upon SARS-CoV-2 infection might be effective in reestablishing the alveolar epithelial barrier and promoting alveolar fluid clearance and thus advantageous in patients with COVID-19-associated ARDS.
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,The Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,The Cardio-Pulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
50
|
Hulswit RJG, Paesen GC, Bowden TA, Shi X. Recent Advances in Bunyavirus Glycoprotein Research: Precursor Processing, Receptor Binding and Structure. Viruses 2021; 13:353. [PMID: 33672327 PMCID: PMC7926653 DOI: 10.3390/v13020353] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 01/04/2023] Open
Abstract
The Bunyavirales order accommodates related viruses (bunyaviruses) with segmented, linear, single-stranded, negative- or ambi-sense RNA genomes. Their glycoproteins form capsomeric projections or spikes on the virion surface and play a crucial role in virus entry, assembly, morphogenesis. Bunyavirus glycoproteins are encoded by a single RNA segment as a polyprotein precursor that is co- and post-translationally cleaved by host cell enzymes to yield two mature glycoproteins, Gn and Gc (or GP1 and GP2 in arenaviruses). These glycoproteins undergo extensive N-linked glycosylation and despite their cleavage, remain associated to the virion to form an integral transmembrane glycoprotein complex. This review summarizes recent advances in our understanding of the molecular biology of bunyavirus glycoproteins, including their processing, structure, and known interactions with host factors that facilitate cell entry.
Collapse
Affiliation(s)
- Ruben J. G. Hulswit
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Guido C. Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Xiaohong Shi
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|