1
|
de Souza Cardoso R, Ono A. The Effects of Viral Structural Proteins on Acidic Phospholipids in Host Membranes. Viruses 2024; 16:1714. [PMID: 39599829 PMCID: PMC11599007 DOI: 10.3390/v16111714] [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: 09/13/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Enveloped viruses rely on host membranes for trafficking and assembly. A substantial body of literature published over the years supports the involvement of cellular membrane lipids in the enveloped virus assembly processes. In particular, the knowledge regarding the relationship between viral structural proteins and acidic phospholipids has been steadily increasing in recent years. In this review, we will briefly review the cellular functions of plasma membrane-associated acidic phospholipids and the mechanisms that regulate their local distribution within this membrane. We will then explore the interplay between viruses and the plasma membrane acidic phospholipids in the context of the assembly process for two enveloped viruses, the influenza A virus (IAV) and the human immunodeficiency virus type 1 (HIV-1). Among the proteins encoded by these viruses, three viral structural proteins, IAV hemagglutinin (HA), IAV matrix protein-1 (M1), and HIV-1 Gag protein, are known to interact with acidic phospholipids, phosphatidylserine and/or phosphatidylinositol (4,5)-bisphosphate. These interactions regulate the localization of the viral proteins to and/or within the plasma membrane and likely facilitate the clustering of the proteins. On the other hand, these viral proteins, via their ability to multimerize, can also alter the distribution of the lipids and may induce acidic-lipid-enriched membrane domains. We will discuss the potential significance of these interactions in the virus assembly process and the property of the progeny virions. Finally, we will outline key outstanding questions that need to be answered for a better understanding of the relationships between enveloped virus assembly and acidic phospholipids.
Collapse
Affiliation(s)
| | - Akira Ono
- Department of Microbiology and Immunology, The University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
2
|
Chaudhury A, Swarnakar S, Pattnaik GP, Varshney GK, Chakraborty H, Basu JK. Peptide-Induced Fusion of Dynamic Membrane Nanodomains: Implications in a Viral Entry. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:17713-17722. [PMID: 38031897 DOI: 10.1021/acs.langmuir.3c02230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Enveloped viruses infect host cells via protein-mediated membrane fusion. However, insights into the microscopic rearrangement induced by the viral proteins and peptides have not yet emerged. Here, we report a new methodology to extract viral fusion peptide (FP)-mediated biomembrane dynamical nanodomain fusion parameter, λ, based on stimulated emission depletion microscopy coupled with fluorescence correlation spectroscopy. We also define another dynamical parameter membrane gradient, defined in terms of the ratio of average lipid diffusion coefficients across dynamic crossover length scales, ξ. Significantly, we observe that λ as well as these mobility gradients are larger in the stiffer liquid-ordered (Lo) phase compared to the liquid-disordered phase and are more effective at the smaller nanodomain interfaces, which are only present in the Lo phase. The results could possibly help to resolve a long-standing puzzle about the enhanced fusogenicity of FP in the Lo phase. Results obtained from the diffusion results have been correlated with the human immunodeficiency virus gp41 FP-induced membrane fusion.
Collapse
Affiliation(s)
- Anurag Chaudhury
- Department of Physics, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shovon Swarnakar
- Department of Physics, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | | | - Gopal K Varshney
- Department of Physics, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Hirak Chakraborty
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Jaydeep Kumar Basu
- Department of Physics, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| |
Collapse
|
3
|
Wang Y, Zhang Y, Zuo W, Bo Z, Zhang C, Zhang X, Wu Y. Avian Reovirus σB Interacts with Caveolin-1 in Lipid Rafts during Dynamin-Dependent Caveolae-Mediated Endocytosis. Viruses 2022; 14:v14102201. [PMID: 36298756 PMCID: PMC9608613 DOI: 10.3390/v14102201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022] Open
Abstract
Caveolin-1 (Cav-1) is the basic component of caveolae, a specialized form of lipid raft that plays an essential role in endocytic viral entry. However, the evidence of direct involvement of caveolae and Cav-1 in avian reovirus (ARV) entry remains insufficient. In this study, the membrane lipid rafts were isolated as detergent-resistant microdomains (DRMs) by sucrose gradient centrifugation, and the capsid protein σB of ARV was found to associate with Cav-1 in DRMs fractions. Additionally, the interaction between ARV σB protein and Cav-1 was demonstrated by immunofluorescence co-localization and co-immunoprecipitation assays. Furthermore, we found that the internalization of ARV is sensitive to caveolae and dynamin inhibitors, while it is insensitive to clathrin inhibitors. In conclusion, these results indicate that the ARV σB protein interacts with Cav-1 during dynamin-dependent caveolae-mediated endocytosis for the entry of ARV.
Collapse
Affiliation(s)
- Yuyang Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Testing Center, Yangzhou University, Yangzhou 225009, China
| | - Yangyang Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Wei Zuo
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zongyi Bo
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Chengcheng Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiaorong Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yantao Wu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
4
|
Tran N, Oh Y, Sutherland M, Cui Q, Hong M. Cholesterol-Mediated Clustering of the HIV Fusion Protein gp41 in Lipid Bilayers. J Mol Biol 2021; 434:167345. [PMID: 34762895 DOI: 10.1016/j.jmb.2021.167345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/26/2021] [Accepted: 10/31/2021] [Indexed: 11/16/2022]
Abstract
The envelope glycoprotein (Env) of the human immunodeficient virus (HIV-1) is known to cluster on the viral membrane surface to attach to target cells and cause membrane fusion for HIV-1 infection. However, the molecular structural mechanisms that drive Env clustering remain opaque. Here, we use solid-state NMR spectroscopy and molecular dynamics (MD) simulations to investigate nanometer-scale clustering of the membrane-proximal external region (MPER) and transmembrane domain (TMD) of gp41, the fusion protein component of Env. Using 19F solid-state NMR experiments of mixed fluorinated peptides, we show that MPER-TMD trimers form clusters with interdigitated MPER helices in cholesterol-containing membranes. Inter-trimer 19F-19F cross peaks, which are indicative of spatial contacts within ∼2 nm, are observed in cholesterol-rich virus-mimetic membranes but are suppressed in cholesterol-free model membranes. Water-peptide and lipid-peptide cross peaks in 2D 1H-19F correlation spectra indicate that the MPER is well embedded in model phosphocholine membranes but is more exposed to the surface of the virus-mimetic membrane. These experimental results are reproduced in coarse-grained and atomistic molecular dynamics simulations, which suggest that the effects of cholesterol on gp41 clustering is likely via indirect modulation of the MPER orientation. Cholesterol binding to the helix-turn-helix region of the MPER-TMD causes a parallel orientation of the MPER with the membrane surface, thus allowing MPERs of neighboring trimers to interact with each other to cause clustering. These solid-state NMR data and molecular dynamics simulations suggest that MPER and cholesterol cooperatively govern the clustering of gp41 trimers during virus-cell membrane fusion.
Collapse
Affiliation(s)
- Nhi Tran
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139, United States
| | - Younghoon Oh
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, United States
| | - Madeleine Sutherland
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139, United States
| | - Qiang Cui
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, United States; Department of Physics, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, United States; Department of Biomedical Engineering, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, United States.
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA 02139, United States. https://twitter.com/MeiHongLab
| |
Collapse
|
5
|
Structural Domains of the Herpes Simplex Type 1 gD Protein that Restrict HIV-1 Particle Infectivity. J Virol 2021; 95:JVI.02355-20. [PMID: 33536165 PMCID: PMC8103709 DOI: 10.1128/jvi.02355-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previously, we showed that the presence of the herpes simplex virus type 1 (HSV-1) gD glycoprotein but not gB potently restricted HIV-1 particle infectivity. This restriction was characterized by incorporation of HSV-1 gD and the exclusion of the HIV-1 gp120/gp41 from budding virus particles. To determine the structural domains involved in gD restriction of HIV-1, a series of deletion mutants and chimeric proteins between gD and the non-restrictive gB were generated. Our results show that deletion of the cytoplasmic tail domain (CTD) of gD or that replacement of the transmembrane domain (TMD) with the TMD from gB slightly reduced restriction activity. However, replacement of the gD CTD with that of gB resulted in lower cell surface expression, significantly less incorporation into HIV-1 particles, and inefficient restriction of the release of infectious HIV-1. Analysis of gB/gD chimeric proteins revealed that removal of the gB CTD or replacement with gD CTD resulted in enhanced surface expression and an increase in restriction activity. Finally, we show that expression of gD without other HSV-1 proteins resulted in gD fractionation into detergent resistant membranes (DRM) and that gD co-localized with the raft marker GM1, which may partially explain its incorporation into budding virus particles. Taken together, our results suggest that expression of gD at the cell surface is likely a major factor but that other intrinsic properties are also involved in the gD-mediated restriction of HIV-1 particle infectivity.IMPORTANCE Previously, we showed that unlike the HSV-1, the presence of the gD glycoprotein in virus producer cells but not gB potently restricted HIV-1 particle infectivity. To better understand the relationship between cell surface expression, virus incorporation and restriction of HIV-1, we analyzed a series of deletion mutants and chimeric proteins in which domains of gD and gB were swapped. Our results indicate that: a) gD/gB chimeras having the cytoplasmic domain (CTD) of gB significantly reduced cell surface expression, release from cells, incorporation into virus, and reduced HIV-1 restriction; b) removal of the gB CTD or replacement with the gD CTD resulted in better surface expression, incorporation into HIV-1, and enhanced restriction; and c) the transmembrane domain of gB can influence transport and ultimately effect incorporation of gB into HIV-1. Overall, these data support a role for gD surface expression as crucial to restriction of infectious HIV-1 release.
Collapse
|
6
|
Wang Y, Zhang Y, Zhang C, Hu M, Yan Q, Zhao H, Zhang X, Wu Y. Cholesterol-Rich Lipid Rafts in the Cellular Membrane Play an Essential Role in Avian Reovirus Replication. Front Microbiol 2020; 11:597794. [PMID: 33224131 PMCID: PMC7667042 DOI: 10.3389/fmicb.2020.597794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/09/2020] [Indexed: 01/07/2023] Open
Abstract
Cholesterol is an essential component of lipid rafts in cellular plasma membranes. Although lipid rafts have been reported to have several functions in multiple stages of the life cycles of many different enveloped viruses, the mechanisms by which non-enveloped viruses, which lack outer lipid membranes, infect host cells remain unclear. In this study, to investigate the dependence of non-enveloped avian reovirus (ARV) infection on the integrity of cholesterol-rich membrane rafts, methyl-β-cyclodextrin (MβCD) was used to deplete cellular membrane cholesterol at the ARV attachment, entry, and post-entry stages. Treatment with MβCD significantly inhibited ARV replication at both the entry and post-entry stages in a dose-dependent manner, but MβCD had a statistically insignificant effect when it was added at the attachment stage. Moreover, MβCD treatment markedly reduced syncytium formation, which occurs at a relatively late stage of the ARV life cycle and is involved in cell-cell transmission and release. Furthermore, the addition of exogenous cholesterol reversed the effects mentioned above. Colocalization data also showed that the ARV proteins σC, μNS, and p10 prefer to localize to cholesterol-rich lipid raft regions during ARV infection. Altogether, these results suggest that cellular cholesterol in lipid rafts plays a critical role in ARV replication.
Collapse
Affiliation(s)
- Yuyang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Testing Center, Yangzhou University, Yangzhou, China
| | - Yangyang Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Chengcheng Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Maozhi Hu
- Testing Center, Yangzhou University, Yangzhou, China
| | - Qiuxiang Yan
- Testing Center, Yangzhou University, Yangzhou, China
| | - Hongyan Zhao
- Testing Center, Yangzhou University, Yangzhou, China
| | - Xiaorong Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yantao Wu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
7
|
Rendezvous at Plasma Membrane: Cellular Lipids and tRNA Set up Sites of HIV-1 Particle Assembly and Incorporation of Host Transmembrane Proteins. Viruses 2020; 12:v12080842. [PMID: 32752131 PMCID: PMC7472227 DOI: 10.3390/v12080842] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/28/2022] Open
Abstract
The HIV-1 structural polyprotein Gag drives the virus particle assembly specifically at the plasma membrane (PM). During this process, the nascent virion incorporates specific subsets of cellular lipids and host membrane proteins, in addition to viral glycoproteins and viral genomic RNA. Gag binding to the PM is regulated by cellular factors, including PM-specific phospholipid PI(4,5)P2 and tRNAs, both of which bind the highly basic region in the matrix domain of Gag. In this article, we review our current understanding of the roles played by cellular lipids and tRNAs in specific localization of HIV-1 Gag to the PM. Furthermore, we examine the effects of PM-bound Gag on the organization of the PM bilayer and discuss how the reorganization of the PM at the virus assembly site potentially contributes to the enrichment of host transmembrane proteins in the HIV-1 particle. Since some of these host transmembrane proteins alter release, attachment, or infectivity of the nascent virions, the mechanism of Gag targeting to the PM and the nature of virus assembly sites have major implications in virus spread.
Collapse
|
8
|
Garrido PF, Calvelo M, Blanco-González A, Veleiro U, Suárez F, Conde D, Cabezón A, Piñeiro Á, Garcia-Fandino R. The Lord of the NanoRings: Cyclodextrins and the battle against SARS-CoV-2. Int J Pharm 2020; 588:119689. [PMID: 32717282 PMCID: PMC7381410 DOI: 10.1016/j.ijpharm.2020.119689] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022]
Abstract
A handful of singular structures and laws can be observed in nature. They are not always evident but, once discovered, it seems obvious how to take advantage of them. In chemistry, the discovery of reproducible patterns stimulates the imagination to develop new functional materials and technological or medical applications. Two clear examples are helical structures at different levels in biological polymers as well as ring and spherical structures of different size and composition. Rings are intuitively observed as holes able to thread elongated structures. A large number of real and fictional stories have rings as inanimate protagonists. The design, development or just discovering of a special ring has often been taken as a symbol of power or success. Several examples are the Piscatory Ring wore by the Pope of the Catholic Church, the NBA Championship ring and the One Ring created by the Dark Lord Sauron in the epic story The Lord of the Rings. In this work, we reveal the power of another extremely powerful kind of rings to fight against the pandemic which is currently affecting the whole world. These rings are as small as ~1 nm of diameter and so versatile that they are able to participate in the attack of viruses, and specifically SARS-CoV-2, in a large range of different ways. This includes the encapsulation and transport of specific drugs, as adjuvants to stabilize proteins, vaccines or other molecules involved in the infection, as cholesterol trappers to destabilize the virus envelope, as carriers for RNA therapies, as direct antiviral drugs and even to rescue blood coagulation upon heparin treatment. “One ring to rule them all. One ring to find them. One ring to bring them all and in the darkness bind them.” J. R. R. Tolkien.
Collapse
Affiliation(s)
- Pablo F Garrido
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, E-15782 Santiago de Compostela, Spain
| | - Martín Calvelo
- Departamento de Química Orgánica, Center for Research in Biological Chemistry and Molecular Materials, Universidade de Santiago de Compostela, Campus Vida s/n, E-15782 Santiago de Compostela, Spain
| | - Alexandre Blanco-González
- Departamento de Química Orgánica, Center for Research in Biological Chemistry and Molecular Materials, Universidade de Santiago de Compostela, Campus Vida s/n, E-15782 Santiago de Compostela, Spain
| | - Uxía Veleiro
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, E-15782 Santiago de Compostela, Spain
| | - Fabián Suárez
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, E-15782 Santiago de Compostela, Spain
| | - Daniel Conde
- Departamento de Química Orgánica, Center for Research in Biological Chemistry and Molecular Materials, Universidade de Santiago de Compostela, Campus Vida s/n, E-15782 Santiago de Compostela, Spain
| | - Alfonso Cabezón
- Departamento de Química Orgánica, Center for Research in Biological Chemistry and Molecular Materials, Universidade de Santiago de Compostela, Campus Vida s/n, E-15782 Santiago de Compostela, Spain
| | - Ángel Piñeiro
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, E-15782 Santiago de Compostela, Spain.
| | - Rebeca Garcia-Fandino
- Departamento de Química Orgánica, Center for Research in Biological Chemistry and Molecular Materials, Universidade de Santiago de Compostela, Campus Vida s/n, E-15782 Santiago de Compostela, Spain.
| |
Collapse
|
9
|
Sengupta P, Lippincott-Schwartz J. Revisiting Membrane Microdomains and Phase Separation: A Viral Perspective. Viruses 2020; 12:v12070745. [PMID: 32664429 PMCID: PMC7412473 DOI: 10.3390/v12070745] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
Retroviruses selectively incorporate a specific subset of host cell proteins and lipids into their outer membrane when they bud out from the host plasma membrane. This specialized viral membrane composition is critical for both viral survivability and infectivity. Here, we review recent findings from live cell imaging of single virus assembly demonstrating that proteins and lipids sort into the HIV retroviral membrane by a mechanism of lipid-based phase partitioning. The findings showed that multimerizing HIV Gag at the assembly site creates a liquid-ordered lipid phase enriched in cholesterol and sphingolipids. Proteins with affinity for this specialized lipid environment partition into it, resulting in the selective incorporation of proteins into the nascent viral membrane. Building on this and other work in the field, we propose a model describing how HIV Gag induces phase separation of the viral assembly site through a mechanism involving transbilayer coupling of lipid acyl chains and membrane curvature changes. Similar phase-partitioning pathways in response to multimerizing structural proteins likely help sort proteins into the membranes of other budding structures within cells.
Collapse
|
10
|
SARS-CoV-2 Evolutionary Adaptation toward Host Entry and Recognition of Receptor O-Acetyl Sialylation in Virus-Host Interaction. Int J Mol Sci 2020; 21:ijms21124549. [PMID: 32604730 PMCID: PMC7352545 DOI: 10.3390/ijms21124549] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/15/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
The recently emerged SARS-CoV-2 is the cause of the global health crisis of the coronavirus disease 2019 (COVID-19) pandemic. No evidence is yet available for CoV infection into hosts upon zoonotic disease outbreak, although the CoV epidemy resembles influenza viruses, which use sialic acid (SA). Currently, information on SARS-CoV-2 and its receptors is limited. O-acetylated SAs interact with the lectin-like spike glycoprotein of SARS CoV-2 for the initial attachment of viruses to enter into the host cells. SARS-CoV-2 hemagglutinin-esterase (HE) acts as the classical glycan-binding lectin and receptor-degrading enzyme. Most β-CoVs recognize 9-O-acetyl-SAs but switched to recognizing the 4-O-acetyl-SA form during evolution of CoVs. Type I HE is specific for the 9-O-Ac-SAs and type II HE is specific for 4-O-Ac-SAs. The SA-binding shift proceeds through quasi-synchronous adaptations of the SA-recognition sites of the lectin and esterase domains. The molecular switching of HE acquisition of 4-O-acetyl binding from 9-O-acetyl SA binding is caused by protein–carbohydrate interaction (PCI) or lectin–carbohydrate interaction (LCI). The HE gene was transmitted to a β-CoV lineage A progenitor by horizontal gene transfer from a 9-O-Ac-SA–specific HEF, as in influenza virus C/D. HE acquisition, and expansion takes place by cross-species transmission over HE evolution. This reflects viral evolutionary adaptation to host SA-containing glycans. Therefore, CoV HE receptor switching precedes virus evolution driven by the SA-glycan diversity of the hosts. The PCI or LCI stereochemistry potentiates the SA–ligand switch by a simple conformational shift of the lectin and esterase domains. Therefore, examination of new emerging viruses can lead to better understanding of virus evolution toward transitional host tropism. A clear example of HE gene transfer is found in the BCoV HE, which prefers 7,9-di-O-Ac-SAs, which is also known to be a target of the bovine torovirus HE. A more exciting case of such a switching event occurs in the murine CoVs, with the example of the β-CoV lineage A type binding with two different subtypes of the typical 9-O-Ac-SA (type I) and the exclusive 4-O-Ac-SA (type II) attachment factors. The protein structure data for type II HE also imply the virus switching to binding 4-O acetyl SA from 9-O acetyl SA. Principles of the protein–glycan interaction and PCI stereochemistry potentiate the SA–ligand switch via simple conformational shifts of the lectin and esterase domains. Thus, our understanding of natural adaptation can be specified to how carbohydrate/glycan-recognizing proteins/molecules contribute to virus evolution toward host tropism. Under the current circumstances where reliable antiviral therapeutics or vaccination tools are lacking, several trials are underway to examine viral agents. As expected, structural and non-structural proteins of SARS-CoV-2 are currently being targeted for viral therapeutic designation and development. However, the modern global society needs SARS-CoV-2 preventive and therapeutic drugs for infected patients. In this review, the structure and sialobiology of SARS-CoV-2 are discussed in order to encourage and activate public research on glycan-specific interaction-based drug creation in the near future.
Collapse
|
11
|
Transmembrane Domain Dissociation Is Required for Hendra Virus F Protein Fusogenic Activity. J Virol 2019; 93:JVI.01069-19. [PMID: 31462574 DOI: 10.1128/jvi.01069-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022] Open
Abstract
Hendra virus (HeV) is a zoonotic paramyxovirus that utilizes a trimeric fusion (F) protein within its lipid bilayer to mediate membrane merger with a cell membrane for entry. Previous HeV F studies showed that transmembrane domain (TMD) interactions are important for stabilizing the prefusion conformation of the protein prior to triggering. Thus, the current model for HeV F fusion suggests that modulation of TMD interactions is critical for initiation and completion of conformational changes that drive membrane fusion. HeV F constructs (T483C/V484C, V484C/N485C, and N485C/P486C) were generated with double cysteine substitutions near the N-terminal region of the TMD to study the effect of altered flexibility in this region. Oligomeric analysis showed that the double cysteine substitutions successfully promoted intersubunit disulfide bond formation in HeV F. Subsequent fusion assays indicated that the introduction of disulfide bonds in the mutants prohibited fusion events. Further testing confirmed that T483C/V484C and V484C/N485C were expressed at the cell surface at levels that would allow for fusion. Attempts to restore fusion with a reducing agent were unsuccessful, suggesting that the introduced disulfide bonds were likely buried in the membrane. Conformational analysis showed that T483C/V484C and V484C/N485C were able to bind a prefusion conformation-specific antibody prior to cell disruption, indicating that the introduced disulfide bonds did not significantly affect protein folding. This study is the first to report that TMD dissociation is required for HeV F fusogenic activity and strengthens our model for HeV fusion.IMPORTANCE The paramyxovirus Hendra virus (HeV) causes severe respiratory illness and encephalitis in humans. To develop therapeutics for HeV and related viral infections, further studies are needed to understand the mechanisms underlying paramyxovirus fusion events. Knowledge gained in studies of the HeV fusion (F) protein may be applicable to a broad span of enveloped viruses. In this study, we demonstrate that disulfide bonds introduced between the HeV F transmembrane domains (TMDs) block fusion. Depending on the location of these disulfide bonds, HeV F can still fold properly and bind a prefusion conformation-specific antibody prior to cell disruption. These findings support our current model for HeV membrane fusion and expand our knowledge of the TMD and its role in HeV F stability and fusion promotion.
Collapse
|
12
|
Sabapathy T, Helmerhorst E, Bottomley S, Babaeff S, Munyard K, Newsholme P, Mamotte CD. Use of virus-like particles as a native membrane model to study the interaction of insulin with the insulin receptor. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1204-1212. [PMID: 30951702 DOI: 10.1016/j.bbamem.2019.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/15/2019] [Accepted: 03/28/2019] [Indexed: 12/15/2022]
Abstract
There is emerging evidence of the utility of virus-like particles (VLPs) as a novel model for the study of receptor-ligand interactions in a native plasma membrane environment. VLPs consist of a viral core protein encapsulated by portions of the cell membrane with membrane proteins and receptors expressed in their native conformation. VLPs can be generated in mammalian cells by transfection with the retroviral core protein (gag). In this study, we used Chinese hamster ovary (CHO T10) cells stably overexpressing the insulin receptor (IR) to generate IR bearing VLPs. The diameter and size uniformity of VLPs were estimated by dynamic light scattering and morphological features examined by scanning electron microscopy. The presence of high affinity IR on VLPs was demonstrated by competitive binding assays (KD: 2.3 ± 0.4 nM, n = 3), which was similar to that on the parental CHO T10 cells (KD: 2.1 ± 0.4 nM, n = 3). We also report that increases or decreases in membrane cholesterol content by treatment with methyl-β-cyclodextrin (MBCD) or cholesterol pre-loaded methyl-β-cyclodextrin (cMBCD), respectively, substantially decreased insulin binding (> 30%) to both VLPs and cells, and we speculate this is due to a change in receptor disposition. We suggest that this novel finding of decreases in insulin binding in response to changes in membrane cholesterol content may largely account for the unexplained decreases in insulin signalling events previously reported elsewhere. Finally, we propose VLPs as a viable membrane model for the study of insulin-IR interactions in a native membrane environment.
Collapse
Affiliation(s)
- Thiru Sabapathy
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| | - Erik Helmerhorst
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| | | | | | - Kylie Munyard
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| | - Cyril D Mamotte
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| |
Collapse
|
13
|
A lipid-based partitioning mechanism for selective incorporation of proteins into membranes of HIV particles. Nat Cell Biol 2019; 21:452-461. [PMID: 30936472 DOI: 10.1038/s41556-019-0300-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022]
Abstract
Particles that bud off from the cell surface, including viruses and microvesicles, typically have a unique membrane protein composition distinct from that of the originating plasma membrane. This selective protein composition enables viruses to evade the immune response and infect other cells. But how membrane proteins sort into budding viruses such as human immunodeficiency virus (HIV) remains unclear. Proteins could passively distribute into HIV-assembly-site membranes producing compositions resembling pre-existing plasma-membrane domains. Here, we demonstrate that proteins instead sort actively into HIV-assembly-site membranes, generating compositions enriched in cholesterol and sphingolipids that undergo continuous remodelling. Proteins are recruited into and removed from the HIV assembly site through lipid-based partitioning, initiated by oligomerization of the HIV structural protein Gag. Changes in membrane curvature at the assembly site further amplify this sorting process. Thus, a lipid-based sorting mechanism, aided by increasing membrane curvature, generates the unique membrane composition of the HIV surface.
Collapse
|
14
|
Burnie J, Guzzo C. The Incorporation of Host Proteins into the External HIV-1 Envelope. Viruses 2019; 11:v11010085. [PMID: 30669528 PMCID: PMC6356245 DOI: 10.3390/v11010085] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
The incorporation of biologically active host proteins into HIV-1 is a well-established phenomenon, particularly due to the budding mechanism of viral egress in which viruses acquire their external lipid membrane directly from the host cell. While this mechanism might seemingly imply that host protein incorporation is a passive uptake of all cellular antigens associated with the plasma membrane at the site of budding, this is not the case. Herein, we review the evidence indicating that host protein incorporation can be a selective and conserved process. We discuss how HIV-1 virions displaying host proteins on their surface can exhibit a myriad of altered phenotypes, with notable impacts on infectivity, homing, neutralization, and pathogenesis. This review describes the canonical and emerging methods to detect host protein incorporation, highlights the well-established host proteins that have been identified on HIV-1 virions, and reflects on the role of these incorporated proteins in viral pathogenesis and therapeutic targeting. Despite many advances in HIV treatment and prevention, there remains a global effort to develop increasingly effective anti-HIV therapies. Given the broad range of biologically active host proteins acquired on the surface of HIV-1, additional studies on the mechanisms and impacts of these incorporated host proteins may inform the development of novel treatments and vaccine designs.
Collapse
Affiliation(s)
- Jonathan Burnie
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| | - Christina Guzzo
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| |
Collapse
|
15
|
HIV-1 Matrix Protein Interactions with tRNA: Implications for Membrane Targeting. J Mol Biol 2018; 430:2113-2127. [PMID: 29752967 DOI: 10.1016/j.jmb.2018.04.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/30/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
The N-terminally myristoylated matrix (MA) domain of the HIV-1 Gag polyprotein promotes virus assembly by targeting Gag to the inner leaflet of the plasma membrane. Recent studies indicate that, prior to membrane binding, MA associates with cytoplasmic tRNAs (including tRNALys3), and in vitro studies of tRNA-dependent MA interactions with model membranes have led to proposals that competitive tRNA interactions contribute to membrane discrimination. We have characterized interactions between native, mutant, and unmyristylated (myr-) MA proteins and recombinant tRNALys3 by NMR spectroscopy and isothermal titration calorimetry. NMR experiments confirm that tRNALys3 interacts with a patch of basic residues that are also important for binding to the plasma membrane marker, phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2]. Unexpectedly, the affinity of MA for tRNALys3 (Kd = 0.63 ± 0.03 μM) is approximately 1 order of magnitude greater than its affinity for PI(4,5)P2-enriched liposomes (Kd(apparent) = 10.2 ± 2.1 μM), and NMR studies indicate that tRNALys3 binding blocks MA association with liposomes, including those enriched with PI(4,5)P2, phosphatidylserine, and cholesterol. However, the affinity of MA for tRNALys3 is diminished by mutations or sample conditions that promote myristate exposure. Since Gag-Gag interactions are known to promote myristate exposure, our findings support virus assembly models in which membrane targeting and genome binding are mechanistically coupled.
Collapse
|
16
|
Cedillo-Barrón L, García-Cordero J, Shrivastava G, Carrillo-Halfon S, León-Juárez M, Bustos Arriaga J, León Valenzuela P, Gutiérrez Castañeda B. The Role of Flaviviral Proteins in the Induction of Innate Immunity. Subcell Biochem 2018; 88:407-442. [PMID: 29900506 DOI: 10.1007/978-981-10-8456-0_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Flaviviruses are positive, single-stranded, enveloped cytoplasmic sense RNA viruses that cause a variety of important diseases worldwide. Among them, Zika virus, West Nile virus, Japanese encephalitis virus, and Dengue virus have the potential to cause severe disease. Extensive studies have been performed to elucidate the structure and replication strategies of flaviviruses, and current studies are aiming to unravel the complex molecular interactions between the virus and host during the very early stages of infection. The outcomes of viral infection and rapid establishment of the antiviral state, depends on viral detection by pathogen recognition receptors and rapid initiation of signalling cascades to induce an effective innate immune response. Extracellular and intracellular pathogen recognition receptors play a crucial role in detecting flavivirus infection and inducing a robust antiviral response. One of the main hallmarks of flaviviral nonstructural proteins is their multiple strategies to antagonise the interferon system. In this chapter, we summarize the molecular characteristics of flaviviral proteins and discuss how viral proteins target different components of the interferon signalling pathway by blocking phosphorylation, enhancing degradation, and downregulating the expression of major components of the Janus kinase/signal transducer and activator of transcription pathway. We also discuss how the interactions of viral proteins with host proteins facilitate viral pathogenesis. Due to the lack of antivirals or prophylactic treatments for many flaviviral infections, it is necessary to fully elucidate how these viruses disrupt cellular processes to influence pathogenesis and disease outcomes.
Collapse
Affiliation(s)
- L Cedillo-Barrón
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico.
| | - J García-Cordero
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico
| | - G Shrivastava
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico
| | - S Carrillo-Halfon
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico
| | - M León-Juárez
- Department of Immunobiochemistry, National Institute of Perinatology, México City, Mexico
| | - J Bustos Arriaga
- Unidad de Biomedicina. Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autonoma de México, Edo. de México, Mexico
| | - Pc León Valenzuela
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico
| | - B Gutiérrez Castañeda
- Immunology Department UMF Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autonoma de México, Edo. de México, Mexico
| |
Collapse
|
17
|
Bagam P, Singh DP, Inda ME, Batra S. Unraveling the role of membrane microdomains during microbial infections. Cell Biol Toxicol 2017; 33:429-455. [PMID: 28275881 PMCID: PMC7088210 DOI: 10.1007/s10565-017-9386-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023]
Abstract
Infectious diseases pose major socioeconomic and health-related threats to millions of people across the globe. Strategies to combat infectious diseases derive from our understanding of the complex interactions between the host and specific bacterial, viral, and fungal pathogens. Lipid rafts are membrane microdomains that play important role in life cycle of microbes. Interaction of microbial pathogens with host membrane rafts influences not only their initial colonization but also their spread and the induction of inflammation. Therefore, intervention strategies aimed at modulating the assembly of membrane rafts and/or regulating raft-directed signaling pathways are attractive approaches for the. management of infectious diseases. The current review discusses the latest advances in terms of techniques used to study the role of membrane microdomains in various pathological conditions and provides updated information regarding the role of membrane rafts during bacterial, viral and fungal infections.
Collapse
Affiliation(s)
- Prathyusha Bagam
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Dhirendra P Singh
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Maria Eugenia Inda
- Departamento de Microbiología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Suipacha, Rosario, Argentina
| | - Sanjay Batra
- Laboratory of Pulmonary Immuno-Toxicology, Department of Environmental Toxicology, Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
18
|
Yang ST, Kreutzberger AJB, Kiessling V, Ganser-Pornillos BK, White JM, Tamm LK. HIV virions sense plasma membrane heterogeneity for cell entry. SCIENCE ADVANCES 2017; 3:e1700338. [PMID: 28782011 PMCID: PMC5489272 DOI: 10.1126/sciadv.1700338] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/12/2017] [Indexed: 05/20/2023]
Abstract
It has been proposed that cholesterol in host cell membranes plays a pivotal role for cell entry of HIV. However, it remains largely unknown why virions prefer cholesterol-rich heterogeneous membranes to uniformly fluid membranes for membrane fusion. Using giant plasma membrane vesicles containing cholesterol-rich ordered and cholesterol-poor fluid lipid domains, we demonstrate that the HIV receptor CD4 is substantially sequestered into ordered domains, whereas the co-receptor CCR5 localizes preferentially at ordered/disordered domain boundaries. We also show that HIV does not fuse from within ordered regions of the plasma membrane but rather at their boundaries. Ordered/disordered lipid domain coexistence is not required for HIV attachment but is a prerequisite for successful fusion. We propose that HIV virions sense and exploit membrane discontinuities to gain entry into cells. This study provides surprising answers to the long-standing question about the roles of cholesterol and ordered lipid domains in cell entry of HIV and perhaps other enveloped viruses.
Collapse
Affiliation(s)
- Sung-Tae Yang
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA
| | - Alex J. B. Kreutzberger
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA
| | - Volker Kiessling
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA
| | - Barbie K. Ganser-Pornillos
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA
| | - Judith M. White
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Lukas K. Tamm
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA
- Corresponding author.
| |
Collapse
|
19
|
Huang R, Zhu G, Zhang J, Lai Y, Xu Y, He J, Xie J. Betanodavirus-like particles enter host cells via clathrin-mediated endocytosis in a cholesterol-, pH- and cytoskeleton-dependent manner. Vet Res 2017; 48:8. [PMID: 28179028 PMCID: PMC5299686 DOI: 10.1186/s13567-017-0412-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/15/2016] [Indexed: 11/10/2022] Open
Abstract
Betanodavirus, also referred to nervous necrosis virus (NNV), is the causative agent of the fatal disease, viral nervous necrosis and has brought significant economic losses in marine and freshwater cultured fish, especially larvae and juveniles. Here, we used an established invasion model with virus-like particle (VLP)-cells, mimicking orange-spotted grouper nervous necrosis virus (OGNNV), to investigate the crucial events of virus entry. VLP were observed in the perinuclear regions of Asian sea bass (SB) cells within 1.5 h after attachment. VLP uptake was strongly inhibited when cells were pretreated with biochemical inhibitors (chlorpromazine and dynasore) blocking clathrin-mediated endocytosis (CME) or transfected with siRNA against clathrin heavy and light chains. Inhibitors against key regulators of caveolae/raft-dependent endocytosis and macropinocytosis had no effect on VLP uptake. In contrast, disruption of cellular cholesterol by methyl-β-cyclodextrin or reduction of cholesterol fluidity by Cholera toxin B subunit significantly decreased VLP entry. Furthermore, VLP entry is dependent on low pH and cytoskeleton, demonstrated by inhibitor (chloroquine, ammonia chloride, cytochalasin D, wiskostatin, and nocodazole) perturbation. Therefore, OGNNV VLP enter SB cells via CME depending on dynamin-2, cholesterol and its fluidity, low pH, and cytoskeleton. In addition, ten more cell lines were screened for VLP entry and VLP can only enter NNV-sensitive cells, GB and SSN-1, via CME, indicating that CME is the common endocytosis pathway for VLP. These results may provide the data for NNV entry without the influence of the viral genome, an ideal model for exploring the behaviour of betanodavirus in cells, and valuable references to vaccine development.
Collapse
Affiliation(s)
- Runqing Huang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guohua Zhu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Zhang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuxiong Lai
- Department of Nephrology, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China
| | - Yu Xu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Jianguo He
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China.,School of Marine Sciences, Sun Yat-sen University, Guangzhou, China
| | - Junfeng Xie
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
20
|
Guo H, Huang M, Yuan Q, Wei Y, Gao Y, Mao L, Gu L, Tan YW, Zhong Y, Liu D, Sun S. The Important Role of Lipid Raft-Mediated Attachment in the Infection of Cultured Cells by Coronavirus Infectious Bronchitis Virus Beaudette Strain. PLoS One 2017; 12:e0170123. [PMID: 28081264 PMCID: PMC5231368 DOI: 10.1371/journal.pone.0170123] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 12/29/2016] [Indexed: 11/19/2022] Open
Abstract
Lipid raft is an important element for the cellular entry of some viruses, including coronavirus infectious bronchitis virus (IBV). However, the exact role of lipid rafts in the cellular membrane during the entry of IBV into host cells is still unknown. In this study, we biochemically fractionated IBV-infected cells via sucrose density gradient centrifugation after depleting plasma membrane cholesterol with methyl-β-cyclodextrin or Mevastatin. Our results demonstrated that unlike IBV non-structural proteins, IBV structural proteins co-localized with lipid raft marker caveolin-1. Infectivity assay results of Vero cells illustrated that the drug-induced disruption of lipid rafts significantly suppressed IBV infection. Further studies revealed that lipid rafts were not required for IBV genome replication or virion release at later stages. However, the drug-mediated depletion of lipid rafts in Vero cells before IBV attachment significantly reduced the expression of viral structural proteins, suggesting that drug treatment impaired the attachment of IBV to the cell surface. Our results indicated that lipid rafts serve as attachment factors during the early stages of IBV infection, especially during the attachment stage.
Collapse
Affiliation(s)
- Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
| | - Mei Huang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Quan Yuan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yanquan Wei
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
| | - Yuan Gao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
| | - Lejiao Mao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
| | - Lingjun Gu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
- College of Animal Science, Yangtze University, Jingzhou, P.R. China
| | - Yong Wah Tan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yanxin Zhong
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Dingxiang Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- * E-mail: (SS); (DL)
| | - Shiqi Sun
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping, Lanzhou, Gansu, The P.R. China
- * E-mail: (SS); (DL)
| |
Collapse
|
21
|
Kraft ML. Sphingolipid Organization in the Plasma Membrane and the Mechanisms That Influence It. Front Cell Dev Biol 2017; 4:154. [PMID: 28119913 PMCID: PMC5222807 DOI: 10.3389/fcell.2016.00154] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 12/27/2016] [Indexed: 11/13/2022] Open
Abstract
Sphingolipids are structural components in the plasma membranes of eukaryotic cells. Their metabolism produces bioactive signaling molecules that modulate fundamental cellular processes. The segregation of sphingolipids into distinct membrane domains is likely essential for cellular function. This review presents the early studies of sphingolipid distribution in the plasma membranes of mammalian cells that shaped the most popular current model of plasma membrane organization. The results of traditional imaging studies of sphingolipid distribution in stimulated and resting cells are described. These data are compared with recent results obtained with advanced imaging techniques, including super-resolution fluorescence detection and high-resolution secondary ion mass spectrometry (SIMS). Emphasis is placed on the new insight into the sphingolipid organization within the plasma membrane that has resulted from the direct imaging of stable isotope-labeled lipids in actual cell membranes with high-resolution SIMS. Super-resolution fluorescence techniques have recently revealed the biophysical behaviors of sphingolipids and the unhindered diffusion of cholesterol analogs in the membranes of living cells are ultimately in contrast to the prevailing hypothetical model of plasma membrane organization. High-resolution SIMS studies also conflicted with the prevailing hypothesis, showing sphingolipids are concentrated in micrometer-scale membrane domains, but cholesterol is evenly distributed within the plasma membrane. Reductions in cellular cholesterol decreased the number of sphingolipid domains in the plasma membrane, whereas disruption of the cytoskeleton eliminated them. In addition, hemagglutinin, a transmembrane protein that is thought to be a putative raft marker, did not cluster within sphingolipid-enriched regions in the plasma membrane. Thus, sphingolipid distribution in the plasma membrane is dependent on the cytoskeleton, but not on favorable interactions with cholesterol or hemagglutinin. The alternate views of plasma membrane organization suggested by these findings are discussed.
Collapse
Affiliation(s)
- Mary L Kraft
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana, IL, USA
| |
Collapse
|
22
|
Sette P, O'Connor SK, Yerramilli VS, Dussupt V, Nagashima K, Chutiraka K, Lingappa J, Scarlata S, Bouamr F. HIV-1 Nucleocapsid Mimics the Membrane Adaptor Syntenin PDZ to Gain Access to ESCRTs and Promote Virus Budding. Cell Host Microbe 2016; 19:336-48. [PMID: 26962944 DOI: 10.1016/j.chom.2016.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/07/2016] [Accepted: 02/11/2016] [Indexed: 01/09/2023]
Abstract
HIV-1 recruits cellular endosomal sorting complexes required for transport (ESCRTs) to bud virions from the membrane. Disruption of the viral nucleocapsid (NC) domain integrity affects HIV-1 budding. However, the molecular mechanisms of NC's involvement in HIV budding remain unclear. We find that NC mimics the PDZ domains of syntenin, a membrane-binding adaptor involved in cell-to-cell contact/communication, to capture the Bro1 domain of ALIX, which is an ESCRTs recruiting cellular adaptor. NC binds membranes via basic residues in either the distal or proximal zinc fingers, and NC-membrane binding is essential for Bro1 capture and HIV-1 budding. Removal of RNA enhances NC membrane binding, suggesting a dynamic competition between membrane lipids and RNA for the same binding sites in NC. Remarkably, syntenin PDZ can substitute for NC function in HIV-1 budding. Thus, NC mimics syntenin PDZs to function as a membrane-binding adaptor critical for HIV-1 budding at specific microdomains of the membrane.
Collapse
Affiliation(s)
- Paola Sette
- Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20894, USA
| | - Sarah K O'Connor
- Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20894, USA
| | - V Siddartha Yerramilli
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Vincent Dussupt
- Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20894, USA
| | - Kunio Nagashima
- Electron Microscope Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Kasana Chutiraka
- Department of Global Health, University of Washington, Seattle, WA 98102, USA
| | - Jaisri Lingappa
- Department of Global Health, University of Washington, Seattle, WA 98102, USA
| | - Suzanne Scarlata
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Fadila Bouamr
- Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20894, USA.
| |
Collapse
|
23
|
Bajimaya S, Hayashi T, Frankl T, Bryk P, Ward B, Takimoto T. Cholesterol reducing agents inhibit assembly of type I parainfluenza viruses. Virology 2016; 501:127-135. [PMID: 27915128 DOI: 10.1016/j.virol.2016.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 11/18/2022]
Abstract
Many enveloped RNA viruses utilize lipid rafts for the assembly of progeny virions, but the role of cholesterol, a major component of rafts, on paramyxovirus budding and virion formation is controversial. In this study, we analyzed the effects of FDA-approved cholesterol-reducing agents, gemfibrozil and lovastatin, on raft formation and assembly of human parainfluenza virus type 1 (hPIV1) and Sendai virus (SeV). Treatment of the human airway epithelial A549 cells with the agents, especially when combined, significantly decreased production of infectious hPIV1 and SeV. Mechanistic analysis indicated that depletion of cellular cholesterol reduced cell surface accumulation of envelope glycoproteins and association of viral matrix and nucleocapsids with raft membrane, which resulted in impaired virus budding and release from the cells. These results indicate that cellular cholesterol is required for assembly and formation of type 1 parainfluenza viruses and suggest that cholesterol could be an attractive target for antiviral agents against hPIV1.
Collapse
Affiliation(s)
- Shringkhala Bajimaya
- Department of Microbiology and Immunology, University of Rochester Medical Center, Box 672, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Tsuyoshi Hayashi
- Department of Microbiology and Immunology, University of Rochester Medical Center, Box 672, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Tünde Frankl
- Department of Microbiology and Immunology, University of Rochester Medical Center, Box 672, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Peter Bryk
- Department of Microbiology and Immunology, University of Rochester Medical Center, Box 672, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Brian Ward
- Department of Microbiology and Immunology, University of Rochester Medical Center, Box 672, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Toru Takimoto
- Department of Microbiology and Immunology, University of Rochester Medical Center, Box 672, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| |
Collapse
|
24
|
Urbanowicz RA, Lacek K, Lahm A, Bienkowska-Szewczyk K, Ball JK, Nicosia A, Cortese R, Pessi A. Cholesterol conjugation potentiates the antiviral activity of an HIV immunoadhesin. J Pept Sci 2016; 21:743-9. [PMID: 26292842 DOI: 10.1002/psc.2802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 01/04/2023]
Abstract
Immunoadhesins are engineered proteins combining the constant domain (Fc) of an antibody with a ligand-binding (adhesion) domain. They have significant potential as therapeutic agents, because they maintain the favourable pharmacokinetics of antibodies with an expanded repertoire of ligand-binding domains: proteins, peptides, or small molecules. We have recently reported that the addition of a cholesterol group to two HIV antibodies can dramatically improve their antiviral potency. Cholesterol, which can be conjugated at various positions in the antibody, including the constant (Fc) domain, endows the conjugate with affinity for the membrane lipid rafts, thus increasing its concentration at the site where viral entry occurs. Here, we extend this strategy to an HIV immunoadhesin, combining a cholesterol-conjugated Fc domain with the peptide fusion inhibitor C41. The immunoadhesin C41-Fc-chol displayed high affinity for Human Embryonic Kidney (HEK) 293 cells, and when tested on a panel of HIV-1 strains, it was considerably more potent than the unconjugated C41-Fc construct. Potentiation of antiviral activity was comparable to what was previously observed for the cholesterol-conjugated HIV antibodies. Given the key role of cholesterol in lipid raft formation and viral fusion, we expect that the same strategy should be broadly applicable to enveloped viruses, for many of which it is already known the sequence of a peptide fusion inhibitor similar to C41. Moreover, the sequence of heptad repeat-derived fusion inhibitors can often be predicted from genomic information alone, opening a path to immunoadhesins against emerging viruses.
Collapse
Affiliation(s)
- Richard A Urbanowicz
- School of Life Sciences, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom.,Nottingham Digestive Diseases Centre Biomedical Research Unit, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Krzysztof Lacek
- CEINGE, Via Gaetano Salvatore 486, 80145, Napoli, Italy.,Laboratory of Virus Molecular Biology, University of Gdansk, 80-822, Gdansk, Poland
| | - Armin Lahm
- PeptiPharma, Viale Città D'Europa 679, 00144, Roma, Italy
| | | | - Jonathan K Ball
- School of Life Sciences, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom.,Nottingham Digestive Diseases Centre Biomedical Research Unit, The University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - Alfredo Nicosia
- CEINGE, Via Gaetano Salvatore 486, 80145, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Napoli, Italy
| | | | - Antonello Pessi
- CEINGE, Via Gaetano Salvatore 486, 80145, Napoli, Italy.,PeptiPharma, Viale Città D'Europa 679, 00144, Roma, Italy.,JV Bio, Via Gaetano Salvatore 486, 80145, Napoli, Italy
| |
Collapse
|
25
|
Mercredi PY, Bucca N, Loeliger B, Gaines CR, Mehta M, Bhargava P, Tedbury PR, Charlier L, Floquet N, Muriaux D, Favard C, Sanders CR, Freed EO, Marchant J, Summers MF. Structural and Molecular Determinants of Membrane Binding by the HIV-1 Matrix Protein. J Mol Biol 2016; 428:1637-55. [PMID: 26992353 DOI: 10.1016/j.jmb.2016.03.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 10/22/2022]
Abstract
Assembly of HIV-1 particles is initiated by the trafficking of viral Gag polyproteins from the cytoplasm to the plasma membrane, where they co-localize and bud to form immature particles. Membrane targeting is mediated by the N-terminally myristoylated matrix (MA) domain of Gag and is dependent on the plasma membrane marker phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2]. Recent studies revealed that PI(4,5)P2 molecules containing truncated acyl chains [tr-PI(4,5)P2] are capable of binding MA in an "extended lipid" conformation and promoting myristoyl exposure. Here we report that tr-PI(4,5)P2 molecules also readily bind to non-membrane proteins, including HIV-1 capsid, which prompted us to re-examine MA-PI(4,5)P2 interactions using native lipids and membrane mimetic liposomes and bicelles. Liposome binding trends observed using a recently developed NMR approach paralleled results of flotation assays, although the affinities measured under the equilibrium conditions of NMR experiments were significantly higher. Native PI(4,5)P2 enhanced MA binding to liposomes designed to mimic non-raft-like regions of the membrane, suggesting the possibility that binding of the protein to disordered domains may precede Gag association with, or nucleation of, rafts. Studies with bicelles revealed a subset of surface and myr-associated MA residues that are sensitive to native PI(4,5)P2, but cleft residues that interact with the 2'-acyl chains of tr-PI(4,5)P2 molecules in aqueous solution were insensitive to native PI(4,5)P2 in bicelles. Our findings call to question extended-lipid MA:membrane binding models, and instead support a model put forward from coarse-grained simulations indicating that binding is mediated predominantly by dynamic, electrostatic interactions between conserved basic residues of MA and multiple PI(4,5)P2 and phosphatidylserine molecules.
Collapse
Affiliation(s)
- Peter Y Mercredi
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Nadine Bucca
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Burk Loeliger
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Christy R Gaines
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Mansi Mehta
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Pallavi Bhargava
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Philip R Tedbury
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, National Cancer Institute at Fredrick, Fredrick, MD 21702-1201, USA
| | - Landry Charlier
- Institut des Biomolécules Max Mousseron, CNRS UMR5247, Université Montpellier, Faculté de Pharmacie, Montpellier Cedex 05, France
| | - Nicolas Floquet
- Institut des Biomolécules Max Mousseron, CNRS UMR5247, Université Montpellier, Faculté de Pharmacie, Montpellier Cedex 05, France
| | - Delphine Muriaux
- Centre d'études d'agents Pathogénes et Biotechnologies pour la Santé CNRS-UMR 5236, Université Montpellier, Montpellier Cedex 5, France
| | - Cyril Favard
- Centre d'études d'agents Pathogénes et Biotechnologies pour la Santé CNRS-UMR 5236, Université Montpellier, Montpellier Cedex 5, France
| | - Charles R Sanders
- Department of Biochemistry, Center for Structural Biology, and Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37240-7917, USA
| | - Eric O Freed
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, National Cancer Institute at Fredrick, Fredrick, MD 21702-1201, USA.
| | - Jan Marchant
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Maryland, Baltimore County, Baltimore, MD 21250, USA.
| | - Michael F Summers
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of Maryland, Baltimore County, Baltimore, MD 21250, USA.
| |
Collapse
|
26
|
Hawkes D, Jones KL, Smyth RP, Pereira CF, Bittman R, Jaworowski A, Mak J. Properties of HIV-1 associated cholesterol in addition to raft formation are important for virus infection. Virus Res 2015; 210:18-21. [DOI: 10.1016/j.virusres.2015.06.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/25/2015] [Accepted: 06/25/2015] [Indexed: 11/28/2022]
|
27
|
Brown LA, Cox C, Baptiste J, Summers H, Button R, Bahlow K, Spurrier V, Kyser J, Luttge BG, Kuo L, Freed EO, Summers MF. NMR structure of the myristylated feline immunodeficiency virus matrix protein. Viruses 2015; 7:2210-29. [PMID: 25941825 PMCID: PMC4452903 DOI: 10.3390/v7052210] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 03/30/2015] [Accepted: 04/21/2015] [Indexed: 11/25/2022] Open
Abstract
Membrane targeting by the Gag proteins of the human immunodeficiency viruses (HIV types-1 and -2) is mediated by Gag's N-terminally myristylated matrix (MA) domain and is dependent on cellular phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2]. To determine if other lentiviruses employ a similar membrane targeting mechanism, we initiated studies of the feline immunodeficiency virus (FIV), a widespread feline pathogen with potential utility for development of human therapeutics. Bacterial co-translational myristylation was facilitated by mutation of two amino acids near the amino-terminus of the protein (Q5A/G6S; myrMAQ5A/G6S). These substitutions did not affect virus assembly or release from transfected cells. NMR studies revealed that the myristyl group is buried within a hydrophobic pocket in a manner that is structurally similar to that observed for the myristylated HIV-1 protein. Comparisons with a recent crystal structure of the unmyristylated FIV protein [myr(-)MA] indicate that only small changes in helix orientation are required to accommodate the sequestered myr group. Depletion of PI(4,5)P2 from the plasma membrane of FIV-infected CRFK cells inhibited production of FIV particles, indicating that, like HIV, FIV hijacks the PI(4,5)P2 cellular signaling system to direct intracellular Gag trafficking during virus assembly.
Collapse
Affiliation(s)
- Lola A Brown
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Cassiah Cox
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Janae Baptiste
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Holly Summers
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Ryan Button
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Kennedy Bahlow
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Vaughn Spurrier
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Jenna Kyser
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Benjamin G Luttge
- Virus-Cell Interaction Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA.
| | - Lillian Kuo
- Virus-Cell Interaction Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA.
| | - Eric O Freed
- Virus-Cell Interaction Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA.
| | - Michael F Summers
- Howard Hughes Medical Institute, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
28
|
Lorenz M, Vollmer B, Unsay JD, Klupp BG, García-Sáez AJ, Mettenleiter TC, Antonin W. A single herpesvirus protein can mediate vesicle formation in the nuclear envelope. J Biol Chem 2015; 290:6962-74. [PMID: 25605719 DOI: 10.1074/jbc.m114.627521] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Herpesviruses assemble capsids in the nucleus and egress by unconventional vesicle-mediated trafficking through the nuclear envelope. Capsids bud at the inner nuclear membrane into the nuclear envelope lumen. The resulting intralumenal vesicles fuse with the outer nuclear membrane, delivering the capsids to the cytoplasm. Two viral proteins are required for vesicle formation, the tail-anchored pUL34 and its soluble interactor, pUL31. Whether cellular proteins are involved is unclear. Using giant unilamellar vesicles, we show that pUL31 and pUL34 are sufficient for membrane budding and scission. pUL34 function can be bypassed by membrane tethering of pUL31, demonstrating that pUL34 is required for pUL31 membrane recruitment but not for membrane remodeling. pUL31 can inwardly deform membranes by oligomerizing on their inner surface to form buds that constrict to vesicles. Therefore, a single viral protein can mediate all events necessary for membrane budding and abscission.
Collapse
Affiliation(s)
- Michael Lorenz
- From the Friedrich Miescher Laboratory of the Max Planck Society, 72076 Tübingen, Germany
| | - Benjamin Vollmer
- From the Friedrich Miescher Laboratory of the Max Planck Society, 72076 Tübingen, Germany
| | - Joseph D Unsay
- the Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany, and
| | - Barbara G Klupp
- the Friedrich Loeffler Institute, Federal Research Institute for Animal Health, 17493 Greifswald, Germany
| | - Ana J García-Sáez
- the Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany, and
| | - Thomas C Mettenleiter
- the Friedrich Loeffler Institute, Federal Research Institute for Animal Health, 17493 Greifswald, Germany
| | - Wolfram Antonin
- From the Friedrich Miescher Laboratory of the Max Planck Society, 72076 Tübingen, Germany,
| |
Collapse
|
29
|
Tedbury PR, Freed EO. HIV-1 gag: an emerging target for antiretroviral therapy. Curr Top Microbiol Immunol 2015; 389:171-201. [PMID: 25731773 DOI: 10.1007/82_2015_436] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
The advances made in the treatment of HIV-1 infection represent a major success of modern biomedical research, prolonging healthy life and reducing virus transmission. There remain, however, many challenges relating primarily to side effects of long-term therapy and the ever-present danger of the emergence of drug-resistant strains. To counter these threats, there is a continuing need for new and better drugs, ideally targeting multiple independent steps in the HIV-1 replication cycle. The most successful current drugs target the viral enzymes: protease (PR), reverse transcriptase (RT), and integrase (IN). In this review, we outline the advances made in targeting the Gag protein and its mature products, particularly capsid and nucleocapsid, and highlight possible targets for future pharmacological intervention.
Collapse
Affiliation(s)
- Philip R Tedbury
- Virus-Cell Interaction Section, HIV Drug Resistance Program, National Cancer Institute, Center for Cancer Research, Frederick, MD, 21702-1201, USA
| | | |
Collapse
|
30
|
Lacek K, Urbanowicz RA, Troise F, De Lorenzo C, Severino V, Di Maro A, Tarr AW, Ferrara F, Ploss A, Temperton N, Ball JK, Nicosia A, Cortese R, Pessi A. Dramatic potentiation of the antiviral activity of HIV antibodies by cholesterol conjugation. J Biol Chem 2014; 289:35015-28. [PMID: 25342747 PMCID: PMC4263897 DOI: 10.1074/jbc.m114.591826] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The broadly neutralizing antibodies HIV 2F5 and 4E10, which bind to overlapping epitopes in the membrane-proximal external region of the fusion protein gp41, have been proposed to use a two-step mechanism for neutralization; first, they bind and preconcentrate at the viral membrane through their long, hydrophobic CDRH3 loops, and second, they form a high affinity complex with the protein epitope. Accordingly, mutagenesis of the CDRH3 can abolish their neutralizing activity, with no change in the affinity for the peptide epitope. We show here that we can mimic this mechanism by conjugating a cholesterol group outside of the paratope of an antibody. Cholesterol-conjugated antibodies bind to lipid raft domains on the membrane, and because of this enrichment, they show increased antiviral potency. In particular, we find that cholesterol conjugation (i) rescues the antiviral activity of CDRH3-mutated 2F5, (ii) increases the antiviral activity of WT 2F5, (iii) potentiates the non-membrane-binding HIV antibody D5 10–100-fold (depending on the virus strain), and (iv) increases synergy between 2F5 and D5. Conjugation can be made at several positions, including variable and constant domains. Cholesterol conjugation therefore appears to be a general strategy to boost the potency of antiviral antibodies, and, because membrane affinity is engineered outside of the antibody paratope, it can complement affinity maturation strategies.
Collapse
Affiliation(s)
- Krzysztof Lacek
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy, the Laboratory of Virus Molecular Biology, University of Gdansk, 80-822 Gdansk, Poland
| | - Richard A Urbanowicz
- the School of Life Sciences and Nottingham Digestive Diseases Centre Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Fulvia Troise
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy
| | - Claudia De Lorenzo
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy, the Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Napoli (NA), Italy
| | - Valeria Severino
- the Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples, Via Vivaldi 43, 81100 Caserta (CE), Italy
| | - Antimo Di Maro
- the Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples, Via Vivaldi 43, 81100 Caserta (CE), Italy
| | - Alexander W Tarr
- the School of Life Sciences and Nottingham Digestive Diseases Centre Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Francesca Ferrara
- the Viral Pseudotype Unit, Infectious Diseases and Allergy group, School of Pharmacy, University of Kent, Kent ME4 4TB, United Kingdom
| | - Alexander Ploss
- the Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, and
| | - Nigel Temperton
- the Viral Pseudotype Unit, Infectious Diseases and Allergy group, School of Pharmacy, University of Kent, Kent ME4 4TB, United Kingdom
| | - Jonathan K Ball
- the School of Life Sciences and Nottingham Digestive Diseases Centre Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Alfredo Nicosia
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy, the Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Napoli (NA), Italy
| | - Riccardo Cortese
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy
| | - Antonello Pessi
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy, JV Bio, Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy
| |
Collapse
|
31
|
Pessi A. Cholesterol-conjugated peptide antivirals: a path to a rapid response to emerging viral diseases. J Pept Sci 2014; 21:379-86. [PMID: 25331523 PMCID: PMC7167725 DOI: 10.1002/psc.2706] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/01/2014] [Accepted: 09/15/2014] [Indexed: 12/18/2022]
Abstract
While it is now possible to identify and genetically fingerprint the causative agents of emerging viral diseases, often with extraordinary speed, suitable therapies cannot be developed with equivalent speed, because drug discovery requires information that goes beyond knowledge of the viral genome. Peptides, however, may represent a special opportunity. For all enveloped viruses, fusion between the viral and the target cell membrane is an obligatory step of the life cycle. Class I fusion proteins harbor regions with a repeating pattern of amino acids, the heptad repeats (HRs), that play a key role in fusion, and HR‐derived peptides such as enfuvirtide, in clinical use for HIV, can block the process. Because of their characteristic sequence pattern, HRs are easily identified in the genome by means of computer programs, providing the sequence of candidate peptide inhibitors directly from genomic information. Moreover, a simple chemical modification, the attachment of a cholesterol group, can dramatically increase the antiviral potency of HR‐derived inhibitors and simultaneously improve their pharmacokinetics. Further enhancement can be provided by dimerization of the cholesterol‐conjugated peptide. The examples reported so far include inhibitors of retroviruses, paramyxoviruses, orthomyxoviruses, henipaviruses, coronaviruses, and filoviruses. For some of these viruses, in vivo efficacy has been demonstrated in suitable animal models. The combination of bioinformatic lead identification and potency/pharmacokinetics improvement provided by cholesterol conjugation may form the basis for a rapid response strategy, where development of an emergency cholesterol‐conjugated therapeutic would immediately follow the availability of the genetic information of a new enveloped virus. Copyright © 2014 European Peptide Society and John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Antonello Pessi
- PeptiPharma, Viale Città D'Europa 679, 00141, Roma, Italy; JV Bio, Via Gaetano Salvatore 486, 80145, Napoli, Italy; CEINGE, Via Gaetano Salvatore 486, 80145, Napoli, Italy
| |
Collapse
|
32
|
Equine viperin restricts equine infectious anemia virus replication by inhibiting the production and/or release of viral Gag, Env, and receptor via distortion of the endoplasmic reticulum. J Virol 2014; 88:12296-310. [PMID: 25122784 DOI: 10.1128/jvi.01379-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Viperin is an endoplasmic reticulum (ER)-associated multifunctional protein that regulates virus replication and possesses broad antiviral activity. In many cases, viperin interferes with the trafficking and budding of viral structural proteins by distorting the membrane transportation system. The lentivirus equine infectious anemia virus (EIAV) has been studied extensively. In this study, we examined the restrictive effect of equine viperin (eViperin) on EIAV replication and investigated the possible molecular basis of this restriction to obtain insights into the effect of this cellular factor on retroviruses. We demonstrated that EIAV infection of primary equine monocyte-derived macrophages (eMDMs) upregulated the expression of eViperin. The overexpression of eViperin significantly inhibited the replication of EIAV in eMDMs, and knockdown of eViperin transcription enhanced the replication of EIAV in eMDMs by approximately 45.8%. Further experiments indicated that eViperin restricts EIAV at multiple steps of viral replication. The overexpression of eViperin inhibited EIAV Gag release. Both the α-helix domain and radical S-adenosylmethionine (SAM) domain were required for this activity. However, the essential motifs in SAM were different from those reported for the inhibition of HIV-1 Gag by human viperin. Furthermore, eViperin disrupted the synthesis of both EIAV Env and receptor, which consequently inhibited viral production and entry, respectively, and this disruption was dependent on the eViperin α-helix domain. Using immunofluorescence assays and electron microscopy, we demonstrated that the α-helix domain is responsible for the distortion of the endoplasmic reticulum (ER). Finally, EIAV did not exhibit counteracting eViperin at the protein level. IMPORTANCE In previous studies, viperin was indicated as restricting virus replications primarily by the inhibition of virus budding. Here, we show that viperin may have multiple antiviral mechanisms, including the reduction of EIAV Gag budding and Env expression, and these activities are dependent on different viperin domains. We especially demonstrate that the overexpression of viperin inhibits EIAV entry by decreasing the level of virus receptor. Therefore, viperin restriction of viruses is determined largely by the dependence of virus on the cellular membrane transportation system.
Collapse
|
33
|
Cui J, Fu X, Xie J, Gao M, Hong M, Chen Y, Su S, Li S. Critical role of cellular cholesterol in bovine rotavirus infection. Virol J 2014; 11:98. [PMID: 24884772 PMCID: PMC4053397 DOI: 10.1186/1743-422x-11-98] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/21/2014] [Indexed: 12/21/2022] Open
Abstract
Background Bovine rotavirus (BRV) is a non-enveloped dsRNA virus that cause neonatal calf diarrhea. Lipid rafts are cholesterol-enrich membrane mircodomains that play a vital role in many cellular processes. In this study, the effect of cellular cholesterol depletion on infection of MA-104 cells with bovine rotavirus was investigated. Results We demonstrated that cholesterol depletion of the plasma membrane by MβCD had no effect on BRV binding to cells but significantly impaired BRV entry in a dose-dependent manner and the effect was partially reversed by addition of exogenous cholesterol, suggesting the reduction of BRV infection by MβCD was specifically due to cholesterol depletion. Cholesterol depletion after virus entry did not reduce BRV replication, whereas affected virus assembly. Conclusions Taken together, our results demonstrate that cell membrane cholesterol is essential to BRV infectivity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shoujun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province 510642, People's Republic of China.
| |
Collapse
|
34
|
Fogarty KH, Berk S, Grigsby IF, Chen Y, Mansky LM, Mueller JD. Interrelationship between cytoplasmic retroviral Gag concentration and Gag-membrane association. J Mol Biol 2013; 426:1611-24. [PMID: 24316368 DOI: 10.1016/j.jmb.2013.11.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/12/2013] [Accepted: 11/23/2013] [Indexed: 12/20/2022]
Abstract
The early events in the retrovirus assembly pathway, particularly the timing and nature of Gag translocation from the site of protein translation to the inner leaflet of the plasma membrane, are poorly understood. We have investigated the interrelationship between cytoplasmic Gag concentration and plasma membrane association using complementary live-cell biophysical fluorescence techniques in real time with both human T-cell leukemia virus type 1 (HTLV-1) and human immunodeficiency virus type 1 (HIV-1) Gag proteins. In particular, dual-color, z-scan fluorescence fluctuation spectroscopy in conjunction with total internal reflection fluorescence and conventional, epi-illumination imaging were utilized. Our results demonstrate that HTLV-1 Gag is capable of membrane targeting and particle assembly at low (i.e., nanomolar) cytoplasmic concentrations and that there is a critical threshold concentration (approaching micromolar) prior to the observation of HIV-1 Gag associated with the plasma membrane. These observations imply fundamental differences between HIV-1 and HTLV-1 Gag trafficking and membrane association.
Collapse
Affiliation(s)
- Keir H Fogarty
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Serkan Berk
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Iwen F Grigsby
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yan Chen
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Louis M Mansky
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; Department of Microbiology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Joachim D Mueller
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA; School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
35
|
Autographa californica Nucleopolyhedrovirus Ac76: a dimeric type II integral membrane protein that contains an inner nuclear membrane-sorting motif. J Virol 2013; 88:1090-103. [PMID: 24198428 DOI: 10.1128/jvi.02392-13] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Our previous study showed that the Autographa californica Nucleopolyhedrovirus (AcMNPV) ac76 gene is essential for both budded virion (BV) and occlusion-derived virion (ODV) development. More importantly, deletion of ac76 affects intranuclear microvesicle formation. However, the exact role by which ac76 affects virion morphogenesis remains unknown. In this report, we characterized the expression, distribution, and topology of Ac76 to further understand the functional role of Ac76 in virion morphogenesis. Ac76 contains an α-helical transmembrane domain, and phase separation showed that it was an integral membrane protein. In AcMNPV-infected cells, Ac76 was detected as a stable dimer that was resistant to SDS and thermal denaturation, and only a trace amount of monomer was detected. A coimmunoprecipitation assay demonstrated the dimerization of Ac76 by high-affinity self-association. Western blot analyses of purified virions and their nucleocapsid and envelope fractions showed that Ac76 was associated with the envelope fractions of both BVs and ODVs. Immunoelectron microscopy revealed that Ac76 was localized to the plasma membrane, endoplasmic reticulum (ER), nuclear membrane, intranuclear microvesicles, and ODV envelope. Amino acids 15 to 48 of Ac76 were identified as an atypical inner nuclear membrane-sorting motif because it was sufficient to target fusion proteins to the ER and nuclear membrane in the absence of viral infection and to the intranuclear microvesicles and ODV envelope during infection. Topology analysis of Ac76 by selective permeabilization showed that Ac76 was a type II integral membrane protein with an N terminus exposed to the cytosol and a C terminus hidden in the ER lumen.
Collapse
|
36
|
Tan KS, Ng WC, Seet JE, Olfat F, Engelward BP, Chow VTK. Investigating the efficacy of pamidronate, a chemical inhibitor of farnesyl pyrophosphate synthase, in the inhibition of influenza virus infection in vitro and in vivo. Mol Med Rep 2013; 9:51-6. [PMID: 24154548 DOI: 10.3892/mmr.2013.1750] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/20/2013] [Indexed: 11/06/2022] Open
Abstract
Influenza A virus has caused significant pandemics in the past decades, including the H1N1‑2009 pandemic. Viperin is an interferon‑inducible protein that acts as a broad‑spectrum antiviral protein via the inhibition of farnesyl pyrophosphate synthase (FPPS). To mimic this activity of viperin, the present study investigated the effectiveness of a commercially available FPPS inhibitor (pamidronate) as an inhibitor of influenza virus infection in vitro and in vivo. HeLaM cells were treated with pamidronate to determine its effect on the replication of influenza virus A/H1N1/WSN/1933. C57BL/6 mice were also subjected to intratracheal pamidronate treatment regimes prior to and following lethal influenza challenge. Treatment with the FPPS inhibitor in vitro resulted in a considerable reduction in the viral titer of ~1 log and diminished lipid raft formation without cellular toxicity, thus mimicking the antiviral effect of viperin. However, pamidronate lacked efficacy in vivo and was associated with increased pulmonary damage, most likely due to the complexity of drug‑host interactions in the infected mice. Further studies are warranted on pamidronate treatment in other infectious diseases that are more susceptible to FPPS inhibition.
Collapse
Affiliation(s)
- Kai Sen Tan
- Host And Pathogen Interactivity Laboratory, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Republic of Singapore
| | | | | | | | | | | |
Collapse
|
37
|
Amir D, Fessler DMT. Boots for Achilles: progesterone's reduction of cholesterol is a second-order adaptation. QUARTERLY REVIEW OF BIOLOGY 2013; 88:97-116. [PMID: 23909226 DOI: 10.1086/670528] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Progesterone and cholesterol are both vital to pregnancy. Among other functions, progesterone downregulates inflammatory responses, allowing for maternal immune tolerance of the fetal allograft. Cholesterol a key component of cell membranes, is important in intracellular transport, cell signaling, nerve conduction, and metabolism Despite the importance of each substance in pregnancy, one exercises an antagonistic effect on the other, as periods of peak progesterone correspond with reductions in cholesterol availability, a consequence of progesterone's negative effects on cholesterol biosynthesis. This arrangement is understandable in light of the threat posed by pathogens early in pregnancy. Progesterone-induced immunomodulation entails increased vulnerability to infection, an acute problem in the first trimester, when fetal development is highly susceptible to insult. Many pathogens rely on cholesterol for cell entry, egress, and replication. Progesterone's antagonistic effects on cholesterol thus partially compensate for the costs entailed by progesterone-induced immunomodulation. Among pathogens to which the host's vulnerability is increased by progesterone's effects, approximately 90% utilize cholesterol, and this is notably true of pathogens that pose a risk during pregnancy. In addition to having a number of possible clinical applications, our approach highlights the potential importance of second-order adaptations, themselves a consequence of the lack of teleology in evolutionary processes.
Collapse
Affiliation(s)
- Dorsa Amir
- Center for Behavior, Evolution, and Culture, Department of Anthropology, University of California, Los Angeles Los Angeles, California 90095-1553, USA.
| | | |
Collapse
|
38
|
Lam SM, Shui G. Lipidomics as a Principal Tool for Advancing Biomedical Research. J Genet Genomics 2013; 40:375-90. [DOI: 10.1016/j.jgg.2013.06.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 06/04/2013] [Accepted: 06/19/2013] [Indexed: 01/22/2023]
|
39
|
Agarwal S, Schroeder C, Schlechtingen G, Braxmeier T, Jennings G, Knölker HJ. Evaluation of steroidal amines as lipid raft modulators and potential anti-influenza agents. Bioorg Med Chem Lett 2013; 23:5165-9. [PMID: 23916260 DOI: 10.1016/j.bmcl.2013.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 07/10/2013] [Accepted: 07/11/2013] [Indexed: 11/25/2022]
Abstract
The influenza A virus (IFV) possesses a highly ordered cholesterol-rich lipid envelope. A specific composition and structure of this membrane raft envelope are essential for viral entry into cells and virus budding. Several steroidal amines were investigated for antiviral activity against IFV. Both, a positively charged amino function and the highly hydrophobic (ClogP≥5.9) ring system are required for IC50 values in the low μM range. An amino substituent is preferential to an azacyclic A-ring. We showed that these compounds either disrupt or augment membrane rafts and in some cases inactivate the free virus. Some of the compounds also interfere with virus budding. The antiviral selectivity improved in the series 3-amino, 3-aminomethyl, 3-aminoethyl, or by introducing an OH function in the A-ring. Steroidal amines show a new mode of antiviral action in directly targeting the virus envelope and its biological functions.
Collapse
Affiliation(s)
- Sameer Agarwal
- JADO Technologies, Tatzberg 47-51, 01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Santos da Silva E, Mulinge M, Perez Bercoff D. The frantic play of the concealed HIV envelope cytoplasmic tail. Retrovirology 2013; 10:54. [PMID: 23705972 PMCID: PMC3686653 DOI: 10.1186/1742-4690-10-54] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/01/2013] [Indexed: 11/18/2022] Open
Abstract
Lentiviruses have unusually long envelope (Env) cytoplasmic tails, longer than those of other retroviruses. Whereas the Env ectodomain has received much attention, the gp41 cytoplasmic tail (gp41-CT) is one of the least studied parts of the virus. It displays relatively high conservation compared to the rest of Env. It has been long established that the gp41-CT interacts with the Gag precursor protein to ensure Env incorporation into the virion. The gp41-CT contains distinct motifs and domains that mediate both intensive Env intracellular trafficking and interactions with numerous cellular and viral proteins, optimizing viral infectivity. Although they are not fully understood, a multiplicity of interactions between the gp41-CT and cellular factors have been described over the last decade; these interactions illustrate how Env expression and incorporation into virions is a finely tuned process that has evolved to best exploit the host system with minimized genetic information. This review addresses the structure and topology of the gp41-CT of lentiviruses (mainly HIV and SIV), their domains and believed functions. It also considers the cellular and viral proteins that have been described to interact with the gp41-CT, with a particular focus on subtype-related polymorphisms.
Collapse
|
41
|
Avadisian M, Gunning PT. Extolling the benefits of molecular therapeutic lipidation. MOLECULAR BIOSYSTEMS 2013; 9:2179-88. [DOI: 10.1039/c3mb70147f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
42
|
Puryear WB, Gummuluru S. Role of glycosphingolipids in dendritic cell-mediated HIV-1 trans-infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 762:131-53. [PMID: 22975874 PMCID: PMC3686569 DOI: 10.1007/978-1-4614-4433-6_5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glycosphingolipids (GSLs) are components of the cell membrane that comprise a membrane bound lipid, ceramide, coupled to an extracellular carbohydrate. GSLs impact numerous aspects of membrane biology, including membrane fluidity, curvature, and organization. The role of these molecules in both chronic inflammation and infectious disease and underlying pathogenic mechanisms are just starting to be recognized. As a component of the cell membrane, GSLs are also incorporated into lipid bilayers of diverse enveloped viruses as they bud out from the host cell and can go on to have a significant influence on viral pathogenesis. Dendritic cell (DC) subsets located in the peripheral mucosal tissues are proposed to be one of the earliest cell types that encounter transmitted viruses and help initiate adaptive immune responses against the invading pathogen by interacting with T cells. In turn, viruses, as obligatory intracellular parasites, rely on host cells for completing their replication cycle, and not surprisingly, HIV has evolved to exploit DC biology for the initial transmission event as well as for its dissemination and propagation within the infected host. In this review, we describe the mechanisms by which GSLs impact DC-mediated HIV trans-infection by either modulating virus infectivity, serving as a direct virus particle-associated host-derived ligand for specific interactions with DCs, or modulating the T cell membrane in such a way as to impact viral entry and thereby productive infection of CD4(+) T cells.
Collapse
Affiliation(s)
- Wendy Blay Puryear
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
43
|
Keller H, Kräusslich HG, Schwille P. Multimerizable HIV Gag derivative binds to the liquid-disordered phase in model membranes. Cell Microbiol 2012; 15:237-47. [PMID: 23121220 DOI: 10.1111/cmi.12064] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 10/18/2012] [Accepted: 10/23/2012] [Indexed: 12/15/2022]
Abstract
During HIV assembly, a protein coat on the inner leaflet of the plasma membrane drives the formation of virus particles, and appears to induce the preferential accumulation of 'raft' lipids in the viral envelope, although the lipid raft concept mainly proposes microdomains of these lipids in the outer leaflet. The common hypothesis is that Gag preferentially associates with, and thereby probably induces, raft-like domains, because the protein is multimerized and specifically linked to two saturated acyl chains. To test this hypothesis, we constructed a minimal in vitro system in which we analysed the interaction of a Gag derivative, which could be triggered to multimerize, with a domain-forming model membrane resembling the inner leaflet of the plasma membrane. Confirming studies with authentic Gag, this Gag derivative only bound to membranes when it was multimerized, myristoylated and when phosphatidylinositol 4,5-bisphosphate was present in the membrane. Unexpectedly, however, the multimerized Gag derivative was largely excluded from ordered domains in model membranes. This suggests that the mechanism of membrane reorganization during HIV assembly does not simply result from a higher affinity of the clustered Gag membrane binding domain to ordered membrane domains, but involves more complex biophysical interactions or possibly also an additional protein machinery.
Collapse
Affiliation(s)
- Heiko Keller
- Biophysics, BIOTEC, Technische Universität Dresden, Dresden, Germany
| | | | | |
Collapse
|
44
|
Tang Y, George A, Taylor T, Hildreth JEK. Cholesterol depletion inactivates XMRV and leads to viral envelope protein release from virions: evidence for role of cholesterol in XMRV infection. PLoS One 2012; 7:e48013. [PMID: 23110160 PMCID: PMC3482229 DOI: 10.1371/journal.pone.0048013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 09/24/2012] [Indexed: 01/24/2023] Open
Abstract
Membrane cholesterol plays an important role in replication of HIV-1 and other retroviruses. Here, we report that the gammaretrovirus XMRV requires cholesterol and lipid rafts for infection and replication. We demonstrate that treatment of XMRV with a low concentration (10 mM) of 2-hydroxypropyl-β-cyclodextrin (2OHpβCD) partially depleted virion-associated cholesterol resulting in complete inactivation of the virus. This effect could not be reversed by adding cholesterol back to treated virions. Further analysis revealed that following cholesterol depletion, virus-associated Env protein was significantly reduced while the virions remained intact and retained core proteins. Increasing concentrations of 2OHpβCD (≥20 mM) resulted in loss of the majority of virion-associated cholesterol, causing disruption of membrane integrity and loss of internal Gag proteins and viral RNA. Depletion of cholesterol from XMRV-infected cells significantly reduced virus release, suggesting that cholesterol and intact lipid rafts are required for the budding process of XMRV. These results suggest that unlike glycoproteins of other retroviruses, the association of XMRV glycoprotein with virions is highly dependent on cholesterol and lipid rafts.
Collapse
Affiliation(s)
- Yuyang Tang
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California Davis, Davis, California, United States of America
| | - Alvin George
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California Davis, Davis, California, United States of America
| | - Thyneice Taylor
- Department of Medicine, School of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - James E. K. Hildreth
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
45
|
Lim ES, Wu LI, Malik HS, Emerman M. The function and evolution of the restriction factor Viperin in primates was not driven by lentiviruses. Retrovirology 2012; 9:55. [PMID: 22734835 PMCID: PMC3414838 DOI: 10.1186/1742-4690-9-55] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 06/26/2012] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Viperin, also known as RSAD2, is an interferon-inducible protein that potently restricts a broad range of different viruses such as influenza, hepatitis C virus, human cytomegalovirus and West Nile virus. Viperin is thought to affect virus budding by modification of the lipid environment within the cell. Since HIV-1 and other retroviruses depend on lipid domains of the host cell for budding and infectivity, we investigated the possibility that Viperin also restricts human immunodeficiency virus and other retroviruses. RESULTS Like other host restriction factors that have a broad antiviral range, we find that viperin has also been evolving under positive selection in primates. The pattern of positive selection is indicative of Viperin's escape from multiple viral antagonists over the course of primate evolution. Furthermore, we find that Viperin is interferon-induced in HIV primary target cells. We show that exogenous expression of Viperin restricts the LAI strain of HIV-1 at the stage of virus release from the cell. Nonetheless, the effect of Viperin restriction is highly strain-specific and does not affect most HIV-1 strains or other retroviruses tested. Moreover, knockdown of endogenous Viperin in a lymphocytic cell line did not significantly affect the spreading infection of HIV-1. CONCLUSION Despite positive selection having acted on Viperin throughout primate evolution, our findings indicate that Viperin is not a major restriction factor against HIV-1 and other retroviruses. Therefore, other viral lineages are likely responsible for the evolutionary signatures of positive selection in viperin among primates.
Collapse
Affiliation(s)
- Efrem S Lim
- Department of Microbiology, Fred Hutchinson Cancer Research Center,University of Washington, Seattle, WA 98109-1024, USA
| | | | | | | |
Collapse
|
46
|
Kyere SK, Mercredi PY, Dong X, Spearman P, Summers MF. The HIV-1 matrix protein does not interact directly with the protein interactive domain of AP-3δ. Virus Res 2012; 169:411-4. [PMID: 22705971 DOI: 10.1016/j.virusres.2012.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/06/2012] [Accepted: 06/06/2012] [Indexed: 11/19/2022]
Abstract
During the late phase of the Human Immunodeficiency Virus Type-1 (HIV-1) replication cycle, viral Gag proteins and the intact RNA genome are trafficked to specific sub-cellular membranes where virus assembly and budding occurs. Targeting to the plasma membranes of T cells and macrophages is mediated by interactions between the N-terminal matrix (MA) domain of Gag and cellular phosphatidylinositol-4,5-bisphosphate [PI(4,5)P(2)] molecules. However, in macrophages and dendritic cells, a subset of Gag proteins appears to be targeted to tetraspanin enriched viral compartments, a process that appears to be mediated by MA interactions with the Delta subunit of the cellular Adaptor Protein AP-3 (AP-3δ). We cloned, overexpressed and purified the protein interactive domain of AP-3δ and probed for MA binding by NMR. Unexpectedly, no evidence of binding was observed in these in vitro experiments, even at relatively high protein concentrations (200μM), suggesting that AP-3δ plays an alternative role in HIV-1 assembly.
Collapse
Affiliation(s)
- Sampson K Kyere
- Howard Hughes Medical Institute, Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | | | | | | | | |
Collapse
|
47
|
Tan KS, Olfat F, Phoon MC, Hsu JP, Howe JLC, Seet JE, Chin KC, Chow VTK. In vivo and in vitro studies on the antiviral activities of viperin against influenza H1N1 virus infection. J Gen Virol 2012; 93:1269-1277. [DOI: 10.1099/vir.0.040824-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Influenza A virus has caused a number of pandemics in past decades, including the recent H1N1-2009 pandemic. Viperin is an interferon (IFN)-inducible protein of innate immunity, and acts as a broad-spectrum antiviral protein. We explored the antiviral activities and mechanisms of viperin during influenza virus (IFV) infection in vitro and in vivo. Wild-type (WT) HeLa and viperin-expressing HeLa cells were infected with influenza A/WSN/33/H1N1 (WSN33) virus, and subjected to virological, light and electron microscopic analyses. Viperin expression reduced virus replication and titres, and restricted viral budding. Young and old viperin-knockout (KO) mice and WT control animals were challenged with influenza WSN33 at lethal doses of 103 and 104 p.f.u. via the intratracheal route. Lungs were subjected to histopathological, virological and molecular studies. Upon lethal IFV challenge, both WT and KO mice revealed similar trends of infection and recovery with similar mortality rates. Viral quantification assay and histopathological evaluation of lungs from different time points showed no significant difference in viral loads and lung damage scores between the two groups of mice. Although the in vitro studies demonstrated the ability of viperin to restrict influenza H1N1 virus replication, the viperin-deficient mouse model indicated that absence of viperin enhanced neither the viral load nor pulmonary damage in the lungs of infected mice. This may be due to the compensation of IFN-stimulated genes in the lungs and/or the influenza non-structural protein 1-mediated IFN antagonism dampening the IFN response, thereby rendering the loss of viperin insignificant. Nevertheless, further investigations that exploit the antiviral mechanisms of viperin as prophylaxis are still warranted.
Collapse
Affiliation(s)
- Kai Sen Tan
- Infectious Diseases Interdisciplinary Research Group, Singapore–Massachusetts Institute of Technology Alliance in Research and Technology, Singapore 117456
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Kent Ridge, Singapore 117597
| | - Farzad Olfat
- Infectious Diseases Interdisciplinary Research Group, Singapore–Massachusetts Institute of Technology Alliance in Research and Technology, Singapore 117456
| | - Meng Chee Phoon
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Kent Ridge, Singapore 117597
| | - Jung Pu Hsu
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Kent Ridge, Singapore 117597
| | - Josephine L. C. Howe
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Kent Ridge, Singapore 117597
| | - Ju Ee Seet
- Department of Pathology, National University Hospital, Singapore 119074
| | - Keh Chuang Chin
- Singapore Immunology Network, Immunos, Biopolis, Singapore 138648
| | - Vincent T. K. Chow
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Kent Ridge, Singapore 117597
| |
Collapse
|
48
|
Pessi A, Langella A, Capitò E, Ghezzi S, Vicenzi E, Poli G, Ketas T, Mathieu C, Cortese R, Horvat B, Moscona A, Porotto M. A general strategy to endow natural fusion-protein-derived peptides with potent antiviral activity. PLoS One 2012; 7:e36833. [PMID: 22666328 PMCID: PMC3353973 DOI: 10.1371/journal.pone.0036833] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 04/07/2012] [Indexed: 01/26/2023] Open
Abstract
Fusion between the viral and target cell membranes is an obligatory step for the infectivity of all enveloped virus, and blocking this process is a clinically validated therapeutic strategy. Viral fusion is driven by specialized proteins which, although specific to each virus, act through a common mechanism, the formation of a complex between two heptad repeat (HR) regions. The HR regions are initially separated in an intermediate termed “prehairpin”, which bridges the viral and cell membranes, and then fold onto each other to form a 6-helical bundle (6HB), driving the two membranes to fuse. HR-derived peptides can inhibit viral infectivity by binding to the prehairpin intermediate and preventing its transition to the 6HB. The antiviral activity of HR-derived peptides differs considerably among enveloped viruses. For weak inhibitors, potency can be increased by peptide engineering strategies, but sequence-specific optimization is time-consuming. In seeking ways to increase potency without changing the native sequence, we previously reported that attachment to the HR peptide of a cholesterol group (”cholesterol-tagging”) dramatically increases its antiviral potency, and simultaneously increases its half-life in vivo. We show here that antiviral potency may be increased by combining cholesterol-tagging with dimerization of the HR-derived sequence, using as examples human parainfluenza virus, Nipah virus, and HIV-1. Together, cholesterol-tagging and dimerization may represent strategies to boost HR peptide potency to levels that in some cases may be compatible with in vivo use, possibly contributing to emergency responses to outbreaks of existing or novel viruses.
Collapse
Affiliation(s)
| | | | | | - Silvia Ghezzi
- Viral Pathogens and Biosafety, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogens and Biosafety, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Guido Poli
- AIDS Immunopathogenesis Units, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Thomas Ketas
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Cyrille Mathieu
- INSERM, Ecole Normale Supérieure de Lyon, Lyon, France
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
| | | | - Branka Horvat
- INSERM, Ecole Normale Supérieure de Lyon, Lyon, France
- IFR128 BioSciences Lyon-Gerland Lyon-Sud, University of Lyon, Lyon, France
| | - Anne Moscona
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Matteo Porotto
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
- * E-mail: (AP); (MP)
| |
Collapse
|
49
|
Ghanam RH, Samal AB, Fernandez TF, Saad JS. Role of the HIV-1 Matrix Protein in Gag Intracellular Trafficking and Targeting to the Plasma Membrane for Virus Assembly. Front Microbiol 2012; 3:55. [PMID: 22363329 PMCID: PMC3281212 DOI: 10.3389/fmicb.2012.00055] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/01/2012] [Indexed: 11/13/2022] Open
Abstract
Human immunodeficiency virus type-1 (HIV-1) encodes a polypeptide called Gag that is able to form virus-like particles in vitro in the absence of any cellular or viral constituents. During the late phase of the HIV-1 infection, Gag polyproteins are transported to the plasma membrane (PM) for assembly. In the past two decades, in vivo, in vitro, and structural studies have shown that Gag trafficking and targeting to the PM are orchestrated events that are dependent on multiple factors including cellular proteins and specific membrane lipids. The matrix (MA) domain of Gag has been the focus of these studies as it appears to be engaged in multiple intracellular interactions that are suggested to be critical for virus assembly and replication. The interaction between Gag and the PM is perhaps the most understood. It is now established that the ultimate localization of Gag on punctate sites on the PM is mediated by specific interactions between the MA domain of Gag and phosphatidylinositol-4,5-bisphosphate [PI(4,5)P(2)], a minor lipid localized on the inner leaflet of the PM. Structure-based studies revealed that binding of PI(4,5)P(2) to MA induces minor conformational changes, leading to exposure of the myristyl (myr) group. Exposure of the myr group is also triggered by binding of calmodulin, enhanced by factors that promote protein self-association like the capsid domain of Gag, and is modulated by pH. Despite the steady progress in defining both the viral and cellular determinants of retroviral assembly and release, Gag's intracellular interactions and trafficking to its assembly sites in the infected cell are poorly understood. In this review, we summarize the current understanding of the structural and functional role of MA in HIV replication.
Collapse
Affiliation(s)
- Ruba H Ghanam
- Department of Microbiology, University of Alabama at Birmingham Birmingham, AL, USA
| | | | | | | |
Collapse
|
50
|
Ivanyi-Nagy R, Darlix JL. Fuzziness in the Core of the Human Pathogenic Viruses HCV and HIV. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 725:142-58. [DOI: 10.1007/978-1-4614-0659-4_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|