Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Elife ; 122024 Mar 25.
Article in English | MEDLINE | ID: mdl-38526940

ABSTRACT

Marburg virus (MARV) is one of the filovirus species that cause deadly hemorrhagic fever in humans, with mortality rates up to 90%. Neutralizing antibodies represent ideal candidates to prevent or treat virus disease. However, no antibody has been approved for MARV treatment to date. In this study, we identified a novel human antibody named AF-03 that targeted MARV glycoprotein (GP). AF-03 possessed a high binding affinity to MARV GP and showed neutralizing and protective activities against the pseudotyped MARV in vitro and in vivo. Epitope identification, including molecular docking and experiment-based analysis of mutated species, revealed that AF-03 recognized the Niemann-Pick C1 (NPC1) binding domain within GP1. Interestingly, we found the neutralizing activity of AF-03 to pseudotyped Ebola viruses (EBOV, SUDV, and BDBV) harboring cleaved GP instead of full-length GP. Furthermore, NPC2-fused AF-03 exhibited neutralizing activity to several filovirus species and EBOV mutants via binding to CI-MPR. In conclusion, this work demonstrates that AF-03 represents a promising therapeutic cargo for filovirus-caused disease.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Marburgvirus , Humans , Antibodies, Viral , Molecular Docking Simulation , Glycoproteins , Hemorrhagic Fever, Ebola/prevention & control , Ebolavirus/chemistry
2.
J Virol ; 97(10): e0059023, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37750724

ABSTRACT

IMPORTANCE: Ebola disease (EBOD) is a public health threat with a high case fatality rate. Most EBOD outbreaks have occurred in remote locations, but the 2013-2016 Western Africa outbreak demonstrated how devastating EBOD can be when it reaches an urban population. Here, the 2022 Sudan virus disease (SVD) outbreak in Mubende District, Uganda, is summarized, and the genetic relatedness of the new variant is evaluated. The Mubende variant exhibited 96% amino acid similarity with historic SUDV sequences from the 1970s and a high degree of conservation throughout the outbreak, which was important for ongoing diagnostics and highly promising for future therapy development. Genetic differences between viruses identified during the Mubende SVD outbreak were linked with epidemiological data to better interpret viral spread and contact tracing chains. This methodology should be used to better integrate discrete epidemiological and sequence data for future viral outbreaks.


Subject(s)
Disease Outbreaks , Ebolavirus , Genetic Variation , Hemorrhagic Fever, Ebola , Humans , Disease Outbreaks/statistics & numerical data , Ebolavirus/chemistry , Ebolavirus/classification , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/transmission , Hemorrhagic Fever, Ebola/virology , Uganda/epidemiology , Contact Tracing
3.
Int J Mol Sci ; 24(5)2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36902059

ABSTRACT

The Ebola virus glycoprotein (GP) gene templates several mRNAs that produce either the virion-associated transmembrane protein or one of two secreted glycoproteins. Soluble glycoprotein (sGP) is the predominant product. GP1 and sGP share an amino terminal sequence of 295 amino acids but differ in quaternary structure, with GP1 being a heterohexamer with GP2 and sGP a homodimer. Two structurally different DNA aptamers were selected against sGP that also bound GP1,2. These DNA aptamers were compared with a 2'FY-RNA aptamer for their interactions with the Ebola GP gene products. The three aptamers have almost identical binding isotherms for sGP and GP1,2 in solution and on the virion. They demonstrated high affinity and selectivity for sGP and GP1,2. Furthermore, one aptamer, used as a sensing element in an electrochemical format, detected GP1,2 on pseudotyped virions and sGP with high sensitivity in the presence of serum, including from an Ebola-virus-infected monkey. Our results suggest that the aptamers interact with sGP across the interface between the monomers, which is different from the sites on the protein bound by most antibodies. The remarkable similarity in functional features of three structurally distinct aptamers suggests that aptamers, like antibodies, have preferred binding sites on proteins.


Subject(s)
Aptamers, Nucleotide , Ebolavirus , Viral Envelope Proteins , Humans , Aptamers, Nucleotide/chemistry , Ebolavirus/chemistry , Viral Envelope Proteins/chemistry , Protein Multimerization
4.
Microscopy (Oxf) ; 72(3): 178-190, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-36242583

ABSTRACT

Filoviruses are filamentous enveloped viruses belonging to the family Filoviridae, in the order Mononegavirales. Some filovirus members, such as Ebola virus and Marburg virus, cause severe hemorrhagic fever in humans and non-human primates. The filovirus ribonucleoprotein complex, called the nucleocapsid, forms a double-layered helical structure in which a non-segmented, single-stranded, negative-sense RNA genome is encapsidated by the nucleoprotein (NP), viral protein 35 (VP35), VP24, VP30 and RNA-dependent RNA polymerase (L). The inner layer consists of the helical NP-RNA complex, acting as a scaffold for the binding of VP35 and VP24 that constitute the outer layer. Recent structural studies using cryo-electron microscopy have advanced our understanding of the molecular mechanism of filovirus nucleocapsid formation. Here, we review the key characteristics of the Ebola virus and Marburg virus nucleocapsid structures, highlighting the similarities and differences between the two viruses. In particular, we focus on the structure of the helical NP-RNA complex, the RNA binding mechanism and the NP-NP interactions in the helix. The structural analyses reveal a possible mechanism of nucleocapsid assembly and provide potential targets for the anti-filovirus drug design.


Subject(s)
Ebolavirus , Marburgvirus , Animals , Cryoelectron Microscopy , Nucleocapsid/chemistry , Nucleocapsid/metabolism , Ebolavirus/chemistry , Ebolavirus/metabolism , Marburgvirus/chemistry , Marburgvirus/metabolism , Viral Proteins/analysis , Viral Proteins/chemistry , Viral Proteins/metabolism , RNA/analysis , RNA/metabolism
5.
J Virol ; 96(17): e0108322, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35993739

ABSTRACT

Ebola virus (EBOV) VP30 regulates viral genome transcription and replication by switching its phosphorylation status. However, the importance of VP30 phosphorylation and dephosphorylation in other viral replication processes such as nucleocapsid and virion assembly is unclear. Interestingly, VP30 is predominantly dephosphorylated by cellular phosphatases in viral inclusions, while it is phosphorylated in the released virions. Thus, uncertainties regarding how VP30 phosphorylation in nucleocapsids is achieved and whether VP30 phosphorylation provides any advantages in later steps in viral replication have arisen. In the present study, to characterize the roles of VP30 phosphorylation in nucleocapsid formation, we used electron microscopic analyses and live cell imaging systems. We identified VP30 localized to the surface of protrusions surrounding nucleoprotein (NP)-forming helical structures in the nucleocapsid, suggesting the involvement in assembly and transport of nucleocapsids. Interestingly, VP30 phosphorylation facilitated its association with nucleocapsid-like structures (NCLSs). On the contrary, VP30 phosphorylation does not influence the transport characteristics and NCLS number leaving from and coming back into viral inclusions, indicating that the phosphorylation status of VP30 is not a prerequisite for NCLS departure. Moreover, the phosphorylation status of VP30 did not cause major differences in nucleocapsid transport in authentic EBOV-infected cells. In the following budding step, the association of VP30 and its phosphorylation status did not influence the budding efficiency of virus-like particles. Taken together, it is plausible that EBOV may utilize the phosphorylation of VP30 for its selective association with nucleocapsids, without affecting nucleocapsid transport and virion budding processes. IMPORTANCE Ebola virus (EBOV) causes severe fevers with unusually high case fatality rates. The nucleocapsid provides the template for viral genome transcription and replication. Thus, understanding the regulatory mechanism behind its formation is important for the development of novel therapeutic approaches. Previously, we established a live-cell imaging system based on the ectopic expression of viral fluorescent fusion proteins, allowing the visualization and characterization of intracytoplasmic transport of nucleocapsid-like structures. EBOV VP30 is an essential transcriptional factor for viral genome synthesis, and, although its role in viral genome transcription and replication is well understood, the functional importance of VP30 phosphorylation in assembly of nucleocapsids is still unclear. Our work determines the localization of VP30 at the surface of ruffled nucleocapsids, which differs from the localization of polymerase in EBOV-infected cells. This study sheds light on the novel role of VP30 phosphorylation in nucleocapsid assembly, which is an important prerequisite for virion formation.


Subject(s)
Ebolavirus , Nucleocapsid , Transcription Factors , Viral Proteins , Virus Assembly , Biological Transport , Ebolavirus/chemistry , Ebolavirus/growth & development , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/virology , Humans , Nucleocapsid/biosynthesis , Nucleocapsid/metabolism , Phosphorylation , Transcription Factors/chemistry , Transcription Factors/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Virion/chemistry , Virion/growth & development , Virion/metabolism
6.
Antiviral Res ; 199: 105267, 2022 03.
Article in English | MEDLINE | ID: mdl-35227759

ABSTRACT

The central role of Ebola virus (EBOV) VP40 in nascent virion assembly and budding from infected host cells makes it an important therapeutic target. The mechanism of dimerization, following oligomerization of VP40 leading to the production of virus-like particles (VLP) has never been investigated for the development of therapeutic candidates against Ebola disease. Molecular dynamics-based computational screening targeted VP40 dimer with 40,000,000 compounds selected 374 compounds. A novel in vitro screening assay selected two compounds, NUSU#1 and NUSU#2. Conventional VLP assays consistently showed that both compounds inhibited EBOV VP40-mediated VLP production. Intriguingly, NUSU#1 inhibited the VP40-mediated VLP production in other ebolavirus species and the Marburg virus, but did not inhibit Lassa virus Z-mediated VLP production. These results strongly suggested that the selected compounds are potential lead drug candidates against Filovirus disease via disruption of VP40-mediated particle production.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Marburgvirus , Ebolavirus/chemistry , Humans , Viral Matrix Proteins/chemistry , Virus Release
7.
STAR Protoc ; 3(1): 101030, 2022 03 18.
Article in English | MEDLINE | ID: mdl-34977676

ABSTRACT

This protocol describes the reconstitution of the filamentous Ebola virus nucleocapsid-like assembly in vitro. This is followed by solving the cryo-EM structure using helical reconstruction, and flexible fitting of the existing model into the 5.8 Å cryo-EM map. The protocol can be applied to other filamentous viral protein assemblies, particularly those with high flexibility and moderate resolution maps, which present technical challenges to model building. For complete details on the use and execution of this profile, please refer to Su et al. (2018).


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Cryoelectron Microscopy/methods , Ebolavirus/chemistry , Humans , Nucleocapsid/chemistry , Virus Assembly
8.
Viruses ; 13(9)2021 09 08.
Article in English | MEDLINE | ID: mdl-34578374

ABSTRACT

Ebola virus (EBOV), one of the most infectious human viruses and a leading cause of viral hemorrhagic fever, imposes a potential public health threat with several recent outbreaks. Despite the difficulties associated with working with this pathogen in biosafety level-4 containment, a protective vaccine and antiviral therapeutic were recently approved. However, the high mortality rate of EBOV infection underscores the necessity to continuously identify novel antiviral strategies to help expand the scope of prophylaxis/therapeutic management against future outbreaks. This includes identifying antiviral agents that target EBOV entry, which could improve the management of EBOV infection. Herein, using EBOV glycoprotein (GP)-pseudotyped particles, we screened a panel of natural medicinal extracts, and identified the methanolic extract of Perilla frutescens (PFME) as a robust inhibitor of EBOV entry. We show that PFME dose-dependently impeded EBOV GP-mediated infection at non-cytotoxic concentrations, and exerted the most significant antiviral activity when both the extract and the pseudoparticles are concurrently present on the host cells. Specifically, we demonstrate that PFME could block viral attachment and neutralize the cell-free viral particles. Our results, therefore, identified PFME as a potent inhibitor of EBOV entry, which merits further evaluation for development as a therapeutic strategy against EBOV infection.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Ebolavirus/physiology , Perilla frutescens/chemistry , Plant Extracts/pharmacology , Viral Envelope Proteins , Virus Internalization/drug effects , Ebolavirus/chemistry , Ebolavirus/genetics , HEK293 Cells , Humans , Methanol/chemistry , Methanol/pharmacology , Plant Extracts/chemistry , Viral Envelope Proteins/genetics
9.
Viruses ; 13(8)2021 08 19.
Article in English | MEDLINE | ID: mdl-34452514

ABSTRACT

Viral interferon (IFN) antagonist proteins mediate evasion of IFN-mediated innate immunity and are often multifunctional, with distinct roles in viral replication. The Ebola virus IFN antagonist VP24 mediates nucleocapsid assembly, and inhibits IFN-activated signaling by preventing nuclear import of STAT1 via competitive binding to nuclear import receptors (karyopherins). Proteins of many viruses, including viruses with cytoplasmic replication cycles, interact with nuclear trafficking machinery to undergo nucleocytoplasmic transport, with key roles in pathogenesis; however, despite established karyopherin interaction, potential nuclear trafficking of VP24 has not been investigated. We find that inhibition of nuclear export pathways or overexpression of VP24-binding karyopherin results in nuclear localization of VP24. Molecular mapping indicates that cytoplasmic localization of VP24 depends on a CRM1-dependent nuclear export sequence at the VP24 C-terminus. Nuclear export is not required for STAT1 antagonism, consistent with competitive karyopherin binding being the principal antagonistic mechanism, while export mediates return of nuclear VP24 to the cytoplasm where replication/nucleocapsid assembly occurs.


Subject(s)
Cell Nucleus/virology , Cytoplasm/virology , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/virology , Interferon Type I/metabolism , Viral Proteins/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Cytoplasm/metabolism , Ebolavirus/chemistry , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/metabolism , Host-Pathogen Interactions , Humans , Interferon Type I/genetics , Nuclear Localization Signals , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics
10.
mBio ; 12(4): e0097221, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34225493

ABSTRACT

Ebola virus (EBOV) VP24 protein is a nucleocapsid-associated protein that inhibits interferon (IFN) gene expression and counteracts the IFN-mediated antiviral response, preventing nuclear import of signal transducer and activator of transcription 1 (STAT1). Proteomic studies to identify additional EBOV VP24 partners have pointed to the nuclear membrane component emerin as a potential element of the VP24 cellular interactome. Here, we have further studied this interaction and its impact on cell biology. We demonstrate that VP24 interacts with emerin but also with other components of the inner nuclear membrane, such as lamin A/C and lamin B. We also show that VP24 diminishes the interaction between emerin and lamin A/C and compromises the integrity of the nuclear membrane. This disruption is associated with nuclear morphological abnormalities, activation of a DNA damage response, the phosphorylation of extracellular signal-regulated kinase (ERK), and the induction of interferon-stimulated gene 15 (ISG15). Interestingly, expression of VP24 also promoted the cytoplasmic translocation and downmodulation of barrier-to-autointegration factor (BAF), a common interactor of lamin A/C and emerin, leading to repression of the BAF-regulated CSF1 gene. Importantly, we found that EBOV infection results in the activation of pathways associated with nuclear envelope damage, consistent with our observations in cells expressing VP24. In summary, here we demonstrate that VP24 acts at the nuclear membrane, causing morphological and functional changes in cells that recapitulate several of the hallmarks of laminopathy diseases. IMPORTANCE The Ebola virus (EBOV) VP24 protein is a nucleocapsid-associated protein with multiple functions. Proteomic studies have identified the cellular nuclear membrane component emerin as a potential VP24 interactor. Here, we demonstrate that VP24 not only interacts with emerin but also with lamin A/C and lamin B, prompting nuclear membrane disruption. This disruption is associated with nuclear morphological abnormalities, activation of a DNA damage response, the phosphorylation of extracellular signal-regulated kinase (ERK), and the induction of interferon-stimulated gene 15 (ISG15). Interestingly, VP24 also promotes the cytoplasmic translocation and downmodulation of barrier-to-autointegration factor (BAF), leading to repression of the BAF-regulated CSF1 gene. Finally, we show that EBOV infection also results in the activation of pathways associated with nuclear envelope damage, consistent with our observations in cells expressing VP24. These results reveal novel activities of EBOV VP24 protein, resulting in a cell phenotype similar to that of most laminopathies, with potential impact on EBOV replication.


Subject(s)
Ebolavirus/pathogenicity , Laminopathies/virology , Lamins/metabolism , Nuclear Envelope/pathology , Viral Proteins/genetics , A549 Cells , Active Transport, Cell Nucleus , Cell Nucleus/pathology , Cell Nucleus/virology , Ebolavirus/chemistry , Ebolavirus/genetics , HEK293 Cells , HeLa Cells , Hemorrhagic Fever, Ebola/virology , Humans , Lamins/classification , Membrane Proteins/metabolism , Nuclear Envelope/virology , Nuclear Proteins/metabolism , Phenotype , Viral Proteins/metabolism , Virus Replication
11.
J Struct Biol ; 213(2): 107742, 2021 06.
Article in English | MEDLINE | ID: mdl-33971285

ABSTRACT

Cryo-electron tomography (cryo-ET) is a pivotal imaging technique for studying the structure of pleomorphic enveloped viruses and their interactions with the host at native conditions. Owing to the limited tilting range of samples with a slab geometry, electron tomograms suffer from so-called missing wedge information in Fourier space. In dual-axis cryo-ET, two tomograms reconstructed from orthogonally oriented tilt series are combined into a tomogram with improved resolution as the missing wedge information is reduced to a pyramid. Volta phase plate (VPP) allows to perform in-focus cryo-ET with high contrast transfer at low-resolution frequencies and thus its application may improve the quality of dual-axis tomograms. Here, we compare dual-axis cryo-ET with and without VPP on Ebola virus-like particles to visualize and segment viral and host cell proteins within the membrane-enveloped filamentous particles. Dual-axis VPP cryo-ET reduces the missing wedge information and ray artifacts arising from the weighted back-projection during tomogram reconstruction, thereby minimizing ambiguity in the analysis of crowded environments and facilitating 3D segmentation. We show that dual-axis VPP tomograms provide a comprehensive description of macromolecular organizations such as nucleocapsid assembly states, the distribution of glycoproteins on the viral envelope and asymmetric arrangements of the VP40 layer in non-filamentous regions of virus-like particles. Our data reveal actin filaments within virus-like particles in close proximity to the viral VP40 scaffold, suggesting a direct interaction between VP40 and actin filaments. Dual-axis VPP cryo-ET provides more complete 3D information at high contrast and allows for better interpretation of macromolecule interactions and pleomorphic organizations.


Subject(s)
Actins/chemistry , Cryoelectron Microscopy/methods , Ebolavirus/chemistry , Viral Matrix Proteins/chemistry , Actins/metabolism , Cell Membrane , Ebolavirus/metabolism , Ebolavirus/ultrastructure , Electron Microscope Tomography/methods , HEK293 Cells , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Host-Pathogen Interactions , Humans , Imaging, Three-Dimensional , Nucleocapsid/chemistry , Viral Matrix Proteins/metabolism
12.
Viruses ; 13(2)2021 02 12.
Article in English | MEDLINE | ID: mdl-33673144

ABSTRACT

Filoviruses Ebola (EBOV) and Marburg (MARV) are devastating high-priority pathogens capable of causing explosive outbreaks with high human mortality rates. The matrix proteins of EBOV and MARV, as well as eVP40 and mVP40, respectively, are the key viral proteins that drive virus assembly and egress and can bud independently from cells in the form of virus-like particles (VLPs). The matrix proteins utilize proline-rich Late (L) domain motifs (e.g., PPxY) to hijack specific host proteins that contain WW domains, such as the HECT family E3 ligases, to facilitate the last step of virus-cell separation. We identified E3 ubiquitin ligase Smad Ubiquitin Regulatory Factor 2 (SMURF2) as a novel interactor with VP40 that positively regulates VP40 VLP release. Our results show that eVP40 and mVP40 interact with the three WW domains of SMURF2 via their PPxY motifs. We provide evidence that the eVP40-SMURF2 interaction is functional as the expression of SMURF2 positively regulates VLP egress, while siRNA knockdown of endogenous SMURF2 decreases VLP budding compared to controls. In sum, our identification of novel interactor SMURF2 adds to the growing list of identified host proteins that can regulate PPxY-mediated egress of VP40 VLPs. A more comprehensive understanding of the modular interplay between filovirus VP40 and host proteins may lead to the development of new therapies to combat these deadly infections.


Subject(s)
Ebolavirus/physiology , Hemorrhagic Fever, Ebola/enzymology , Marburg Virus Disease/enzymology , Marburgvirus/physiology , Ubiquitin-Protein Ligases/metabolism , Viral Matrix Proteins/metabolism , Virus Release , Amino Acid Motifs , Animals , Ebolavirus/chemistry , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Humans , Marburg Virus Disease/genetics , Marburg Virus Disease/virology , Marburgvirus/chemistry , Marburgvirus/genetics , Protein Binding , Ubiquitin-Protein Ligases/genetics , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Virion/genetics , Virion/physiology , Virus Assembly
13.
J Microbiol ; 59(4): 410-416, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33630249

ABSTRACT

Zaire ebolavirus, commonly called Ebola virus (EBOV), is an RNA virus that causes severe hemorrhagic fever with high mortality. Viral protein 35 (VP35) is a virulence factor encoded in the EBOV genome. VP35 inhibits host innate immune responses and functions as a critical cofactor for viral RNA replication. EBOV VP35 contains a short conserved motif that interacts with dynein light chain 8 (LC8), which serves as a regulatory hub protein by associating with various LC8-binding proteins. Herein, we present the crystal structure of human LC8 bound to the peptide comprising residues 67-76 of EBOV VP35. Two VP35 peptides were found to interact with homodimeric LC8 by extending the central ß-sheets, constituting a 2:2 complex. Structural analysis demonstrated that the intermolecular binding between LC8 and VP35 is mainly sustained by a network of hydrogen bonds and supported by hydrophobic interactions in which Thr73 and Thr75 of VP35 are involved. These findings were verified by binding measurements using isothermal titration calorimetry. Biochemical analyses also verified that residues 67-76 of EBOV VP35 constitute a core region for interaction with LC8. In addition, corresponding motifs from other members of the genus Ebolavirus commonly bound to LC8 but with different binding affinities. Particularly, VP35 peptides originating from pathogenic species interacted with LC8 with higher affinity than those from noninfectious species, suggesting that the binding of VP35 to LC8 is associated with the pathogenicity of the Ebolavirus species.


Subject(s)
Cytoplasmic Dyneins/chemistry , Ebolavirus/chemistry , Nucleocapsid Proteins/chemistry , Amino Acid Sequence , Calorimetry , Computer Simulation , Crystallization , Crystallography, X-Ray , Hemorrhagic Fever, Ebola/virology , Host Microbial Interactions , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Multimerization , Viral Proteins/chemistry , Virulence Factors/chemistry
14.
mBio ; 12(1)2021 01 12.
Article in English | MEDLINE | ID: mdl-33436438

ABSTRACT

Ebola virus (EBOV) entry into host cells comprises stepwise and extensive interactions of the sole viral surface glycoprotein (GP) with multiple host factors. During the intricate process, following virus uptake and trafficking to late endosomal/lysosomal compartments, GP is proteolytically processed to cleaved GP (GPCL) by the endosomal proteases cathepsin B and L, unmasking GP's receptor-binding site. Engagement of GPCL with the universal filoviral intracellular receptor Niemann-Pick C1 (NPC1) eventually culminates in fusion between viral and cellular membranes, cytoplasmic escape of the viral nucleocapsid, and subsequent infection. Mechanistic delineation of the indispensable GPCL-NPC1-binding step has been severely hampered by the unavailability of a robust cell-based assay assessing interaction of GPCL with full-length endosomal NPC1. Here, we describe a novel in situ assay to monitor GPCL-NPC1 engagement in intact, infected cells. Visualization of the subcellular localization of binding complexes is based on the principle of DNA-assisted, antibody-mediated proximity ligation. Virus-receptor binding monitored by proximity ligation was contingent on GP's proteolytic cleavage and was sensitive to perturbations in the GPCL-NPC1 interface. Our assay also specifically decoupled detection of virus-receptor binding from steps post-receptor binding, such as membrane fusion and infection. Testing of multiple FDA-approved small-molecule inhibitors revealed that drug treatments inhibited virus entry and GPCL-NPC1 recognition by distinctive mechanisms. Together, here we present a newly established proximity ligation assay, which will allow us to dissect cellular and viral requirements for filovirus-receptor binding and to delineate the mechanisms of action of inhibitors on filovirus entry in a cell-based system.IMPORTANCE Ebola virus causes episodic but increasingly frequent outbreaks of severe disease in Middle Africa, as shown by the recently overcome second largest outbreak on record in the Democratic Republic of Congo. Despite considerable effort, FDA-approved anti-filoviral therapeutics or targeted interventions are not available yet. Virus host-cell invasion represents an attractive target for antivirals; however, our understanding of the inhibitory mechanisms of novel therapeutics is often hampered by fragmented knowledge of the filovirus-host molecular interactions required for viral infection. To help close this critical knowledge gap, here, we report an in situ assay to monitor binding of the EBOV glycoprotein to its receptor NPC1 in intact, infected cells. We demonstrate that our in situ assay based on proximity ligation represents a powerful tool to delineate receptor-viral glycoprotein interactions. Similar assays can be utilized to examine receptor interactions of diverse viral surface proteins whose studies have been hampered until now by the lack of robust in situ assays.


Subject(s)
Ebolavirus/chemistry , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Binding Sites , Cell Line , Endosomes/metabolism , Gene Knockout Techniques , Glycoproteins , Humans , Lysosomes/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Niemann-Pick C1 Protein , Protein Binding , Protein Domains , Protein Transport , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virion , Virus Internalization
15.
RNA Biol ; 18(4): 523-536, 2021 04.
Article in English | MEDLINE | ID: mdl-32882148

ABSTRACT

Ebola virus (EBOV) RNA has the potential to form hairpin structures at the transcription start sequence (TSS) and reinitiation sites of internal genes, both on the genomic and antigenomic/mRNA level. Hairpin formation involving the TSS and the spacer sequence between promotor elements (PE) 1 and 2 was suggested to regulate viral transcription. Here, we provide evidence that such RNA structures form during RNA synthesis by the viral polymerase and affect its activity. This was analysed using monocistronic minigenomes carrying hairpin structure variants in the TSS-spacer region that differ in length and stability. Transcription and replication were measured via reporter activity and by qRT-PCR quantification of the distinct viral RNA species. We demonstrate that viral RNA synthesis is remarkably tolerant to spacer extensions of up to ~54 nt, but declines beyond this length limit (~25% residual activity for a 66-nt extension). Minor incremental stabilizations of hairpin structures in the TSS-spacer region and on the mRNA/antigenomic level were found to rapidly abolish viral polymerase activity, which may be exploited for antisense strategies to inhibit viral RNA synthesis. Finally, balanced viral transcription and replication can still occur when any RNA structure formation potential at the TSS is eliminated, provided that hexamer phasing in the promoter region is maintained. Altogether, the findings deepen and refine our insight into structure and length constraints within the EBOV transcription and replication promoter and suggest a remarkable flexibility of the viral polymerase in recognition of PE1 and PE2.


Subject(s)
Ebolavirus/genetics , RNA Stability/genetics , RNA, Viral/chemistry , Virus Replication/genetics , Ebolavirus/chemistry , Ebolavirus/physiology , Genome, Viral/physiology , HEK293 Cells , Hemorrhagic Fever, Ebola/virology , Humans , Nucleic Acid Conformation , Promoter Regions, Genetic , RNA, Viral/genetics , Transcription Initiation Site , Transcription, Genetic
16.
J Chem Phys ; 153(15): 155102, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33092380

ABSTRACT

Ebola virus (EBOV) is a human pathogen with the ability to cause hemorrhagic fever and bleeding diathesis in hosts. The life cycle of EBOV depends on its nucleocapsid. The Ebola nucleocapsid consists of a helical assembly of nucleoproteins (NPs) encapsidating single-stranded viral RNA (ssRNA). Knowledge of the molecular determinants of Ebola nucleocapsid stability is essential for the development of therapeutics against EBOV. However, large degrees of freedom associated with the Ebola nucleocapsid helical assembly pose a computational challenge, thereby limiting the previous simulation studies to the level of monomers. In the present work, we have performed all atom molecular dynamics (MD) simulations of the helical assembly of EBOV nucleoproteins in the absence and presence of ssRNA. We found that ssRNA is essential for maintaining structural integrity of the nucleocapsid. Other molecular determinants observed to stabilize the nucleocapsid include NP-RNA and NP-NP interactions and ion distributions. Additionally, the structural and dynamical behavior of the nucleocapsid monomer depends on its position in the helical assembly. NP monomers present on the longitudinal edges of the helical tube are more exposed, flexible, and have weaker NP-NP interactions than those residing in the center. This work provides key structural features stabilizing the nucleocapsid that may serve as therapeutic targets.


Subject(s)
Ebolavirus/chemistry , Molecular Dynamics Simulation , Nucleocapsid/analysis , Humans
17.
Elife ; 92020 10 05.
Article in English | MEDLINE | ID: mdl-33016878

ABSTRACT

Filoviruses such as Ebola and Marburg virus bud from the host membrane as enveloped virions. This process is achieved by the matrix protein VP40. When expressed alone, VP40 induces budding of filamentous virus-like particles, suggesting that localization to the plasma membrane, oligomerization into a matrix layer, and generation of membrane curvature are intrinsic properties of VP40. There has been no direct information on the structure of VP40 matrix layers within viruses or virus-like particles. We present structures of Ebola and Marburg VP40 matrix layers in intact virus-like particles, and within intact Marburg viruses. VP40 dimers assemble extended chains via C-terminal domain interactions. These chains stack to form 2D matrix lattices below the membrane surface. These lattices form a patchwork assembly across the membrane and suggesting that assembly may begin at multiple points. Our observations define the structure and arrangement of the matrix protein layer that mediates formation of filovirus particles.


Subject(s)
Ebolavirus/physiology , Marburgvirus/physiology , Protein Multimerization , Viral Matrix Proteins/chemistry , Cell Membrane/physiology , Ebolavirus/chemistry , Marburgvirus/chemistry
18.
mBio ; 11(5)2020 09 15.
Article in English | MEDLINE | ID: mdl-32934085

ABSTRACT

Membrane-associated RING-CH-type 8 (MARCH8) strongly blocks human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env) incorporation into virions by downregulating its cell surface expression, but the mechanism is still unclear. We now report that MARCH8 also blocks the Ebola virus (EBOV) glycoprotein (GP) incorporation via surface downregulation. To understand how these viral fusion proteins are downregulated, we investigated the effects of MARCH8 on EBOV GP maturation and externalization via the conventional secretion pathway. MARCH8 interacted with EBOV GP and furin when detected by immunoprecipitation and retained the GP/furin complex in the Golgi when their location was tracked by a bimolecular fluorescence complementation (BiFC) assay. MARCH8 did not reduce the GP expression or affect the GP modification by high-mannose N-glycans in the endoplasmic reticulum (ER), but it inhibited the formation of complex N-glycans on the GP in the Golgi. Additionally, the GP O-glycosylation and furin-mediated proteolytic cleavage were also inhibited. Moreover, we identified a novel furin cleavage site on EBOV GP and found that only those fully glycosylated GPs were processed by furin and incorporated into virions. Furthermore, the GP shedding and secretion were all blocked by MARCH8. MARCH8 also blocked the furin-mediated cleavage of HIV-1 Env (gp160) and the highly pathogenic avian influenza virus H5N1 hemagglutinin (HA). We conclude that MARCH8 has a very broad antiviral activity by prohibiting different viral fusion proteins from glycosylation and proteolytic cleavage in the Golgi, which inhibits their transport from the Golgi to the plasma membrane and incorporation into virions.IMPORTANCE Enveloped viruses express three classes of fusion proteins that are required for their entry into host cells via mediating virus and cell membrane fusion. Class I fusion proteins are produced from influenza viruses, retroviruses, Ebola viruses, and coronaviruses. They are first synthesized as a type I transmembrane polypeptide precursor that is subsequently glycosylated and oligomerized. Most of these precursors are cleaved en route to the plasma membrane by a cellular protease furin in the late secretory pathway, generating the trimeric N-terminal receptor-binding and C-terminal fusion subunits. Here, we show that a cellular protein, MARCH8, specifically inhibits the furin-mediated cleavage of EBOV GP, HIV-1 Env, and H5N1 HA. Further analyses uncovered that MARCH8 blocked the EBOV GP glycosylation in the Golgi and inhibited its transport from the Golgi to the plasma membrane. Thus, MARCH8 has a very broad antiviral activity by specifically inactivating different viral fusion proteins.


Subject(s)
Ebolavirus/chemistry , Glycoproteins/antagonists & inhibitors , HIV-1/chemistry , Hemagglutinins, Viral/metabolism , Influenza A Virus, H5N1 Subtype/chemistry , Ubiquitin-Protein Ligases/genetics , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/physiology , Animals , Cell Line , Chlorocebus aethiops , Ebolavirus/physiology , Glycosylation , HEK293 Cells , HIV-1/physiology , HeLa Cells , Hep G2 Cells , Humans , Influenza A Virus, H5N1 Subtype/physiology , Protein Binding , THP-1 Cells , Ubiquitin-Protein Ligases/metabolism , Vero Cells , Viral Fusion Proteins/antagonists & inhibitors , Viral Fusion Proteins/metabolism
19.
Infect Genet Evol ; 85: 104464, 2020 11.
Article in English | MEDLINE | ID: mdl-32681997

ABSTRACT

Ebola virus (EBOV) is a rare but fatal disease that has been a burden to mankind for over 40 years. EBOV exhibits several symptoms including severe bleeding, organ failure and if left untreated causes death. It is assumed that fruit bats of the Pteropodidae family are natural hosts for the virus. Over the years, there has been no effective vaccine that can confer immunity to this virus. Considering the necessity of a vaccine against EBOV, this study to develop a multi-epitope subunit vaccine for the EBOV using the immunoinformatics approach was conducted. The construct was designed using structural and non-structural proteins of EBOV. Class I and Class II MHC epitopes were predicted and linked along with ß defensin and compatible linkers. B-cell linear epitopes were also assessed and the physiological parameters of the vaccine were determined. The vaccine was capable of administration to humans and also is capable of an immune response. The vaccine was modeled further and affinity towards the TLR4 receptor was studied by docking and simulation for 20 ns. The trajectory analysis high affinity between the vaccine and the construct with an average hydrogen bond of 18. For ease of purification, the vaccine construct was ligated into pET28a(+) vector with His-tag. Concluding from the results, the vaccine construct has the potentiality to help develop immunity against the Ebola virus. Furthermore, experimental and immunological investigations will be required to verify the feasibility of the multi-epitope subunit construct as a commercial vaccine.


Subject(s)
Ebolavirus/chemistry , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Viral Vaccines/chemistry , Viral Vaccines/immunology , Amino Acid Sequence , Computational Biology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Humans , Immunogenicity, Vaccine , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/immunology , Vaccines, Subunit
20.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32611759

ABSTRACT

Ebola virus (EBOV) entry into cells is mediated by its spike glycoprotein (GP). Following attachment and internalization, virions traffic to late endosomes where GP is cleaved by host cysteine proteases. Cleaved GP then binds its cellular receptor, Niemann-Pick C1. In response to an unknown cellular trigger, GP undergoes conformational rearrangements that drive fusion of viral and endosomal membranes. The temperature-dependent stability (thermostability) of the prefusion conformers of class I viral fusion glycoproteins, including those of filovirus GPs, has provided insights into their propensity to undergo fusion-related rearrangements. However, previously described assays have relied on soluble glycoprotein ectodomains. Here, we developed a simple enzyme-linked immunosorbent assay (ELISA)-based assay that uses the temperature-dependent loss of conformational epitopes to measure thermostability of GP embedded in viral membranes. The base and glycan cap subdomains of all filovirus GPs tested suffered a concerted loss of prefusion conformation at elevated temperatures but did so at different temperature ranges, indicating virus-specific differences in thermostability. Despite these differences, all of these GPs displayed reduced thermostability upon cleavage to GP conformers (GPCL). Surprisingly, acid pH enhanced, rather than decreased, GP thermostability, suggesting it could enhance viral survival in hostile endo/lysosomal compartments. Finally, we confirmed and extended previous findings that some small-molecule inhibitors of filovirus entry destabilize EBOV GP and uncovered evidence that the most potent inhibitors act through multiple mechanisms. We establish the epitope-loss ELISA as a useful tool for studies of filovirus entry, engineering of GP variants with enhanced stability for use in vaccine development, and discovery of new stability-modulating antivirals.IMPORTANCE The development of Ebola virus countermeasures is challenged by our limited understanding of cell entry, especially at the step of membrane fusion. The surface-exposed viral protein, GP, mediates membrane fusion and undergoes major structural rearrangements during this process. The stability of GP at elevated temperatures (thermostability) can provide insights into its capacity to undergo these rearrangements. Here, we describe a new assay that uses GP-specific antibodies to measure GP thermostability under a variety of conditions relevant to viral entry. We show that proteolytic cleavage and acid pH have significant effects on GP thermostability that shed light on their respective roles in viral entry. We also show that the assay can be used to study how small-molecule entry inhibitors affect GP stability. This work provides a simple and readily accessible assay to engineer stabilized GP variants for antiviral vaccines and to discover and improve drugs that act by modulating GP stability.


Subject(s)
Ebolavirus/drug effects , Niemann-Pick C1 Protein/antagonists & inhibitors , Receptors, Virus/antagonists & inhibitors , Viral Envelope Proteins/antagonists & inhibitors , Viral Fusion Proteins/antagonists & inhibitors , Virion/drug effects , Animals , Binding Sites , Biological Assay , Chlorocebus aethiops , Clomiphene/chemistry , Clomiphene/pharmacology , Ebolavirus/chemistry , Ebolavirus/genetics , Ebolavirus/metabolism , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Hot Temperature , Hydrogen-Ion Concentration , Molecular Docking Simulation , Niemann-Pick C1 Protein/chemistry , Niemann-Pick C1 Protein/genetics , Niemann-Pick C1 Protein/metabolism , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Tertiary , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/chemistry , Tamoxifen/pharmacology , Toremifene/chemistry , Toremifene/pharmacology , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virion/chemistry , Virion/genetics , Virion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...