Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 336
Filter
1.
Nat Commun ; 15(1): 8479, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39353909

ABSTRACT

The majority of viruses classified as pandemic threats are enveloped viruses which enter the cell through receptor-mediated endocytosis and take advantage of endosomal acidification to activate their fusion machinery. Here we report that the endosomal fusion of low pH-requiring viruses is highly dependent on TRPM7, a widely expressed TRP channel that is located on the plasma membrane and in intracellular vesicles. Using several viral infection systems expressing the envelope glycoproteins of various viruses, we find that loss of TRPM7 protects cells from infection by Lassa, LCMV, Ebola, Influenza, MERS, SARS-CoV-1, and SARS-CoV-2. TRPM7 ion channel activity is intrinsically necessary to acidify virus-laden endosomes but is expendable for several other endosomal acidification pathways. We propose a model wherein TRPM7 ion channel activity provides a countercurrent of cations from endosomal lumen to cytosol necessary to sustain the pumping of protons into these virus-laden endosomes. This study demonstrates the possibility of developing a broad-spectrum, TRPM7-targeting antiviral drug to subvert the endosomal fusion of low pH-dependent enveloped viruses.


Subject(s)
Endosomes , TRPM Cation Channels , Virus Internalization , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Endosomes/metabolism , Endosomes/virology , Hydrogen-Ion Concentration , Humans , Animals , HEK293 Cells , SARS-CoV-2/physiology , SARS-CoV-2/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Ebolavirus/physiology , Ebolavirus/metabolism , Lymphocytic choriomeningitis virus/physiology , Chlorocebus aethiops , Viral Envelope/metabolism , Lassa virus/metabolism , Lassa virus/physiology
2.
Cell ; 187(20): 5587-5603.e19, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39293445

ABSTRACT

Filoviruses, including the Ebola and Marburg viruses, cause hemorrhagic fevers with up to 90% lethality. The viral nucleocapsid is assembled by polymerization of the nucleoprotein (NP) along the viral genome, together with the viral proteins VP24 and VP35. We employed cryo-electron tomography of cells transfected with viral proteins and infected with model Ebola virus to illuminate assembly intermediates, as well as a 9 Å map of the complete intracellular assembly. This structure reveals a previously unresolved third and outer layer of NP complexed with VP35. The intrinsically disordered region, together with the C-terminal domain of this outer layer of NP, provides the constant width between intracellular nucleocapsid bundles and likely functions as a flexible tether to the viral matrix protein in the virion. A comparison of intracellular nucleocapsids with prior in-virion nucleocapsid structures reveals that the nucleocapsid further condenses vertically in the virion. The interfaces responsible for nucleocapsid assembly are highly conserved and offer targets for broadly effective antivirals.


Subject(s)
Ebolavirus , Electron Microscope Tomography , Nucleocapsid , Virus Assembly , Ebolavirus/ultrastructure , Ebolavirus/chemistry , Ebolavirus/metabolism , Ebolavirus/physiology , Nucleocapsid/metabolism , Nucleocapsid/ultrastructure , Nucleocapsid/chemistry , Humans , Cryoelectron Microscopy/methods , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/ultrastructure , Nucleoproteins/chemistry , Nucleoproteins/metabolism , Nucleoproteins/ultrastructure , Animals , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/ultrastructure , Models, Molecular , Virion/ultrastructure , Virion/metabolism , Hemorrhagic Fever, Ebola/virology , Chlorocebus aethiops
3.
PLoS Pathog ; 20(8): e1012444, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39173055

ABSTRACT

The Ebola virus (EBOV) has emerged as a significant global health concern, notably during the 2013-2016 outbreak in West Africa. Despite the clinical approval of two EBOV antibody drugs, there is an urgent need for more diverse and effective antiviral drugs, along with comprehensive understanding of viral-host interactions. In this study, we harnessed a biologically contained EBOVΔVP30-EGFP cell culture model which could recapitulate the entire viral life cycle, to conduct a genome-wide CRISPR/Cas9 screen. Through this, we identified PIK3C3 (phosphatidylinositide 3-kinase) and SLC39A9 (zinc transporter) as crucial host factors for EBOV infection. Genetic depletion of SLC39A9 and PIK3C3 lead to reduction of EBOV entry, but not impact viral genome replication, suggesting that SLC39A9 and PIK3C3 act as entry factors, facilitating viral entry into host cells. Moreover, PIK3C3 kinase activity is indispensable for the internalization of EBOV virions, presumably through the regulation of endocytic and autophagic membrane traffic, which has been previously recognized as essential for EBOV internalization. Notably, our study demonstrated that PIK3C3 kinase inhibitor could effectively block EBOV infection, underscoring PIK3C3 as a promising drug target. Furthermore, biochemical analysis showed that recombinant SLC39A9 protein could directly bind viral GP protein, which further promotes the interaction of viral GP protein with cellular receptor NPC1. These findings suggests that SLC39A9 plays dual roles in EBOV entry. Initially, it serves as an attachment factor during the early entry phase by engaging with the viral GP protein. Subsequently, SLC39A9 functions an adaptor protein, facilitating the interaction between virions and the NPC1 receptor during the late entry phase, prior to cathepsin cleavage on the viral GP. In summary, this study offers novel insights into virus-host interactions, contributing valuable information for the development of new therapies against EBOV infection.


Subject(s)
CRISPR-Cas Systems , Ebolavirus , Hemorrhagic Fever, Ebola , Virus Internalization , Animals , Humans , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Class III Phosphatidylinositol 3-Kinases/genetics , Ebolavirus/genetics , Ebolavirus/physiology , Ebolavirus/metabolism , HEK293 Cells , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/genetics , Virus Replication
4.
Nat Commun ; 15(1): 6953, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138205

ABSTRACT

Filovirus-host interactions play important roles in all stages of the virus lifecycle. Here, we identify LATS1/2 kinases and YAP, key components of the Hippo pathway, as critical regulators of EBOV transcription and egress. Specifically, we find that when YAP is phosphorylated by LATS1/2, it localizes to the cytoplasm (Hippo "ON") where it sequesters VP40 to prevent egress. In contrast, when the Hippo pathway is "OFF", unphosphorylated YAP translocates to the nucleus where it transcriptionally activates host genes and promotes viral egress. Our data reveal that LATS2 indirectly modulates filoviral VP40-mediated egress through phosphorylation of AMOTp130, a positive regulator of viral egress, but more surprisingly that LATS1/2 kinases directly modulate EBOV transcription by phosphorylating VP30, an essential regulator of viral transcription. In sum, our findings highlight the potential to exploit the Hippo pathway/filovirus axis for the development of host-oriented countermeasures targeting EBOV and related filoviruses.


Subject(s)
Ebolavirus , Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , Transcription, Genetic , Virus Release , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Phosphorylation , Ebolavirus/physiology , Ebolavirus/genetics , Ebolavirus/metabolism , HEK293 Cells , Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , YAP-Signaling Proteins/metabolism , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/metabolism , Host-Pathogen Interactions , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics
5.
Virus Res ; 347: 199430, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38964470

ABSTRACT

A multistep priming process involving furin and endosomal cathepsin B and L (CatB/L) has been described for the Orthoebolavirus zairense (EBOV) glycoprotein GP. Inhibition or knockdown of either furin or endosomal cathepsins, however, did not prevent virus multiplication in cell cultures. Moreover, an EBOV mutant lacking the furin cleavage motif (RRTRR→AGTAA) was able to replicate and cause fatal disease in nonhuman primates, indicating that furin cleavage may be dispensable for virus infectivity. Here, by using protease inhibitors and EBOV GP-carrying recombinant vesicular stomatitis virus (VSV) and transcription and replication-competent virus-like particles (trVLPs) we found that processing of EBOV GP is mediated by different proteases in different cell lines depending on the protease repertoire available. Endosomal cathepsins were essential for EBOV GP entry in Huh-7 but not in Vero cells, in which trypsin-like proteases and stably expressed trypsin-like transmembrane serine protease 2 (TMPRSS2) supported wild-type EBOV GP and EBOV GP_AGTAA mutant entry. Furthermore, we show that the EBOV GP_AGTAA mutant is cleaved into fusion-competent GP2 by TMPRSS2 and by CatL at a so far unknown site. Fluorescence microscopy co-localization studies indicate that EBOV GP cleavage by TMPRSS2 may occur in the TGN prior to virus release or in the late endosome at the stage of virus entry into a new cell. Our data show that EBOV GP must be proteolytically activated to support virus entry but has even greater flexibility in terms of proteases and the precise cleavage site than previously assumed.


Subject(s)
Cathepsin L , Ebolavirus , Furin , Serine Endopeptidases , Viral Envelope Proteins , Virus Internalization , Cathepsin L/metabolism , Cathepsin L/genetics , Furin/metabolism , Furin/genetics , Ebolavirus/genetics , Ebolavirus/physiology , Ebolavirus/metabolism , Animals , Humans , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Chlorocebus aethiops , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Proteolysis , Vero Cells , Cell Line , Endosomes/metabolism , Endosomes/virology
6.
PLoS One ; 19(7): e0307579, 2024.
Article in English | MEDLINE | ID: mdl-39052567

ABSTRACT

VP30 and VP40 proteins of Ebola and Marburg viruses have been recognized as potential targets for antiviral drug development due to their essential roles in the viral lifecycle. Targeting these proteins could disrupt key stages of the viral replication process, inhibiting the viruses' ability to propagate and cause disease. The current study aims to perform molecular docking and virtual screening on deep-sea fungal metabolites targeting Marburg virus VP40 Dimer, matrix protein VP40 from Ebola virus Sudan, Ebola VP35 Interferon Inhibitory Domain, and VP35 from Marburg virus. The top ten compounds for each protein target were chosen using the glide score. All the compounds obtained indicate a positive binding interaction. Furthermore, AdmetSAR was utilized to investigate the pharmacokinetics of the inhibitors chosen. Gliotoxin was used as a ligand with Marburg virus VP40 Dimer, Austinol with matrix protein VP40 from Ebola virus Sudan, Ozazino-cyclo-(2,3-dihydroxyl-trp-tyr) with Ebola VP35 Interferon Inhibitory Domain, and Dehydroaustinol with VP35 from Marburg virus. MD modeling and MMPBSA studies were used to provide a better understanding of binding behaviors. Pre-clinical experiments can assist validate our in-silico studies and assess whether the molecule can be employed as an anti-viral drug.


Subject(s)
Antiviral Agents , Ebolavirus , Marburgvirus , Molecular Docking Simulation , Ebolavirus/drug effects , Ebolavirus/metabolism , Marburgvirus/drug effects , Marburgvirus/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/chemistry , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Humans , Computer Simulation , Molecular Dynamics Simulation , Viral Regulatory and Accessory Proteins
7.
J Virol ; 98(6): e0052424, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38757972

ABSTRACT

Ebola virus glycoprotein (EBOV GP) is one of the most heavily O-glycosylated viral glycoproteins, yet we still lack a fundamental understanding of the structure of its large O-glycosylated mucin-like domain and to what degree the host O-glycosylation capacity influences EBOV replication. Using tandem mass spectrometry, we identified 47 O-glycosites on EBOV GP and found similar glycosylation signatures on virus-like particle- and cell lysate-derived GP. Furthermore, we performed quantitative differential O-glycoproteomics on proteins produced in wild-type HEK293 cells and cell lines ablated for the three key initiators of O-linked glycosylation, GalNAc-T1, -T2, and -T3. The data show that 12 out of the 47 O-glycosylated sites were regulated, predominantly by GalNAc-T1. Using the glycoengineered cell lines for authentic EBOV propagation, we demonstrate the importance of O-linked glycan initiation and elongation for the production of viral particles and the titers of progeny virus. The mapped O-glycan positions and structures allowed to generate molecular dynamics simulations probing the largely unknown spatial arrangements of the mucin-like domain. The data highlight targeting GALNT1 or C1GALT1C1 as a possible way to modulate O-glycan density on EBOV GP for novel vaccine designs and tailored intervention approaches.IMPORTANCEEbola virus glycoprotein acquires its extensive glycan shield in the host cell, where it is decorated with N-linked glycans and mucin-type O-linked glycans. The latter is initiated by a family of polypeptide GalNAc-transferases that have different preferences for optimal peptide substrates resulting in a spectrum of both very selective and redundant substrates for each isoform. In this work, we map the exact locations of O-glycans on Ebola virus glycoprotein and identify subsets of sites preferentially initiated by one of the three key isoforms of GalNAc-Ts, demonstrating that each enzyme contributes to the glycan shield integrity. We further show that altering host O-glycosylation capacity has detrimental effects on Ebola virus replication, with both isoform-specific initiation and elongation playing a role. The combined structural and functional data highlight glycoengineered cell lines as useful tools for investigating molecular mechanisms imposed by specific glycans and for steering the immune responses in future vaccine designs.


Subject(s)
Ebolavirus , Polysaccharides , Virus Replication , Ebolavirus/physiology , Ebolavirus/metabolism , Humans , HEK293 Cells , Glycosylation , Polysaccharides/metabolism , Viral Envelope Proteins/metabolism , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/metabolism , N-Acetylgalactosaminyltransferases/metabolism , N-Acetylgalactosaminyltransferases/genetics , Glycoproteins/metabolism , Polypeptide N-acetylgalactosaminyltransferase
8.
Commun Biol ; 7(1): 634, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796621

ABSTRACT

Ebola virus (EBOV) matrix protein VP40 can assemble and bud as virus-like particles (VLPs) when expressed alone in mammalian cells. Nucleoprotein (NP) could be recruited to VLPs as inclusion body (IB) when co-expressed, and increase VLP production. However, the mechanism behind it remains unclear. Here, we use a computational approach to study NP-VP40 interactions. Our simulations indicate that NP may enhance VLP production through stabilizing VP40 filaments and accelerating the VLP budding step. Further, both the relative timing and amount of NP expression compared to VP40 are important for the effective production of IB-containing VLPs. We predict that relative NP/VP40 expression ratio and time are important for efficient production of IB-containing VLPs. We conclude that disrupting the expression timing and amount of NP and VP40 could provide new avenues to treat EBOV infection. This work provides quantitative insights into EBOV proteins interactions and how virion generation and drug efficacy could be influenced.


Subject(s)
Ebolavirus , Viral Core Proteins , Ebolavirus/metabolism , Viral Core Proteins/metabolism , Viral Core Proteins/genetics , Humans , Virion/metabolism , Virion/genetics , Nucleoproteins/metabolism , Nucleoproteins/genetics , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/metabolism
9.
ACS Infect Dis ; 10(5): 1590-1601, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38684073

ABSTRACT

Ebola virus (EBOV) is an enveloped virus that must fuse with the host cell membrane in order to release its genome and initiate infection. This process requires the action of the EBOV envelope glycoprotein (GP), encoded by the virus, which resides in the viral envelope and consists of a receptor binding subunit, GP1, and a membrane fusion subunit, GP2. Despite extensive research, a mechanistic understanding of the viral fusion process is incomplete. To investigate GP-membrane association, a key step in the fusion process, we used two approaches: high-throughput measurements of single-particle diffusion and single-molecule measurements with optical tweezers. Using these methods, we show that the presence of the endosomal Niemann-Pick C1 (NPC1) receptor is not required for primed GP-membrane binding. In addition, we demonstrate this binding is very strong, likely attributed to the interaction between the GP fusion loop and the membrane's hydrophobic core. Our results also align with previously reported findings, emphasizing the significance of acidic pH in the protein-membrane interaction. Beyond Ebola virus research, our approach provides a powerful toolkit for studying other protein-membrane interactions, opening new avenues for a better understanding of protein-mediated membrane fusion events.


Subject(s)
Ebolavirus , Viral Envelope Proteins , Ebolavirus/metabolism , Ebolavirus/physiology , Ebolavirus/genetics , Ebolavirus/chemistry , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Humans , Protein Binding , Virus Internalization , Niemann-Pick C1 Protein/metabolism , Cell Membrane/metabolism , Cell Membrane/virology , Hemorrhagic Fever, Ebola/virology , Hydrogen-Ion Concentration
10.
Protein Sci ; 33(5): e4978, 2024 May.
Article in English | MEDLINE | ID: mdl-38591637

ABSTRACT

The Ebola virus (EBOV) is a lipid-enveloped virus with a negative sense RNA genome that can cause severe and often fatal viral hemorrhagic fever. The assembly and budding of EBOV is regulated by the matrix protein, VP40, which is a peripheral protein that associates with anionic lipids at the inner leaflet of the plasma membrane. VP40 is sufficient to form virus-like particles (VLPs) from cells, which are nearly indistinguishable from authentic virions. Due to the restrictions of studying EBOV in BSL-4 facilities, VP40 has served as a surrogate in cellular studies to examine the EBOV assembly and budding process from the host cell plasma membrane. VP40 is a dimer where inhibition of dimer formation halts budding and formation of new VLPs as well as VP40 localization to the plasma membrane inner leaflet. To better understand VP40 dimer stability and critical amino acids to VP40 dimer formation, we integrated computational approaches with experimental validation. Site saturation/alanine scanning calculation, combined with molecular mechanics-based generalized Born with Poisson-Boltzmann surface area (MM-GB/PBSA) method and molecular dynamics simulations were used to predict the energetic contribution of amino acids to VP40 dimer stability and the hydrogen bonding network across the dimer interface. These studies revealed several previously unknown interactions and critical residues predicted to impact VP40 dimer formation. In vitro and cellular studies were then pursued for a subset of VP40 mutations demonstrating reduction in dimer formation (in vitro) or plasma membrane localization (in cells). Together, the computational and experimental approaches revealed critical residues for VP40 dimer stability in an alpha-helical interface (between residues 106-117) as well as in a loop region (between residues 52-61) below this alpha-helical region. This study sheds light on the structural origins of VP40 dimer formation and may inform the design of a small molecule that can disrupt VP40 dimer stability.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Ebolavirus/genetics , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/metabolism , Cell Membrane/metabolism , Molecular Dynamics Simulation , Amino Acids/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism
11.
J Biol Chem ; 300(5): 107213, 2024 May.
Article in English | MEDLINE | ID: mdl-38522519

ABSTRACT

Ebola virus (EBOV) is a filamentous negative-sense RNA virus, which causes severe hemorrhagic fever. There are limited vaccines or therapeutics for prevention and treatment of EBOV, so it is important to get a detailed understanding of the virus lifecycle to illuminate new drug targets. EBOV encodes for the matrix protein, VP40, which regulates assembly and budding of new virions from the inner leaflet of the host cell plasma membrane (PM). In this work, we determine the effects of VP40 mutations altering electrostatics on PM interactions and subsequent budding. VP40 mutations that modify surface electrostatics affect viral assembly and budding by altering VP40 membrane-binding capabilities. Mutations that increase VP40 net positive charge by one (e.g., Gly to Arg or Asp to Ala) increase VP40 affinity for phosphatidylserine and phosphatidylinositol 4,5-bisphosphate in the host cell PM. This increased affinity enhances PM association and budding efficiency leading to more effective formation of virus-like particles. In contrast, mutations that decrease net positive charge by one (e.g., Gly to Asp) lead to a decrease in assembly and budding because of decreased interactions with the anionic PM. Taken together, our results highlight the sensitivity of slight electrostatic changes on the VP40 surface for assembly and budding. Understanding the effects of single amino acid substitutions on viral budding and assembly will be useful for explaining changes in the infectivity and virulence of different EBOV strains, VP40 variants that occur in nature, and for long-term drug discovery endeavors aimed at EBOV assembly and budding.


Subject(s)
Cell Membrane , Ebolavirus , Virus Assembly , Virus Release , Humans , Amino Acid Substitution , Cell Membrane/metabolism , Ebolavirus/metabolism , Ebolavirus/genetics , HEK293 Cells , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/virology , Mutation , Nucleoproteins , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylserines/metabolism , Phosphatidylserines/chemistry , Protein Binding , Static Electricity , Viral Core Proteins/metabolism , Viral Core Proteins/chemistry , Viral Core Proteins/genetics , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/chemistry , Virion/metabolism , Virion/genetics
12.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(3): 159464, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38360201

ABSTRACT

The Ebola virus matrix protein VP40 is responsible for the formation of the viral matrix by localizing at the inner leaflet of the human plasma membrane (PM). Various lipid types, including PI(4,5)P2 (i.e. PIP2) and phosphatidylserine (PS), play active roles in this process. Specifically, the negatively charged headgroups of both PIP2 and PS interact with the basic residues of VP40 and stabilize it at the membrane surface, allowing for eventual egress. Phosphatidic acid (PA), resulting from the enzyme phospholipase D (PLD), is also known to play an active role in viral development. In this work, we performed a biophysical and computational analysis to investigate the effects of the presence of PA on the membrane localization and association of VP40. We used coarse-grained molecular dynamics simulations to quantify VP40 hexamer interactions with the inner leaflet of the PM. Analysis of the local distribution of lipids shows enhanced lipid clustering when PA is abundant in the membrane. We observed that PA lipids have a similar role to that of PS lipids in VP40 association due to the geometry and charge. Complementary experiments performed in cell culture demonstrate competition between VP40 and a canonical PA-binding protein for the PM. Also, inhibition of PA synthesis reduced the detectable budding of virus-like particles. These computational and experimental results provide new insights into the early stages of Ebola virus budding and the role that PA lipids have on the VP40-PM association.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/metabolism , Cell Membrane/metabolism , Molecular Dynamics Simulation , Lipids/analysis
13.
J Lipid Res ; 65(3): 100512, 2024 03.
Article in English | MEDLINE | ID: mdl-38295986

ABSTRACT

Ebola virus (EBOV) causes severe hemorrhagic fever in humans and is lethal in a large percentage of those infected. The EBOV matrix protein viral protein 40 kDa (VP40) is a peripheral binding protein that forms a shell beneath the lipid bilayer in virions and virus-like particles (VLPs). VP40 is required for virus assembly and budding from the host cell plasma membrane. VP40 is a dimer that can rearrange into oligomers at the plasma membrane interface, but it is unclear how these structures form and how they are stabilized. We therefore investigated the ability of VP40 to form stable oligomers using in vitro and cellular assays. We characterized two lysine-rich regions in the VP40 C-terminal domain (CTD) that bind phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) and play distinct roles in lipid binding and the assembly of the EBOV matrix layer. The extensive analysis of VP40 with and without lipids by hydrogen deuterium exchange mass spectrometry revealed that VP40 oligomers become extremely stable when VP40 binds PI(4,5)P2. The PI(4,5)P2-induced stability of VP40 dimers and oligomers is a critical factor in VP40 oligomerization and release of VLPs from the plasma membrane. The two lysine-rich regions of the VP40 CTD have different roles with respect to interactions with plasma membrane phosphatidylserine (PS) and PI(4,5)P2. CTD region 1 (Lys221, Lys224, and Lys225) interacts with PI(4,5)P2 more favorably than PS and is important for VP40 extent of oligomerization. In contrast, region 2 (Lys270, Lys274, Lys275, and Lys279) mediates VP40 oligomer stability via lipid interactions and has a more prominent role in release of VLPs.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/metabolism , Lysine/metabolism , Binding Sites , Lipids , Protein Binding
14.
Nat Commun ; 14(1): 6785, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37880247

ABSTRACT

Marburg and Ebola filoviruses are two of the deadliest infectious agents and several outbreaks have occurred in the last decades. Although several receptors and co-receptors have been reported for Ebola virus, key host factors remain to be elucidated. In this study, using a haploid cell screening platform, we identify the guanine nucleotide exchange factor CCZ1 as a key host factor in the early stage of filovirus replication. The critical role of CCZ1 for filovirus infections is validated in 3D primary human hepatocyte cultures and human blood-vessel organoids, both critical target sites for Ebola and Marburg virus tropism. Mechanistically, CCZ1 controls early to late endosomal trafficking of these viruses. In addition, we report that CCZ1 has a role in the endosomal trafficking of endocytosis-dependent SARS-CoV-2 infections, but not in infections by Lassa virus, which enters endo-lysosomal trafficking at the late endosome stage. Thus, we have identified an essential host pathway for filovirus infections in cell lines and engineered human target tissues. Inhibition of CCZ1 nearly completely abolishes Marburg and Ebola infections. Thus, targeting CCZ1 could potentially serve as a promising drug target for controlling infections caused by various viruses, such as SARS-CoV-2, Marburg, and Ebola.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Marburg Virus Disease , Marburgvirus , Vesicular Transport Proteins , Animals , Humans , Ebolavirus/metabolism , Lysosomes , Marburg Virus Disease/genetics , Marburg Virus Disease/metabolism , Marburgvirus/metabolism , Vesicular Transport Proteins/metabolism
15.
J Mol Biol ; 435(20): 168241, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37598728

ABSTRACT

Viral inclusion bodies (IBs) are potential sites of viral replication and assembly. How viral IBs form remains poorly defined. Here we describe a combined biophysical and cellular approach to identify the components necessary for IB formation during Ebola virus (EBOV) infection. We find that the eNP0VP35 complex containing Ebola nucleoprotein (eNP) and viral protein 35 (eVP35), the functional equivalents of nucleoprotein (N) and phosphoprotein (P) in non-segmented negative strand viruses (NNSVs), phase separates to form inclusion bodies. Phase separation of eNP0VP35 is reversible and modulated by ionic strength. The multivalency of eVP35, and not eNP, is also critical for phase separation. Furthermore, overexpression of an eVP35 peptide disrupts eNP0VP35 complex formation, leading to reduced frequency of IB formation and limited viral infection. Together, our results show that upon EBOV infection, the eNP0VP35 complex forms the minimum unit to drive IB formation and viral replication.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Inclusion Bodies , Nucleoproteins , Virus Replication , Humans , Ebolavirus/metabolism , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Inclusion Bodies/virology , Nucleoproteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism
16.
Biotechnol Lett ; 45(10): 1327-1337, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37526868

ABSTRACT

PURPOSE: Viruses, such as Ebola virus (EBOV), evolve rapidly and threaten the human health. There is a great demand to exploit efficient gene-editing techniques for the identification of virus to probe virulence mechanism for drug development. METHODS: Based on lambda Red recombination in Escherichia coli (E. coli), counter-selection, and in vitro annealing, a high-efficiency genetic method was utilized here for precisely engineering viruses. EBOV trVLPs assay and dual luciferase reporter assay were used to further test the effect of mutations on virus replication. RESULTS: Considering the significance of matrix protein VP24 in EBOV replication, the types of mutations within vp24, including several single-base substitutions, one double-base substitution, two seamless deletions, and one targeted insertion, were generated on the multi-copy plasmid of E. coli. Further, the length of the homology arms for recombination and in vitro annealing, and the amount of DNA cassettes and linear plasmids were optimized to create a more elaborate and cost-efficient protocol than original approach. The effects of VP24 mutations on the expression of a reporter gene (luciferase) from the EBOV minigenome were determined, and results indicated that mutations of key sites within VP24 have significant impacts on EBOV replication. CONCLUSION: This precise mutagenesis method will facilitate effective and simple editing of viral genes in E. coli.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Viral Proteins/genetics , Hemorrhagic Fever, Ebola/genetics , Ebolavirus/genetics , Ebolavirus/metabolism , Genetic Engineering , Recombination, Genetic
17.
J Infect Dis ; 228(Suppl 7): S488-S497, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37551415

ABSTRACT

The 3' untranslated regions (UTRs) of Ebola virus (EBOV) mRNAs are enriched in their AU content and therefore represent potential targets for RNA binding proteins targeting AU-rich elements (ARE-BPs). ARE-BPs are known to fine-tune RNA turnover and translational activity. We identified putative AREs within EBOV mRNA 3' UTRs and assessed whether they might modulate mRNA stability. Using mammalian and zebrafish embryo reporter assays, we show a conserved, ARE-BP-mediated stabilizing effect and increased reporter activity with the tested EBOV 3' UTRs. When coexpressed with the prototypic ARE-BP tristetraprolin (TTP, ZFP36) that mainly destabilizes its target mRNAs, the EBOV nucleoprotein (NP) 3' UTR resulted in decreased reporter gene activity. Coexpression of NP with TTP led to reduced NP protein expression and diminished EBOV minigenome activity. In conclusion, the enrichment of AU residues in EBOV 3' UTRs makes them possible targets for cellular ARE-BPs, leading to modulation of RNA stability and translational activity.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Animals , 3' Untranslated Regions/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ebolavirus/genetics , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/genetics , Zebrafish/metabolism , RNA Stability/genetics , Mammals
18.
J Phys Chem B ; 127(29): 6449-6461, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37458567

ABSTRACT

The Ebola virus (EBOV) is a filamentous virus that acquires its lipid envelope from the plasma membrane of the host cell it infects. EBOV assembly and budding from the host cell plasma membrane are mediated by a peripheral protein, known as the matrix protein VP40. VP40 is a 326 amino acid protein with two domains that are loosely linked. The VP40 N-terminal domain (NTD) contains a hydrophobic α-helix, which mediates VP40 dimerization. The VP40 C-terminal domain has a cationic patch, which mediates interactions with anionic lipids and a hydrophobic region that mediates VP40 dimer-dimer interactions. The VP40 dimer is necessary for trafficking to the plasma membrane inner leaflet and interactions with anionic lipids to mediate the VP40 assembly and oligomerization. Despite significant structural information available on the VP40 dimer structure, little is known on how the VP40 dimer is stabilized and how residues outside the NTD hydrophobic portion of the α-helical dimer interface contribute to dimer stability. To better understand how VP40 dimer stability is maintained, we performed computational studies using per-residue energy decomposition and site saturation mutagenesis. These studies revealed a number of novel keystone residues for VP40 dimer stability just adjacent to the α-helical dimer interface as well as distant residues in the VP40 CTD that can stabilize the VP40 dimer form. Experimental studies with representative VP40 mutants in vitro and in cells were performed to test computational predictions that reveal residues that alter VP40 dimer stability. Taken together, these studies provide important biophysical insights into VP40 dimerization and may be useful in strategies to weaken or alter the VP40 dimer structure as a means of inhibiting the EBOV assembly.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Hemorrhagic Fever, Ebola/metabolism , Ebolavirus/genetics , Ebolavirus/metabolism , Dimerization , Mutagenesis , Lipids/chemistry , Viral Matrix Proteins/chemistry
19.
Structure ; 31(9): 1038-1051.e7, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37392738

ABSTRACT

The Ebola virus matrix protein VP40 mediates viral budding and negatively regulates viral RNA synthesis. The mechanisms by which these two functions are exerted and regulated are unknown. Using a high-resolution crystal structure of Sudan ebolavirus (SUDV) VP40, we show here that two cysteines in the flexible C-terminal arm of VP40 form a stabilizing disulfide bridge. Notably, the two cysteines are targets of posttranslational redox modifications and interact directly with the host`s thioredoxin system. Mutation of the cysteines impaired the budding function of VP40 and relaxed its inhibitory role for viral RNA synthesis. In line with these results, the growth of recombinant Ebola viruses carrying cysteine mutations was impaired and the released viral particles were elongated. Our results revealed the exact positions of the cysteines in the C-terminal arm of SUDV VP40. The cysteines and/or their redox status are critically involved in the differential regulation of viral budding and viral RNA synthesis.


Subject(s)
Ebolavirus , Viral Matrix Proteins , Ebolavirus/genetics , Ebolavirus/metabolism , Mutation , Oxidation-Reduction , Sudan , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virus Assembly , Humans
20.
J Infect Dis ; 228(Suppl 7): S479-S487, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37119290

ABSTRACT

BACKGROUND: Our previous study demonstrated that the fruit bat (Yaeyama flying fox)-derived cell line FBKT1 showed preferential susceptibility to Ebola virus (EBOV), whereas the human cell line HEK293T was similarly susceptible to EBOV and Marburg virus (MARV). This was due to 3 amino acid differences of the endosomal receptor Niemann-Pick C1 (NPC1) between FBKT1 and HEK293T (ie, TET and SGA, respectively, at positions 425-427), as well as 2 amino acid differences at positions 87 and 142 of the viral glycoprotein (GP) between EBOV and MARV. METHODS/RESULTS: To understand the contribution of these amino acid differences to interactions between NPC1 and GP, we performed molecular dynamics simulations and binding free energy calculations. The average binding free energies of human NPC1 (hNPC1) and its mutant having TET at positions 425-427 (hNPC1/TET) were similar for the interaction with EBOV GP. In contrast, hNPC1/TET had a weaker interaction with MARV GP than wild-type hNPC1. As expected, substitutions of amino acid residues at 87 or 142 in EBOV and MARV GPs converted the binding affinity to hNPC1/TET. CONCLUSIONS: Our data provide structural and energetic insights for understanding potential differences in the GP-NPC1 interaction, which could influence the host tropism of EBOV and MARV.


Subject(s)
Chiroptera , Ebolavirus , Hemorrhagic Fever, Ebola , Marburgvirus , Animals , Humans , Niemann-Pick C1 Protein , Marburgvirus/metabolism , HEK293 Cells , Virus Internalization , Glycoproteins/metabolism , Ebolavirus/metabolism , Amino Acids
SELECTION OF CITATIONS
SEARCH DETAIL