Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.182
Filter
1.
Proc Natl Acad Sci U S A ; 121(37): e2404250121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39231203

ABSTRACT

Human cytomegalovirus (HCMV) glycoprotein B (gB) is a class III membrane fusion protein required for viral entry. HCMV vaccine candidates containing gB have demonstrated moderate clinical efficacy, but no HCMV vaccine has been approved. Here, we used structure-based design to identify and characterize amino acid substitutions that stabilize gB in its metastable prefusion conformation. One variant containing two engineered interprotomer disulfide bonds and two cavity-filling substitutions (gB-C7), displayed increased expression and thermostability. A 2.8 Å resolution cryoelectron microscopy structure shows that gB-C7 adopts a prefusion-like conformation, revealing additional structural elements at the membrane-distal apex. Unlike previous observations for several class I viral fusion proteins, mice immunized with postfusion or prefusion-stabilized forms of soluble gB protein displayed similar neutralizing antibody titers, here specifically against an HCMV laboratory strain on fibroblasts. Collectively, these results identify initial strategies to stabilize class III viral fusion proteins and provide tools to probe gB-directed antibody responses.


Subject(s)
Cytomegalovirus , Viral Envelope Proteins , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Cytomegalovirus/immunology , Humans , Animals , Mice , Cryoelectron Microscopy , Protein Conformation , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Virus Internalization , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Protein Stability , Cytomegalovirus Vaccines/immunology , Amino Acid Substitution , Models, Molecular
2.
Nature ; 633(8030): 704-709, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39232163

ABSTRACT

Fifty-eight million individuals worldwide are affected by chronic hepatitis C virus (HCV) infection, a primary driver of liver cancer for which no vaccine is available1. The HCV envelope proteins E1 and E2 form a heterodimer (E1/E2), which is the target for neutralizing antibodies2. However, the higher-order organization of these E1/E2 heterodimers, as well as that of any Hepacivirus envelope protein complex, remains unknown. Here we determined the cryo-electron microscopy structure of two E1/E2 heterodimers in a homodimeric arrangement. We reveal how the homodimer is established at the molecular level and provide insights into neutralizing antibody evasion and membrane fusion by HCV, as orchestrated by E2 motifs such as hypervariable region 1 and antigenic site 412, as well as the organization of the transmembrane helices, including two internal to E1. This study addresses long-standing questions on the higher-order oligomeric arrangement of Hepacivirus envelope proteins and provides a critical framework in the design of novel HCV vaccine antigens.


Subject(s)
Hepacivirus , Protein Multimerization , Viral Envelope Proteins , Humans , Amino Acid Motifs , Antibodies, Neutralizing/immunology , Cryoelectron Microscopy , Hepacivirus/chemistry , Hepacivirus/immunology , Hepacivirus/metabolism , Hepacivirus/ultrastructure , Immune Evasion/immunology , Membrane Fusion , Models, Molecular , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/ultrastructure , Virus Internalization , Viral Hepatitis Vaccines/chemistry , Viral Hepatitis Vaccines/immunology
3.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273370

ABSTRACT

Zika virus (ZIKV; family, Flaviviridae), which causes congenital Zika syndrome, Guillain-Barré Syndrome, and other severe diseases, is transmitted mainly by mosquitoes; however, the virus can be transmitted through other routes. Among the three structural and seven nonstructural proteins, the surface envelope (E) protein of ZIKV plays a critical role in viral entry and pathogenesis, making it a key target for the development of effective entry inhibitors. This review article describes the life cycle, genome, and encoded proteins of ZIKV, illustrates the structure and function of the ZIKV E protein, summarizes E protein-targeting entry inhibitors (with a focus on those based on natural products and small molecules), and highlights challenges that may potentially hinder the development of effective inhibitors of ZIKV infection. Overall, the article will provide useful guidance for further development of safe and potent ZIKV entry inhibitors targeting the viral E protein.


Subject(s)
Antiviral Agents , Viral Envelope Proteins , Virus Internalization , Zika Virus Infection , Zika Virus , Zika Virus/drug effects , Zika Virus/physiology , Virus Internalization/drug effects , Humans , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/antagonists & inhibitors , Zika Virus Infection/virology , Zika Virus Infection/drug therapy , Antiviral Agents/pharmacology , Animals
4.
Proc Natl Acad Sci U S A ; 121(39): e2408078121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39292744

ABSTRACT

The Pentamer complex of Human Cytomegalovirus (HCMV) consists of the viral glycoproteins gH, gL, UL128, UL130, and UL131 and is incorporated into infectious virions. HCMV strains propagated extensively in vitro in fibroblasts carry UL128, UL130, or UL131 alleles that do not make a functional complex and thus lack Pentamer function. Adding functional Pentamer to such strains decreases virus growth in fibroblasts. Here, we show that the Pentamer inhibits productive HCMV replication in fibroblasts by repressing viral Immediate Early (IE) transcription. We show that ectopic expression of the viral IE1 protein, a target of Pentamer-mediated transcriptional repression, complements the growth defect of a Pentamer-positive virus. Furthermore, we show that the Pentamer also represses viral IE transcription in cell types where HCMV in vitro latency is studied. Finally, we identify UL130 as a functional subunit of the Pentamer for IE transcriptional repression and demonstrate that cyclic AMP Response Element (CRE) and NFkB sites within the Major Immediate Early Promoter that drives IE1 transcription contribute to this repression. We conclude that the HCMV Pentamer represses viral IE transcription.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Fibroblasts , Immediate-Early Proteins , Transcription, Genetic , Viral Envelope Proteins , Humans , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus/metabolism , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , Fibroblasts/virology , Fibroblasts/metabolism , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Gene Expression Regulation, Viral , Virus Replication/genetics , Glycoproteins/metabolism , Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Genes, Immediate-Early , Promoter Regions, Genetic
5.
Nature ; 633(8030): 695-703, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39232167

ABSTRACT

Viral glycoproteins drive membrane fusion in enveloped viruses and determine host range, tissue tropism and pathogenesis1. Despite their importance, there is a fragmentary understanding of glycoproteins within the Flaviviridae2, a large virus family that include pathogens such as hepatitis C, dengue and Zika viruses, and numerous other human, animal and emergent viruses. For many flaviviruses the glycoproteins have not yet been identified, for others, such as the hepaciviruses, the molecular mechanisms of membrane fusion remain uncharacterized3. Here we combine phylogenetic analyses with protein structure prediction to survey glycoproteins across the entire Flaviviridae. We find class II fusion systems, homologous to the Orthoflavivirus E glycoprotein in most species, including highly divergent jingmenviruses and large genome flaviviruses. However, the E1E2 glycoproteins of the hepaciviruses, pegiviruses and pestiviruses are structurally distinct, may represent a novel class of fusion mechanism, and are strictly associated with infection of vertebrate hosts. By mapping glycoprotein distribution onto the underlying phylogeny, we reveal a complex evolutionary history marked by the capture of bacterial genes and potentially inter-genus recombination. These insights, made possible through protein structure prediction, refine our understanding of viral fusion mechanisms and reveal the events that have shaped the diverse virology and ecology of the Flaviviridae.


Subject(s)
Evolution, Molecular , Flaviviridae , Glycoproteins , Phylogeny , Viral Envelope Proteins , Animals , Humans , Flaviviridae/chemistry , Flaviviridae/classification , Glycoproteins/chemistry , Glycoproteins/classification , Glycoproteins/metabolism , Models, Molecular , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/classification , Viral Envelope Proteins/metabolism
6.
Int J Mol Sci ; 25(18)2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39337476

ABSTRACT

Host restriction factor GBP2 suppresses the replication of the ecotropic Moloney murine leukemia virus (E-MLV) by inhibiting furin protease, which cleaves the viral envelope glycoprotein (Env) into surface (SU) and transmembrane (TM) subunits. We analyzed the impacts of GBP2 on the infection efficiency mediated by MLV Envs of different strains of ecotropic Moloney, polytropic Friend, amphotropic, and xenotropic MLV-related (XMRV) viruses. Interestingly, the Envs of ecotropic Moloney and polytropic Friend MLV were sensitive to the antiviral activity of GBP2, while XMRV and amphotropic Envs showed resistance. Consistent with the sensitivity to GBP2, the amino acid sequences of the sensitive Envs at the SU-TM cleavage site were similar, as were the sequences of the resistant Envs. SU-TM cleavage of the GBP2-sensitive Env protein was inhibited by furin silencing, whereas that of GBP2-resistant Env was not. The substitution of the ecotropic Moloney cleavage site sequence with that of XMRV conferred resistance to both GBP2 and furin silencing. Reciprocally, the substitution of the XMRV cleavage site sequence with that of the ecotropic sequence conferred sensitivity to GBP2 and furin silencing. According to the SU-TM cleavage site sequence, there were sensitive and resistant variants among ecotropic, polytropic, and xenotropic MLVs. This study found that the dependence of MLV Env proteins on furin cleavage and GBP2-mediated restriction is determined by the amino acid sequences at the SU-TM cleavage site.


Subject(s)
Furin , Viral Envelope Proteins , Furin/metabolism , Animals , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Mice , Humans , Amino Acid Sequence , Leukemia Virus, Murine/genetics , Virus Replication/drug effects , Retroviridae Infections/virology , Retroviridae Infections/metabolism
7.
Viruses ; 16(9)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39339951

ABSTRACT

Baculoviral vectors (BVs) derived from Autographa californica multiple nucleopolyhedrovirus (AcMNPV) are an attractive tool for multigene delivery in mammalian cells, which is particularly relevant for CRISPR technologies. Most applications in mammalian cells rely on BVs that are pseudotyped with vesicular stomatitis virus G-protein (VSV-G) to promote efficient endosomal release. VSV-G expression typically occurs under the control of the hyperactive polH promoter. In this study, we demonstrate that polH-driven VSV-G expression results in BVs characterised by reduced stability, impaired morphology, and VSV-G induced toxicity at high multiplicities of transduction (MOTs) in target mammalian cells. To overcome these drawbacks, we explored five alternative viral promoters with the aim of optimising VSV-G levels displayed on the pseudotyped BVs. We report that Orf-13 and Orf-81 promoters reduce VSV-G expression to less than 5% of polH, rescuing BV morphology and stability. In a panel of human cell lines, we elucidate that BVs with reduced VSV-G support efficient gene delivery and CRISPR-mediated gene editing, at levels comparable to those obtained previously with polH VSV-G-pseudotyped BVs (polH VSV-G BV). These results demonstrate that VSV-G hyperexpression is not required for efficient transduction of mammalian cells. By contrast, reduced VSV-G expression confers similar transduction dynamics while substantially improving BV integrity, structure, and stability.


Subject(s)
Genetic Vectors , Nucleopolyhedroviruses , Promoter Regions, Genetic , Transduction, Genetic , Viral Envelope Proteins , Humans , Nucleopolyhedroviruses/genetics , Nucleopolyhedroviruses/physiology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Genetic Vectors/genetics , Animals , Cell Line , Baculoviridae/genetics , Gene Editing/methods , HEK293 Cells , CRISPR-Cas Systems , Membrane Glycoproteins
8.
Viruses ; 16(9)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39339958

ABSTRACT

The human cytomegalovirus (HCMV) glycoprotein B (gB) is the viral fusogen required for entry into cells and for direct cell-to-cell spread of the virus. We have previously demonstrated that the exchange of the carboxy-terminal domain (CTD) of gB for the CTD of the structurally related fusion protein G of the vesicular stomatitis virus (VSV-G) resulted in an intrinsically fusion-active gB variant (gB/VSV-G). In this present study, we employed a dual split protein (DSP)-based cell fusion assay to further characterize the determinants of fusion activity in the CTD of gB. We generated a comprehensive library of gB CTD truncation mutants and identified two mutants, gB-787 and gB-807, which were fusion-competent and induced the formation of multinucleated cell syncytia in the absence of other HCMV proteins. Structural modeling coupled with site-directed mutagenesis revealed that gB fusion activity is primarily mediated by the CTD helix 2, and secondarily by the recruitment of cellular SH2/WW-domain-containing proteins. The fusion activity of gB-807 was inhibited by gB-specific monoclonal antibodies (MAbs) targeting the antigenic domains AD-1 to AD-5 within the ectodomain and not restricted to MAbs directed against AD-4 and AD-5 as observed for gB/VSV-G. This finding suggested a differential regulation of the fusion-active conformational state of both gB variants. Collectively, our findings underscore a pivotal role of the CTD in regulating the fusogenicity of HCMV gB, with important implications for understanding the conformations of gB that facilitate membrane fusion, including antigenic structures that could be targeted by antibodies to block this essential step in HCMV infection.


Subject(s)
Cytomegalovirus , Protein Domains , Viral Envelope Proteins , Virus Internalization , Humans , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/chemistry , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Giant Cells/virology , Cell Line , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/chemistry , Mutagenesis, Site-Directed , Cell Fusion
9.
Sci Rep ; 14(1): 20924, 2024 09 09.
Article in English | MEDLINE | ID: mdl-39251790

ABSTRACT

Human herpes viruses (HHV) are ubiquitous and have been implicated in numerous long-term health conditions. Since the association between viral exposure and long-term health impacts is partially influenced by variation in human leukocyte antigen (HLA) genes, we evaluated in silico the binding affinities of 9 HHV envelope glycoproteins with 127 common HLA Class I and Class II molecules. The findings show substantial variability in HHV binding affinity across viruses, HLA Class, HLA genes, and HLA alleles. Specific findings were as follows: (1) the predicted binding affinities of HHVs were characterized by four distinct groupings-[HHV1, HHV2], [HHV3, HHV4, HHV5], [HHV6A], [HHV6B, HHV7, HHV8]-with relatively lower binding affinities for HHV1, HHV2, and HHV6a compared to other HHVs; (2) significantly higher binding affinity was found for HLA Class I relative to Class II; (3) analyses within each class demonstrated that alleles of the C gene (for Class I) and DRB1 gene (for Class II) had the highest binding affinities; and (4) for each virus, predicted binding affinity to specific alleles varied, with HHV6a having the lowest affinity for HHV-HLA complexes, and HHV3, HHV4, and HHV5 having the highest. Since HLA-antigen binding is the first step in initiating an immune response to foreign antigens, these relative differences in HHV binding affinities are likely to influence long-term health impacts such that the cells infected with viruses associated with higher binding affinities across common HLA alleles may be more reduced in numbers, thereby lowering the potential for long-term sequelae of their infections.


Subject(s)
Alleles , Viral Envelope Proteins , Humans , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Herpesviridae/immunology , Herpesviridae/genetics , HLA Antigens/genetics , HLA Antigens/immunology , Protein Binding , Immunogenetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology
10.
Virus Res ; 349: 199458, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39187047

ABSTRACT

Coronaviruses (CoVs) are significant animal and human pathogens, characterized by being enveloped RNA viruses with positive-sense single-stranded RNA. The Coronaviridae family encompasses four genera, among which gammacoronaviruses pose a major threat to the poultry industry, which infectious bronchitis virus (IBV) being the most prominent of these threats. Particularly, IBV adversely affects broiler growth and egg production, causing substantial losses. The IBV strains currently circulating in Taiwan include the IBV Taiwan-I (TW-I) serotype, IBV Taiwan-II (TW-II) serotype, and vaccine strains. Therefore, ongoing efforts have focused on developing novel vaccines and discovering antiviral agents. The envelope (E) proteins of CoVs accumulate in the endoplasmic reticulum-Golgi intermediate compartment prior to virus budding. These E proteins assemble into viroporins, exhibiting ion channel activity that leads to cell membrane disruption, making them attractive targets for antiviral therapy. In this study, we investigated the E proteins of IBV H-120, as well as IBV serotypes TW-I and TW-II. E protein expression resulted in inhibited bacteria growth, increased permeability of bacteria to ß-galactosidase substrates, and blocked protein synthesis of bacteria by hygromycin B (HygB). Furthermore, in the presence of E proteins, HygB also impeded protein translation in DF-1 cells and damaged their membrane integrity. Collectively, these findings confirm the viroporin activity of the E proteins from IBV H-120, IBV serotype TW-I, and IBV serotype TW-II. Next, the viroporin inhibitors, 5-(N,N-hexamethylene) amiloride (HMA) and 4,4'-diisothiocyano stilbene-2,2'-disulphonic acid (DIDS) were used to inhibit the viroporin activities of the E proteins of IBV H-120, IBV serotype TW-I, and IBV serotype TW-II. In chicken embryos and chickens infected with IBV serotypes TW-I and IBV TW-II, no survivors were observed at 6 and 11 days post-infection (dpi), respectively. However, treatments with both DIDS and HMA increased the survival rates in infected chicken embryos and chickens and mitigated histopathological lesions in the trachea and kidney. Additionally, a 3D pentameric structure of the IBV E protein was constructed via homology modeling. As expected, both inhibitors were found to bind to the lipid-facing surface within the transmembrane domain of the E protein, inhibiting ion conduction. Taken together, our findings provide comprehensive evidence supporting the use of viroporin inhibitors as promising antiviral agents against IBV Taiwan isolates.


Subject(s)
Antiviral Agents , Infectious bronchitis virus , Infectious bronchitis virus/drug effects , Infectious bronchitis virus/genetics , Antiviral Agents/pharmacology , Taiwan , Animals , Coronavirus Infections/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Chickens , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Poultry Diseases/virology , Poultry Diseases/drug therapy , Viroporin Proteins/antagonists & inhibitors
11.
J Virol ; 98(9): e0054024, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39162433

ABSTRACT

Systemic viral infection of insects typically begins with the primary infection of midgut epithelial cells (enterocytes) and subsequent transit of the progeny virus in an apical-to-basal orientation into the hemocoel. For insect-vectored viruses, an oppositely oriented process (basal-to-apical transit) occurs upon secondary infection of salivary glands and is necessary for virus transmission to non-insect hosts. To examine this inversely oriented virus transit in these polarized tissues, we assessed the intracellular trafficking of two model viral envelope proteins (baculovirus GP64 and vesicular stomatitis virus G) in the midgut and salivary gland cells of the model insect, Drosophila melanogaster. Using fly lines that inducibly express either GP64 or VSV G, we found that each protein, expressed alone, was trafficked basally in midgut enterocytes. In salivary gland cells, VSV G was trafficked apically in most but not all cells, whereas GP64 was consistently trafficked basally. We demonstrated that a YxxØ motif present in both proteins was critical for basal trafficking in midgut enterocytes but dispensable for trafficking in salivary gland cells. Using RNAi, we found that clathrin adaptor protein complexes AP-1 and AP-3, as well as seven Rab GTPases, were involved in polarized VSV G trafficking in midgut enterocytes. Our results indicate that these viral envelope proteins encode the requisite information and require no other viral factors for appropriately polarized trafficking. In addition, they exploit tissue-specific differences in protein trafficking pathways to facilitate virus egress in the appropriate orientation for establishing systemic infections and vectoring infection to other hosts. IMPORTANCE: Viruses that use insects as hosts must navigate specific routes through different insect tissues to complete their life cycles. The routes may differ substantially depending on the life cycle of the virus. Both insect pathogenic viruses and insect-vectored viruses must navigate through the polarized cells of the midgut epithelium to establish a systemic infection. In addition, insect-vectored viruses must also navigate through the polarized salivary gland epithelium for transmission. Thus, insect-vectored viruses appear to traffic in opposite directions in these two tissues. In this study, we asked whether two viral envelope proteins (VSV G and baculovirus GP64) alone encode the signals necessary for the polarized trafficking associated with their respective life cycles. Using Drosophila as a model to examine tissue-specific polarized trafficking of these viral envelope proteins, we identified one of the virus-encoded signals and several host proteins associated with regulating the polarized trafficking in the midgut epithelium.


Subject(s)
Drosophila melanogaster , Protein Transport , Salivary Glands , Viral Envelope Proteins , Animals , Salivary Glands/virology , Salivary Glands/metabolism , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Drosophila melanogaster/virology , Drosophila melanogaster/metabolism , Insect Vectors/virology , Insect Vectors/metabolism , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Enterocytes/virology , Enterocytes/metabolism , Gastrointestinal Tract/virology , Gastrointestinal Tract/metabolism
12.
Cell Stem Cell ; 31(9): 1298-1314.e8, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39146934

ABSTRACT

Endogenous retroviruses (ERVs) occupy a significant part of the human genome, with some encoding proteins that influence the immune system or regulate cell-cell fusion in early extra-embryonic development. However, whether ERV-derived proteins regulate somatic development is unknown. Here, we report a somatic developmental function for the primate-specific ERVH48-1 (SUPYN/Suppressyn). ERVH48-1 encodes a fragment of a viral envelope that is expressed during early embryonic development. Loss of ERVH48-1 led to impaired mesoderm and cardiomyocyte commitment and diverted cells to an ectoderm-like fate. Mechanistically, ERVH48-1 is localized to sub-cellular membrane compartments through a functional N-terminal signal peptide and binds to the WNT antagonist SFRP2 to promote its polyubiquitination and degradation, thus limiting SFRP2 secretion and blocking repression of WNT/ß-catenin signaling. Knockdown of SFRP2 or expression of a chimeric SFRP2 with the ERVH48-1 signal peptide rescued cardiomyocyte differentiation. This study demonstrates how ERVH48-1 modulates WNT/ß-catenin signaling and cell type commitment in somatic development.


Subject(s)
Cell Differentiation , Endogenous Retroviruses , Membrane Proteins , Myocytes, Cardiac , Wnt Signaling Pathway , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Endogenous Retroviruses/metabolism , Endogenous Retroviruses/genetics , Animals , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Primates , HEK293 Cells , Mesoderm/metabolism
13.
Microb Pathog ; 195: 106849, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147215

ABSTRACT

The white spot syndrome virus (WSSV), a rapidly replicating and highly lethal pathogen that targets Penaeid shrimp, has emerged as one of the most widespread viruses globally due to its high virulence. With effective chemotherapeutics still unavailable, the pursuit of novel and viable strategies against WSSV remains a crucial focus in the field of shrimp farming. The envelope proteins of WSSV are essential for virus entry, serving as excellent targets for the development of antiviral therapeutics. Novel strategies in the design of inhibitory peptides, especially those targeting envelope protein (VP28) located on the surface of the virus particle, play a critical role as a significant virulence factor during the early stages of inherent WSSV infection in shrimp. In this direction, the current computational study focused on identifying self-inhibitory peptides from the hydrophobic membrane regions of the VP28 protein, employing peptide docking and molecular dynamics simulation (MDS) approaches. Such inhibitory peptides could be useful building blocks for the rational engineering of inhibitory therapeutics since they imitate the mechanism of binding to homologous partners used by their origin domain to interact with other molecules. The N-terminal sequence of VP28 has been reported as the potential site for membrane interactions during the virus entry. Moreover, drug delivery systems mediated by chitosan and gold nanoparticles are being developed to enhance the therapeutic efficacy of anti-viral peptides. These systems can increase the solubility, stability, and selectivity of peptides, possessing better qualities than conventional delivery methods. This computational study on self-inhibitory peptides could be a valuable resource for further in vitro and in vivo studies on anti-viral therapeutics in the aquaculture industry.


Subject(s)
Antiviral Agents , Molecular Docking Simulation , Molecular Dynamics Simulation , Penaeidae , Peptides , White spot syndrome virus 1 , White spot syndrome virus 1/drug effects , White spot syndrome virus 1/genetics , Antiviral Agents/pharmacology , Animals , Peptides/pharmacology , Peptides/chemistry , Penaeidae/virology , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/chemistry , Virus Internalization/drug effects
14.
PLoS Pathog ; 20(8): e1012468, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39146367

ABSTRACT

Genetic editing of the germline using CRISPR/Cas9 technology has made it possible to alter livestock traits, including the creation of resistance to viral diseases. However, virus adaptability could present a major obstacle in this effort. Recently, chickens resistant to avian leukosis virus subgroup J (ALV-J) were developed by deleting a single amino acid, W38, within the ALV-J receptor NHE1 using CRISPR/Cas9 genome editing. This resistance was confirmed both in vitro and in vivo. In vitro resistance of W38-/- chicken embryonic fibroblasts to all tested ALV-J strains was shown. To investigate the capacity of ALV-J for further adaptation, we used a retrovirus reporter-based assay to select adapted ALV-J variants. We assumed that adaptive mutations overcoming the cellular resistance would occur within the envelope protein. In accordance with this assumption, we isolated and sequenced numerous adapted virus variants and found within their envelope genes eight independent single nucleotide substitutions. To confirm the adaptive capacity of these substitutions, we introduced them into the original retrovirus reporter. All eight variants replicated effectively in W38-/- chicken embryonic fibroblasts in vitro while in vivo, W38-/- chickens were sensitive to tumor induction by two of the variants. Importantly, receptor alleles with more extensive modifications have remained resistant to the virus. These results demonstrate an important strategy in livestock genome engineering towards antivirus resistance and illustrate that cellular resistance induced by minor receptor modifications can be overcome by adapted virus variants. We conclude that more complex editing will be necessary to attain robust resistance.


Subject(s)
Avian Leukosis Virus , Avian Leukosis , Chickens , Animals , Avian Leukosis Virus/genetics , Avian Leukosis Virus/physiology , Chickens/virology , Avian Leukosis/virology , Avian Leukosis/genetics , Poultry Diseases/virology , Poultry Diseases/genetics , Disease Resistance/genetics , CRISPR-Cas Systems , Gene Editing , Chick Embryo , Evolution, Molecular , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Fibroblasts/virology , Fibroblasts/metabolism
15.
Nat Commun ; 15(1): 6548, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095394

ABSTRACT

Eastern equine encephalitis virus (EEEV) is the most virulent alphavirus that infects humans, and many survivors develop neurological sequelae, including paralysis and intellectual disability. Alphavirus spike proteins comprise trimers of heterodimers of glycoproteins E2 and E1 that mediate binding to cellular receptors and fusion of virus and host cell membranes during entry. We recently identified very-low density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2) as cellular receptors for EEEV and a distantly related alphavirus, Semliki Forest virus (SFV). Here, we use single-particle cryo-electron microscopy (cryo-EM) to determine structures of the EEEV and SFV spike glycoproteins bound to the VLDLR ligand-binding domain and found that EEEV and SFV interact with the same cellular receptor through divergent binding modes. Our studies suggest that the ability of LDLR-related proteins to interact with viral spike proteins through very small footprints with flexible binding modes results in a low evolutionary barrier to the acquisition of LDLR-related proteins as cellular receptors for diverse sets of viruses.


Subject(s)
Cryoelectron Microscopy , Encephalitis Virus, Eastern Equine , Receptors, LDL , Receptors, LDL/metabolism , Receptors, LDL/chemistry , Encephalitis Virus, Eastern Equine/metabolism , Encephalitis Virus, Eastern Equine/ultrastructure , Humans , Animals , Semliki forest virus/metabolism , Protein Binding , Receptors, Virus/metabolism , Receptors, Virus/chemistry , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/ultrastructure , Models, Molecular
16.
Viruses ; 16(8)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39205190

ABSTRACT

Transduction of producer cells during lentiviral vector (LVV) production causes the loss of 70-90% of viable particles. This process is called retro-transduction and it is a consequence of the interaction between the LVV envelope protein, VSV-G, and the LDL receptor located on the producer cell membrane, allowing lentiviral vector transduction. Avoiding retro-transduction in LVV manufacturing is crucial to improve net production and, therefore, the efficiency of the production process. Here, we describe a method for quantifying the transduction of producer cells and three different strategies that, focused on the interaction between VSV-G and the LDLR, aim to reduce retro-transduction.


Subject(s)
Genetic Vectors , Lentivirus , Receptors, LDL , Transduction, Genetic , Genetic Vectors/genetics , Lentivirus/genetics , Humans , Receptors, LDL/metabolism , Receptors, LDL/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , HEK293 Cells , Membrane Glycoproteins
17.
Viruses ; 16(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39205211

ABSTRACT

The West Nile virus (WNV) subtype Kunjin virus (WNVKUN) is endemic to Australia. Here, we characterized the classical WNVKUN strain, OR393. The original OR393 strain contained two types of viruses: small plaque-forming virus (SP) and large plaque-forming virus (LP). The amino acid residues at positions 156 and 332 in the E protein (E156 and E332) of SP were Ser and Lys (E156S/332K), respectively, whereas those in LP were Phe and Thr (E156F/332T). SP grew slightly faster than LP in vitro. The E protein of SP was N-glycosylated, whereas that of LP was not. Analysis using two recombinant single-mutant LP viruses, rKUNV-LP-EF156S and rKUNV-LP-ET332K, indicated that E156S enlarged plaques formed by LP, but E332K potently reduced them, regardless of the amino acid at E156. rKUNV-LP-EF156S showed significantly higher neuroinvasive ability than LP, SP, and rKUNV-LP-ET332K. Our results indicate that the low-pathogenic classical WNVKUN can easily change its pathogenicity through only a few amino acid substitutions in the E protein. It was also found that Phe at E156 of the rKUNV-LP-ET332K was easily changed to Ser during replication in vitro and in vivo, suggesting that E156S is advantageous for the propagation of WNVKUN in mammalian cells.


Subject(s)
Viral Envelope Proteins , Viral Plaque Assay , West Nile Fever , West Nile virus , Animals , West Nile virus/genetics , West Nile virus/pathogenicity , West Nile virus/physiology , Mice , West Nile Fever/virology , Virulence , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/chemistry , Amino Acids/metabolism , Amino Acids/genetics , Virus Replication , Chlorocebus aethiops , Amino Acid Substitution , Vero Cells , Female , Humans , Australia , Cell Line
18.
Int J Mol Sci ; 25(16)2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39201685

ABSTRACT

An efficient gene transfer and expression tool is lacking for shrimps and shrimp cells. To solve this, this study has developed a shrimp DNA virus-mediated gene transfer and expression system, consisting of insect Sf9 cells for viral packaging, the shrimp viral vector of pUC19-IHHNV-PH-GUS and the baculoviral vector of Bacmid or Bacmid-VP28 encoding the shrimp WSSV envelope protein VP28. The pUC19-IHHNV-PH-GUS vector was constructed by assembling the genomic DNA of shrimp infectious hypodermal and hematopoietic necrosis virus (IHHNV), which has shortened inverted terminal repeats, into a pUC19 backbone, and then an expression cassette of baculoviral polyhedron (PH) promoter-driven GUS (ß-glucuronidase) reporter gene was inserted immediately downstream of IHHNV for proof-of-concept. It was found that the viral vector of pUC19-IHHNV-PH-GUS could be successfully packaged into IHHNV-like infective virions in the Sf9 cells, and the gene transfer efficiency of this system was evaluated and verified in three systems of Sf9 cells, shrimp hemolymph cells and tissues of infected shrimps, but the GUS expression could only be detected in cases where the viral vector was co-transfected or co-infected with a baculovirus of Bacmid or Bacmid-VP28 due to the Bacmid-dependence of the PH promoter. Moreover, the packaging and infection efficiencies could be significantly improved when Bacmid-VP28 was used instead of Bacmid.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Penaeidae , Animals , Penaeidae/virology , Penaeidae/genetics , Sf9 Cells , Genetic Vectors/genetics , Baculoviridae/genetics , Promoter Regions, Genetic , Spodoptera/virology , Densovirinae/genetics , Gene Expression , White spot syndrome virus 1/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Glucuronidase/genetics , Glucuronidase/metabolism
19.
Genes (Basel) ; 15(8)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39202352

ABSTRACT

Atypical porcine pestivirus (APPV) can cause congenital tremor type A-II in neonatal piglets, posing a significant threat to swine herd health globally. Our previous study demonstrated that the Mut domains, comprising 112 amino acids at the N-terminus, are the primary functional regions of the E2 protein of APPV. This study identified 14 host cellular proteins that exhibit potential interactions with the Mut domains of the E2 protein using yeast two-hybrid screening. Using bioinformatics analysis, we discovered that the Mut domains of the E2 protein might exert regulatory effects on apoptosis by modulating energy metabolism within the mitochondria. We also conducted co-immunoprecipitation, glutathione S-transferase pull-down, and immunofluorescence assays to confirm the interaction between the Mut domains of the E2 protein and cathepsin H and signal sequence receptor subunit 4 (SSR4). Ultimately, SSR4 enhanced APPV replication in vitro. In summary, our study successfully elucidated the interactions between the Mut domains of the E2 protein and host cell protein, predicted the potential pathways implicated in these interactions, and demonstrated SSR4 involvement in APPV infection. These significant findings contribute valuable knowledge toward a deeper understanding of APPV pathogenesis and the role of the Mut domains of the E2 protein in this intricate process.


Subject(s)
Pestivirus Infections , Pestivirus , Animals , Pestivirus/genetics , Pestivirus/metabolism , Swine , Pestivirus Infections/veterinary , Pestivirus Infections/virology , Pestivirus Infections/genetics , Swine Diseases/virology , Swine Diseases/genetics , Swine Diseases/metabolism , Host-Pathogen Interactions/genetics , Protein Domains , Virus Replication/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Humans , Protein Interaction Maps/genetics
20.
Sci Rep ; 14(1): 19585, 2024 08 23.
Article in English | MEDLINE | ID: mdl-39179615

ABSTRACT

The World Health Organization (WHO) has declared the monkeypox outbreak a public health emergency, as there is no specific therapeutics for monkeypox virus (MPXV) disease. This study focused on docking various commercial drugs and plant-derived compounds against the E8 envelope protein crucial for MPXV attachment and pathogenesis. The target protein structure was modeled based on the vaccinia virus D8L protein. Notably, maraviroc and punicalagin emerged as potential ligands, with punicalagin exhibiting higher binding affinity (- 9.1 kcal/mol) than maraviroc (- 7.8 kcal/mol). Validation through 100 ns molecular dynamics (MD) simulations demonstrated increased stability of the E8-punicalagin complex, with lower RMSD, RMSF, and Rg compared to maraviroc. Enhanced hydrogen bonding, lower solvent accessibility, and compact motions also attributed to higher binding affinity and stability of the complex. MM-PBSA calculations revealed van der Waals, electrostatic, and non-polar solvation as principal stabilizing energies. The binding energy decomposition per residue favored stable interactions between punicalagin and the protein's active site residues (Arg20, Phe56, Glu228, Tyr232) compared to maraviroc. Overall study suggests that punicalagin can act as a potent inhibitor against MPXV. Further research and experimental investigations are warranted to validate its efficacy and safety.


Subject(s)
Maraviroc , Molecular Docking Simulation , Molecular Dynamics Simulation , Viral Envelope Proteins , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/antagonists & inhibitors , Maraviroc/chemistry , Maraviroc/pharmacology , Monkeypox virus/chemistry , Monkeypox virus/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Hydrogen Bonding , Protein Binding , Hydrolyzable Tannins/chemistry , Hydrolyzable Tannins/pharmacology , Hydrolyzable Tannins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL