Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 23(21)2022 Oct 30.
Article in English | MEDLINE | ID: mdl-36361998

ABSTRACT

Porcine reproductive and respiratory syndrome virus is a positive-stranded RNA virus of the family Arteriviridae. The Gp5/M dimer, the major component of the viral envelope, is required for virus budding and is an antibody target. We used alphafold2, an artificial-intelligence-based system, to predict a credible structure of Gp5/M. The short disulfide-linked ectodomains lie flat on the membrane, with the exception of the erected N-terminal helix of Gp5, which contains the antibody epitopes and a hypervariable region with a changing number of carbohydrates. The core of the dimer consists of six curved and tilted transmembrane helices, and three are from each protein. The third transmembrane regions extend into the cytoplasm as amphiphilic helices containing the acylation sites. The endodomains of Gp5 and M are composed of seven ß-strands from each protein, which interact via ß-strand seven. The area under the membrane forms an open cavity with a positive surface charge. The M and Orf3a proteins of coronaviruses have a similar structure, suggesting that all four proteins are derived from the same ancestral gene. Orf3a, like Gp5/M, is acylated at membrane-proximal cysteines. The role of Gp5/M during virus replication is discussed, in particular the mechanisms of virus budding and models of antibody-dependent virus neutralization.


Subject(s)
Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Swine , Animals , Porcine respiratory and reproductive syndrome virus/genetics , Viral Envelope Proteins/metabolism , Epitopes , Virus Replication
2.
Int J Mol Sci ; 23(9)2022 May 03.
Article in English | MEDLINE | ID: mdl-35563480

ABSTRACT

Lipid modification of viral proteins with fatty acids of different lengths (S-acylation) is crucial for virus pathogenesis. The reaction is catalyzed by members of the DHHC family and proceeds in two steps: the autoacylation is followed by the acyl chain transfer onto protein substrates. The crystal structure of human DHHC20 (hDHHC20), an enzyme involved in the acylation of S-protein of SARS-CoV-2, revealed that the acyl chain may be inserted into a hydrophobic cavity formed by four transmembrane (TM) α-helices. To test this model, we used molecular dynamics of membrane-embedded hDHHC20 and its mutants either in the absence or presence of various acyl-CoAs. We found that among a range of acyl chain lengths probed only C16 adopts a conformation suitable for hDHHC20 autoacylation. This specificity is altered if the small or bulky residues at the cavity's ceiling are exchanged, e.g., the V185G mutant obtains strong preferences for binding C18. Surprisingly, an unusual hydrophilic ridge was found in TM helix 4 of hDHHC20, and the responsive hydrophilic patch supposedly involved in association was found in the 3D model of the S-protein TM-domain trimer. Finally, the exchange of critical Thr and Ser residues in the spike led to a significant decrease in its S-acylation. Our data allow further development of peptide/lipid-based inhibitors of hDHHC20 that might impede replication of Corona- and other enveloped viruses.


Subject(s)
Acyltransferases , COVID-19 , Acyl Coenzyme A/metabolism , Acylation , Acyltransferases/chemistry , Acyltransferases/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Humans , Molecular Dynamics Simulation , SARS-CoV-2 , Substrate Specificity/physiology
3.
Vector Borne Zoonotic Dis ; 21(12): 1003-1006, 2021 12.
Article in English | MEDLINE | ID: mdl-34958267

ABSTRACT

Background: The endemic character of Rift Valley fever (RVF) disease points toward an interepidemic reservoir. Although not yet identified, bats and rodents may be implicated in RVF virus (RVFV) epidemiology. In this study, we investigated the putative role of Egyptian frugivorous and insectivorous bats in RVFV epidemiology in Egypt. Methods: From 2019 to 2021, 200 bats of two different species from six Egyptian governorates were tested for phleboviruses using real-time RT-PCR (rRT-PCR) and sequence analysis. Results: Screening through rRT-PCR showed evidence of the RVFV genome only in insectivorous bats. Partial sequence and phylogenetic analysis based on S and M genome segments showed that these viruses are genetically similar to those circulating (clade A) in livestock and humans during previously reported RVFV outbreaks in 1977/78 and 2003 in Egypt. Conclusions: Our molecular data suggest that the bat Pipistrellus deserti could play a role in RVFV ecology in Egypt.


Subject(s)
Chiroptera , Rift Valley Fever , Rift Valley fever virus , Animals , Egypt/epidemiology , Phylogeny , Rift Valley Fever/epidemiology , Rift Valley fever virus/genetics
4.
Cell Microbiol ; 23(6): e13322, 2021 06.
Article in English | MEDLINE | ID: mdl-33629465

ABSTRACT

Influenza A viruses contain two S-acylated proteins, the ion channel M2 and the glycoprotein hemagglutinin (HA). Acylation of the latter is essential for virus replication. Here we analysed the expression of each of the 23 members of the family of ZDHHC acyltransferases in human airway cells, the site of virus replication. RT-PCR revealed that every ZDHHC acyltransferase (except ZDHHC19) is expressed in A549 and Calu cells. Interestingly, expression of one ZDHHC, ZDHHC22, is upregulated in virus-infected cells; this effect is more pronounced after infection with an avian compared to a human virus strain. The viral protein NS1 triggers ZDHHC22 expression in transfected cells, whereas recombinant viruses lacking a functional NS1 gene did not cause ZDHHC22 upregulation. CRISPR/Cas9 technology was then used to knock-out the ZDHHC22 gene in A549 cells. However, acylation of M2 and HA was not reduced, as analysed for intracellular HA and M2 and the stoichiometry of S-acylation of HA incorporated into virus particles did not change according to MALDI-TOF mass spectrometry analysis. Comparative mass spectrometry of palmitoylated proteins in wt and ΔZDHHC22 cells identified 25 potential substrates of ZDHHC22 which might be involved in virus replication.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Influenza A virus/physiology , Membrane Proteins/genetics , Up-Regulation , Viral Nonstructural Proteins/genetics , A549 Cells , Acylation , Animals , CRISPR-Cas Systems , Cell Line , Dogs , Gene Knockout Techniques , Humans , Madin Darby Canine Kidney Cells , Virus Replication
5.
Bio Protoc ; 10(4): e3523, 2020 Feb 20.
Article in English | MEDLINE | ID: mdl-33654747

ABSTRACT

Non-covalent binding of cholesterol to the transmembrane region of proteins affect their functionalities, but methods to prove such an interaction are rare. We describe our protocol to label the hemagglutinin (HA) of Influenza virus with a cholesterol derivative in living cells or with immunoprecipitated protein. We synthesized a "clickable" photocholesterol compound, which closely mimics authentic cholesterol. It contains a reactive diazirine group that can be activated by UV-illumination to form a covalent bond with amino acids in its vicinity. Incorporation of photocholesterol into HA is then visualized by "clicking" it to a fluorophore, which can be detected in an SDS-gel by fluorescence scanning. This method provides a convenient and practical way to demonstrate cholesterol-binding to other proteins and probably to identify the binding site.

6.
Expert Opin Drug Discov ; 15(2): 159-177, 2020 02.
Article in English | MEDLINE | ID: mdl-31809605

ABSTRACT

Introduction: S-acylation is the attachment of fatty acids not only to cysteines of cellular, but also of viral proteins. The modification is often crucial for the protein´s function and hence for virus replication. Transfer of fatty acids is mediated by one or several of the 23 members of the ZDHHC family of proteins. Since their genes are linked to various human diseases, they represent drug targets.Areas covered: The authors explore whether targeting acylation of viral proteins might be a strategy to combat viral diseases. Many human pathogens contain S-acylated proteins; the ZDHHCs involved in their acylation are currently identified. Based on the 3D structure of two ZDHHCs, the regulation and the biochemistry of the palmitolyation reaction and the lipid and protein substrate specificities are discussed. The authors then speculate how ZDHHCs might recognize S-acylated membrane proteins of Influenza virus.Expert opinion: Although many viral diseases can now be treated, the available drugs bind to viral proteins that rapidly mutate and become resistant. To develop inhibitors for the genetically more stable cellular ZDHHCs, their binding sites for viral substrates need to be identified. If only a few cellular proteins are recognized by the same binding site, development of specific inhibitors may have therapeutic potential.


Subject(s)
Acyltransferases/metabolism , Antiviral Agents/pharmacology , Virus Diseases/drug therapy , Acylation/physiology , Animals , Binding Sites , Drug Development , Fatty Acids/metabolism , Humans , Lipoylation/physiology , Viral Proteins/metabolism , Virus Diseases/enzymology , Virus Diseases/virology
7.
Biochem J ; 477(1): 285-303, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31872235

ABSTRACT

Hemagglutinin (HA), a glycoprotein of Influenza A viruses and its proton channel M2 are site-specifically modified with fatty acids. Whereas two cysteines in the short cytoplasmic tail of HA contain only palmitate, stearate is exclusively attached to one cysteine located at the cytoplasmic border of the transmembrane region (TMR). M2 is palmitoylated at a cysteine positioned in an amphiphilic helix near the TMR. The enzymes catalyzing acylation of HA and M2 have not been identified, but zinc finger DHHC domain-containing (ZDHHC) palmitoyltransferases are candidates. We used a siRNA library to knockdown expression of each of the 23 human ZDHHCs in HA-expressing HeLa cells. siRNAs against ZDHHC2 and 8 had the strongest effect on acylation of HA as demonstrated by Acyl-RAC and confirmed by 3H-palmitate labeling. CRISPR/Cas9 knockout of ZDHHC2 and 8 in HAP1 cells, but also of the phylogenetically related ZDHHCs 15 and 20 strongly reduced acylation of group 1 and group 2 HAs and of M2, but individual ZDHHCs exhibit slightly different substrate preferences. These ZDHHCs co-localize with HA at membranes of the exocytic pathway in a human lung cell line. ZDHHC2, 8, 15 and 20 are not required for acylation of the HA-esterase-fusion protein of Influenza C virus that contains only stearate at one transmembrane cysteine. Knockout of these ZDHHCs also did not compromise acylation of HA of Influenza B virus that contains two palmitoylated cysteines in its cytoplasmic tail. Results are discussed with respect to the acyl preferences and possible substrate recognition features of the identified ZDHHCs.


Subject(s)
Acyltransferases/metabolism , Gammainfluenzavirus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/metabolism , Influenza B virus/metabolism , Influenza, Human/virology , A549 Cells , Acylation , Animals , Dogs , HeLa Cells , Humans , Madin Darby Canine Kidney Cells
8.
J Microbiol Biotechnol ; 24(12): 1719-27, 2014 Dec 28.
Article in English | MEDLINE | ID: mdl-25112319

ABSTRACT

In the present study, whispovirus immediate early 1 promoter (ie-1) was used to initiate surface expression of the hemagglutinin (HA) protein of Egyptian H5N1 avian influenza virus (AIV) by using the baculovirus expression vector system. The HA gene and whispovirus ie-1 promoter sequence were synthesized as a fused expression cassette (ie1-HA) and successfully cloned into the pFastBac-1 transfer vector. The recombinant vector was transformed into DH10Bac competent cells, and the recombinant bacmid was generated via site-specific transposition. The recombinant bacmid was used for transfection of Spodoptera frugiperda (Sf-9) insect cells to construct the recombinant baculovirus and to induce expression of the HA protein of H5N1 AIV. The recombinant glycoprotein expressed in Sf-9 cells showed hemadsorption activity. Hemagglutination activity was also detected in both extra- and intracellular recombinant HAs. Both the HA and hemadsorption activities were inhibited by reference polyclonal anti-H5 sera. Significant expression of the recombinant protein was observed on the surface of infected insect cells by using immunofluorescence. SDS-PAGE analysis of the expressed protein revealed the presence of a visually distinguishable band of ~63 kDa in size, which was absent in the non-infected cell control. Western blot analysis confirmed that the distinct 63 kDa band corresponded to the recombinant HA glycoprotein of H5N1 AIV. This study reports the successful expression of the HA protein of H5N1 AIV. The expressed protein was displayed on the plasma membrane of infected insect cells under the control of whispovirus ie-1 promoter by using the baculovirus expression vector system.


Subject(s)
Cell Surface Display Techniques , Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Animals , Baculoviridae/genetics , Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/genetics , Nimaviridae/genetics , Promoter Regions, Genetic , Sf9 Cells , Spodoptera , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...