2016
DOI: 10.1128/jvi.01563-16
|View full text |Cite
|
Sign up to set email alerts
|

The Tetherin Antagonism of the Ebola Virus Glycoprotein Requires an Intact Receptor-Binding Domain and Can Be Blocked by GP1-Specific Antibodies

Abstract: The glycoprotein of Ebola virus (EBOV GP), a member of the family Filoviridae, facilitates viral entry into target cells. In addition, EBOV GP antagonizes the antiviral activity of the host cell protein tetherin, which may otherwise restrict EBOV release from infected cells. However, it is unclear how EBOV GP antagonizes tetherin, and it is unknown whether the GP of Lloviu virus (LLOV), a filovirus found in dead bats in Northern Spain, also counteracts tetherin. Here, we show that LLOV GP antagonizes tetherin,… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

2
31
0

Year Published

2017
2017
2021
2021

Publication Types

Select...
6
1

Relationship

2
5

Authors

Journals

citations
Cited by 21 publications
(33 citation statements)
references
References 63 publications
2
31
0
Order By: Relevance
“…We identified single amino acid residues in the RBD that are important for GP-virosome formation ( Figures 5A and 5B). Notably, the same residues have been previously demonstrated to be involved in GP-mediated entry and tetherin antagonism but were dispensable for GP virion incorporation and augmentation of VP40-driven VLP release (Brinkmann et al, 2016). A thus far unknown RBD-binding partner is supposed to be involved in GP-driven entry (Dube et al, 2008(Dube et al, , 2010 and tetherin counteraction (Brinkmann et al, 2016), and according to our results the same factor might also be involved in regulating GP-virosome release.…”
Section: Discussionsupporting
confidence: 75%
See 2 more Smart Citations
“…We identified single amino acid residues in the RBD that are important for GP-virosome formation ( Figures 5A and 5B). Notably, the same residues have been previously demonstrated to be involved in GP-mediated entry and tetherin antagonism but were dispensable for GP virion incorporation and augmentation of VP40-driven VLP release (Brinkmann et al, 2016). A thus far unknown RBD-binding partner is supposed to be involved in GP-driven entry (Dube et al, 2008(Dube et al, , 2010 and tetherin counteraction (Brinkmann et al, 2016), and according to our results the same factor might also be involved in regulating GP-virosome release.…”
Section: Discussionsupporting
confidence: 75%
“…Fruit bat tetherins, Epo and Hyp tetherin, where cloned from fruit bat cell lines EpoNi/22.1 and HypNi/1.1 (Hoffmann et al, 2013), respectively, and sequences have been submitted to GenBank under accession numbers MG792836 and MG792837. The plasmids coding for the GP RBD mutants (F88A, L111A, L122A, W104A), GP LXXXL, LASV-GPC, ELE and LEL GP chimera, wild-type GP as well as the GP A82V mutant derived from the 2014 Ebola outbreak in West Africa are described elsewhere (Brinkmann et al, 2016;Gnirß et al, 2014;Hacke et al, 2015;Hoffmann et al, 2017). The GP-coding sequences of those mutants were amplified by PCR (forward 5 0 -CGTCTAGAATATGGGTGTTACAGGAATATTGC-3 0 and reverse 5 0 -TACGCGTTTCTAAAA GACAAATTTGCATATACAG-3 0 ) and subsequently cloned into the pCG-IRES-GFP vector using the restriction enzymes MluI and XbaI.…”
Section: Methods Detailsmentioning
confidence: 99%
See 1 more Smart Citation
“…Plasmids, mutagenesis, and transfection. Expression plasmids for HIV-1 p55-Gag (Gag), HIV-1 Vpu (Vpu), EBOV-GP, VSV glycoprotein (VSV-G), Nipah virus fusion (F), and attachment glycoprotein (G), EBOV-VP40 harboring an N-terminal cMYC tag, and DC-SIGN have been described elsewhere (35,44,(48)(49)(50). To generate expression plasmids for human, gorilla, African green monkey, pig, rat, mouse, and artificial tetherin, the respective ORFs were amplified from existing plasmids (32,51,52) and inserted into the pCAGGS expression vector.…”
Section: Cells and Viruses Hek293t (Human Kidney) And A549 (Human mentioning
confidence: 99%
“…As mentioned above, HIV-1 Vpu is the prototype antagonist of BST2 (2,3); Vpu mistraffics BST2 away from the plasma membrane, removes it from virion assembly sites, and targets it for degradation (13,(16)(17)(18)(19)(20)(21). On the other hand, the Ebola virus envelope glycoprotein (GP 1,2 ) antagonizes BST2 to enhance virion release through an unclear mechanism that requires neither the removal of BST2 from the cell surface nor its degradation but might instead involve dissociation of BST2 from the Ebola virus matrix protein, VP40; this activity requires proper N-linked glycosylation of the GP1 subunit and the interaction of BST2 with the GP2 subunit (12,(22)(23)(24)(25)(26)(27)(28).…”
Section: Abstract Bst2 Ebola Virus Nf-b Vp40mentioning
confidence: 99%