HIV-1 Vpu is a potent transcriptional suppressor of NF-κB-elicited antiviral immune responses

  1. Simon Langer
  2. Christian Hammer
  3. Kristina Hopfensperger
  4. Lukas Klein
  5. Dominik Hotter
  6. Paul D De Jesus
  7. Kristina M Herbert
  8. Lars Pache
  9. Nikaïa Smith
  10. Johannes A van der Merwe
  11. Sumit K Chanda
  12. Jacques Fellay
  13. Frank Kirchhoff
  14. Daniel Sauter  Is a corresponding author
  1. Ulm University Medical Center, Germany
  2. Sanford Burnham Prebys Medical Discovery Institute, United States
  3. École Polytechnique Fédérale de Lausanne, Switzerland
  4. Swiss Institute of Bioinformatics, Switzerland
4 figures, 1 table and 5 additional files

Figures

Figure 1 with 2 supplements
Generation of Vpu mutants that fail to inhibit NF-κB activation or to counteract tetherin.

(A) Vpu-mediated inhibition of NF-κB activation via two independent mechanisms. Asterisks illustrate mutations in Vpu that were introduced to selectively abrogate tetherin counteraction (orange) or inhibition of NF-κB activation downstream of tetherin (blue). (B) Wt and mutant HIV-1 clones used in this study. MSM, man having sex with men; WSM, woman having sex with men. (C) Vpu-mediated inhibition of NF-κB activation. HEK293T cells were co-transfected with the indicated proviral constructs, a firefly luciferase-based NF-κB reporter vector, a Gaussia luciferase construct for normalization, and an expression vector for a constitutively active mutant of IKKβ as NF-κB inducer. Two days post-transfection, luciferase activity was determined. Mean values of three to seven independent experiments, each performed in triplicate ±SEM are shown (*p<0.05; **p<0.01; RM one-way ANOVA with Greenhouse-Geisser correction and Dunnett’s multiple comparison test). (D) Vpu-mediated down-modulation of tetherin. Human PBMCs were infected with the indicated VSV-G pseudotyped HIV-1 strains. Three days post-infection, tetherin surface levels of p24 positive cells were determined by flow cytometry. Mean values of three to five independent experiments ± SEM are shown (*p<0.05; **p<0.01; ***p<0.001; RM one-way ANOVA with Greenhouse-Geisser correction and Dunnett’s multiple comparison test). (E) Vpu-mediated enhancement of infectious virus yield. HEK293T cells were co-transfected with the indicated proviral constructs and increasing amounts of an expression plasmid for human tetherin. Two days post-transfection, infectious virus yield was determined by infection of TZM-bl reporter cells. Mean values of three to four independent experiments ± SEM are shown. (F) Expression of Vpu. HEK293T cells were transfected with the indicated proviral constructs. Two days post-transfection, cells were harvested and analyzed by Western Blotting.

https://doi.org/10.7554/eLife.41930.003
Figure 1—figure supplement 1
Vpu alignment and Vpu-mediated inhibition of nuclear translocation of p65.

(A) Amino acid sequence alignment of CH293, CH077 and STCO1 Vpu. Residues that were mutated to abrogate the anti-tetherin activity, ability to inhibit NF-κB activation or expression of Vpu are highlighted in orange, blue and green, respectively. Phosphorylated serine residues are shown in grey. (B) HEK293T cells were transfected with the indicated proviral constructs or an empty vector control. Two days post-transfection, cells were stimulated with 5 ng/ml TNFα for 15 min before fixation and staining for intracellular p65 (red), HIV-1 p24/Gag (green) and DNA (blue). Exemplary images are shown on top (scale bar: 10 µm). Arrows indicate exemplary cells containing high and low levels of cytoplasmic p65 in cells expressing wild type and vpu-deficient HIV-1, respectively. To quantify nuclear translocation, the average p65 signal intensity in the nucleus was determined and divided by the p65 signal intensity in the cytoplasm. The bottom panel shows the quantification of 475 cells from one of two independent experiments. Mean values ± SEM are shown (*p<0.05; **p<0.01; ***p<0.001; ordinary one-way ANOVA with Dunnett’s multiple comparison test).

https://doi.org/10.7554/eLife.41930.004
Figure 1—figure supplement 2
Vpu-mediated counteraction of tetherin.

(A) Human PBMCs were infected with the indicated VSV-G pseudotyped HIV-1 strains. Three days post-infection, tetherin surface levels of p24 positive cells were determined by flow cytometry. Exemplary primary data of the results shown in Figure 1D illustrating the gating strategy are shown. The mean fluorescence intensity of tetherin surface staining and the percentage of infected cells are shown in black and blue, respectively. (B) Area under the curve (AUC) of the data shown in Figure 1E was calculated. Mean values of three to four independent experiments ± SEM are shown (*p<0.05; **p<0.01; ***p<0.001; ordinary one-way ANOVA with Dunnett’s multiple comparison test).

https://doi.org/10.7554/eLife.41930.005
Figure 2 with 1 supplement
RNA sequencing of CD4 +T cells infected with wt or vpu-mutated HIV-1.

(A) Experimental setup. CD4 +T cells of four healthy donors were infected with wt or vpu-mutated HIV-1. Three days post-infection, cells were harvested and RNA sequencing was performed. (B) Principal component analysis demonstrating that samples are clustered by donor (A - D) and virus (CH077, CH293, STCO1) rather than the vpu genotype. (C) MA plot providing a global view of differentially expressed genes between Vpu wt and mock infected samples (combined analysis of CH077, CH293, and STCO1). Red dots represent significant genes with a q value < 0.1 after Benjamini-Hochberg correction. (D) KEGG pathways significantly down- or up-regulated upon infection with HIV-1 wt (combined analysis of CH077, CH293, and STCO1) (* q < 0.1; ** q < 0.01; *** q < 0.001). HIV-1 suppresses pathways involved in transcription/translation (red), cell division (orange) and energy metabolism (purple), but induces immune responses (yellow), signaling cascades (green) and pathways involved in cell-to-cell interactions and motility (blue). Pathway analysis was performed using GAGE (Luo et al., 2009).

https://doi.org/10.7554/eLife.41930.006
Figure 2—figure supplement 1
Isolation, infection and RNA sequencing of CD4 +T cells.

(A) Purity of isolated CD4 +T cells. After negative isolation, cells were stained for surface CD4 and CD11c and analyzed by flow cytometry. In the left panel, the percentage of CD4+ (CD11c-) T cells for donors A, B, C and D is shown. An exemplary dot plot of donor D illustrating the gating strategy is shown on the right. (B) Number of reads per sample, unmapped and mapped to the human genome. (C) Cook’s distances of all 52 samples (see Supplementary file 1 for sample identity). (D) Heatmap of the sample distance matrix, showing hierarchical clustering of sample-to-sample distances. (E) Infection rates of CD4 +T cells used for RNA sequencing. CD4 +T cells were infected with the indicated VSV-G pseudotyped HIV-1 strains. Three days post-infection, infection rates were determined by intracellular staining for p24 and subsequent flow cytometric analysis. (F) Venn diagram illustrating the overlap of differentially expressed genes (q < 0.1) for the three viruses analyzed (HIV-1 wt infected vs. mock infected control).

https://doi.org/10.7554/eLife.41930.007
Figure 3 with 1 supplement
Vpu suppresses the induction of NF-κB-dependent immune responses.

(A) Volcano plots illustrating differentially expressed genes (red) in vpu stop vs. wt (left panel) or NF-κB-def. Vpu mutant vs. wt (right panel) infected CD4 +T cells. (B) Pie charts illustrating the relative size of gene sets that are targeted by a specific transcription factor. Genes whose expression is significantly increased or decreased upon loss of vpu were analyzed in the left and right panel, respectively. NF-κB target gene sets are highlighted in pink. (C) Pie charts illustrating the percentage of ISGs among different gene sets. The analysis is based on a list of ISGs obtained through the interferome v2.1 database (Rusinova et al., 2013) (***p<0.001; *p<0.05; Fisher’s exact test). (D) Top10 KEGG and GO pathways that are induced upon complete loss of Vpu (left panel) or selective loss of Vpu-mediated NF-κB inhibition (right panel). Modulation of two custom-defined gene sets (i.e. Restriction factors and NF-κB target genes) is shown at the bottom. Pathways involved in intrinsic, innate or adaptive immunity are highlighted in yellow. Pathway analysis was performed using GAGE (Luo et al., 2009) (* q < 0.1; ** q < 0.01; *** q < 0.001). (E) n-fold modulation of host restriction factor and IFN-β mRNA levels upon loss of Vpu. Mean values of four independent infections ± SEM are shown. Statistical significance was determined using negative binomial generalized linear models as implemented in DESeq2 (Love et al., 2014) (* q < 0.1; ** q < 0.01; *** q < 0.001). The dotted line indicates the mean change of restriction factor expression upon loss of Vpu.

https://doi.org/10.7554/eLife.41930.008
Figure 3—figure supplement 1
Modulation of NF-κB-dependent gene expression by Vpu.

(A) MA plots illustrating the number of differentially expressed genes (red) between each vpu mutant and wt, using a P value threshold of 0.1 after Benjamini-Hochberg correction. (B) Pie charts illustrating the percentage of known NF-κB target genes among different gene sets. The analysis is based on a list of NF-κB inducible genes obtained through the NF-κB target gene database (70) (***p<0.001; n.s. not significant; Fisher’s exact test). (C) Venn diagram illustrating the overlap of NF-κB inducible genes (70) and genes, whose expression is suppressed by Vpu. (D) Genes that are differentially expressed in vpu stop vs. wt (top panel) or NF-κB-def. Vpu mutant vs. wt (bottom panel) infected CD4 +T cells. NF-κB inducible genes (70) and ISGs (Rusinova et al., 2013) are highlighted in pink and purple, respectively. Statistical significance was determined using negative binomial generalized linear models as implemented in DESeq2 (* q < 0.1; ** q < 0.01; *** q < 0.001) (Love et al., 2014). Arrows highlight previously identified targets of Vpu.

https://doi.org/10.7554/eLife.41930.009
Figure 4 with 1 supplement
Vpu suppresses pro-inflammatory cytokine production.

(A) Vpu inhibits IFN-β expression. CD4 +T cells were infected with the indicated VSV-G pseudotyped HIV-1 strains. Three days post-infection, IFN-β mRNA levels were quantified by qRT-PCR and normalized to the respective mock control. In the top panels, mean values of three to four independent experiments ± SEM are shown. The bottom panels show correlation analyses of IFN-β mRNA levels and NF-κB activity (determined in Figure 1C) (*p<0.05; RM one-way ANOVA with Greenhouse-Geisser correction and Dunnett’s multiple comparison test). (B) Vpu suppresses release of CXCL10 and IL-6. CD4 +T cells were infected with the indicated VSV-G pseudotyped HIV-1 STCO1 mutants. Three days post-infection, cytokine concentrations in the cell culture supernatant were quantified by flow cytometry using a bead-based immunoassay, in which the analytes are captured between two antibodies. In the top panels, each data point represents results obtained with one of three donors, normalized to the wild type control (100%). Error bars indicate SEM. Representative primary data of one donor are shown in the bottom panels. Numbers indicate mean fluorescence intensity. (C) Vpu exerts broad suppressive effects on cytokine release. CD4 +T cells were infected with the indicated VSV-G pseudotyped HIV-1 STCO1 mutants. Three days post-infection, cytokine release was determined using a cytokine array, based on antibody-coated membranes. The heat map on the left illustrates relative cytokine release and is based on the mean of three independent experiments. Cytokines that were not detectable in more than one donor were omitted from the heat map. Known NF-κB inducible genes (70) are shown in pink. Arrows indicate CXCL10 and IL-6 that were also analyzed in panel (B). Mean changes ± SEM in cytokine release relative to HIV-1 wt are shown in the right panel. (**p<0.01; ***p<0.001; RM one-way ANOVA with Greenhouse-Geisser correction and Dunnett’s multiple comparison test).

https://doi.org/10.7554/eLife.41930.010
Figure 4—figure supplement 1
Infection rates of CD4 +T cells.

(A) Infection rates of donors analyzed in Figure 4A. (B) Infection rates of donors analyzed in Figure 4B and C. Human CD4 +T cells were infected with the indicated VSV-G pseudotyped HIV-1 strains. Three days post-infection, infection rates were determined by intracellular staining for p24 and subsequent flow cytometric analysis. Horizontal lines indicate mean values of three to four donors.

https://doi.org/10.7554/eLife.41930.011

Tables

Key resources table
Reagent type
(species)
or resource
DesignationSource or referenceIdentifiersAdditional information
Gene (H. sapiens)Custom-defined gene
set of NF-κB target
genes
see
Supplementary file 2
of this paper
N/A
Gene (H. sapiens)Custom-defined
gene set
of host restriction
factors
see
Supplementary file 3
of this paper
N/A
Strain, strain
background (E. coli)
XL-2 blueStratageneCat#200150
Strain, strain
background
(E. coli)
XL2-Blue MRF’ TM
Ultracompetent cells
Agilent TechnologiesCat#200151
Cell line
(H. sapiens)
HEK293T, femaleATCCCat#CRL-3216;
RRID: CVCL_0063
Cell line
(H. sapiens)
TZM-bl, femaleNIHCat#8129;
RRID: CVCL_B478
Biological
sample
(H. sapiens)
peripheral blood
mononuclear
cells (donors A
and C: male;
donors B and
D: female)
DRK-Blutspendedienst
Baden-Württemberg-Hessen,
Ulm, Germany
N/Aderived from four
healthy donors
Antibodyanti-HIV-1 p24 (capsid
protein), FITC-conjugated
(mouse monoclonal)
Beckman CoulterCat#6604665;
RRID: AB_1575987
FACS (1:25)
Antibodyanti-BST-2/tetherin, APC-conjugated
(mouse monoclonal)
BiolegendCat#348410;
RRID:AB_2067121
FACS (1:20)
Antibodyanti-Human CD4, PerCP-conjugated
(mouse monoclonal)
BD PharmingenCat#550631;
RRID:AB_393791
FACS (1:12.5)
Antibodyanti-CD11c [3.9], FITC-conjugated
(mouse monoclonal)
AbcamCat#ab82445-100;
RRID:AB_1859733
FACS (1:12.5)
Antibodyanti-CD11c [BU15], FITC-conjugated (mouse monoclonal)AbcamCat#ab22540FACS (1:5)
AntibodyIgG1, κ, APC-conjugated (mouse)BiolegendCat#400122;
RRID:AB_326443
FACS (1:20)
AntibodyHIV-1 subtype C serumNIHCat#11942WB (1:5000)
AntibodyHIV-1 NL4-3 Vpu antiserumN/AN/AWB (1:500);
kindly provided
by S. Bolduan
AntibodyAnti-HIV1 p24 antibody [39/5.4A]AbcamCat#ab9071;
RRID:AB_306981
WB (1:5000)
AntibodyPurified anti-GAPDH
antibody
BiolegendCat#607902;
RRID:AB_2734503
WB (1:1000)
AntibodyIRDye 800CW Goat anti-Mouse
IgG (H + L)
LI-CORCat#926–32210;
RRID:AB_621842
WB (1:20000)
AntibodyIRDye 680RD Goat anti-Rabbit
IgG (H + L)
LI-CORCat#925–68071;
RRID:AB_2721181
WB (1:20000)
AntibodyIRDye 800CW Goat anti-Rat
IgG (H + L)
LI-CORCat#925–32219;
RRID:AB_2721932
WB (1:20000)
Antibodyanti-HIV-1 p24 (mouse monoclonal)AbcamCat#ab9071;
RRID:AB_306981
IF (1:500)
Antibodyanti-p65
(rabbit polyclonal IgG)
Santa CruzCat#sc-372;
RRID:AB_632037
IF (1:150)
AntibodyDonkey anti-Mouse
IgG (H + L), AF647-
conjugated
Thermo ScientificCat#A31571;
RRID:AB_162542
IF (1:350)
AntibodyGoat anti-Rabbit
IgG (H + L), AF568-
conjugated
Thermo ScientificCat#A11011;
RRID:AB_143157
IF (1:350)
Recombinant
DNA reagent
pBR322_HIV-1
M subtype
B STCO1 wt (plasmid)
PMID: 23542380N/Akindly provided
by B. Hahn
Recombinant
DNA reagent
pBR322_HIV-1 M subtype B STCO1 vpu
stop (plasmid)
PMID: 27531907N/A
Recombinant
DNA reagent
pBR322_HIV-1 M subtype B STCO1 Vpu
R45K (plasmid)
PMID: 29324226N/A
Recombinant
DNA reagent
pBR322_HIV-1 M subtype B STCO1 Vpu
A15L/A19L (plasmid)
PMID: 27531907N/A
Recombinant
DNA reagent
pUC57rev_HIV-1 M subtype C CH293 w8
wt (plasmid)
PMID: 23542380N/Akindly provided by B. Hahn
Recombinant
DNA reagent
pUC57rev_HIV-1 M subtype C CH293 w8
vpu stop (plasmid)
PMID: 2562070N/A
Recombinant
DNA reagent
pUC57rev_HIV-1 M subtype C CH293
w8 Vpu
R50K (plasmid)
this paperN/Aderived from pUC57rev_HIV-1 M subtype C
CH293 w8 wt (plasmid)
Recombinant
DNA reagent
pUC57rev_HIV-1
M subtype C CH293 w8 Vpu A20L/A24L (plasmid)
this paperN/Aderived from pUC57rev_HIV-1 M subtype C CH293 w8 wt (plasmid)
Recombinant
DNA reagent
pCR-XL-TOPO_HIV-1 M subtype B CH077 wt
(plasmid)
PMID: 22190722N/Akindly provided by B. Hahn
Recombinant
DNA reagent
pCR-XL-TOPO_HIV-1 M subtype B CH077
vpu stop
(plasmid)
PMID: 27531907N/A
Recombinant
DNA reagent
pCR-XL-TOPO_HIV-1 M subtype B CH077
Vpu R45K
(plasmid)
this paperN/Aderived from pCR-XL-TOPO_HIV-1 M subtype B CH077 wt (plasmid)
Recombinant
DNA reagent
pCR-XL-TOPO_HIV-1 M subtype B CH077
Vpu A15L/A19L
(plasmid)
PMID: 27531907N/A
Recombinant
DNA reagent
p_human IKKβ,
constitutively active mutant (S177E, S181E) (plasmid)
PMID: 23552418N/Akindly provided by B. Baumann
Recombinant
DNA reagent
p_NF-κB(3x)-Firefly Luciferase (plasmid)PMID: 23552418N/Akindly provided by B. Baumann
Recombinant
DNA reagent
pTAL_Gaussia Luciferase (plasmid)PMID: 23552418N/A
Recombinant
DNA reagent
pCG_human Tetherin IRES DsRed2 (plasmid)PMID: 19917496N/A
Recombinant
DNA reagent
pHIT-G_VSV-G (vesicular stomatitis virus glycoprotein) (plasmid)PMID: 9303297N/A
Sequence-
based reagent
Primers used for mutagenesis of vpusee
Supplementary file 4
of this paper
N/A
Sequence-
based reagent
TaqMan Gene
Expression Assay for IFN-beta
Thermo Fisher
Scientific
Cat#Hs01077958_s1
Sequence-
based reagent
Human GAPD (GAPDH) Endogenous Control (VIC/TAMRA probe, primer limited)Thermo Fisher
Scientific
Cat#4310884E
Peptide,
recombinant
protein
IRDye 800CW
streptavidin, 0.5 mg
LI-CORCat#926–32230
Commercial
assay or kit
GalScreenApplied BioscienceCat#T1027
Commercial
assay or kit
PrimeScript
RT-PCR Kit
TAKARACat#RR014A
Commercial
assay or kit
RNeasy Plus
Mini kit
QIAGENCat#74136
Commercial
assay or kit
RosetteSep
Human CD4 + T Cell
Enrichment
Cocktail
Stem Cell
Technologies
Cat#15062
Commercial
assay or kit
Luciferase Assay
System 10-pack
PromegaCat#E1501
Commercial
assay or kit
LEGENDplex Human Anti-Virus Response
Panel
BioLegendCat#740390
Commercial
assay or kit
RayBio C-Series
Human Cytokine Antibody
Array C5
RayBiotechCat#AAH-CYT-5–8
Commercial
assay or kit
FIX and PERM Kit
(CE-IVD) (1000 Tests)
Nordic-MUbioCat#GAS-002–1
Commercial
assay or kit
DNA-free
DNA Removal Kit
ThermoFisher
Scientific
Cat#AM1906
Commercial
assay or kit
QuikChange II XL Site-Directed Mutagenesis
Kit
AgilentCat#200522
Commercial
assay or kit
TruSeq Stranded
mRNA Sample Prep Kit
illuminaCat#RS-122–2101
Commercial
assay or kit
Thermo Script
RT-PCR system
InvitrogenCat#11146–016
Commercial
assay or kit
Signal Enhancer
HIKARI kit for Western
Blotting and ELISA
Nacalai TesqueCat#02267–41
Chemical
compound, drug
Human IL-2 IS,
premium grade
MACS Miltenyi
Biotec
Cat#130-097-745
Chemical
compound, drug
Remel PHA purifiedThermoFisher
Scientific
Cat#R30852801
Chemical
compound, drug
Recombinant
human TNFα
Sigma AldrichCat#H8916-10UG
Chemical
compound, drug
DAPISigma AldrichCat#D9542-1MG
Software,
algorithm
BD FACSDiva
Version 8.0
BD Bioscienceshttps://www.bdbiosciences.com; RRID: SCR_001456
Software,
algorithm
Corel DRAW 2017Corel Corporationhttps://www.coreldraw.com/
Software,
algorithm
GraphPad Prism
Version 5.03
GraphPad Software, Inc.https://www.graphpad.com; RRID: SCR_002798
Software,
algorithm
ImageJOpen sourcehttp://imagej.nih.gov/ij/
Software,
algorithm
LI-COR Image Studio Lite Version 3.1LI-CORwww.licor.com/; RRID: SCR_013715
Software,
algorithm
FlowJo_V10Tree Star, Inc.https://www.flowjo.com
Software,
algorithm
ZENZeisshttps://www.zeiss.com/microscopy/int/products/microscope-software/zen.html; RRID:SCR_013672
Software,
algorithm
FijiOpen source
(Max Planck
Institute of Molecular
Cell Biology and
genetics,
Dresden, Germany)
https://imagej.net/Fiji; RRID:SCR_002285
Software,
algorithm
GeneMANIAOpen sourcehttp://genemania.org/
Software,
algorithm
HeatmapperOpen sourcehttp://www2.heatmapper.ca/expression/
Software,
algorithm
ROpen sourcehttps://www.r-project.org/
Software,
algorithm
KallistoPMID: 27043002https://pachterlab.github.io/kallisto/
Software,
algorithm
DESeq2PMID: 25516281https://bioconductor.org/packages/release/bioc/html/DESeq2.html
Software,
algorithm
GAGEPMID: 19473525https://bioconductor.org/packages/release/bioc/html/gage.html
Software,
algorithm
StepOne and StepOnePlus Software
v2.3
Thermo Fisher
Scientific
https://www.thermofisher.com/de/de/home/technical-resources/software-downloads/StepOne-and-StepOnePlus-Real-Time-PCR-System.html
OtherRel/NF-κB target
gene database
Open sourcehttp://bioinfo.lifl.fr/NF-KB/
OtherRNAseq Datathis paperGEO accession GSE117655
OtherInterferome
database
PMID: 23203888http://www.interferome.org/interferome/home.jspx

Additional files

Supplementary file 1

Identity and integrity of RNA-Seq samples.

https://doi.org/10.7554/eLife.41930.012
Supplementary file 2

Custom-defined gene set of NF-κB target genes.

https://doi.org/10.7554/eLife.41930.013
Supplementary file 3

Custom-defined gene set of host restriction factors.

https://doi.org/10.7554/eLife.41930.014
Supplementary file 4

Primers used for mutagenesis of vpu

https://doi.org/10.7554/eLife.41930.015
Transparent reporting form
https://doi.org/10.7554/eLife.41930.016

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Simon Langer
  2. Christian Hammer
  3. Kristina Hopfensperger
  4. Lukas Klein
  5. Dominik Hotter
  6. Paul D De Jesus
  7. Kristina M Herbert
  8. Lars Pache
  9. Nikaïa Smith
  10. Johannes A van der Merwe
  11. Sumit K Chanda
  12. Jacques Fellay
  13. Frank Kirchhoff
  14. Daniel Sauter
(2019)
HIV-1 Vpu is a potent transcriptional suppressor of NF-κB-elicited antiviral immune responses
eLife 8:e41930.
https://doi.org/10.7554/eLife.41930