Next Article in Journal
Initial Establishment of Warning Model for Epidemic Intensity of Norovirus GII Associated with Acute Gastroenteritis in Beijing Based on Synthetic Index Method
Previous Article in Journal
HTLV-1 p13 Protein Hijacks Macrophage Polarization and Promotes T-Cell Recruitment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Analysis of Factors That Regulate HIV-1 Fusion in Reverse

Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(4), 472; https://doi.org/10.3390/v17040472
Submission received: 12 February 2025 / Revised: 24 March 2025 / Accepted: 25 March 2025 / Published: 26 March 2025
(This article belongs to the Section Animal Viruses)

Abstract

:
Based on observations that HIV-1 envelope (Env) proteins on the surfaces of cells have the capacity to fuse with neighboring cells or enveloped viruses that express CD4 receptors and CXCR4 co-receptors, we tested factors that affect the capacities of lentiviral vectors pseudotyped with CD4 and CXCR4 variants to infect Env-expressing cells. The process, which we refer to as fusion in reverse, involves the binding and activation of cellular Env proteins to fuse membranes with lentiviruses carrying CD4 and CXCR4 proteins. We have found that infection via fusion in reverse depends on cell surface Env levels, is inhibitable by an HIV-1-specific fusion inhibitor, and preferentially requires lentiviral pseudotyping with a glycosylphosphatidylinositol (GPI)-anchored CD4 variant and a cytoplasmic tail-truncated CXCR4 protein. We have demonstrated that latently HIV-1-infected cells can be specifically infected using this mechanism, and that activation of latently infected cells increases infection efficiency. The fusion in reverse approach allowed us to characterize how alteration of CD4 plus CXCR4 lipid membranes affected Env protein activities. In particular, we found that perturbation of membrane cholesterol levels did not affect Env activity. In contrast, viruses assembled in cells deficient for long-chain sphingolipids showed increased infectivities, while viruses that incorporated a lipid scramblase were non-infectious. Our results yield new insights into factors that influence envelope protein functions.

1. Introduction

The trafficking of wild type (WT) HIV-1 envelope (Env) proteins to viral assembly sites at the plasma membranes (PMs) of infected cells is a complicated process; it involves travel via a vesicular transport pathway, trimerization enroute, and processing in the Golgi by furin or a furin-like protease into their surface (SU; gp120) and transmembrane (TM; gp41) components [1,2,3,4,5,6,7,8,9]. Evidence also indicates that because their cytoplasmic tails (CTs) carry YxxL and LL internalization signals, they can be internalized into endosomes where they may be shunted off for lysosomal degradation or returned to the PM [1,2,3,4,5,6,7,8,9]. The result of this is a reduced number of WT Env trimers at the PMs of infected cells, and surprisingly few WT Env proteins are incorporated into virus particles [1,10,11,12]. In this regard, it is noteworthy that some Env variants with CT truncations are present in PMs and virus particles at significantly higher levels than WT proteins, and one of these (which we refer to as 753Stop) still retains its YxxL internalization signal [11,12,13].
Once assembled into virions, Env proteins must operate in a lipid membrane environment that is considerably different than most PM regions [14,15,16,17,18,19,20,21,22,23,24]. In addition to phosphatidylinositol-(4,5)-bisphosphate (PI(4,5)P2), which is the binding partner of the HIV-1 precursor Gag (PrGag) matrix (MA) domain, viral membranes are highly enriched in phosphatidylserine (PS), phosphatidylethanolamine (PE), saturated phosphatidylcholines (PCs), cholesterol, and sphingolipids (SLs), including ceramide (Cer), sphingomyelin (SM), and hexosylceramides (HEX) [14,15,16]. The importance of this lipid composition is underscored by the effects of tampering with it. In particular, we have shown that the production of viruses in ceramide synthase 2 knockout (CerS2−/−) cells (which reduces the levels of long-chain SLs) impairs the fusion functions of both WT and CT deletion (ΔCT) Env proteins [25]. We and others have also demonstrated that treatment of HIV-1 virions with the cholesterol-binding compound amphotericin B methyl ester (AME) dramatically reduces the fusion activity of WT, but not ΔCT viruses [13,26,27,28]. Finally, the Env fusion function is also dependent on the membrane leaflet lipid asymmetry in virions, and scramblase incorporation into HIV-1 virions blocks viral infection [29,30,31,32,33,34].
Interestingly, the Env receptor-binding and fusion functions can be active in the PMs of Env-expressing cells [35,36,37,38,39]. Notably, when Env-expressing cells are in the presence of cells that carry the HIV-1 Env CD4 receptor plus a co-receptor (CXCR4 or CCR5), the combination can lead to cell–cell fusion and syncytia formation [35,36,37,38,39]. Similarly, enveloped viruses carrying CD4 and a co-receptor have been shown to infect Env-expressing cells in a process we refer to as fusion in reverse [40,41,42,43,44,45,46,47]. Recently, using cells expressing a different viral surface protein, the Spike protein of SARS-CoV-2, we demonstrated that lentiviruses decorated with a Spike-binding nanobody could specifically infect SARS-CoV-2-infected cells via a fusion in reverse mechanism [48]. Based on these results, we reasoned that it might be possible to infect HIV-1 Env-expressing cells in a related fashion.
We now report the ability to specifically infect HIV-1 Env-expressing cells with lentiviral vectors via a fusion in reverse mechanism. The optimized vectors utilize HIV-1 Gag and Pol proteins, and employ a C-terminally truncated CXCR4 protein, as well as a CD4 variant that is anchored to membranes by virtue of glycosylphosphatidylinositol (GPI) tail [48,49,50,51]. The lentiviruses preferentially infect HIV-1 infected cells, or cells independently expressing HIV-1 Env, and infection efficiency is dependent on Env surface expression levels. Using this fusion in reverse approach, it was possible to assess how lipid variations in membranes containing the HIV-1 receptor and co-receptor affected the HIV-1 Env activity. Interestingly, we found that reducing the leaflet asymmetry of viral membranes containing receptors plus co-receptors effectively inhibited infection, that reduction of long-chain SLs increased infectivity, and that AME treatment of receptor-containing membranes did not impair the activity of PM Env. We believe that this system will be useful in the analysis of other factors involved in Env-mediated membrane fusion, and may reinvigorate efforts for directly killing HIV-1-infected cells, or delivering antiviral or immunomodulatory genes to them [40,41,42,43,44,45,46,47].

2. Materials and Methods

2.1. Recombinant DNA Constructs

The lentivirus GagPol packaging plasmid (psPAX2), the GFP expression plasmids (GFP-PS, GFP-NoPS, GFP-C1-PLCdelta-PH, pEGFP-GFP11-ClathrinLightChain, pcDNA3.1-GFP [1,2,3,4,5,6,7,8,9,10]), the VSV G expression vector pMD.G, the CD4 expression plasmid pCAGGS-CD4-Myc, and the CXCR4 expression plasmid pcDNA3-CXCR4 were all obtained from Addgene, and were the gifts of Drs. Didier Trono, Jennifer Doudna, Tobias Meyer, Bo Huang, Simon Davis, Erik Procko, and Jacob Yount [48,49,50,51,52,53,54]. The TMEM16F scramblase expression vector, mTMEM-GY, was kindly provided by Dr. Walther Mothes [34]. The mammalian expression plasmid pcDNA3.1 (Invitrogen), the HIV-1 envelope expression vector pSVIII-Env (from the NIH HIV Reagent Program), the pLVX-puro-Xho-ATG-β-gal β-galactosidase lentivirus vector, and the wild type (WT) and 753Stop HIV-1 NL4-3 proviral constructs have been described previously [13,55,56,57]. The pMDG-NL43-Env-WT expression vector was created from the pMD.G backbone [52]. The pMD.G EcoRI site at nt 3774 was first killed by insertion of an A residue after nt 3775, giving the sequence 5′ tctagaaattc 3′, where the 5′ nt corresponds to pMD.G nt 3771, the 3′ nt corresponds to nt 3880, and the inserted nt is in bold. After that, the VSV G-encoding EcoRI fragment was replaced with an EcoRI fragment containing the NL4-3 envelope protein-coding region from the NL4-3 nt 6153 SspI site to the nt 8887 XhoI site. The 5′ juncture sequence is 5′ gaattcaagc ttgtcgacct gcaggcatgc aagcttcaga tt 3′, where the EcoRI site is italicized and NL4-3 nt 6156 is in bold. The 3′ juncture sequence is 5′ ctcgag gaattc 3′, where the NL4-3 XhoI site is in bold and the EcoRI site is italicized. The pMDG-NL43-Env-753Stop construct is identical to pMDG-NL43-Env-WT, except that it carries a termination codon after the HIV-1 Env residue 753, yielding the reading frame between NL4-3 residues 8465 and 8476 5′ gga tcc TAA gca 3′ [57]. The pCAGGS-CD4-GPI construct was generated from pCAGGS-CD4-Myc by replacement of the CD4 transmembrane and cytoplasmic domains with the GPI anchor from the human placental alkaline phosphatase (PLAP) protein [55,58,59]. To do so, CD4 was truncated at codon 393 and fused to the AscI-XhoI PLAP GPI fragment described in the construction of the previously described pMDG-Nano-GPI plasmid [55]. The reading frame at the CD4-GPI juncture is as follows, where the bold codon indicates CD4 codon 393: 5′ tgg tcc acc ccg gcg cgc cag 3′. For generation of the pcDNA3-CXCR4-309Stop and pcDNA3-CXCR4-318Stop cytoplasmic truncations of CXCR4, the parental pcDNA-CXCR4 plasmid was modified. The reading frame at the CXCR4 C-terminus of pcDNA3-CXCR4-309Stop is 5′ gcc aaa ttc TGA ggc ctc gag 3′, where the codon for residue 309 is in bold, and the stop codon is capitalized. The reading frame at the CXCR4 C-terminus of pcDNA3-CXCR4-318Stop is 5′ ctc acc TGA ccc ggg ctc gag 3′, where the codon for residue 318 is in bold, and the stop codon is capitalized. The CXCR4 variants with C-terminal fusions to the phospholipase C delta pleckstrin homology domain (PLCδPH) were based on a variant of GFP-C1-PLCdelta-PH [49,60,61] in which the GFP N-terminal coding region from the NheI site to the BamHI site at the N-terminus of the PLCδPH domain was replaced with CXCR4 fragments. For CXCR4-309-PH the CXCR4-PLCδPH juncture sequence is 5′ gcc aaa ttc ggg gga ggc ggc gga tcc ATG 3′, where the codon in bold encodes CXCR4 residue 309, and the capitalized codon is the start of the PLCδPH. For CXCR4-318-PH, the CXCR4-PLCδPH juncture sequence is 5′ ctc acc tgg gaa ttc ggg gga ggc ggc gga tcc ATG 3′, where the codon in bold encodes CXCR4 residue 318, and the capitalized codon is the start of the PLCδPH. Our split GFP partners also represent fusions to the PLCδPH. For pGFP-H11-PLCdeltaPH, where the eleventh helix of GFP is fused to a C-terminal PLCδPH, the pEGFP-GFP11-ClathrinLightChain [48] plasmid EcoRI-HpaI clathrin-encoding fragment was replaced with the PLCδPH EcoRI-HpaI fragment from GFP-C1-PLCdelta-PH. The resulting GFP helix 11-PLCδPH juncture reading frame is 5′ aca ggc gga tcc gga ctc aga tct cga gct caa gct tcg aat tct gca gtc gac ggt acc gcg ggc ccg gga tcc ATG 3′, where the codon in bold represents the last residue of GFP helix 11 and the capitalized codon is the start of the PLCδPH. For pcDNA3.1-PLCdeltaPH-GFP-H1-10, the PLCδPH coding region was inserted in frame and upstream of the GFP helix 1–10 sequence in pcDNA3.1-GFP(1-10) [48]. The reading frame at the PLCδPH N-terminus is 5′ gct agc gcc acc atg gcg gcc gcg gga tcc ATG 3′, where the italicized sequence is the pcDNA3.1 NheI site, the capitalized sequence is the PLCδPH start codon, and the bold sequence is an alternative in-frame start codon. The PLCδPH-GFP juncture sequence is 5′ aag gag ctc ggg ata tct agc aag ctt ccc ggg ctc gag gaa ttc gtt aac aga tct ATG 3′, where the bold codon indicates a PLCδPH C-terminal residue corresponding to residue 433 of the EGFP-C1-PLCdeltaPH fusion protein, and the ATG indicates the beginning of the GFP helix 1–10 sequence.

2.2. Cell Culture

Human embryonic kidney 293T cells (293; [62]) were obtained from the American Type Culture Collection (ATCC), HIV-1 reporter TZM-bl cells [63,64,65] were from the National Institutes of Health (NIH) AIDS Reagent Program, and 293-CerS2−/− (CerS2−/−) knockout cells, which carry premature stop codons after 63 residues of both CerS2 alleles, were obtained from Dr. Antony H. Futerman [25,66]. The Jurkat T cell line [67] was from the BEI Resources HIV Reagent Program, as was the J1.1 cell line, which is an HIV-1-infected Jurkat derivative [68,69,70,71,72]. Cells were routinely grown in humidified 5% carbon dioxide air at 37 °C in Dulbecco’s Modified Eagle’s Media (DMEM; 293, TZM-bl and CerS2−/−) or Roswell Park Memorial Institute (RPMI; Jurkat and J1.1) media, both of which were supplemented with 10% fetal bovine sera (FBS) plus 10 mM Hepes, pH 7.3, penicillin, and streptomycin [13,25,55].
Transfections of cells were performed using polyethyleneimine (PEI) as described previously [13,25,55,73], and typically employed 24 μg total DNA for 5 million cells on 10 cm plates, or 6 μg total DNA for 1 million cells on polylysine-coated (0.01%; Sigma P4707) coverslips (Fisher 22 × 22 − 1.0) in 6-well plates with 35 mm well diameters. Unless otherwise noted, in cases where cells were co-transfected with multiple plasmids, equal DNA amounts in weight for each plasmid were used to a total of 24 μg. When infectious NL4-3 viruses were scored for infectivity, TZM-bl cells were seeded onto wells of 6-well plates (1 million cells per well) 24 h before infections. For the generation of 293-derived target cells to be infected, cells were seeded 1 day post-transfection into 6-well plate wells at 1 million cells per well, and grown an additional 2 days prior to infection. For cells to be processed for immunofluorescence, cells transfected on coverslips were refed one day post-transfection and processed 2 days later. Alternatively, cells on 10 cm plates were split onto polylysine-coated coverslips 1 day post-transfection (1 million cells per coverslip) and processed 2 days later. A third approach was employed for cell–cell fusion assays. In this case, 24 h after separately transfecting cells on 10 cm plates, cells were co-seeded onto polylysine-coated coverslips at 1 million cells per cell fusion partner, and incubated an additional 48 h prior to processing for microscopy.
For the production of NL4-3 virus stocks and pseudotyped lentivirus vectors, transfected 293 or CerS2−/− cells were refed 1 day post-transfection, and cells and virus-containing media supernatants were collected at 3 days post transfection. Cell samples for protein analyses were pelleted and frozen at −80 °C prior to use. Viral samples were filtered through 0.45 micron filters and either used immediately or stored at −80 °C. In some cases, viral samples were processed for protein analyses. For this, filtered virus-containing media samples were concentrated by centrifugation (1 h at 197,000× g; 35,000 rpm, Beckman SW41 rotor) through 2 mL cushions of 20% or 30% sucrose in phosphate-buffered saline (PBS; 9.5 mM sodium potassium phosphate [pH 7.4], 137 mM NaCl, 2.7 mM KCl), suspended in 0.1 mL PBS, and stored at −80 °C prior to use.
Viral infections were performed with cells seeded in 6-well plates and filtered viruses. Typically, cells were infected for 3–4 h with 1 mL viral stocks at 37 °C, supplemented with 1 mL media, and incubated an additional 72 h prior to collection for β-galactosidase (β-gal) assays, which were performed as described previously [55,74]. Briefly, media on cells in 6-well plate wells (35 mm diameter) were removed, and cells were scraped into 1.0 mL PBS and pelleted. Cell pellets were suspended in 150 μL PBS containing 0.1% sodium dodecyl sulfate (SDS), vortexed, supplemented with 600 μL PM-2 buffer (33 mM NaH2PO4, 66 mM Na2HPO4, 2 mM MgSO4, 0.1 mM MnCl2, 40 mM β-mercaptoethanol [BME]), vortexed, supplemented with 150 mL 4 mg/mL 2-nitrophenyl-β-d-galactopyranoside (ONPG) in PM-2 buffer, and incubated at 37 °C. Reactions were stopped by addition of 375 μL 1 M Na2CO3 and flash freezing on dry ice powder. Samples then were thawed, and 420 nm light absorbances were read spectrophotometrically to calculate β-galactosidase activities (1 unit = 1 nMole ONPG hydrolyzed per minute = 420 nm absorbance × 285/minutes of incubation) as a measure of infectivity [55,74]. To calculate significance values, means and standard deviations from multiple experiments were determined and converted to Z values, which were used to calculate probabilities.
In addition to the above infection protocol, three variations also were employed. When viruses were treated with amphotericin B methyl ester (AME; kindly provided by the National Cancer Institute Developmental Therapeutics Program [NCI DTP; NCI #252624] and used after minimum freeze–thaw cycles to avoid degradation), they were incubated for 2 h at 37 °C in the presence of 10 μM AME before infection [13,26,27,28]. For testing of the effects of T-20 (Enfuvirtide; BEI Resources HIV Reagent Program; [75,76,77]) on infections, cells were infected 24 h in 1 mL media plus one ml virus plus either 10 μM T-20 in dimethylsulfoxide (DMSO) or DMSO (final 1%). After 24 h, infection mixes were removed, cells were washed once with media, refed with media without T-20 or DMSO, and incubated an additional 48 h prior to assay. Infections of non-adherent Jurkat and J1.1 cells also employed a slightly modified approach. Here, cells either were mock treated or treated for 48 h with 10 ng/mL tumor necrosis factor alpha (TNF-α; Stemcell Technologies #78068.1). After this treatment, cells were infected for 72 h at 37 °C in incubations containing 1 mL media, 1 mL virus, and 3 million cells in the presence or absence of 10 ng/mL TNF-α. Following infections, pelleted cells were processed for β-gal assays as described above.

2.3. Microscopy

For direct viewing of cell–cell fusion incubations, cells on coverslips were washed with PBS, fixed 20 min at room temperature in 4% paraformaldehyde (PFA; Sigma) in PBS, washed three times in PBS, incubated 5 min at room temperature with 200 ng/mL 4′,6-diamidino-2-phenylindole (DAPI; Sigma) in PBS, washed three times in PBS, and mounted onto microscope slides in Fluoro-G (Thermo-Fisher) mounting medium. For indirect immunofluorescent detection of cell surface Env proteins, cells on coverslips were washed and fixed as described above and then processed for immunofluorescence detection as described previously [55,61,78], except for excluding the Triton X-100 permeabilization step. Primary antibodies were human anti-gp120 VRC03 and 2G12, used at 1.67 μg/mL and 0.67 μg/mL, respectively; the secondary antibody was an AlexaFluor594-conjugated goat anti-human IgG(H + L) antibody (Invitrogen A11014), used at 1:1000 (2 μg/mL).
Fluorescence viewing was conducted on a Keyence BZ-X710 fluorescence microscope using either Plan Fluor 20× or Plan Fluor 40× (oil) lenses and the following filter sets: DAPI, excitation 360 nm/40 nm slit width, emission 460 nm/50 nm slit width; GFP, excitation 470 nm/40 nm slit width, emission 525 nm/50 nm slit width; AlexaFluor594, excitation 560 nm/40 nm slit width, emission, 630 nm/75 nm slit width. All imaging employed 100% gain settings, and samples to be compared used identical exposure settings, unless otherwise noted.
For cell–cell syncytia counting of each sample, numbers of GFP+ syncytia per five 0.4 mm2 fields were counted and averaged. For determination of Env+ cell percentages, the red-staining cell counts and total cell counts from a minimum of nine images for each sample were used in calculations. For comparison of cell brightnesses, background-subtracted gray scale brightness values were determined for boxed Env+ cells using (Image J [Fiji]; [79]) software. Raw brightness values then were divided by image exposure times to yield the final brightness values. Note that for Env-753Stop-expressing cells, exposure times were typically 10 ms, while for Env-WT-expressing cells, exposure times were 67–100 ms. Note also that brightness value averages and standard deviations were calculated from a minimum of 14 cells for the VRC03 primary antibody, and a minimum of 36 cells for the 2G12 primary antibody.

2.4. Protein Analysis

For analysis of concentrated virus samples, pelleted and resuspended viral samples were mixed with an equal volume of 2× sample buffer (12.5 mM Tris-HCl [pH 6.8], 2% SDS, 20% glycerol, 0.25% bromphenol blue) plus 5% β-mercaptoethanol (BME), and frozen at −80 °C. Cell samples for protein analysis were prepared by collecting cells in PBS, pelleting 20% of the cell sample, suspension in 50 μL IPB (20 mM Tris-HCl [pH 7.5], 150 mM NaCl, 1 mM ethylenediamine tetraacetic acid [EDTA], 0.1% sodium dodecyl sulfate [SDS], 0.5% sodium deoxycholate, 1.0% TritonX-100, 0.02% sodium acetate), vortexing, incubation on ice for 5 min, pelleting 15 min at 13,000× g to remove insoluble debris, mixing with 50 μL 2× sample buffer plus 0.1 volume of BME, and stored frozen at −80 °C prior to analysis.
Samples for protein analysis were subjected to SDS–polyacrylamide gel electrophoresis (SDS-PAGE) as described previously [13,25,55,61,74,78], except that samples were heated only to 37 °C before electrophoresis, and acrylamide concentrations were either 10% or 7.5%. Typically, 10% of total viral samples or 4% of total cell samples were subjected to electrophoresis in parallel with molecular weight size standards (Bio-Rad). After SDS-PAGE fractionation, proteins were electroblotted and immunoblotted following previously described methods [13,25,55,61,74,78]. The employed primary antibodies were as follows: mouse anti-HIV-CA hybridoma media (Hy183, kindly provided by Dr. Bruce Chesebro) at 1:15; mouse anti-gp41(CT) hybridoma media (Chessie 8, recognizing CT residues 727-732 [PDRPEG], from the NIH AIDS Reagent Program) at 1:15; rabbit anti-CD4 D2E6M monoclonal antibody (Cell Signaling Technology, 93518T), used at 1:2000; polyclonal rabbit anti-CXCR4 antibody (ProSci, #1009), used at 1:2000. Secondary reagents were alkaline phosphatase-conjugated anti-mouse, anti-human, or anti-rabbit IgG antibodies (Promega) used at 1:15,000. Color reactions for visualization of antibody-bound bands employed nitrobluetetrazolium plus 5-bromo-4-chloro-3-indolyl phosphate in AP buffer (1000 mM Tris-hydrochloride [pH 9.5], 100 mM NaCl, 5 mM MgCl2; [13,25,55,61,74,78]). For quantitation, immunoblots were air-dried and scanned using an Epson Perfection V600 scanner, and band intensities of scanned TIFF images were determined using NIH Image J software [79].

3. Results

3.1. Analysis of Env-Mediated Cell–Cell Fusion

We recently observed that HIV-1-derived lentivirus vectors decorated with a nanobody against the SARS-CoV-2 Spike protein selectively infected SARS-CoV-2-infected cells via a fusion in reverse mechanism, in which nanobody–Spike binding activated the fusion function of Spike [55]. These results were reminiscent of efforts to infect HIV-1 Env-expressing cells with membrane-enveloped viruses carrying receptors and co-receptors [40,41,42,43,44,45,46,47]. Based on this, we reasoned that it might be possible to employ a fusion in reverse approach to examine factors in target membranes that affect HIV-1 Env functions. As a preliminary test, we examined the capacities of Env, receptor, and co-receptor combinations to function in cell–cell fusion assays. The method employed (Figure 1A) is similar to one previously described [80], and takes advantage of split green fluorescent protein (GFP) fragments to assemble into active fluorescent proteins [48,80]. As shown (Figure 1A), one set of cells was co-transfected with an Env expression plasmid plus a PM-targeted large helix 1–10 GFP fragment, while a second set of cells was co-transfected with CD4 receptor-CXCR4 co-receptor pairs plus a PM-targeted small (helix 11) GFP fragment. After mixing the cells, fusion was assayed via the appearance of GFP-positive (GFP+) syncytia.
For our initial purposes, we employed human embryonic kidney 293 (293) cells [62], which are negative for CD4 and CXCR4. For transfections, an expression vector for the WT Env (Figure 1B, Env-WT) protein was used, along with a vector for the 753Stop Env protein (Env-753Stop), which carries a CT truncation, behaves similarly to a complete ΔCT Env protein, and is incorporated at high levels into HIV-1 particles [11,12,13]. Our CD4 vectors expressed a WT variant (Figure 1B, CD4) or a GPI-anchored CD4 (CD4-GPI), since GPI-anchored CD4 previously was shown to function as an HIV-1 receptor, and because GPI-anchored proteins may be assembled efficiently into HIV-1 particles [40,41,42,43,44,45,46,47,81,82,83]. For co-receptors, we tested WT CXCR4 (CXCR4) (which has been shown to be delivered, under some circumstances, to exosomes and enveloped viruses), and a C-terminally truncated variant (CXCR4-318), to remove residues involved in phosphorylation and internalization [40,41,42,43,44,45,46,47,84,85,86,87,88].
Examples of our cell–cell fusion assays are shown in Figure 2A–F. In each case, one set of fusion-partner cells expressed the helix 11 GFP fragment plus CD4-GPI and CXCR4-318, while the other sets of cells expressed the large GFP fragment plus Env-WT (A,B), Env-753Stop (C,D), or no Env (E,F). Cells were monitored in parallel for nuclei using 4′,6-diamidino-2-phenylindole (DAPI) stain (A,C,E) or for GFP+ syncytia (B,D,F). Not surprisingly, few if any GFP+ syncytia were observed for the no Env incubation (Figure 2F), whereas some were seen with Env-WT (Figure 2B), and syncytia were numerous in the Env-753Stop incubation (Figure 2D). These observations were borne out when syncytia were quantified (Figure 2G). Notably, numbers of Env-WT syncytia were twenty times higher than the background, and Env-753Stop syncytia numbers were ten times higher than those for Env-WT. It also was apparent that all of the receptor plus co-receptor combinations yielded similar results.
What might account for the tenfold difference in syncytia formation with the Env-753Stop versus Env-WT proteins? Because an HIV-1 Env protein with a truncation similar to Env-753Stop had been selected for enhanced cell surface expression [11,12], we speculated that our observed increase in syncytia formation with Env-753Stop might stem from a high level of surface expression. To test this possibility, target cells expressing Env-WT versus Env-753Stop proteins were subjected to immunofluorescent detection of cell surface Env. Although we did not observe discernable differences in the localization patterns of the different Env proteins (Figure 3A,B), it was noteworthy that Env imaging required higher exposure times for Env-WT versus Env-753Stop (67 ms in Figure 3B versus 10 ms in Figure 3A). To quantify our observations, we measured percentages of fluorescent cells (Figure 3C) and relative brightness levels (Figure 3D) of the Envs with two different anti-gp120(SU) antibodies (VRC03, 2G12). Both antibodies detected moderately lower (2.5-fold) percentages of Env-WT fluorescent cells, but substantial reductions (10-fold) in Env-WT brightness levels that correlate with surface expression levels. Given that differences in Env-WT and Env-753Stop surface expression levels matched the differences we observed in cell–cell fusion capacities (Figure 2), we conclude that the increased frequency of Env-753Stop-mediated fusion is related to its high surface availability.

3.2. Viral Infection via Fusion in Reverse

Given that CD4-GPI and CXCR4-318 seemed likely candidates to be incorporated into HIV-1 particles [55,81,82,83,84,85,86,87,88], and that they efficiently mediated cell–cell fusion reactions (Figure 2), we tested their propensity for targeting the infection of Env-expressing cells. To do so, viruses produced by co-transfection of cells with psPAX2, pLVX-puro-Xho-ATG-β-gal, CD4-GPI, and CXCR4-318 plasmids were used to infect Env-expressing cells (Figure 4A), scoring for infection by β-gal expression. As shown in Figure 4B, our results clearly showed that the CD4-GPI plus CXCR4-318 pseudotyped viruses specifically infected Env-expressing cells. In particular, infection of Env-WT cells was over ten times greater than Env-minus (mock) cells, while infection of Env-753Stop cells appeared to be ten times more efficient than infection of the Env-WT cells. These results closely mirrored our cell–cell fusion results (Figure 2), implying that infection efficiency also is dependent on cell surface Env protein levels (Figure 3). To verify that infections were dependent on the Env fusion function, separate infections were performed in the absence or presence of the HIV-1 Env fusion inhibitor T-20 (Enfuvirtide; [75,76,77]). As depicted in Figure 4C, T-20 almost completely abolished infectivity, supporting the interpretation that infection occurred by a fusion in reverse process.
To help optimize the receptor and co-receptor requirements for infection, different CD4 and CXCR4 combinations (Figure 1B) were tested for their capacities to infect cells expressing Env-753Stop (Figure 5A) and Env-WT (Figure 5B), and normalized to results of the CD4-GPI (gpi) plus CXCR4-318 (318). As expected, little or no infection was observed when either receptor or co-receptor was used alone (gpi/-; -/318). Surprisingly, substitution of WT CD4 for CD4-GPI also dramatically reduced infection efficiency (CD4/318). In contrast, replacement of CXCR4-318 with WT CXCR4, or with CXCR4 carrying a nearly complete CT truncation (CXCR4-309; 309), or with CXCR4 variants bearing C-terminal fusions to a PI(4,5)P2-binding pleckstrin homology (PH; [62,63,64]) domain (CXCR4-318PH [318PH], CXCR4-309PH [309PH]) had little effect on infectivities. Nevertheless, the most efficient combination for infection either Env-753Stop or Env-WT cells remained the CD4-GPI plus CXCR4-318 combination.
Because none of the CXCR4 variants gave dramatically different infectivity results (Figure 5), and because our anti-CXCR4 antibodies were ineffective in immunoblotting, we did not examine the virion incorporation of the CXCR4 variants. Nevertheless, all of the CXCR4 variants gave similar cellular immunofluorescence staining patterns (data not shown), suggesting similar expression levels. However, because of the significant disparity between the cell–cell fusion (Figure 2) and fusion in reverse infection results (Figure 5) for WT CD4, it was worthwhile to examine CD4 and CD4-GPI virion incorporation levels in greater detail. Initially, virions from cells expressing HIV-1 Gag and GagPol plus either CD4 or CD4-GPI were collected by sedimentation through 20% sucrose cushions, and subjected to immunoblot detection of PrGag, CA, and CD4 proteins. As shown in Figure 6A, CD4 migrated (as expected) at a slightly slower mobility than CD4-GPI, and the observed viral CD4-to-CA ratio appeared greater than that of CD4-GPI. As a more stringent test, the two CD4 variants were expressed in the presence or absence of Gag plus GagPol proteins, and media samples were sedimented through 30% sucrose cushions to reduce possible exosome or membrane vesicle contamination. Cell and viral samples so prepared were processed for immunodetection with anti-CA (Figure 6B) and anti-CD4 (Figure 6C) antibodies. Our anti-CA immunoblots gave the expected PrGag and CA bands in samples derived from cells transfected with the psPAX2 (psPAX) Gag and GagPol expression construct. The CD4 and CD4-GPI proteins also were readily detected in cellular samples (Figure 6C, top panel). Importantly, both WT CD4 and CD4-GPI were present in virus samples from cells co-transfected with psPAX2, while only trace amounts were present in samples processed similarly from cells not expressing Gag and GagPol proteins. Based on the virus-to-cell CD4 ratios, it appeared that CD4-GPI might be preferentially incorporated into virions, consistent with observations with other GPI-tagged proteins [55]. However, when we calculated viral CD4-to-CA ratios, the WT CD4 and CD4-GPI protein values were within 10% of each other. These results agree with studies that have demonstrated CD4 incorporation into HIV-1 virions [42,44,45]. Nevertheless, our observations strongly suggest that in the context of HIV-1 viral membranes, CD4-GPI is considerably more competent than CD4 in fostering Env-mediated fusion.

3.3. Targeted Transduction of HIV-1 Infected Cells

Beyond demonstrating that cells transfected to express HIV-1 Env could be targeted for infection, it was of interest to verify that HIV-1-infected cells could be similarly targeted. For these experiments, we employed the HIV-1-infected J1.1 Jurkat T cell line that has proven useful in latency reactivation studies, and can be induced by treatment with tumor necrosis factor alpha (TNFα; [68,69,70,71,72]). As expected, and as illustrated in Figure 7, TNFα treatment of J1.1 cells increased cellular levels of both Gag (Figure 7A; PrGag, CA) and Env (Figure 7B; gp160, gp41) proteins. Thus, for our purposes, we tested the propensity of CD4-GPI plus CXCR4-318 pseudotyped lentivirus vectors to transduce Jurkat, J1.1, and TNFα-induced J1.1 (J1.1 + TNFα) cells. As illustrated in Figure 7C, relative to the background transduction of Jurkat cells, β-gal delivery to J1.1 cells via fusion in reverse was sevenfold higher, and infection levels rose to tenfold over the background with induced J1.1 recipients. These results are consistent with previous results in showing that the fusion in reverse mechanism can be utilized to target HIV-1-infected cells [40,41,42,43,44,45,46,47].

3.4. Effects of Membrane Lipid Compositions

As mentioned in the Introduction, HIV-1 particle membranes are highly enriched in PI(4,5)P2, PS, PE, saturated PCs, cholesterol, Cer, SM, and HEX ([14,15,16,17,18,19,20,21,22,23,24]; Figure 8A). Like cellular PMs, the internal leaflets of viral membranes are enriched in PI(4,5)P2, PS and PE lipids, while external leaflets have higher concentrations of SM, HEX, and saturated PC lipids ([14,15,16,17,18,19,20,21,22,23,24]; Figure 8A). We and others have shown that perturbation of viral cholesterol with AME inhibits the replication of Env-WT viruses, but not ΔCT viruses [13,26,27,28]. Additionally, depletion of long-chain SLs by production of viruses in CerS2−/− cells inhibits both WT and ΔCT virus fusion capabilities, while assembly of a scramblase into virions blocks virus replication [25,34].
The availability of viruses capable of reverse fusion gave us the unique opportunity to examine how perturbation of receptor plus co-receptor containing membranes might affect HIV-1 replication. For these investigations, we compared how AME, CerS2−/− knockout, and TMEM16F scramblase incorporation affected normal HIV-1 infections versus fusion in reverse infections (Figure 8B–D). Our results with AME perturbation of viral cholesterol are graphed in Figure 8B. As expected [13,26,27,28], AME treatment of WT HIV-1 dramatically reduced infection, but did not inhibit infection of Env-753Stop viruses, which behaved similarly to ΔCT viruses. Interestingly, AME treatment of receptor plus co-receptor pseudotyped virions did not impair infectivity, regardless of whether the target cells carried Env-WT or Env-753Stop. These results suggest that the HIV-1 Env fusion machinery can tolerate tampering with cholesterol levels in receptor- plus co-receptor-containing membranes.
Our results with viruses produced from cells expressing the PM-targeted TMEM16F scramblase are shown in Figure 8C. As previously demonstrated [34], WT HIV-1 virions produced in the presence of this TMEM16F variant were essentially blocked for replication. Our observations showing an inhibition of Env-753Stop viruses also are consistent previous results that demonstrated a significant scramblase inhibition of ΔCT HIV-1 replication [34]. Importantly, the TMEM16F scramblase also dramatically reduced fusion in reverse infections for both Env-WT and Env-753Stop target cells. Thus, the HIV-1 Env fusion activity appears to require asymmetric target membranes.
The third lipid variation we tried was a comparison of viruses produced from WT 293 cells as opposed to 293 CerS2−/− knockout cells (Figure 8D). For the WT virus, as seen previously [25], production in CerS2−/− cells reduced viral infectivity two to threefold. In contrast, and surprisingly, production in CerS2−/− cells increased the efficiency of fusion in reverse, implying that the presence of long-chain SLs in receptor plus co-receptor membranes may have a negative effect on HIV-1 Env activities. These results are discussed in more detail below.

4. Discussion

Ordinarily, a small number of trimeric HIV-1 Env proteins are incorporated into HIV-1 particles, and must perform their receptor-binding, co-receptor-binding, and fusion duties within the atypical viral lipid membrane environment that features high levels of PI(4,5)P2, PS, PE, saturated PCs, cholesterol, and sphingolipids ([1,2,3,4,5,6,7,8,9,14,15,16,17,18,19,20,21,22,23,24]; Figure 8A). However, it has long been known that Env trimers on the surfaces of infected cells can mediate cell–cell fusion with neighbor receptor- plus co-receptor-expressing cells, resulting in syncytia formation [35,36,37,38,39]. Using a split GFP reporter system (Figure 1A), we examined the capacities of different CD4 receptor plus CXCR4 co-receptor combinations to participate in cell–cell fusion reactions with cells expressing either WT or C-terminally truncated (753Stop) Env proteins. Among our observations were that CD4-GPI and truncated CXCR4-318 proteins were competent for fusion reactions, and that fusion efficiency with Env-753Stop was significantly higher than with Env-WT. Because Env CTs can influence Env fusogenic properties [13,27,28,89,90,91,92,93], it is possible that an increase in Env-753Stop fusogenicity could account for some of the differences in efficiency observed between WT- and 753Stop-directed cell–cell fusion. However, the tenfold difference in cell–cell fusion efficiencies directly matched the cell surface expression differences of the two Env proteins (Figure 2 and Figure 3), which suggests that Env surface expression is responsible for the disparity.
Based on previous observations of fusion in reverse [40,41,42,43,44,45,46,47,55], we tested CD4 and CXCR4 variants for their abilities to be assembled into HIV-1-based lentivirus vectors and accomplish fusion in reverse infections. Our results clearly demonstrate that this depends on both a receptor and co-receptor (Figure 5), and on the fusion function of Env (Figure 4). In addition to the truncated CXCR4-318 variant, other CXCR4 variants also were capable of mediating infection, including WT CXCR4 (Figure 5). Considering previous results [40,41,42,43,44,45,46,47,87,88], this is not surprising. In contrast, while WT CD4 facilitated cell–cell fusion reactions (Figure 2), it was deficient for fusion in reverse reactions (Figure 5). Given that CD4 and CD4-GPI virion incorporation levels appeared similar (Figure 6), our results imply that in the context of HIV-1 membranes, CD4-GPI proteins are more active than WT CD4 proteins either in binding Env or collaboration with CXCR4 proteins in directing fusion. In this regard, it is worth noting that previous work with a B16 mouse melanoma cell line expressing CXCR4, CCR5, and WT CD4 demonstrated that it was resistant to HIV-1 Env-mediated fusion because of the reduced CD4 lateral mobility in these cells [94]. Interestingly, replacement of WT CD4 with a CD4 cytoplasmic domain mutant rescued Env fusion [94]. Given these observations, we believe that a reduced WT CD4 mobility in virus membranes may impair fusion by restricting the formation of fusogenic Env–CD4–CXCR4 complexes [94]. Nevertheless, it is pertinent to mention that a previous study [45] did not see a difference in CD4- and CD4-GPI-directed fusion in reverse. We speculate that this could be due to specific differences in vectors, protocols, and detection methods employed; also, note that viral CD4 and CD4-GPI levels were not monitored in those investigations [45].
When we extended our analyses to include HIV-1-infected cells, we observed that latently infected Jurkat J1.1 cells [68,69,70,71,72] were specifically infected, and that induction of HIV-1 expression increased the efficiency of infection (Figure 7). These results again [40,41,42,43,44,45,46,47] demonstrate that a fusion in reverse approach permits the specific transduction of cells harboring HIV-1, albeit with the caveat that the efficiency depends greatly on Env surface expression levels (Figure 3, Figure 4 and Figure 7; [42]). In addition to CXCR4-tropic envelopes, others have shown that CCR5-tropic Envs [95,96,97] can be targeted [42,43,47], but it is important to point out that experiments have predominantly been performed with lab-adapted strains, rather than primary isolates, which adopt more closed Env trimer conformations [98]. Ultimately, the targeting of viral vectors to cells harboring HIV-1 in vivo could be employed to help eradicate viral reservoirs [40,41,42,43,44,45,46,47], and our results may facilitate the development of more efficient strategies. In this regard, it is tempting to speculate that some Env-binding nanobodies [99] may precisely replicate co-receptor properties and stimulate fusion in reverse to a greater degree than we have observed, but no evidence in favor of this possibility is currently available.
With respect to other applications of the HIV-1 fusion in reverse approach, we have demonstrated that it can allow the examination of factors that determine whether membranes carrying receptors and co-receptors are capable of Env-directed fusion. Interestingly, we did not find that AME perturbation of membrane cholesterol affected infection [13,26,27,28]. Nevertheless, given previous results [100,101,102,103,104], it will be of interest to test the effects of other cholesterol-targeting agents such as β-cyclodextrin, cholesterol oxidase, and 25-hydroxycholesterol. In contrast with AME, the assembly of virions in the presence of the PM-targeted TMEM16F scramblase [34] effectively killed the fusion in reverse process (Figure 8). This intriguing observation seems at odds with the notion that TMEM16F-induced PS scrambling in target PMs promotes membrane fusion events [105,106], and suggests that further examination of leaflet asymmetry effects on Env-directed fusion will be of interest. Another result that deserves further scrutiny is the observation that rather than inhibiting infectivity [25], expression of fusion in reverse virions in cells with a deficiency in very long-chain SLs increased infectivity (Figure 8). While it is tempting to speculate that SL-mediated alterations in membrane fluidity may be responsible for this phenomenon [107,108], it is pertinent to note that we have not directly demonstrated lipidome differences in viruses produced from WT versus CerS2−/− cells. It also is relevant that experiments performed with GPI-anchored CD4 may or may not reflect what might occur with WT CD4. Nevertheless, we believe that the HIV-1 fusion in reverse system will prove useful in the dissection of other factors that regulate HIV-1 Env-driven membrane fusion. We believe this knowledge will prove applicable in the deployment of lipid-targeted therapeutics, and in the design of lipid nanoparticles for therapeutic purposes.

Author Contributions

E.B. and A.A. conceptualized, performed, and analyzed the experiments, and wrote the manuscript. E.B. also was involved in funding acquisition and providing resources. R.L.B. was involved in methodology development and performing experiments. F.G.T. provided funding and resources, analyzed data, and helped in manuscript revision. All authors have read and agreed to the published version of the manuscript.

Funding

E.B. and F.G.T. are grateful for funding from the National Institutes of Health through grants R01 AI152579 (E.B.) and R01 AI141549 (F.G.T.).

Data Availability Statement

All data used to generate this report are provided in the figures of the paper. All plasmids reported here are available to academic researchers on request.

Acknowledgments

We are grateful to Antony Futerman for providing the CerS2−/− cells, and to Addgene, the NIH AIDS Reagent Program, BEI Resources and Didier Trono, Jennifer Doudna, Tobias Meyer, Bo Huang, Simon Davis, Erik Procko, and Jacob Yount for making reagents and recombinant DNA constructs available.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AMEamphotericin B methyl ester
β-galβ-galactosidase
Cerceramide
CerS2ceramide synthase 2
CTcytoplasmic tail
Envenvelope protein
GFPgreen fluorescent protein
GPIglycosylphosphatidylinositol
HEXhexosylceramide
MAmatrix
PCphosphatidylcholine
PE phosphatidylethanolamine
PHPleckstrin homology
PI(4,5)P2phosphatitylinositol-(4,5)-bisphosphate
PSphosphatidylserine
SLsphingolipid
SMsphingomyelin

References

  1. Checkley, M.; Luttge, B.; Freed, E. HIV-1 envelope glycoprotein biosynthesis, trafficking, and incorporation. J. Mol. Biol. 2011, 410, 582–608. [Google Scholar] [CrossRef] [PubMed]
  2. Ohno, H.; Aguilar, R.; Fournier, M.; Hennecke, S.; Cosson, P.; Bonifacino, J. Interaction of endocytic signals from the HIV-1 envelope glycoprotein complex with members of the adaptor medium chain family. Virology 1997, 238, 305–315. [Google Scholar] [CrossRef] [PubMed]
  3. Boge, M.; Wyss, S.; Bonifacino, J.; Thali, M. A membrane-proximal tyrosine-based signal mediates internalization of the HIV-1 envelope glycoprotein via interaction with the AP-2 clathrin adaptor. J. Biol. Chem. 1998, 273, 15773–15778. [Google Scholar] [CrossRef]
  4. Wyss, S.; Berlioz-Torrent, C.; Boge, M.; Blot, G.; Höning, S.; Benarous, R.; Thali, M. The highly conserved C-terminal dileucine motif in the cytosolic domain of the human immunodeficiency virus type 1 envelope glycoprotein is critical for its association with the AP-1 clathrin adaptor. J. Virol. 2001, 75, 2982–2992. [Google Scholar] [CrossRef] [PubMed]
  5. Bhakta, S.; Shang, L.; Prince, D.; Hunter, E. Mutagenesis of tyrosine and di-leucine motifs in the HIV-1 envelope cytoplasmic domain results in a loss of Env-mediated fusion and infectivity. Retrovirology 2011, 8, 37. [Google Scholar] [CrossRef]
  6. Zhang, S.; Nguyen, H.T.; Ding, H.; Wang, J.; Zou, S.; Liu, L.; Guha, D.; Gabuzda, D.; Ho, D.D.; Kappes, J.C.; et al. Dual Pathways of Human Immunodeficiency Virus Type 1 Envelope Glycoprotein Trafficking Modulate the Selective Exclusion of Uncleaved Oligomers from Virions. J. Virol. 2021, 95, e01369-20. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  7. Qi, M.; Williams, J.; Chu, H.; Chen, X.; Wang, J.; Ding, L.; Akhirome, E.; Wen, X.; Lapierre, L.; Goldenring, J.; et al. Rab11-FIP1C and Rab14 direct plasma membrane sorting and particle incorporation of the HIV-1 envelope glycoprotein complex. PLoS Pathog. 2013, 9, e1003278. [Google Scholar] [CrossRef]
  8. Qi, M.; Chu, H.; Chen, X.; Choi, J.; Wen, X.; Hammonds, J.; Ding, L.; Hunter, E.; Spearman, E. A tyrosine-based motif in the HIV-1 envelope glycoprotein tail mediates cell-type- and Rab11-FIP1C-dependent incorporation into virions. Proc. Natl. Acad. Sci. USA 2015, 112, 7575–7580. [Google Scholar] [CrossRef]
  9. Kirschman, J.; Qi, M.; Ding, L.; Hammonds, J.; Dienger-Stambaugh, K.; Wang, J.; Lapierre, L.; Goldenring, J.; Spearman, P. HIV-1 envelope glycoprotein trafficking through the endosomal recycling compartment is required for particle incorporation. J. Virol. 2018, 92, e01893-17. [Google Scholar] [CrossRef]
  10. Zhu, P.; Liu, J.; Bess, J., Jr.; Chertova, E.; Lifson, J.D.; Grisé, H.; Ofek, G.A.; Taylor, K.A.; Roux, K.H. Distribution and three-dimensional structure of AIDS virus envelope spikes. Nature 2006, 441, 847–852. [Google Scholar] [CrossRef] [PubMed]
  11. Stano, A.; Leaman, D.P.; Kim, A.S.; Zhang, L.; Autin, L.; Ingale, J.; Gift, S.K.; Truong, J.; Wyatt, R.T.; Olson, A.J.; et al. Dense Array of Spikes on HIV-1 Virion Particles. J. Virol. 2017, 91, e00415-17. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Mangala Prasad, V.; Leaman, D.P.; Lovendahl, K.N.; Croft, J.T.; Benhaim, M.A.; Hodge, E.A.; Zwick, M.B.; Lee, K.K. Cryo-ET of Env on intact HIV virions reveals structural variation and positioning on the Gag lattice. Cell 2022, 185, 641–653.e17. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  13. Alfadhli, A.; Romanaggi, C.; Barklis, R.L.; Barklis, E. Analysis of HIV-1 envelope cytoplasmic tail effects on viral replication. Virology 2023, 579, 54–66. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  14. Brugger, B.; Glass, B.; Haberkant, P.; Liebrecht, I.; Wieland, F.; Krausslich, H.-G. The HIV lipidome: A raft with an unusual composition. Proc. Natl. Acad. Sci. USA 2006, 103, 2641–2646. [Google Scholar] [CrossRef]
  15. Chan, R.; Uchil, P.D.; Jin, J.; Shui, G.; Ott, D.E.; Mothes, W.; Wenk, M.R. Retroviruses human immunodeficiency virus and murine leukemia virus are enriched in phosphoinositides. J. Virol. 2008, 82, 11228–11238. [Google Scholar]
  16. Lorizate, M.; Sachsenheimer, T.; Glass, B.; Habermann, A.; Gerl, M.J.; Krausslich, H.-G.; Brugger, B. Comparative lipidomics analysis of HIV-1 particles and their producer cell membrane in different cell lines. Cell. Microbiol. 2013, 15, 292–304. [Google Scholar]
  17. Nguyen, D.H.; Hildreth, J.E. Evidence for budding of human immunodeficiency virus type 1 selectively from glycolipid-enriched membrane lipid rafts. J. Virol. 2000, 74, 3264–3272. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  18. Ono, A.; Freed, E.O. Plasma membrane rafts play a critical role in HIV-1 assembly and release. Proc. Natl. Acad. Sci. USA 2001, 98, 13925–13930. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  19. Ono, A.; Ablan, S.D.; Lockett, S.J.; Nagashima, K.; Freed, E.O. Phosphatidylinositol (4,5) bisphosphate regulates HIV-1 Gag targeting to the plasma membrane. Proc. Natl. Acad. Sci. USA 2004, 101, 14889–14894. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  20. Krementsov, D.N.; Rassam, P.; Margeat, E.; Roy, N.H.; Schneider-Schaulies, J.; Milhiet, P.E.; Thali, M. HIV-1 assembly differentially alters dynamics and partitioning of tetraspanins and raft components. Traffic 2010, 11, 1401–1414. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  21. Hogue, I.B.; Grover, J.R.; Soheilian, F.; Nagashima, K.; Ono, A. Gag induces the coalescence of clustered lipid rafts and tetraspanin-enriched microdomains at HIV-1 assembly sites on the plasma membrane. J. Virol. 2011, 85, 9749–9766. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  22. Inlora, J.; Collins, D.R.; Trubin, M.E.; Chung, J.Y.; Ono, A. Membrane binding and subcellular localization of retroviral Gag proteins are differentially regulated by MA interactions with phosphatidylinositol-(4,5)-bisphosphate and RNA. mBio 2014, 5, e02202. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  23. Patil, A.; Gautam, A.; Bhattacharya, J. Evidence that Gag facilitates HIV-1 envelope association both in GPI-enriched plasma membrane and detergent resistant membranes and facilitates envelope incorporation onto virions in primary CD4+ T cells. Virol. J. 2010, 7, 3. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. Tomishige, N.; Bin Nasim, M.; Murate, M.; Pollet, B.; Didier, P.; Godet, J.; Richert, L.; Sako, Y.; Mély, Y.; Kobayashi, T. HIV-1 Gag targeting to the plasma membrane reorganizes sphingomyelin-rich and cholesterol-rich lipid domains. Nat. Commun. 2023, 14, 7353. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  25. Barklis, E.; Alfadhli, A.; Kyle, J.E.; Bramer, L.M.; Bloodsworth, K.J.; Barklis, R.L.; Leier, H.C.; Petty, R.M.; Zelnik, I.D.; Metz, T.O.; et al. Ceramide synthase 2 deletion decreases the infectivity of HIV-1. J. Biol. Chem. 2021, 296, 100340. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  26. Waheed, A.; Ablan, S.; Mankowski, M.; Cummins, J.; Ptak, R.; Schaffner, C.; Freed, E. Inhibition of HIV-1 replication by amphotericin B methyl ester. J. Biol. Chem. 2006, 281, 28699–28711. [Google Scholar]
  27. Waheed, A.; Ablan, S.; Roser, J.; Sowder, R.; Schaffner, C.; Chertova, E.; Freed, E. HIV-1 escape from the entry-inhibiting effects of a cholesterol-binding compound via cleavage of gp41 by the viral protease. Proc. Natl. Acad. Sci. USA 2007, 104, 8467–8471. [Google Scholar]
  28. Waheed, A.; Ablan, S.; Sowder, R.; Roser, J.; Schaffner, C.; Chertova, E.; Freed, E. Effect of mutations in the human immunodeficiency virus 1 protease on cleavage of the gp41 cytoplasmic tail. J. Virol. 2010, 84, 3121–3126. [Google Scholar]
  29. Contreras, F.X.; Sánchez-Magraner, L.; Alonso, A.; Goñi, F.M. Transbilayer (flip-flop) lipid motion and lipid scrambling in membranes. FEBS Lett. 2010, 584, 1779–1786. [Google Scholar] [CrossRef] [PubMed]
  30. Suzuki, J.; Umeda, M.; Sims, P.J.; Nagata, S. Calcium-dependent phospholipid scrambling by TMEM16F. Nature 2010, 468, 834–838. [Google Scholar] [CrossRef] [PubMed]
  31. Segawa, K.; Suzuki, J.; Nagata, S. Constitutive exposure of phosphatidylserine on viable cells. Proc. Natl. Acad. Sci. USA 2011, 108, 19246–19251. [Google Scholar] [CrossRef] [PubMed]
  32. Kunzelmann, K.; Nilius, B.; Owsianik, G.; Schreiber, R.; Ousingsawat, J.; Sirianant, L.; Wanitchakool, P.; Bevers, E.M.; Heemskerk, J.W. Molecular functions of anoctamin 6 (TMEM16F): A chloride channel, cation channel, or phospholipid scramblase? Pflugers Arch. 2014, 466, 407–414. [Google Scholar] [CrossRef] [PubMed]
  33. Arndt, M.; Alvadia, C.; Straub, M.S.; Clerico Mosina, V.; Paulino, C.; Dutzler, R. Structural basis for the activation of the lipid scramblase TMEM16F. Nat Commun. 2022, 13, 6692. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  34. Leonhardt, S.A.; Purdy, M.D.; Grover, J.R.; Yang, Z.; Poulos, S.; McIntire, W.E.; Tatham, E.A.; Erramilli, S.K.; Nosol, K.; Lai, K.K.; et al. Antiviral HIV-1 SERINC restriction factors disrupt virus membrane asymmetry. Nat. Commun. 2023, 14, 4368. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  35. Camerini, D.; Seed, B. A CD4 domain important for HIV-mediated syncytium formation lies outside the virus binding site. Cell 1990, 60, 747–754. [Google Scholar] [CrossRef] [PubMed]
  36. Chowdhury, M.I.; Koyanagi, Y.; Suzuki, M.; Kobayashi, S.; Yamaguchi, K.; Yamamoto, N. Increased production of human immunodeficiency virus (HIV) in HIV-induced syncytia formation: An efficient infection process. Virus Genes 1992, 6, 63–78. [Google Scholar] [CrossRef] [PubMed]
  37. Valentin, A.; Trivedi, H.; Lu, W.; Kostrikis, L.G.; Pavlakis, G.N. CXCR4 mediates entry and productive infection of syncytia-inducing (X4) HIV-1 strains in primary macrophages. Virology 2000, 269, 294–304. [Google Scholar] [CrossRef] [PubMed]
  38. Symeonides, M.; Murooka, T.T.; Bellfy, L.N.; Roy, N.H.; Mempel, T.R.; Thali, M. HIV-1-Induced Small T Cell Syncytia Can Transfer Virus Particles to Target Cells through Transient Contacts. Viruses 2015, 7, 6590–6603. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  39. Starling, T.; Padilla-Parra, S. HIV-1 Induced Cell-to-Cell Fusion or Syncytium Formation. In Syncytia: Origin, Structure, and Functions. Results and Problems in Cell Differentiation; Kloc, M., Uosef, A., Eds.; Springer: Cham, Switzerland, 2024; Volume 71. [Google Scholar] [CrossRef]
  40. Mebatsion, T.; Finke, S.; Weiland, F.; Conzelmann, K.K. A CXCR4/CD4 pseudotype rhabdovirus that selectively infects HIV-1 envelope protein-expressing cells. Cell 1997, 90, 841–847. [Google Scholar] [CrossRef] [PubMed]
  41. Schnell, M.J.; Johnson, J.E.; Buonocore, L.; Rose, J.K. Construction of a novel virus that targets HIV-1-infected cells and controls HIV-1 infection. Cell 1997, 90, 849–857. [Google Scholar] [CrossRef] [PubMed]
  42. Endres, M.J.; Jaffer, S.; Haggarty, B.; Turner, J.D.; Doranz, B.J.; O’Brien, P.J.; Kolson, D.L.; Hoxie, J.A. Targeting of HIV- and SIV-infected cells by CD4-chemokine receptor pseudotypes. Science 1997, 278, 1462–1464. [Google Scholar] [CrossRef] [PubMed]
  43. Somia, N.V.; Miyoshi, H.; Schmitt, M.J.; Verma, I.M. Retroviral vector targeting to human immunodeficiency virus type 1-infected cells by receptor pseudotyping. J. Virol. 2000, 74, 4420–4424. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Bittner, A.; Mitnacht-Kraus, R.; Schnierle, B.S. Specific transduction of HIV-1 envelope expressing cells by retroviral vectors pseudotyped with hybrid CD4/CXCR4 receptors. J. Virol. Methods 2002, 104, 83–92. [Google Scholar] [CrossRef] [PubMed]
  45. Ye, Z.; Harmison, G.G.; Ragheb, J.A.; Schubert, M. Targeted infection of HIV-1 Env expressing cells by HIV(CD4/CXCR4) vectors reveals a potential new rationale for HIV-1 mediated down-modulation of CD4. Retrovirology 2005, 2, 80. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  46. Lee, C.L.; Dang, J.; Joo, K.I.; Wang, P. Engineered lentiviral vectors pseudotyped with a CD4 receptor and a fusogenic protein can target cells expressing HIV-1 envelope proteins. Virus Res. 2011, 160, 340–350. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  47. Peretti, S.; Schiavoni, I.; Pugliese, K.; Federico, M. Selective elimination of HIV-1-infected cells by Env-directed, HIV-1-based virus-like particles. Virology 2006, 345, 115–126. [Google Scholar] [CrossRef] [PubMed]
  48. Kamiyama, D.; Sekine, S.; Barsi-Rhyne, B.; Hu, J.; Chen, B.; Gilbert, L.A.; Ishikawa, H.; Leonetti, M.D.; Marshall, W.F.; Weissman, J.S.; et al. Versatile protein tagging in cells with split fluorescent protein. Nat. Commun. 2016, 7, 11046. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Stauffer, T.P.; Ahn, S.; Meyer, T. Receptor-induced transient reduction in plasma membrane PtdIns(4,5)P2 concentration monitored in living cells. Curr. Biol. 1998, 8, 343–346. [Google Scholar] [CrossRef] [PubMed]
  50. Zuffery, R.; Nagy, D.; Mandel, R.; Naldini, L.; Trono, D. Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. Nat. Biotechnol. 1997, 15, 871–875. [Google Scholar]
  51. Syed, A.M.; Taha, T.Y.; Tabata, T.; Chen, I.P.; Ciling, A.; Khalid, M.M.; Sreekumar, B.; Chen, P.Y.; Hayashi, J.M.; Soczek, K.M.; et al. Rapid assessment of SARS-CoV-2-evolved variants using virus-like particles. Science 2021, 374, 1626–1632. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  52. Sušac, L.; Vuong, M.T.; Thomas, C.; von Bülow, S.; O’Brien-Ball, C.; Santos, A.M.; Fernandes, R.A.; Hummer, G.; Tampé, R.; Davis, S.J. Structure of a fully assembled tumor-specific T cell receptor ligated by pMHC. Cell 2022, 185, 3201–3213.e19. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. Heredia, J.D.; Park, J.; Brubaker, R.J.; Szymanski, S.K.; Gill, K.S.; Procko, E. Mapping Interaction Sites on Human Chemokine Receptors by Deep Mutational Scanning. J. Immunol. 2018, 200, 3825–3839. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Chesarino, N.M.; McMichael, T.M.; Hach, J.C.; Yount, J.S. Phosphorylation of the antiviral protein interferon-inducible transmembrane protein 3 (IFITM3) dually regulates its endocytosis and ubiquitination. J. Biol. Chem. 2014, 289, 11986–11992. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  55. Alfadhli, A.; Bates, T.A.; Barklis, R.L.; Romanaggi, C.; Tafesse, F.G.; Barklis, E. A nanobody interaction with SARS-COV-2 Spike allows the versatile targeting of lentivirus vectors. J. Virol. 2024, 98, e0079524. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  56. López, C.S.; Sloan, R.; Cylinder, I.; Kozak, S.L.; Kabat, D.; Barklis, E. RRE-dependent HIV-1 Env RNA effects on Gag protein expression, assembly and release. Virology 2014, 462–463, 126–134. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Adachi, A.; Gendelman, H.E.; Koenig, S.; Folks, T.; Willey, R.; Rabson, A.; Martin, M.A. Production of acquired immunodeficiency syndrome-associated retrovirus in human and nonhuman cells transfected with an infectious molecular clone. J. Virol. 1986, 59, 284–291. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  58. Millán, J.L. Molecular cloning and sequence analysis of human placental alkaline phosphatase. J. Biol. Chem. 1986, 261, 3112–3125, Erratum in J. Biol. Chem. 1991, 266, 4023. [Google Scholar] [CrossRef] [PubMed]
  59. Mace, M.C.; Hansen, M.; Whiting, S.; Wang, C.T.; Barklis, E. Retroviral envelope protein fusions to secreted and membrane markers. Virology 1992, 188, 869–874. [Google Scholar] [CrossRef] [PubMed]
  60. Scholz, I.; Still, A.; Dhenub, T.C.; Coday, K.; Webb, M.; Barklis, E. Analysis of human immunodeficiency virus matrix domain replacements. Virology 2008, 371, 322–335. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  61. Barklis, E.; Staubus, A.O.; Mack, A.; Harper, L.; Barklis, R.L.; Alfadhli, A. Lipid biosensor interactions with wild type and matrix deletion HIV-1 Gag proteins. Virology 2018, 518, 264–271. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. DuBridge, R.B.; Tang, P.; Hsia, H.C.; Leong, P.M.; Miller, J.H.; Calos, M.P. Analysis of mutation in human cells by using an Epstein-Barr virus shuttle system. Mol. Cell Biol. 1987, 7, 379–387. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  63. Platt, E.J.; Wehrly, K.; Kuhmann, S.E.; Chesebro, B.; Kabat, D. Effects of CCR5 and CD4 cell surface concentrations on infections by macrophagetropic isolates of human immunodeficiency virus type 1. J. Virol. 1998, 72, 2855–2864. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  64. Derdeyn, C.A.; Decker, J.M.; Sfakianos, J.N.; Wu, X.; O’Brien, W.A.; Ratner, L.; Kappes, J.C.; Shaw, G.M.; Hunter, E. Sensitivity of human immunodeficiency virus type 1 to the fusion inhibitor T-20 is modulated by coreceptor specificity defined by the V3 loop of gp120. J. Virol. 2000, 74, 8358–8367. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Wei, X.; Decker, J.M.; Liu, H.; Zhang, Z.; Arani, R.B.; Kilby, J.M.; Saag, M.S.; Wu, X.; Shaw, G.M.; Kappes, J.C. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob. Agents Chemother. 2002, 46, 1896–1905. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  66. Tidhar, R.; Zelnik, I.D.; Volpert, G.; Ben-Dor, S.; Kelly, S.; Merrill, A.H., Jr.; Futerman, A.H. Eleven residues determine the acyl chain specificity of ceramide synthases. J. Biol. Chem. 2018, 293, 9912–9921. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. Schneider, U.; Schwenk, H.U.; Bornkamm, G. Characterization of EBV-genome negative “null” and “T” cell lines derived from children with acute lymphoblastic leukemia and leukemic transformed non-Hodgkin lymphoma. Int. J. Cancer 1977, 19, 621–626. [Google Scholar] [CrossRef] [PubMed]
  68. Rodari, A.; Poli, G.; Van Lint, C. Jurkat-Derived (J-Lat, J1.1, and Jurkat E4) and CEM-Derived T Cell Lines (8E5 and ACH-2) as Models of Reversible Proviral Latency. Methods Mol. Biol. 2022, 2407, 3–15. [Google Scholar] [CrossRef] [PubMed]
  69. Saayman, S.M.; Lazar, D.C.; Scott, T.A.; Hart, J.R.; Takahashi, M.; Burnett, J.C.; Planelles, V.; Morris, K.V.; Weinberg, M.S. Potent and Targeted Activation of Latent HIV-1 Using the CRISPR/dCas9 Activator Complex. Mol. Ther. 2016, 24, 488–498. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Okutomi, T.; Minakawa, S.; Hirota, R.; Katagiri, K.; Morikawa, Y. HIV Reactivation in Latently Infected Cells With Virological Synapse-Like Cell Contact. Viruses 2020, 12, 417. [Google Scholar] [CrossRef]
  71. Liu, H.; Chen, C.; Liao, S.; Sohaii, D.K.; Cruz, C.R.Y.; Burdo, T.H.; Cradick, T.J.; Mehta, A.; Barrero, C.; Florez, M.; et al. Strategic self-limiting production of infectious HIV particles by CRISPR in permissive cells. Mol. Ther. Nucleic Acids 2023, 32, 1010–1025. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  72. Molnar, S.M.; Kim, Y.; Wieczorek, L.; Williams, A.; Patil, K.A.; Khatkar, P.; Santos, M.F.; Mensah, G.; Lorico, A.; Polonis, V.R.; et al. Extracellular vesicle isolation methods identify distinct HIV-1 particles released from chronically infected T-cells. J. Extracell. Vesicles 2024, 13, e12476. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  73. Longo, P.A.; Kavran, J.M.; Kim, M.S.; Leahy, D.J. Transient mammalian cell transfection with polyethylenimine (PEI). Methods Enzymol. 2013, 529, 227–240. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  74. Jones, T.A.; Blaug, G.; Hansen, M.; Barklis, E. Assembly of gag-beta-galactosidase proteins into retrovirus particles. J. Virol. 1990, 64, 2265–2279. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  75. Wild, C.; Greenwell, T.; Matthews, T. A synthetic peptide from HIV-1 gp41 is a potent inhibitor of virus-mediated cell-cell fusion. AIDS Res. Hum. Retrovir. 1993, 9, 1051–1053. [Google Scholar] [CrossRef] [PubMed]
  76. Wild, C.T.; Shugars, D.C.; Greenwell, T.K.; McDanal, C.B.; Matthews, T.J. Peptides corresponding to a predictive alpha-helical domain of human immunodeficiency virus type 1 gp41 are potent inhibitors of virus infection. Proc. Natl. Acad. Sci. USA 1994, 91, 9770–9774. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  77. Kilby, J.M.; Hopkins, S.; Venetta, T.M.; DiMassimo, B.; Cloud, G.A.; Lee, J.Y.; Alldredge, L.; Hunter, E.; Lambert, D.; Bolognesi, D.; et al. Potent suppression of HIV-1 replication in humans by T-20, a peptide inhibitor of gp41-mediated virus entry. Nat. Med. 1998, 4, 1302–1307. [Google Scholar] [CrossRef] [PubMed]
  78. Alfadhli, A.; Romanaggi, C.; Barklis, R.L.; Barklis, E. Second site reversion of HIV-1 envelope protein baseplate mutations maps to the matrix protein. J. Virol. 2024, 98, e0174223. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  79. Schneider, C.A.; Rasband, W.S.; Eliceiri, K.W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 2012, 9, 671–675. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  80. Wang, J.; Kondo, N.; Long, Y.; Iwamoto, A.; Matsuda, Z. Monitoring of HIV-1 envelope-mediated membrane fusion using modified split green fluorescent proteins. J. Virol. Methods 2009, 161, 216–222. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  81. Diamond, D.C.; Finberg, R.; Chaudhuri, S.; Sleckman, B.P.; Burakoff, S.J. Human immunodeficiency virus infection is efficiently mediated by a glycolipid-anchored form of CD4. Proc. Natl. Acad. Sci. USA 1990, 87, 5001–5005. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  82. Marshall, W.L.; Mittler, E.S.; Avery, P.; Lawrence, J.P.; Finberg, R.W. Glycosylphosphatidylinositol-anchored CD4 supports human immunodeficiency virus type 1 replication, but not cytopathic effect, in T-cell transfectants. J. Virol. 1994, 68, 4039–4042. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  83. Kost, T.A.; Kessler, J.A.; Patel, I.R.; Gray, J.G.; Overton, L.K.; Carter, S.G. Human immunodeficiency virus infection and syncytium formation in HeLa cells expressing glycophospholipid-anchored CD4. J. Virol. 1991, 65, 3276–3283. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Busillo, J.M.; Benovic, J.L. Regulation of CXCR4 signaling. Biochim. Biophys. Acta 2007, 1768, 952–963. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  85. Cronshaw, D.G.; Nie, Y.; Waite, J.; Zou, Y.R. An essential role of the cytoplasmic tail of CXCR4 in G-protein signaling and organogenesis. PLoS ONE 2010, 5, e15397. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  86. García-Cuesta, E.M.; Rodríguez-Frade, J.M.; Gardeta, S.R.; D’Agostino, G.; Martínez, P.; Soler Palacios, B.; Cascio, G.; Wolf, T.; Mateos, N.; Ayala-Bueno, R.; et al. Altered CXCR4 dynamics at the cell membrane impairs directed cell migration in WHIM syndrome patients. Proc. Natl. Acad. Sci. USA 2022, 119, e2119483119. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  87. Kang, K.; Ma, R.; Cai, W.; Huang, W.; Paul, C.; Liang, J.; Wang, Y.; Zhao, T.; Kim, H.W.; Xu, M.; et al. Exosomes Secreted from CXCR4 Overexpressing Mesenchymal Stem Cells Promote Cardioprotection via Akt Signaling Pathway following Myocardial Infarction. Stem Cells Int. 2015, 2015, 659890. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  88. Luo, H.; Chen, D.; Li, R.; Li, R.; Teng, Y.; Cao, Y.; Zou, X.; Wang, W.; Zhou, C. Genetically engineered CXCR4-modified exosomes for delivery of miR-126 mimics to macrophages alleviate periodontitis. J. Nanobiotechnol. 2023, 21, 116. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  89. Edwards, T.G.; Wyss, S.; Reeves, J.D.; Zolla-Pazner, S.; Hoxie, J.A.; Doms, R.W.; Baribaud, F. Truncation of the cytoplasmic domain induces exposure of conserved regions in the ectodomain of human immunodeficiency virus type 1 envelope protein. J. Virol. 2002, 76, 2683–2691. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  90. Wyma, D.J.; Jiang, J.; Shi, J.; Zhou, J.; Lineberger, J.E.; Miller, M.D.; Aiken, C. Coupling of human immunodeficiency virus type 1 fusion to virion maturation: A novel role of the gp41 cytoplasmic tail. J. Virol. 2004, 78, 3429–3435. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  91. Wyss, S.; Dimitrov, A.S.; Baribaud, F.; Edwards, T.G.; Blumenthal, R.; Hoxie, J.A. Regulation of human immunodeficiency virus type 1 envelope glycoprotein fusion by a membrane-interactive domain in the gp41 cytoplasmic tail. J. Virol. 2005, 79, 12231–12241. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  92. Chen, J.; Kovacs, J.M.; Peng, H.; Rits-Volloch, S.; Lu, J.; Park, D.; Zablowsky, E.; Seaman, M.S.; Chen, B. HIV-1 ENVELOPE. Effect of the cytoplasmic domain on antigenic characteristics of HIV-1 envelope glycoprotein. Science 2015, 349, 191–195. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  93. Piai, A.; Fu, Q.; Cai, Y.; Ghantous, F.; Xiao, T.; Shaik, M.M.; Peng, H.; Rits-Volloch, S.; Chen, W.; Seaman, M.S.; et al. Structural basis of transmembrane coupling of the HIV-1 envelope glycoprotein. Nat. Commun. 2020, 11, 2317. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  94. Rawat, S.S.; Zimmerman, C.; Johnson, B.T.; Cho, E.; Lockett, S.J.; Blumenthal, R.; Puri, A. Restricted lateral mobility of plasma membrane CD4 impairs HIV-1 envelope glycoprotein mediated fusion. Mol. Membr. Biol. 2008, 25, 83–94. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  95. Benkirane, M.; Jin, D.Y.; Chun, R.F.; Koup, R.A.; Jeang, K.T. Mechanism of transdominant inhibition of CCR5-mediated HIV-1 infection by ccr5delta32. J. Biol. Chem. 1997, 272, 30603–30606. [Google Scholar] [CrossRef] [PubMed]
  96. Venkatesan, S.; Petrovic, A.; Locati, M.; Kim, Y.O.; Weissman, D.; Murphy, P.M. A membrane-proximal basic domain and cysteine cluster in the C-terminal tail of CCR5 constitute a bipartite motif critical for cell surface expression. J. Biol. Chem. 2001, 276, 40133–40145. [Google Scholar] [CrossRef] [PubMed]
  97. Shaik, M.M.; Peng, H.; Lu, J.; Rits-Volloch, S.; Xu, C.; Liao, M.; Chen, B. Structural basis of coreceptor recognition by HIV-1 envelope spike. Nature 2019, 565, 318–323. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  98. Montefiori, D.C.; Roederer, M.; Morris, L.; Seaman, M.S. Neutralization tiers of HIV-1. Curr. Opin. HIV AIDS 2018, 13, 128–136. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  99. Weiss, R.A.; Verrips, C.T. Nanobodies that Neutralize HIV. Vaccines 2019, 7, 77. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  100. Liao, Z.; Cimakasky, L.M.; Hampton, R.; Nguyen, D.H.; Hildreth, J.E. Lipid rafts and HIV pathogenesis: Host membrane cholesterol is required for infection by HIV type 1. AIDS Res. Hum. Retrovir. 2001, 17, 1009–1019. [Google Scholar] [CrossRef] [PubMed]
  101. Liao, Z.; Graham, D.R.; Hildreth, J.E. Lipid rafts and HIV pathogenesis: Virion-associated cholesterol is required for fusion and infection of susceptible cells. AIDS Res. Hum. Retrovir. 2003, 19, 675–687. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, S.Y.; Aliyari, R.; Chikere, K.; Li, G.; Marsden, M.D.; Smith, J.K.; Pernet, O.; Guo, H.; Nusbaum, R.; Zack, J.A.; et al. Interferon-inducible cholesterol-25-hydroxylase broadly inhibits viral entry by production of 25-hydroxycholesterol. Immunity 2013, 38, 92–105. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  103. Zhang, J.; Zhu, Y.; Wang, X.; Wang, J. 25-hydroxycholesterol: An integrator of antiviral ability and signaling. Front. Immunol. 2023, 14, 1268104. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  104. Nieto-Garai, J.A.; Arboleya, A.; Otaegi, S.; Chojnacki, J.; Casas, J.; Fabriàs, G.; Contreras, F.X.; Kräusslich, H.G.; Lorizate, M. Cholesterol in the Viral Membrane is a Molecular Switch Governing HIV-1 Env Clustering. Adv. Sci. 2020, 8, 2003468. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  105. Whitlock, J.M.; Chernomordik, L.V. Flagging fusion: Phosphatidylserine signaling in cell-cell fusion. J. Biol. Chem. 2021, 296, 100411. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  106. Braga, L.; Ali, H.; Secco, I.; Chiavacci, E.; Neves, G.; Goldhill, D.; Penn, R.; Jimenez-Guardeño, J.M.; Ortega-Prieto, A.M.; Bussani, R.; et al. Drugs that inhibit TMEM16 proteins block SARS-CoV-2 spike-induced syncytia. Nature 2021, 594, 88–93. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  107. Finnegan, C.M.; Rawat, S.S.; Cho, E.H.; Guiffre, D.L.; Lockett, S.; Merrill, A.H., Jr.; Blumenthal, R. Sphingomyelinase restricts the lateral diffusion of CD4 and inhibits human immunodeficiency virus fusion. J. Virol. 2007, 81, 5294–5304. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  108. Vieira, C.R.; Munoz-Olaya, J.M.; Sot, J.; Jiménez-Baranda, S.; Izquierdo-Useros, N.; Abad, J.L.; Apellániz, B.; Delgado, R.; Martinez-Picado, J.; Alonso, A.; et al. Dihydrosphingomyelin impairs HIV-1 infection by rigidifying liquid-ordered membrane domains. Chem. Biol. 2010, 17, 766–775. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cell–cell fusion assays and recombinant envelope and receptor constructs. (A). Cell–cell fusion assays involved transfection of one set of cells with an Env expression construct (in red) plus a PM-targeted split GFP helix 1–10 expression construct (pcDNA3.1-PLCdeltaPH-GFP-H1-10; green), and transfection of a separate set of cells with expression vectors for CD4 (blue) and CXCR4 (pink) variants, plus a PM-targeted split GFP helix 11 expression construct (For pGFP-H11-PLCdeltaPH; green). One day post-transfection cells were seeded together onto coverslips, and scored for GFP+ syncytia two days later. (B). HIV-1 Env expression constructs included one that produced the WT HIV-1 NL4-3 envelope protein (Env-WT: pMDG-NL43-Env-WT or SVIII-Env-WT) and one that produced an Env protein (Env-753stop: pMDG-NL43-Env-753stop), truncated at Env residue 753, before the CT amphipathic helices. CD4 expression constructs included one that expressed a Myc-tagged WT human CD4 protein (CD4: pCAGGS-CD4-Myc) or a variant in which the CD4 transmembrane and cytoplasmic tail (TMCT) domains were replaced with a GPI anchor (CD4-GPI: pCAGGS-CD4-GPI). CXCR4 expression constructs included a WT human CXCR4 expression construct (CXCR4: pcDNA3-CXCR4), constructs with partial (CXCR4-318: pcDNA3-CXCR4-318Stop) or complete (CXCR4-309: pcDNA3-CXCR4-309Stop) CXCR4 cytoplasmic tail (CT) deletions, and constructs in which CT truncations were fused to the PLCδPH that preferentially binds to PI(4,5)P2 (CXCR4-318-PH and CXCR4-309-PH).
Figure 1. Cell–cell fusion assays and recombinant envelope and receptor constructs. (A). Cell–cell fusion assays involved transfection of one set of cells with an Env expression construct (in red) plus a PM-targeted split GFP helix 1–10 expression construct (pcDNA3.1-PLCdeltaPH-GFP-H1-10; green), and transfection of a separate set of cells with expression vectors for CD4 (blue) and CXCR4 (pink) variants, plus a PM-targeted split GFP helix 11 expression construct (For pGFP-H11-PLCdeltaPH; green). One day post-transfection cells were seeded together onto coverslips, and scored for GFP+ syncytia two days later. (B). HIV-1 Env expression constructs included one that produced the WT HIV-1 NL4-3 envelope protein (Env-WT: pMDG-NL43-Env-WT or SVIII-Env-WT) and one that produced an Env protein (Env-753stop: pMDG-NL43-Env-753stop), truncated at Env residue 753, before the CT amphipathic helices. CD4 expression constructs included one that expressed a Myc-tagged WT human CD4 protein (CD4: pCAGGS-CD4-Myc) or a variant in which the CD4 transmembrane and cytoplasmic tail (TMCT) domains were replaced with a GPI anchor (CD4-GPI: pCAGGS-CD4-GPI). CXCR4 expression constructs included a WT human CXCR4 expression construct (CXCR4: pcDNA3-CXCR4), constructs with partial (CXCR4-318: pcDNA3-CXCR4-318Stop) or complete (CXCR4-309: pcDNA3-CXCR4-309Stop) CXCR4 cytoplasmic tail (CT) deletions, and constructs in which CT truncations were fused to the PLCδPH that preferentially binds to PI(4,5)P2 (CXCR4-318-PH and CXCR4-309-PH).
Viruses 17 00472 g001
Figure 2. HIV-1 Env cell–cell fusion assays. Cell–cell fusion assays were performed with 293 cells as illustrated in Figure 1. Panels (AF) show coincubations of cells transfected with pGFP-H11-PLCdeltaPH plus CD4-GPI plus CXCR4-318 mixed with cells transfected with pcDNA3.1-PLCdeltaPH-GFP-H1-10 plus either Env-WT (A,B), Env-753Stop (C,D), or a mock (E,F). Panels (AF) are matched photos imaged for DAPI nuclear stain (A,C,E) and GFP (B,D,F). Note that the size bar (panel (F)) pertains to all fluorescent images (AF). In panel (G), numbers of syncytia per 0.4 mm2 are averaged with standard deviations from five images each of coincubations of cells co-transfected with split GFP constructs, plus the indicated Env or CD4 plus CXCR4 expression construct variants. Note that numbers of syncytia for Env-753 were approximately tenfold higher than syncytia for Env-WT, and that syncytia for Env-WT were approximately twenty-fold higher than syncytia for the Env-minus (No Env) control. Differences between the sets were highly significant (p < 0.001), and were obtained by conversion of means and standard deviations to Z values for probability calculations.
Figure 2. HIV-1 Env cell–cell fusion assays. Cell–cell fusion assays were performed with 293 cells as illustrated in Figure 1. Panels (AF) show coincubations of cells transfected with pGFP-H11-PLCdeltaPH plus CD4-GPI plus CXCR4-318 mixed with cells transfected with pcDNA3.1-PLCdeltaPH-GFP-H1-10 plus either Env-WT (A,B), Env-753Stop (C,D), or a mock (E,F). Panels (AF) are matched photos imaged for DAPI nuclear stain (A,C,E) and GFP (B,D,F). Note that the size bar (panel (F)) pertains to all fluorescent images (AF). In panel (G), numbers of syncytia per 0.4 mm2 are averaged with standard deviations from five images each of coincubations of cells co-transfected with split GFP constructs, plus the indicated Env or CD4 plus CXCR4 expression construct variants. Note that numbers of syncytia for Env-753 were approximately tenfold higher than syncytia for Env-WT, and that syncytia for Env-WT were approximately twenty-fold higher than syncytia for the Env-minus (No Env) control. Differences between the sets were highly significant (p < 0.001), and were obtained by conversion of means and standard deviations to Z values for probability calculations.
Viruses 17 00472 g002
Figure 3. Cell surface expression of Env proteins. (A,B) 293 cells transfected with the indicated Env expression plasmids were subjected to indirect immunofluorescent detection of surface Env proteins in the absence of cell permeabilization with the human 2G12 anti-Env antibody as a primary antibody, and a fluorescent anti-human IgG secondary antibody. Note that the size bar in panel (B) pertains to both panels, and that the exposure times for panels (A,B) were 10 and 67 ms, respectively. Panels (C,D) respectively indicate percentages of fluorescent cells and relative brightness values for fluorescent cells, using the cells transfected with the indicated Env expression constructs and either VRC03 or 2G12 anti-Env antibodies for detection. Averages and standard deviations were obtained from a minimum of 9 images for panel (C), a minimum of 14 cells for panel (D) (VRC03), and a minimum of 36 cells for panel (D) (2G12). Differences between all pairs were highly significant (p < 0.001), and were obtained by conversion of means and standard deviations to Z values for probability calculations.
Figure 3. Cell surface expression of Env proteins. (A,B) 293 cells transfected with the indicated Env expression plasmids were subjected to indirect immunofluorescent detection of surface Env proteins in the absence of cell permeabilization with the human 2G12 anti-Env antibody as a primary antibody, and a fluorescent anti-human IgG secondary antibody. Note that the size bar in panel (B) pertains to both panels, and that the exposure times for panels (A,B) were 10 and 67 ms, respectively. Panels (C,D) respectively indicate percentages of fluorescent cells and relative brightness values for fluorescent cells, using the cells transfected with the indicated Env expression constructs and either VRC03 or 2G12 anti-Env antibodies for detection. Averages and standard deviations were obtained from a minimum of 9 images for panel (C), a minimum of 14 cells for panel (D) (VRC03), and a minimum of 36 cells for panel (D) (2G12). Differences between all pairs were highly significant (p < 0.001), and were obtained by conversion of means and standard deviations to Z values for probability calculations.
Viruses 17 00472 g003
Figure 4. Infection of Env-expressing cells via fusion in reverse. (A) Target 293 cells expressing different Env variants are infected with a β-gal-transducing, HIV-1-based, lentivirus vector pseudotyped with a GPI-anchored CD4 variant (CD4-GPI) plus a C-terminally-truncated CXCR4 protein (CXCR4-318). Receptor plus co-receptor binding to cellularly expressed Env activate the Env fusion machinery, resulting in virus infection. (B) Shown are β-gal activity-derived infectivities of cells expressing Env-753Stop, Env-WT, or no Env (mock), normalized to the Env-753Stop cells. Note that results are averaged from five independent experiments each, are plotted on a log scale graph to facilitate comparison, and that standard deviations for the Env-753Stop and mock cells were too small to be observed on the graph. (C) Cells expressing the indicated Env proteins were infected with CD4-GPI plus CXCR4-318 pseudotyped β-gal-transducing HIV-1 lentivirus vectors in the absence (753, WT) or presence (753 + T20, WT + T20) of HIV-1 Env fusion inhibitor T20. Results for pairs of infections are normalized to the infectivities of viruses in the absence of T20. Averages and standard deviations derive from three separate infections, and differences between 753 and WT and WT and mock were highly significant (p < 0.001), as were differences between untreated and treated samples. Probabilities were obtained by conversion of means and standard deviations to Z values, which were used for calculations.
Figure 4. Infection of Env-expressing cells via fusion in reverse. (A) Target 293 cells expressing different Env variants are infected with a β-gal-transducing, HIV-1-based, lentivirus vector pseudotyped with a GPI-anchored CD4 variant (CD4-GPI) plus a C-terminally-truncated CXCR4 protein (CXCR4-318). Receptor plus co-receptor binding to cellularly expressed Env activate the Env fusion machinery, resulting in virus infection. (B) Shown are β-gal activity-derived infectivities of cells expressing Env-753Stop, Env-WT, or no Env (mock), normalized to the Env-753Stop cells. Note that results are averaged from five independent experiments each, are plotted on a log scale graph to facilitate comparison, and that standard deviations for the Env-753Stop and mock cells were too small to be observed on the graph. (C) Cells expressing the indicated Env proteins were infected with CD4-GPI plus CXCR4-318 pseudotyped β-gal-transducing HIV-1 lentivirus vectors in the absence (753, WT) or presence (753 + T20, WT + T20) of HIV-1 Env fusion inhibitor T20. Results for pairs of infections are normalized to the infectivities of viruses in the absence of T20. Averages and standard deviations derive from three separate infections, and differences between 753 and WT and WT and mock were highly significant (p < 0.001), as were differences between untreated and treated samples. Probabilities were obtained by conversion of means and standard deviations to Z values, which were used for calculations.
Viruses 17 00472 g004
Figure 5. Fusion in reverse infectivities for different Env, receptor, and co-receptor combinations. 293 cells expressing the 753Stop (A) or WT HIV-1 Env (B) were infected with equivalent volumes of β-gal-transducing HIV-1 particles carrying the indicated receptor plus co-receptor combinations (CD4, CD4-GPI [gpi],—[mock], CXCR4, 318 [CXCR4-318], 309 [CXCR4-309], 318PH [CXCR4-318-PH], 309PH [CXCR4-309-PH]). At 72 h post-infection, cells were subjected to β-gal assays and β-gal activities are plotted as infectivities relative to viruses carrying the CD4-GPI/CXCR4-318 receptor/co-receptor combinations. Averages and standard deviations derive from the following numbers of experiments: 753/gpi/318, 9; 753/gpi/-, 2; 753/-/318, 2; 753/CD4/325, 2; 753/gpi/CXCR4, 4; 753/gpi/309, 3; 753/gpi/318PH, 5; 753/gpi/309PH, 5; WT/gpi/318, 9; WT/gpi/-, 2; WT/-/318, 2; WT/CD4/318, 3; WT/gpi/CXCR4, 6; WT/gpi/309, 4; WT/gpi/318PH, 5; WT/gpi/309PH, 5. Note that differences between the gpi/318 samples versus the gpi/-, -/318, and CD4/318 samples had probabilities of p < 0.001, as determined by conversion of means and standard deviations to Z values for probability calculations.
Figure 5. Fusion in reverse infectivities for different Env, receptor, and co-receptor combinations. 293 cells expressing the 753Stop (A) or WT HIV-1 Env (B) were infected with equivalent volumes of β-gal-transducing HIV-1 particles carrying the indicated receptor plus co-receptor combinations (CD4, CD4-GPI [gpi],—[mock], CXCR4, 318 [CXCR4-318], 309 [CXCR4-309], 318PH [CXCR4-318-PH], 309PH [CXCR4-309-PH]). At 72 h post-infection, cells were subjected to β-gal assays and β-gal activities are plotted as infectivities relative to viruses carrying the CD4-GPI/CXCR4-318 receptor/co-receptor combinations. Averages and standard deviations derive from the following numbers of experiments: 753/gpi/318, 9; 753/gpi/-, 2; 753/-/318, 2; 753/CD4/325, 2; 753/gpi/CXCR4, 4; 753/gpi/309, 3; 753/gpi/318PH, 5; 753/gpi/309PH, 5; WT/gpi/318, 9; WT/gpi/-, 2; WT/-/318, 2; WT/CD4/318, 3; WT/gpi/CXCR4, 6; WT/gpi/309, 4; WT/gpi/318PH, 5; WT/gpi/309PH, 5. Note that differences between the gpi/318 samples versus the gpi/-, -/318, and CD4/318 samples had probabilities of p < 0.001, as determined by conversion of means and standard deviations to Z values for probability calculations.
Viruses 17 00472 g005
Figure 6. CD4 incorporation into virions. (A). Media samples from 293 cells transfected with psPAX2 (psPAX) plus the CD4 or CD4-gpi expression constructs were pelleted through 20% sucrose cushions, separated by electrophoresis and immunoblotted for detection of CA and CD4 immunoreactive proteins. PrGag, p41 (Gag intermediate), CA, and CD4 proteins were identified based on their immunoreactivities and migration mobilities relative to protein molecular weight standards run in parallel. (B,C). Cell samples (top panels) and media samples that were pelleted through 30% sucrose cushions (bottom panels) were collected from cells transfected with the indicated constructs, separated by electrophoresis, and immunoblotted for detection of PrGag, p41 (Gag intermediate) and CA (B) or CD4 and CD4-gpi (C). Note that viral CD4-to-CA ratios (expressed as percentages) were 33% for (A), and 25% for (B,C); while viral CD4-gpi-to-CA ratios were 26% for (A), and 34% for (B,C).
Figure 6. CD4 incorporation into virions. (A). Media samples from 293 cells transfected with psPAX2 (psPAX) plus the CD4 or CD4-gpi expression constructs were pelleted through 20% sucrose cushions, separated by electrophoresis and immunoblotted for detection of CA and CD4 immunoreactive proteins. PrGag, p41 (Gag intermediate), CA, and CD4 proteins were identified based on their immunoreactivities and migration mobilities relative to protein molecular weight standards run in parallel. (B,C). Cell samples (top panels) and media samples that were pelleted through 30% sucrose cushions (bottom panels) were collected from cells transfected with the indicated constructs, separated by electrophoresis, and immunoblotted for detection of PrGag, p41 (Gag intermediate) and CA (B) or CD4 and CD4-gpi (C). Note that viral CD4-to-CA ratios (expressed as percentages) were 33% for (A), and 25% for (B,C); while viral CD4-gpi-to-CA ratios were 26% for (A), and 34% for (B,C).
Viruses 17 00472 g006
Figure 7. Infection of HIV-1-infected cells. Jurkat T cells, HIV-1-infected J1.1 Jurkat cells, and J1.1 cells induced with tumor necrosis factor alpha (J1.1 + TNFa) were subjected to immunoblot detection of Gag proteins (A) with an anti-CA antibody, immunoblot detection of Env proteins (B) with an anti-gp41(TM) antibody, and infection with a lentivirus vector (C) generated by transfection of 293 cells with psPAX2, pLVX-puro-Xho-ATG-β-gal, CD4-GPI, and CXCR4-328 plasmids. (A,B) Migration positions of PrGag, CA, gp160 and gp41 proteins are indicated, as are molecular weight standards (in kDa) that were electrophoresced in parallel. (C) Infectivity results represent normalized transduced β-gal activity readings normalized to results for induced J1.1 cells. Averages and standard deviations derive from duplicate (Jurkat, J1.1) or triplicate (J1.1 + TNFα) infections, and differences between Jurkat and J1.1, as well as J1.1 versus J1.1 + TNFα were highly significant (p < 0.001), as determined by conversion of means and standard deviations to Z values that were used in probability calculations.
Figure 7. Infection of HIV-1-infected cells. Jurkat T cells, HIV-1-infected J1.1 Jurkat cells, and J1.1 cells induced with tumor necrosis factor alpha (J1.1 + TNFa) were subjected to immunoblot detection of Gag proteins (A) with an anti-CA antibody, immunoblot detection of Env proteins (B) with an anti-gp41(TM) antibody, and infection with a lentivirus vector (C) generated by transfection of 293 cells with psPAX2, pLVX-puro-Xho-ATG-β-gal, CD4-GPI, and CXCR4-328 plasmids. (A,B) Migration positions of PrGag, CA, gp160 and gp41 proteins are indicated, as are molecular weight standards (in kDa) that were electrophoresced in parallel. (C) Infectivity results represent normalized transduced β-gal activity readings normalized to results for induced J1.1 cells. Averages and standard deviations derive from duplicate (Jurkat, J1.1) or triplicate (J1.1 + TNFα) infections, and differences between Jurkat and J1.1, as well as J1.1 versus J1.1 + TNFα were highly significant (p < 0.001), as determined by conversion of means and standard deviations to Z values that were used in probability calculations.
Viruses 17 00472 g007
Figure 8. Effects of lipid variations on fusion in reverse infections. (A) The HIV-1 viral membrane is highly enriched in cholesterol (CHOL), and has substantial amounts of ceramide (CER). The inner leaflets are enriched for phosphatidylserine (PS), phosphatidylinositol-4,5-bisphosphate (PI[4,5]P2; PIP2), and phosphatidylethanolamine (PE), while the outer leaflet is enriched in sphingomyelin (SM), hexosylceramides (HEX), and saturated phosphatidylcholines (PCs). (BD) Infectivities of equivalent volumes of virus were scored via β-gal activities, and were normalized either to untreated (−) samples, or for convenience in the panel (D) CD/CX infections, to viruses from CerS−/− cells. In all panels, CD/CX viruses refer to lentivirus vectors generated by co-transfection of 293 cells with a GagPol expression vector (psPAX2), a β-gal-transducing vector (pLVX-puro-Xho-ATG-β-gal), as well as CD4-GPI and CXCR4-318 plasmids. Also, in all panels, Env-WT and Env-753 targets cells refer to 293 cells transfected to express those proteins. For panels (B,C), WT and 753 viruses are the NL4-3 variants and were used to infect TZM-bl reporter cells; for panel (D), WT refers to viruses produced from cells transfected with psPAX2, pLVX-puro-Xho-ATG-β-gal, plus a WT Env expression vector, while targets were 293 cells transfected to express the CD4-GPI and CXCR4-318 proteins. In panel (B), AME refers to treatment of viruses with 10 mM AME prior to infections. In panel (C), TMEM refers to co-transfection of virus producing cells with (+) or without (−) the mTMEM-GY scramblase expression vector. In panel (D), CerS2−/− pertains to producing viruses either from 293 cells (−) or from 293-CerS2−/− knockout cells (+). For panels (BD), averages and standard deviations are as indicated, noting that where error bars are missing, it is because the standard deviations were too small to be depicted. The following significant differences were obtained by conversion of means and standard deviations to Z values for probability calculations: Panel (B) WT/TZMBL, AME −/+, p < 0.001; Panel (B) CDCX/Env-WT, AME −/+, p < 0.001; Panel C all pairs p < 0.001; Panel (D) CDCX/Env-WT, CerS2−/−, −/+, p < 0.01; all other Panel D pairs, p < 0.001. Note that in panel (D), if normalizations were based on CDCX viruses produced from WT 293 cells and infections of Env-753Stop-expressing cells, infectivities of CDCX viruses from virus–cell combinations would be as follows: from WT 293 cells into Env-753Stop cells, 100%; from CerS2−/− cells into Env-753Stop cells 500%; from WT 293 cells into Env-WT cells, 8%; from CerS2−/− cells into Env-WT cells, 19%.
Figure 8. Effects of lipid variations on fusion in reverse infections. (A) The HIV-1 viral membrane is highly enriched in cholesterol (CHOL), and has substantial amounts of ceramide (CER). The inner leaflets are enriched for phosphatidylserine (PS), phosphatidylinositol-4,5-bisphosphate (PI[4,5]P2; PIP2), and phosphatidylethanolamine (PE), while the outer leaflet is enriched in sphingomyelin (SM), hexosylceramides (HEX), and saturated phosphatidylcholines (PCs). (BD) Infectivities of equivalent volumes of virus were scored via β-gal activities, and were normalized either to untreated (−) samples, or for convenience in the panel (D) CD/CX infections, to viruses from CerS−/− cells. In all panels, CD/CX viruses refer to lentivirus vectors generated by co-transfection of 293 cells with a GagPol expression vector (psPAX2), a β-gal-transducing vector (pLVX-puro-Xho-ATG-β-gal), as well as CD4-GPI and CXCR4-318 plasmids. Also, in all panels, Env-WT and Env-753 targets cells refer to 293 cells transfected to express those proteins. For panels (B,C), WT and 753 viruses are the NL4-3 variants and were used to infect TZM-bl reporter cells; for panel (D), WT refers to viruses produced from cells transfected with psPAX2, pLVX-puro-Xho-ATG-β-gal, plus a WT Env expression vector, while targets were 293 cells transfected to express the CD4-GPI and CXCR4-318 proteins. In panel (B), AME refers to treatment of viruses with 10 mM AME prior to infections. In panel (C), TMEM refers to co-transfection of virus producing cells with (+) or without (−) the mTMEM-GY scramblase expression vector. In panel (D), CerS2−/− pertains to producing viruses either from 293 cells (−) or from 293-CerS2−/− knockout cells (+). For panels (BD), averages and standard deviations are as indicated, noting that where error bars are missing, it is because the standard deviations were too small to be depicted. The following significant differences were obtained by conversion of means and standard deviations to Z values for probability calculations: Panel (B) WT/TZMBL, AME −/+, p < 0.001; Panel (B) CDCX/Env-WT, AME −/+, p < 0.001; Panel C all pairs p < 0.001; Panel (D) CDCX/Env-WT, CerS2−/−, −/+, p < 0.01; all other Panel D pairs, p < 0.001. Note that in panel (D), if normalizations were based on CDCX viruses produced from WT 293 cells and infections of Env-753Stop-expressing cells, infectivities of CDCX viruses from virus–cell combinations would be as follows: from WT 293 cells into Env-753Stop cells, 100%; from CerS2−/− cells into Env-753Stop cells 500%; from WT 293 cells into Env-WT cells, 8%; from CerS2−/− cells into Env-WT cells, 19%.
Viruses 17 00472 g008
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alfadhli, A.; Barklis, R.L.; Tafesse, F.G.; Barklis, E. Analysis of Factors That Regulate HIV-1 Fusion in Reverse. Viruses 2025, 17, 472. https://doi.org/10.3390/v17040472

AMA Style

Alfadhli A, Barklis RL, Tafesse FG, Barklis E. Analysis of Factors That Regulate HIV-1 Fusion in Reverse. Viruses. 2025; 17(4):472. https://doi.org/10.3390/v17040472

Chicago/Turabian Style

Alfadhli, Ayna, Robin Lid Barklis, Fikadu G. Tafesse, and Eric Barklis. 2025. "Analysis of Factors That Regulate HIV-1 Fusion in Reverse" Viruses 17, no. 4: 472. https://doi.org/10.3390/v17040472

APA Style

Alfadhli, A., Barklis, R. L., Tafesse, F. G., & Barklis, E. (2025). Analysis of Factors That Regulate HIV-1 Fusion in Reverse. Viruses, 17(4), 472. https://doi.org/10.3390/v17040472

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop