Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 21.
Published in final edited form as: Cell Chem Biol. 2024 Jan 16;31(3):487–501.e7. doi: 10.1016/j.chembiol.2023.12.017

Bioorthogonal click labeling of an amber-free HIV-1 provirus for in-virus single molecule imaging

Yuanyun Ao 1, Jonathan R Grover 2, Levi Gifford 3, Yang Han 1, Guohua Zhong 1, Revansiddha Katte 1, Wenwei Li 2, Rajanya Bhattacharjee 4,5, Baoshan Zhang 6, Stephanie Sauve 7, Wenyi Qin 1, Dibya Ghimire 1, Anzarul Haque 1, James Arthos 8, Mahmoud Moradi 7, Walther Mothes 2, Edward A Lemke 4,9, Peter D Kwong 6, Gregory B Melikyan 3, Maolin Lu 1,10,*
PMCID: PMC10960674  NIHMSID: NIHMS1959381  PMID: 38232732

Summary

Structural dynamics of HIV-1 envelope (Env) glycoprotein mediates cell entry and facilitates immune evasion. Single-molecule FRET using peptides for Env labeling revealed structural dynamics of Env, but peptide use risks potential effects on structural integrity/dynamics. While incorporating noncanonical amino acids (ncAAs) into Env by amber stop-codon suppression, followed by click chemistry, offers a minimally invasive mean, this approach proved technically challenging for HIV-1. Here, we developed an intact amber-free HIV-1 system that overcomes hurdles of preexisting viral amber codons. We achieved dual-ncAA incorporation into Env on amber-free virions, enabling smFRET studies of click-labeled Env that validated the previous peptide-based labeling approaches by confirming the intrinsic propensity of Env to dynamically sample multiple conformational states. Amber-free click-labeled Env also enables real-time tracking of single virion internalization and trafficking in cells. Our system thus permits in-virus bioorthogonal labeling of proteins, compatible with studies of virus entry, trafficking, and egress from cells.

In Brief

Ao et al. developed an intact amber-free HIV-1 system that enables minimally invasive Env tagging by genetic code expansion and in-virus bioorthogonal click labeling for single-molecule FRET structural dynamics and advanced microscopy studies of virus entry, trafficking, and egress in living cells.

Graphical Abstract:

graphic file with name nihms-1959381-f0001.jpg

Introduction

The sparsely distributed envelope (Env) glycoprotein on human immunodeficiency virus 1 (HIV-1) is responsible for the host cellular receptor binding and cell entry while escaping neutralizing antibodies. Consequently, Env is a main target for designing vaccines, antibodies, and antiviral drugs. Env interacts with the host cellular receptor CD4 and coreceptor CCR5/CXCR4 to mediate viral membrane fusion into target cells through a series of large-scale conformational changes.110 Functional Env is a homotrimer-of-heterodimer consisting of the gp120 exterior subunits and gp41 transmembrane subunits.1116 The gp120 subunit binds the primary receptor CD4 and coreceptor CCR5/CXCR4 on the susceptible target cells.4 The binding of CD4 induces opening of gp120 and the formation of binding sites for coreceptors. After binding to CCR5/CXCR4, additional conformational changes in gp120 are relayed to the membrane-anchored gp41 subunit. Subsequent conformational refolding of gp41 induces fusion of the viral membrane with the host cell membrane.1722 Besides mediating viral entry, Env harbors virus-specific antigenic epitopes and thus serves as the main target for antibody responses during viral infection.1,2327 Nevertheless, Env has evolved to successfully evade the host immune responses through multiple mechanisms, including extensive glycan shielding, regions of hypermutation, and conformational flexibility.2831 Collectively, Env exhibits a high capacity to undergo conformational switching upon interacting with host cells, which is essential for virus entry and immune evasion during HIV-1 infection life cycle. Thus, a better understanding of conformational dynamics of native Env on infectious viruses will inform the development of HIV-1 prevention and treatment strategies, such as vaccines, antibodies, and small-molecule inhibitors.1,710

Single-molecule Förster resonance energy transfer (smFRET) imaging has provided unique insights into the HIV-1 entry mechanism by visualizing real-time conformational changes of Env tagged with a donor/acceptor pair of fluorophores undergoing distance-sensitive energy transfer on intact virions (Figure 1A).3235 Attaching donor/acceptor fluorophores to two sites of a molecule within their Förster distance of a few nanometers is a prerequisite for smFRET.36 We have used enzymatic labeling for smFRET imaging of HIV-1 Env,32,3739 in which a customized Cy3/Cy5 FRET pair was used to label gp120 variable loops V1 and V4, respectively. This approach uses enzymes that site-specifically recognize the introduced short peptides (Q3 and A1) and transfer dye-conjugated substrates to specific residues within these peptides.40,41 This approach requires 6 and 12 amino acids insertion into the gp120 V1 and V4, respectively. These insertions have generated concerns regarding the potential impact on structural integrity and unpredictable interaction with antibodies.42 Although these concerns have been addressed by extensive functional validations,32,33,43 there are still some unreconciled differences between virus-associated states of Env identified by smFRET810 and the structures revealed by cryo-electron microscopy (cryo-EM).7,39 The pretriggered conformational state of Env reported by smFRET using peptide labeling tags has not been structurally observed by cryoEM.7,39 There was a concern that the existence of this “missing” state might be promoted by peptide tags used for labeling. Thus, a minimal tagging approach for attaching dyes to Env is critical to directly address this concern. Such a labeling approach holds promise for further advancing our knowledge of Env-mediated virus entry in future smFRET studies.

Figure 1. Experimental design for minimally invasive smFRET of HIV-1 Env on the intact virion using amber suppression and bioorthogonal click labeling.

Figure 1.

(A) smFRET of Env trimer in the virus context. The Env structure is adapted from PDB 4ZMJ (dually labeled gp120, brown; non-labeled gp120, cyan; gp41, blue).

(B, C) Theoretical (B) and experimental (C) schematics of using two-step minimal genetically encoded amber (TAG) tags for fluorescence labeling - amber suppression (step 1) followed by click chemistry (step 2). Amber suppressor tRNA and tRNA synthetase (tRNAPyl/NESPylRSAF) introduce ncAA trans-cyclooct-2-en – L – lysine (TCO*) into the desired TAG position in Env (POI) on an assembled intact virion generated in mammalian cells. The supply of TCO* to the transfected cells allows the incorporation of TCO* at the TAG insertion site in Env, to which clickable fluorophores can be attached.

(D) Bioorthogonal click labeling via SPIEDAC. Tetrazine-conjugated Cy3 and Cy5 derivatives (LD555-TTZ and LD655-TTZ) react with the strained alkene of the TCO* via strain-promoted inverse electron-demand Diels-Alder cycloaddition (SPIEDAC). Fluorophores/dyes are colored green and red, respectively.

Design

The recently developed amber (TAG) stop-codon suppression through genetic code expansion in mammalian cells and copper-free bioorthogonal click chemistry to label dyes on proteins by only altering a single amino acid4450 seems an ideal labeling strategy for Env. This approach includes suppressing amber stop codons with clickable noncanonical/unnatural amino acids (ncAAs) by an engineered suppressor tRNA and its cognate tRNA synthetase (tRNA/RS) and the subsequent click labeling of dyes onto ncAAs.44,51 The use of amber suppression, advantageous over peptide tags, minimizes the impact of tagging on the structural integrity of proteins, thus offering a less invasive means for site-specific labeling of Env (Table S1). Despite its obvious advances, click labeling of HIV-1 proteins in their close-to-native states has been rare, except for the two published pioneering reports.44,52 Sakin and colleagues have generated singly click-labeled Env expressed on mammalian cells for monitoring Env mobility at the plasma membrane.44 However, this pioneered study did not achieve click labeling of Env in the viral context, due in part to low single ncAA incorporation into Env at each of the three optimized sites.44 Single-site click labeling of capsids in the viral context has been recently achieved, enabling tracking the journey of click-labeled capsids from the plasma membrane to the nucleus.52 The drastic differences in the number of copies of capsid vs. Env (thousands vs. a few) in/on each virion is likely the reason singly click-labeled Env in the viral context has been so difficult. In addition, smFRET requires dually click-labeled Env at proximal sites, contingent upon successful incorporation of two ncAAs into a single gp120 protomer that does not interfere with efficient virus assembly and release. This stringency imposes additional technical barriers. Thus, coupling a single or a pair of dyes to Env on virions through genetic code expansion–click chemistry has been lacking due to lower copies of Env on HIV-1, in addition to the low amber-suppression efficiency in mammalian cells.

Here, we aimed to establish a two-step minimally invasive labeling strategy for smFRET imaging on the virus via genetically encoding ncAA tags in gp120 and click labeling ncAAs with Cy3/Cy5 donor/acceptor dyes for prism-based total internal reflection fluorescence (prism-TIRF) microscopy (Figure 1). Incorporation of ncAAs into Env on intact HIV-1 virions can be achieved by co-transfecting HEK293T cells with the amber suppression machinery plasmid encoding for tRNAPyl/NESPylRSAF and an amber-tagged Env plasmid, in the presence of ncAAs.44,46,53,54 We introduced ncAA trans-cyclooct-2-ene-L-lysine (the A isomer, hereafter abbreviated TCO*) at an amber (TAG) stop codon engineered into gp120 (Figure 1B and C). TCO* reacts with tetrazine-conjugated Cy3 or Cy5 fluorophores by click chemistry via strain-promoted inverse electron-demand Diels-Alder cycloaddition (SPIEDAC, Figure 1D).44,48

Results

Construction of dually amber-suppressed click-labeled Env on intact HIV-1 virions

We screened for amber suppression (ncAA incorporation) efficiency of Env TAG variants in the presence of tRNAPyl/NESPylRSAF in HEK293T by comparing the HIV-1Q23 BG505 5557 infectivity on TZM-bl cells with the counterpart without tRNAPyl/NESPylRSAF. To identify amber-tolerable positions in gp120, we generated four full-length virus Env TAG variants that each carried single TAG insertion at different sites within V1 loop, denoted as N133TAG, N136TAG, N137TAG, and T139TAG. The amber-inserted positions were at the same or adjacent to the insertion sites of peptide tags.33,43 N136TAG showed better amber tolerance than the other three, as manifested by a higher ratio of infectivity under amber-suppression vs. non-suppression conditions (Figure S1). We combined N136TAG with six amber insertion sites in the gp120 V4 loop and generated six dually amber-tagged HIV-1Q23 BG505 clones. Among six clones, N136TAG S401TAG, and N136TAG S413TAG can better tolerate amber insertions in Env by showing slight increases in infectivity under suppression conditions compared to non-suppression conditions (Figure S1). Attempts to visualize truncated (at Amber stop) and suppressed full-length Env by western blotting failed, likely because of insufficient virus particles released and low suppression efficiency. Related, we noticed that HIV-1Q23 BG505 wild-type infectivity exhibited an approximate 50- to 100-fold reduction (Figures 2 and S1) in the presence of the amber suppressor system compared to the results without. This reduction had limited our ability to screen amber-tolerable sites but provided a possible clue for improving amber suppression efficiency and ncAA incorporation into Env.

Figure 2. Construction and validation of an amber-free primary HIV-1 system.

Figure 2.

(A) Alternations of four amber TAG termination codons of pol, vif, vpu, rev open reading frames (ORFs) in full-length wildtype HIV-1Q23 BG505 to ochre TAA termination codons, generating an amber-free HIV-1 genome.

(B–D) Amber-free HIV-1Q23 BG505 yields an approximate 10-fold increase in released infectivity (B) and an enhanced Env expression (C) and retains similar susceptibility to potent dodecameric CD4 molecules (sCD4D1D2–Igαtp, D) compared to the amber-abundant wildtype counterpart. (B) Release of virus infectivity (mean ± SD) of the mutants carrying a termination codon change at Pol from amber to ochre (TAG to TAA) (Left), and amber-free viral particles (Right) in comparison to that of the wildtype counterpart. The red arrow points to a 5- to 10-fold increase in infectivity caused by a single termination codon change at Pol. For comparisons, infectivity for the wildtype HIV-1 was measured separately and thus shown twice in graph (B). Where indicated as “Amber-Suppression” or “S,” a plasmid encoding tRNAPyl/NESPylRSAF was co-transfected with the HIV-1 genome, and the TCO* at 250 μM was added to the media. In contrast, only HIV-1 genome was transfected under the “Non-Suppression” or “NS” condition (grey bar in B). WT or wildtype, the original amber-abundant HIV-1Q23 BG505.

(C) Env processing and incorporation (Figure S2) of viral particles were assessed by SDS-PAGE of virus supernatants, followed by western blotting using the polyclonal anti-gp120 and anti-HIV-IG. Repeated twice.

(D) Neutralization curves (mean ± SD) of amber-free and amber-abundant viral particles by SCD4D1D2-Igαtp molecules.

Development of an intact amber-free HIV-1 system for improving amber suppression efficiency in the target protein

We then explored the cause for 50 to 100-fold reduction of viral infectivity. Analysis of the HIV-1 Q23 genome revealed that the full-length HIV-1Q23 BG505 contains four pre-existing amber termination codons in the Pol, Vif, Vpu, and Rev open reading frames (Figure 2A and Table S2). These ambers might incorporate TCO* by amber suppressors against intrinsic cellular translation machinery. Consequently, the read-through of supposed-to-terminate-translation amber codons is problematic through the generation of additional amino acids at the C termini of the above proteins before termination that could exhibit dominant-negative effects (Table S2). To test our hypothesis, given the critical role of Pol in the HIV-1 infection lifecycle, our first attempt was made to change amber to ochre (TAG to TAA) in Pol. Strikingly, a single TAG-to-TAA (amber to ochre) alternation in Pol improved infectivity under the suppression condition by approximately 5 to 10-fold (Figure 2B - left), confirming significant issues arising from the preexisting ambers in the HIV-1 genome. Thus, we reasoned that the previously observed unsatisfying efficiency of amber suppression and the resulting undetectable ncAA incorporated Env on gels were likely in part due to problematic suppression of preexisting ambers in HIV-1.

We then constructed an amber-free HIV-1 system by modifying all four termination codons from amber to ochre presented in the Pol, Vif, Vpu, and Rev genes (Figure 2A). A more than 10-fold increase in infectivity was obtained with the amber-free HIV-1Q23 BG505 construct, compared with the wildtype – amber-abundant (Figure 2B - right). Env expression and incorporation into virions of amber-free and amber-abundant HIV-1 constructs were further verified by immunoblots of virus supernatants using the polyclonal anti-gp120 and anti-HIV IgG (Human serum). Consistently, gp120 and Gag/p24 expression levels were higher in amber-free than in amber-abundant virions under the same suppression conditions (Figures 2C and S2). In control experiments, amber-free virions were as sensitive to neutralization by a potent soluble dodecameric sCD4D1D2–Igαtp as wild-type virions were (Figure 2D). Thus, the developed amber-free system provides an optimal backbone for inserting genetically encoded amber tags in Env.

Intact amber-free HIV-1 carrying clickable dual-ncAA Env retains sensitivities to trimer-specific neutralizing antibodies

We next reconstructed and reevaluated amber-suppression efficiencies of previously screened Env TAG variants in the new amber-free backbone. We identified thus far superior amber-tolerable positions: N136TAG in the V1 loop, S401TAG and S413TAG in the V4 loop (Figure S3). We generated four dually tagged Env constructs in the context of an intact amber-free HIV-1, including our choice of two dual-amber N136TAG S401TAG and N136TAG S413TAG and two hybrid amber/peptide N136TAG V4A1 and V1Q3 S401TAG mutants as comparisons (Figure 3A). Single-amber Env variants N136TAG, S401TAG, and S413TAG retained ~ 30% of infectivity (suppression efficiency) compared to the wildtype (Figure 3B), similar to the reported 5 ~ 20% infectivity of three different NL4–3 Env modified with single amber introduced into gp120.44 Hybrid amber/peptide N136TAG V4A1 and V1Q3 S401TAG showed ~20% suppression efficiency (Figure 3B, left and middle panels). Comparable to infectivity of hybrid Env, dual-amber Env (N136TAG S401TAG and N136TAG S413TAG) were successfully suppressed with efficiencies of 10 to 20% relative to the wild-type virus (Figure 3B, right panel). In contrast to bacteria, the efficiency of amber suppression or ncAA incorporation in mammalian cells is known to be challenging,44,46,58, and for Env it might be unlikely to reach 100%. Truncated Env proteins due to translation termination following incomplete suppression might exhibit dominant-negative effects on Env trimer folding and reduce infectivity. Therefore, it is not unexpected that the infectivity of virus carrying amber-tagged virus Env can be somewhat lower than peptide-tagged virus Env (V1Q3 V4 A1 tags on BG505 did not affect viral infectivity in our previous study43). Western blotting of post-transfected cell supernatants revealed that all four dually tagged Env variants were successfully suppressed, proteolytically processed (gp120 signal), and incorporated onto amber-free virions, in contrast to no traceable Env expression under non-suppression conditions (Figure 3C, top). By comparison, both gp120 and gp160 expressions were observed in post-transfected cell lysates (Figure 3C, bottom). The presence of gp160 in cell lysate and its absence in cell supernatant implies that precursor gp160 Env is largely (if not entirely) proteolytically cleaved to produce mature gp120/gp41 on the released virions. The morphology of packaged virus particles is unaffected in 2D EM images acquired by cryo-electron tomography (Figure 3D). Nanoparticle Tracking Analysis (NTA) showed the viral particles with a mode diameter size of 109.2 ± 15.9, 125.7 ± 3.4, and 121.8 ± 4.2 nm for amber-free virions carrying no-amber or two dual-amber Envs, respectively (Figure S4). These mode diameters are close to the diameter of 120 nm reported for native HIV particles59, suggesting the normal assembly of ncAA-incorporated virions.

Figure 3. Efficient ncAA incorporation into dually amber-tagged Env on the amber-free HIV-1 virus that retains sensitivities to trimer-specific neutralizing antibodies.

Figure 3.

(A) Scheme showing tag insertion sites and denotations of labeling tags in BG505 gp120 subunit.

(B) Release of infectivity (mean ± SD) on TZM-bl cells from HEK293T cells transfected with plasmids encoding singly amber-tagged, hybrid peptide/amber-tagged, dually amber-tagged EnvBG505 in the context of an amber-free Q23 backbone (HIV-1Q23 BG505).

(C) Env processing and incorporation of amber-free, dually amber-tagged Env (N136TAG S401TAG and N136TAG S413TAG), or hybrid peptide/amber-tagged Env (N136TA V4A1 and V1Q3 S401TAG) viral particles. Proteins were analyzed by immunoblotting using a polyclonal antiserum against HIV-1 gp120.

(D) A representative cryoET tomographic slice of amber-free and two dually amber-tagged HIV-1 viral particles, respectively. Scale bar: 50 nm.

(E) Neutralization curves (mean ± SD) of amber-free and dually amber-tagged HIV-1 viral particles by trimer-specific antibodies, including PG9, PG16, and PGT151. Dose-dependent relative infectivity was normalized to the mean value determined in the absence of the corresponding ligand. All viral particles generated in this study were prepared using 100% of indicated HIV-1 constructs during transfection.

We next tested the sensitivities of full-length dual-ncAA Env virions to trimer-specific neutralizing antibodies, including Env apex-targeting PG9 and PG16 and gp120/gp41 interface-directed PGT151.33,60,61 Neutralization curves of dual-amber Env viral particles resembled that of amber-free wildtype ones (Figure 3E), indicating the preservation of neutralization sensitivities of dual-ncAA incorporated Env to trimer-specific antibodies. Thus, we demonstrated that our constructed dual-amber Env variants were successfully expressed with efficient ncAA insertions, processed, and incorporated into intact amber-free virions that retained neutralization sensitivities to trimeric Env-directed antibodies. The consistent immunoblots and neutralization results indicate efficient trimerization and maturation of full-length dual-ncAA incorporated Env proteins, which present properly on intact HIV-1 virions. These functional validations pave the way for minimally invasive smFRET of Env.

smFRET imaging of dually click-labeled Env on the intact amber-free HIV-1 virion

We next performed smFRET imaging of dually click-labeled Env on amber-free intact virions (Figures 1A and 4). Three different types of donor/acceptor-dually labeled Env were investigated by smFRET imaging, including 1) dually click-labeled Env (N136TAG S401TAG and N136TAG S413TAG), 2) hybrid click/peptide Env (N136TAG V4A1 and V1Q3 S401TAG), and 3) dual-peptide Env (V1Q3 V4A1). Dually click-labeled Env was generated by randomly coupling tetrazine-conjugated Cy3/Cy5 derivatives LD555-TTZ and LD655-TTZ to TCO* ncAAs that are incorporated into Env V1V4 loops (exampled in Figure 4A or flipped sites of labeling). Two hybrid click/peptide Env were used as comparisons, in which one label was attached by click chemistry and the other by enzymatic modification of a peptide tag. Two labels of dual-peptide Env, LD555-CD and LD655-CoA were attached by enzymatic modification of two peptide tags as previously described.32,33,43,62

Figure 4. Representative fluorescence and FRET trajectories of unliganded dually click-labeled Env on amber-free virions at the single-molecule level.

Figure 4.

(A) A donor/acceptor-paired labeled protomer within an Env trimer at indicated sites on an intact virion was immobilized on a quartz slide and imaged by prism-TIRF microscope. Donor dye was excited with an incident laser and transferred its energy to nearby acceptor dye depending on its proximity. The structure of the Env trimer (PDB 4ZMJ)15 was fitted into in-situ electron density map of the ligand-free trimer (EMD-21412)34. Dually labeled gp120 monomer, brown; unlabeled gp120 monomer, cyan; gp41, blue; donor fluorophore, green; acceptor fluorophore, red.

(B, C) Two exampled fluorescence (Top panel) and derived FRET trajectories (Bottom panel) of individual dually click-labeled amber-free virions. LD555-TTZ and LD655-TTZ were randomly attached to TCO* at sites N136TAG and S413TAG (Top panel). Their corresponding FRET trajectory (blue) with overlaid idealization was generated using Hidden Markov Modeling (HMM) (purple) (Bottom panel). The monitored individual gp120 monomers undergo transitions between three FRET-identified conformational states (State 1, low FRET; State 2, high-FRET; State 3, intermediate-FRET). Photobleaching points (B, the acceptor bleaches first; C, the donor bleaches first) indicate time-course fluorescence/FRET monitoring at the single-molecule level.

As smFRET monitors a single FRET-labelled gp120 protomer within an Env trimer in otherwise wild-type trimers on a single virion, we could achieve 1 labeled-protomer:1 trimer:1 virion by adjusting the co-transfecting ratio of tagged vs. wild-type Env plasmids in HEK293T cells.32,33,63 Based on the double-amber suppression efficiency, we used an approximate 4:1 ratio of amber-free wildtype and dual-amber virus Env constructs to achieve ideal conditions for smFRET: single FRET-labelled gp120 protomer embedded in an otherwise wildtype environment. Fluorescently labeled virions were immobilized on a streptavidin-coated quartz plate-coverslip sample chamber through the incorporation of biotinylated lipids into the virus membrane. The LD555-TTZ label on immobilized virions was excited, and fluorescence intensities of LD555-TTZ and LD655-TTZ were recorded simultaneously by our customized prism-TIRF microscope. As in smFRET studies of enveloped viruses38, single-color virions were automatically filtered based on the cross-correlation between two fluorescence trajectories, whereas multi-labeled protomers in a trimer were manually filtered based on stepwise photobleaching events. We extracted hundreds of fluorescence trajectories with anti-correlated features between donor and acceptor fluorescence intensities (Figure 4B and C; top). Anti-correlation is the revealing signature of energy transfer between a donor and an acceptor, reflecting conformational changes of Env on individual virions. The donor or acceptor photobleaching point, either acceptor bleaching first (Figure 4B) or donor bleaching first (Figure 4C), indicates the detection of a single FRET-labeled Env on an immobilized intact virion. We further derived the FRET efficiency (FRET traces/trajectories, exampled in Figures 4B and 4C; bottom), documenting real-time conformational changes over time of individual Env on virions.

Consolidation of Env conformational profiles by minimally invasive amber-click smFRET systems in the context of intact amber-free virions

Qualitative and quantitative smFRET analysis of dually click-labeled Env in the amber-free virus context revealed consistent three-state conformational profiles of Env on the viruses with results obtained from previous dual-peptide Env and two hybrid click/peptide Env (Figures 5, S5, S6, and Table S3). A pair of donor/acceptor dyes attached to different Env constructs include LD555-TTZ/LD655-TTZ (R0 ~61.0 Å) for labeling both N136TAG S401TAG and N136TAG S413TAG, LD555-TTZ/LD655-CoA (R0 ~60.0 Å) for N136TAG V4A1, LD555-CD/LD655-TTZ (R0 ~61.0 Å) for V1Q3 S401TAG, and LD555-CD/LD655-CoA (R0 ~60.0 Å) for V1Q3 V4A1. In bulk photophysical measurements, no noticeable differences were found in the spectra of different LD555 or LD655 derivatives (Figure S5A) or shifts in excitation or emission spectra for all dyes used after attaching to Env constructs. For example, the spectra of single-color N136TAG S401TAG that was singly labeled by LD555-TTZ or LD655-TTZ were almost identical to the spectra of free dyes (Figure S5B). As expected, the fluorescence spectra of dual-color N136TAG S401TAG labeled by LD555-TTZ and LD655 showed emission peaks of both dyes (Figure S5C).

Figure 5. Consolidation of conformational landscapes of Env by smFRET imaging of click-labeled Env on amber-free virions.

Figure 5.

(A–C) FRET histograms of dually click-labeled Env at sites N136TAG S401TAG in the context of intact virions under different conditions: ligand-free (A), PGT151-bound (B), and sCD4D1D2–Igαtp-bound (C). The number (Nm) of individual trajectories was compiled into a conformation-population FRET histogram and fitted into a 3-state Gaussian distribution (solid gray line) centered at ~0.1-FRET (dashed cyan), ~0.3-FRET (dashed red), ~0.6-FRET (dashed green). The fitted distributions (solid gray line) were overlaid with the corresponding referenced distributions of dual-peptide Env V1Q3 V4A1, where ref stands for the referenced distribution.

(D–F) Experiments as in A–C, revealing conformational landscapes for the ligand-free (D) and upon binding of PGT151 (E) or sCD4D1D2–Igαtp (F) to dually click-labeled Env N136TAG S401TAG.

(G) Bar graph of relative conformation-occupancy of virus Env using different FRET probes. FRET histograms represent mean ± SEM, determined from three randomly assigned populations of FRET traces under indicated experimental conditions. Relative occupancies of each conformational state and fitting parameters were summarized in Table S3.

In single-molecule experiments, previous smFRET using peptide tags showed that native Env on the virus is intrinsically dynamic, transiting from the pre-triggered conformation (State 1) through a partially open intermediate (State 2) to the fully activated open conformation (State 3) upon binding to CD4 molecules.32,33,38,43 FRET trajectories report time-correlated relative donor-acceptor distance changes associated with conformational changes between V1 and V4 regions within the Env monomer. Compiling hundreds of FRET traces obtained under different conditions led to respective merged histograms fitted with Gaussian functions, indicating conformational distributions with state occupancies (Figures 4 and 5). FRET histograms of Env revealed by dually click-labeled N136TAG S401TAG (Figure 5AC), N136TAG S413TAG (Figure 5DF), as well as hybrid click/peptide N136TAG V4A1 or V1Q3 S401TAG (Figure S6), were in good agreement with previous results from dual-peptide Env V1Q3 V4A1 (overlaid lines). Similarly, the three-state conformational ensemble contained State 1 (low-FRET with mean 0.11 ± 0.02), State 2 (high-FRET with mean 0.63 ± 0.05), and State 3 (intermediate-FRET with mean 0.28 ± 0.04). Transitions between low- and high-FRET states and high- and intermediate-FRET states occurred frequently, but the low- and intermediate-FRET states were rarely observed, as shown in the representative traces (Figures 4B, 4C, and S6A). The unliganded N136TAG S401TAG, N136TAG S413TAG, N136TAG V4A1, and V1Q3 S401TAG tagged Env in the amber-free virus context all predominantly resided in a low-FRET State 1 with the mean occupations of 41%, 50%, 42%, and 52%, respectively, similar to the previous studies (Figures 5, S6, and Table S3).33,43 Potent sCD4D1D2–Igαtp shifted the conformational landscape of Env towards intermediate-FRET State 3-dominant distributions with the occupations of respective 38%, 47%, 45%, and 50% from the above-mentioned four smFRET systems (Figures 5, S6, and Table S3). Adding the PGT151 antibody that asymmetrically targets the interface between gp120 and gp41 resulted in the preferential conformation to be high-FRET State 2 with the mean occupations of 54%, 58%, 57%, and 50% (Figures 5, S6, and Table S3). Donor/acceptor dyes coupled to distinct pairs of sites on Env may contribute to the observed variations in the relative occupancy of three conformations in different FRET systems.

The minimal tags used in dually click-labeled Env did not appear to narrow the FRET histograms compared to the relatively longer peptide tags used in dual-peptide Env (Figure 5). The similarity of FRET histograms using different tags could be explained by the high degrees of flexibility and fast motions of Env variable loops V1V4 in which tags are inserted. The sampling space of tags is likely within that of V1V4 loops; thus, the impact of using different tags is likely to be buried by the intrinsic flexibility of V1V4 loops. The motions of V1V4 loops (including tags) are at a faster time scale (nanoseconds to microseconds) and are beyond our temporal resolution of 40 milliseconds, which are also averaged out during global milliseconds-to-seconds conformational motions of Env. In summary, the FRET histogram similarities could be due in part to the fast-sampling flexibility of V1V4 loops, freely rotated dyes, and similar R0 values (~60.0 vs. 61.0 Å) of different donor/acceptor pairs. All factors mentioned above likely contributed to the consistency in conformational propensities of Env probed using all established FRET systems (Figure 5G). These results reinforce our previous finding of Env sampling three primary large-scale milliseconds-to-seconds global conformations. Altogether, minimally invasive amber-click smFRET systems of Env in the context of intact amber-free virions consolidated conformational landscapes of Env and ruled out the possibility that Env conformational states are affected by peptide tagging.

Application of amber-free HIV-1 Env labeling for single virus tracking in cells

We further explored the potential of the amber-free HIV-1 system for adopting click labeling in live cell imaging settings. For the purposes of prolonged single virus tracking, virions containing 100% amber N136TAG-Cy5 mutant Env were produced and labeled with LD655-tetrazine. Live cell confocal microscopy of single-labeled amber-free HIV-1 N136TAG-Cy5 revealed virus internalization into target TZM-bl cells labeled with SPY555-Actin. Viruses were pre-bound to cells in the cold, unbound particles washed away, and their entry was initiated by shifting to 37 °C. The trajectory and the Cy5 and SPY555 intensity profiles of a representative single virion are shown in Figure 6A, B, and Movie 1. The virion lingered at the cell periphery for about 20 min and then was rapidly internalized, as evidenced by initial colocalization with the peripheral actin cortex followed by net displacement to the center of the cell (Fig. 6B, C). Consistent with endosomal trafficking, the virus’ speed reached 0.17 μm/s. (Figure 6D). The super-linear mean square displacement (MSD) profile for the trajectory further supports a net directional movement of the virus away from the cell periphery. (Figure 6E). These findings support the notion that HIV-1 is internalized and transported in endosomes.64,65 Our results thus demonstrate the versatility of the amber-free virus labeling approach that can be applied to live-cell tracking of HIV-1 entry into cells.

Figure 6. Trafficking of amber-free HIV-1Q23 BG505 virions carrying 100% click-labeled Env N136TAG-Cy5 in TZM-bl HeLa cells.

Figure 6.

(A) Live cell imaging of SPY555-Actin (magenta) labeled TZM-bl cells with an internalization event of a single virion with click-labeled Env N136TAG-Cy5 (green). The HIV-1 particle of interest initially associated with the cell periphery is marked with a dashed yellow circle.

(B) Representative single particle trajectory of the click-labeled HIV-1 internalization event from panel A. The single HIV-1 trajectory is overlayed on the source image to demonstrate the depth of virus internalization into the cell.

(C) Representative single particle intensity trace for the particle of interest from panel A. The internalization event is indicated with the black arrowhead on the panel.

(D) Instantaneous particle speed demonstrating an increase in virus trafficking speed during internalization into the cell, consistent with virus transport in endosomes. The internalization event is signified with an arrow.

(E) Mean squared displacement (MSD) plot of the particle from panel A demonstrating that the particle of interest undergoes directional motion for a fraction of its trajectory coinciding with the internalization even.

Discussion

Here, we developed and optimized an intact amber-free HIV-1 provirus that significantly improves the incorporation of ncAAs into Env as a minimally invasive method to label viral proteins at single-residue precision by genetic code expansion. The developed provirus further enabled bio-orthogonal click labeling of Env and the subsequent establishment of the minimal invasive amber-click smFRET systems for Env. The amber-free HIV-1 system is versatile and valuable for in-virus protein biorthogonal click labeling, particularly as lentiviral cores are often used to study glycoproteins from other viruses by pseudotying.62,63,6668

Demands for minimally invasive labeling strategy in smFRET imaging of Env

To achieve smFRET of Env, we had previously deployed an alternative approach of introducing short peptide tags 40,41 into variable loops of gp120.32,33,43 Nevertheless, the introduction of a few amino acids still holds the possibility of affecting Env conformations in unpredictable ways.42 There was also a concern, or controversy, that the use of peptide tags for FRET labeling could be the cause for the observation of an additional conformational state at the smFRET level that has not been structurally characterized. The amber-click strategy for site-specific incorporation of ncAAs and labeling facilitates the identification of additional, more permissive labeling sites that lower the risk of functional impact.51,69,70 This approach is also promising for accessing the sterically restricted gp41 subunit that does not tolerate peptide insertions in our hands.

Challenges in amber-click labeling the sparse Env on the virus

The challenges in achieving dually amber-click labeling of Env on the virus were initially underestimated and impeded our long-sought-after minimally invasive smFRET imaging of Env. No success of singly click-labeled Env on intact HIV-1 virions was reported. On the positive side, progress has been made with click-labeling of pseudotyped influenza A hemagglutinin63 and Ebolavirus glycoprotein.66 However, the number of Env copies on HIV-1 is much lower than influenza A hemagglutinin or Ebolavirus glycoproteins. Labeling of HIV-1 capsids has recently been reported52 and in a new preprint71 posted at the time of this submission. Again, a few copies of Env on each virion are in sharp contrast to thousands of copies of capsid protein (p24).13,7274 Apparently, major challenges, such as low copies of Env on virions, incomplete suppression in mammalian cells, and high Env structural complexity, have impeded the application of ncAAs to Env.

Pinpointing a limiting factor of amber suppression in the HIV-1 genome

We realized that many HIV-1 proviruses carry an abundant number of amber codons in their genomes. These preexisting ambers could result in aberrant extensions of viral proteins, resulting in the synthesis of dominant-negative proteins. We thus constructed an intact amber-free HIV-1 system by genetically modifying naturally occurring amber codons (TAG) to ochre codons (TAA) in critical protein-coding genes, warranting their correct translating terminations. Altering the amber codon of Pol largely rescued virus infectivity (Figure 2B), implying that the aberrant extension of Pol is likely the major reason for the poor production/infectivity of naturally amber-abundant HIV-1Q23 (Table S2). Changing the amber of Vpu to ochre causes a point mutation in Env, but it is within the signal peptide and does not appear to interfere with Env secretion (Figure 2C). Modifying the amber of Rev to ochre results in a mutation in the C-tail of Env gp41, but it does not seem to affect Env conformational dynamics (Figures 5 and S6).

While we could not biochemically document problematic suppression of amber stop codons within the HIV-1 genome under suppression conditions, our reasoning that it is likely the main hurdle of applying genetic code expansion to HIV-1 is based on the following observations: 1) all amber stop codons are expected to be indiscriminately read through by suppressor tRNAs inside the cytosol of producer cells including ambers in the HIV-1 genome; 2) indeed, we observed a strong rescue of infectivity by a single change from amber to ochre in Pol and further rescue by modifying all ambers to ochres in intact HIV-1 genome (Figure 2B); 3) our constructed amber-free system enabled the first-ever visualization of singly and dually amber-suppressed virus Env that can be detected by western blotting (Figure 3C), in contrast to unsuccessful attempts made on the amber-abundant virions by us and others.

The amber-free HIV-1 system is compatible with click-labeling of dually amber-suppressed Env, enabling minimally invasive smFRET imaging of Env on the virus

Built upon our newly developed amber-free HIV-1 provirus, we further optimized two candidate clones encoding dually TAG-tagged Env. In contrast to the pioneering study44, our infectivity and immunoblot results provided evidence of enhanced suppression efficiency and resulting effective ncAA incorporation in Env (Figure 3). smFRET results of dually click-labeled Env in parallel with hybrid click/peptide Env were in global agreement with our previous observations (Figures 4 and 5).32,33,43. These results further rule out the scenarios that sCD4D1D2–Igαtp and PGT151 interact with peptide tags/dyes, and that peptide tags affect the presence of smFRET-identified conformational states of Env. Our results directly prove that the Env states observed at the level of smFRET are an intrinsic dynamic property of Env and not due to a labeling artifact using peptide tags.

The amber-free context also holds great promise for smFRET imaging of the highly constrained transmembrane subunit gp41 of Env, for which the fusion-inducing conformational events remain elusive. The amber-free HIV-1 system is expected to significantly increase our success rate for site-specifically introducing a pair of clickable donor/acceptor dyes into conformational switching regions of gp41 by only altering a single amino acid per site. Therefore, the potential of amber-click Env in the amber-free context will be exploited in our future smFRET imaging of conformational dynamics and allostery during virus entry.

Application of an amber-free HIV-1 system for in-virus protein bioorthogonal labeling

The amber-free clinically relevant infectious virus system is highly versatile for screening amber-tolerable sites in any HIV-1 proteins, offering a high chance of efficient labeling of proteins in/on HIV-1 virions. It, therefore, permits visualizing/tracking under-investigated viral proteins within the context of infectious virus particles close to their native states during the trafficking of viruses in cells using live-cell microscopy, correlative imaging, and super-resolution microscopy. For example, the identified amber-tolerable N136TAG, S401TAG, and S413TAG sites on Env in the amber-free Q23 virus context can be used for virus tracking in living cells using confocal, TIRF, super-resolution microscopy. Searching for bioorthogonal clickable sites in the capsid in the context of our amber-free clinically relevant HIV-1Q23 infectious system, which is more relevant than a lab-adapted HIV-1 isolate,52,71 is promising for tracking capsid uncoating and other imaging applications.

In-virus protein labeling commonly involves the use of peptide tags, fluorescent proteins, or antibody staining. Peptide tags attached to viral proteins or fluorescent proteins packaged into virions could affect the trafficking speed or trajectories of virions. Antibodies used in antibody staining could trigger unpredictable cellular responses, which further complicates the understanding of virus-host interactions. Click-labeled virions have the potential to minimize these effects. The broad application of click labeling will open gates for visualizing complicated processes in the virus lifecycle, as demonstrated by the recent inspiring HIV-1 studies.44,52,71 Importantly, live cell tracking of single HIV-1 entry and fusion has been commonly performed using pseudoviruses over-incorporating Env (see, for example 64,75). The use of full-length HIV-1 proviruses carrying Env trimers at natural levels is likely more presentative of bona fide virions. Our study directly labels and tracks native HIV-1 particles carrying a proper number of copies of a nearly fully proteolytically processed Env precursor.

To demonstrate the potential applications of our amber-free HIV-1 system, we tracked single viruses in live cells (Figure 6 and Movie 1). These experiments demonstrate the feasibility of long-term tracking of single virions with only a few Env trimers incorporated. These studies are significant in the light of evidence supporting productive HIV-1 entry through an endocytic route in HeLa-derived and primary CD4+ T-cells.64,75 Further studies are required to deeply understand HIV-1 internalization dynamics and its relevance to productive infection.

Limitations of the study

The amber-suppressing efficiencies of Env are still not perfectly satisfactory. Two factors likely limit amber suppression in mammalian cells. Firstly, the efficiency of incorporating ncAAs is still limited by unpredictable competition with cellular amber codons translations, e.g., release factors.44,7680 Secondly, the tRNApyl/PylRSAF and its advanced NES systems from Methanosarcina species are not exploited fully in the eukaryotic cells,44 leading to the decrease of amber-suppression efficiency, with the expectation of the exponential decrease efficiency for a duall amber-suppression in the same protein.81 Future efforts to develop promiscuously amber-suppressed systems with higher suppression efficiency are thus required. This may include an improved pylRS system with enhanced translations46 and the design of an orthogonal tRNA/pylRS pair permitting simultaneous incorporation of two distinct unnatural amino acids against different stop codons.45 Due to the instrumental limitations, we have not yet explored their full potential in advanced microscopic studies of click-labeled viral proteins beyond smFRET imaging of Env. Nevertheless, the success of dually click labeling Env at the single-molecule level in this study holds great promise for bioorthogonal click labeling other viral proteins with high copies at the ensemble or single-molecule levels.

STAR ★ METHODS

RESOURCE AVAILABILITY

Lead Contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Maolin Lu (maolin.lu@uthct.edu). This study generates new recombinant DNA (plasmids).

Materials Availability

All primers used for constructing amber-free HIV-1 and amber-tagged Env plasmids are shown in Table S4. All template and constructed recombinant DNA are listed in the key resources table. The replication-incompetent amber-free HIV-1 provirus plasmid is available from Addgene (plasmid ID: 213007) or from the lead contact upon request. All other unique reagents generated in this study are available from the lead contact upon request.

KEY RESOURCES TABLE.
REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
PG9 NIH HIV Reagent Program Cat # ARP-12149
PG16 NIH HIV Reagent Program Cat # ARP-12150
PGT151 Peter D. Kwong, NIH/NIAID N/A
Anti-HIV-1-gp120 NIH HIV Reagent Program Cat # ARP-288
Anti-HIV-1-p24 NIH HIV Reagent Program Cat # ARP-3537
HRP-conjugated rabbit anti-sheep IgG secondary antibodies Invitrogen Cat # 31240
HRP-conjugated rabbit anti-goat IgG secondary antibodies Invitrogen Cat # 31402
Chemicals, peptides, and recombinant proteins
Dulbecco’s Modified Eagle Medium Gibco Cat # 2509022
Fetal bovine serum Atlanta Biologicals Cat # S11550
L-glutamine Gibco Cat # 25030-081
Penicillin-streptomycin Gibco Cat # 15140-122
0.05% Trypsin-EDTA, phenol red Gibco Cat # 25300054
Dulbecco’s Phosphate Buffered Saline Gibco Cat # 14190144
Opti-MEM reduced serum medium Gibco Cat # 31985070
Polyethyleneimine Polyscience Cat # NC1014320
Fugene 6 Promega Cat # E2311
Unnatural amino acid transcyclooct-2-ene lysine (TCO*) Sichem Cat # SC8008
4–12% Bis Tris polyacrylamide Bio-Rad Cat # NP0322BOX
MOPS SDS running Buffer Invitrogen Cat # NP0001
Trans-Blot Turbo Trans Buffer Bio-Rad Cat # 10026938
Precision plus protein dual color standards Invitrogen Cat # 1610374
Tween20 Invitrogen Cat # BP337-500
Nonfat-Dried Milk Bovine Sigma Cat # SLBZ5377
Clarity western ECL substrate Bio-Rad Cat # 170-5060
Sucrose Sigma Cat # S0389
Transglutaminase Sigma Cat # 80146-85-6
AcpS (acyl-carrier-protein synthase) Maolin Lu Lab, University of Texas at Tyler N/A
PEG-passivated, streptavidin-coated quartz slides This paper N/A
Trolox Sigma Cat # 238813
Cyclooctatetraene Sigma Cat # 138924
Nitrobenzyl alcohol Sigma Cat # N12821
Protocatechuic acid Sigma Cat # 37580
Protoatechuate 3,4-deoxygenase Sigma Cat # P8279
LD555-Tetrazine Lumidyne Technologies N/A
LD655-Tetrazine Lumidyne Technologies N/A
LD555-COA Lumidyne Technologies N/A
LD6555-CD Lumidyne Technologies N/A
BCN-OH quencher Sigma Cat # 742678
PEG2000-biotin Avanti Polar Lipids Cat # 880129P
Streptavidin Invitrogen Cat # S888
60% (w/v) Opti-prep Sigma Cat # D1556
Nitrobenzyl alcohol Sigma Cat # N12821
Protocatechuic acid Sigma Cat # 37580
Protocatechuic-3,4-dioxygenase Sigma Cat # P8279
Acetone, EM-Grade, Glass-Distilled Electron Microscopy Sciences Cat # 10015
SPY555-Actin Cytoskeleton CAT # CY-SC202
Critical commercial assays
KAPA SYBR FAST qPCR Master Mix (2X) Kit KAPA Biosystems Cat # KK4600
Pierce Gaussia Luciferase Glow Assay Kit Thermo Fisher Scientific Cat # 16158
Experimental models: cell lines
HEK293T ATCC Cat # CRL-3216
TZM-bl NIH HIV Reagent Program Cat # ARP-8129
Experimental models: organisms/strains
HIV-1 lentiviral particles carrying envelope This paper N/A
Oligonucleotides
Primers for constructing plasmids, see Table S4 This paper N/A
Recombinant DNA
Wildtype HIV-1Q23 BG505 Julie Overbaugh Lab, Fred Hutch Cancer Center N/A
Amber-free HIV-1Q23 BG505 This paper N/A
tRNAPyl/NESPylRSAF Edward Lemke Lab, Johannes Gutenberg –University Mainz N/A
Amber-free HIV-1Q23 BG505 N136TAG V4A1 This paper N/A
Amber-free HIV-1Q23 BG505 V1Q3 S401TAG This paper N/A
Amber-free HIV-1Q23 BG505 N136TAG S401TAG This paper N/A
Amber-free HIV-1Q23 BG505 N136TAG S413TAG This paper N/A
HIV-1-lnGluc Walther Mothes Lab, Yale University N/A
Deposited data
Replication-incompetent amber-free HIV-1Q23 BG505 deltaRT Addgene Cat # 213007
Replication-competent amber-free HIV-1Q23 BG505 This paper Mendeley Data (https://doi.org/10.17632/d9ww5mh46d.1)
Software and algorithms
GraphPad Prism v8.4.3 GraphPad https://www.graphpad.com/
MATLAB Mathworks https://www.mathworks.com/
PyMOL Schrödinger https://pymol.org/2/
Chimera University of California, San Francisco https://bio3d.colorado.edu/SerialEM/
SerialEM software package David N. Mastronarde, University of Colorado Boulder https://bio3d.colorado.edu/SerialEM/
IMOD software package David N. Mastronarde, University of Colorado Boulder https://bio3d.colorado.edu/imod/
Other
Trans-Blot Turbo system Bio-Rad N/A
ProFlex PCR System Applied biosystems N/A
NanoSight instrument Malvern Panalytical N/A
Bio-Rad ChemiDoc XRS+ System Bio-Rad N/A
BioTek Synergy H1 Microplate Reader BioTek N/A
Prism-based TIRF Microscope Maolin Lu Lab, University of Texas at Tyler N/A
Gradient master 108 Biocomp N/A
FluoroMax - Spectrofluorometer Horiba Scientific N/A
Gravity-driven plunger apparatus Walther Mothes Lab, Yale University N/A
QUANTIFOIL® holey carbon grids Electron Microscopy Sciences Cat # Q250-CR1
Titan Krios G3 microscope Thermo Fisher Scientific N/A
Zeiss LSM880 laser scanning confocal microscope Gregory Melikian Lab, Emory University N/A
96-well white plates for luciferase assays Costar Cat # 3917
ThermalGrid Rigid Strip PCR tubes Denville Scientific Inc Cat # C18064
Acrodisc 32 mm Syringe Filter w/0.45 μm Supor Memberane Non-Pyrogenic. PALL Life Sciences Cat # 4654
Immun-Blot PVDF membrane Bio-Rad Cat # 1620174
Mini Trans-Blot Filter Paper Bio-Rad Cat # 1703932

Data and Code Availability

  • Sequence and map of replication-incompetent amber-free HIV-1 provirus plasmid have been deposited into Addgene under plasmid number 213007 listed in the key resources table. Sequences and maps of both replication-competent and incompetent amber-free HIV-1 provirus have been deposited at Mendeley. The DOI is listed in the key resources table. Other data reported in this paper will be shared by the lead contact upon request.

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS

Cell Lines and Culture Conditions

HEK293T cells (human embryonic kidney, ATCC) were used for producing replication-complement and replication-incompetent HIV-1 viruses. TZM-bl cells (generated from HeLa cells) that stably express receptor CD4 and co-receptors CCR5 and CXCR4 were used as target cells for quantifying the infection abilities of produced HIV-1 viruses82. These cell lines were cultured in high glucose Dulbecco’s Modified Eagle Medium supplemented with 10% (v/v) fetal bovine serum, 100 μg/mL penicillin-streptomycin, and 6 mM L-glutamine, and maintained in a 37 °C incubator supplied with 5% CO2.

METHOD DETAILS

Plasmid construction

The wildtype HIV-1Q23 BG505, an infectious clade A clone, contains full-length EnvBG505 with Q23 as the backbone5557 (a gift from Julie Overbaugh, Fred Hutchinson Cancer Center, Seattle). BG505 is a clinical Clade A transmitted/founder Env strain. HIV-1Q23 BG505 ΔRT is a replication-incompetent clone that lacks the reverse transcriptase gene32,33,43. Amber-free HIV-1Q23 BG505 plasmid was generated from HIV-1Q23 BG505 plasmid in which TAG stop codons presented in the Pol, Vif, Vpu and Rev genes were altered to TAA stop codons. Tagged-EnvBG505 plasmids with amber-free Q23 as backbone include amber-free HIV-1Q23 BG505 N136TAGV4A1, V1Q3S401TAG, N136TAGS401TAG and N136TAGS413TAG. Amber-free N136TAGV4A1 plasmid was tagged with a single amber codon TAG at the position of codon 136 (Asn) in the variable V1 loop and an A1 peptide tag (A1, GDSLDMLEWSLM) in the variable V4 loop within gp120 of Env, while plasmid amber-free V1Q3 S401TAG contained the insertions of Q3 peptide tag (Q3, GQQQLG) in V1 and a single amber codon at the position of 401 (Ser) in the V4 of gp120. Similarly, N136TAGS401TAG contains two ambers inserted at positions 136 (Asn) in V1 and 401 (Ser) in V4, respectively. N136TAGS413TAG was tagged by two ambers introduced at positions 136 (Asn) in V1 and 413 (Ser) in V4, respectively. These plasmids were constructed from amber-free HIV-1Q23 BG505 and HIV-1Q23 BG505 ΔRT for generating amber-click labeled Env in the context of intact virions. During the initial screening of amber-tolerance sites on EnvBG505 V1 and V4 loops, single amber- or dual amber-tagged Env were generated from wildtype amber-abundant HIV-1Q23 BG505. We generated different versions of amber-free HIV-1Q23 BG505 clones, including ΔRT, ΔEnv (a new virus packing system with BG505 Env deletion), and a ΔRT ΔEnv. All the plasmids were sequenced before being used.

Viral infectivity measurements

Viral infectivity of HIV-1Q23 BG505 was determined by using a Gaussia luciferase reporter83,84 and quantified using a Gaussia luciferase flash assay kit, according to the manufacturer’s instructions. Briefly, amber-abundant, or amber-free HIV-1Q23 BG505 viruses, or Env-tagged ones, were produced, respectively, in HEK293T cells that were co-transfected with the corresponding HIV-1Q23 BG505 plasmid, and intron-regulated luciferase plasmid (HIV-1-InGluc) using transfection reagent polyethyleneimine (PEI Max) or Fugene 6. Under amber suppression conditions, in addition to the HIV-1Q23 BG505 and HIV-1-InGluc plasmids, the transfection solution also includes a tRNAPyl/NESPylRSAF plasmid (1:3 HIV-1Q23 BG505 plasmid) supplemented with 250 μM unnatural amino acid (ncAA) transcyclooct-2-ene lysine (TCO*). The plasmid tRNAPyl/NESPylRSAF is composed of a pyrrolysine tRNA and its cognate aminoacyl tRNA synthetase pair. PylRSAF is the double mutant (Y306A, Y384F) of the M. mazei pyrrolysine tRNA synthetase, accommodating bulky side-chain moieties in ncAAs. tRNAPyl/NESPylRSAF plasmid can be expressed in mammalian cells, such as HEK293T cells.45,46 Viruses in the cell supernatants were harvested at 40 hour (h) post-transfection, filtered through 0.2 μm filters, and concentrated by ultracentrifugation at 25,000 rpm for 2 h. Viruses diluted in the medium were added onto TZM-bl cells pre-seeded in a 48-well plate. At 40 to 48 h post-infection, 100 μl cell supernatant was transferred to 96-well plates for the measurement of gaussian luciferase activity using a Gaussia luciferase flash assay kit. Relative viral infectivity was determined by normalizing relative light units (RLU) to the control group in each assay.

Immunoblotting

Viral particles of amber-abundant, amber-free HIV-1Q23 BG505, and amber-free Env-tagged N136TAG S401TAG, N136TAG S413TAG, V1Q3 S401TAG or N136TAG V4A1 produced in HEK293T cells were verified by anti-gp120 western blotting under both suppression and non-suppression experimental conditions. Viral particles of amber-abundant and amber-free HIV-1Q23 BG505 were also analyzed using anti-Gag immunoblotting. Viral particles were produced similarly as that used in the infectivity assay, except lack of HIV-1-InGluc. For immunoblot assays, both the supernatants and cells were harvested after 40–48 h post-transfection of HEK293T cells. Virus supernatants were followed by ultracentrifugation of 40,000 rpm for 1.5 h, and virus pellets were resuspended in PBS buffer. Protein samples from virus supernatants and cell lysates were loaded on a 4–12% BisTris gel and transferred onto PVDF membrane by semi-dry blotting (Bio-Rad trans-blot turbo transfer system, 1.4 A constant and 20V for 7 min) with precision plus protein dual color standards PVDF membranes were blocked with 5% (w/v) skimmed milk powder in PBS for 1 h at room temperature and then incubated with primary antibodies of anti-HIV-1-gp120, and anti-HIV-1-p2485, diluted in PBST (PBS with 0.05% Tween 20) buffer overnight at 4 °C. Membranes were followed by incubation with HRP-conjugated rabbit anti-sheep/-goat IgG secondary antibodies for 1 h at room temperature. Detection and quantitation of bound antibodies were conducted using clarity western ECL substrate and Bio-Rad ChemiDoc XRS+ System with Image Lab Software.

Neutralization assays

HEK293T cells were seeded and co-transfected with a mix of plasmids encoding either amber-abundant wildtype or amber-free HIV-1Q23 BG505, amber-free N136TAG S401TAG, or amber-free N136TAG S413TAG, together with HIV-1-InGluc and tRNAPyl/NESPylRSAF. 250 μM TCO* was added to the culture medium. At 40 h post-transfection, produced viruses in cell supernatants were collected, filtered, and concentrated by ultracentrifugation. Prior to adding onto the target TZM-bl cells, respective HIV-1 viruses as indicated in figures were incubated with sCD4D1D2–lgαtp, PG960, PG1660 or PGT151 at the 10-fold serial dilutions (102 to 10−2 μg/mL) for 60 min at room temperature, as previously described33. Viruses in the presence of different concentrations of ligands were then added on pre-seeded TZM-bl cells in the 48-well plate and were further incubated for another 40–48 h. Approximately two days later, cell supernatants were transferred to 96-well plates for the measurement of Gaussia luciferase activity, as described earlier in the viral infectivity measurements. Relative infectivity (mean ± SD) was normalized to the mean value determined from the control counterpart in each neutralization group.

2D cryo-electron microscopy

After 40 h post-transfection of 293T cells, 120 mL viral supernatants of amber-free HIV-1Q23 BG505, amber-free N136TAG S401TAG, and amber-free N136TAG S413TAG were harvested, filtered and centrifugated on a 15% sucrose cushion by ultracentrifugation at 25,000 rpm for 2 h. Virus pellets were resuspended with 30 μL DPBS. Viral particles were mixed with a 6 nm gold tracer at a 1:3 ratio. 5 μL of the mixture was placed onto freshly glow discharged holey carbon grids (Quantifoil R 2/1 on 200 copper mesh) and incubated for 1 min. Grids were blotted with filter paper and plunge-frozen into liquid ethane using a homemade gravity-driven plunger apparatus. Samples were screened with a Titan Krios G3 microscope (Titan Krios, Thermo Fisher Scientific) operating at 300 kV on a Gatan K3 camera in counting mode, at a magnification of 64,000×, with a calibrated pixel size of 1.346 Å.

Diameter size measurement of produced virus particles

HEK293T cells were seeded in 10 cm dishes with 12 mL complete growth medium. On the next day, cells with 80% confluency were transfected with 12 μg DNA plasmid and 1 mg/mL PEI Max at a molar ratio of 1:3 (w: w). Plasmid DNA and PEI were separately diluted in Opti-MEM reduced serum medium (Thermal Fisher) for 5 min at room temperature (RT). PEI was then mixed well with plasmid DNA and incubated for 15 min at room temperature. The final mixture was added dropwise to the cells. For the amber suppression, HEK293T cells transfected with HIV-1 plasmid DNA, tRNAPyl/NESPylRSAF and PEI at a ratio of 3: 1: 12 (w: w: w) were maintained in complete growth medium supplemented with a final concentration of 250 μM TCO*. The exchange of fresh complete growth medium (supplemented with 250 μM TCO* for amber suppression) was carried out at 6 h post-transfection. At 16 h post-transfection, an additional 250 μM TCO* was added to the amber suppression transfection systems. After 40 h post-transfection, the supernatants containing virions were harvested and centrifugated at a low speed of 200 g for 10 min. The supernatants were purified on a 15% sucrose cushion (w/v phosphate-buffered saline, PBS) by ultracentrifugation at 40,000 rpm for 1.5 h, using Bechman SW-41 Ti Swing Rotor. Virus pellets were resuspended with 200 μL borate buffered saline (BBS) buffer. The diameter size distribution of virus particles diluted in borate buffered saline (BBS) buffer was measured by Nanoparticle Tracking Analysis (NTA) using NanoSight instrument. Three measurements were carried out with 30 seconds duration for each measurement and the data were analyzed by NTA 3.3 Dev Build 3.3.301. Detected tracks were translated into a size distribution using maximum likelihood estimation with an assumed distribution (FTLA method).

Preparation of intact HIV-1 virions for smFRET imaging

HIV-1Q23 BG505 virions incorporated with tagged EnvBG505 (amber-tagged, amber/peptide-tagged, or peptide-tagged) in V1 and V4 loops were prepared in a similar way as previously described.33 HIV-1 virions used for smFRET imaging were prepared as replication-incompetent viral particles that lack reverse transcriptase (ΔRT), meaning that all the full-length HIV-1Q23 BG505 plasmids used for transfection are ΔRT versions. In this study, we generated three different types of viruses with peptide-, hybrid amber/peptide-, or amber-tagged Env: 1) dual-peptide V1Q3 V4A1; 2) hybrid click/peptide V1Q3 S401TAG and N136TAG V4A1 in the amber-free virus context; 3) dual-amber N136TAG S401TAG and N136TAG S413TAG in the amber-free virus context. V1Q3 V4A1 Env carries a Q3 tag in V1 and an A1 tag in V4. As previously described,33,43 V1Q3 V4A1 virions were produced by co-transfecting HEK293T cells with a 40:1 plasmid: plasmid ratio of wild-type tag-free Env virion: V1Q3 V4A1, to ensure that on average only one dually peptide-tagged protomer within a trimer was available for labeling on a single virion. The plasmid: plasmid ratio of tag-free: tagged Env virions was adjusted according to the reduction in amber suppression efficiency of amber-tagged or amber/peptide-tagged Env. N136TAG S401TAG, N136TAG S413TAG, V1Q3 S401TAG, N136TAG V4A1 on the amber-free virions were made similarly, by co-transfecting HEK293T cells with a 4:1 ratio of amber-free tag-free Env to Env-tagged plasmids, along with an amber suppressor plasmid supplemented with unnatural amino acids. For instance, amber-free N136TAG S413TAG viruses, carrying two clickable sites at N136TAG in V1 and S413TAG in V4, were produced by co-transfecting HEK293T cells with a tag-free ΔRT plasmid, an Env-tagged variant N136TAG S413TAG ΔRT plasmid, and a bi-cistronic plasmid tRNAPyl/NESPylRSAF encoding the amber suppressor tRNA and PylRSAF. The amount of tRNAPyl/NESPylRSAF used is 1/3 of the total amount of plasmids that encode Env (including untagged and tagged). The unnatural amino acid TCO* (250 μM) was supplemented to transfected HEK293T cells during the transfection and re-supplemented 12-h post-transfection.

Fluorescent labeling Env on intact HIV-1 virions

After 40 h post-transfection, viruses in the supernatants were filtered, harvested, and concentrated on the 15% sucrose cushion by ultracentrifugation at 25,000 rpm (SW28, Beckman Coulter) for 2 h. Virus pellets were resuspended in the labeling buffer containing 50 mM HEPES, 10 mM MgCl2 and 10 mM CaCl2.32,33,43 Fluorescently labeling, including enzymatic labeling and click labeling, of virus-associated Env has been previously described32,33,43. For enzymatic labeling, the resuspended V1Q3 V4A1 viruses in labeling buffer were supplied with cadaverine-conjugated Cy3 (LD555-CD, 0.5 μM) and coenzyme A (CoA)-conjugated Cy5 (LD655-CoA, 0.65 μM)41, and AcpS (5 μM)40, and then incubated overnight at room temperature. For click labeling, resuspended amber-free N136TAG S401TAG or N136TAG S413TAG viruses that carry two click-chemistry-reactive TCO* were fluorescently labeled in a reaction mixture of 0.1 μM tetrazine-conjugated Cy3 and Cy5 derivatives (LD555-TTZ and LD655-TTZ). For enzymatic/click labeling, resuspended amber-free N136TAG V4A1 or V1Q3 S401TAG viruses that carry one peptide and one clickable TCO* were fluorescently labeled by the corresponding enzymatically conjugated dye along with the specific enzyme and the FRET-paired click dye. For the click-labeled or enzymatic/click-labeled viruses, a quenching step of 10 min incubation with 1μM BCN-OH quencher followed the labeling step. PEG2000-biotin was subsequently added to the labeling reaction at a final concentration of 0.1 mg/ml and followed by incubation at room temperature for 30 min with rotation. Excessive label and lipid were removed by ultracentrifugation over a 6–18% Optiprep gradient at 40,000 rpm for 1 h. The labeling virions were stored at −80 °C until further study.

smFRET imaging data acquisition

All smFRET experiments of Env on intact HIV-1 virions were performed on a customized prism-based total internal reflection fluorescence (prism-TIRF) microscope, as previously described.33,62,68,86 Fluorescently labeled HIV-1 viruses were incubated in the absence or presence of 0.1 mg/ml sCD4D1D2–lgαtp or 0.1 mg/ml PGT151 in the imaging buffer containing 50 mM Tris (pH 7.4), 50 mM NaCl, a cocktail of triplet-state quenchers, 2 mM protocatechuic acid (PCA), and 8 nM protocatechuic-3,4-dioxygenase (PCD) at room temperature for 30 min before smFRET imaging, as previously described.32,33,43,87 The ligand and antibody concentrations were approximately 5-fold above the 95% inhibitory concentration. HIV-1 viruses carrying fluorescently labeled Env were immobilized on a PEG-passivated biotinylated quartz-coverslip imaging chamber coated with streptavidin. Based on the refractive indexes at the interface between quartz and sample solution, the evanescent field was generated by the total internal reflection of 532-nm single-wavelength laser excitation (Ventus, Laser Quantum) directed to a prism. The donor fluorophore labeled on Env on intact HIV-1 virions is further directly excited by the generated TIRF field. Fluorescence from both donor and acceptor fluorophores labeled on Env was collected through a water-immersion Nikon objective (1.27-NA 60x) and then optically separated by a MultiCam LS image splitter (Cairn Research) with a dichroic filter (Chroma). The separated fluorescence signals from the donor and acceptor passed through two emission filters (ET590/50, ET690/50, Chroma) mounted on the image splitter, respectively. Fluorescence signals were recorded simultaneously on two synchronized sCMOS cameras (Hamamatsu ORCA-Flash4.0 V3) at 25 Hz for 80 seconds. Where indicated, viruses were pre-incubated with the corresponding antibody/ligand for 30 mins at room temperature before imaging and that antibody/ligand was continuously present during smFRET imaging.

smFRET data analysis

smFRET data were viewed, processed, and analyzed by a customized SPARTAN software package that was generously shared by Scott Blanchard laboratory (https://www.scottcblanchardlab.com/software)86 and customized MATLAB-based scripts. Image stacks of smFRET data were extracted as individual fluorescence time series trajectories (fluorescence traces) of donor and acceptor labeled on HIV-1 virions. At the single-molecule level, the background signal was evaluated and then subtracted from recorded signals according to the level of signals at the single-step fluorophore bleaching points. Fluorescence traces of the donor and acceptor over 80 seconds were then corrected for the donor to acceptor crosstalk. The time-correlated donor-to-acceptor energy transfer efficiency (FRET values or FRET in graphs) was determined based on FRET= IA/(γID+IA). ID and IA are the fluorescence of the donor and acceptor, respectively. The correlation coefficient γ compensates for detection efficiencies variations between donor and acceptor channels, as the response of optics and cameras to different wavelengths varies slightly. FRET traces, real-time FRET values over time, were further derived from corresponding fluorescence traces of paired donor and acceptor. FRET traces indicate the relative distance changes between donor and acceptor over time, which are ultimately translated to real-time global conformational dynamics of the donor/acceptor labeled Env in the context of an intact virion. We use stringent criteria to filter out noisy signals. Fluorescence or its corresponding FRET Traces were automatically excluded from further analysis if it lacked signals from either donor or acceptor or both, and if it contained multiple labeled donors/ acceptors. After the initial automatic filtering, the remaining traces with sufficient signal-to-noise ratio need to display anti-correlation between donor and acceptor fluorescence before being manually included for further data analysis. The time-correlated anti-correlated feature is a signature of donor-to-acceptor energy transfer in response to real-time conformational changes of the host molecule Env, thus is an indicator of fully active/dynamic Env on the virion. The use of automatic and manual filters ensures that only one dually FRET-labeled protomer within one Env trimer on one virion is included for further analysis. FRET traces that pass our filters were included and compiled into FRET histograms/distributions, reflecting ensembles of multiple conformational states – conformational ensembles. FRET histograms - conformational ensembles represent mean ± SEM, determined from three randomly assigned populations of FRET traces under indicated experimental conditions. FRET histograms were further fitted into the sum of three Gaussian distributions using the least-squares fitting algorithm in MATLAB, based on visual inspection of traces that show clear state-to-state transitions and the idealization of individual traces using 3-state hidden Markov modeling.32,33,43,88 Each Gaussian distribution represents one distinct conformational state of Env on the virus. The area under each Gaussian estimates the relative probability of Env occupying that conformational state, displayed as relative state occupancy.

Calculation of R0 values

The Förster distance, R0, is calculated by the following equation:89

R0=(9ln(10)128π5NA*k2ΦDn4*1017*Jλ)16(in)

where n is the refractive index of the medium, NA is Avogadro’s constant, ΦD is the quantum yield of the donor in the absence of the acceptor, k2 is the orientation factor, Jλ is the spectral overlap integral. For biomolecules in water, the medium used in these experiments, n is assumed to be 1.3390, within the dynamic isotropic averaging assumption k2 is 2/3 (freely rotated dyes with flexible linkers)91. Jλ-J(λ) is the spectral overlap integral between the steady-state donor emission and the steady-state acceptor absorption. J(λ) can itself be defined using the equation: 89

J(λ)=0fD(λ)εA(λ)λ4dλ

where fD(λ) is the normalized emission spectrum of the donor and εA(λ) is the extinction of the acceptor at wavelength λ. To integrate J(λ), the trapezoid rule was used. The steady-state emission spectra of the donor and the excitation spectra of the acceptor were measured on a Horiba FluoroMax spectrofluorometer (model FluoroMax Plus-C-SP). No spectra shifts were observed for free dyes vs. dyes labeled on viruses. R0 values were similar for different donor/acceptor pairs used in our study, in which R0 values were calculated to be 60.0 Å, 61.0 Å, 61.0 Å, and 60.0 Å for donor/acceptor-paired LD555-CD/LD655-CoA, LD555-tetrazine/LD655-tetrazine, LD555-CD/LD655-tetrazine, and LD555-tetrazine/LD655-CoA, respectively.

Live cell imaging of click-labeled virions

Amber-free HIV-1Q23 BG505 N136TAG -TCO* viral particles made using 100% mutant Env construct were click-labeled by incubating for 6 hours at room temperature with 0.1 μM LD655-TTZ (Cy5-TTZ). Unreacted dye was removed by washing with PBS buffer, followed by centrifugation (25,000 rpm, 2 hr). Trafficking of amber-free HIV-1Q23 BG505 N136TAG virions labeled with Cy5-TTZ in HeLa-derived TZM-bl cells 9294 was visualized with live cell confocal microscopy. Briefly, 50,000 TZM-bl cells were plated onto collagen-coated imaging dishes with #1.5 cover glass inserts (Matek, CAT# P35G-1.5–10-C). Labeled virions were prebound to TZM-bl cells via centrifugation for 20 min at 4°C at 1,550×g in DPBS with Ca2+ and Mg2+. After virus-cell binding, cells were washed with cold DPBS, and transferred to pre-warmed Fluorobrite DMEM medium supplemented with 10% fetal bovine serum and 20 mM HEPES, pH 7.2. The imaging medium contained the SPY555-Actin (Cytoskeleton, CAT# CY-SC202) live cell actin probe, and imaging was performed in the continued presence of the probe to visualize the actin cortex, as per the manufacturer’s instructions. 3D time-lapse live cell imaging was carried out on a Zeiss LSM880 laser scanning confocal microscope equipped with a humidity and temperature-controlled environmental chamber and using a C-Apo 63x/1.4 NA oil immersion objective. All live cell imaging experiments were performed using the highly attenuated 561 nm (for SPY555) & 633 nm (for Cy5-TTZ) laser lines, and the emitted light was detected by gallium-arsenide spectral detectors with 0.22 × 0.22 μm pixel size. Live cell micrographs were collected using 10–12 Z-sections (0.7 μm spacing), with each frame being imaged every 15–17 seconds.

QUANTIFICATION AND STATISTICAL ANALYSIS

Quantification and statistical analysis are detailed in the figure legends and the STAR Methods. As indicated, analysis was performed using GraphPad Prism 9, MATLAB, or EXCEL software. Data (mean ± SD or SEM) were derived from at least three independent observations.

Supplementary Material

1

Movie 1. Internalization of an amber-free click-labeled HIV-1Q23 BG505 Env N136TAG-Cy5 virion into a live cell, related to Figure 6.

Representative movie of a single Env N136TAG-Cy5 (green) virion entering into a TZM-bl cell labeled with SPY555-Actin (magenta). The virion initially binds to the cell periphery and later undergoes internalization into the peripheral actin cortex, where high degrees of mobility and displacement are observed within the cell.

Download video file (34MB, avi)
2
3

Highlights.

  • We constructed an intact amber-free HIV-1 provirus for in-virus click labeling

  • System enables dual-ncAA insertion for biorthogonal click labeling on intact virions

  • smFRET of Env reinforced its structural dynamics observed via conventional labeling

  • Live cell imaging tracked amber-free click-labeled Env virions in cells

SIGNIFICANCE.

The minimally invasive bioorthogonal click labeling approach based on genetic code expansion combined with click chemistry has been rarely applied to fluorescently labeling HIV-1 proteins partially due to intrinsic technical limitations. Conformational dynamics of sparsely distributed envelope (Env) glycoprotein on HIV-1 mediate cell entry and facilitate antibody evasion. Dually fluorescent labeling Env is a prerequisite for enabling a unique “spectroscopic ruler” or “molecular ruler” approach to access spatiotemporal information on conformations sampled by Env on the virus, so least-invasive click labeling has been highly desirable. Here, we overcame a technical bottleneck of genetic code expansion application for HIV-1 protein labeling by establishing new full-length clinically relevant HIV-1 strains, which enabled, for the first time, singly and dually click labeling of Env in the context of virions. More specifically, we successfully incorporated two adjacent clickable unnatural amino acids into Env on intact HIV-1 virions. Using click-labeled amber-free virions, real-time single-molecule/virus spectroscopic imaging of Env on virions consolidated the presence of multiple conformations as an intrinsic property of Env dynamics that is not due to labeling artifacts and tracked the internalization route of HIV-1 into host cells. The amber-free system could open new opportunities for click labeling of functional proteins in HIV-1 and of surface glycoproteins of other viruses.

ACKNOWLEDGMENTS

The following reagents were obtained through the NIH HIV Reagent Program, Division of AIDS, NIAID, NIH: Anti-Human Immunodeficiency Virus 1 gp120 Monoclonal Antibody PG9 (ARP-12149) and PG16 (ARP-12150), contributed by International AIDS Vaccine Initiative. We thank Dr. L. Vijaya Mohan Rao and Dr. Torry Tuck for all their continued support and advice. We thank members of NIAID/VRC SBS-SBIS for their valuable feedback on our manuscript. We are grateful to Dr. Scott Blanchard for sharing software and the original design of their prism-TIRF microscope. We thank Polyhedron Optics LLC for the microscope consulting service. This work was supported by NIH/NIAID R56 AI170101, R01 AI181600, a collaborative development award from Duke Center for HIV Structural Biology (U54 AI170752), a Gilead Research Scholar award, Mathilde Krim awards from the American Foundation for AIDS Research to M.L., an NIH/NIAID R01 AI129862 to G.B.M., an award from ERC ADG Multiorganelledesign to E.A.L., the Intramural Research Program of the Vaccine Research Center, NIAID, NIH to B.Z. and P.D.K., an NIH/NIAID grant R37 AI150560 to W.M., an NIH/NIGMS R35GM147423 to M.M., and the Intramural Research Program of the Division of Intramural Research, NIAID, NIH to J.A.

Footnotes

DECLARATION OF INTERESTS

We declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Wyatt R, and Sodroski J (1998). The HIV-1 envelope glycoproteins: fusogens, antigens, and immunogens. Science 280, 1884–1888. 10.1126/science.280.5371.1884. [DOI] [PubMed] [Google Scholar]
  • 2.Dalgleish AG, Beverley PC, Clapham PR, Crawford DH, Greaves MF, and Weiss RA (1984). The CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus. Nature 312, 763–767. 10.1038/312763a0. [DOI] [PubMed] [Google Scholar]
  • 3.Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Di Marzio P, Marmon S, Sutton RE, Hill CM, et al. (1996). Identification of a major co-receptor for primary isolates of HIV-1. Nature 381, 661–666. 10.1038/381661a0. [DOI] [PubMed] [Google Scholar]
  • 4.Wu L, Gerard NP, Wyatt R, Choe H, Parolin C, Ruffing N, Borsetti A, Cardoso AA, Desjardin E, Newman W, et al. (1996). CD4-induced interaction of primary HIV-1 gp120 glycoproteins with the chemokine receptor CCR-5. Nature 384, 179–183. 10.1038/384179a0. [DOI] [PubMed] [Google Scholar]
  • 5.Choe H, Farzan M, Sun Y, Sullivan N, Rollins B, Ponath PD, Wu L, Mackay CR, LaRosa G, Newman W, et al. (1996). The beta-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 85, 1135–1148. 10.1016/s0092-8674(00)81313-6. [DOI] [PubMed] [Google Scholar]
  • 6.Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, Cayanan C, Maddon PJ, Koup RA, Moore JP, and Paxton WA (1996). HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 381, 667–673. 10.1038/381667a0. [DOI] [PubMed] [Google Scholar]
  • 7.Chen B (2019). Molecular Mechanism of HIV-1 Entry. Trends Microbiol 27, 878–891. 10.1016/j.tim.2019.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wang Q, Finzi A, and Sodroski J (2020). The Conformational States of the HIV-1 Envelope Glycoproteins. Trends Microbiol 28, 655–667. 10.1016/j.tim.2020.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Zhao C, Li H, Swartz TH, and Chen BK (2022). The HIV Env Glycoprotein Conformational States on Cells and Viruses. mBio 13, e0182521. 10.1128/mbio.01825-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Murakami T, and Ono A (2021). HIV-1 entry: Duels between Env and host antiviral transmembrane proteins on the surface of virus particles. Curr Opin Virol 50, 59–68. 10.1016/j.coviro.2021.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lee JH, Ozorowski G, and Ward AB (2016). Cryo-EM structure of a native, fully glycosylated, cleaved HIV-1 envelope trimer. Science 351, 1043–1048. 10.1126/science.aad2450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kwong PD, Wyatt R, Robinson J, Sweet RW, Sodroski J, and Hendrickson WA (1998). Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody. Nature 393, 648–659. 10.1038/31405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Liu J, Bartesaghi A, Borgnia MJ, Sapiro G, and Subramaniam S (2008). Molecular architecture of native HIV-1 gp120 trimers. Nature 455, 109–113. 10.1038/nature07159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Julien JP, Cupo A, Sok D, Stanfield RL, Lyumkis D, Deller MC, Klasse PJ, Burton DR, Sanders RW, Moore JP, et al. (2013). Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science 342, 1477–1483. 10.1126/science.1245625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kwon YD, Pancera M, Acharya P, Georgiev IS, Crooks ET, Gorman J, Joyce MG, Guttman M, Ma X, Narpala S, et al. (2015). Crystal structure, conformational fixation and entry-related interactions of mature ligand-free HIV-1 Env. Nat Struct Mol Biol 22, 522–531. 10.1038/nsmb.3051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chan DC, Fass D, Berger JM, and Kim PS (1997). Core structure of gp41 from the HIV envelope glycoprotein. Cell 89, 263–273. 10.1016/s0092-8674(00)80205-6. [DOI] [PubMed] [Google Scholar]
  • 17.Herschhorn A, Gu C, Moraca F, Ma X, Farrell M, Smith AB 3rd, Pancera M, Kwong PD, Schon A, Freire E, et al. (2017). The beta20-beta21 of gp120 is a regulatory switch for HIV-1 Env conformational transitions. Nat Commun 8, 1049. 10.1038/s41467-017-01119-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ozorowski G, Pallesen J, de Val N, Lyumkis D, Cottrell CA, Torres JL, Copps J, Stanfield RL, Cupo A, Pugach P, et al. (2017). Open and closed structures reveal allostery and pliability in the HIV-1 envelope spike. Nature 547, 360–363. 10.1038/nature23010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Trkola A, Dragic T, Arthos J, Binley JM, Olson WC, Allaway GP, Cheng-Mayer C, Robinson J, Maddon PJ, and Moore JP (1996). CD4-dependent, antibody-sensitive interactions between HIV-1 and its co-receptor CCR-5. Nature 384, 184–187. 10.1038/384184a0. [DOI] [PubMed] [Google Scholar]
  • 20.Wang H, Cohen AA, Galimidi RP, Gristick HB, Jensen GJ, and Bjorkman PJ (2016). Cryo-EM structure of a CD4-bound open HIV-1 envelope trimer reveals structural rearrangements of the gp120 V1V2 loop. Proc Natl Acad Sci U S A 113, E7151–E7158. 10.1073/pnas.1615939113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pancera M, Majeed S, Ban YE, Chen L, Huang CC, Kong L, Kwon YD, Stuckey J, Zhou T, Robinson JE, et al. (2010). Structure of HIV-1 gp120 with gp41-interactive region reveals layered envelope architecture and basis of conformational mobility. Proc Natl Acad Sci U S A 107, 1166–1171. 10.1073/pnas.0911004107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Blumenthal R, Durell S, and Viard M (2012). HIV entry and envelope glycoprotein-mediated fusion. J Biol Chem 287, 40841–40849. 10.1074/jbc.R112.406272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.West AP Jr., Scharf L, Scheid JF, Klein F, Bjorkman PJ, and Nussenzweig MC (2014). Structural insights on the role of antibodies in HIV-1 vaccine and therapy. Cell 156, 633–648. 10.1016/j.cell.2014.01.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Guttman M, Cupo A, Julien JP, Sanders RW, Wilson IA, Moore JP, and Lee KK (2015). Antibody potency relates to the ability to recognize the closed, pre-fusion form of HIV Env. Nat Commun 6, 6144. 10.1038/ncomms7144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Veillette M, Desormeaux A, Medjahed H, Gharsallah NE, Coutu M, Baalwa J, Guan Y, Lewis G, Ferrari G, Hahn BH, et al. (2014). Interaction with cellular CD4 exposes HIV-1 envelope epitopes targeted by antibody-dependent cell-mediated cytotoxicity. J Virol 88, 2633–2644. 10.1128/JVI.03230-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ding S, Gasser R, Gendron-Lepage G, Medjahed H, Tolbert WD, Sodroski J, Pazgier M, and Finzi A (2019). CD4 Incorporation into HIV-1 Viral Particles Exposes Envelope Epitopes Recognized by CD4-Induced Antibodies. J Virol 93. 10.1128/JVI.01403-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Schoofs T, Klein F, Braunschweig M, Kreider EF, Feldmann A, Nogueira L, Oliveira T, Lorenzi JC, Parrish EH, Learn GH, et al. (2016). HIV-1 therapy with monoclonal antibody 3BNC117 elicits host immune responses against HIV-1. Science 352, 997–1001. 10.1126/science.aaf0972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kwong PD, Doyle ML, Casper DJ, Cicala C, Leavitt SA, Majeed S, Steenbeke TD, Venturi M, Chaiken I, Fung M, et al. (2002). HIV-1 evades antibody-mediated neutralization through conformational masking of receptor-binding sites. Nature 420, 678–682. 10.1038/nature01188. [DOI] [PubMed] [Google Scholar]
  • 29.Wei X, Decker JM, Wang S, Hui H, Kappes JC, Wu X, Salazar-Gonzalez JF, Salazar MG, Kilby JM, Saag MS, et al. (2003). Antibody neutralization and escape by HIV-1. Nature 422, 307–312. 10.1038/nature01470. [DOI] [PubMed] [Google Scholar]
  • 30.Wyatt R, Kwong PD, Desjardins E, Sweet RW, Robinson J, Hendrickson WA, and Sodroski JG (1998). The antigenic structure of the HIV gp120 envelope glycoprotein. Nature 393, 705–711. 10.1038/31514. [DOI] [PubMed] [Google Scholar]
  • 31.Chung AW, Isitman G, Navis M, Kramski M, Center RJ, Kent SJ, and Stratov I (2011). Immune escape from HIV-specific antibody-dependent cellular cytotoxicity (ADCC) pressure. Proc Natl Acad Sci U S A 108, 7505–7510. 10.1073/pnas.1016048108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Munro JB, Gorman J, Ma X, Zhou Z, Arthos J, Burton DR, Koff WC, Courter JR, Smith AB 3rd, Kwong PD, et al. (2014). Conformational dynamics of single HIV-1 envelope trimers on the surface of native virions. Science 346, 759–763. 10.1126/science.1254426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lu M, Ma X, Castillo-Menendez LR, Gorman J, Alsahafi N, Ermel U, Terry DS, Chambers M, Peng D, Zhang B, et al. (2019). Associating HIV-1 envelope glycoprotein structures with states on the virus observed by smFRET. Nature 568, 415–419. 10.1038/s41586-019-1101-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Li Z, Li W, Lu M, Bess J Jr., Chao CW, Gorman J, Terry DS, Zhang B, Zhou T, Blanchard SC, et al. (2020). Subnanometer structures of HIV-1 envelope trimers on aldrithiol-2-inactivated virus particles. Nat Struct Mol Biol 27, 726–734. 10.1038/s41594-020-0452-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Herschhorn A, Ma X, Gu C, Ventura JD, Castillo-Menendez L, Melillo B, Terry DS, Smith AB 3rd, Blanchard SC, Munro JB, et al. (2016). Release of gp120 Restraints Leads to an Entry-Competent Intermediate State of the HIV-1 Envelope Glycoproteins. mBio 7. 10.1128/mBio.01598-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Roy R, Hohng S, and Ha T (2008). A practical guide to single-molecule FRET. Nat Methods 5, 507–516. 10.1038/nmeth.1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lampe M, Briggs JA, Endress T, Glass B, Riegelsberger S, Krausslich HG, Lamb DC, Brauchle C, and Muller B (2007). Double-labelled HIV-1 particles for study of virus-cell interaction. Virology 360, 92–104. 10.1016/j.virol.2006.10.005. [DOI] [PubMed] [Google Scholar]
  • 38.Lu M (2021). Single-Molecule FRET Imaging of Virus Spike-Host Interactions. Viruses 13. 10.3390/v13020332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lu M, Ma X, and Mothes W (2019). Illuminating the virus life cycle with single-molecule FRET imaging. Adv Virus Res 105, 239–273. 10.1016/bs.aivir.2019.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yin J, Lin AJ, Golan DE, and Walsh CT (2006). Site-specific protein labeling by Sfp phosphopantetheinyl transferase. Nat Protoc 1, 280–285. 10.1038/nprot.2006.43. [DOI] [PubMed] [Google Scholar]
  • 41.Lin CW, and Ting AY (2006). Transglutaminase-catalyzed site-specific conjugation of small-molecule probes to proteins in vitro and on the surface of living cells. J Am Chem Soc 128, 4542–4543. 10.1021/ja0604111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zhang Z, Wang Q, Nguyen HT, Chen HC, Chiu TJ, Smith Iii AB, and Sodroski JG (2023). Alterations in gp120 glycans or the gp41 fusion peptide-proximal region modulate the stability of the human immunodeficiency virus (HIV-1) envelope glycoprotein pretriggered conformation. J Virol 97, e0059223. 10.1128/jvi.00592-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ma X, Lu M, Gorman J, Terry DS, Hong X, Zhou Z, Zhao H, Altman RB, Arthos J, Blanchard SC, et al. (2018). HIV-1 Env trimer opens through an asymmetric intermediate in which individual protomers adopt distinct conformations. Elife 7. 10.7554/eLife.34271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Sakin V, Hanne J, Dunder J, Anders-Osswein M, Laketa V, Nikic I, Krausslich HG, Lemke EA, and Muller B (2017). A Versatile Tool for Live-Cell Imaging and Super-Resolution Nanoscopy Studies of HIV-1 Env Distribution and Mobility. Cell Chem Biol 24, 635–645 e635. 10.1016/j.chembiol.2017.04.007. [DOI] [PubMed] [Google Scholar]
  • 45.Nikic I, Plass T, Schraidt O, Szymanski J, Briggs JA, Schultz C, and Lemke EA (2014). Minimal tags for rapid dual-color live-cell labeling and super-resolution microscopy. Angew Chem Int Ed Engl 53, 2245–2249. 10.1002/anie.201309847. [DOI] [PubMed] [Google Scholar]
  • 46.Nikic I, Estrada Girona G, Kang JH, Paci G, Mikhaleva S, Koehler C, Shymanska NV, Ventura Santos C, Spitz D, and Lemke EA (2016). Debugging Eukaryotic Genetic Code Expansion for Site-Specific Click-PAINT Super-Resolution Microscopy. Angew Chem Int Ed Engl 55, 16172–16176. 10.1002/anie.201608284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Nikic I, Kang JH, Girona GE, Aramburu IV, and Lemke EA (2015). Labeling proteins on live mammalian cells using click chemistry. Nat Protoc 10, 780–791. 10.1038/nprot.2015.045. [DOI] [PubMed] [Google Scholar]
  • 48.Plass T, Milles S, Koehler C, Schultz C, and Lemke EA (2011). Genetically encoded copper-free click chemistry. Angew Chem Int Ed Engl 50, 3878–3881. 10.1002/anie.201008178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sletten EM, and Bertozzi CR (2011). From mechanism to mouse: a tale of two bioorthogonal reactions. Acc Chem Res 44, 666–676. 10.1021/ar200148z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shandell MA, Tan Z, and Cornish VW (2021). Genetic Code Expansion: A Brief History and Perspective. Biochemistry 60, 3455–3469. 10.1021/acs.biochem.1c00286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lang K, and Chin JW (2014). Cellular incorporation of unnatural amino acids and bioorthogonal labeling of proteins. Chem Rev 114, 4764–4806. 10.1021/cr400355w. [DOI] [PubMed] [Google Scholar]
  • 52.Schifferdecker S, Zila V, Muller TG, Sakin V, Anders-Osswein M, Laketa V, Krausslich HG, and Muller B (2022). Direct Capsid Labeling of Infectious HIV-1 by Genetic Code Expansion Allows Detection of Largely Complete Nuclear Capsids and Suggests Nuclear Entry of HIV-1 Complexes via Common Routes. mBio 13, e0195922. 10.1128/mbio.01959-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Plass T, Milles S, Koehler C, Szymanski J, Mueller R, Wiessler M, Schultz C, and Lemke EA (2012). Amino acids for Diels-Alder reactions in living cells. Angew Chem Int Ed Engl 51, 4166–4170. 10.1002/anie.201108231. [DOI] [PubMed] [Google Scholar]
  • 54.Sakin V, Paci G, Lemke EA, and Muller B (2016). Labeling of virus components for advanced, quantitative imaging analyses. FEBS Lett 590, 1896–1914. 10.1002/1873-3468.12131. [DOI] [PubMed] [Google Scholar]
  • 55.Wu X, Parast AB, Richardson BA, Nduati R, John-Stewart G, Mbori-Ngacha D, Rainwater SM, and Overbaugh J (2006). Neutralization escape variants of human immunodeficiency virus type 1 are transmitted from mother to infant. J Virol 80, 835–844. 10.1128/JVI.80.2.835-844.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Poss M, and Overbaugh J (1999). Variants from the diverse virus population identified at seroconversion of a clade A human immunodeficiency virus type 1-infected woman have distinct biological properties. J Virol 73, 5255–5264. 10.1128/JVI.73.7.5255-5264.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Haddox HK, Dingens AS, Hilton SK, Overbaugh J, and Bloom JD (2018). Mapping mutational effects along the evolutionary landscape of HIV envelope. Elife 7. 10.7554/eLife.34420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Reinkemeier CD, and Lemke EA (2021). Synthetic biomolecular condensates to engineer eukaryotic cells. Curr Opin Chem Biol 64, 174–181. 10.1016/j.cbpa.2021.08.005. [DOI] [PubMed] [Google Scholar]
  • 59.Staropoli I, Dufloo J, Ducher A, Commere PH, Sartori-Rupp A, Novault S, Bruel T, Lorin V, Mouquet H, Schwartz O, and Casartelli N (2020). Flow Cytometry Analysis of HIV-1 Env Conformations at the Surface of Infected Cells and Virions: Role of Nef, CD4, and SERINC5. J Virol 94. 10.1128/JVI.01783-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, Mitcham JL, et al. (2009). Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326, 285–289. 10.1126/science.1178746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Blattner C, Lee JH, Sliepen K, Derking R, Falkowska E, de la Pena AT, Cupo A, Julien JP, van Gils M, Lee PS, et al. (2014). Structural delineation of a quaternary, cleavage-dependent epitope at the gp41-gp120 interface on intact HIV-1 Env trimers. Immunity 40, 669–680. 10.1016/j.immuni.2014.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lu M, Uchil PD, Li W, Zheng D, Terry DS, Gorman J, Shi W, Zhang B, Zhou T, Ding S, et al. (2020). Real-Time Conformational Dynamics of SARS-CoV-2 Spikes on Virus Particles. Cell Host Microbe 28, 880–891 e888. 10.1016/j.chom.2020.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Das DK, Govindan R, Nikic-Spiegel I, Krammer F, Lemke EA, and Munro JB (2018). Direct Visualization of the Conformational Dynamics of Single Influenza Hemagglutinin Trimers. Cell 174, 926–937 e912. 10.1016/j.cell.2018.05.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Miyauchi K, Kim Y, Latinovic O, Morozov V, and Melikyan GB (2009). HIV enters cells via endocytosis and dynamin-dependent fusion with endosomes. Cell 137, 433–444. 10.1016/j.cell.2009.02.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Melikyan GB (2014). HIV entry: a game of hide-and-fuse? Curr Opin Virol 4, 1–7. 10.1016/j.coviro.2013.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Das DK, Bulow U, Diehl WE, Durham ND, Senjobe F, Chandran K, Luban J, and Munro JB (2020). Conformational changes in the Ebola virus membrane fusion machine induced by pH, Ca2+, and receptor binding. PLoS Biol 18, e3000626. 10.1371/journal.pbio.3000626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Diaz-Salinas MA, Li Q, Ejemel M, Yurkovetskiy L, Luban J, Shen K, Wang Y, and Munro JB (2022). Conformational dynamics and allosteric modulation of the SARS-CoV-2 spike. Elife 11. 10.7554/eLife.75433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yang Z, Han Y, Ding S, Shi W, Zhou T, Finzi A, Kwong PD, Mothes W, and Lu M (2022). SARS-CoV-2 Variants Increase Kinetic Stability of Open Spike Conformations as an Evolutionary Strategy. mBio 13, e0322721. 10.1128/mbio.03227-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Nikic I, and Lemke EA (2015). Genetic code expansion enabled site-specific dual-color protein labeling: superresolution microscopy and beyond. Curr Opin Chem Biol 28, 164–173. 10.1016/j.cbpa.2015.07.021. [DOI] [PubMed] [Google Scholar]
  • 70.Serwa RA, Sekine E, Brown J, Teo SHC, Tate EW, and O’Hare P (2019). Analysis of a fully infectious bio-orthogonally modified human virus reveals novel features of virus cell entry. PLoS Pathog 15, e1007956. 10.1371/journal.ppat.1007956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gifford LB, and Melikyan GB (2023). Human Immunodeficiency Virus 1 Capsid Uncoating in the Nucleus Progresses Through Defect Formation in the Capsid Lattice. bioRxiv, 2023.2008.2022.553958. 10.1101/2023.08.22.553958. [DOI] [Google Scholar]
  • 72.Chertova E, Bess JW Jr., Crise BJ, Sowder IR, Schaden TM, Hilburn JM, Hoxie JA, Benveniste RE, Lifson JD, Henderson LE, and Arthur LO (2002). Envelope glycoprotein incorporation, not shedding of surface envelope glycoprotein (gp120/SU), Is the primary determinant of SU content of purified human immunodeficiency virus type 1 and simian immunodeficiency virus. J Virol 76, 5315–5325. 10.1128/jvi.76.11.5315-5325.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zhu P, Chertova E, Bess J Jr., Lifson JD, Arthur LO, Liu J, Taylor KA, and Roux KH (2003). Electron tomography analysis of envelope glycoprotein trimers on HIV and simian immunodeficiency virus virions. Proc Natl Acad Sci U S A 100, 15812–15817. 10.1073/pnas.2634931100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chojnacki J, Staudt T, Glass B, Bingen P, Engelhardt J, Anders M, Schneider J, Muller B, Hell SW, and Krausslich HG (2012). Maturation-dependent HIV-1 surface protein redistribution revealed by fluorescence nanoscopy. Science 338, 524–528. 10.1126/science.1226359. [DOI] [PubMed] [Google Scholar]
  • 75.Sharma M, Marin M, Wu H, Prikryl D, and Melikyan GB (2023). Human Immunodeficiency Virus 1 Preferentially Fuses with pH-Neutral Endocytic Vesicles in Cell Lines and Human Primary CD4+ T-Cells. ACS Nano 17, 17436–17450. 10.1021/acsnano.3c05508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wang K, Neumann H, Peak-Chew SY, and Chin JW (2007). Evolved orthogonal ribosomes enhance the efficiency of synthetic genetic code expansion. Nat Biotechnol 25, 770–777. 10.1038/nbt1314. [DOI] [PubMed] [Google Scholar]
  • 77.Reinkemeier CD, Girona GE, and Lemke EA (2019). Designer membraneless organelles enable codon reassignment of selected mRNAs in eukaryotes. Science 363. 10.1126/science.aaw2644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Reinkemeier CD, and Lemke EA (2021). Dual film-like organelles enable spatial separation of orthogonal eukaryotic translation. Cell 184, 4886–4903 e4821. 10.1016/j.cell.2021.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Schmied WH, Elsasser SJ, Uttamapinant C, and Chin JW (2014). Efficient multisite unnatural amino acid incorporation in mammalian cells via optimized pyrrolysyl tRNA synthetase/tRNA expression and engineered eRF1. J Am Chem Soc 136, 15577–15583. 10.1021/ja5069728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Arsic A, Hagemann C, Stajkovic N, Schubert T, and Nikic-Spiegel I (2022). Minimal genetically encoded tags for fluorescent protein labeling in living neurons. Nat Commun 13, 314. 10.1038/s41467-022-27956-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Neumann H, Wang K, Davis L, Garcia-Alai M, and Chin JW (2010). Encoding multiple unnatural amino acids via evolution of a quadruplet-decoding ribosome. Nature 464, 441–444. 10.1038/nature08817. [DOI] [PubMed] [Google Scholar]
  • 82.Charneau P, Alizon M, and Clavel F (1992). A second origin of DNA plus-strand synthesis is required for optimal human immunodeficiency virus replication. J Virol 66, 2814–2820. 10.1128/JVI.66.5.2814-2820.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zhong P, Agosto LM, Ilinskaya A, Dorjbal B, Truong R, Derse D, Uchil PD, Heidecker G, and Mothes W (2013). Cell-to-cell transmission can overcome multiple donor and target cell barriers imposed on cell-free HIV. PLoS One 8, e53138. 10.1371/journal.pone.0053138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Jin J, Sherer NM, Heidecker G, Derse D, and Mothes W (2009). Assembly of the murine leukemia virus is directed towards sites of cell-cell contact. PLoS Biol 7, e1000163. 10.1371/journal.pbio.1000163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Park S, Auyeung A, Lee DL, Lambert PF, Carchman EH, and Sherer NM (2021). HIV-1 Protease Inhibitors Slow HPV16-Driven Cell Proliferation through Targeted Depletion of Viral E6 and E7 Oncoproteins. Cancers (Basel) 13. 10.3390/cancers13050949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Juette MF, Terry DS, Wasserman MR, Altman RB, Zhou Z, Zhao H, and Blanchard SC (2016). Single-molecule imaging of non-equilibrium molecular ensembles on the millisecond timescale. Nat Methods 13, 341–344. 10.1038/nmeth.3769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Aitken CE, Marshall RA, and Puglisi JD (2008). An oxygen scavenging system for improvement of dye stability in single-molecule fluorescence experiments. Biophys J 94, 1826–1835. 10.1529/biophysj.107.117689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.McKinney SA, Joo C, and Ha T (2006). Analysis of single-molecule FRET trajectories using hidden Markov modeling. Biophys J 91, 1941–1951. 10.1529/biophysj.106.082487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Barth A, Opanasyuk O, Peulen TO, Felekyan S, Kalinin S, Sanabria H, and Seidel CAM (2022). Unraveling multi-state molecular dynamics in single-molecule FRET experiments. I. Theory of FRET-lines. J Chem Phys 156, 141501. 10.1063/5.0089134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hosoda H, Mori H, Sogoshi N, Nagasawa A, and Nakabayashi S (2004). Refractive indices of water and aqueous electrolyte solutions under high magnetic fields. J Phys Chem A 108, 1461–1464. 10.1021/jp0310145. [DOI] [Google Scholar]
  • 91.Sasmal DK, Pulido LE, Kasal S, and Huang J (2016). Single-molecule fluorescence resonance energy transfer in molecular biology. Nanoscale 8, 19928–19944. 10.1039/c6nr06794h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wei X, Decker JM, Liu H, Zhang Z, Arani RB, Kilby JM, Saag MS, Wu X, Shaw GM, and Kappes JC (2002). Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob Agents Chemother 46, 1896–1905. 10.1128/AAC.46.6.1896-1905.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Derdeyn CA, Decker JM, Sfakianos JN, Wu XY, O’Brien WA, Ratner L, Kappes JC, Shaw GM, and Hunter E (2000). 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 74, 8358–8367. Doi 10.1128/Jvi.74.18.8358-8367.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Platt EJ, Wehrly K, Kuhmann SE, Chesebro B, and Kabat D (1998). Effects of CCR5 and CD4 cell surface concentrations on infections by macrophagetropic isolates of human immunodeficiency virus type 1. J Virol 72, 2855–2864. 10.1128/JVI.72.4.2855-2864.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

Movie 1. Internalization of an amber-free click-labeled HIV-1Q23 BG505 Env N136TAG-Cy5 virion into a live cell, related to Figure 6.

Representative movie of a single Env N136TAG-Cy5 (green) virion entering into a TZM-bl cell labeled with SPY555-Actin (magenta). The virion initially binds to the cell periphery and later undergoes internalization into the peripheral actin cortex, where high degrees of mobility and displacement are observed within the cell.

Download video file (34MB, avi)
2
3

Data Availability Statement

  • Sequence and map of replication-incompetent amber-free HIV-1 provirus plasmid have been deposited into Addgene under plasmid number 213007 listed in the key resources table. Sequences and maps of both replication-competent and incompetent amber-free HIV-1 provirus have been deposited at Mendeley. The DOI is listed in the key resources table. Other data reported in this paper will be shared by the lead contact upon request.

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

RESOURCES