Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 8.
Published in final edited form as: Science. 2016 Jun 23;353(6295):172–175. doi: 10.1126/science.aaf7066

Structural Basis for Membrane Anchoring of HIV-1 Envelope Spike

Jyoti Dev 1,5,#, Donghyun Park 2,#, Qingshan Fu 1, Jia Chen 2,3, Heather Jiwon Ha 2,3, Fadi Ghantous 4, Tobias Herrmann 5, Weiting Chang 2, Zhijun Liu 6, Gary Frey 2,3, Michael S Seaman 4, Bing Chen 2,3,*, James J Chou 1,6
PMCID: PMC5085267  NIHMSID: NIHMS802647  PMID: 27338706

Abstract

HIV-1 envelope spike (Env) is a type I membrane protein that mediates viral entry. We use NMR to determine an atomic structure of the transmembrane (TM) domain of HIV-1 Env reconstituted in bicelles that mimic a lipid bilayer. The TM forms a well-ordered trimer that protects a conserved membrane-embedded arginine. An N-terminal coiled-coil and a C-terminal hydrophilic core stabilize the trimer. Individual mutations of conserved residues did not disrupt the TM trimer and minimally affected membrane fusion and infectivity. Major changes in the hydrophilic core, however, altered the antibody sensitivity of Env. These results show how a TM domain anchors, stabilizes and modulates a viral envelope spike and suggest that its influence on Env conformation is an important consideration for HIV-1 immunogen design.


HIV-1 envelope spike [Env; trimeric (gp160)3, cleaved to (gp120/gp41)3] fuses viral and host cell membranes to initiate infection (1). Binding of gp120 to receptor (CD4) and coreceptor (e.g., CCR5 or CXCR4) trigger large conformational changes, leading to a cascade of refolding events in gp41 and ultimately to membrane fusion (2-4). Mature Env spikes, (gp120/gp41)3, are the sole antigens on the virion surface; they often induce strong antibody responses in infected individuals (5, 6). A vast amount of structural information is available for the ectodomain of Env, a primary target of the host immune system, but much less for its transmembrane domain (TMD), membrane proximal external region (MPER) and cytoplasmic tail (CT), in the context of lipid bilayer. The cryoEM (electron microscopy) structure of a detergent-solubilized clade B JR-FL EnvΔCT construct without the CT has been described recently (7), but its MPER and TMD are disordered probably because detergent micelles failed to mimic a membrane environment.

The HIV-1 TMD is more conserved than a typical membrane anchor (Fig. S1). Previous studies showed that mutations and truncations in the TMD indeed affect membrane fusion and viral infectivity (8-11). Presence of a GxxxG motif, often implicated in oligomeric assembly of TM helices (12), suggests clustering of TMDs in membrane (Fig. S1). The presence of a conserved, positively charged residue (usually arginine) near the middle of the TMD suggests functions other than just spanning a bilayer. TM helices of many cell surface receptors are not merely inert anchors but play essential roles in receptor assembly and signal transmission. For example, we have shown that CT truncation affects the antigenic surface of the ectodomain of HIV-1 Env on the opposite side of the membrane (13). Thus, understanding the physical coupling (conformation and/or dynamics) between the CT and the ectodomain mediated by the TMD may guide design of immunogens that mimic native, functional Env and induce broadly neutralizing antibodies (bnAbs).

To characterize the TMD structure by NMR, we used a fragment of gp41 (residues 677-716; HXB2 numbering, Fig. S1), derived from a clade D HIV-1 isolate 92UG024.2 (14). This construct, gp41HIV1D(677-716), contains a short stretch of MPER (residues 677-683), the TM segment (residues 684-705), defined by hydrophobicity, and a fragment previously assigned to the CT domain (residues 706-716, containing a tyrosine-based sorting motif (15, 16)). The gp41HIV1D(677-716) protein was purified and reconstituted into bicelles (Fig. S2A and S2B) (17-19) with an expected lipid-bilayer diameter of ~44 Å (Fig. S2C) (20, 21), thereby incorporating the refolded gp41HIV1D(677-716) into a membrane-like environment. The bicelle-reconstituted gp41HIV1D(677-716) migrated on SDS-PAGE with a size close to that of a trimer (theoretical M.W. 14.1 kDa) (Fig. S2D), suggesting that the protein was trimeric and resistant to SDS denaturation. The reconstituted gp41HIV1D(677-716) protein in bicelles generated NMR spectrum with excellent chemical shift dispersion (Fig. S3A). The equivalent protein constructs from isolates 92BR025.9 (clade C) and 92RU131.16 (clade G) gave similar NMR spectra (Fig. S3B and C), suggesting that the TMDs of most HIV-1 Envs have similar structures when reconstituted in bicelles. We completed the NMR structure of gp41HIV1D(677-716) using a previously described protocol (Figs. S4 and S5) (22, 23). The final ensemble of structures converged to RMSD of 0.95 Å and 1.44 Å for backbone and all heavy atoms, respectively (Fig. S6, Table S1).

gp41HIV1D(677-716) is a tightly assembled trimer ~54 Å long, with the conserved arginine (R696) near its midpoint (Fig. 1A). It shows a packing arrangement not seen in any other known TM helix dimers or trimers: its N- and C-terminal halves have different modes of assembly, with an intervening kink. The N-terminal region is a conventional three-chain coiled-coil formed by residues 686-696 (Fig. 1B), including the GxxxG motif. The C-terminal half does not show classic “knobs-into-holes” interactions, but instead is held together by a network of polar contacts, mainly involving R707 and Q710, at the trimer interface of the kinked helical segments (residues 704-712) (Fig. 1C). We call this interface the “hydrophilic core”.

Figure 1. NMR structure of the gp41HIV1D(677-716) trimer in bicelles.

Figure 1

(A) Ribbon representation of the lowest energy structure from the calculated ensemble. The sphere representation of the top view (lower right) shows that the trimer has no ion permeable holes. (B) The N-terminal half of the structure with hydrophobic residues (orange) arranged in the coiled-coil pattern (right panel). (C) The C-terminal half of the structure showing an array of polar residues that form the C-terminal hydrophilic core. The network of polar contacts is hypothesized to stabilize the trimer. (D) Zoomed middle region of the structure showing the intramembrane R696 and its surrounding hydrophobic residues, as well as the backbone oxygen of L692.

R696, near the middle of each TM helix (Fig. 1D), produces three unbalanced charges at the center of the membrane. This residue occupies a ‘d’ position in the heptad sequence (Fig. 1B). Its Cβ points toward the three-fold axis of the trimer, while the rest of the sidechain bends away from the axis, placing the guanidinium group in a peripheral hydrophobic pocket formed by L692, L695, and I697 (Fig. 1D). The backbone carbonyl of L692 may form a hydrogen bond with one of the guanidinium NH of R696. Hε of R696 showed water NOE in a 15N-edited NOE spectrum (Fig. S4), indicating the presence of an adjacent structured water. Thus, the guanidinium group, presumably charged at pH 6.7 under the NMR conditions, is partially neutralized by hydrogen bonding with the electronegative backbone oxygen of L692 and the water molecule. The polarizability of the hydrophobic pocket that surrounds the guanidinium may also lower its pKa from its value in aqueous solution. Although well accommodated in the TMD trimer, the intramembrane R696 may modulate the stability of the helical trimer if the helices dissociate at any stage in assembly or fusion. The 1H-15N correlation spectrum of the gp41HIV1D(677-716) trimer showed inhomogeneous peak linewidth, with the N-terminal half near R696 having the most severe peak broadening, consistent with conformational fluctuation (Fig. S7).

To confirm membrane partition of the TMD trimer, we used a paramagnetic probe, Gd(DOTA) (24), to measure solvent exposure of the four arginine residues in the gp41HIV1D(677-716) trimer. These arginines are distributed at different positions along the TM helices and thus serve as four depth markers. We measured intensity decrease of the arginine Hε-Nε correlation peaks at increasing concentrations of Gd(DOTA). The most solvent-exposed R707 showed the highest sensitivity to Gd(DOTA), whereas the most buried R696 was the least sensitive (Fig. 2A). R683 and R709 are near opposite lipid headgroup regions in the structure. R709 showed a greater resonance broadening than did R683, indicating that the latter is more deeply buried. We placed the gp41HIV1D(677-716) trimer in the lipid bilayer so that the four arginine positions were consistent with their respective sensitivity to Gd(DOTA) (Fig. 2B). This placement, which is consistent with the surface distribution of hydrophobic, polar, and charged residues (Fig. 2C), places R696 in a fully hydrophobic environment, slightly closer to the cytoplasmic side of the membrane. The MPER segment is in the headgroup region of the outer leaflet. The C-terminal segment, previously assigned to the CT, is at the headgroup-water interface of the inner leaflet.

Figure 2. Partition of the gp41HIV1D(677-716) trimer in lipid bilayer.

Figure 2

(A) Measurement of membrane immersion depth of the four arginines using the water-soluble and membrane-inaccessible paramagnetic probe Gd-DOTA. The 1H-15N correlation peaks of the arginine Hε-Nε recorded in q=0.5 bicelles (left) showed reduced peak intensity with increasing Gd-DOTA concentration (right) due to paramagnetic relaxation enhancement (PRE). R683 is shown in green, R696 in blue, R707 in magenta and R709 on orange. Error bars represent spectra noise level normalized against maximum reference peak intensity. (B) Placement of the trimer structure in the presumed DMPC lipid bilayer, which is slightly thinner than the characterized thickness of DOPC bilayer (35), such that the positions of the four arginines are in accordance with the PRE results in (A). The positions of the arginine sidechains are indicated by the Hε (spheres) in the ensemble of 15 low energy structures. The color scheme is the same as in (A). (C) Surface representation of the lowest energy structure positioned in DMPC bilayer as in (B) for showing the surface distribution of hydrophobic, polar, and charged amino acids. The hydrophobic residues (yellow) include A, I, L, F, V, P, and G; the polar residues (cyan) include Q, N, H, S, T, Y, C, M, and W; the charged residues (blue) include K, R, D, and E.

To assess contribution of specific residues to TMD stability, we generated 12 gp41HIV1D(677-716) mutants with single or double mutations, mainly in the trimer interface. To introduce large-scale changes in the hydrophilic core, we also created mutants Δ(705-716) and G690L/Δ(705-716) with residues 705-716 deleted, and mutants 704-713 and G690L/704-713 with residues 704-713 mutated (Fig. 3 and Table S2). We analyzed these mutants by SDS-PAGE after reconstitution in bicelles (Fig. S8). Most simple mutations, including I686A, I693A and R696N, did not disrupt the trimer completely, but only shifted the band to M.W. positions lower than that of the wildtype, indicating only partial trimer destabilization (Fig. S8). We saw a similar pattern for mutants 704-713, G690L/704-713, and Δ705-716. The only mutant that migrated as a monomer was G690L/Δ705-716, with the GxxxG motif mutated and the entire hydrophilic-core region truncated (Fig. S8). Thus, both the coiled-coil and the hydrophilic core contribute to the extraordinary stability of the TMD, and either one of them is sufficient to prevent the trimer from complete dissociation in bicelles.

Figure 3. Effect of mutations in the TMD of HIV-1 Env on its antibody sensitivity.

Figure 3

(A) Antibody inhibition of cell-cell fusion mediated by the wildtype 92UG037.8 Env (left) and the TMD mutant 704-713 (right; residues 704-713 (VINRVRQGYS) were mutated to SSAASAAGSA.) was analyzed using both non-neutralizing antibodies, including b6 (CD4 binding site; blue), 3791 (V3; cyan) and 17b (CD4-induced; purple), and trimer-specific bnAbs, including PG9 (orange), PG16 (red) and PGT145 (magenta). The CD4 binding site bnAb VRC01 (green) was a control antibody. The experiment was carried out in triplicate and repeated at least twice with similar results. Error bars indicate the standard deviation calculated by the Excel STDEV function. (B) Antibody neutralization of pseudoviruses containing either the 92UG037.8 Env (left) or the TMD mutant 704-713 (right) was determined using antibodies, b6, 3791, 17b, PG9, PG16, and PGT145 shown in the same color scheme as in (A). The CD4 binding site bnAb 3BNC117 was a control antibody shown in green. The experiment was performed in duplicate.

To elucidate functional roles of the structural elements in the TMD, we mutated each of them in the intact Env spike and analyzed the impact on Env biogenesis, membrane fusion and viral infectivity. We generated 27 Env mutants, using the sequence of a clade A isolate 92UG037.8, with single, double or triple mutations in the coiled-coil region, R696 and its protecting residues, the kink, the hydrophilic core, or combinations of these elements (Table S2). We also produced mutants 704-713 and G690L/704-713. When transiently transfected in 293T cells, all mutants expressed comparable levels of Env, with similar extents of cleavage between gp120 and gp41, as well as similar cell-surface levels (Fig. S9 and S10). At a high Env expression level, at which cell-cell fusion was resistant to most neutralizing antibodies, the fusion activity of the TMD mutants was indistinguishable (93-109%) from that of the wildtype (Fig. S11). When the expression level was reduced to mimic the low surface density on HIV-1 virions (25), several TMD mutants, including R696A, had significantly lower cell-cell fusion activity than did wildtype (Table S2 and Fig. S12). Likewise, when these mutations were introduced into pseudoviruses, there were no major differences in Env incorporation and processing, but limited changes in infectivity for most of them (Table S2, Figs. S13 and S14). R696A had almost full wildtype infectivity, while 704-713 and G690L/704-713 had substantially less -- just the opposite of their effects on cell-cell fusion (Table S2 and Fig. S14).

To determine whether mutations in the TMD can influence the antigenic structure of the Env ectodomain and whether they might affect its antibody sensitivity, we used the cell-cell fusion assay to test inhibition of each Env mutant by a trimer-specific bnAb PG16 and by a non-neutralizing V3 antibody 3791 (Table S3). Most mutants were essentially identical to the wildtype in their sensitivity to either antibody; mutants I686A, Y712A, P714N, G690L/Y712A, G690L/P714N, G690L/P714K, and G690L/P714H exhibited detectable differences (Fig. S15). We observed the most striking differences for mutants 704-713 and G690L/704-713, for which PG16 and 3791 switched phenotypes -- the former became inactive and the latter inhibitory (Fig. S15). All mutations that affect antibody inhibition are located in either the coiled-coil or the hydrophilic core. We infer that changes in TMD stability influence the antigenic structure of the ectodomain of the functional Env. When tested in a pseudovirus-based neutralization assay with bnAbs PG9 (trimer specific), 3BNC117 (CD4 binding site), 10-1074 (glycan- and V3-dependent), 10E8 (MPER); and 3791 (Table S3), most mutants, except for 704-713 and G690/704-713, were unchanged in their sensitivity to PG9, 3BNC117, 10-1074 and 3791, but most became more sensitive to 10E8 (Table S4). This result suggests that the changes produced by all the mutations tested, except for 704-713 and G690/704-713, are limited to local structure. In contrast, the mutants 704-713 and G690/704-713 became resistant to PG9 and sensitive to 3791 -- reflecting their properties in the cell-cell fusion assay. Mutant 704-713 was analyzed with additional antibodies. For cell-cell fusion, wildtype Env is sensitive to trimer-specific bnAbs PG9, PG16 and PGT145 and resistant to non-neutralizing antibodies b6 (CD4 binding site), 3791, and 17b (CD4-induced) (Table S3; Figs. 3A and S16). The antibody inhibition pattern is reversed, however, for the fully functional mutant 704-713, indicating that the hydrophilic core of the TMD plays a significant role in stabilizing and modulating the antigenic structure of the Env spike. Similar phenotypes were also observed with a 704-713 mutant derived from a clade C strain C97ZA012 (Fig. S17). The pseudovirus neutralization assay gave similar, but less pronounced, results for the mutant 704-713 (Fig. 3B and Table S4).

The most important finding from this study relevant to vaccine development is how the TMD modulates the antigenic surfaces of the Env spike. We reported previously that truncation of the CT domain of HIV-1 Env reshapes the antigenic surfaces of its ectodomain (13). We now show that mutations destabilizing the hydrophilic core of the TMD trimer resemble the CT deletion in altering the sensitivity of the functional Env to both non-neutralizing and trimer-specific neutralizing antibodies. In particular, the trimer-specific bnAbs, which neutralize by stabilizing the native conformation of Env (26, 27), do not recognize the Env spike when its TMD has been destabilized. We suggest that the TMD mediates conformational coupling between the ectodomain and the CT and that the trimeric structure seen by NMR represents the conformation of the TMD adopted by a native Env spike in a membrane.

The placement of gp41HIV1D(677-716) in a lipid bilayer reveals clear boundaries of the TM segment and settles a contentious issue (10, 28). Part of the 10E8 epitope (residues 677-683) is embedded in the headgroup layer of the outer leaflet, consistent with lack of accessibility of this epitope on the native Env (13, 29). The hydrophilic core, which was thought to be part of the CT, is similarly protected by the headgroup layer of the inner leaflet. This hydrophilic region contains a tyrosine-based sorting signal (712YSPL715), which may participate in Env internalization by endocytosis (15, 16, 30). Our structure indicates that Y712 and P714 in this motif on one TM protomer interact with L704 and V708 of the adjacent protomer, respectively, thereby also contributing to trimer stability.

The TMD is required not only for membrane anchoring and fusion, but also for stability of the entire Env spike. This observation can explain why most soluble Env preparations with the TMD deleted, except for those of a few selected strains (31, 32), are unstable and conformationally heterogeneous, unless they have specific, stabilizing modifications (26, 33, 34). To design immunogens that mimic optimally native viral spikes, one must not ignore structural constraints imposed by the TMD on the ectodomain. The high-resolution structure of the HIV-1 Env TMD trimer presented here can be a guide for engineering more effective immunogens.

Supplementary Material

SM

Acknowledgments

We thank S. Harrison and S. Rits-Volloch for generous advice and assistance, D. Barouch, B. Haynes and A. Carfi for critical reading of the manuscript, and the NIH AIDS Reagent Program, Division of AIDS, NIAID, NIH for reagents. The data presented in this manuscript are tabulated in the main paper and in the supplementary materials. The atomic structure coordinate and structural constraints are deposited in the Protein Data Bank (PDB) under the accession number 5JYN. The chemical shift values are deposited in the Biological Magnetic Resonance Data Bank (BMRB) under the accession number 30090. This work was supported by NIH grants AI084794 (to B.C. and Dan H. Barouch), GM083680 (to B.C.), AI106488 (to B.C.), HL103526 (to J.J.C.), Collaboration for AIDS Vaccine Discovery (CAVD) grant OPP1040741 (to Dan H. Barouch from the Bill and Melinda Gates Foundation) and the Center for HIV/AIDS Vaccine Immunology – Immunogen Design AI-100645 (to Barton F. Haynes). The NMR data were collected at the NMR facility of NCPSS (supported by CAS grant XDB08030301) and MIT-Harvard CMR (supported by NIH grant P41 EB-002026).

References and Notes

  • 1.Harrison SC. Viral membrane fusion. Nature structural & molecular biology. 2008;15:690–698. doi: 10.1038/nsmb.1456. published online EpubJul (10.1038/nsmb.1456) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Chan DC, Fass D, Berger JM, Kim PS. Core structure of gp41 from the HIV envelope glycoprotein. Cell. 1997;89:263–273. doi: 10.1016/s0092-8674(00)80205-6. [DOI] [PubMed] [Google Scholar]
  • 3.Weissenhorn W, Dessen A, Harrison SC, Skehel JJ, Wiley DC. Atomic structure of the ectodomain from HIV-1 gp41. Nature. 1997;387:426–430. doi: 10.1038/387426a0. [DOI] [PubMed] [Google Scholar]
  • 4.Pancera M, Zhou T, Druz A, Georgiev IS, Soto C, Gorman J, Huang J, Acharya P, Chuang GY, Ofek G, Stewart-Jones GB, Stuckey J, Bailer RT, Joyce MG, Louder MK, Tumba N, Yang Y, Zhang B, Cohen MS, Haynes BF, Mascola JR, Morris L, Munro JB, Blanchard SC, Mothes W, Connors M, Kwong PD. Structure and immune recognition of trimeric pre-fusion HIV-1 Env. Nature. 2014;514:455–461. doi: 10.1038/nature13808. published online EpubOct 23 (10.1038/nature13808) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wei X, Decker JM, Wang S, Hui H, Kappes JC, Wu X, Salazar-Gonzalez JF, Salazar MG, Kilby JM, Saag MS, Komarova NL, Nowak MA, Hahn BH, Kwong PD, Shaw GM. Antibody neutralization and escape by HIV-1. Nature. 2003;422:307–312. doi: 10.1038/nature01470. published online EpubMar 20. [DOI] [PubMed] [Google Scholar]
  • 6.Richman DD, Wrin T, Little SJ, Petropoulos CJ. Rapid evolution of the neutralizing antibody response to HIV type 1 infection. Proc Natl Acad Sci U S A. 2003;100:4144–4149. doi: 10.1073/pnas.0630530100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lee JH, Ozorowski G, Ward AB. Cryo-EM structure of a native, fully glycosylated, cleaved HIV-1 envelope trimer. Science. 2016;351:1043–1048. doi: 10.1126/science.aad2450. published online EpubMar 4 (10.1126/science.aad2450) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Owens RJ, Burke C, Rose JK. Mutations in the membrane-spanning domain of the human immunodeficiency virus envelope glycoprotein that affect fusion activity. J Virol. 1994;68:570–574. doi: 10.1128/jvi.68.1.570-574.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Shang L, Yue L, Hunter E. Role of the membrane-spanning domain of human immunodeficiency virus type 1 envelope glycoprotein in cell-cell fusion and virus infection. J Virol. 2008;82:5417–5428. doi: 10.1128/JVI.02666-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Yue L, Shang L, Hunter E. Truncation of the membrane-spanning domain of human immunodeficiency virus type 1 envelope glycoprotein defines elements required for fusion, incorporation, and infectivity. J Virol. 2009;83:11588–11598. doi: 10.1128/JVI.00914-09. published online EpubNov (10.1128/JVI.00914-09) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Helseth E, Olshevsky U, Gabuzda D, Ardman B, Haseltine W, Sodroski J. Changes in the transmembrane region of the human immunodeficiency virus type 1 gp41 envelope glycoprotein affect membrane fusion. J Virol. 1990;64:6314–6318. doi: 10.1128/jvi.64.12.6314-6318.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Teese MG, Langosch D. Role of GxxxG Motifs in Transmembrane Domain Interactions. Biochemistry. 2015;54:5125–5135. doi: 10.1021/acs.biochem.5b00495. published online EpubAug 25 (10.1021/acs.biochem.5b00495) [DOI] [PubMed] [Google Scholar]
  • 13.Chen J, Kovacs JM, Peng H, Rits-Volloch S, Lu J, Park D, Zablowsky E, Seaman MS, Chen B. HIV-1 ENVELOPE. Effect of the cytoplasmic domain on antigenic characteristics of HIV-1 envelope glycoprotein. Science. 2015;349:191–195. doi: 10.1126/science.aaa9804. published online EpubJul 10 (10.1126/science.aaa9804) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gao F, Morrison SG, Robertson DL, Thornton CL, Craig S, Karlsson G, Sodroski J, Morgado M, Galvao-Castro B, von Briesen H, et al. Molecular cloning and analysis of functional envelope genes from human immunodeficiency virus type 1 sequence subtypes A through G. The WHO and NIAID Networks for HIV Isolation and Characterization. J Virol. 1996;70:1651–1667. doi: 10.1128/jvi.70.3.1651-1667.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rowell JF, Stanhope PE, Siliciano RF. Endocytosis of endogenously synthesized HIV-1 envelope protein. Mechanism and role in processing for association with class II MHC. J Immunol. 1995;155:473–488. [PubMed] [Google Scholar]
  • 16.Boge M, Wyss S, Bonifacino JS, Thali M. A membrane-proximal tyrosine-based signal mediates internalization of the HIV-1 envelope glycoprotein via interaction with the AP-2 clathrin adaptor. J Biol Chem. 1998;273:15773–15778. doi: 10.1074/jbc.273.25.15773. [DOI] [PubMed] [Google Scholar]
  • 17.Call ME, Schnell JR, Xu C, Lutz RA, Chou JJ, Wucherpfennig KW. The structure of the zetazeta transmembrane dimer reveals features essential for its assembly with the T cell receptor. Cell. 2006;127:355–368. doi: 10.1016/j.cell.2006.08.044. published online EpubOct 20 (10.1016/j.cell.2006.08.044) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Call ME, Wucherpfennig KW, Chou JJ. The structural basis for intramembrane assembly of an activating immunoreceptor complex. Nat Immunol. 2010;11:1023–1029. doi: 10.1038/ni.1943. published online EpubNov (10.1038/ni.1943) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.To produce the protein, we expressed the His-tagged TrpLE-gp41HIV1D(677-716) fusion protein in E. coli as inclusion bodies, purified the solubilized protein by Ni-affinity chromatography, removed the TrpLE tag with cyanogen bromide, and separated the product by reverse phase HPLC. Bicelles were made of 1,2-Dimyristoyl-sn-Glycero-3-Phosphocholine (DMPC; lipid) and 1,2-Dihexanoyl sn-Glycero-3-Phosphocholine (DHPC; detergent) at a ratio (q) of 0.5. In this report, we use gp41HIV1D(677-716) and TMD interchangeably for convenience.
  • 20.Glover KJ, Whiles JA, Wu G, Yu N, Deems R, Struppe JO, Stark RE, Komives EA, Vold RR. Structural evaluation of phospholipid bicelles for solution-state studies of membrane-associated biomolecules. Biophys J. 2001;81:2163–2171. doi: 10.1016/s0006-3495(01)75864-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sanders CR, 2nd, Schwonek JP. Characterization of magnetically orientable bilayers in mixtures of dihexanoylphosphatidylcholine and dimyristoylphosphatidylcholine by solid-state NMR. Biochemistry. 1992;31:8898–8905. doi: 10.1021/bi00152a029. [DOI] [PubMed] [Google Scholar]
  • 22.OuYang B, Xie S, Berardi MJ, Zhao X, Dev J, Yu W, Sun B, Chou JJ. Unusual architecture of the p7 channel from hepatitis C virus. Nature. 2013;498:521–525. doi: 10.1038/nature12283. published online EpubJun 27 (10.1038/nature12283) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.For structure determination, we proceeded with the clade D construct because its expression level was the highest. The approach involves determination of local structures of the monomers and assembly of the trimer with intermonomer distance restraints derived from nuclear Overhauser enhancements (NOEs) between structurally equivalent but isotopically differently labeled subunits. We could identify 8 inter-monomer NOEs using the isotopically mixed labeled sample to calculate a unique assembly solution, which was further validated and refined using other conventional NOE data.
  • 24.Gd(DOTA) is a water-soluble and membrane-inaccessible molecule, so that the paramagnetic relaxation enhancement (PRE) it generates decreases with distance from the bilayer surface.
  • 25.Zhu P, Liu J, Bess J, Jr., Chertova E, Lifson JD, Grise H, Ofek GA, Taylor KA, Roux KH. Distribution and three-dimensional structure of AIDS virus envelope spikes. Nature. 2006;441:847–852. doi: 10.1038/nature04817. published online EpubJun 15 (nature04817 [pii] 10.1038/nature04817) [DOI] [PubMed] [Google Scholar]
  • 26.Sanders RW, Derking R, Cupo A, Julien JP, Yasmeen A, de Val N, Kim HJ, Blattner C, de la Pena AT, Korzun J, Golabek M, de Los Reyes K, Ketas TJ, van Gils MJ, King CR, Wilson IA, Ward AB, Klasse PJ, Moore JP. A next-generation cleaved, soluble HIV-1 Env Trimer, BG505 SOSIP.664 gp140, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS pathogens. 2013;9:e1003618. doi: 10.1371/journal.ppat.1003618. published online EpubSep (10.1371/journal.ppat.1003618) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Julien JP, Lee JH, Cupo A, Murin CD, Derking R, Hoffenberg S, Caulfield MJ, King CR, Marozsan AJ, Klasse PJ, Sanders RW, Moore JP, Wilson IA, Ward AB. Asymmetric recognition of the HIV-1 trimer by broadly neutralizing antibody PG9. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:4351–4356. doi: 10.1073/pnas.1217537110. published online EpubMar 12 (10.1073/pnas.1217537110) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.West JT, Johnston PB, Dubay SR, Hunter E. Mutations within the putative membrane-spanning domain of the simian immunodeficiency virus transmembrane glycoprotein define the minimal requirements for fusion, incorporation, and infectivity. J Virol. 2001;75:9601–9612. doi: 10.1128/JVI.75.20.9601-9612.2001. published online EpubOct (10.1128/JVI.75.20.9601-9612.2001) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chen J, Frey G, Peng H, Rits-Volloch S, Garrity J, Seaman MS, Chen B. Mechanism of HIV-1 neutralization by antibodies targeting a membrane-proximal region of gp41. Journal of virology. 2014;88:1249–1258. doi: 10.1128/JVI.02664-13. published online EpubJan (10.1128/JVI.02664-13) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Berlioz-Torrent C, Shacklett BL, Erdtmann L, Delamarre L, Bouchaert I, Sonigo P, Dokhelar MC, Benarous R. Interactions of the cytoplasmic domains of human and simian retroviral transmembrane proteins with components of the clathrin adaptor complexes modulate intracellular and cell surface expression of envelope glycoproteins. J Virol. 1999;73:1350–1361. doi: 10.1128/jvi.73.2.1350-1361.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jeffs SA, Goriup S, Kebble B, Crane D, Bolgiano B, Sattentau Q, Jones S, Holmes H. Expression and characterisation of recombinant oligomeric envelope glycoproteins derived from primary isolates of HIV-1. Vaccine. 2004;22:1032–1046. doi: 10.1016/j.vaccine.2003.08.042. published online EpubFeb 25. [DOI] [PubMed] [Google Scholar]
  • 32.Kovacs JM, Noeldeke E, Ha HJ, Peng H, Rits-Volloch S, Harrison SC, Chen B. Stable, uncleaved HIV-1 envelope glycoprotein gp140 forms a tightly folded trimer with a native-like structure. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:18542–18547. doi: 10.1073/pnas.1422269112. published online EpubDec 30 (10.1073/pnas.1422269112) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Guenaga J, Dubrovskaya V, de Val N, Sharma SK, Carrette B, Ward AB, Wyatt RT. Structure-guided redesign increases the propensity of HIV Env to generate highly stable soluble trimers. J Virol. 2015 doi: 10.1128/JVI.02652-15. published online EpubDec 30 (10.1128/JVI.02652-15) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kwon YD, Pancera M, Acharya P, Georgiev IS, Crooks ET, Gorman J, Joyce MG, Guttman M, Ma X, Narpala S, Soto C, Terry DS, Yang Y, Zhou T, Ahlsen G, Bailer RT, Chambers M, Chuang GY, Doria-Rose NA, Druz A, Hallen MA, Harned A, Kirys T, Louder MK, O'Dell S, Ofek G, Osawa K, Prabhakaran M, Sastry M, Stewart-Jones GB, Stuckey J, Thomas PV, Tittley T, Williams C, Zhang B, Zhao H, Zhou Z, Donald BR, Lee LK, Zolla-Pazner S, Baxa U, Schon A, Freire E, Shapiro L, Lee KK, Arthos J, Munro JB, Blanchard SC, Mothes W, Binley JM, McDermott AB, Mascola JR, Kwong PD. Crystal structure, conformational fixation and entry-related interactions of mature ligand-free HIV-1 Env. Nat Struct Mol Biol. 2015;22:522–531. doi: 10.1038/nsmb.3051. published online EpubJul (10.1038/nsmb.3051) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wiener MC, White SH. Structure of a fluid dioleoylphosphatidylcholine bilayer determined by joint refinement of x-ray and neutron diffraction data. III. Complete structure. Biophys J. 1992;61:434–447. doi: 10.1016/S0006-3495(92)81849-0. published online EpubFeb (10.1016/S0006-3495(92)81849-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Delaglio F, Grzesiek S, Vuister GW, Zhu G, Pfeifer J, Bax A. NMRPipe: a multidimensional spectral processing system based on UNIX pipes. Journal of biomolecular NMR. 1995;6:277–293. doi: 10.1007/BF00197809. [DOI] [PubMed] [Google Scholar]
  • 37.Vranken WF, Boucher W, Stevens TJ, Fogh RH, Pajon A, Llinas M, Ulrich EL, Markley JL, Ionides J, Laue ED. The CCPN data model for NMR spectroscopy: development of a software pipeline. Proteins. 2005;59:687–696. doi: 10.1002/prot.20449. published online EpubJun 1 (10.1002/prot.20449) [DOI] [PubMed] [Google Scholar]
  • 38.Bartels C, Xia TH, Billeter M, Guntert P, Wuthrich K. The program XEASY for computer-supported NMR spectral analysis of biological macromolecules. Journal of biomolecular NMR. 1995;6:1–10. doi: 10.1007/BF00417486. published online EpubJul (10.1007/BF00417486) [DOI] [PubMed] [Google Scholar]
  • 39.Salzmann M, Wider G, Pervushin K, Wuthrich K. Improved sensitivity and coherence selection for [15N,1H]-TROSY elements in triple resonance experiments. Journal of biomolecular NMR. 1999;15:181–184. doi: 10.1023/a:1008358030477. [DOI] [PubMed] [Google Scholar]
  • 40.Schwieters CD, Kuszewski JJ, Tjandra N, Clore GM. The Xplor-NIH NMR molecular structure determination package. Journal of magnetic resonance. 2003;160:65–73. doi: 10.1016/s1090-7807(02)00014-9. [DOI] [PubMed] [Google Scholar]
  • 41.Shen Y, Delaglio F, Cornilescu G, Bax A. TALOS+: a hybrid method for predicting protein backbone torsion angles from NMR chemical shifts. Journal of biomolecular NMR. 2009;44:213–223. doi: 10.1007/s10858-009-9333-z. published online EpubAug (10.1007/s10858-009-9333-z) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kovacs JM, Nkolola JP, Peng H, Cheung A, Perry J, Miller CA, Seaman MS, Barouch DH, Chen B. HIV-1 envelope trimer elicits more potent neutralizing antibody responses than monomeric gp120. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:12111–12116. doi: 10.1073/pnas.1204533109. published online EpubJul 24 (10.1073/pnas.1204533109) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Li M, Gao F, Mascola JR, Stamatatos L, Polonis VR, Koutsoukos M, Voss G, Goepfert P, Gilbert P, Greene KM, Bilska M, Kothe DL, Salazar-Gonzalez JF, Wei X, Decker JM, Hahn BH, Montefiori DC. Human immunodeficiency virus type 1 env clones from acute and early subtype B infections for standardized assessments of vaccine-elicited neutralizing antibodies. J Virol. 2005;79:10108–10125. doi: 10.1128/JVI.79.16.10108-10125.2005. published online EpubAug. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Holland AU, Munk C, Lucero GR, Nguyen LD, Landau NR. Alpha-complementation assay for HIV envelope glycoprotein-mediated fusion. Virology. 2004;319:343–352. doi: 10.1016/j.virol.2003.11.012. published online EpubFeb 20 (10.1016/j.virol.2003.11.012) [DOI] [PubMed] [Google Scholar]
  • 45.Sarzotti-Kelsoe M, Bailer RT, Turk E, Lin CL, Bilska M, Greene KM, Gao H, Todd CA, Ozaki DA, Seaman MS, Mascola JR, Montefiori DC. Optimization and validation of the TZM-bl assay for standardized assessments of neutralizing antibodies against HIV-1. Journal of immunological methods. 2014;409:131–146. doi: 10.1016/j.jim.2013.11.022. published online EpubJul (10.1016/j.jim.2013.11.022) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Scheid JF, Mouquet H, Ueberheide B, Diskin R, Klein F, Oliveira TY, Pietzsch J, Fenyo D, Abadir A, Velinzon K, Hurley A, Myung S, Boulad F, Poignard P, Burton DR, Pereyra F, Ho DD, Walker BD, Seaman MS, Bjorkman PJ, Chait BT, Nussenzweig MC. Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science. 2011;333:1633–1637. doi: 10.1126/science.1207227. published online EpubSep 16 (10.1126/science.1207227) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, Mitcham JL, Lehrman JK, Priddy FH, Olsen OA, Frey SM, Hammond PW, Kaminsky S, Zamb T, Moyle M, Koff WC, Poignard P, Burton DR. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science. 2009;326:285–289. doi: 10.1126/science.1178746. published online EpubOct 9 (1178746 [pii] 10.1126/science.1178746) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R, Julien JP, Wang SK, Ramos A, Chan-Hui PY, Moyle M, Mitcham JL, Hammond PW, Olsen OA, Phung P, Fling S, Wong CH, Phogat S, Wrin T, Simek MD, Koff WC, Wilson IA, Burton DR, Poignard P. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature. 2011;477:466–470. doi: 10.1038/nature10373. published online EpubSep 22 (10.1038/nature10373) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Mouquet H, Scharf L, Euler Z, Liu Y, Eden C, Scheid JF, Halper-Stromberg A, Gnanapragasam PN, Spencer DI, Seaman MS, Schuitemaker H, Feizi T, Nussenzweig MC, Bjorkman PJ. Complex-type N-glycan recognition by potent broadly neutralizing HIV antibodies. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:E3268–3277. doi: 10.1073/pnas.1217207109. published online EpubNov 20 (10.1073/pnas.1217207109) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Thali M, Moore JP, Furman C, Charles M, Ho DD, Robinson J, Sodroski J. Characterization of conserved human immunodeficiency virus type 1 gp120 neutralization epitopes exposed upon gp120-CD4 binding. J Virol. 1993;67:3978–3988. doi: 10.1128/jvi.67.7.3978-3988.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Swetnam J, Shmelkov E, Zolla-Pazner S, Cardozo T. Comparative magnitude of cross-strain conservation of HIV variable loop neutralization epitopes. PLoS ONE. 2010;5(e15994) doi: 10.1371/journal.pone.0015994. 10.1371/journal.pone.0015994) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Huang J, Ofek G, Laub L, Louder MK, Doria-Rose NA, Longo NS, Imamichi H, Bailer RT, Chakrabarti B, Sharma SK, Alam SM, Wang T, Yang Y, Zhang B, Migueles SA, Wyatt R, Haynes BF, Kwong PD, Mascola JR, Connors M. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature. 2012;491:406–412. doi: 10.1038/nature11544. published online EpubNov 15 (10.1038/nature11544) [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

SM

RESOURCES