HIV-1 Nef is a multifunctional protein that optimizes virus spread and promotes immune evasion of infected cells to accelerate disease progression in AIDS patients. As one of its activities, Nef reduces the motility of infected CD4+ T lymphocytes in confined space. In vivo, Nef restricts T lymphocyte homing to lymph nodes as it reduces the ability for extravasation at the diapedesis step. Effects of Nef on T lymphocyte motility are typically mediated by its ability to reduce actin remodeling. However, interference with diapedesis does not depend on residues in Nef required for inhibition of host cell actin dynamics. In search for an alternative mechanism by which Nef could alter T lymphocyte extravasation, we noted that the viral protein interferes with the polarization of primary human CD4+ T lymphocytes upon infection with HIV-1. Expression of Nef alone is sufficient to disrupt T cell polarization, and this effect is conserved among lentiviral Nef proteins. Nef acts by arresting the oscillation of CD4+ T cells between polarized and nonpolarized morphologies. Mapping studies identified the binding site for the Nef-associated kinase complex (NAKC) as critical determinant of this Nef activity and a NAKC-binding–deficient Nef variant fails to impair CD4+ T lymphocyte extravasation and homing to lymph nodes. These results thus imply the disruption of T lymphocyte polarity via its NAKC binding site as a novel mechanism by which lentiviral Nef proteins alter T lymphocyte migration in vivo.

Negative factor (Nef) is a 25–34-kDa myristoylated accessory protein encoded by the primate lentiviruses HIV-1, HIV-2, and SIV. Although Nef is not needed for virus replication in cell culture, it optimizes virus replication in the infected host and thus significantly contributes to disease progression to AIDS. To mediate this critical function in AIDS pathogenesis, Nef acts as versatile protein interaction adaptor that manipulates a remarkable range of host cell processes, including signal transduction and vesicular transport pathways, by genetically separable but incompletely defined molecular mechanisms (15). Nef mediates its functions via various interactions with host cell proteins, thereby inducing changes in central intracellular transport and signaling pathways of HIV-infected cells (2, 6). This includes reducing cell surface densities of transmembrane receptors and peripheral membrane proteins by molecular mechanisms that affect endocytosis, anterograde transport, and/or protein stability (712). By modulating surface exposure of cell-surface receptors such as MHC class I and II, CD4, chemokine receptors, costimulatory molecules such as CD80 and CD86, tetraspanins, and NK cell ligands, Nef acts to evade host cell immune responses and prevents superinfection of infected cells (8, 1017). In addition, Nef affects activation state and responsiveness of T lymphocytes to TCR signaling by modifying their vesicular transport and actin remodeling pathways (1826). These alterations reduce activation-induced cell death and thus prolong the survival of productively infected cells (27). Finally, Nef enhances the infectivity of HIV particles by antagonizing the restriction factors SERINC 3 and 5 via yet to be determined mechanisms (2831).

In addition to activities that are directly linked to the replicative abilities of the virus, HIV-1 Nef was also shown to interfere with the motility of infected CD4+ T lymphocytes (3234), which, together with macrophages, constitute the main target cells of the virus. We previously identified that the mechanism by which Nef affects T lymphocyte motility involves its ability to inhibit dynamic remodeling of host cell actin. At least in part, this reduction of actin remodeling results from the association of Nef with cellular PAK2 kinase that induces phosphorylation, and thus inactivation, of the actin-severing factor cofilin (35). Subsequent work suggested that this block to T lymphocyte motility in vitro translates into a potent block to lymph node homing of T lymphocytes when Nef-expressing CD4+ T lymphocytes were adoptively transferred into recipient mice (36, 37). The multistep homing process of T lymphocytes via extravasation through high endothelial venules initiates with the selectin-mediated trapping and rolling with the direction of blood flow subsequent engagement of CCR7 by its endothelium-presented chemokine ligands CCL21 and CCL19 result in integrin activation and firm arrest of T lymphocytes on the inner vessel wall. Following acquisition of a polarized phenotype and an optional crawling step (irrespective of the direction of blood flow) to find permissive sites for diapedesis, T lymphocytes enter the lymph node parenchyma and resume migration (38, 39). Three-dimensional visualization and intravital microscopy demonstrated that the Nef-mediated block in lymph node homing was primarily due to a strong inhibition of extravasation, whereas subsequent parenchymal motility was only slightly reduced. Consistently, ex vivo analyses of T lymphocyte extravasation across an endothelial cell monolayer under physiological shear stress revealed that Nef potently disrupts T lymphocyte polarization and interfered specifically with the diapedesis step. The use of an Nef mutant (F195A) that is specifically deficient in PAK2 association, and thus inhibiting actin remodeling, but exerts all other known Nef activities (37, 4043) demonstrated that Nef affects extravasation via a dual mechanism: although inhibition of subendothelial migration appears to be achieved via the known deregulation of actin dynamics, this mechanism was dispensable for the effects of Nef on T cell polarization during diapedesis (37). Because it remains unknown how the viral protein induces these effects, the goal of this study was to gain insight into the mechanisms and relevance of Nef-mediated disruption of T cell polarization and diapedesis.

The plasmids encoding GFP fusion proteins of HIV-1SF2 Nef wild type (WT) and mutants thereof as well as the panel of expression constructs encoding for GFP or YFP fusion proteins of HIV-1, HIV-2, and SIV Nef alleles were described previously (10, 22, 27, 35, 4447). The proviral constructs used are based on HIV-1NL4–3 and either lack Nef expression (HIV-1 ∆Nef), or encode for WT Nef from HIV-1SF2 (HIV-1 WT) or mutants thereof as described previously (35, 44). Proviral plasmids pHIV-1NL4–3 WT, pHIV-1NL4–3 Vpustop (ΔVpu), pHIV-1NL4–3 Nefstop (ΔNef), and pHIV-1NL4–3 Vpustop Nefstop (ΔNefΔVpu) were kindly provided by F. Kirchhoff. The expression plasmids for pSTITCH-GFP, pHit60 (murine leukemia virus [MLV]–Gag/Pol) as well as for ecotropic (pHit123), and amphotropic MLV-Env were kindly provided by R. Debets (Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands). As described (37), pSTITCH-GFP was used to generate pSTITCH-IRESΔNGFR, pSTITCH-NefSF2WT IRESΔNGFR, and pSTITCH-NefSF2F195A IRES∆NGFR, which express ∆NGFR at the cell surface for cell sorting. pSTITCH-NefSF2∆12–39 IRES∆NGFR was generated accordingly by inserting the respective nef gene.

The following Abs were used: rat anti-CD44 (clone IM7; BioLegend), rabbit anti–HIV-1 p24 capsid [p24CA; kindly provided by H.-G. Kräusslich (44)], mouse anti-transferrin receptor (clone H68.4; Life Technologies), and rat anti-GFP (clone 3H9; ChromoTek). Secondary fluorophore-coupled Abs (Alexa Fluor) were purchased from Invitrogen. Fibronectin was purchased from Sigma-Aldrich. CellTracker Blue (CMAC), CellTracker Orange (CMTMR), and CellTracker Deep Red were purchased from Thermo Fisher Scientific, CFSE from Molecular Probes, eFluor 670 from eBioscience, and PKH26 membrane dye and Mowiol 4-88 from Sigma-Aldrich.

Human PBMCs from healthy donors were obtained from the Heidelberg University Hospital Blood Bank and purified by Ficoll gradient centrifugation as described (24). Primary human T lymphocytes and A3.01 T cells were cultivated in RPMI 1640 medium plus GlutaMAX-I, supplemented with 10% FCS, 1% l-glutamine, and 1% penicillin/streptomycin (all from Invitrogen). Primary murine T lymphocytes were isolated from spleen, peripheral, and mesenteric lymph nodes of donor mice and kept in complete mouse medium (CMM) as described previously (37). 293T cells were cultivated in DMEM with 10% FCS and 1% penicillin/streptomycin (all from Invitrogen). For transient transfection, 1 × 107 A3.01 T cells were electroporated using 18 μg of plasmid DNA (950 μF, 350 V, Gene Pulser Xcell; Bio-Rad).

The 107 A3.01 cells were electroporated with the indicated Nef.GFP expression plasmids as described above. Twenty-four hours later, transfection efficiency was determined by flow cytometry (BD FACSVerse), and numbers of transfected cells, as well as total cell numbers, were adjusted by addition of untransfected cells. Cells were pelleted and directly lysed in 300 μl of 2×SDS sample buffer. Forty microliters of the cell lysate were loaded per sample on a 14% SDS-PAGE. Membranes were probed against GFP to detect expression of Nef.GFP fusion constructs and transferrin receptor as an internal loading control.

Virus stocks were generated by transfection (jetPEI, PEQLAB - Life Science) of proviral HIV-1 plasmid DNA into 293T cells, which were seeded on 15-cm culture dishes 1 d before transfection (6 × 106 cells/dish). Twenty five micrograms of proviral DNA together with 75 μl of jetPEI (1 mg/ml) was used. Supernatants were harvested 2 and 3 d after transfection and filtered using 0.45-μm pore size filters. Typically, virus was subjected to concentration by ultracentrifugation of the cleared supernatant through a 20% sucrose cushion (24,000 rpm, 1.5–2 h). Virus titers were evaluated by p24 ELISA or SYBR Green I-based PERT (SG-PERT) assay, and infectivity was determined by Blue Cell Assay as described before (29, 44, 48, 49).

For infection, primary human lymphocytes were activated for 72 h with 2 μg/ml PHA and 10 ng/ml IL-2 and spin-infected with virus in the presence of 4 μg/ml polybrene (multiplicity of infection between 1 and 3) in 96-well plates with a total volume of 100 μl/well. The plates were then centrifuged at 2000 rpm for 90 min at 37°C and incubated 4–6 h at 37°C and 5% CO2 prior to medium change. Cells were cultured for 72 h in supplemented RPMI 1640 containing 10 ng/ml IL-2 before further analyses.

Six hours posttransfection or 72 h postinfection, 3–5 × 105 A3.01 cells or monocyte-depleted, standard activated PBMCs were analyzed for polarization. To exclude dead cells from the analysis, they were stained with CMAC CellTracker dye (1 μM for 15 min at 37°C) and discriminated from viable cells by the characteristic loss of the dye. Cells were seeded in 100 μl of supplemented RPMI 1640 onto fibronectin-coated cover glasses (30 μg/ml, 1 h at room temperature) and placed in the incubator for 2 h to allow polarization, followed by fixation with 3% paraformaldehyde in PBS (w/v) and incubated for another 15–30 min at room temperature. Cells were then permeabilized with 0.1% Triton X-100 for 2 min and blocked in 1% BSA in PBS (w/v) for 30 min at room temperature. Following washing with 1× PBS, rat anti-CD44 Ab (1:800, in 1% BSA in PBS; BioLegend) was added and incubated for 2 h at room temperature. Cover glasses were washed three times with 1× PBS, and incubation with the secondary goat anti-mouse Ab coupled to Alexa 568 (1:2000, in 1× PBS; Invitrogen) was carried out for 1 h at room temperature protected from light. After washing with 1× PBS, cover glasses were mounted using Mowiol, dried for 2 h at room temperature protected from light, and stored at 4°C until analyzed by microscopy. Polarization of cells was typically quantified based on overall cell morphology [polarized cells have a polarization index (cell length/cell width) ≥2 (37)]. Analysis based on the enrichment of the uropod marker CD44 at one pole of the cell gave similar results (see Supplemental Fig. 1), and CD44 staining was included in most experiments as quality control.

C57BL/6 (JANVIER LABS) and LifeAct-GFP (50) mice were used in this study, and all the experiments were carried out in accordance with the standards approved by central animal facilities of the Universities of Heidelberg (G252/14 and T45/14) and Bern. Mice were used for the experiments at 5–10 wk of age.

Effects of Nef on T lymphocyte homing to lymph nodes in mice were investigated by adoptive transfer of mouse T cells following transduction and sorting as described previously (37). Briefly, for virus production, 293T cells were transfected using jetPEI transfection reagent (PEQLAB - Life Science) according to manufacturer’s instructions with 20 μg pSTITCH construct, 20 μg pHit60, 5 μg pHit123, and 5 μg amphotropic MLV-Env. Viral supernatant was collected after 48 h, filtered (0.45-μm pore size filter; Roth), and used immediately for transduction of murine T cells.

Single-cell suspensions were obtained from spleen and peripheral lymph node by mechanical disruption (70-μm cell strainer; Falcon) and cultured for 24 h in CMM (RPMI 1640 supplemented with 10% FCS, 100 U/ml penicillin, 100 μg/ml streptomycin, 2 mM glutamine, 50 μM 2-ME, 0.1 mM nonessential amino acids, and 0.1 mM sodium pyruvate [all from Life Technologies, Invitrogen]) in the presence of 2 μg/ml Con A (Sigma-Aldrich) and 10 ng/ml IL-2 (Biomol). Activated T lymphocytes (2 × 106 cells/well) were spin-infected at 2300 rpm for 90 min at 37°C in the presence of viral supernatant in 24-well plate precoated with 16 μg/ml RetroNectin (TaKaRa Bio, MoBiTec), cultured with viral supernatant for 3–5 h at 37°C 5% CO2, followed by replacing supernatant with CMM supplemented with 10 ng/ml IL-2. One day later, transduced cells (NGFR-positive) were magnetically sorted (STEMCELL Technologies) and labeled with either CFSE or eFluor 670 (5 μM for 20 min at 37°C). Control T cells (pSTITCH-IRES∆NGFR) were mixed with Nef WT (pSTITCH-NefSF2WT IRES∆NGFR) or Nef mutant–expressing T cells in 1:1 ratio, and a total of 5–10 × 106 cells were adoptively transferred into 5–8-wk-old C57BL/6 recipient mice by i.v. injection. Four and twenty-four hours after cell transfer, spleen, mesenteric, and peripheral lymph nodes were harvested separately and analyzed by flow cytometry (FACSVerse; BD Bioscience). Homing efficiency was calculated relative to mixed input cells and normalized to cotransferred control cells.

Transendothelial migration or extravasation assays were essentially performed as described previously (37). Isolation and culture of primary mouse brain microvascular endothelial cells (pMBMECs) from LifeAct-GFP mice was performed exactly as described before (51, 52). Cytokine stimulation of pMBMECs was done for 16–20 h with 10 ng/ml TNF-α. All experiments were performed in migration assay medium (DMEM, 5% calf serum, and 25 mM HEPES) at 37°C. For live imaging, a parallel flow chamber connected to an automated syringe pump (Harvard Apparatus) was mounted on TNF-α–stimulated pMBMECs previously overlaid with 1 μM CCL21 and placed on the heating stage of an inverted microscope (Axio Observer Z1; ZEISS) (53).

Primary murine T lymphocytes were activated, transduced, and NGFR-sorted as described above, and 1 × 107 cells/ml were labeled with CellTracker dyes (CMTMR, 5 μM; CMAC, 20 μM; Deep Red, 0.5 μM; for 20 min at 37°C). Dyes were exchanged between samples for successive replicates to exclude adverse effects of the dyes. The three differentially labeled cell populations were mixed 1:1:1 and allowed to accumulate at low shear stress (0.1 dyne/cm2) for 4 min, then shear stress was increased to physiological strength (1.5 dyne/cm2), and T lymphocytes were recorded over 20 min using a 10× objective with phase contrast and fluorescent illumination. Time-lapse videos were created from one frame every 10 s.

Adherence to the endothelium was calculated based on the ratio of input cells, precisely defined by flow cytometry (FACSCalibur; BD Biosciences), relative to the ratio of cells adhered to the endothelium immediately after the increase of shear stress. Cells were tracked and phenotypes counted manually using a customized ImageJ Annotation Plugin developed by J. -P. Bergeest (Karl Rohr Laboratory, Heidelberg), which combines the ability of the Cell Counter plugin and the Manual Tracking plugin. We differentiated the following: 1) cells crawling on top of the endothelium, 2) diapedesis, 3) migration under the endothelium, 4) cells that detached, and 5) cells crawling out of view. Exported tracks were filtered according to the phenotype annotations using Microsoft Excel to calculate relative frequencies of phenotypes. Percentage of cells staying on top of the endothelium were calculated by dividing the number of cells defined as action 1, which do not show other behaviors during the course of the experiment (cells that migrate out of view on top of the endothelium were not taken into account), by the total number of tracked cells ([no. 1 − no. 2 − no. 4 − no. 5] / total no. tracks). Percentages of cells that were washed off and that undergo diapedesis were calculated by dividing the number of tracks with actions 4 and 2 (cells that migrate out of view afterward are also counted) by the total number of tracked cells, respectively. Polarization index of cells migrating on top of the endothelium was calculated by dividing the cell length by the cell width, measured manually with Fiji (37).

Cell polarization was assessed using an epifluorescence microscope (IX81; Olympus). Cells were considered polarized when cell length exceeded cell width by at least a factor of 2. At least 100 transfected cells or 30 infected cells were evaluated per experiment and condition. Polarization of an equal number of untransfected or uninfected neighboring cells on the same coverslip was also determined, and values of transfected/infected cells were normalized to this internal control.

For time-lapse microscopy videos, A3.01 cells were transfected and 6 h later, stained with the PKH26 membrane dye and seeded on fibronectin-coated, eight-well chambered cover glasses (Lab-Tek). Live microscopy was performed with a Nikon Ti PerkinElmer UltraVIEW VoX Spinning Disc Confocal Microscope equipped with a perfect focus system, a 60× oil numerical aperture = 1.49 objective, Hamamatsu ORCA Flash4.0 scientific complementary metal-oxide-semiconductor camera, and an environmental control chamber (37°C, 5% CO2). Each frame contained a Z-stack with 1 μm of spacing. Acquisition of approximately 1 frame/min resulted in time-lapse videos of ∼1 h length. For representation of change in cell morphology over time, all single frames of one video were color coded and projected into one image.

Morphological dynamics and modes of deformation have been analyzed from binarized time-lapse videos according to previous power spectrum analyses assessing the energy consumption by shape change of mononuclear cells (5456) and anuclear cells (57, 58). The peripheral edge of the cells was defined in each single frame and plotted in polar coordinates with the center of mass being the origin and r the radial distance to the edge of the cell. Morphological dynamics of each target cell were analyzed through the radial distance r as a function of angle θ = 0 − 360° and time t. The deformation was defined as the normalized radial distance: R(θ,t)=r(θ,t)r(θ,t)θ. The characteristic spatiotemporal patterns were identified by calculating the autocorrelation function (ACF) Γ of R(θ,t):

ΓRR(Δθ,Δt)=R(θ+Δθ,t+Δt)R(θ,t)[R(θ,t)]2.

The deformation of cells involves active, energy-consuming processes such as the bending of membranes, the rearrangements of lipids and proteins, and, most importantly, the remodeling of cytoskeletal structures. To assess the energy consumed by these processes, the power spectral density Γ^m was calculated via spatial frequency domain analysis. In this equation, the deformation R(θ,t) was expanded in a Fourier series over spatial modes m to calculate the power spectrum Γ^m of the morphological dynamics (54, 56):

Γ^m=FTm(R(θ,t))FTm(R(θ,t))¯t.

Note that isotropic expansion/contraction (m = 0) and translational motion (m = 1) were excluded from the deformation analysis because of the normalization to r(θ,t)θ and the center of mass being the origin in the inertial frame. Thus, Γ^m is presented in units of min−1, as a dimensionless average over time. Because the power spectral density is proportional to the mechanical power dissipated by morphological dynamics (59), this approach allows the comparative analysis of cell deformation under different conditions. With the spectrum corresponding to the energy dissipation broken down by each mode of deformation, power spectrum analysis provides quantitative information about the predominant mode of deformation.

A3.01 lymphocytes were either transfected or transduced to express GFP and NefSF2.GFP. For transfection, A3.01 cells were electroporated (250 V, 950 μF) with GFP/NefSF2.GFP expression plasmids and pDisplay-YFP (60) in a 1:6 ratio. On the next day, cells were MACS sorted using GFP microBeads (Miltenyi Biotec), cultured overnight, and subjected to Seahorse measurement using Seahorse XFe96 FluxPaks (inc. mini) and Seahorse XF Cell Mito Stress Test Kit (Agilent Technologies). For transduction, A3.01 cells were spin-inoculated for 90 min in the presence of polybrene with sucrose cushion–concentrated, vesicular stomatitis virus glycoprotein G–pseudotyped lentiviral particles produced in 293T in saturating amounts [see (61) for details]. Transduced cells were directly subjected to Seahorse measurement 48 h after transduction. Transduction/transfection efficiency at the time of the Seahorse measurement was analyzed by FACS and ranged between 30–60%. Ninety-six–well plates for Seahorse measurement were coated with fibronectin (12 μg/ml in PBS equals the same final concentration as on the cover glasses) and air dried overnight. 7.5 × 105 transduced/transfected cells were seeded per well in Seahorse medium supplemented with 10 mM glucose and 2 mM l-glutamine and measured in a Seahorse XFe96 Analyzer (Agilent Technologies) using the standard Agilent Seahorse Instrument Protocol settings. Total duration of measurement was ∼140 min. After three successive measurements of baseline oxygen consumption rate (OCR; basal respiration) and extracellular acidification rate (ECAR; glycolysis), 2 μM of oligomycin (inhibits ATP synthase/complex IV) were injected (block of mitochondrial respiration) and measured three times, followed by injection of 1.5 μM carbonyl cyanide-4 (trifluoromethoxy) phenylhydrazone (FCCP; uncoupling agent that collapses the proton gradient and disrupts the mitochondrial membrane potential, resulting in maximal oxygen consumption, which equals maximal respiration). After three measurement steps, 1.5 μM of rotenone were injected (complex I inhibitor, shuts down mitochondrial respiration, which equals nonmitochondrial respiration), followed by six measurement steps. Four to eight replicate wells were analyzed per experiment.

Data analysis and statistical calculations were performed using Microsoft Excel and GraphPad Prism. Image editing was carried out using Adobe Photoshop and Illustrator CS4 or CS6. Video editing was performed with Fiji. Statistical significance was calculated by Mann–Whitney U test analysis (***p < 0.001, **p < 0.01, *p < 0.05).

Our previous results had identified that isolated expression of HIV-1 Nef impairs the polarization of mouse CD4+ T cells during the extravasation process (37). To test the potential relevance of this observation for HIV infection, we infected activated primary human CD4+ T lymphocytes with HIV-1 WT (HIV-1NL4.3 carrying the nef gene of HIV-1SF2) (44) or its counterpart lacking expression of a functional Nef protein (HIV-1 ΔNef) and allowed them to polarize on fibronectin-coated surfaces (Fig. 1A, 1B). Dead cells were excluded based on their inability to retain the cell viability dye CMAC (Fig. 1A, bottom panel). Productively infected cells were identified by immunostaining against p24CA, and the polarization state of cells was typically assessed by the overall cell morphology. Cells were considered polarized when formation of CD44-rich uropod and leading edge structures were apparent in elongated cells (polarized cells have a polarization index [cell length/cell width] ≥2; Fig. 1A, middle panel), whereas cells of round shape (Fig. 1A, top panel) were considered to be not polarized. Because the overall percentage of uninfected polarized cells slightly varied between donors but also between individual samples of cells from the same donor (typically ranging between 40 and 60% of cells), the percentage of polarized cells positive for p24CA was determined relative to that of uninfected neighboring cells (Fig. 1B). Polarization of cells productively infected with HIV-1 WT was markedly reduced for all donors analyzed with differences in the magnitude of this effect between cells from individual donors (0–60% of uninfected cells) (Fig. 1B, 1D). Although scoring slightly divergent overall amounts of polarized cells, quantification of cell polarization based on by the distribution of the uropod marker CD44 confirmed this pronounced interference of HIV-1 infection with CD4 T cell polarization (Supplemental Fig. 1). Notably, disruption of T cell polarization was markedly less pronounced for cells infected with HIV-1 ΔNef (Fig. 1B, 1D). Depending on the donor (D), the lack of Nef in cells infected with HIV-1 ΔNef resulted in partially (e.g., D1, D2, D5, D6, and D7) or fully restored polarization rates (D3 and D4) compared with uninfected control cells. These results identify Nef as a key viral determinant for disruption of polarization of HIV-1–infected CD4+ T lymphocytes but also suggest that additional viral parameters that can affect cell polarization may exist. Because Nef and Vpu share a number of biological activities (15, 16, 62), we tested the involvement of Vpu in polarization disruption (Fig. 1C, 1D). In these experiments, cell polarization was unaffected by the presence of Vpu, and a lack of Nef expression was associated with polarization frequencies that were virtually indistinguishable from uninfected cells. We conclude that Nef is the central viral determinant governing the disruption of CD4+ T cell polarization in the context of HIV-1 infection.

FIGURE 1.

HIV-1 Nef impairs polarity of infected primary human CD4+ T lymphocytes. Human PBMCs were activated for 72 h with 2 μg/ml PHA and 10 ng/ml IL-2 and spin-infected with HIV-1 in the presence of 4 μg/ml polybrene. After 72 h, cells were seeded on fibronectin-coated cover glasses, incubated for 2 h at 37°C, and fixed. Cells were stained for CD44 and HIV-1 p24CA, as polarity and infection markers, respectively. For donors 5 to 10, CellTracker Blue CMAC viability dye was used to discriminate dead cells. (A) Representative epifluorescence micrographs of the studied morphotypes of PBMCs infected with WT and ΔNef versions of HIV-1NL4.3 SF2 Nef: not polarized (upper row) or polarized (middle row). Dead cells (lower row) were not considered. (B) Histogram of morphological (polarization index ≥2) polarization values observed for HIV-1 WT-infected (black) and ΔNef-infected (gray) cells, calculated relative to the polarization frequency of uninfected control cells on the same cover glass. At least 30 cells were counted per condition. (C) Representative epifluorescence micrographs of the studied morphotypes of PBMCs infected with HIV-1NL4.3 SF2 Nef and its ΔNef, ΔVpu, and ΔNefΔVpu derivatives. (D) Morphological polarity quantification. Bars indicate polarization levels of infected cells relative to the polarization levels of uninfected cells from the same cover glasses. One hundred cells were counted per condition. Scale bars, 5 μm.

FIGURE 1.

HIV-1 Nef impairs polarity of infected primary human CD4+ T lymphocytes. Human PBMCs were activated for 72 h with 2 μg/ml PHA and 10 ng/ml IL-2 and spin-infected with HIV-1 in the presence of 4 μg/ml polybrene. After 72 h, cells were seeded on fibronectin-coated cover glasses, incubated for 2 h at 37°C, and fixed. Cells were stained for CD44 and HIV-1 p24CA, as polarity and infection markers, respectively. For donors 5 to 10, CellTracker Blue CMAC viability dye was used to discriminate dead cells. (A) Representative epifluorescence micrographs of the studied morphotypes of PBMCs infected with WT and ΔNef versions of HIV-1NL4.3 SF2 Nef: not polarized (upper row) or polarized (middle row). Dead cells (lower row) were not considered. (B) Histogram of morphological (polarization index ≥2) polarization values observed for HIV-1 WT-infected (black) and ΔNef-infected (gray) cells, calculated relative to the polarization frequency of uninfected control cells on the same cover glass. At least 30 cells were counted per condition. (C) Representative epifluorescence micrographs of the studied morphotypes of PBMCs infected with HIV-1NL4.3 SF2 Nef and its ΔNef, ΔVpu, and ΔNefΔVpu derivatives. (D) Morphological polarity quantification. Bars indicate polarization levels of infected cells relative to the polarization levels of uninfected cells from the same cover glasses. One hundred cells were counted per condition. Scale bars, 5 μm.

Close modal

We next tested if expression of Nef alone is sufficient to disrupt CD4+ T cell polarization and transiently expressed GFP or an Nef.GFP fusion proteins that was previously characterized as functional equivalent of nonfusion Nef (22, 35, 63, 64) in A3.01 T cells (Fig. 2). Although ∼50% of GFP-expressing control cells polarized, polarization efficiency was markedly reduced for cells expressing HIV-1SF2 Nef.GFP (Fig. 2A, 2B). This experimental system allowed us to screen a panel of HIV-1, HIV-2, and SIV Nef.YFP proteins for their ability to disrupt T cell polarization. When segregated into Nef proteins with the ability to downregulate cell-surface CD3 (group 2) versus Nef proteins that lack this activity (group 1) (27, 46), T cell polarity disruption was pronounced and well conserved among group 2 Nef proteins. Most group 1 Nef proteins also significantly reduced T cell polarization, albeit with slightly lower efficacy (Fig. 2C). Nef from HIV-1 YBF30, which is also deficient in rerouting the TCR proximal kinase Lck and SIVcpz Tan 3, which is defective in most Nef activities, including TCR-induced actin remodeling (46, 65), were impaired in affecting CD4+ T cell polarization. Nef from SIVcpz Gab2 also displayed weak polarity disrupting activity but is fully active for other Nef activities (27, 46, 65). Besides these exceptions, the ability to disrupt CD4+ T cell polarization appears to be a conserved feature of functional lentiviral Nef proteins.

FIGURE 2.

Polarity disruption is an evolutionarily conserved activity of Nef proteins. A3.01 T lymphocytes were transiently transfected with expression plasmids for GFP, NefSF2.GFP, or YFP versions of various SIV, HIV-1, and HIV-2 Nef alleles. Six hours posttransfection, cells were seeded on fibronectin-coated cover glasses and incubated for 1 h at 37°C, fixed, and stained for CD44. At least 100 cells were counted for each condition. (A) Representative epifluorescence micrographs of transfected cells. Scale bar, 5 μm. (B) Histogram bars depict morphological polarization levels of transfected cells. Nef alleles were categorized into two groups: group 1, or Nef alleles able to downregulate the CD3 surface receptor, and group 2, or Nef alleles unable to downregulate CD3. Shown are arithmetic mean values and SEs from three independent experiments. (C) Batch comparison of the two groups of Nef alleles. Bars indicate mean values for each group. Statistical analysis was performed by the Mann–Whitney U test. *p < 0.05, ***p < 0.001.

FIGURE 2.

Polarity disruption is an evolutionarily conserved activity of Nef proteins. A3.01 T lymphocytes were transiently transfected with expression plasmids for GFP, NefSF2.GFP, or YFP versions of various SIV, HIV-1, and HIV-2 Nef alleles. Six hours posttransfection, cells were seeded on fibronectin-coated cover glasses and incubated for 1 h at 37°C, fixed, and stained for CD44. At least 100 cells were counted for each condition. (A) Representative epifluorescence micrographs of transfected cells. Scale bar, 5 μm. (B) Histogram bars depict morphological polarization levels of transfected cells. Nef alleles were categorized into two groups: group 1, or Nef alleles able to downregulate the CD3 surface receptor, and group 2, or Nef alleles unable to downregulate CD3. Shown are arithmetic mean values and SEs from three independent experiments. (C) Batch comparison of the two groups of Nef alleles. Bars indicate mean values for each group. Statistical analysis was performed by the Mann–Whitney U test. *p < 0.05, ***p < 0.001.

Close modal

Next, we sought to determine the molecular surfaces in Nef required for disruption of CD4+ T cell polarity. To this end, we tested a panel of HIV-1SF2 Nef.GFP proteins in which individual functional motifs are disrupted (Fig. 3A, 3B). Residues required for phosphorylation of Nef (serine 6 to alanine, S6A) (47), downregulation of cell surface MHC class I molecules (M20A) (66), or with the clathrin endocytosis machinery (LL168/169AA, ED178/179AA) (67) were dispensable for T cell polarity disruption and amino acid exchanges that reduce the ability of Nef to interact with SH3 domains (E466–69A4, VGF70–72AAA) (40, 45, 68) only moderately reduced the disruption of polarization as the activity of these mutants was not statistically significant from WT Nef. As for most Nef activities (2), myristoylation of glycine 2, and thus efficient interaction with cellular membranes, was essential for T cell polarity disruption (G2A). In line with our previous study, mutation of the PAK2 interaction motif (F195A) resulted in loss of polarity interference by Nef (37). Similarly, mutation of the SH3 domain interaction surface in Nef, which also disrupts Nef PAK2 association but also causes defects in many other Nef functions (P76xxP79 mutated to alanine, AxxA) (7, 69), impaired T cell polarization disruption by Nef. Importantly, the deletion of the interaction surface with the Nef-associated kinase complex (NAKC; Nef mutant Δ12–39) also abrogated the ability of Nef to interfere with T cell polarization. This relevance of PAK2 and NAKC interaction surfaces was also observed in the context of HIV-1 infection of activated primary human CD4+ T lymphocytes (Fig. 3C, 3D). Because Nef ∆12–39 reduces TCR-induced actin remodeling (22) and abrogates T cell chemotaxis (data not shown), the requirement for the NAKC interaction for Nef-mediated disruption of T cell polarity suggests that inhibition of actin remodeling and cell polarization involve distinct mechanisms.

FIGURE 3.

The NAKC interaction site in Nef is required for disruption of T cell polarity. A3.01 cells were transfected with expression plasmids for GFP, NefSF2.GFP, or various mutants. Six hours after transfection, cells were incubated for 1 h at 37°C on fibronectin-coated cover glasses, fixed, and stained for CD44. (A) Representative epifluorescence micrographs of transfected cells. (B) Histogram showing polarization levels of cells transfected with the indicated mutants. Bars correspond to the arithmetic mean and SE of three independent experiments. At least 100 cells were counted for each condition. Black and dark gray bars are statistically indistinguishable from Nef.GFP or GFP, respectively. (C) The NAKC interaction surface of Nef is also relevant in the context of infection. Representative epifluorescence micrographs of PBMCs infected with HIV-1NL4.3 SF2 Nef WT, the indicated Nef mutants, or Nef deleted (∆Nef). Cells were allowed to polarize on fibronectin-coated surfaces, fixed, and stained for CD44 and HIV-1 p24CA. (D) Histogram of polarization levels of cells shown in (C) relative to uninfected cells from the same cover glass. At least 100 cells were counted for each condition. Scale bars, 5 μm.

FIGURE 3.

The NAKC interaction site in Nef is required for disruption of T cell polarity. A3.01 cells were transfected with expression plasmids for GFP, NefSF2.GFP, or various mutants. Six hours after transfection, cells were incubated for 1 h at 37°C on fibronectin-coated cover glasses, fixed, and stained for CD44. (A) Representative epifluorescence micrographs of transfected cells. (B) Histogram showing polarization levels of cells transfected with the indicated mutants. Bars correspond to the arithmetic mean and SE of three independent experiments. At least 100 cells were counted for each condition. Black and dark gray bars are statistically indistinguishable from Nef.GFP or GFP, respectively. (C) The NAKC interaction surface of Nef is also relevant in the context of infection. Representative epifluorescence micrographs of PBMCs infected with HIV-1NL4.3 SF2 Nef WT, the indicated Nef mutants, or Nef deleted (∆Nef). Cells were allowed to polarize on fibronectin-coated surfaces, fixed, and stained for CD44 and HIV-1 p24CA. (D) Histogram of polarization levels of cells shown in (C) relative to uninfected cells from the same cover glass. At least 100 cells were counted for each condition. Scale bars, 5 μm.

Close modal

To gain more insight into how Nef affects T cell polarization, we recorded the morphological dynamics of A3.01 T cells transiently expressing GFP or HIV-1SF2 Nef.GFP on fibronectin-coated surfaces by live-cell spinning-disc microscopy (Fig. 4). Over the 60-min recording time, GFP-expressing control cells underwent frequent morphological changes and thus oscillated between polarized and nonpolarized states (Fig. 4A, also see Supplemental Video 1). These morphological dynamics were quantitatively assessed by the spatiotemporal analysis of active deformation (54). As presented in Fig. 4B, the peripheral edges of a GFP-expressing T lymphocyte monitored over time imply that the cell undergoes a rigorous shape deformation after receiving a polarization cue. The ACF of the GFP-expressing cell exhibits several characteristic features, as indicated in Fig. 4C: ① indicates ACF that shows two distinct minima that are separated by Δθ = 180°, suggesting an elliptic deformation; ② indicates the position of minima that shows a drift over time, corresponding to the continuous rotation of an elliptic cell; and ③ indicates the positions of minima “jump” (e.g., Δθjump = 90°) with a characteristic time interval of Δt ≈ 10 min, indicating that the direction of elliptic deformation periodically changes. In contrast, T cells expressing Nef.GFP exhibit no major shape deformation (Fig. 4D). In fact, the calculated ACF (Fig. 4E) is featureless, and the uniformly high correlation over Δt and Δθ clearly implies that cells expressing Nef.GFP adopt a round shape over time.

FIGURE 4.

Interference of Nef with T cell polarity oscillation is associated with significant reduction in energy consumption. (A) A3.01 cells were transfected with the GFP or Nef.GFP expression plasmids, stained with the PKH26 membrane dye 6 h later, and seeded on fibronectin-coated MatTek dishes for live-cell spinning-disc microscopy. Acquisition of one frame every 1 min results in time-lapse videos of ∼1 h in length. Time projection of Supplemental Video 1 shown as temporal color code. Scale bar, 5 μm. (B) Tracking of the cell periphery and (C) the corresponding autocorrelation map of a GFP-expressing T cell, showing a pronounced shape deformation. (D and E) Corresponding data set for a T cell expressing Nef.GFP, showing a drastic suppression of shape deformation. (F) Power spectra of T cells expressing GFP (black) or Nef.GFP (red). GFP-expressing cells predominantly undergo an elliptic deformation (m = 2). (G) Total power of deformation for T cells expressing GFP (black) and Nef.GFP (red). In case of Nef.GFP, the median energy consumption by active cell deformation decreased by a factor of 4.5 compared with GFP-expressing cells. Each dot represents one cell. Shown are box plots with median values and range. (H) Mitochondrial stress test of transfected and GFP microBead–sorted A3.01 cells seeded onto fibronectin-coated Seahorse microplates and measured in a Seahorse XFe analyzer to assess mitochondrial respiration. Time points of drug injection (arrows), as well as the respective parameters that are assessed by the drug treatments, are indicated. Shown are the mean OCR with SD from four to five replicate wells derived from one out of three representative experiments. Transfection efficiencies were 80 and 56% for GFP and Nef.GFP, respectively.

FIGURE 4.

Interference of Nef with T cell polarity oscillation is associated with significant reduction in energy consumption. (A) A3.01 cells were transfected with the GFP or Nef.GFP expression plasmids, stained with the PKH26 membrane dye 6 h later, and seeded on fibronectin-coated MatTek dishes for live-cell spinning-disc microscopy. Acquisition of one frame every 1 min results in time-lapse videos of ∼1 h in length. Time projection of Supplemental Video 1 shown as temporal color code. Scale bar, 5 μm. (B) Tracking of the cell periphery and (C) the corresponding autocorrelation map of a GFP-expressing T cell, showing a pronounced shape deformation. (D and E) Corresponding data set for a T cell expressing Nef.GFP, showing a drastic suppression of shape deformation. (F) Power spectra of T cells expressing GFP (black) or Nef.GFP (red). GFP-expressing cells predominantly undergo an elliptic deformation (m = 2). (G) Total power of deformation for T cells expressing GFP (black) and Nef.GFP (red). In case of Nef.GFP, the median energy consumption by active cell deformation decreased by a factor of 4.5 compared with GFP-expressing cells. Each dot represents one cell. Shown are box plots with median values and range. (H) Mitochondrial stress test of transfected and GFP microBead–sorted A3.01 cells seeded onto fibronectin-coated Seahorse microplates and measured in a Seahorse XFe analyzer to assess mitochondrial respiration. Time points of drug injection (arrows), as well as the respective parameters that are assessed by the drug treatments, are indicated. Shown are the mean OCR with SD from four to five replicate wells derived from one out of three representative experiments. Transfection efficiencies were 80 and 56% for GFP and Nef.GFP, respectively.

Close modal

In general, the deformation of cells is an active, nonequilibrium process accompanied by energy consumption (5456). To quantitatively assess energy consumption by cell deformation in individual cells, we calculated power spectra (Fig. 4F) following Eq. 2. The power spectrum of GFP-expressing cells (black) exhibits a clear peak at m = 2. The contribution of higher modes, such as m ≥ 5, is negligibly small. This confirms that polarizing, GFP-expressing cells predominantly undergo an elliptic deformation. In contrast, the power spectrum in the presence of Nef.GFP (red) is much less pronounced, suggesting Nef.GFP-expressing cells do not consume the energy required for shape deformation of GFP-expressing control cells. In fact, the calculated total power m=210Γ^ m demonstrated that the median energy dissipated through shape deformation of Nef.GFP-expressing cells is 4.5-fold lower compared with that of GFP-expressing T lymphocytes over the 1-h observation period (Fig. 4G). The disruption of dynamic morphological T lymphocyte polarization by HIV-1 Nef is, therefore, paralleled by a reduction in energy consumption at time when cell polarization would typically be initiated.

To test whether this reduced energy consumption of individual Nef.GFP-expressing cells reflects a global effect of the viral protein on T cell metabolism, we analyzed the OCR of mitochondrial respiration and the ECAR of glycolysis performing a mitochondrial stress test in bulk cultures of GFP-expressing control and Nef.GFP-expressing cells (Fig. 4H, Supplemental Fig. 2). A3.01 cells were cotransfected with expression plasmids for GFP or Nef.GFP and pDisplay.YFP, sorted using GFP microBeads and plated onto Fibronectin-coated Seahorse microplates. OCR and ECAR were measured at various time points in bulk cells before treatment, upon oligomycin, FCCP, and rotenone treatment, as highlighted in Fig. 4H and Supplemental Fig. 2 (arrows), to assess basal respiration, ATP production, proton leak, maximal respiration, spare capacity, and nonmitochondrial respiration, as indicated (Fig. 4H). No significant differences between GFP- and Nef.GFP-expressing cultures were detectable in mitochondrial respiration (Fig. 4H) and glycolysis (Supplemental Fig. 2). We conclude that although the disruption of cell polarization by Nef is accompanied by reduced energy consumption in individual cells at the initiation of cell polarization, Nef does not imprint general metabolic changes in CD4 T lymphocytes.

Plotting the duration of each phase of polarized/nonpolarized morphology revealed that GFP-expressing control cells displayed polarized morphology for the majority of the time analyzed (Fig. 5A, 5C, 5G). In contrast, Nef.GFP-expressing cells almost constantly displayed nonpolarized morphology and only rarely switched to short intervals of polarization (Fig. 5A, 5C, 5D, 5G). Mutation of the PAK2 or NAKC interaction motifs in Nef resulted in significantly more frequent and longer-lasting polarization intervals (Fig. 5A, 5E, 5F, 5G). Despite comparable levels of expression of both mutants that exceeded those of WT Nef.GFP in this experiment (Fig. 5B), cells expressing NefΔ12–39 had a tendency to display more expanded periods of polarization; however, this difference was not statistically significant to NefF195A (Fig. 5G). Expression of Nef, hence, reduces both the frequency with which T cells undergo polarization as well as the duration of intervals with polarized cell morphology in a manner that depends on the interaction surface with NAKC and PAK2.

FIGURE 5.

Determinants in Nef for association with NAKC as well as PAK2 reduce T cell polarity oscillations. (A) Time projection of live-cell spinning-disc microscopy videos of A3.01 cells transiently expressing the indicated proteins as in Fig. 4A. Scale bars, 5 μm. (B) Western blot analyses of the cells shown in (A) adjusted for similar amounts of GFP-positive cells and probed for GFP and transferrin receptor (TfR). (CF) Polarization plots of A3.01 cells analyzed as in (A) over a 1-h observation period. Polarization (black) and no polarization (white) periods for individual cells are depicted over the acquisition time (in minutes). Fifteen cells were analyzed per condition. (G) Relative time in a polarized morphology of the cells shown in (C)–(F). Shown are mean values with SD. The p values have been calculated using a one-way ANOVA with Holm Sidak multiple comparison test. **p < 0.01, ***p < 0.001.

FIGURE 5.

Determinants in Nef for association with NAKC as well as PAK2 reduce T cell polarity oscillations. (A) Time projection of live-cell spinning-disc microscopy videos of A3.01 cells transiently expressing the indicated proteins as in Fig. 4A. Scale bars, 5 μm. (B) Western blot analyses of the cells shown in (A) adjusted for similar amounts of GFP-positive cells and probed for GFP and transferrin receptor (TfR). (CF) Polarization plots of A3.01 cells analyzed as in (A) over a 1-h observation period. Polarization (black) and no polarization (white) periods for individual cells are depicted over the acquisition time (in minutes). Fifteen cells were analyzed per condition. (G) Relative time in a polarized morphology of the cells shown in (C)–(F). Shown are mean values with SD. The p values have been calculated using a one-way ANOVA with Holm Sidak multiple comparison test. **p < 0.01, ***p < 0.001.

Close modal

In our previous work, we showed that the inability of Nef-expressing T cells to polarize was associated with their failure to undergo diapedesis (37). Therefore, we tested how the NAKC-binding–deficient mutant Nef Δ12–39 would score in these assays. To this end, we made use of a previously established experimental system in which mouse CD4+ T lymphocytes are transduced with MLV vectors with nefIRES∆ngfr expression cassettes, sorted via NGFR cell-surface expression, and labeled with different CellTracker dyes. This approach has previously been validated to allow for functional analysis of homogenously Nef-expressing primary mouse T lymphocytes in which Nef is expressed to similar levels and exerts its activities comparably to Nef expressed in the context of HIV-1 infection of primary human T lymphocytes (37). Cells were mixed and perfused over a pMBMEC monolayer overlaid with CCL21 to image the extravasation process by video microscopy over 20 min (Fig. 6A, Supplemental Video 2). Frame-by-frame offline analysis of cell behavior under flow enabled us to dissect which step of the extravasation process is influenced by Nef. As observed previously (37), Nef expression had no effect on the cells’ ability to arrest on CCL21-overlaid pMBMECs and Nef-expressing cells resisted detachment equally well during an observation period of 20 min (Fig. 6B, 6E [detached]) but resulted in a marked reduction of cell polarization on the endothelium (Fig. 6C, 6D). Nef expression also induced a mild, but statistically significant, reduction in the number of overall diapedesis events (Fig. 6E, diapedesis) and a delay in transmigration kinetics (Fig. 6F). Interestingly, polarization and diapedesis of cells expressing Nef Δ12–39 were virtually indistinguishable from that of control cells (Fig. 6C–F), indicating that disruption of the NAKC-binding site in Nef results in a loss of its ability to interfere with CD4+ T cell polarization and diapedesis.

FIGURE 6.

The NAKC interaction site in Nef is critical for Nef-mediated interference of T cell extravasation and homing to lymph nodes. Twenty-four hours posttransduction and sorting, primary T lymphocytes were labeled with one of three different CellTracker dyes (CMTMR, red; Deep Red, green; CMAC, blue), mixed in equal amounts, and applied to a TNF-α–stimulated, CCL21-overlaid primary murine brain endothelium under low shear stress (0.1 dyne/cm2). After 4 min, cell attachment had occurred, shear stress was increased (1.5 dyne/cm2), and extravasation was monitored over 20 min with one image acquired every 10 s. (A) Still images of Supplemental Video 2 showing a representative field of view of an extravasation experiment. Shown is an endothelial cell monolayer with control, Nef WT, and Nef ∆12–39-expressing T lymphocytes, pseudocolored in green, red, and blue, respectively. Colored lines highlight migration tracks of an Nef-expressing cell (red line), two control cells (dark green line, light green following diapedesis), and an Nef ∆12–39-expressing cell (dark blue line, light blue following diapedesis). Scale bar, 10 μm. Images were acquired using a 10× objective. (B) Relative efficiency of arrest on the endothelium under shear stress. Shown is the percentage of input cells that attached to the endothelial cell monolayer at the time point when higher shear stress rates are first applied. (C) Still image of a representative extravasation time-lapse video with indicated measurements of cell length and width as used for calculation of polarization indices (calculated by dividing cell length by cell width). (D) Polarization indices of single cells at one representative time point, measured as shown in (C). Red bars indicate mean values. The p values were calculated using a one-tailed ANOVA test with a Newman–Keuls posttest. (E) Quantification of the percentage of cells that perform diapedesis, remain on top of the endothelium (on top), or were detached from the endothelium by shear stress (detached). Shown are mean values of three experiments with SD. The p values were calculated by performing a Student t test. (F) Kinetics of diapedesis during the 20-min time course of imaging. Shown are mean values from three experiments with SD. The p values of Nef WT relative to control and relative to Nef ∆12–39 are shown in red and black, respectively. The p values were calculated by performing a Student t test. (G and H) To evaluate the role of NAKC interacting motif of Nef on T cell homing block, primary murine T cells were transduced, sorted, labeled with cell tracing dyes, and adoptively transferred into recipient mice. For calculating relative homing efficiencies, control T cells were coinjected with T cells overexpressing WT or Nef mutants. Cells were harvested from peripheral lymph nodes (pLN) (G) or mesenteric lymph nodes (mLN) (H) after 4 or 24 h and analyzed by flow cytometry. The homing ratio in each mouse was calculated relative to coinjected control cells, arbitrarily set to 1.0 (indicated by dashed line). Each symbol represents data from one animal, and Mann–Whitney rank sum test was used to calculate p values. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 6.

The NAKC interaction site in Nef is critical for Nef-mediated interference of T cell extravasation and homing to lymph nodes. Twenty-four hours posttransduction and sorting, primary T lymphocytes were labeled with one of three different CellTracker dyes (CMTMR, red; Deep Red, green; CMAC, blue), mixed in equal amounts, and applied to a TNF-α–stimulated, CCL21-overlaid primary murine brain endothelium under low shear stress (0.1 dyne/cm2). After 4 min, cell attachment had occurred, shear stress was increased (1.5 dyne/cm2), and extravasation was monitored over 20 min with one image acquired every 10 s. (A) Still images of Supplemental Video 2 showing a representative field of view of an extravasation experiment. Shown is an endothelial cell monolayer with control, Nef WT, and Nef ∆12–39-expressing T lymphocytes, pseudocolored in green, red, and blue, respectively. Colored lines highlight migration tracks of an Nef-expressing cell (red line), two control cells (dark green line, light green following diapedesis), and an Nef ∆12–39-expressing cell (dark blue line, light blue following diapedesis). Scale bar, 10 μm. Images were acquired using a 10× objective. (B) Relative efficiency of arrest on the endothelium under shear stress. Shown is the percentage of input cells that attached to the endothelial cell monolayer at the time point when higher shear stress rates are first applied. (C) Still image of a representative extravasation time-lapse video with indicated measurements of cell length and width as used for calculation of polarization indices (calculated by dividing cell length by cell width). (D) Polarization indices of single cells at one representative time point, measured as shown in (C). Red bars indicate mean values. The p values were calculated using a one-tailed ANOVA test with a Newman–Keuls posttest. (E) Quantification of the percentage of cells that perform diapedesis, remain on top of the endothelium (on top), or were detached from the endothelium by shear stress (detached). Shown are mean values of three experiments with SD. The p values were calculated by performing a Student t test. (F) Kinetics of diapedesis during the 20-min time course of imaging. Shown are mean values from three experiments with SD. The p values of Nef WT relative to control and relative to Nef ∆12–39 are shown in red and black, respectively. The p values were calculated by performing a Student t test. (G and H) To evaluate the role of NAKC interacting motif of Nef on T cell homing block, primary murine T cells were transduced, sorted, labeled with cell tracing dyes, and adoptively transferred into recipient mice. For calculating relative homing efficiencies, control T cells were coinjected with T cells overexpressing WT or Nef mutants. Cells were harvested from peripheral lymph nodes (pLN) (G) or mesenteric lymph nodes (mLN) (H) after 4 or 24 h and analyzed by flow cytometry. The homing ratio in each mouse was calculated relative to coinjected control cells, arbitrarily set to 1.0 (indicated by dashed line). Each symbol represents data from one animal, and Mann–Whitney rank sum test was used to calculate p values. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

Finally, we sought to assess whether the disruption of T cell polarization and diapedesis may be linked to the ability of Nef to restrict T lymphocyte motility in vivo. To this end, mouse CD4+ T lymphocytes were transduced, sorted via NGFR cell-surface expression, and then adoptively transferred into recipient mice as established previously (37). Transduced and sorted cell populations were labeled with cell-tracing dyes, and control T cells were mixed with WT or mutant Nef–expressing T cells. Nef WT and NAKC-binding deficient Nef Δ12–39 were compared with Nef F195A, which is deficient in preventing T cell actin remodeling and lymph node homing (37). Nef-expressing cells were mixed with control cells and adoptively cotransferred into C57BL/6 recipient mice, and a small fraction of cells from each sample were analyzed by flow cytometry to estimate the input ratio that was used to normalize the homing efficiency. Four and twenty-four hours after cell transfer, spleen, peripheral, and mesenteric lymph nodes were harvested and analyzed independently for homing efficiency by flow cytometry. In line with our previous results (37), Nef expression did not have any statistically significant effect on T cell recruitment to spleen, which does not require the crossing of an endothelial barrier (70) (data not shown). In contrast, homing of Nef-expressing T lymphocytes to peripheral and mesenteric lymph nodes was markedly reduced at 4 h (42 ± 16% or 40 ± 16% residual homing compared with control) and 24 h (45 ± 20% and 41 ± 23% residual homing compared with control) posttransfer. As shown previously, T lymphocytes expressing Nef F195A only displayed a mild reduction of homing efficiencies 4 h posttransfer, whereas homing at 24 h was slightly enhanced as compared with control cells (37). In contrast, T lymphocytes expressing Nef Δ12–39 displayed full homing potential as early as 4 h posttransfer and were indistinguishable from control cells at both time points analyzed. NAKC and PAK2 interaction surfaces are thus both critical for the ability of Nef to interfere with CD4+ T lymphocyte trafficking in vivo.

The goal of this study was to gain insight into the actin-remodeling independent mechanism by which the HIV-1 pathogenesis factor Nef impairs extravasation of CD4+ T lymphocytes at the diapedesis step. We find that Nef potently restricts the polarization of infected primary, as well as immortalized-transfected, CD4+ T lymphocytes on adhesive substrates and results in rounded, instead of elongated, cell morphology. Because this effect was not observed in cells with nef-deficient HIV, and isolated expression of Nef alone impaired cell polarization, Nef is necessary and sufficient for disruption of CD4+ T lymphocyte polarity. Polarity disruption was observed, albeit to variable extent, for a wide variety of HIV-1, HIV-2, and SIV Nef proteins, suggesting that this activity is overall conserved during lentiviral evolution. Polarity disruption resulted from arrest of dynamic changes in T lymphocyte morphology and was paralleled with a reduced need for energy to support these alterations in cell morphology. In contrast, mitochondrial respiration was not globally altered in Nef-expressing CD4+ T cells. Mapping studies revealed the binding site for NAKC as important molecular determinant for this Nef activity. NAKC-binding–deficient Nef, hence, did not disrupt T cell polarization of CD4+ T lymphocytes primed for extravasation under shear stress, did not restrict subsequent diapedesis, and failed to affect T lymphocyte homing to lymph nodes. These results suggest disruption of T cell polarization as a novel mechanism by which HIV-1 Nef impairs T lymphocyte motility in vivo and identifies the NAKC biding site as critical determinant for this activity.

A central finding of this study was that disruption of T cell polarity by Nef correlates with the inability of these cells to undergo efficient extravasation and homing to lymph nodes. Considering that 1) morphological plasticity and polarization is a well-established prerequisite for T cell diapedesis (39), and 2) extravasation represents an essential step in the homing process (38), it is conceivable that polarity disruption directly contributes to the reduced homing potential of Nef-expressing CD4+ T lymphocytes. Conceivably, Nef-induced polarity disruption results in impaired diapedesis, and this effect likely synergizes with the reduction of host cell actin dynamics, which in turn, limits intralymph node motility of cells that successfully underwent diapedesis (37). Consistently, protein interaction motifs essential for interaction of Nef with NAKC (∆12–39) and for interference with actin remodeling (F195A) are both required for Nef’s ability to interfere with lymph node homing. However, cells expressing NAKC-binding–deficient Nef Δ12–39 homed to lymph nodes with efficiencies comparable to that of control cells already early after adoptive transfer (4 h), whereas Nef F195A–expressing cells reached full homing efficiency, or are even slightly enriched, after 24 h. Temporally altered homing efficiencies may result from a concerted residual impairment of homing and egress by the F195A, but not ∆12–39, mutant of Nef. Of note, mutation of either the PAK2 or NAKC interaction motif in Nef abrogated its ability to disrupt T cell polarization, but only Nef Δ12–39 had lost the ability to block diapedesis. This indicates that Nef, via its PAK2 as well as NAKC binding sites, targets essential, but distinct, aspects of morphological T lymphocyte polarization. However, only interactions occurring via the NAKC binding surface lead to functional disruption and thus compromise diapedesis.

These findings raise the question about the molecular mechanism by which the NAKC binding site enables Nef to disrupt T cell polarity and extravasation. An obvious hypothesis is that this reflects the association of Nef with NAKC. NAKC assembly is best known to mediate the enhancement of HIV transcription (19, 47, 7173) as well as the release of extracellular vesicles with proinflammatory molecules (74, 75) by Nef. These effects are likely unrelated to T lymphocyte polarization and diapedesis. However, with Lck, PI3-K, and PKC θ (19, 47, 76), NAKC contains at least three kinases with known roles in T lymphocyte polarization toward the immunological synapse (77, 78), and the association with NAKC enables Nef to affect integrin signaling (72, 79) and thus a central pathway–regulating cell polarity. Our attempts to test the relevance of individual NAKC components for the disruption of T cell polarization by silencing their expression and/or pharmacologically inhibiting their activity were hampered by the interference of these measures with cell polarization even in the absence of Nef, which precluded meaningful analysis (data not shown). Dissecting whether NAKC is directly involved in these effects of Nef will therefore require the identification of mutants that uncouple intrinsic roles in T cell polarity from Nef-mediated disruption thereof. Because the NAKC biding site is recognized as an important determinant for an increasing number of Nef activities, including antagonism of the host cell restriction factor SERINC5 (29) and promoting CD4+ T cell depletion in ex vivo cultures of human tonsils (80); the 27-aa stretch deleted in Nef Δ12–39 may also mediate additional, yet unknown, interactions with cellular ligands other than NAKC.

An additional observation made in the course of this study is that the perceived disruption of cell polarization by Nef at any given time point reflects the inhibition of frequent morphological oscillations of T lymphocytes between elongated and rounded shapes, which expression of Nef arrests at the rounded stage. Our measurements revealed that this reduction in morphological alterations is associated with a reduced requirement for energy in individual cells when shape changes are initiated, which, however, does not translate into appreciable differences in the metabolism of the corresponding cell culture when analyzed in bulk. This seeming discrepancy likely reflects that cell polarization of individual cells in the culture occurs in an asynchronous manner, and differences imprinted by Nef in the few cells in which polarization is initiated per time point cannot be detected on the background metabolism of the bulk of cells in the culture. Importantly, however, this indicates that effects of Nef on energy consumption are limited to phases of cell shape transition, suggesting that Nef is not a major determinant of HIV-induced alterations of T lymphocyte metabolism (8184). Whether Nef induces specific mechanisms to depleting energy to prevent morphological oscillations, or the reduced energy consumption at phases of shape transition is a consequence of the inhibition of host cell vesicular trafficking and cytoskeleton dynamics by the viral protein remains to be determined.

In sum, this study defines disruption of T cell polarity and extravasation as a novel activity of lentiviral Nef proteins. This mechanism is essential for the ability of the viral protein to interfere with T lymphocyte homing to lymph nodes. The fact that Nef contains two molecular surfaces that interfere with lymph node homing via independent mechanisms and the evolutionary conservation of this activity suggests that this activity may be critical for the role Nef plays in AIDS pathogenesis. Interference of Nef with homing by T cell polarity disruption may reduce the frequency of HIV-infected cells in distal areas of the lymph node and thereby locally enrich HIV-infected cells in the T cell–rich paracortex to facilitate viral spread and/or immune evasion of infected cells (36).

We are grateful to Reno Debets, Hans-Georg Kräusslich, and Frank Kirchhoff for the kind gift of reagents and to Jan-Philip Bergeest for the ImageJ Annotation Plugin. M.T. thanks the Nakatani Foundation for support. This work benefitted from optical setups of the Microscopy Imaging Center of the University of Bern.

This work was supported by Deutsche Forschungsgemeinschaft Grants FA378/10-2 and SFB1129 (to O.T.F.) and SFB1129 (to M.T.), and the Nakatani Foundation (to M.T.). N. Tsopoulidis was supported by a Heidelberg Biosciences International Graduate School fellowship. O.T.F. and M.T. are members of the CellNetworks Cluster of Excellence (EXO81).

The online version of this article contains supplemental material.

Abbreviations used in this article:

ACF

autocorrelation function

CMM

complete mouse medium

D

donor

ECAR

extracellular acidification rate

MLV

murine leukemia virus

NAKC

Nef-associated kinase complex

OCR

oxygen consumption rate

pMBMEC

primary mouse brain microvascular endothelial cell

WT

wild type.

1
Deacon
,
N. J.
,
A.
Tsykin
,
A.
Solomon
,
K.
Smith
,
M.
Ludford-Menting
,
D. J.
Hooker
,
D. A.
McPhee
,
A. L.
Greenway
,
A.
Ellett
,
C.
Chatfield
, et al
.
1995
.
Genomic structure of an attenuated quasi species of HIV-1 from a blood transfusion donor and recipients.
Science
270
:
988
991
.
2
Geyer
,
M.
,
O. T.
Fackler
,
B. M.
Peterlin
.
2001
.
Structure--function relationships in HIV-1 Nef.
EMBO Rep.
2
:
580
585
.
3
Kestler
, III,
H. W.
,
D. J.
Ringler
,
K.
Mori
,
D. L.
Panicali
,
P. K.
Sehgal
,
M. D.
Daniel
,
R. C.
Desrosiers
.
1991
.
Importance of the nef gene for maintenance of high virus loads and for development of AIDS.
Cell
65
:
651
662
.
4
Kirchhoff
,
F.
,
T. C.
Greenough
,
D. B.
Brettler
,
J. L.
Sullivan
,
R. C.
Desrosiers
.
1995
.
Brief report: absence of intact nef sequences in a long-term survivor with nonprogressive HIV-1 infection.
N. Engl. J. Med.
332
:
228
232
.
5
Laguette
,
N.
,
C.
Brégnard
,
S.
Benichou
,
S.
Basmaciogullari
.
2010
.
Human immunodeficiency virus (HIV) type-1, HIV-2 and simian immunodeficiency virus Nef proteins.
Mol. Aspects Med.
31
:
418
433
.
6
Kirchhoff
,
F.
,
M.
Schindler
,
A.
Specht
,
N.
Arhel
,
J.
Münch
.
2008
.
Role of Nef in primate lentiviral immunopathogenesis.
Cell. Mol. Life Sci.
65
:
2621
2636
.
7
Abraham
,
L.
,
O. T.
Fackler
.
2012
.
HIV-1 Nef: a multifaceted modulator of T cell receptor signaling.
Cell Commun. Signal.
10
:
39
.
8
Chaudhry
,
A.
,
S. R.
Das
,
S.
Jameel
,
A.
George
,
V.
Bal
,
S.
Mayor
,
S.
Rath
.
2008
.
HIV-1 Nef induces a Rab11-dependent routing of endocytosed immune costimulatory proteins CD80 and CD86 to the Golgi.
Traffic
9
:
1925
1935
.
9
Garcia
,
J. V.
,
A. D.
Miller
.
1991
.
Serine phosphorylation-independent downregulation of cell-surface CD4 by nef.
Nature
350
:
508
511
.
10
Michel
,
N.
,
I.
Allespach
,
S.
Venzke
,
O. T.
Fackler
,
O. T.
Keppler
.
2005
.
The Nef protein of human immunodeficiency virus establishes superinfection immunity by a dual strategy to downregulate cell-surface CCR5 and CD4.
Curr. Biol.
15
:
714
723
.
11
Schwartz
,
O.
,
V.
Maréchal
,
S.
Le Gall
,
F.
Lemonnier
,
J. M.
Heard
.
1996
.
Endocytosis of major histocompatibility complex class I molecules is induced by the HIV-1 Nef protein.
Nat. Med.
2
:
338
342
.
12
Stumptner-Cuvelette
,
P.
,
S.
Morchoisne
,
M.
Dugast
,
S.
Le Gall
,
G.
Raposo
,
O.
Schwartz
,
P.
Benaroch
.
2001
.
HIV-1 Nef impairs MHC class II antigen presentation and surface expression.
Proc. Natl. Acad. Sci. USA
98
:
12144
12149
.
13
Cerboni
,
C.
,
F.
Neri
,
N.
Casartelli
,
A.
Zingoni
,
D.
Cosman
,
P.
Rossi
,
A.
Santoni
,
M.
Doria
.
2007
.
Human immunodeficiency virus 1 Nef protein downmodulates the ligands of the activating receptor NKG2D and inhibits natural killer cell-mediated cytotoxicity.
J. Gen. Virol.
88
:
242
250
.
14
Galaski
,
J.
,
F.
Ahmad
,
N.
Tibroni
,
F. M.
Pujol
,
B.
Müller
,
R. E.
Schmidt
,
O. T.
Fackler
.
2016
.
Cell surface downregulation of NK cell ligands by patient-derived HIV-1 Vpu and Nef alleles.
J. Acquir. Immune Defic. Syndr.
72
:
1
10
.
15
Haller
,
C.
,
B.
Müller
,
J. V.
Fritz
,
M.
Lamas-Murua
,
B.
Stolp
,
F. M.
Pujol
,
O. T.
Keppler
,
O. T.
Fackler
.
2014
.
HIV-1 Nef and Vpu are functionally redundant broad-spectrum modulators of cell surface receptors, including tetraspanins.
J. Virol.
88
:
14241
14257
.
16
Matheson
,
N. J.
,
J.
Sumner
,
K.
Wals
,
R.
Rapiteanu
,
M. P.
Weekes
,
R.
Vigan
,
J.
Weinelt
,
M.
Schindler
,
R.
Antrobus
,
A. S.
Costa
, et al
.
2015
.
Cell surface proteomic map of HIV infection reveals antagonism of amino acid metabolism by Vpu and Nef.
Cell Host Microbe
18
:
409
423
.
17
Matusali
,
G.
,
M.
Potestà
,
A.
Santoni
,
C.
Cerboni
,
M.
Doria
.
2012
.
The human immunodeficiency virus type 1 Nef and Vpu proteins downregulate the natural killer cell-activating ligand PVR.
J. Virol.
86
:
4496
4504
.
18
Abraham
,
L.
,
P.
Bankhead
,
X.
Pan
,
U.
Engel
,
O. T.
Fackler
.
2012
.
HIV-1 Nef limits communication between linker of activated T cells and SLP-76 to reduce formation of SLP-76-signaling microclusters following TCR stimulation.
J. Immunol.
189
:
1898
1910
.
19
Baur
,
A. S.
,
G.
Sass
,
B.
Laffert
,
D.
Willbold
,
C.
Cheng-Mayer
,
B. M.
Peterlin
.
1997
.
The N-terminus of Nef from HIV-1/SIV associates with a protein complex containing Lck and a serine kinase.
Immunity
6
:
283
291
.
20
Fackler
,
O. T.
,
A. S.
Baur
.
2002
.
Live and let die: Nef functions beyond HIV replication.
Immunity
16
:
493
497
.
21
Haller
,
C.
,
S.
Rauch
,
O. T.
Fackler
.
2007
.
HIV-1 Nef employs two distinct mechanisms to modulate Lck subcellular localization and TCR induced actin remodeling.
PLoS One
2
:
e1212
.
22
Haller
,
C.
,
S.
Rauch
,
N.
Michel
,
S.
Hannemann
,
M. J.
Lehmann
,
O. T.
Keppler
,
O. T.
Fackler
.
2006
.
The HIV-1 pathogenicity factor Nef interferes with maturation of stimulatory T-lymphocyte contacts by modulation of N-Wasp activity.
J. Biol. Chem.
281
:
19618
19630
.
23
Pan
,
X.
,
M. M.
Geist
,
J. M.
Rudolph
,
W.
Nickel
,
O. T.
Fackler
.
2013
.
HIV-1 Nef disrupts membrane-microdomain-associated anterograde transport for plasma membrane delivery of selected Src family kinases.
Cell. Microbiol.
15
:
1605
1621
.
24
Pan
,
X.
,
J. M.
Rudolph
,
L.
Abraham
,
A.
Habermann
,
C.
Haller
,
J.
Krijnse-Locker
,
O. T.
Fackler
.
2012
.
HIV-1 Nef compensates for disorganization of the immunological synapse by inducing trans-Golgi network-associated Lck signaling.
Blood
119
:
786
797
.
25
Silva
,
J. G.
,
N. P.
Martins
,
R.
Henriques
,
H.
Soares
.
2016
.
HIV-1 Nef impairs the formation of calcium membrane territories controlling the signaling nanoarchitecture at the immunological synapse.
J. Immunol.
197
:
4042
4052
.
26
Thoulouze
,
M. I.
,
N.
Sol-Foulon
,
F.
Blanchet
,
A.
Dautry-Varsat
,
O.
Schwartz
,
A.
Alcover
.
2006
.
Human immunodeficiency virus type-1 infection impairs the formation of the immunological synapse.
Immunity
24
:
547
561
.
27
Schindler
,
M.
,
J.
Münch
,
O.
Kutsch
,
H.
Li
,
M. L.
Santiago
,
F.
Bibollet-Ruche
,
M. C.
Müller-Trutwin
,
F. J.
Novembre
,
M.
Peeters
,
V.
Courgnaud
, et al
.
2006
.
Nef-mediated suppression of T cell activation was lost in a lentiviral lineage that gave rise to HIV-1.
Cell
125
:
1055
1067
.
28
Rosa
,
A.
,
A.
Chande
,
S.
Ziglio
,
V.
De Sanctis
,
R.
Bertorelli
,
S. L.
Goh
,
S. M.
McCauley
,
A.
Nowosielska
,
S. E.
Antonarakis
,
J.
Luban
, et al
.
2015
.
HIV-1 Nef promotes infection by excluding SERINC5 from virion incorporation.
Nature
526
:
212
217
.
29
Trautz
,
B.
,
V.
Pierini
,
R.
Wombacher
,
B.
Stolp
,
A. J.
Chase
,
M.
Pizzato
,
O. T.
Fackler
.
2016
.
The antagonism of HIV-1 Nef to SERINC5 particle infectivity restriction involves the counteraction of virion-associated pools of the restriction factor.
J. Virol.
90
:
10915
10927
.
30
Trautz
,
B.
,
H.
Wiedemann
,
C.
Lüchtenborg
,
V.
Pierini
,
J.
Kranich
,
B.
Glass
,
H. G.
Kräusslich
,
T.
Brocker
,
M.
Pizzato
,
A.
Ruggieri
, et al
.
2017
.
The host-cell restriction factor SERINC5 restricts HIV-1 infectivity without altering the lipid composition and organization of viral particles.
J. Biol. Chem.
292
:
13702
13713
.
31
Usami
,
Y.
,
Y.
Wu
,
H. G.
Göttlinger
.
2015
.
SERINC3 and SERINC5 restrict HIV-1 infectivity and are counteracted by Nef.
Nature
526
:
218
223
.
32
Choe
,
E. Y.
,
E. S.
Schoenberger
,
J. E.
Groopman
,
I. W.
Park
.
2002
.
HIV Nef inhibits T cell migration.
J. Biol. Chem.
277
:
46079
46084
.
33
Janardhan
,
A.
,
T.
Swigut
,
B.
Hill
,
M. P.
Myers
,
J.
Skowronski
.
2004
.
HIV-1 Nef binds the DOCK2-ELMO1 complex to activate rac and inhibit lymphocyte chemotaxis.
PLoS Biol.
2
:
E6
.
34
Park
,
I. W.
,
J. J.
He
.
2009
.
HIV-1 Nef-mediated inhibition of T cell migration and its molecular determinants.
J. Leukoc. Biol.
86
:
1171
1178
.
35
Stolp
,
B.
,
M.
Reichman-Fried
,
L.
Abraham
,
X.
Pan
,
S. I.
Giese
,
S.
Hannemann
,
P.
Goulimari
,
E.
Raz
,
R.
Grosse
,
O. T.
Fackler
.
2009
.
HIV-1 Nef interferes with host cell motility by deregulation of Cofilin.
Cell Host Microbe
6
:
174
186
.
36
Fackler
,
O. T.
,
T. T.
Murooka
,
A.
Imle
,
T. R.
Mempel
.
2014
.
Adding new dimensions: towards an integrative understanding of HIV-1 spread.
Nat. Rev. Microbiol.
12
:
563
574
.
37
Stolp
,
B.
,
A.
Imle
,
F. M.
Coelho
,
M.
Hons
,
R.
Gorina
,
R.
Lyck
,
J. V.
Stein
,
O. T.
Fackler
.
2012
.
HIV-1 Nef interferes with T-lymphocyte circulation through confined environments in vivo.
Proc. Natl. Acad. Sci. USA
109
:
18541
18546
.
38
Girard
,
J. P.
,
C.
Moussion
,
R.
Förster
.
2012
.
HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes.
Nat. Rev. Immunol.
12
:
762
773
.
39
Schnoor
,
M.
,
P.
Alcaide
,
M. B.
Voisin
,
J. D.
van Buul
.
2015
.
Crossing the vascular wall: common and unique mechanisms exploited by different leukocyte subsets during extravasation.
Mediators Inflamm.
2015
:
946509
.
40
Imle
,
A.
,
L.
Abraham
,
N.
Tsopoulidis
,
B.
Hoflack
,
K.
Saksela
,
O. T.
Fackler
.
2015
.
Association with PAK2 enables functional interactions of lentiviral Nef proteins with the exocyst complex.
MBio
6
:
e01309-e01315
.
41
O’Neill
,
E.
,
L. S.
Kuo
,
J. F.
Krisko
,
D. R.
Tomchick
,
J. V.
Garcia
,
J. L.
Foster
.
2006
.
Dynamic evolution of the human immunodeficiency virus type 1 pathogenic factor, Nef.
J. Virol.
80
:
1311
1320
.
42
Rauch
,
S.
,
K.
Pulkkinen
,
K.
Saksela
,
O. T.
Fackler
.
2008
.
Human immunodeficiency virus type 1 Nef recruits the guanine exchange factor Vav1 via an unexpected interface into plasma membrane microdomains for association with p21-activated kinase 2 activity.
J. Virol.
82
:
2918
2929
.
43
Schindler
,
M.
,
D.
Rajan
,
A.
Specht
,
C.
Ritter
,
K.
Pulkkinen
,
K.
Saksela
,
F.
Kirchhoff
.
2007
.
Association of Nef with p21-activated kinase 2 is dispensable for efficient human immunodeficiency virus type 1 replication and cytopathicity in ex vivo-infected human lymphoid tissue.
J. Virol.
81
:
13005
13014
.
44
Fackler
,
O. T.
,
A.
Moris
,
N.
Tibroni
,
S. I.
Giese
,
B.
Glass
,
O.
Schwartz
,
H. G.
Kräusslich
.
2006
.
Functional characterization of HIV-1 Nef mutants in the context of viral infection.
Virology
351
:
322
339
.
45
Meuwissen
,
P. J.
,
B.
Stolp
,
V.
Iannucci
,
J.
Vermeire
,
E.
Naessens
,
K.
Saksela
,
M.
Geyer
,
G.
Vanham
,
K. K.
Arien
,
O. T.
Fackler
,
B.
Verhasselt
.
2012
.
Identification of a highly conserved valine-glycine-phenylalanine amino acid triplet required for HIV-1 Nef function.
Retrovirology
9
:
34
.
46
Rudolph
,
J. M.
,
N.
Eickel
,
C.
Haller
,
M.
Schindler
,
O. T.
Fackler
.
2009
.
Inhibition of T-cell receptor-induced actin remodeling and relocalization of Lck are evolutionarily conserved activities of lentiviral Nef proteins.
J. Virol.
83
:
11528
11539
.
47
Wolf
,
D.
,
S. I.
Giese
,
V.
Witte
,
E.
Krautkrämer
,
S.
Trapp
,
G.
Sass
,
C.
Haller
,
K.
Blume
,
O. T.
Fackler
,
A. S.
Baur
.
2008
.
Novel (n)PKC kinases phosphorylate Nef for increased HIV transcription, replication and perinuclear targeting.
Virology
370
:
45
54
.
48
Keppler
,
O. T.
,
I.
Allespach
,
L.
Schüller
,
D.
Fenard
,
W. C.
Greene
,
O. T.
Fackler
.
2005
.
Rodent cells support key functions of the human immunodeficiency virus type 1 pathogenicity factor Nef.
J. Virol.
79
:
1655
1665
.
49
Pizzato
,
M.
,
O.
Erlwein
,
D.
Bonsall
,
S.
Kaye
,
D.
Muir
,
M. O.
McClure
.
2009
.
A one-step SYBR Green I-based product-enhanced reverse transcriptase assay for the quantitation of retroviruses in cell culture supernatants.
J. Virol. Methods
156
:
1
7
.
50
Riedl
,
J.
,
K. C.
Flynn
,
A.
Raducanu
,
F.
Gärtner
,
G.
Beck
,
M.
Bösl
,
F.
Bradke
,
S.
Massberg
,
A.
Aszodi
,
M.
Sixt
,
R.
Wedlich-Söldner
.
2010
.
Lifeact mice for studying F-actin dynamics.
Nat. Methods
7
:
168
169
.
51
Lyck
,
R.
,
N.
Ruderisch
,
A. G.
Moll
,
O.
Steiner
,
C. D.
Cohen
,
B.
Engelhardt
,
V.
Makrides
,
F.
Verrey
.
2009
.
Culture-induced changes in blood-brain barrier transcriptome: implications for amino-acid transporters in vivo.
J. Cereb. Blood Flow Metab.
29
:
1491
1502
.
52
Steiner
,
O.
,
C.
Coisne
,
R.
Cecchelli
,
R.
Boscacci
,
U.
Deutsch
,
B.
Engelhardt
,
R.
Lyck
.
2010
.
Differential roles for endothelial ICAM-1, ICAM-2, and VCAM-1 in shear-resistant T cell arrest, polarization, and directed crawling on blood-brain barrier endothelium.
J. Immunol.
185
:
4846
4855
.
53
Coisne
,
C.
,
R.
Lyck
,
B.
Engelhardt
.
2013
.
Live cell imaging techniques to study T cell trafficking across the blood-brain barrier in vitro and in vivo.
Fluids Barriers CNS
10
:
7
.
54
Burk
,
A. S.
,
C.
Monzel
,
H. Y.
Yoshikawa
,
P.
Wuchter
,
R.
Saffrich
,
V.
Eckstein
,
M.
Tanaka
,
A. D.
Ho
.
2015
.
Quantifying adhesion mechanisms and dynamics of human hematopoietic stem and progenitor cells.
Sci. Rep.
5
:
9370
.
55
Maeda
,
Y. T.
,
J.
Inose
,
M. Y.
Matsuo
,
S.
Iwaya
,
M.
Sano
.
2008
.
Ordered patterns of cell shape and orientational correlation during spontaneous cell migration.
PLoS One
3
:
e3734
.
56
Partin
,
A. W.
,
J. S.
Schoeniger
,
J. L.
Mohler
,
D. S.
Coffey
.
1989
.
Fourier analysis of cell motility: correlation of motility with metastatic potential.
Proc. Natl. Acad. Sci. USA
86
:
1254
1258
.
57
Betz
,
T.
,
M.
Lenz
,
J. F.
Joanny
,
C.
Sykes
.
2009
.
ATP-dependent mechanics of red blood cells.
Proc. Natl. Acad. Sci. USA
106
:
15320
15325
.
58
Gov
,
N. S.
,
S. A.
Safran
.
2005
.
Red blood cell membrane fluctuations and shape controlled by ATP-induced cytoskeletal defects.
Biophys. J.
88
:
1859
1874
.
59
Norton
,
M. P.
,
D. G.
Karczub
.
2003
.
Fundamentals of Noise and Vibration Analysis for Engineers, 2nd Ed.
Cambridge University Press
,
Cambridge, U.K.
60
Baldauf
,
H. M.
,
L.
Stegmann
,
S. M.
Schwarz
,
I.
Ambiel
,
M.
Trotard
,
M.
Martin
,
M.
Burggraf
,
G. M.
Lenzi
,
H.
Lejk
,
X.
Pan
, et al
.
2017
.
Vpx overcomes a SAMHD1-independent block to HIV reverse transcription that is specific to resting CD4 T cells.
Proc. Natl. Acad. Sci. USA
114
:
2729
2734
.
61
Geist
,
M. M.
,
X.
Pan
,
S.
Bender
,
R.
Bartenschlager
,
W.
Nickel
,
O. T.
Fackler
.
2014
.
Heterologous Src homology 4 domains support membrane anchoring and biological activity of HIV-1 Nef.
J. Biol. Chem.
289
:
14030
14044
.
62
Sugden
,
S. M.
,
M. G.
Bego
,
T. N.
Pham
,
E. A.
Cohen
.
2016
.
Remodeling of the host cell plasma membrane by HIV-1 Nef and Vpu: a strategy to ensure viral fitness and persistence.
Viruses
8
:
67
.
63
Giese
,
S. I.
,
I.
Woerz
,
S.
Homann
,
N.
Tibroni
,
M.
Geyer
,
O. T.
Fackler
.
2006
.
Specific and distinct determinants mediate membrane binding and lipid raft incorporation of HIV-1(SF2) Nef.
Virology
355
:
175
191
.
64
Krautkrämer
,
E.
,
S. I.
Giese
,
J. E.
Gasteier
,
W.
Muranyi
,
O. T.
Fackler
.
2004
.
Human immunodeficiency virus type 1 Nef activates p21-activated kinase via recruitment into lipid rafts.
J. Virol.
78
:
4085
4097
.
65
Stolp
,
B.
,
L.
Abraham
,
J. M.
Rudolph
,
O. T.
Fackler
.
2010
.
Lentiviral Nef proteins utilize PAK2-mediated deregulation of cofilin as a general strategy to interfere with actin remodeling.
J. Virol.
84
:
3935
3948
.
66
Akari
,
H.
,
S.
Arold
,
T.
Fukumori
,
T.
Okazaki
,
K.
Strebel
,
A.
Adachi
.
2000
.
Nef-induced major histocompatibility complex class I down-regulation is functionally dissociated from its virion incorporation, enhancement of viral infectivity, and CD4 down-regulation.
J. Virol.
74
:
2907
2912
.
67
Manrique
,
S.
,
D.
Sauter
,
F. A.
Horenkamp
,
S.
Lülf
,
H.
Yu
,
D.
Hotter
,
K.
Anand
,
F.
Kirchhoff
,
M.
Geyer
.
2017
.
Endocytic sorting motif interactions involved in Nef-mediated downmodulation of CD4 and CD3.
Nat. Commun.
8
:
442
.
68
Baugh
,
L. L.
,
J. V.
Garcia
,
J. L.
Foster
.
2008
.
Functional characterization of the human immunodeficiency virus type 1 Nef acidic domain.
J. Virol.
82
:
9657
9667
.
69
Saksela
,
K.
,
G.
Cheng
,
D.
Baltimore
.
1995
.
Proline-rich (PxxP) motifs in HIV-1 Nef bind to SH3 domains of a subset of Src kinases and are required for the enhanced growth of Nef+ viruses but not for down-regulation of CD4.
EMBO J.
14
:
484
491
.
70
Stein
,
J. V.
,
C.
Nombela-Arrieta
.
2005
.
Chemokine control of lymphocyte trafficking: a general overview.
Immunology
116
:
1
12
.
71
Witte
,
V.
,
B.
Laffert
,
P.
Gintschel
,
E.
Krautkrämer
,
K.
Blume
,
O. T.
Fackler
,
A. S.
Baur
.
2008
.
Induction of HIV transcription by Nef involves Lck activation and protein kinase C theta raft recruitment leading to activation of ERK1/2 but not NF kappa B.
J. Immunol.
181
:
8425
8432
.
72
Witte
,
V.
,
B.
Laffert
,
O.
Rosorius
,
P.
Lischka
,
K.
Blume
,
G.
Galler
,
A.
Stilper
,
D.
Willbold
,
P.
D’Aloja
,
M.
Sixt
, et al
.
2004
.
HIV-1 Nef mimics an integrin receptor signal that recruits the polycomb group protein Eed to the plasma membrane.
Mol. Cell
13
:
179
190
.
73
Wolf
,
D.
,
V.
Witte
,
P.
Clark
,
K.
Blume
,
M. G.
Lichtenheld
,
A. S.
Baur
.
2008
.
HIV Nef enhances Tat-mediated viral transcription through a hnRNP-K-nucleated signaling complex.
Cell Host Microbe
4
:
398
408
.
74
Lee
,
J. H.
,
S.
Schierer
,
K.
Blume
,
J.
Dindorf
,
S.
Wittki
,
W.
Xiang
,
C.
Ostalecki
,
N.
Koliha
,
S.
Wild
,
G.
Schuler
, et al
.
2016
.
HIV-Nef and ADAM17-containing plasma extracellular vesicles induce and correlate with immune pathogenesis in chronic HIV infection.
EBioMedicine
6
:
103
113
.
75
Muratori
,
C.
,
L. E.
Cavallin
,
K.
Krätzel
,
A.
Tinari
,
A.
De Milito
,
S.
Fais
,
P.
D’Aloja
,
M.
Federico
,
V.
Vullo
,
A.
Fomina
, et al
.
2009
.
Massive secretion by T cells is caused by HIV Nef in infected cells and by Nef transfer to bystander cells.
Cell Host Microbe
6
:
218
230
.
76
Wolf
,
D.
,
V.
Witte
,
B.
Laffert
,
K.
Blume
,
E.
Stromer
,
S.
Trapp
,
P.
d’Aloja
,
A.
Schürmann
,
A. S.
Baur
.
2001
.
HIV-1 Nef associated PAK and PI3-kinases stimulate Akt-independent Bad-phosphorylation to induce anti-apoptotic signals.
Nat. Med.
7
:
1217
1224
.
77
Nombela-Arrieta
,
C.
,
R. A.
Lacalle
,
M. C.
Montoya
,
Y.
Kunisaki
,
D.
Megías
,
M.
Marqués
,
A. C.
Carrera
,
S.
Mañes
,
Y.
Fukui
,
C.
Martínez-A
,
J. V.
Stein
.
2004
.
Differential requirements for DOCK2 and phosphoinositide-3-kinase gamma during T and B lymphocyte homing.
Immunity
21
:
429
441
.
78
Quann
,
E. J.
,
X.
Liu
,
G.
Altan-Bonnet
,
M.
Huse
.
2011
.
A cascade of protein kinase C isozymes promotes cytoskeletal polarization in T cells.
Nat. Immunol.
12
:
647
654
.
79
Lee
,
J. H.
,
S.
Wittki
,
T.
Bräu
,
F. S.
Dreyer
,
K.
Krätzel
,
J.
Dindorf
,
I. C.
Johnston
,
S.
Gross
,
E.
Kremmer
,
R.
Zeidler
, et al
.
2013
.
HIV Nef, paxillin, and Pak1/2 regulate activation and secretion of TACE/ADAM10 proteases.
Mol. Cell
49
:
668
679
.
80
Homann
,
S.
,
N.
Tibroni
,
I.
Baumann
,
S.
Sertel
,
O. T.
Keppler
,
O. T.
Fackler
.
2009
.
Determinants in HIV-1 Nef for enhancement of virus replication and depletion of CD4+ T lymphocytes in human lymphoid tissue ex vivo.
Retrovirology
6
:
6
.
81
Hegedus
,
A.
,
M.
Kavanagh Williamson
,
M. B.
Khan
,
J.
Dias Zeidler
,
A.
Da Poian
,
T.
El Bacha
,
E.
Struys
,
H.
Huthoff
.
2017
.
Evidence for altered glutamine metabolism in human immunodeficiency virus type 1 infected primary human CD4+ T cells.
AIDS Res. Hum. Retroviruses
33
:
1236
1247
.
82
Matheson
,
N. J.
,
E. J.
Greenwood
,
P. J.
Lehner
.
2016
.
Manipulation of immunometabolism by HIV-accessories to the crime?
Curr. Opin. Virol.
19
:
65
70
.
83
Drakesmith
,
H.
,
N.
Chen
,
H.
Ledermann
,
G.
Screaton
,
A.
Townsend
,
X. N.
Xu
.
2005
.
HIV-1 Nef down-regulates the hemochromatosis protein HFE, manipulating cellular iron homeostasis.
Proc. Natl. Acad. Sci. USA
102
:
11017
11022
.
84
Koppensteiner
,
H.
,
K.
Höhne
,
M. V.
Gondim
,
F. X.
Gobert
,
M.
Widder
,
S.
Gundlach
,
A.
Heigele
,
F.
Kirchhoff
,
M.
Winkler
,
P.
Benaroch
,
M.
Schindler
.
2014
.
Lentiviral Nef suppresses iron uptake in a strain specific manner through inhibition of Transferrin endocytosis.
Retrovirology
11
:
1
.

The authors have no financial conflicts of interest.