supplementary materials for - science advances...checked the ability of gag to induce pi(4,5)p2...

15
advances.sciencemag.org/cgi/content/full/5/10/eaaw8651/DC1 Supplementary Materials for HIV-1 Gag specifically restricts PI(4,5)P2 and cholesterol mobility in living cells creating a nanodomain platform for virus assembly C. Favard, J. Chojnacki, P. Merida, N. Yandrapalli, J. Mak, C. Eggeling*, D. Muriaux* *Corresponding author. Email: [email protected] (D.M.); [email protected] (C.E.) Published 2 October 2019, Sci. Adv. 5, eaaw8651 (2019) DOI: 10.1126/sciadv.aaw8651 The PDF file includes: Sections S1 and S2. Characterization of fluorescent lipid analogs Section S3. MHC-I mobility at HIV-1 assembly sites in NL4.3 Gag-iGFP HIV-1–infected Jurkat T cells Section S4. STED microscope calibration Section S5. PI(4,5)P2 clustering and trapping by full-length HIV-1 Gag on biomimetic membranes Section S6. Budding efficiency comparison in Jurkat T cells transfected with Gag.eGFP or with a mixture of Gag.eGFP/Gag (ratio, 1:3) Section S7. Cumulative frequency distributions observed in infected cells for the different lipids Section S8. Cumulative frequency distributions observed in transfected cells for the different lipids Fig. S1. Fluorescent lipid analog structures. Fig. S2. Fluorescent lipid analog mobility in Jurkat T cells. Fig. S3. MHC-I mobility at HIV-1 assembly sites in NL4.3 Gag-iGFP HIV-1–infected Jurkat T cells. Fig. S4. STED microscope calibration results. Fig. S5. Changes in the lateral mobility of ATTO647N-PI(4,5)P2 upon addition of Gag on SLBs. Fig. S6. Jurkat T cell coelectroporation with Gag.eGFP and Gag plasmids. Fig. S7. Cumulative frequency distributions of diffusion coefficient observed in HIV-1–infected T cells. Fig. S8. Cumulative frequency distributions of diffusion coefficient observed in HIV-1 Gag- transfected T cells. Legend for movie S1 References (62, 63)

Upload: others

Post on 10-Feb-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

  • advances.sciencemag.org/cgi/content/full/5/10/eaaw8651/DC1

    Supplementary Materials for

    HIV-1 Gag specifically restricts PI(4,5)P2 and cholesterol mobility in living cells

    creating a nanodomain platform for virus assembly

    C. Favard, J. Chojnacki, P. Merida, N. Yandrapalli, J. Mak, C. Eggeling*, D. Muriaux*

    *Corresponding author. Email: [email protected] (D.M.); [email protected] (C.E.)

    Published 2 October 2019, Sci. Adv. 5, eaaw8651 (2019) DOI: 10.1126/sciadv.aaw8651

    The PDF file includes:

    Sections S1 and S2. Characterization of fluorescent lipid analogs Section S3. MHC-I mobility at HIV-1 assembly sites in NL4.3 Gag-iGFP HIV-1–infected Jurkat T cells Section S4. STED microscope calibration Section S5. PI(4,5)P2 clustering and trapping by full-length HIV-1 Gag on biomimetic membranes Section S6. Budding efficiency comparison in Jurkat T cells transfected with Gag.eGFP or with a mixture of Gag.eGFP/Gag (ratio, 1:3) Section S7. Cumulative frequency distributions observed in infected cells for the different lipids Section S8. Cumulative frequency distributions observed in transfected cells for the different lipids Fig. S1. Fluorescent lipid analog structures. Fig. S2. Fluorescent lipid analog mobility in Jurkat T cells. Fig. S3. MHC-I mobility at HIV-1 assembly sites in NL4.3 Gag-iGFP HIV-1–infected Jurkat T cells. Fig. S4. STED microscope calibration results. Fig. S5. Changes in the lateral mobility of ATTO647N-PI(4,5)P2 upon addition of Gag on SLBs. Fig. S6. Jurkat T cell coelectroporation with Gag.eGFP and Gag plasmids. Fig. S7. Cumulative frequency distributions of diffusion coefficient observed in HIV-1–infected T cells. Fig. S8. Cumulative frequency distributions of diffusion coefficient observed in HIV-1 Gag-transfected T cells. Legend for movie S1 References (62, 63)

  • Other Supplementary Material for this manuscript includes the following: (available at advances.sciencemag.org/cgi/content/full/5/10/eaaw8651/DC1)

    Movie S1 (.avi format). Drift stabilized time lapse movie of a representative NL4.3 Gag-iGFP HIV-1–infected Jurkat T-cell showing already present and newly developing virus assembly sites.

  • Supplementary Materials Sections S1 and S2. Characterization of fluorescent lipid analogs Scanning STED FCS requires dedicated lipid analogs with photostable and bright dye labels

    such as Atto647N or KK114. Figure S1 shows the structures of the lipid analogs used throughout the present study. These analogs have been used multiple times before and in their

    cases the dye label shown to have minimal influence on the lipids’ membrane interaction

    dynamics (34–37) as confirmed by using classical spot FCS (fig. S2), highlighting that the

    mobility of these lipid analogs in CD4+ T-cells was comparable between different dye labels,

    specifically to the mobility observed with corresponding BODIPY labelled lipids that we

    previously used to monitor the effect of Gag self-assembly on model membranes (10)

    Fig. S1. Fluorescent lipid analog structures. From left to right, structures of all the fluorescent sterols and lipid analogs used in this study, a- KK114 Chol, b- TopFluor Cholesterol, c- Bodipy TMR C16 PI(4,5)P2, d- Atto647N- PI(4,5)P2, e- TopFluor PI(4,5)P2, f- TopFluor SM, g- Atto647N SM, h-Atto647N-PE, i- KK114-PE.

  • Fig. S2. Fluorescent lipid analog mobility in Jurkat T cells. a – Transmission (upper part) and fluorescent (lower part) image of Jurkat T-cells labelled with Atto647N-SM or KK114-

    Chol (scalebar=5 µm). b- Examples of normalized correlograms and their corresponding fit using equation 1 obtained in the labelled Jurkat T-cells for KK114-Chol (red line) and

    Atto647N-SM (black line). c- Box-plot of the apparent diffusion coefficients observed in Jurkat T-cells labelled with different lipid analogs for PI(4,5)P2 (red boxes), Cholesterol

    (green boxes) and sphingomyelin (blue boxes) and measured using confocal spot-FCS (empty

    boxes) and compared to scanning confocal FCS (diagonal pattern filled boxes).

  • Section S3. MHC-I mobility at HIV-1 assembly sites in NL4.3 Gag-iGFP HIV-1–infected Jurkat T cells In order to further reinforce the idea that our experiments are not observing fully

    assembled/budded viruses on the cell membrane, we have performed an experiment

    measuring MHC-I mobility at HIV-1 virus assembly sites in infected Jurkat T-cells. For the

    observation of MHC-I mobility, NL4.3 Gag-iGFP infected cells were stained for MHC-I in

    suspension at 16 °C using W6/32 anti-MHC-I Fab fragments and anti-human Abberior STAR

    RED (KK114) conjugated Fab fragments for 1 h each in 0.5% BSA/L-15 Medium (3).

    Mobility analysis inside and outside selected virus assembly sites shows that there is no

    significant MHC-I mobility change at these sites compared to areas outside and uninfected

    control cells. In a previous study (3), we shown that MHC-I mobility is highly reduced on

    fully assembled and budded viruses (Dmedian = 0.0027 μm2/s). These results indicate that virus

    assembly sites analysed in this study do not represent fully assembled and budded virus

    particles. Small mobility reduction seen between inside and outside assembly site areas may

    be due to the early stages of the virus assembly beginning to affect MHC-I diffusion in these

    areas.

  • Fig. S3. MHC-I mobility at HIV-1 assembly sites in NL4.3 Gag-iGFP HIV-1–infected Jurkat T cells. Median MHC-I diffusion coefficient (D) was determined by sSTED-FCS measurements of 20 sites each from two independent virus preparations and infections. Box

    and whisker plots (horizontal line – median, box – 25-75 % percentiles or interquartile range

    (IQR) and whiskers – 10-90 % measurements) shows D values inside assembly sites (red),

    outside assembly sites (green) and in non-infected control cells (blue). Statistical significance

    was assessed by Wilcoxon rank-sum test.

  • Section S4. STED microscope calibration

    The diameters of the observation spots employed in the scanning STED-FCS was estimated at

    37 °C following a standard calibration procedure on supported lipid bilayers (SLBs) (61),

    comparing transit times of freely diffusing fluorescent lipid (KK114-DPPE) in confocal and

    STED microscopy modes (fig. S4).

    Fig. S4. STED microscope calibration results. (a) Mean ± SD of observation spot diameters at different STED laser powers was determined by STED-FCS calibration measurements of

    KK114-DPPE diffusion in supported lipid bilayers (SLBs). Results represent 10

    measurements each from 2 separate preparations. For the virus assembly sites measurements,

    a minimum STED laser power (90 mW) was set to generate an observation spot diameter (100

    nm) below the diameter of HIV-1 particles (≈ 140 nm diameter) and virus assembly sites,

    while maintaining as high as possible signal-to-noise ratio and minimising the laser light

    exposure of live Jurkat T cells.

  • Section S5. PI(4,5)P2 clustering and trapping by full-length HIV-1 Gag on biomimetic membranes

    Yandrapalli et al.(10) previously reported the ability of purified full length Gag to segregate

    PI(4,5)P2 and cholesterol while self-assembling on supported lipid bilayers (SLB). Here, we

    checked the ability of Gag to induce PI(4,5)P2 clustering and trapping on SLBs with the basic

    lipid composition described in Yandrapalli et al.(10) by imaging the change in ATTO647N-

    PI(4,5)P2 fluorescence before and after addition of Gag on SLBs (fig. S2a and movie S1). As

    previously reported, we observed the generation of ATTO647N-PI(4,5)P2 enriched domains

    few minutes after injection of recombinant full length Gag (fig. S5a). Concomitantly, confocal

    line scanning FCS (sFCS) was performed on SLBs in order to monitor the changes in lateral

    PI(4,5)P2 mobility induced by Gag self-assembly. We first measured the lateral diffusion of

    ATTO647N-PI(4,5)P2 before Gag injection (fig. S5b, blue box) and compared it to the lateral

    diffusion observed after Gag injection inside (fig. S5b, red box) and outside (fig. S5b, green

    box) the generated Gag aggregation sites. We observed a significant decrease in ATTO647N-

    PI(4,5)P2 lateral mobility at (Da = 0.15 µm2.s-1) compared to outside (Da = 1.1 µm2.s-1) the

    sites or before the addition of Gag (Da = 0.95 µm2.s-1), highlighting strong trapping of

    ATTO647N-PI(4,5)P2 in lipid domains generated by Gag.

    We then applied the confinement index calculation to determine the enrichment of

    ATTO647N-PI(4,5)P2 in Gag induced clusters. We obtained values in line with either HIV-1

    infected cells or HIV-1 Gag.eGFP transfected cells. (Figs. 2 and 3)

  • Fig. S5. Changes in the lateral mobility of ATTO647N-PI(4,5)P2 upon addition of Gag on SLBs. (a) Confocal images of PC/PS/PI(4,5)P2 supported lipid bilayers labelled with ATTO647N-PI(4,5)P2 before (upper part) and 3 minutes after addition of the full length HIV-

    1 Gag protein. (b) Box plot of the diffusion coefficients of ATTO647N-PI(4,5)P2 observed before addition of Gag (blue) and 3 minutes after addition of Gag inside (red) or outside

    ATTO647N-PI(4,5)P2 clusters (green), (boxes are median, first and third quartile and 10-90%

    whiskers). Values of the confinement indices of PI(4,5)P2 analog in SLB for the three

    different diffusion regimes (slow Da < 0.1 µm².s-1, intermediates 0.1 < Da < 1µm².s-1) and fast

    (Da > 1 µm².s-1). As in the case of infected and transfected cells, ATTO647N-PI(4,5)P2 is

    strongly enriched in the clusters observed 3 min after addition of Gag.

  • Section S6. Budding efficiency comparison in Jurkat T cells transfected with Gag.eGFP or with a mixture of Gag.eGFP/Gag (ratio, 1:3) We attempted to transfect Jurkat T cells, using electroporation, with a mixture of tagged and

    untagged Gag expressing plasmids, similarly to studies done on highly transfectable adherent

    cell lines, such as HeLa or 293T cells (see Jouvenet et al., (62) and Ivanchenko et al., (32)),

    but never on CD4 T cells. When electroporated with 2 plasmids we observed, using anti-CA

    (p24) immunoblots, that one of the plasmids is better expressed than the other: here, it is the

    pGag plasmid – producing unlabelled Gag proteins (fig. S6a). Moreover, with a ratio of 1 to 3

    Gag.eGFP/Gag expressing plasmids we were not able to detect GFP(+) cells producing

    fluorescent VLPs and none of them were producing detectable virus assembly sites. This lack

    of production of fluorescent dots (virus assembly) at their cell surface thus prevents us from

    doing STED-FCS in and out of the virus assembly sites. In these conditions Gag.eGFP stays

    diffuse in the cytosol even 40h post-transfection, indicating probably that the unlabelled Gag

    protein is assembling preferentially, leaving the Gag.eGFP poorly incorporated into the viral

    budding site (fig. S6b). This data indicates that we are unable to do a plasmid mixture

    experiments on electroporated Jurkat T cells.

    Nonetheless, we believe that Gag.eGFP only experiment is still useful in the context of our

    study. This is because, while constructs such as pKHIV.eGFP or NL43.eGFP indeed require a

    rescue (50% of untagged Gag) to get a fully infectious virus, early studies using these

    constructs suggest that the infectivity defects in Gag.eGFP-only particles arise from incorrect

    Gag processing (maturation) following assembly rather than Gag assembly itself (63).

    Furthermore, in a recently study (33), we have shown that in Jurkat T cells expressing 100%

    mEOS2-tagged(i)Gag, we were able to monitor at the single molecule level, the kinetic of

    Gag assembly and it was found to resemble the one described in HeLa cells (5 min of

    assembly + 10 min of particle residence at the cell membrane and release).

    We would also like to emphasise that similar lipid mobility results were also obtained using

    fully infectious NL43 Gag.iGFP plasmid. This plasmid is described in the study of Hubner et

    al. (31), which characterize fully infectious NL4.3 Gag.iGFP virus particles. Although the

    authors do not comment on the assembly and release competency of the particles they indicate

    that the construct and expression of the proviral clone HIV Gag-iGFP, with the GFP inserted

    between MA and CA and with 2 protease cleavage sites have normal expression, viral

    production and infectivity, very close to wild-type HIV-1. The authors show that NL4.3

  • Gag.iGFP retains full infectivity as wild type and that the distribution of Gag on cells surface

    is the same (by confocal microscopy). They also indicate correct assembly/budding pathway.

    This make us confident that even if maturation and infectivity defects exist when using 100%

    GFP-tagged Gag, this tagged protein is still useful for our measurements which target only an

    early assembly phase.

  • Fig. S6. Jurkat T cell coelectroporation with Gag.eGFP and Gag plasmids. (a) Jurkat T cells were electroporated with empty plasmid (mock), GFP, Gag.eGFP or a mixture of

    Gag.eGFP+Gag (ratio 1:3) expressing plasmids and analysed 40 hours post-electroporation,

    as seen in images using low magnification fluorescence microscopy. (b) A Western blot anti-CAp24 of the electroporated Jurkat T cells in the different conditions. It can be seen that in

    the mixture condition, Gag is highly expressed as compared to Gag.eGFP, lowering the

    probability to get a T cell producing GFP(+) viruses. (c) Confocal microscopy images of electroporated Jurkat T cells. Cells were rinsed, resuspended in L15 transparent media,

    adhered on Poly-L coverslip at 37°C for 1h and imaged by confocal microscopy. A typical

    living GFP(+) T cell is shown for the condition Gag.eGFP and for the condition

    Gag.eGFP+Gag (ratio 1:3) as indicated. Results demonstrate that, viral assembly sites

    produced by the T cell can be observed only in the condition with Gag.eGFP only. It is never

    the case for Jurkat Tcells electroporated with the mixture Gag.eGFP/Gag. LUT scale:

    Blue/purple: low fluorescence intensity, yellow: high fluorescence intensity.

  • Section S7. Cumulative frequency distributions observed in infected cells for the different lipids

    Fig. S7. Cumulative frequency distributions of diffusion coefficient observed in HIV-1–infected T cells. Cumulative frequency distributions of values of apparent diffusion coefficients Da measured inside (red) and outside (blue) assembly sites of infected cells, and

    in non-infected cells (green) for the different fluorescent lipid analogs, ATTO647N-PI(4,5)P2

    (a), Cholesterol (Chol-PEG-KK114) (b), sphingomyelin (ATTO647N-SM) (c) and DPPE analogs (d).

  • Section S8. Cumulative frequency distributions observed in transfected cells for the different lipids

    Fig. S8. Cumulative frequency distributions of diffusion coefficient observed in HIV-1 Gag-transfected T cells. Cumulative frequency distributions of the Da values measured inside (red) and outside (blue) assembly sites of transfected cells, and in non-transfected cells

    (green) for the different lipids, ATTO647N-PI(4,5)P2 (a), Cholesterol (Chol-PEG-KK114) (b), sphingomyelin (ATTO647N-SM) (c) and DPPE analogs (d).

  • Movie S1. Drift stabilized time lapse movie of a representative NL4.3 Gag-iGFP HIV-1–infected Jurkat T-cell showing already present and newly developing virus assembly sites. Timer indicates minutes post cell adherence and LUT: blue-green-yellow from lowest to highest fluorescent signal intensity.

    aaw8651_SMaaw8651_SupplementalMaterial_v2