Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Biology

High-throughput Confocal Imaging of Quantum Dot-Conjugated SARS-CoV-2 Spike Trimers to Track Binding and Endocytosis in HEK293T Cells

Published: April 21, 2022 doi: 10.3791/63202

Summary

In this protocol, quantum dots conjugated to recombinant SARS-CoV-2 spike enable cell-based assays to monitor spike binding to hACE2 at the plasma membrane and subsequent endocytosis of the bound proteins into the cytoplasm.

Abstract

The development of new technologies for cellular fluorescence microscopy has facilitated high-throughput screening methods for drug discovery. Quantum dots are fluorescent nanoparticles with excellent photophysical properties imbued with bright and stable photoluminescence as well as narrow emission bands. Quantum dots are spherical in shape, and with the proper modification of the surface chemistry, can be used to conjugate biomolecules for cellular applications. These optical properties, combined with the ability to functionalize them with biomolecules, make them an excellent tool for investigating receptor-ligand interactions and cellular trafficking. Here, we present a method that uses quantum dots to track the binding and endocytosis of SARS-CoV-2 spike protein. This protocol can be used as a guide for experimentalists looking to utilize quantum dots to study protein-protein interactions and trafficking in the context of cellular physiology.

Introduction

Fluorescence microscopy enables researchers to peer into the inner workings of the cell using specialized dyes1, genetically encoded fluorescent proteins2, and fluorescent nanoparticles in the form of quantum dots (QDs)3. For the severe acute respiratory syndrome coronavirus of 2019 (SARS-CoV-2) global pandemic, researchers have employed fluorescence microscopy to understand how the virus interacts with the cell both at the plasma membrane and in the cytoplasm. For example, researchers have been able to gain insights into the binding of the SARS-CoV-2 Spike protein on the virion's surface to human angiotensin-converting enzyme 2 (hACE2) on the surface of human cells, subsequent internalization via fusion at the plasma membrane, and endocytosis of the Spike:hACE2 protein complex4,5. Great insights have also been gained into the SARS-CoV-2 egress from cells via the lysosome using cellular fluorescence imaging, a unique feature of coronaviruses previously thought to occur via traditional vesicle budding from the Golgi, as it is with many other viruses6. A mainstay of almost all aspects of biological research, the cellular fluorescence microscopy technique has necessarily advanced in its breadth and scope of applications from super-resolution imaging of whole animals to automated high-content multi-parametric imaging for drug screening. Here, automated high-content confocal microscopy is applied to the study of SARS-CoV-2 cell entry using fluorescent QDs conjugated to the viral spike protein.

High-content analysis of images generated by biological imaging platforms allows for greater extraction of valuable biological insights than single parameters such as whole-well intensity, that one would obtain using a multi-modal plate reader7. By separating the objects in a field of view using automated segmentation algorithms, each object or a population of objects can be analyzed for parameters such as intensity, area, and texture in each available fluorescence channel8. Combining many measurements into multivariate datasets is a useful approach for phenotypic profiling. When the desired phenotype is known, such as QD internalization in the form of puncta, one can use the measurements related to puncta such as size, number, and intensity to assess the efficacy of a treatment.

Cloud-based high content imaging analysis software can accommodate a large variety of instrument data outputs, including the high content imaging platform. By using a cloud-based server for image storage and online analysis, the user is able to upload their data from either the imaging instrument or from the network drive where the data is stored. The analysis portion of the protocol is conducted within the cloud software environment, and data can be exported in a variety of file formats for downstream data visualization.

The SARS-CoV-2 virus is composed of nonstructural and structural proteins that aid in its assembly and replication. SARS-CoV-2 spike has two domains called S1 and S2, with S1 containing the receptor-binding domain responsible for hACE2 interactions at the plasma membrane9. Spike has also been found to interact with other molecules at the plasma membrane that may act as co-receptors in addition to hACE210,11. Throughout the spike protein sequence and particularly at the S1/S2 interface, there are protease cleavage sites that enable fusion at the membrane after the transmembrane serine protease 2 (TMPRSS2)12. Various recombinant SARS-CoV-2 Spike proteins have been produced from individual receptor binding domains, to S1, S2, S1 with S2, and whole spike trimers from multiple commercial vendors for use in research activities13.

In this work, the surface of QDs was functionalized with recombinant spike trimers that contain a histidine tag (QD-Spike). The QDs produced by Naval Research Laboratory Optical Nanomaterials Section contain a cadmium selenide core and a zinc sulfide shell14,15. The zinc on the QD surface coordinates the histidine residues within the recombinant protein to form a functionalized QD that resembles a SARS-CoV-2 viral particle in form and function. The generation of the nanoparticles and protein conjugation was previously described using the QD-conjugated receptor binding domain15. This method describes the cell culture preparations, QD treatment, image acquisition, and data analysis protocol that can guide a researcher in studying SARS-CoV-2 Spike activity in the physiological context of a human cell.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

The HEK293T cell line used in this study is an immortalized cell line. No human or animal subjects were used in this study.

1. Cell culturing and seeding

  1. Inside a sterile biosafety cabinet, wearing personal protective equipment (including lab gloves, lab coat, and safety glasses), prepare cell culture medium by supplementing Dulbecco's Modified Eagle Medium (DMEM) with 10% fetal bovine serum (FBS), 1% penicillin/streptomycin (P/S), and 250 µg/mL G418.
    1. For 500 mL of media, add 443.75 mL of DMEM, 50 mL of FBS, 5 mL of P/S, and 1.25 mL of G418.
    2. Filter through a 0.2 µm filter flask and maintain sterility to avoid bacterial contamination.
  2. Inside a sterile biosafety cabinet, seed the interior 60 wells of a black, clear-bottom, poly-D-lysine coated 96-well plate with 20,000 cells in 100 µL per well of cell culture medium. Fill the outer 36 wells with 100 µL per well of phosphate-buffered saline (PBS).
    1. To count the cells, add 2 µL of acridine orange and propidium iodide stain to 18 µL of cell suspension.
    2. Load 10 µL of this solution into one side of a cell counting slide.
    3. Repeat steps 1.2.1. and 1.2.2. to load both sides of the slide.
    4. Place the slide into an automated cell counter.
    5. Select the fluorescence counting protocol and press Count. Count both sides of the slide and calculate the average of the two live-cell densities.
    6. To calculate the volume of cell suspension required, divide the total number of cells needed by the average cell density.
  3. Inspect the wells after seeding under a light microscope to ensure that proper density and distribution have been achieved.
  4. Incubate the plate overnight in a humidified incubator at 37 °C with 5% CO2.

2. Treatment of cells with QD-Spike

  1. Prepare 0.1% bovine serum albumin (BSA) by diluting in imaging media.
    1. For 10 mL of imaging media, add 130 µL of 7.5% BSA and mix.
  2. In a 12-well reservoir or assay plate, dilute the 440 nM QD-Spike (SARS-CoV-2, Isolate USA-WA1/2020) stock to 20 nM using 0.1% BSA in imaging media.
    1. For 1 well of QD-Spike, add 2.27 µL of 440 nM QD-Spike to 47.73 µL of 0.1% BSA imaging media for a final concentration of 20 nM.
    2. To generate a six-point 1:3 serial dilution (in triplicate), add 14.26 µL of QD-Spike to 285.74 µL of 0.1% BSA imaging media to make the highest concentration of 20 nM. Add 100 µL of 20 nM QD-Spike to 200 µL of 0.1% BSA media to make the second dilution of 6.67 nM QD-Spike. Repeat four times to generate the six concentration points.
  3. Using a multi-channel aspirator, remove all spent media from each well. Using a multi-channel pipette, wash once with imaging media (100 µL/well).
  4. Aspirate 100 µL of imaging media and add back 50 µL/well of QD-Spike solution.
  5. Incubate the plate for 3 h in a humidified incubator at 37 °C with 5% CO2. Continue to step 4.

3. Fixation and nuclei staining

  1. Inside a sterile biosafety cabinet, wearing personal protective equipment (including lab gloves, lab coat, and safety glasses), prepare 4% paraformaldehyde (PFA) in 0.1% BSA imaging media.
  2. Aspirate 50 µL of QD-Spike from each well and add 100 µL/well of 4% PFA.
    1. Do not let the wells dry out. Use an automated multi-channel pipette to avoid drying of the wells (recommended).
  3. Incubate for 15 min at room temperature.
  4. Wash three times with 1x phosphate-buffered saline (PBS).
  5. Prepare the deep red nuclear dye by diluting the 5 mM stock solution to 1:1000 dilution in PBS.
  6. Aspirate out the PBS and add back 50 µL/well of diluted nuclear dye.
  7. Incubate for 30 min at room temperature.
  8. Wash three times with PBS.
  9. Image the plate or seal using a plate sealer for imaging at a later date. Store the plate at 4 °C. Fluorescence should be stable for several weeks or more.

4. Acquisition set-up and imaging

  1. Start the software for the imaging platform. Turn on the high content imaging platform, and the light on the status bar should be on. If the machine is not connected, the software will go into offline analysis mode.
  2. Login into the imaging platform.
  3. Create a new acquisition protocol.
    1. Select Plate Type as 96-well clear bottom imaging plate.
      NOTE: The plate selection in the instrument may vary and can be customized by loading the correct plate definitions that include plate dimensions such as height, width, and other distances to define well spacing and focal points.
    2. Select Optical Mode as confocal and magnification as 40x water immersion.
    3. Select Binning as 1.
    4. Choose the channels and fluorescent excitation/emission wavelengths as described in steps 4.3.5-4.3.8. Take a snapshot when the settings are selected to view the image and ensure the settings are appropriate.
      NOTE: Digital Phase Contrast (DPC) will allow visualization of live cells without a cell stain or fluorescent dye. It is not recommended for fixed cells.
    5. Select the Mode for DPC such as High Contrast to produce well-defined cell bodies. Take a snapshot to confirm that this setting is appropriate, as seen in the image window.
    6. Select the FITC Channel for use with the ACE2-GFP cell line that allows visualization of ACE2 trafficking within the cell.
    7. To make a custom channel for the QD channel, select the triangle drop-down menu for the channel and choose the excitation in the 405 nm range and emission in the 608 nm range.
    8. If cells were fixed and a deep red nuclear dye was used, select the far-red channel with emission greater than 630 nm to further delineate the cell body and act as a cell mask.
    9. Select a well with a strong cytoplasmic QD signal to set the exposure time, laser power, and Z-height position. Follow steps from 4.3.10-4.3.13.
    10. Check whether the default height produces an image where the cells are in the desired focal plane. If the endocytosed puncta are the target organelle, the Z-position will be lower than if the plasma membrane is the target organelle.
    11. Choose an exposure time that produces a bright image with gray levels at least three-fold greater than the background signal. This is typically between 100 and 300 ms. At 20 nM, the gray levels should be approximately 6000 a.u. using an exposure time of 200 ms and laser power of 80%.
    12. Check gray levels by right-clicking on the image produced with the Snapshot feature in each channel and selecting Show Intensity. Then, left-click on the object of interest or background to view the gray level for that pixel.
    13. Adjust the laser power to fine-tune the intensity of the objects of interest.
  4. Save the acquisition protocol.
  5. Switch to Run Experiment and enter the Plate Name.
  6. Run the experiment.

5. Data analysis

  1. Import data onto the imaging software.
    1. After acquiring all images, export measurements to the imaging software. This requires a server to be established and an account to be created. Create a screen for the data to be exported into the imaging software.
  2. In the imaging software, select Image Analysis to begin building the analysis protocol.
  3. Load the measurements from the imaging experiment by right-clicking on the file in the screens list and clicking on Select. The plate map of imaged wells and their associated fields should be visible at the bottom left-hand corner of the window.
  4. Choose a well with bright cytoplasmic QD spots to begin image segmentation.
  5. In the Input Image Building Block, select Flatfield Correction.
  6. Add the next building block to the protocol by selecting Find Nuclei.
    1. Here, use the DPC or far-red channel as the Nuclei marker.
    2. Select the method that accurately segments the objects first, and then fine-tune the segmentation using the drop-down menu and adjusting the sliders.
      NOTE: Good segmentation accurately defines the region of interest (ROI) for the nucleus, cell, and spots. Only the pixels that are positive for the marker should be captured within an individual ROI. Background signal should be excluded. If the background is captured in the ROI, the sensitivity of the method can be decreased, or the threshold of intensity can be increased to increase the stringency of the segmentation algorithm. This is applicable to all Find building blocks.
  7. Next, add the Find Cytoplasm Building Block to identify the cytoplasm.
    1. In this case, use the ACE2-GFP channel for cytoplasmic segmentation.
    2. Select the method that best identifies the cytoplasm of each cell.
  8. Next, add the Find Spots Building Block to identify the QD-Spike puncta.
    1. Select the Nuclei as the ROI population.
    2. Select the Cell as the ROI region. This captures the puncta within the entire cell.
    3. Select the method that best identifies the QD puncta in each cell.
  9. Once all the objects in the image have been segmented and identified accurately in this well, choose other wells to ensure the building blocks and settings can be generally applied to the other wells and other conditions.
  10. Add the Calculate Intensity Properties for all segmented objects (nuclei, cytoplasm/cells, and spots).
  11. Add the Calculate Morphology Properties for all segmented objects (nuclei, cytoplasm/cells, and spots).
  12. Add the Calculate Texture Properties for all segmented objects (nuclei, cytoplasm/cells, and spots).
  13. Define results by selecting the parameters for each population, including nuclei and spots. The number of objects can be used as an indirect measure of cell viability.

6. Export data

  1. Click on Batch analysis. Wait for the analysis to finish before proceeding to the next step (step 6.2).
    NOTE: The batch analysis allows the image analysis software server to load the analysis protocol using the selected measurements to analyze the data remotely.
  2. Export the dataset to a local computer or network drive.
    1. Connect the image analysis software to a helper application on the computer by downloading and opening a connection file.
    2. Select the results file type (.txt, .csv, .html, or Native XML).
    3. Choose the File Folder settings with the drop-down menu.

7. Analyze the data in a spreadsheet

  1. Open the exported file. If it is a .csv file, save it as a .xls file format to enable the use of pivot tables. If the file path is too long, save the .xls file higher up in the folder hierarchy to avoid errors in saving.
  2. Add columns to the spreadsheet that designate the conditions for each well. For example, the cell type, QD-Spike variants, QD-Spike concentrations, incubation time, etc.
  3. Choose the Pivot Table function and build the table using the added conditions as Rows, and the measured parameters as Columns. Select the calculation for each parameter i.e., Average, Standard Deviation, Median, Min, Max, or Count.
  4. Normalize the data to control samples, including unconjugated QDs as 0% and the highest concentration of QD-Spike as 100%.
    NOTE: If assessing the efficacy of inhibitors such as neutralizing antibodies, normalize the data to media-only treated cells (100% efficacy) and QD-Spike without inhibitor (0% efficacy).
  5. Plot the data in graphing software.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

Upon treatment, the QDs will be internalized as the nanoparticle will bind to ACE2 on the plasma membrane and induce endocytosis. Using an ACE2-GFP expressing cell line, translocation of both QDs and ACE2 can be visualized using fluorescence microscopy. Once internalized, the two QD and ACE2 signals show strong colocalization. From these images, image segmentation and subsequent analysis can be performed to extract relevant parameters such as spot count (Figure 1, Figure 2B). Figure 1A is a montage of cells treated with different concentrations of QD-Spike or media only as a control. Three channels were used, including digital phase contrast, FITC, and the custom QD channel with excitation at 405 nm and emission at 608 nm. DPC provides an image of the general cell shape. The DPC image provides an approximate indication of the entire cell morphology. The area of interest overlaps with the DPC signal and is sufficient for detecting internalized QDs. The FITC channel shows the ACE2-GFP changing localization and accumulating at regions that colocalize with QD-Spike. As the concentration of QD-Spike decreases, the QDs are no longer visible and the ACE2-GFP signal is similar to control. The merge channel demonstrates the colocalization of ACE2-GFP with QD-Spike. These objects are a mixture of spots and larger accumulations that can be segmented with the analysis software. Fine-tuning of the software analysis method used for image segmentation can be used to segment the objects of interest.

Here a concentration-response experiment with six different concentrations of QD-Spike was performed, starting at 20 nM, to determine optimal concentrations of QD (Figure 2A). QDs can be used as low as 2.22 nM and still show binding and internalization. However, it is recommended to use concentrations of 20 nM or higher to ensure a robust response.

During conjugation to the QD, protein aggregation may occur. The main issue with aggregates is that they will accumulate on top of cells and cause artifacts in the image segmentation step. Aggregates will not be able to enter cells, and the nanoparticles as a whole will no longer resemble a viral particle (Figure 3). Aggregates can be spotted in the QD solution as bright, clumped precipitates.

This assay can also be used to assess biologics, such as neutralizing antibodies that block viral entry. QDs were incubated with neutralizing antibodies (Figure 4A), starting at 30 µg/mL, for 30 min at room temperature before addition to cells. Antibodies raised against the reference Washington strain, SARS-CoV-2 RBD, were used. They blocked binding, internalization, and caused a reduction in the measured spot count compared to cells treated with QD-only (Figure 4B).

Figure 1
Figure 1: High-content image analysis and segmentation of ACE2-GFP cells treated with QD608-Spike. Representative images of accurate segmentation. First, nuclei are segmented from the channel containing the nuclear marker to create an ROI population. From the ROI population, an ROI region outlining the cell is segmented using the ACE2-GFP channel. Lastly, QD608-Spike spots are segmented from the Cell ROI region. Please click here to view a larger version of this figure.

Figure 2
Figure 2: QD608-Spike binds hACE2 and is endocytosed in hACE2-GFP HEK293T cells. (A) Image montage of hACE2-GFP (yellow) HEK293T cells treated with several concentrations of QD608-Spike (magenta) or media-only. Digital phase contrast (cyan) was used to visualize the cell body. Scale bar: 20 µm. (B) High content analysis of QD608-Spike Spot Counts normalized to 20 nM QD608-Spike (100%) and the control (0%). N = triplicate wells. Error bars indicate standard deviation (S.D.). Please click here to view a larger version of this figure.

Figure 3
Figure 3: Aggregated QD608-Spike is not internalized into hACE2-GFP HEK293T cells. Image montage of hACE2-GFP HEK293T cells treated with 10 nM QD608-Spike or media-only. Scale bar: 20 µm. Please click here to view a larger version of this figure.

Figure 4
Figure 4: Neutralizing antibody blocks endocytosis of QD608-Spike in hACE2-GFP HEK293T cells. (A) Image montage of hACE2-GFP (yellow) HEK293T cells treated with QD608-Spike (magenta) preincubated with decreasing concentrations of neutralizing antibodies, using digital phase contrast (cyan) to identify cell bodies. Scale bar: 20 µm. (B) High content analysis of QD608-Spike Spot Counts normalized to media-only control (100%) and 10 nM QD608-Spike only (0%). N = triplicate wells. Please click here to view a larger version of this figure.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

The method described in this article provides the necessary steps for imaging functionalized QDs in human cells using high-throughput confocal microscopy. This method is best suited for cells where endocytosis is the main route of viral entry rather than the activity of TMPRSS2 and membrane fusion, as it enables the study of SARS-CoV-2 Spike and hACE2 endocytosis. Because of the nature of the QD model and the C-terminal His-tag on the commercially available Spike trimer, any TMPRSS2 cleavage of Spike S1 and S2 domains would leave the QD attached to the S2 domain only12. This may prevent internalization, given that the RBD is found in S1. Therefore, if the sequence of events at the cell surface were precise, where hACE2 is bound and then TMPRSS2 cleaves Spike, a negative signal with no internalization is expected.

As this protocol deals with imaging cellular processes, the cultured cells must be permissive to viral infection with the expression of hACE2. hACE2 may be transiently transfected into cells, or a stable cell line expressing hACE2 may be generated17. It is recommended to verify hACE2 expression and localization using immunofluorescence with antibodies against hACE2 unless the hACE2 has a fluorescent protein tag such as GFP that can be observed using microscopy. GFP-tagged hACE2 confers the added benefit of visualizing hACE2 trafficking following QD-Spike endocytosis. Some cell lines with endogenous hACE2 expression may be used, but this should be confirmed using immunocytochemistry. In some cases, the endogenous expression does not provide enough hACE2 binding partners for QD-Spike and may result in a signal that is poorly detected by high-throughput confocal microscopy.

One critical step in the protocol includes the procurement of high-quality QDs conjugated to the recombinant His-tagged SARS-CoV-2 Spike. The prerequisite for this is a properly purified protein that can be conjugated to the QDs without causing aggregation. Aggregated QDs will have several problems that prevent a successful experiment. Therefore, testing of QD-Spike using analysis tools (e.g., UV-visible spectroscopy, transmission electron microscopy, or dynamic light scattering) prior to full experimentation is highly recommended to preserve valuable resources if testing precious reagents16. During the labeling of QDs with spike, specific concentrations of QD and Spike are mixed to achieve a specific ratio of molecules. Only QD and Spike are mixed, and both solutions are highly purified. To assess the labeling of QDs, an acrylamide gel can be cast and loaded with QD alone as well as QD-conjugated to Spike, which results in heavier molecular weight bands.

The QDs used in this protocol have a fluorescence excitation/emission spectrum tuned to 608 nm emission following UV excitation (≤405 nm) since most QDs have high absorption near the UV range producing high photoluminescence18. This is an unconventional excitation/emission combination that requires a microscope to have customizable channels. Many traditional confocal microscopes can be set up with the right filters and laser lines to achieve this excitation/emission. Alternatively, excitation of the QD at the first absorption maximum, approximately 10 to 20 nm away from the emission peak (e.g., 592 nm for QD608 used here), will also be able to produce sufficient photoluminescence.

The cloud-based software used in this high-content analysis protocol uses a naming scheme that builds on previous steps. For example, the first objects that are segmented are nuclei, which create a population called Nuclei. Following this, the cell or cytoplasm can be identified as an ROI region within the population Nuclei. The terminology used in the image analysis software sets the name of the population of objects segmented using the nucleus building block as Nuclei. However, they do not necessarily have to be nuclei and can be cell bodies if no nuclear dye is available. This naming scheme can also be changed and customized within each building block output name.

Our protocol did not account for membrane interactions, but this could be done by adding an additional membrane stain that is independent of ACE2-GFP trafficking. To block nonspecific binding, 0.1% BSA is included in the assay media (DMEM + 0.1% BSA), and the cells are grown in 10% FBS as well. Mutant spike proteins could be used, but we were limited to commercially available spike proteins. In this case, a SARS-CoV-2 mutant non-ACE2 binding Spike protein was not available.

The QD nanoparticle method presents a powerful technology to study the binding and internalization of viruses that rely on Spike-mediated entry. This assay can be used for high-throughput screening in an analogous manner, as shown in Figure 4, to repurpose and identify potent antivirals that block cellular entry. While this protocol only demonstrated the use of QDs conjugated to the Washington WA-1 reference strain of SARS-CoV-2, this assay can be easily adapted to study the binding properties of newly emerging variants such as Alpha, Beta, Gamma, and Delta through the use of their respective spike proteins conjugated to QDs.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have no conflicts of interest to disclose.

Acknowledgments

This research was supported in part by the Intramural Research Program of the National Center for Advancing Translational Sciences, NIH. Naval Research Laboratory provided funding via its internal Nanoscience Institute. Reagent preparation was supported via the NRL COVID-19 base fund.

Materials

Name Company Catalog Number Comments
32% Paraformaldehyde Electron Microscopy Sciences 15714 Used for fixing cells after quantum dot treatment, final concentration 3.2%
Used for stabilizing QDs in Optimem I and preventing non-specific interactions, final concentration 0.1%
7.5% Bovine Serum Albumin Gibco 15260-037 Used as a cell viability dye for fluorescence cell counting
Acridine Orange / Propidium Iodide Stain Logos Biosystems F23001 Microwell plates used for seeding cells and assaying QD-Spike
Black clear bottom 96 well coated plate coated with poly-D-lysine Greiner 655946 Used to support cell culture, DMEM supplement
Characterized Fetal Bovine Serum Cytiva/HyClone SH30071.03 Cloud-based high-content image analysis software; V2.9.1
Columbus Analyzer Perkin Elmer NA Used for labeling cell nuclei and cell bodies after fixation, deep red nuclear dye
DRAQ5 (5 mM) ThermoFisher Scientific 62252 Basal media for HEK293T cell culture
Dulbecco's Minimal Essential Media, D-glucose (4.5g/L), L-glutamine, sodium pyruvate (110 mg/L), phenol red Gibco 11995-065 Used for arranging data after export from Columbus; V2110 Microsoft 365
Excel Microsoft NA Used to continue selection of hACE2-GFP positive cells, DMEM supplement
G418 InvivoGen ant-gn-5 Human embryonic kidney cell line stably expression human angiotensin converting enzyme 2 tagged with GFP
HEK293T hACE2-GFP Codex Biosolutions CB-97100-203 Automated cell counter
Luna Automated Cell Counter Logos Biosystems NA Used for fluorescence cell counting
Luna Cell Counting Slides Logos Biosystems L12001 High-content imaging platform
Opera Phenix Perkin Elmer NA Imaging media, used for incubating cells with quantum dots
Opti-MEM I Reduced Serum Medium Gibco 11058-021 Phosphate-buffered saline without calcium or magnesium used for washing cells during passaging and assaying
PBS -/- Gibco 10010-023 Used to prevent bacterial contamination of cell culture, DMEM supplement
Penicillin Streptomycin Gibco 15140-122 Used for graphing, data visualization, and statistical analysis;V9.1.0
Prism GraphPad NA Used for assaying SARS-Cov-2 Spike binding to hACE2 and monitoring Spike endocytosis
Quantum Dot 608 nm-Spike (QD608-Spike) custom made by Naval Research Laboratory Used for inhibition of SARS-Cov-2 Spike binding to hACE2
SARS-CoV-2 (2019-nCoV) Spike Neutralizing Antibody, Mouse Mab Sino Biological 40592-MM57 Used to dissociate cells from flask during passaging
TrypLE Express Gibco 12605-010

DOWNLOAD MATERIALS LIST

References

  1. Chazotte, B. Labeling Lysosomes in Live Cells with LysoTracker. Cold Spring Harbor Protocols. 2011 (2), 5571 (2011).
  2. Mehta, S., Zhang, J. Biochemical activity architectures visualized-using genetically encoded fluorescent biosensors to map the spatial boundaries of signaling compartments. Accounts of Chemical Research. 54 (10), 2409-2420 (2021).
  3. Barroso, M. M. Quantum dots in cell biology. The Journal of Histochemistry and Cytochemistry: Official Journal of the Histochemistry Society. 59 (3), 237-251 (2011).
  4. Cuervo, N. Z., Grandvaux, N. ACE2: Evidence of role as entry receptor for SARS-CoV-2 and implications in comorbidities. eLife. 9, 61390 (2020).
  5. Shang, J., et al. Cell entry mechanisms of SARS-CoV-2. Proceedings of the National Academy of Sciences of the United States of America. 117 (21), 11727-11734 (2020).
  6. Ghosh, S., et al. β-Coronaviruses use lysosomes for egress instead of the biosynthetic secretory pathway. Cell. 183 (6), 1520-1535 (2020).
  7. Buchser, W., et al. Assay development guidelines for image-based high content screening, high content analysis and high content imaging. Assay Guidance Manual. Markossian, S., et al. , Eli Lilly & Company and the National Center for Advancing Translational Sciences. (2012).
  8. Chandrasekaran, S. N., Ceulemans, H., Boyd, J. D., Carpenter, A. E. Image-based profiling for drug discovery: due for a machine-learning upgrade. Nature Reviews. Drug Discovery. 20 (2), 145-159 (2021).
  9. Huang, Y., Yang, C., Xu, X. F., Xu, W., Liu, S. W. Structural and functional properties of SARS-CoV-2 spike protein: potential antivirus drug development for COVID-19. Acta Pharmacologica Sinica. 41 (9), 1141-1149 (2020).
  10. Gao, C., et al. SARS-CoV-2 spike protein interacts with multiple innate immune receptors. bioRxiv: the preprint server for biology. , 227462 (2020).
  11. Zhang, Q., et al. Heparan sulfate assists SARS-CoV-2 in cell entry and can be targeted by approved drugs in vitro. Cell Discovery. 6 (1), 80 (2020).
  12. Hoffmann, M., et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and Is blocked by a clinically proven protease inhibitor. Cell. 181 (2), 271-280 (2020).
  13. Cai, Y., et al. Distinct conformational states of SARS-CoV-2 spike protein. Science. 369 (6511), New York, N.Y. 1586-1592 (2020).
  14. Oh, E., et al. Meta-analysis of cellular toxicity for cadmium-containing quantum dots. Nature Nanotechnology. 11 (5), 479-486 (2016).
  15. Gorshkov, K., et al. Quantum dot-conjugated SARS-CoV-2 spike pseudo-virions enable tracking of angiotensin Converting enzyme 2 binding and endocytosis. ACS Nano. 14 (9), 12234-12247 (2020).
  16. Narayanan, S. S., Pal, S. K. Aggregated CdS quantum dots: Host of biomolecular ligands. The Journal of Physical Chemistry B. 110 (48), 24403-24409 (2006).
  17. Wang, S., et al. Endocytosis of the receptor-binding domain of SARS-CoV spike protein together with virus receptor ACE2. Virus Research. 136 (1), 8-15 (2008).
  18. Hildebrandt, N., et al. Energy transfer with semiconductor quantum dot bioconjugates: A versatile platform for biosensing, energy harvesting, and other developing applications. Chemical Reviews. 117 (2), 536 (2017).

Tags

High-throughput Confocal Imaging Quantum Dot-Conjugated SARS-CoV-2 Spike Trimers Binding Endocytosis HEK293T Cells Platform Technology Spike Mediated Cell Recognition Cellular Viral Infection Quantum Dots Functionalized Surface Visualize Measure Internalization BSA Serial Dilution Triplicate Concentration Nanomole Multi-channel Aspirator Multi-channel Pipette Imaging Media Incubator Carbon Dioxide Protective Equipment PFA
High-throughput Confocal Imaging of Quantum Dot-Conjugated SARS-CoV-2 Spike Trimers to Track Binding and Endocytosis in HEK293T Cells
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Tran, B. N., Oh, E., Susumu, K.,More

Tran, B. N., Oh, E., Susumu, K., Wolak, M., Gorshkov, K. High-throughput Confocal Imaging of Quantum Dot-Conjugated SARS-CoV-2 Spike Trimers to Track Binding and Endocytosis in HEK293T Cells. J. Vis. Exp. (182), e63202, doi:10.3791/63202 (2022).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter