Antibody Validation for Flow Cytometry

By Guest Blogger

Antibody validation is to confirm (or refute) that the antibody is selectively detecting the target-of-interest in your assay and sample-of-interest. The approaches available broadly map onto the five pillars of antibody validation (see: Uhlen et al., 2016). In this post, we will describe the approaches that can be used to determine selectivity of an antibody for flow cytometry experiments. This will include a discussion of the Human Leucocyte Differentiation Antigens (HCDM) workshop approach, which focuses on validating antibodies against markers of the human blood leukocyte populations, as well as a pragmatic approach to determining the selectivity of antibodies for other sample types and for non-surface (intracellular) antigens.

Unfortunately, antibodies frequently do not work as assumed (Ayoubi et al., 2023). Additionally, their performance depends on the specific protocol used and the abundance of the epitope in the sample, relative to cross-reactive antigens – that is, an antibody shown to perform well in one set of conditions may not perform as well in other conditions. For these reasons, it is essential to test the selectivity of your antibody. But testing is not one size fits all. The approaches used will be different depending on the sample and target type.

To start the validation process, one must first confirm that a candidate antibody can detect the antigen of interest in the given protocol. This requires use of one of the five pillars of antibody validation, and it is recommended to screen multiple candidate antibodies when possible. If cost is prohibitive, consider an approach that prioritizes more reproducible/renewable reagents, such as recombinant or hybridoma-derived monoclonal antibodies. It is also important to seek out supportive data on candidate antibodies from the manufacturer and/or in the published literature. Note that the antibody being published doesn’t always indicate the presence of supportive data. To be supportive, the data must show that the antigen being detected in the intended application — here, flow cytometry — is the desired target, using one or more of the approaches outlined below.

Subscribe to the Addgene blog

Knockout cell line approach

Genetic knockout-based validation can conclusively prove the ability of an antibody to detect the antigen of interest when expressed at endogenous levels (Laflamme et al., 2019). CRISPR-Cas9 approaches are commonly used to produce KO cell lines, some of which are commercially available. Various proteomic and transcriptomic datasets can be used to select a candidate cell line (e.g. DepMap) – you’ll need to ensure that the parental cell lines of your chosen KO line are likely to express the target at a robust level, similar to that in samples of interest. If you are working with cells that are amenable to genetic knockout, this represents a very robust approach to antibody validation. If your protein of interest is essential to the cell’s function, it likely is not amenable to knockout approaches. In these cases, we would recommend a knockdown approach instead.

In this approach, you’re looking for an antibody that detects the target in the parental cell line, but not in the iso-genic knockout control. This can involve cell surface labeling and/or intracellular labeling (or both). It is important to test for the protocol you intend to use in your experiment. An antibody that demonstrates selectivity for cell surface labeling may become less selective if used in intracellular labeling because the intracellular compartment may contain cross-reacting antigens not present on the cell surface. The presentation of antigens can also be affected by cell fixation and permeabilization techniques  and so it may be necessary to try multiple techniques.

For example, we have found that some antibodies perform well in a PFA-saponin fixation and permeabilization protocol, but very poorly in a methanol fixation-permeabilization protocol. Although the expected location of the target is an important factor in deciding which protocol is likely to work, we have found that it is often best to determine this by testing them. For intracellular targets, we usually test 3 different fix/perms (methanol, PFA-saponin, PFA-triton). The fix/perm solutions can make a significant difference in the efficacy of your antibody. When compared, 4% PFA and 0.1% saponin; 4% PFA and 0.1% Triton X-100; and methanol, we found that the optimal perm/fix solution depended on the specific antibody:target combination of interest (Figure 1). It can therefore be helpful to try all three when looking to label an intracellular target.

 

Eleven flow plots, each testing a different antibody against WT and KO cells (see figure legend for details). In some plots, the peaks for WT and KO completely overlap (plots 1, 2, 4, 5, 6, 11). In others, the peaks are separate with overlap only at the shoulders (plots 3, 8, 9, and 10). Plot 7 shows incomplete overlap between the two peaks. In each plot, a secondary only control is to the left of the WT and usually to the left of the KO. Plot 10 shows the KO and secondary only peaks completely overlapping.

Figure 1: HCT 116 WT and SYT1 KO cells were labelled with a green or violet, fluorescent dye, respectively. WT and KO cells were mixed in a 1:1 ratio and fixed in 4% PFA and permeabilized in 0.1% saponin. 400,000 cells were stained with the indicated Synaptotagmin-1 antibodies and corresponding Multi-rAb CoraLite® Plus 647 secondary antibodies. Antibody staining was quantified using the Attune NxT Flow Cytometer with representative images showing the staining intensity in the KO population (pink histogram) compared to the WT cells (green histogram). The unfilled histograms show cells that have only been stained with secondary antibodies, showing the level of background staining. All antibodies were diluted to 1 µg/mL except for 14558, which was used at 0.35 µg/mL (1/100). * = monoclonal antibody, ** = recombinant antibody. Figure adapted from Biddle et al., 2024.

 

Once an antibody is found that shows separation between the wild-type and the KO population (Figure 1), further optimization can be performed to both maximize the separation and reduce any background staining. This can include using: different blocking reagents; titration of antibody concentration (this can also save money!); and optimizing fixation/permeabilization reagents for intracellular targets.

Since knockout experiments can be time-consuming and expensive, it may be helpful to find an antibody that has already been validated for flow cytometry through this approach. If you’re looking for a flow antibody, the YCharOS (Antibody Characterization through Open Science) initiative has now started producing flow cytometry characterization data for selected antibodies, using the knockout approach. You can see an example of their work on their synaptotagmin 1 page. The data produced is available on the F1000 gateway and Zenodo community. 

Knockdown approach

For cell types where knockout studies are not feasible, an alternative approach is RNA interference (RNAi). The most common method is transfection of short interfering RNA (siRNA). Knockdown caused by siRNA is also transient, and as such, the optimal knockdown of the protein target will depend on the protein turnover rate. If you need a more permanent knockdown, consider using shRNA vectors instead. With both siRNA and shRNA, it can be difficult to achieve sufficient knockdown to enable robust assessment of antibody selectivity. Additionally, it can be a real challenge to troubleshoot RNAi failures. Potential explanations for RNAi not reducing the antibody signal include:

  • The antibody is not selective
  • The knockdown is not efficient at the RNA level
  • The knockdown is not efficient at the protein level at the timepoint studied
  • Off-target activities of the RNAi

Correctly interpreting your data therefore necessitates careful design of RNAi sequences, confirmation of RNA knockdown by RT-qPCR, and potentially the need to study protein expression at multiple timepoints. When the target of interest is critical to cell survival or proliferation, an additional challenge is that only partial or ephemeral knockdown will be possible, and the small difference in protein expression makes confirmation of antibody selectivity even more challenging.

Correlation with RNA or Proteomic data from multiple cell lines/ types with different expression

Flow cytometry is often used with mixed populations of cells, like a blood sample containing many different leukocytes. Where data is available from antibody-independent methods, like RNAseq or proteomics, it is possible to compare antibody labeling of different cell types within a sample to expected labeling (Figure 2). The panel of antibodies used in this approach should also allow for the clear identification of the cell types of interest. By correlating your data sets, you can increase confidence that the labeling observed represents the expression level of the target of interest. This approach maps onto the orthogonal approach within the five pillars and is frequently used in combination with other approaches.

If several cell lines express the target at different levels, it is possible to design experiments where the performance of the antibody can be assessed through its ability to separate the cell lines based on expression of the target. In this case, you could compare the antibody’s labeling of the cell lines to the -omics data. The cell lines with higher expression of the target protein, as determined by the -omics data, should have more antibodies labeling them; the cell lines with lower expression should have less antibodies labeling them. We find that cell tracker dyes are helpful in designing such panels, so that pre-stained cell lines can be mixed together and then labeled with the antibody in the same tube.

 

Schematic: Five cells have been analyzed with transcriptomics and antibody staining, with bar graphs showing a correlation between the two in each cell line. Linear regression is performed, and the genomic mean fluorescence intensity and normalized transcript per million are positively correlated, with an R-squared of 0.9906,

 

Figure 2: Schematic showing the use of an orthogonal approach to understand the relationship between antibody staining intensity by flow cytometry, and mRNA expression.

 

If your protein of interest is expressed at similar levels in your cell lines, you can consider a cell treatment approach. If there is a cell treatment (e.g. PMA or a specific cytokine) known to either induce or suppress the expression of a target of interest, you can add an additional control containing treated cells. The expected change in expression should then be reflected in antibody labeling. This approach does allow you to determine expression levels in the same cell lines/background. However, the cell treatment will frequently induce/suppress the expression of many related proteins, not just your protein of interest, which can affect your experimental readouts.

Be aware that in any version of this approach, you are relying on correlations between actual antibody labeling and expected labeling. Correlation cannot prove antibody selectivity, and for that reason, it is best practice to combine correlation with other approaches when possible.

Finally, correlation of -omics data can also be used in conjunction with an assessment of labeling pattern between multiple antibody clones targeting the same protein (independent antibodies). If two antibodies, that recognize different epitopes of the same protein, show the same pattern of labeling, this is supportive of their specificity for the protein. A challenge to this approach is that the identity of the epitope is often not available to confirm that they recognize different epitopes.

Detection of overexpressed (tagged or untagged) protein

When expression plasmids are available, or the necessary expertise to clone them, it can be useful to produce cell lines that overexpress the target of interest. This approach often involves the transient transfection of expression plasmids in a given cell line. For flow cytometry, it is useful to produce a sample with mixed expression, with the expression measurable through an epitope tag such as FLAG/His/GFP. Fluorescent protein tags, which correlate closely with target expression, are often preferred for this approach.

A highly selective antibody should be able to detect even in cells transfected with low efficacy. But since there can be large differences between endogenous expression and transfected overexpression, this approach cannot confirm the antibody’s ability to detect endogenous levels of the target protein. It may only be able to detect the target protein at the high levels caused by overexpression. Additionally, if the cell line used has endogenous levels of expression, this can make the results more difficult to interpret, and a knockout/ or knockdown approach should be preferred. The overexpression approach is therefore best suited to cell lines without any expression of the target of interest, which can be difficult to find for many proteins.

In practice, it was necessary for us to combine overexpression with the use of orthogonal approaches, cell treatments, and independent antibody approaches in different assay systems to validate flow antibodies (Virk et al., 2019, 2021). In those studies, we used this combination to find suitable antibodies for a target with very low expression, due to induced toxicity caused by the protein of interest.

HLDA workshop approved antibody clones

The antibody validation process represents a high level of effort and time, which may not be available to every scientist looking to do a flow cytometry experiment. But validation is critical to ensuring accurate interpretation of your results. Therefore, taking advantage of antibody validation efforts and information provided by external organizations like the Human Cell Differentiation Molecules (HCDM) can be quite useful.

HCDM, among other things, works to test flow cytometry antibodies through their HLDA workshops. Let’s use TIM-1 (Single pass type-1 membrane protein), which has been designated CD365 in the most recent HLDA workshop (HLDA10), as an example. For CD365, they examined two different antibody clones produced by different vendors. The specific epitope recognized by either antibody was not shared. Tables describe that the antibodies both recognized the target when transiently overexpressed in CHO cells and displayed similar labeling patterns on different primary blood leukocytes. The file shared also contains information about the labeling pattern of several cell lines using these two antibodies. This is the most usual level of evidence supplied to support the suitability of an antibody for its target.

HCDM as a whole is focused on characterizing cell surface molecules expressed on human blood leukocytes, including antibodies against these markers. The 371 CD markers characterized so far, and the additional subtypes, are available here, and include information on tested antibodies against the target. Helpfully, this also includes clone names, which can allow you to search different providers for the same clone. This can be particularly useful when designing larger multicolour panels, where different manufacturers may have the same clone available with different conjugates.

Summary

Flow cytometry is a powerful tool to delineate cell populations and to study relative protein expression. Its reliability will be entirely dependent on the selectivity of the antibody used in the specific sample and protocol. This can be a challenge to confirm. As such, the approach used may need to vary according to the target, sample type, and available resources. The approach we suggest is summarized in Figure 3. The principle is to confirm the antibody can detect the antigen of interest in the protocol of interest, ideally at endogenously expressed levels, and then adjust the approach to the specific end-use of an individual experiment.

 

a flow chart summarizing the text of the post.
Figure 3: Our recommendations for an approach that may be used to determine whether an antibody is suitable for your flow experiment. We find it useful to start with a screening procedure in easily genetically modified cell types (top). The specific approach will depend on the characteristics of the target and whether suitable cell types are available. Further evidence is then needed to confirm that the staining represents the target in your sample of interest (bottom). The approach maps onto the five pillars, with some specific recommendations for how to use these in practice.

""This post was written by Harvinder Virk, MA (Oxon), PhD, MRCP, from Only Good Antibodies, with significant input from Michael Biddle, PhD, also from Only Good Antibodies. The Only Good Antibodies community works to make best practices in research antibody selection and use possible, easy, and rewarded. 

The OGA community produces antibody characterisation data as part of the YCharOS open science ecosystem. It has received in-kind support to produce this data from commercial antibody manufacturers.

References and resources

References

Ayoubi, R., Ryan, J., Biddle, M. S., Alshafie, W., Fotouhi, M., Bolivar, S. G., Ruiz Moleon, V., Eckmann, P., Worrall, D., McDowell, I., Southern, K., Reintsch, W., Durcan, T. M., Brown, C., Bandrowski, A., Virk, H., Edwards, A. M., McPherson, P., & Laflamme, C. (2023). Scaling of an antibody validation procedure enables quantification of antibody performance in major research applications. eLife, 12, RP91645. https://doi.org/10.7554/eLife.91645

Biddle, M. S., Alende, C., Fotouhi, M., Jones, C., Ayoubi, R., Southern, K., Laflamme, C., Virk, H., NeuroSGC/YCharOS/EDDU Collaborative Group, & ABIF Consortium. (2024). A guide to selecting high-performing antibodies for Synaptotagmin-1 (Uniprot ID P21579) for use in western blot, immunoprecipitation, immunofluorescence and flow cytometry. F1000Research, 13, 817. https://doi.org/10.12688/f1000research.154034.1

Laflamme, C., McKeever, P. M., Kumar, R., Schwartz, J., Kolahdouzan, M., Chen, C. X., You, Z., Benaliouad, F., Gileadi, O., McBride, H. M., Durcan, T. M., Edwards, A. M., Healy, L. M., Robertson, J., & McPherson, P. S. (2019). Implementation of an antibody characterization procedure and application to the major ALS/FTD disease gene C9ORF72. eLife, 8, e48363. https://doi.org/10.7554/eLife.48363

Uhlen, M., Bandrowski, A., Carr, S., Edwards, A., Ellenberg, J., Lundberg, E., Rimm, D. L., Rodriguez, H., Hiltke, T., Snyder, M., & Yamamoto, T. (2016). A proposal for validation of antibodies. Nature Methods, 13(10), 823–827. https://doi.org/10.1038/nmeth.3995

Virk, H. S., Biddle, M. S., Smallwood, D. T., Weston, C. A., Castells, E., Bowman, V. W., McCarthy, J., Amrani, Y., Duffy, S. M., Bradding, P., & Roach, K. M. (2021). TGFβ1 induces resistance of human lung myofibroblasts to cell death via down-regulation of TRPA1 channels. British Journal of Pharmacology, 178(15), 2948–2962. https://doi.org/10.1111/bph.15467

Virk, H. S., Rekas, M. Z., Biddle, M. S., Wright, A. K. A., Sousa, J., Weston, C. A., Chachi, L., Roach, K. M., & Bradding, P. (2019). Validation of antibodies for the specific detection of human TRPA1. Scientific Reports, 9(1), 18500. https://doi.org/10.1038/s41598-019-55133-7

More resources from the Addgene blog

Introduction to Gating in Flow Cytometry

Antibodies 101: Flow Cytometry

Antibodies 101: Validation

Resources on addgene.org

Addgene's Antibody Guide

Addgene's Antibody Protocols

Topics: Antibodies, antibodies 101

Leave a Comment

Sharing science just got easier... Subscribe to our blog