Review Article

The Application of Single-Cell RNA Sequencing in Vaccinology

Box 4: Recommendations for the implementation of scRNA-seq in vaccine research

Hypothesis generation: areas in vaccinology that lend themselves to investigation by scRNA-seq
 (i) Cellular responses to various adjuvants and vaccines
 (ii) Transcriptional and antigen-specific responses to adjuvants and vaccines
 (iii) Site-specific immunity induced by vaccinations (for example, at mucosal surfaces following HIV vaccination, in the liver following liver-stage malaria vaccination, or in the lungs following tuberculosis vaccination [147])
   (a) Spatial transcriptomics, soon to provide resolution at the single-cell level, could also be used
 (iv) Single-cell transcriptomic signatures associated with neutralising antibody responses
 (v) The accordance between protective transcriptional signatures in vaccine human challenge studies in nonendemic and endemic countries
Experimental design: exploit the technology and make the most of precious samples
 (i) Assess the need for scRNA-seq as the primary experimental technique and contemplate whether the question can be answered by established techniques (e.g., ELISAs, ELISpots, and flow cytometry)
 (ii) Consider using bulk RNA-seq as an adjunct to scRNA-seq (e.g., bulk RNA-seq on all samples, with scRNA-seq on a subset to allow computational deconvolution)
 (iii) Tailor the particular type of scRNA-seq to the experimental question (e.g., will alternative splicing be of interest? Use full-length transcript profiling if so)
 (iv) Longitudinal gene expression and TCR/BCR profiling to track antigen-specific clones
 (v) Define heterogeneity of cellular response in protected individuals
 (vi) Generate monoclonal antibodies from TCR/BCR sequences for rational vaccine redesign
 (vii) Use scRNA-seq as “backbone” to other technologies (e.g., G&T seq/CITE-seq)
 (viii) Combine scRNA-seq with flow cytometry/FACS and/or magnetically assisted cell separation to isolate rare or specific cell types [148]
 (ix) Multiplex samples according to genotype to reduce sample preparation time, reagent and sequencing costs, and batch effects [149, 150]
 (x) Preserve leftover cells either by sorting into a plate or by preserving in fixative for later use (for example, in the reanalysis of an interesting sample)
Analytical considerations: bioinformatic requirements/suggestions
 (i) Plan with, budget, and include bioinformaticians who are capable of working with scRNA-seq data from study conception onwards
 (ii) Use freely available packages that are regularly maintained. See Table 1 in the review by Zeng and Dai [129] for a list of computational tools for scRNA-seq analyses
 (iii) Upload data files to Gene Expression Omnibus [151] with as much metadata as possible (and not contravening human subject guidelines, if using human samples) as both processed and raw data
 (iv) Consider submission of data to other relevant databases such as Human Cell Atlas and/or TCGA
 (v) Consider analysing a similar published dataset with the experimental dataset to increase statistical power and/or to ensure a novel pipeline reproduces results in a previously published study
 (vi) Make freely available the code which relates to bespoke analyses
Box 4: Recommendations for the implementation of scRNA-seq in vaccine research