Immunotranscriptomic profiling the acute and clearance phases of a human challenge dengue virus serotype 2 infection model

John P Hanley, Huy A Tu, Julie A Dragon, Dorothy M Dickson, Roxana Del Rio-Guerra, Scott W Tighe, Korin M Eckstrom, Nicholas Selig, Samuel V Scarpino, Stephen S Whitehead, Anna P Durbin, Kristen K Pierce, Beth D Kirkpatrick, Donna M Rizzo, Seth Frietze, Sean A Diehl, John P Hanley, Huy A Tu, Julie A Dragon, Dorothy M Dickson, Roxana Del Rio-Guerra, Scott W Tighe, Korin M Eckstrom, Nicholas Selig, Samuel V Scarpino, Stephen S Whitehead, Anna P Durbin, Kristen K Pierce, Beth D Kirkpatrick, Donna M Rizzo, Seth Frietze, Sean A Diehl

Abstract

About 20-25% of dengue virus (DENV) infections become symptomatic ranging from self-limiting fever to shock. Immune gene expression changes during progression to severe dengue have been documented in hospitalized patients; however, baseline or kinetic information is difficult to standardize in natural infection. Here we profile the host immunotranscriptome response in humans before, during, and after infection with a partially attenuated rDEN2Δ30 challenge virus (ClinicalTrials.gov NCT02021968). Inflammatory genes including type I interferon and viral restriction pathways are induced during DENV2 viremia and return to baseline after viral clearance, while others including myeloid, migratory, humoral, and growth factor immune regulation factors pathways are found at non-baseline levels post-viremia. Furthermore, pre-infection baseline gene expression is useful to predict rDEN2Δ30-induced immune responses and the development of rash. Our results suggest a distinct immunological profile for mild rDEN2Δ30 infection and offer new potential biomarkers for characterizing primary DENV infection.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Viremia and antibody levels in…
Fig. 1. Viremia and antibody levels in response to experimental infection of humans with rDEN2Δ30 (Tonga 74 strain).
a DENV2 viremia in a cohort of flavivirus-naïve subjects, n = 11. Viremia was measured by culture of serum on Vero81 cells and mean viral titer plaque-forming units per mL of serum (PFU/mL) ± standard deviation (approx. 67th percentile) above zero are shown. b Serum neutralizing antibodies (PRNT50, mean ± standard deviation) to DENV2-NGC and DENV2-Tonga after infection with rDEN2Δ30.
Fig. 2. Clinical laboratory assessment of rDEN2Δ30…
Fig. 2. Clinical laboratory assessment of rDEN2Δ30 infection.
a Leukocyte populations expressed as absolute counts, (×1000 cells/cm2, left) and as a percent of white blood cells (right) are shown for study visits occurring every other day over two ~2-week periods: 6 months preinfection and after infection of flavivirus-naive healthy volunteers (n = 11) with rDEN2Δ30, including day of infection. b Hematology labs, including red blood cell counts, serum hemoglobin, hematocrit, mean corpuscular hemoglobin (MCH), MCH concentration (MCHC), and mean corpuscular volume (MCV) are shown for pre- and post-infection as in (a). c Coagulation labs, platelet counts, mean platelet volume (MPV), Prothrombin time, international normalized ratio (INR), and partial thromboplastin time (PTT) are shown for pre- and post-infection as in (a). d Metabolic labs including alanine aminotransferase (ALT), Creatinine, and glomerular filtration rate (GFR) are shown for pre- and post-infection as in (a). Data shown for all subjects are shown as non-parametric LOESS (LOcal regrESSion) smoothing. All data also plotted by individual subject in Source Data. ULN upper limit of normal (solid lines), LLN lower limit of normal (dashed lines). For all panels, LOESS-smoothed lines and 95% confidence intervals are shown. Source data are provided as a Source Data file.
Fig. 3. Group analysis of temporal gene…
Fig. 3. Group analysis of temporal gene expression analysis after rDEN2Δ30 primary infection.
a Principal component analysis by timepoint. One symbol per subject. b The top 2400 variable genes clustered on heatmap with column annotations for timepoints post infection. TMM values ln(x + 1)-transformed. Rows are centered; unit variance scaling is applied to rows. Both rows and columns are clustered using correlation distance and average linkage. c F × E plots (log2 fold-change × expression, indicated by log2 counts per million (CPM) reads) of pairwise timepoint comparisons. Differential gene expression threshold is significance level of P < 0.05, FDR < 0.1, and transcript level ≥ 4 CPM. Examples of highly regulated genes are labeled (d) Pathway analysis of differentially expressed genes in pairwise comparisons by timepoint after infection. Predicted pathway directionality (green up-pointing triangle, upregulated; red down-pointing triangle, downregulated) was determined by the behavior of the genes exhibiting |≥1.5-fold change|. Source data are provided as a Source Data file.
Fig. 4. Immune-cell dynamics before, during, and…
Fig. 4. Immune-cell dynamics before, during, and after rDEN2Δ30 infection.
a Deconvolution of gene expression using a leukocyte gene signatures matrix to obtain median proportions of select adaptive and innate white blood cells at each timepoint. Center lines show the medians; box limits indicate the 25th and 75th percentiles; whiskers represent 95% confidence interval. Kruskal–Wallis one-way ANOVA was used to determine P values. *P < 0.05; #P < 0.1. bg Flow cytometric analysis of peripheral blood mononuclear cells from rDEN2Δ30-infected subjects. Data are expressed as fold change in cell frequency at days 8, and 28 after infection relative to day 0 baseline. Center lines show the medians; box limits indicate the 25th and 75th percentiles and all data points are shown. Kruskal–Wallis one-way ANOVA was used to determine P values indicated as *P < 0.05 and #P < 0.1. b Lineage-negative (CD3−CD19−CD56−CD14−CD16−) HLA-DR+ dendritic cells (DC), HLA-DRhi activated DC (DC_act); c monocytes: classical (c_mono), intermediate (i_mono), and non-classical (nc_mono); d natural killer T cells (nkt), NK1-5 populations (defined by CD56 and CD16 staining as outlined in Supplementary Fig. 4); e CD3+ T cells (t cells), CD4+ T cells (cd4), and CD8+ T cells (cd8); f CD8+ T-cell populations: activated CD279+ (cd8_act), cytotoxic CD57+ (cd8_cyto), CCR7−CD45RA+ T-effector re-expressing CD45RA (cd8t_emra), CCR7+CD45RA+ naive (cd8t_naive), CCR7−CD45RA− effector memory (cd8t_em), CCR7+CD45RA− central memory (cd8t_cm), and homeostatic CD127+ (cd8_homeo); g CD4+ T-cell populations: activated CD279+ (cd4_act), cytotoxic CD57+ (cd4_cyto), activation-induced marker (AIM)-positive CD154+CD134+(cd4_AIM), CD25+CD134+ (cd4_AIM_old), regulatory T cells CD127–CD25+ (t_regs), homeostatic CD127+ (cd4_homeo), CCR7–CD45RA+ T effector re-expressing CD45RA (cd4t_emra), CCR7+CD45RA+ naive (cd4t_naive), CCR7–CD45RA– effector memory (cd4t_em), CCR7+CD45RA– central memory (cd4t_cm); h CD19+ B cells (bcells), plasmablasts CD19+ CD38hiCD27hi (pb), IgM+CD27– naïve B cells (naive_b), and IgM–CD27+ switched memory B cells (s_mbc). See Fig. S4 for gating. Source data are provided as a Source Data file.
Fig. 5. Temporal gene expression analysis after…
Fig. 5. Temporal gene expression analysis after rDEN2Δ30 infection by logical conjunction analysis (LCA).
a Schematic overview of LCA. Genes have undergone regulation if for all 11 subjects the regulation occurred in the same direction (e.g., up) and had a P value < 0.05 and FDR < 0.1. No minimum fold-change criteria was applied for Threshold. b F × E plots (log2 fold-change × expression, in counts per million reads (CPM)) of pairwise timepoint comparisons for genes meeting threshold of P < 0.05 and FDR < 0.1 and ≥4 counts per million reads (CPM). Examples of genes regulated |≥1.5-fold| are labeled (c) Venn diagram of gene expression changes by timepoint. Triangles indicate directionality of gene regulation in timepoint comparison. d DAVID Pathway analysis of Viremia-Tracking genes (e) DAVID pathway analysis of post-viremia genes. Directionality is not shown because the pathway definition incorporates three timepoints. Source data are provided as a Source Data file.
Fig. 6. Association of preinfection baseline gene…
Fig. 6. Association of preinfection baseline gene expression with clinical features of rDEN2Δ30 infection.
a Genome-wide absolute baseline gene expression level counts (rows) were queried against ten clinical laboratory parameters (columns) associated with rDEN2Δ30 infection including peak viremia, neutralizing antibodies and changes in blood cell counts across the 11 subjects. Only genes (rows) that had an absolute correlation >0.9 for at least one of the columns are pictured. Darker colors represent a stronger correlation; while blue and red represent positive and negative correlation, respectively. Unsupervised clustering identified 49 genes and three major groups (I–III) distinguishing amongst the parameters. bd Pathway analysis and GO terms associated with Group 1 (b), Group II (c), and Group III (d) gene associations and unadjusted P values with FDR < 0.1 are shown. Source data are provided as a Source Data file.
Fig. 7. Association of preinfection gene expression…
Fig. 7. Association of preinfection gene expression with rash following rDEN2Δ30 infection.
a Hierarchical clustering of transcript levels (expressed) counts per million (cpm) for genes that have a day 0 cpm that is linearly separable for subjects that have rash and those that do not have rash. Darker shades of red indicate a higher median expression (log2cpm) on day 0. b Genes that distinguish rash versus non-rash based on linear separability greater than 20 (i.e., the median Δ cpm for rash versus non-rash is |≥20|) with corresponding P values (square symbols). c DAVID pathway analysis for all significant rash-distinguishing linear separable genes (n = 56). Source data are provided as a Source Data file.
Fig. 8. Distinct gene regulation in different…
Fig. 8. Distinct gene regulation in different phases of rDEN2Δ30 infection and in severe dengue.
Venn diagram of genes associated with severe dengue from Robinson et al. (ref. ) in relation to viremia-tracking genes (n = 74) and to those that were upregulated (n = 60) or downregulated (n = 57) post-viremia compared to baseline following rDEN2Δ30 infection. Source data are provided as a Source Data file.

References

    1. Bhatt S, et al. The global distribution and burden of dengue. Nature. 2013;496:504–507. doi: 10.1038/nature12060.
    1. WHO. Dengue guidelines for diagnosis, treatment, prevention and control: new edition.) World Health Organization (2009).
    1. Wilder-Smith A, Ooi EE, Horstick O, Wills B. Dengue. Lancet. 2019;393:350–363. doi: 10.1016/S0140-6736(18)32560-1.
    1. Guzman MG, Gubler DJ, Izquierdo A, Martinez E, Halstead SB. Dengue infection. Nat. Rev. Dis. Prim. 2016;2:16055. doi: 10.1038/nrdp.2016.55.
    1. Brathwaite Dick, O. et al. The history of dengue outbreaks in the Americas. Am. J. Trop. Med. Hyg.87, 584–593 (2012).
    1. Gibbons RV, et al. Analysis of repeat hospital admissions for dengue to estimate the frequency of third or fourth dengue infections resulting in admissions and dengue hemorrhagic fever, and serotype sequences. Am. J. Trop. Med. Hyg. 2007;77:910–913. doi: 10.4269/ajtmh.2007.77.910.
    1. Whitehead SS, Blaney JE, Durbin AP, Murphy BR. Prospects for a dengue virus vaccine. Nat. Rev. Microbiol. 2007;5:518–528. doi: 10.1038/nrmicro1690.
    1. Katzelnick LC, et al. Antibody-dependent enhancement of severe dengue disease in humans. Science. 2017;358:929–932. doi: 10.1126/science.aan6836.
    1. St John AL, Rathore APS. Adaptive immune responses to primary and secondary dengue virus infections. Nat. Rev. Immunol. 2019;19:218–230. doi: 10.1038/s41577-019-0123-x.
    1. Devignot S, et al. Genome-wide expression profiling deciphers host responses altered during dengue shock syndrome and reveals the role of innate immunity in severe dengue. PLoS ONE. 2010;5:e11671. doi: 10.1371/journal.pone.0011671.
    1. Hoang LT, et al. The early whole-blood transcriptional signature of dengue virus and features associated with progression to dengue shock syndrome in Vietnamese children and young adults. J. Virol. 2010;84:12982–12994. doi: 10.1128/JVI.01224-10.
    1. Kwissa M, et al. Dengue virus infection induces expansion of a CD14(+)CD16(+) monocyte population that stimulates plasmablast differentiation. Cell Host Microbe. 2014;16:115–127. doi: 10.1016/j.chom.2014.06.001.
    1. Loke P, et al. Gene expression patterns of dengue virus-infected children from nicaragua reveal a distinct signature of increased metabolism. PLoS Negl. Trop. Dis. 2010;4:e710. doi: 10.1371/journal.pntd.0000710.
    1. Long HT, et al. Patterns of gene transcript abundance in the blood of children with severe or uncomplicated dengue highlight differences in disease evolution and host response to dengue virus infection. J. Infect. Dis. 2009;199:537–546. doi: 10.1086/596507.
    1. Nascimento EJ, et al. Gene expression profiling during early acute febrile stage of dengue infection can predict the disease outcome. PLoS ONE. 2009;4:e7892. doi: 10.1371/journal.pone.0007892.
    1. Simmons CP, et al. Patterns of host genome-wide gene transcript abundance in the peripheral blood of patients with acute dengue hemorrhagic fever. J. Infect. Dis. 2007;195:1097–1107. doi: 10.1086/512162.
    1. Sun P, et al. Sequential waves of gene expression in patients with clinically defined dengue illnesses reveal subtle disease phases and predict disease severity. PLoS Negl. Trop. Dis. 2013;7:e2298. doi: 10.1371/journal.pntd.0002298.
    1. van de Weg CA, et al. Time since onset of disease and individual clinical markers associate with transcriptional changes in uncomplicated dengue. PLoS Negl. Trop. Dis. 2015;9:e0003522. doi: 10.1371/journal.pntd.0003522.
    1. Zanini F, et al. Virus-inclusive single-cell RNA sequencing reveals the molecular signature of progression to severe dengue. Proc. Natl Acad. Sci. U.S.A. 2018;115:E12363–E12369. doi: 10.1073/pnas.1813819115.
    1. Robinson M, et al. A 20-gene set predictive of progression to severe dengue. Cell Rep. 2019;26:1104–1111.e1104. doi: 10.1016/j.celrep.2019.01.033.
    1. Clapham H, et al. Epidemiology of infant dengue cases illuminates serotype-specificity in the interaction between immunity and disease, and changes in transmission dynamics. PLoS Negl. Trop. Dis. 2015;9:e0004262. doi: 10.1371/journal.pntd.0004262.
    1. Durbin AP, Whitehead SS. The dengue human challenge model: has the time come to accept this challenge? J. Infect. Dis. 2013;207:697–699. doi: 10.1093/infdis/jis749.
    1. Larsen CP, Whitehead SS, Durbin AP. Dengue human infection models to advance dengue vaccine development. Vaccine. 2015;33:7075–7082. doi: 10.1016/j.vaccine.2015.09.052.
    1. Mammen MP, et al. Evaluation of dengue virus strains for human challenge studies. Vaccine. 2014;32:1488–1494. doi: 10.1016/j.vaccine.2013.12.040.
    1. Gunther VJ, et al. A human challenge model for dengue infection reveals a possible protective role for sustained interferon gamma levels during the acute phase of illness. Vaccine. 2011;29:3895–3904. doi: 10.1016/j.vaccine.2011.03.038.
    1. Gubler DJ, Reed D, Rosen L, Hitchcock JR., Jr. Epidemiologic, clinical, and virologic observations on dengue in the Kingdom of Tonga. Am. J. Trop. Med. Hyg. 1978;27:581–589. doi: 10.4269/ajtmh.1978.27.581.
    1. Blaney JE, Jr, Hanson CT, Hanley KA, Murphy BR, Whitehead SS. Vaccine candidates derived from a novel infectious cDNA clone of an American genotype dengue virus type 2. BMC Infect. Dis. 2004;4:39. doi: 10.1186/1471-2334-4-39.
    1. Durbin AP, Kirkpatrick BD, Pierce KK, Schmidt AC, Whitehead SS. Development and clinical evaluation of multiple investigational monovalent DENV vaccines to identify components for inclusion in a live attenuated tetravalent DENV vaccine. Vaccine. 2011;29:7242–7250. doi: 10.1016/j.vaccine.2011.07.023.
    1. Kirkpatrick BD, et al. The live attenuated dengue vaccine TV003 elicits complete protection against dengue in a human challenge model. Sci. Transl. Med. 2016;8:330ra336. doi: 10.1126/scitranslmed.aaf1517.
    1. Grifoni A, et al. Patterns of Cellular Immunity Associated with Experimental Infection with rDEN2Delta30 (Tonga/74) Support Its Suitability as a Human Dengue Virus Challenge Strain. J. Virol. 2017;91:e02133–02116.
    1. Halstead SB. Dengue. Lancet. 2007;370:1644–1652. doi: 10.1016/S0140-6736(07)61687-0.
    1. Newman AM, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods. 2015;12:453–457. doi: 10.1038/nmeth.3337.
    1. Tran TM, et al. A molecular signature in blood reveals a role for p53 in regulating malaria-induced inflammation. Immunity. 2019;51:750–765.e710. doi: 10.1016/j.immuni.2019.08.009.
    1. Frankenberger M, Sternsdorf T, Pechumer H, Pforte A, Ziegler-Heitbrock HW. Differential cytokine expression in human blood monocyte subpopulations: a polymerase chain reaction analysis. Blood. 1996;87:373–377. doi: 10.1182/blood.V87.1.373.373.
    1. Angelo MA, et al. Human CD4(+) T cell responses to an attenuated tetravalent dengue vaccine parallel those induced by natural infection in magnitude, HLA restriction, and antigen specificity. J. Virol. 2017;91:e02147–02116. doi: 10.1128/JVI.02147-16.
    1. Graham N, et al. Rapid induction and maintenance of virus-specific CD8(+) TEMRA and CD4(+) TEM cells following protective vaccination against dengue virus challenge in humans. Front Immunol. 2020;11:479. doi: 10.3389/fimmu.2020.00479.
    1. Weiskopf D, et al. The human CD8+ T cell responses induced by a live attenuated tetravalent dengue vaccine are directed against highly conserved epitopes. J. Virol. 2015;89:120–128. doi: 10.1128/JVI.02129-14.
    1. Weiskopf D, et al. Comprehensive analysis of dengue virus-specific responses supports an HLA-linked protective role for CD8+ T cells. Proc. Natl Acad. Sci. U.S.A. 2013;110:E2046–E2053. doi: 10.1073/pnas.1305227110.
    1. Weiskopf D, et al. Dengue virus infection elicits highly polarized CX3CR1+ cytotoxic CD4+ T cells associated with protective immunity. Proc. Natl Acad. Sci. U.S.A. 2015;112:E4256–E4263. doi: 10.1073/pnas.1505956112.
    1. Perng YC, Lenschow DJ. ISG15 in antiviral immunity and beyond. Nat. Rev. Microbiol. 2018;16:423–439. doi: 10.1038/s41579-018-0020-5.
    1. Nivarthi UK, et al. Longitudinal analysis of acute and convalescent B cell responses in a human primary dengue serotype 2 infection model. EBioMedicine. 2019;41:465–478. doi: 10.1016/j.ebiom.2019.02.060.
    1. Simmons JS, St John JH, Reynolds FHK. Numerical and morphological alterations of the leukocytes during dengue fever. Phillipine J. Sci. 1931;44:128–146.
    1. Unger G, Chor B. Linear separability of gene expression data sets. IEEE/ACM Trans. Comput Biol. Bioinform. 2010;7:375–381. doi: 10.1109/TCBB.2008.90.
    1. Popper SJ, et al. Temporal dynamics of the transcriptional response to dengue virus infection in Nicaraguan children. PLoS Negl. Trop. Dis. 2012;6:e1966. doi: 10.1371/journal.pntd.0001966.
    1. Vaughn DW, et al. Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity. J. Infect. Dis. 2000;181:2–9. doi: 10.1086/315215.
    1. Durbin AP, et al. Phenotyping of peripheral blood mononuclear cells during acute dengue illness demonstrates infection and increased activation of monocytes in severe cases compared to classic dengue fever. Virology. 2008;376:429–435. doi: 10.1016/j.virol.2008.03.028.
    1. Schmid MA, Diamond MS, Harris E. Dendritic cells in dengue virus infection: targets of virus replication and mediators of immunity. Front Immunol. 2014;5:647. doi: 10.3389/fimmu.2014.00647.
    1. Michlmayr D, Andrade P, Gonzalez K, Balmaseda A, Harris E. CD14(+)CD16(+) monocytes are the main target of Zika virus infection in peripheral blood mononuclear cells in a paediatric study in Nicaragua. Nat. Microbiol. 2017;2:1462–1470. doi: 10.1038/s41564-017-0035-0.
    1. Roth H, et al. Flavivirus infection uncouples translation suppression from cellular stress responses. mBio. 2017;8:e02150–16.
    1. Hishiki T, et al. Interferon-mediated ISG15 conjugation restricts dengue virus 2 replication. Biochem. Biophys. Res. Commun. 2014;448:95–100. doi: 10.1016/j.bbrc.2014.04.081.
    1. Chan KR, et al. Metabolic perturbations and cellular stress underpin susceptibility to symptomatic live-attenuated yellow fever infection. Nat. Med. 2019;25:1218–1224. doi: 10.1038/s41591-019-0510-7.
    1. Popper SJ, et al. Early transcriptional responses after dengue vaccination mirror the response to natural infection and predict neutralizing antibody titers. J. Infect. Dis. 2018;218:1911–1921. doi: 10.1093/infdis/jiy434.
    1. Santos JC, Broz P. Sensing of invading pathogens by GBPs: at the crossroads between cell-autonomous and innate immunity. J. Leukoc. Biol. 2018;104:729–735. doi: 10.1002/JLB.4MR0118-038R.
    1. Durbin AP, et al. A single dose of any of four different live attenuated tetravalent dengue vaccines is safe and immunogenic in flavivirus-naive adults: a randomized, double-blind clinical trial. J. Infect. Dis. 2013;207:957–965. doi: 10.1093/infdis/jis936.
    1. Kirkpatrick BD, et al. Robust and balanced immune responses to all 4 dengue virus serotypes following administration of a single dose of a live attenuated tetravalent dengue vaccine to healthy, flavivirus-naive adults. J. Infect. Dis. 2015;212:702–710. doi: 10.1093/infdis/jiv082.
    1. Durbin AP, et al. rDEN4delta30, a live attenuated dengue virus type 4 vaccine candidate, is safe, immunogenic, and highly infectious in healthy adult volunteers. J. Infect. Dis. 2005;191:710–718. doi: 10.1086/427780.
    1. Pertea M, Kim D, Pertea GM, Leek JT, Salzberg SL. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat. Protoc. 2016;11:1650–1667. doi: 10.1038/nprot.2016.095.
    1. ENCODE. ENCODE experimental guidelines for ENCODE3 RNA-seq. (2017).
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. doi: 10.1093/bioinformatics/btp616.
    1. McCarthy DJ, Chen Y, Smyth GK. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 2012;40:4288–4297. doi: 10.1093/nar/gks042.
    1. Fu J., Frazee A. C., Collado-Torres L., Jaffe A. E., Leek J. T. ballgown: flexible, isoform-level differential expression analysis.). 2.18.0 edn. R package version (2019).
    1. Metsalu T, Vilo J. ClustVis: a web tool for visualizing clustering of multivariate data using Principal Component Analysis and heatmap. Nucleic Acids Res. 2015;43:W566–W570. doi: 10.1093/nar/gkv468.
    1. Li H, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–930. doi: 10.1093/bioinformatics/btt656.
    1. Brodin P, Davis MM. Human immune system variation. Nat. Rev. Immunol. 2017;17:21–29. doi: 10.1038/nri.2016.125.
    1. Dalman MR, Deeter A, Nimishakavi G, Duan ZH. Fold change and p-value cutoffs significantly alter microarray interpretations. BMC Bioinform. 2012;13(Suppl 2):S11. doi: 10.1186/1471-2105-13-S2-S11.
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009;4:44–57. doi: 10.1038/nprot.2008.211.
    1. Liao Y, Wang J, Jaehnig EJ, Shi Z, Zhang B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res. 2019;47:W199–W205. doi: 10.1093/nar/gkz401.
    1. Mi H, et al. Protocol update for large-scale genome and gene function analysis with the PANTHER classification system (v.14.0) Nat. Protoc. 2019;14:703–721. doi: 10.1038/s41596-019-0128-8.
    1. Moncunill G, Han H, Dobano C, McElrath MJ, De Rosa SC. OMIP-024: pan-leukocyte immunophenotypic characterization of PBMC subsets in human samples. Cytom. A. 2014;85:995–998. doi: 10.1002/cyto.a.22580.
    1. Dan JM, et al. A cytokine-independent approach to identify antigen-specific human germinal center T follicular helper cells and rare antigen-specific CD4+ T cells in blood. J. Immunol. 2016;197:983–993. doi: 10.4049/jimmunol.1600318.
    1. Rydyznski Moderbacher C, et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell. 2020;183:996–1012.e1019. doi: 10.1016/j.cell.2020.09.038.

Source: PubMed

3
Iratkozz fel