Skip to main content
PLOS Neglected Tropical Diseases logoLink to PLOS Neglected Tropical Diseases
. 2019 Dec 9;13(12):e0007908. doi: 10.1371/journal.pntd.0007908

Helminth-induced Th2 cell dysfunction is distinct from exhaustion and is maintained in the absence of antigen

Johanna A Knipper 1,2, Alasdair Ivens 1,2, Matthew D Taylor 1,2,*
Editor: Benjamin L Makepeace3
PMCID: PMC6922449  PMID: 31815932

Abstract

T cell-intrinsic regulation, such as anergy, adaptive tolerance and exhaustion, is central to immune regulation. In contrast to Type 1 and Type 17 settings, knowledge of the intrinsic fate and function of Th2 cells in chronic Type 2 immune responses is lacking. We previously showed that Th2 cells develop a PD-1/PD-L2-dependent intrinsically hypo-responsive phenotype during infection with the filarial nematode Litomosoides sigmodontis, denoted by impaired functionality and parasite killing. This study aimed to elucidate the transcriptional changes underlying Th2 cell-intrinsic hypo-responsiveness, and whether it represents a unique and stable state of Th2 cell differentiation. We demonstrated that intrinsically hypo-responsive Th2 cells isolated from L. sigmodontis infected mice stably retained their dysfunctional Th2 phenotype upon transfer to naïve recipients, and had a divergent transcriptional profile to classical Th2 cells isolated prior to hypo-responsiveness and from mice exposed to acute Type 2 stimuli. Hypo-responsive Th2 cells displayed a distinct transcriptional profile to exhausted CD4+ T cells, but upregulated Blimp-1 and the anergy/regulatory-associated transcription factors Egr2 and c-Maf, and shared characteristics with tolerised T cells. Hypo-responsive Th2 cells increased mRNA expression of the soluble regulatory factors Fgl2, Cd38, Spp1, Areg, Metrnl, Lgals3, and Csf1, and a subset developed a T-bet+IFN-γ+ Th2/Th1 hybrid phenotype, indicating that they were not functionally inert. Contrasting with their lost ability to produce Th2 cytokines, hypo-responsive Th2 cells gained IL-21 production and IL-21R blockade enhanced resistance to L. sigmodontis. IL-21R blockade also increased the proportion of CD19+PNA+ germinal centre B cells and serum levels of parasite specific IgG1. This indicates a novel regulatory role for IL-21 during filarial infection, both in controlling protection and B cell responses. Thus, Th2 cell-intrinsic hypo-responsiveness is a distinct and stable state of Th2 cell differentiation associated with a switch from a classically active IL-4+IL-5+ Th2 phenotype, to a non-classical dysfunctional and potentially regulatory IL-21+Egr2+c-Maf+Blimp-1+IL-4loIL-5loT-bet+IFN-γ+ Th2 phenotype. This divergence towards alternate Th2 phenotypes during chronicity has broad implications for the outcomes and treatment of chronic Type 2-related infections and diseases.

Author summary

Helminth parasites chronically infect over 1 billion people, with filarial nematodes accounting for 120 million. Protection is mediated by Th2 cells, but infection invokes dominant down-regulatory immune responses forming a major barrier to the development of protective immunity. We previously demonstrated that during murine filarial infection Th2 cells change phenotype, developing a novel form of Th2 cell-intrinsic dysfunction that impairs parasite clearance. Here we investigate the gene expression profile of Th2 cell dysfunction, and whether it represents a unique and stable form of T cell differentiation. Dysfunctional Th2 cells had a different gene expression profile to effector Th2 cells, and retained their unresponsive phenotype in the absence of antigen and active infection when transferred to a naïve recipient. Dysfunctional Th2 cells produced IL-21, through which they may inhibit protective immunity, as IL-21R neutralisation increased parasite killing. In comparison to known forms of T cell intrinsic regulation, dysfunctional Th2 cells had a distinct gene expression profile to exhausted T cells, but shared characteristics with anergy and tolerance. Together this indicates that Th2 cell dysfunction represents a unique and stable state of Th2 cell differentiation, which has important implications for understanding the outcomes of helminth infections and designing therapies for Th2-mediated allergies.

Introduction

Human helminth infections are characterized by impaired Type 2 immune responses, the combined result of immune suppression by the parasite and immune regulation by the host [1]. This results in chronic infections in over 1 billion individuals worldwide, with protective immunity taking decades to develop [2]. Immune regulation during helminth infections, and immune tolerance to allergens, both associate with a change from a classical inflammatory Type 2 phenotype to a modified or tolerant Type 2 phenotype; denoted by increased IL-10 and a switch from inflammatory IgE to non-inflammatory IgG4 [1,3,4]. Thus, allergic inflammation and helminth infection share similar activation pathways, and in both cases immune regulation during chronicity associates with a change in Type 2 phenotype rather than a global downregulation of Type 2 responses. The mechanisms underlying this phenotype change are still largely unknown.

T cell-intrinsic regulation, e.g. exhaustion, anergy, and adaptive tolerance, is central to the development of immune tolerance [5], and the control of immune responses to infections and tumours [6]. The various forms of T cell-intrinsic unresponsiveness represent distinct states of T cell differentiation with unique functional properties and transcriptional signatures [58], and can lead to the development of tolerised or exhausted memory responses [810]. Thus, the development of T cell intrinsic regulation or dysfunction can have profound consequences on infection susceptibility and development of protective memory. Whilst a range of Th2 cell-extrinsic regulatory mechanisms have been characterised, e.g. regulatory T and B cells, and alternatively-activated macrophages [1,3,11,12], little is known of how Type 2 immune responses are regulated by intrinsic changes in Th2 cell function and fate.

There is some evidence for Th2 cell-intrinsic regulation during helminth infections. T cells from patients with chronic nematode infections show impaired TCR signalling [13,14], and suppression in human filariasis is associated with the upregulation of T cell anergy factors [15]. Th2 cells become intrinsically hypo-responsive or dysfunctional during murine schistosomiasis [16], and exposure to Fasciola hepatica tegumental antigens induces an anergic-like T cell phenotype [17,18]. We demonstrated that Th2 cell-intrinsic hypo-responsiveness develops during murine infection with the filarial nematode Litomosoides sigmodontis, and that blockade of the PD-1/PD-L2 pathway increases resistance and recovers Th2 functionality [19,20]. Hypo-responsive Th2 cells in both schistosomiasis and filariasis show an intrinsically-impaired ability to produce Th2 cytokines (IL-4, IL-5, IL-10, and IL-13) and to proliferate, both in the presence of antigen and mitogen [16,19,20]. Thus, immune regulation during chronic helminth infections associates with an intrinsically hypo-responsive or dysfunctional Th2 cell phenotype, which influences both resistance and pathology.

It has not been established whether Th2 cell-intrinsic hypo-responsiveness represents a known form of T cell dysfunction or a hitherto undescribed differentiation state, and the underlying mechanisms are not well defined. The involvement of PD-1 suggests similarities with T cell exhaustion. However, in contrast with exhaustion, PD-1 acts in conjunction with PD-L2 rather than PD-L1 [19]. Susceptibility to L. sigmodontis associates with the recruitment of PD-L2+ monocytes to the infection site that control Th2 cell functional quality, suggesting that monocytes may play a role in induction of Th2 cell-intrinsic hypo-responsiveness [21]. In murine schistosomiasis, Th2 cell intrinsic hypo-responsiveness is dependent upon the anergy factor Grail, suggesting similarities with adaptive tolerance [16].

In this study we identify the transcriptional changes associated with the development of L. sigmodontis-elicited Th2 cell-intrinsic hypo-responsiveness, and test whether it represents a unique and stable state of Th2 cell differentiation. We find that hypo-responsive Th2 cells retained their dysfunctional phenotype upon transfer to a naïve recipient indicating that their unresponsive state is stably maintained in the absence of antigen and active infection. We demonstrate that hypo-responsive Th2 cells have a divergent transcriptional profile to classical Th2 cells isolated either prior to the onset of hypo-responsiveness, or from mice acutely infected with the nematode Nippostrongylus brasiliensis. Although sharing characteristics with anergy and tolerance, hypo-responsive Th2 cells had a global transcriptional profile that differentiated them from exhausted, anergic, and tolerised CD4+ T cells. Contrasting with their loss of ability to produce Th2 cytokines, hypo-responsive Th2 cells gained IL-21 production, and IL-21R blockade increased resistance to L. sigmodontis infection, indicating a novel regulatory role for IL-21. This indicates that Th2 cell-intrinsic hypo-responsiveness represents a distinct and stable state of T cell differentiation, and that hypo-responsive Th2 cells may inhibit Type 2 immunity via IL-21.

Materials and methods

Ethics statement

All animal work was approved by the University of Edinburgh Ethics Committee (PL02-10) and by the UK Home Office (PPL70/8548), and conducted in accordance with the Animals (Scientific Procedures) Act 1986.

Animals, parasites, and cell isolations

Female BALB/c and IL-4gfp 4get reporter mice on the BALB/c background were bred in-house and maintained under specific pathogen-free conditions at the University of Edinburgh. Mice were used at 6–12 weeks of age, and randomly assigned to experimental groups. The L. sigmodontis life cycle was maintained in gerbils (Meriones unguiculatus) using the mite vector Ornithonyssus bacoti [22]. Mice were infected s.c. on the upper back with 30 L. sigmodontis L3 larvae. Adult or larval parasites were recovered by lavage of the thoracic cavity. L. sigmodontis antigen (LsAg) was prepared by collecting the PBS-soluble fraction of homogenized adult male and female worms. To quantify blood microfilariae, 30 μL of tail blood was collected in FACS lysing solution (Becton-Dickinson), and microfilaria counted using a dark field optical microscopy (Axiovert 25, Zeiss). N. brasiliensis was maintained in Sprague-Dawley rats as previously described [23]. Mice were infected by s.c. injection with 200 N. brasiliensis L3 larvae. The parathymic, posterior, mediastinal and paravertebral LN, were taken as a source of thoracic LN (tLN) draining the pleural cavity (PleC). PleC cells were recovered by lavage. TLN cells were dissociated and washed in RPMI-1640 (invitrogen) supplemented with 0.5% mouse sera (Caltag-Medsystems), 100 U/ml penicillin, 100 μg/ml streptomycin and 2 mM L-glutamine.

Flow cytometry and intracellular cytokine staining

The following antibodies were used: Pacific Blue- or Alexa 700-conjugated anti-CD4 (RM4-5), polyclonal anti-GFP (Ebioscience), Alexafluor488-conjugated goat anti-rabbit IgG (Invitrogen), phycoerythrin (Pe)-conjugated anti-IL-4 (11B11, Biolegend), allophycocyanine (APC)-conjugated anti-IL-5 (TRFK5, Biolegend), biotinylated anti-CXCR5 (RF8B2, BD Biosciences), PcPCy5.5-conjugated anti-CD44 (IM7, Biolegend), Pacific Blue-conjugated anti-CD62L (MEL-14, Biolegend), PeCy7-conjugated anti-CD25 (PC61, Biolegend), Pe-conjugated anti-CD45RB (C363-16A, Biolegend), Fitc-conjugated anti-IFNg (XMG1.2, Biolegend), Pe-conjugated anti-T-bet (4B10 Ebioscience), Pe-conjugated anti-Blimp1 (5E7, Biolegend), Pe-conjugated anti-c-Maf (sym0F1, Ebioscience), Pe-conjugated anti-Egr2 (erongr2, Ebioscience), Pe-conjugated anti-gp49 (H1.1, Biolegend), Alexa Fluor 647-conjugated anti-Granzyme B (GB11, Biolegend), Pe-conjugated anti-IL-21 (FFA21, Ebioscience), Fitc-conjugated anti-Gata3 (REA174, Miltenyi) and APC-conjugated Streptavidin (Biolegend). Non-specific binding was blocked with 4 μg of rat IgG/ 1x106 cells. Staining of transcription factors was performed using the Foxp3/Transcription Factor Staining Buffer Set (Ebioscience). For intracellular cytokine staining 2x106 cells/well were stimulated in in RPMI-1640 media (Invitrogen), containing 100 U/mL Penicillin, 100 μg/ mL Streptomycin, 2 mM L-Glutamine, 0.5 μg/mL PMA and 1 μg/mL Ionomycin (both Sigma) at 37°C and 5% CO2 for 4 hours. Brefeldin-A (Invitrogen) was added at a final concentration of 10 μg/mL for the final 2 hours. Dead cells were excluded by the Zombie Aqua Fixable Viability kit (Biolegend), and the cells fixed and permeabilized using Biolegend Fixation Buffer in combination with the Intracellular Permeabilisation Wash Buffer. Flowcytometric acquisition was performed by using a FACS Canto II (BD Biosciences), data was analysed using Flowjo Software (Tree Star).

L. sigmodontis specific antibody ELISA

ELISA plates (NUNC) were coated with 5 μg/ml LsAg diluted in 0.45M NaHCO3/0.18M Na2CO3 (Sigma-Aldrich). Plates were incubated with serial dilutions of serum, and a representative dilution from the linear section of the dilution curve selected for each isotype (1/3200 for IgM and IgG2a, 1/6400 for IgG1). Detection of Ab isotypes was performed using HRP-conjugated anti-mouse IgG1, IgG2a or IgM (Southern Biotechnology Associates) and ABTS peroxidase substrate system (KPL).

Adoptive transfers

CD4 T cells were purified by magnetic positive selection using anti-CD4 micro beads (Miltenyi Biotec). Sorted cells were > 93% positive for CD4. Due to limiting cell numbers, tLN and PleC cells were pooled from 12–18 infected mice for transfer into 9 recipients. The proportion of CD4+IL-4gfp+ Th2 cells in the day (d) 20 and d 60 samples were quantified by flow cytometry, and samples adjusted to transfer 4.3x105 d 20 or d 60 CD4+IL-4gfp+ cells i.v. into wild-type BALB/c recipients. Recipients were challenged with L. sigmodontis 2-weeks post-transfer, and autopsies performed 3-weeks post-transfer. Parasite numbers were quantified to confirm infections.

RNA preparation and microarray

CD4 T cells were enriched from the PleC of L. sigmodontis infected mice, tLN of L. sigmodontis and N. brasiliensis infected mice, and spleen of naïve mice by magnetic negative selection (DynaMag) using anti-CD8 (53–6.72), anti-CD19 (ID3), anti-MHC class II (M5/114.15.2), and anti-Mac1 (M1/70) followed by sheep anti-rat beads (Invitrogen). CD4+IL-4gfp+ and CD4+IL-4gfp+CXCR5- Th2 cells were then purified from the enriched PleC and tLN CD4+ populations using a FACSAria flow sorter (BD Biosciences). Naïve splenic T cells were sorted based on CD4+IL-4gfp-CXCR5-CD25-CD44-CD62L+CD45RB+. Due to limiting cell numbers, each biological replicate was created by pooling cells from 20–31 mice for day 20 L. sigmodontis infection, 16–21 mice for day 60 L. sigmodontis infection, 20–23 for N. brasiliensis infection, and 3 naïve mice.

Purities of sorted cells were: naïve spleen (97–99%), N. brasiliensis tLN (94–97%), L. sigmodontis d 20 tLN (87–96%), L. sigmodontis d 60 tLN (94–98%), L. sigmodontis d 20 PleC (91–98%), and L. sigmodontis d 60 PleC (96–99.5%). Biological replicates were created using independent infections. Purified cells were shock-frozen and stored at -80°C until RNA purification using the RNeasy Kit (Qiagen) and RNA quantitated by Qubit (ThermoFisher Scientific). Samples were processed and hybridised to an Affymetrix 2.1 ST mouse array by Edinburgh Genomics. All subsequent array data processing and analyses were undertaken in the R environment using Bioconductor packages. Array quality was assessed using the arrayQualityMetrics package in Bioconductor to identify outliers [24]. One replicate failed the hybridisation process and 2 replicates failed quality control resulting in 5 independent biological replicates for naïve spleen, N. brasiliensis tLN, d 20 L. sigmodontis tLN, and d 60 L. sigmodontis tLN, 4 independent replicates for d 20 L. sigmodontis PleC, and 3 independent replicates for d 60 L. sigmodontis PleC.

Microarray data analysis

Data were normalized using the robust multi-array average expression measure [25]. Pair-wise group comparisons, using the linear modelling methods, with subsequent Bayesian approaches, provided in the limma Bioconductor package, were used to assess differential gene expression, with p-value adjustment for multiple testing. Adjusted p<0.05 was used to define significance for all gene array analysis. Fold-changes (log2-based) between groups, with filtering by adjusted (adj) P value (adj. p<0.05) as appropriate, were plotted using the heatmap.2 function. Principal component analyses were undertaken separately for all normalised and statistically significant data using the prcomp function prior to plotting using scatterplot3d. Correlations were performed on log2 normalised sample data using Standard Pearson correlation. Gene array data is accessible at the Gene Expression Omnibus with accession code GSE114308. To identify genes expressed by exhausted T cells during LCMV infection [26] we downloaded the GEO GSE41870 dataset, and identified genes that significantly changed (adj. p < 0.05) between effector (D8, Armstrong) and exhausted (D30, Clone 13) CD4+ T cells. To identify genes expressed by anergic T cells [27] we used the GEO GSE46242 dataset, and identified genes that significantly changed (adj. p < 0.05) between control EV and Anergic EV samples.

In vivo IL-21R blockade

Mice received i.p. injections of 500 μg anti-IL-21R antibody (4A9, Bioxcell), or an equivalent dose of rat IgG (Sigma-Aldrich), on d 45, 48, 52, and 55 pi.

Statistics for non-microarray data

Power calculations for group sizes were performed using G*power. Statistical analysis was performed using JMP version 12 (SAS). Parametric analysis of combined data from multiple repeat experiments, or of experiments containing more than two groups, was performed using ANOVA followed by Tukey’s post-hoc tests when required. Adult parasite numbers were analysed using a GLM with a poisson distribution. When using two-way ANOVA or GLM to combine data from multiple experiments, experimental effects were controlled for in the analysis and it was verified that there were no significant qualitative interactions between experimental and treatment effects.

Results

Intrinsically hypo-responsive Th2 cells retain their dysfunctional phenotype in the absence of antigen and active infection

Filarial parasites are highly immunosuppressive and it is not known whether Th2 cell-intrinsic hypo-responsiveness is actively maintained by the parasite. The dependence of hypo-responsive Th2 cells on antigen is also unknown. Using IL-4gfp 4get reporter mice to track Th2 cells, we previously demonstrated that IL-4gfp+ Th2 cells become functionally hypo-responsive between days (d) 20 and 60 of L. sigmodontis infection, showing a progressive loss in their intrinsic ability to proliferate and produce Th2 cytokines [19,20,28]. Thus, day 20 pi represents a time point before the onset of hypo-responsiveness when Th2 cells are still functionally active, whilst at d 60 pi Th2 cells are intrinsically hypo-responsive [19].

To test whether the maintenance of L. sigmodontis-induced Th2 cell-intrinsic hypo-responsiveness requires the presence of antigen or active infection, or is stably maintained in their absence, we purified CD4+ T cells from L. sigmodontis infected BALB/c IL-4gfp mice at d 20 and 60 post-infection (pi) and adoptively transferred them into WT recipients. IL-4gfp mice are a valuable tool for tracking Th2 cells because they report IL-4 mRNA independently of IL-4 protein, and retain GFP expression for at least 4-weeks after clearance of infection [19,29]. Whole CD4+ T cells were pooled from the infection site, the pleural cavity (PleC), and the LN within the thoracic cavity (tLN) that drain the PleC [19,20], as Th2 cell-intrinsic hypo-responsiveness occurs at both locations and cell numbers were limiting. GFP expression was used as a surrogate marker allowing us to specifically track and functionally assess the transferred L. sigmodontis-elicited IL-4 mRNA+ Th2 cells. The total number of transferred CD4+ T cells was adjusted to ensure transfer of equal numbers of d 20 and d 60 CD4+IL-4gfp+ T cells. Recipients were challenged with L. sigmodontis two-weeks post-transfer or left unchallenged. The survival and function of IL-4gfp+ Th2 cells recovered from the PleC, brachial LN draining the skin inoculation site, and tLN, were assessed at d 7 post-challenge (Fig 1A).

Fig 1. Hypo-responsive Th2 cells retain their dysfunctional phenotype in the absence of antigen.

Fig 1

CD4+ T cells from L. sigmodontis infected IL-4gfp mice were isolated at d 20 (circles) and d 60 (squares) pi and transferred into naive WT recipients. WT recipients were either left unchallenged (open symbols), or challenged (closed symbols) with L. sigmodontis 2-weeks post-transfer, and IL-4gfp+ Th2 cell function assessed 3 weeks post-transfer. Figures show data from two independent experiments (n = 5–6 for unchallenged, n = 12 for challenged). Symbols represent individual animals, and bars show mean values. (A) Schematic of experimental approach. (B) Total numbers of CD4+IL-4gfp+ donor cells within the PleC, tLN, and brachical LN of recipient. *p<0.05, **p<0.01 (ANOVA based on combined data from two independent experiments). (C) Flow plots show representative expression of CD62L and CD44 by donor CD4+IL-4gfp+ Th2 cells recovered from the PleC. Figures show proportions of CD44+CD62L+ and CD4+CD62L- CD4+IL-4gfp+ Th2 cells. (D) Proportion of CD4+IL-4gfp+ PleC Th2 cells producing IL-5. **Significant effect of timepoint, p<0.05 (ANOVA based on combined data from two independent experiments). (E) Proportion of CD4+IL-4gfp+ PleC Th2 cells producing IL-4. *Significant effect of timepoint, p<0.05 (ANOVA based on combined data from two independent experiments).

In the absence of L. sigmodontis challenge, transferred d 20 and d 60 CD4+IL-4gfp+ T cells were detectable in the brachial LN, tLN, and PleC, and showed equivalent survival after three weeks (Fig 1B). Similarly, they showed equivalent expression of memory markers, with the majority of transferred IL-4gfp+ Th2 cells displaying a CD62L-CD44hi effector memory phenotype or CD62L+CD44hi activated/central memory phenotype, independently of whether or not they received challenge infection (Fig 1C).

Upon challenge with L. sigmodontis, functional differences became apparent, and d 60 CD4+IL-4gfp+ Th2 cells showed a significantly reduced ability to expand (Fig 1B), and produce IL-4 and IL-5 protein (Fig 1D & 1E). Thus, hypo-responsive d 60 IL-4gfp+ Th2 cells showed equivalent survival and expression of memory markers compared to active d 20 Th2 cells upon transfer to a naïve recipient. However, they retained their dysfunctional phenotype, confirming an intrinsic functional change and that this phenotype is maintained for at least 2 weeks in the absence of antigen and active infection.

Intrinsically hypo-responsive Th2 cells and classically active Th2 cells have divergent transcriptional profiles

To investigate whether Th2 cell-intrinsic hypo-responsiveness associates with the development of a unique gene expression profile, an Affymetrix 2.1 mouse gene ST array platform was used to compare mRNA expression of CD4+IL-4gfp+ Th2 cells purified from the PleC and tLN of L. sigmodontis-infected mice pre- (d 20) and post- (d 60) onset of hypo-responsiveness (Fig 2A & 2B, S1A Fig). TLN Th2 cells were also defined as being CXCR5- to exclude CXCR5+ T follicular helper (Tfh) cells, which are IL-4gfp+ during helminth infection [30,31]. To act as a reference effector Th2 cell population, CD4+IL-4gfp+CXCR5- Th2 cells were purified from the tLN of mice infected with Nippostrongylus brasiliensis (S1B Fig). N. brasiliensis is cleared within 7–9 days, stimulates a prototypical acute Th2 response, and the tLN drain the lung migration stage of infection. To identify the gene expression profile of naïve T cells, CD4+IL-4gfp-CXCR5-CD25-CD44-CD62L+CD45RB+ T cells were purified from the spleens of uninfected mice (S1C Fig).

Fig 2. Hypo-responsive Th2 cells have a distinct gene expression profile.

Fig 2

(A) Schematic showing time points at which active and hypo-responsive IL-4gfp+ Th2 cells were purified from L. sigmodontis infected mice. (B) Representative flow plots showing pre- and post- sorts for CD4+IL-4gfp+ Th2 cells from the PleC of L. sigmodontis infected mice at d 20 and d 60 pi. (C) Unbiased hierarchical clustering of IL-4gfp+ Th2 cells purified from the PleC and tLN of mice infected with L. sigmondontis (Ls) and from the tLN of N. brasiliensis (Nb) infected animals d 6 pi., and naïve splenic IL-4gfp-CD4+ T cells. Clustering was performed based on Euclidean distance using the fold change of loci that exhibited an 8-fold or greater change in one or more comparisons. (D) Principal component analysis based on all normalised data. Symbols represent individual samples. (E) Venn diagrams showing the number of loci from L. sigmodontis (Ls) and N. brasiliensis (Nb) Th2 cells that significantly differ compared to naïve (adjusted p<0.05).

To determine the relatedness of the individual samples to each other we performed unsupervised clustering using hierarchical analysis (Euclidean distance measure) with normalised gene expression levels as input. The individual samples clustered back into their original groups demonstrating the reliability of the gene expression data, and that the transcriptional differences between groups are greater than between biological replicates (Fig 2C). As expected, naïve CD4+ T cells had a distinct mRNA expression profile to the IL-4gfp+ Th2 cell populations and clustered separately. This was confirmed by principal component analysis, which also showed that N. brasiliensis Th2 cells clustered separately from L. sigmodontis Th2 cell populations indicating infection-specific transcriptional changes (Fig 2D). IL-4gfp+ Th2 cells from the tLN and PleC of L. sigmodontis infected mice also showed different transcriptional profiles indicating location specific gene expression (Fig 2C & 2D). Importantly, the transcriptional profiles of L. sigmodontis IL-4gfp+ Th2 cells differed over time, consistent with their phenotype change. In particular, the hypo-responsive d 60 PleC IL-4gfp+ Th2 cells segregated separately from all the other Th2 cell populations, including the d 20 PleC IL-4gfp+ Th2 cells (Fig 2C & 2D).

Th2 cells from different contexts show both common and specific gene expression profiles [32]. Similarly, based on PleC IL-4gfp+ Th2 cells, 4124 common loci significantly changed upon infection with L. sigmodontis and N. brasiliensis, whilst 4155 and 2308 unique loci significantly changed upon L. sigmodontis and N. brasiliensis infections respectively (Fig 2E). A similar pattern was seen for tLN IL-4gfp+ Th2 cells, although there were fewer L. sigmodontis unique loci. Only 51% of loci that significantly changed in PleC IL-4gfp+ Th2 cells at d 60 of L. sigmodontis infection were common with those that changed at d 20 pi.

These transcriptional changes over time during L. sigmodontis infection associated with the differential expression of transcription factors. Investigating transcription factors that had significantly differential gene expression compared with naïve T cells demonstrated that d 20 and d 60 IL-4gfp+ Th2 cells had unique expression of 43 and 138 transcription factors respectively (S2 Fig), and had 152 commonly expressed transcription factors (S3 Fig). Day 60 IL-4gfp+ Th2 cells also showed enrichment of particular KEGG pathways compared with d 20 IL-4gfp+ Th2 cells (S1 Table). In particular, d 60 PleC IL-4gfp+ Th2 cells showed enrichment of metabolism pathway genes, and both d 60 PleC and tLN IL-4gfp+ Th2 cells showed distinct enrichment of DNA repair pathway genes. Altogether this indicates that intrinsically hypo-responsive d 60 IL-4gfp+ Th2 cells are transcriptionally divergent from classically active Th2 cells from both d 20 of L. sigmodontis infection and from N. brasiliensis infection.

Hypo-responsive Th2 cells retained a Th2 transcriptional profile, while a subset upregulated T-bet and produced IFN-γ

The loss of Th2 cytokines by hypo-responsive IL-4gfp+ Th2 cells could indicate a switch in phenotype towards a different T cell subset rather than a manifestation of Th2 cell dysfunction. To address this, the expression of the major transcription factors for Th2 (Gata3, Irf4), Th1 (Tbx21), Th9 (Spi1), Th17 (Rorc), Treg (Foxp3), Tr1 (Irf1), and Tfh (Bcl6) cells were assessed. All IL-4gfp+ Th2 cells from L. sigmodontis and N. brasiliensis infected mice showed significant upregulation of the Th2 transcription factors Gata3 and Irf4 compared to naïve T cells, and their expression remained constant between d 20 and d 60 of L. sigmodontis infection (Fig 3A). Bcl6 was significantly increased in CD4+IL-4gfp+CXCR5- tLN T cell populations from both L. sigmodontis and N. brasiliensis infected mice, suggesting they may contain IL-4gfp+Bcl6+CXCR5- Tfh cells. Importantly, neither d 20 or d 60 IL-4gfp+ Th2 cells from the PleC, where Th2 cell-intrinsic hypo-responsiveness predominates, showed elevated Bcl6 mRNA compared with naïve T cells. Thus, the hypo-responsive PleC d 60 Th2 population did not take on the characteristics of Tfh cells.

Fig 3. A subset of d 60 IL-4gfp+ Th2 cells take on Th1 characteristics.

Fig 3

(A & B) Heatmaps showing fold change in expression of transcription factors (A) and cytokines (B) in L. sigmodontis (Ls) and N. brasiliensis (Nb) IL-4gfp+ cells relative to naïve IL-4gfp-CD4+ T cells, or in d 60 L. sigmodontis IL-4gfp+ Th2 cells relative to d 20. *adj. p<0.05). (C) Representative staining for production of IL-4, IL-5 and IFN-γ by PleC IL-4gfp+ from naïve and L. sigmodontis infected mice (d 20 and d 60 pi) (D) Proportion of IL-4gfp+ Th2 cells from the PleC of naïve and L. sigmodontis infected mice singly or co-expressing IL-4 and IFN-γ, or IL-5 and IFN-γ. Data shown from one representative experiment of two (n = 3 for naïve and 6 for L. sigmodontis infected mice). (E) Proportion of IL-4gfp+ Th2 cells from the PleC of naïve (open symbols) or d 20 and d 60 L. sigmodontis infected mice (closed symbols) producing IFN-γ. Symbols show individual mice and bars represent mean values. Graph shows combined data from two independent experiments. ***Significant effect between groups (p<0.001, ANOVA), # p<0.05 (Tukey’s HSD). (F) Proportion of PleC CD4+ T cells from naïve and L. sigmodontis infected mice expressing T-bet. Symbols show individual mice and bars represent mean values. Graph shows combined data from two independent experiments. ***Significant effect between groups (p<0.001, ANOVA), # p<0.05 (Tukey’s HSD). (G) CD4+ T cells from L. sigmodontis infected IL-4gfp mice were isolated at d 20 (circles) and d 60 (squares) pi and transferred into naive WT recipients. Recipients were either left unchallenged (open symbols), or challenged (closed symbols) with L. sigmodontis two weeks post-transfer, and the proportion of PleC IL-4gfp+ Th2 cells producing IFN-γ assessed 3 weeks post-transfer. *Significant effect of time point independent of challenge status (p<0.05, ANOVA based on combined data from two independent experiments).

Expression of mRNA for Spi1 and Rorc were not significantly upregulated in any of the IL-4gfp+ T cell populations relative to naïve T cells. Irf1 mRNA levels were significantly reduced in IL-4gfp+ Th2 cells compared to naïve T cells, although there was a significant increase between d 20 and 60 of L. sigmodontis infection. Interestingly, despite impaired production of Th2 cytokines and IL-10 at the protein level [19,20], expression of Il5 mRNA remained constant between d 20 and 60, and expression of Il10, Il13, and Il4 mRNA significantly increased in PleC IL-4gfp+ Th2 cells (Fig 3B). There was no significant upregulation of mRNA for IL-9 or IL-17 during L. sigmodontis infection. Thus, the dysfunctional d 60 IL-4gfp+ T cell population does not take on characteristics of Tfh, Th9, Th17, Tr1, or Foxp3+ Treg populations, and instead retains a typical Th2 cell mRNA phenotype with the inhibition of Th2 cytokines occurring post-transcriptionally.

Tbx21 (T-bet) was significantly upregulated between d 20 and 60 of L. sigmodontis infection (Fig 3A), and Ifng was the most upregulated loci (27-fold, adjusted p = 4.1e-6) between PleC d 60 and d 20 IL-4gfp+ Th2 cells (Fig 3B). Consistent with the development of a Th1 phenotype, Ingenuity Pathway Analysis (IPA) predicted IFN-γ (p = 2.7e-24, F = 4.1) (S4 Fig), IL-12 (complex) (p = 2.7e-24, F = 3.9), IL-12A (p = 6.93e-11, F = 2.6), IL-12B (p = 6.93e-11, F = 2.6), and IL-18 (p = 3.17e-11, F = 3.2) to be activated upstream regulators, explaining some of the observed gene expression changes between d 60 and d 20 PleC IL-4gfp+ Th2 cells.

Flow cytometry was used to test whether cells with a mixed Th1/Th2 phenotype develop during L. sigmodontis infection, as described for other helminth infections [33]. In naïve mice, just under 40% of PleC IL-4gfp+ Th2 cells produced IFN-γ protein (Fig 3C–3E), with the majority of IFN-γ+ cells co-producing IL-4 (Fig 3C & 3D). Upon L. sigmodontis infection, the proportion of PleC IL-4gfp+IFN-γ+ Th2 cells significantly reduced to less than 5% at d 20 consistent with a dominant Th2 response at this time point, before significantly increasing during the hypo-responsive stage at d 60 pi (Fig 3C–3E). Although co-staining for T-bet and IL-4gfp was not possible, expression of T-bet by CD4+ T cells significantly increased between d 20 and 60 pi (Fig 3F). The IFN-γ production by d 60 IL-4gfp+ Th2 cells was retained following transfer to a naïve recipient and rechallenge with L. sigmodontis (Figs 1A & 3G), indicating that the hypo-responsive IL4gfp+ Th2 cells also stably retained a Th1 phenotype.

During L. sigmodontis infection the d 60 IL-4gfp+ Th2 cells that produced IFN-γ were distinct from those producing IL-4 and IL-5 (Fig 3C & 3D), contrasting with the published Th1/Th2 hybrid phenotype [33]. Importantly, the loss of IL-4 (15.8%) and IL-5 (16.4%) producing Th2 cells was greater than the gain in IFN-γ producing Th2 cells (5.5%) (Fig 3D), indicating that Th2 cell intrinsic hypo-responsiveness does not represent a switch to a Th1 phenotype. This suggests that a proportion of the hypo-responsive d 60 IL-4gfp+ Th2 population took on Th1 characteristics. However, as a whole, the d 60 IL-4gfp+ Th2 cells retained a Th2 phenotype and a Th2 to Th1 switch was not responsible for their dysfunctional phenotype.

Intrinsically hypo-responsive CD4+ Th2 cells express Blimp-1 and c-Maf, but have a distinct transcriptional profile to exhausted CD4+ T cells

PD-1 co-inhibition is a key mechanism of T cell exhaustion [34], suggesting that PD-1-dependent Th2 cell-intrinsic hypo-responsiveness represents a form of exhaustion. To investigate this hypothesis, we compared the gene expression profiles of hypo-responsive Th2 cells and exhausted CD4+ T cells. Published CD4+ T cell gene expression data from LCMV infection was used as a well-defined exhaustion model [26]. We initially assessed whether intrinsically hypo-responsive Th2 cells and exhausted CD4+ T cells upregulate the same transcription factors. Of the 17 exhaustion-associated transcription factors, only 5 (Maf, Mcm6, Baz1a, Mdfic, and Prdm1) were significantly upregulated in PleC IL-4gfp+ Th2 cells between d 20 and 60 pi, and all five showed similar expression levels in our reference active N. brasiliensis IL-4gfp+ Th2 cells (Fig 4A). Expression of Blimp1 and c-Maf protein did correlate with Th2 cell hypo-responsiveness as they were highly expressed in CD4+ T cells at d 60, but not d 20, pi (Fig 4B & 4C), and IPA predicted that c-Maf was an activated upstream regulator of PleC d 60 IL-4gfp+ Th2 cells (z-score = 2.2, p = 6e-6). However, d 60 IL-4gfp+ Th2 cells showed an inverse gene expression pattern to exhausted CD4+ T cells (r = -0.170, p<0.001), based on genes that are significantly differentially expressed in exhausted CD4+ T cells [26] (Fig 4D). Thus, hyporesponsive Th2 cells and exhausted T cells have common expression of Blimp1 and c-Maf, but overall show inverse gene expression profiles indicating that they represent distinct T cell activation states.

Fig 4. Hypo-responsive Th2 cells are distinct from exhausted CD4+ T cells.

Fig 4

(A) Heatmap showing fold change in expression of exhaustion-associated transcription factors in L. sigmodontis (Ls) and N. brasiliensis (Nb) IL-4gfp+ cells relative to naïve IL-4gfp-CD4+ T cells, or in d 60 L. sigmodontis IL-4gfp+ Th2 cells relative to d 20. *adj. p<0.05. (B & C) Proportion of PleC CD4+ T cells expressing Blimp1 (B) and c-Maf (C) in naïve and L. sigmodontis infected mice at 20 and 60 d pi. Symbols represent individual animals, bars show mean. Naïve animals from d 20 and d 60 timepoints were pooled. Combined data from two experiments shown. ***Significant group effect (p<0.001, ANOVA using combined data from both experiments), # p<0.05 (Tukey HSD). (D) Fold-change of genes significantly differentially expressed (adj. p<0.05) in exhausted CD4+ T cells correlated against their fold-change expression in hypo-responsive Th2 cells (Pearson Rank Correlation).

Th2 cell-intrinsic hypo-responsiveness has similarities with T cell anergy and tolerance

Immune suppression during filariasis and other helminth infections is associated with increases in T cell anergy markers, including upregulation of the E3 ubiquitin ligases Nedd4, Itch, C-cbl, Cbl-b, and GRAIL [1316]. To test whether Th2 cell-intrinsic hypo-responsiveness represents a form of anergy, the expression of anergy-associated E3 ubiquitin ligases was assessed (Fig 5A). Nedd4 and Rnf128 (GRAIL) mRNA were significantly increased in PleC IL-4gfp+ Th2 cells at both d 20 and d 60 of L. sigmodontis infection in comparison to naïve CD4+ T cells, and were not significantly upregulated in N. brasiliensis IL-4gfp+ Th2 cells. However, their expression did not change significantly between d 20 and d 60 of L. sigmodontis infection. Ndfip1 was significantly upregulated during L. sigmodontis infection, and its expression significantly increased between d 20 and d 60 (Fig 5A). However, it was also upregulated to a similar extent in in N. brasiliensis IL-4gfp+ Th2 cells. Expression of other anergy-associated ubiquitin ligases (Rc3h1, Cbl, Cblb, Cblc, Itch, and Traf6) either decreased upon L. sigmodontis infection or remained unchanged.

Fig 5. Hypo-responsive Th2 cells have similarities with T cell anergy and tolerance.

Fig 5

(A) Heatmap showing fold change in expression of anergy-associated ubiquitin ligases in L. sigmodontis (Ls) and N. brasiliensis (Nb) IL-4gfp+ cells respectively, relative to naïve IL-4gfp-CD4+ T cells, and in d 60 L. sigmodontis IL-4gfp+ Th2 cells relative to d 20 and N. brasiliensis Th2 cells. Data is from PleC IL-4gfp+ Th2 cells for L. sigmodontis infection. *adjusted p<0.05. (B) Representative flow staining and proportion of CD4+ T cells expressing Egr2 in naïve and L. sigmodontis infected mice 20 and 60 d pi. Figure shows data from two independent experiments. Symbols represent individual animals and bars show means. *** Significant difference between groups (p<0.001, ANOVA using combined data from both experiments), # p<0.05 (Tukey HSD). (C) Fold-change of genes significantly up- or down- regulated in anergic CD4+ T cells correlated against their fold-change expression in hypo-responsive Th2 cells (Pearson Rank Correlation). (D) Unbiased hierarchical clustering of IL-4gfp+ Th2 cells purified from the PleC and tLN of mice infected with L. sigmondontis (Ls) and from the tLN of N. brasiliensis (Nb) infected animals. Clustering was performed based on Euclidean distance using the fold change of genes that are significantly (adj. p < 0.05) increased in anergic CD4+ T cells.

Egr2 is a key anergy associated transcription factor [27,35], and PleC IL-4gfp+ Th2 cells significantly increased expression of Egr2 protein between d 20 and d 60 pi (Fig 5B). Comparing the fold change of genes that significantly differed in CD4+ T cells following Egr2-dependent anergy induction [27] against their fold-change in IL-4gfp+ Th2 cells between d 20 and d 60 of L. sigmodontis infection showed a significant positive correlation (r = 0.376, p<0.001) (Fig 5C). When compared with d 20, d 60 PleC IL-4gfp+ Th2 cells also significantly upregulated 9 out of the 10 key genes associated with the induction of T cell tolerance following peptide immunotherapy [36], including Lag3, Tigit, Havcr2, Icos, Il10, Il21, Maf, Nfil3, and Ahr. However, L. sigmodontis IL-4gfp+ Th2 cells only upregulated a subset of the total genes upregulated in Egr2-dependent anergy (Fig 5D, S2 Table) or peptide-induced tolerance (S5 Fig & S3 Table), and the d 60 IL-4gfp+ Th2 cells showed similar expression levels to the reference N. brasiliensis IL-4gfp+ Th2 cells. Together this indicates that IL-4gfp+ Th2 cells upregulate some anergy and tolerance associated genes during L. sigmodontis infection, and that d 60 IL-4gfp+ Th2 cells have developed a gene expression profile more similar to that of anergic or tolerised CD4+ T cells compared with d 20 IL-4gfp+ Th2 cells. However, despite a partial overlap with tolerised and anergic T cells, the gross transcriptional profile of Th2 cell-intrinsic hypo-responsiveness was dissimilar to T cell anergy and tolerance.

Identification of immune regulatory genes associated with Th2 cell-intrinsic hypo-responsiveness

1132 and 257 loci significantly changed in PleC and tLN IL-4gfp+ Th2 cells respectively between d 20 and d 60 of L. sigmodontis infection (S4 Table & S5 Table). We predicted that genes responsible for the dysfunctional Th2 cell phenotype should show a different expression profile to the reference N. brasiliensis Th2 cells. 471 of the PleC loci were also significantly differentially expressed compared with N. brasiliensis Th2 cells, and thus formed a core list of candidate loci (S6 Table). Th2 cell-intrinsic hypo-responsiveness is detected to a lesser extent in the tLN, so we predicted that candidate loci should show a similar expression pattern in the tLN, but to a lower level. Thus, the core candidate list was refined based on their expression in the tLN. 308 loci showed a similar expression profile in the tLN, of which 58 changed significantly (S7 Table). 18 loci (17 unique genes) matched our full prediction by having a similar expression pattern in the tLN and being expressed to significantly lower levels in the tLN compared to the PleC (Fig 6A).

Fig 6. Identification of genes associated with Th2 cell dysfunction.

Fig 6

(A) Heatmap showing fold change in expression of candidate genes that significantly differ between d 20 and d 60 of L. sigmodontis (Ls) infection in both the PleC and tLN, have significantly higher expression levels in the PleC versus tLN, and significantly differ from N. brasiliensis (Nb) Th2 cells. (B) Heatmap showing fold change of translation factors that uniquely or commonly significantly differ in L. sigmodontis PleC IL-4gfp+ Th2 cells and N. brasiliensis Th2 cells compared to naïve CD4+ T cells. (C) Heatmaps showing fold-change in expression of core candidate genes with known immune regulatory functions. Fold-change is calculated relative to Th2 cells from d 20 of L. sigmodontis infection, N. brasiliensis Th2 cells, and tLN Th2 cells from d 60 of L. sigmodontis infection. (D & E) Percentage of PleC CD4+ T cells expressing ILT3 (D) and CD4+IL-4gfp+ Th2 cells expressing Granzyme B (E) in naïve and L. sigmodontis infected mice at d 20 and d 60 pi. Graphs show combined data from two independent experiments. Symbols represent individual animals and bars show means. ***Significant effect between groups (p<0.001, ANOVA based on combined data from two independent experiments), # p<0.05 (Tukey HSD). (F) Heatmap showing fold-change in expression of core candidate genes with known secreted immune regulatory functions. Fold-change is calculated relative to Th2 cells from d 20 of L. sigmodontis infection, N. brasiliensis Th2 cells, and tLN Th2 cells from d 60 of L. sigmodontis infection.

As the gene array data indicated post-transcriptional inhibition of Th2 cytokines (Fig 3B) we investigated the expression of translation-associated genes based on KEGG pathways, IPA-defined ‘translation regulators’, and literature searches. Nine translation associated-genes were identified amongst the candidate genes (Pwp2, Riok1, Gtpbp4, Lsg1, Mdn1, Utp20, Paip2b, Nup43, Ears2), of which all but Riok1 and Nup43 were downregulated at d 60 compared with d 20. Interestingly, 28 eukaryotic type translation factors (KEGG Brite Ko03012) were significantly downregulated in d 60 PleC IL-4gfp+ Th2 cells compared with naïve T cells, but only 7 of these were downregulated in N. brasiliensis Th2 cells (Fig 6B). Conversely, 10 translation factors were significantly upregulated in N. brasiliensis Th2 cells, of which only 2 were upregulated during L. sigmodontis infection. However, the expression of these 38 translation factors did not significantly differ between d 20 and d 60 of L. sigmodontis infection, indicating that their expression was specific to infection type rather than the change in Th2 cell phenotype over time. Nevertheless, L. sigmodontis infection associates with a reduction in expression of translation factors in Th2 cells when compared with acute N. brasiliensis infection.

Literature searches were used to identify and group candidate genes with known immune regulatory functions. In common with other forms of T cell dysfunction, various inhibitory receptors were upregulated (Fig 6C). Despite its central role in driving Th2 cell-intrinsic hypo-responsiveness [19], Pd1 was not identified as a core candidate gene as its expression also increased in N. brasiliensis infection. Candidate genes were also identified that inhibit Th2 responses, T cell function and cytokine production, are associated with Treg suppression, and that play important roles in the development of immune tolerance (Fig 6C). ILT3 (Lilrb4) is expressed by Foxp3+ Tregs during L. sigmodontis infection [37], and we confirmed that expression of ILT3 protein was significantly increased on the surface of d 60 CD4+ Th2 cells (Fig 6D).

Granzyme B inhibits protective immunity to L. sigmodontis [38], and was upregulated by PleC Th2 cells between d 20 and 60 pi, both at the mRNA (13-fold increase, p<9e-7), and protein levels (Fig 6E). However, Granzyme B was not represented in the core candidate gene list as it was also highly upregulated in N. brasiliensis Th2 cells compared to naive (37-fold, adj. p < 1.6e-13). Furin is a pro-TGF-β1 converting enzyme [39], and TGF-β signalling genes (Snx9 & Skil) were upregulated in the core candidate gene list. IPA upstream regulator analysis of the core PleC candidate genes, based on fold change between d 60 and d 20 pi, indicated that the TGF-β1 pathway was significantly enriched and predicted to be activated (p = 1.58e-14, F = 2.0) (S6 Fig).

The candidate gene list also contained a range of secreted factors with immune regulatory functions (Fig 6F). Of these, IPA identified CD38 (p = 3.4e-13), IL-21 (p = 4.93e-18), CSF1 (p = 2.25e-11), Spp1 (p = 2.3e-4), and Lgals3 (p = 2.4e-6) as potential upstream regulators of the candidate genes, suggestive of autocrine activity. Furthermore, the CD38 (F = 3.3), CSF1 (F = 2.8) and IL-21 (F = 2.0) pathways were predicted to be activated. This suggests that intrinsically hypo-responsive Th2 cells are not functionally inert, and have developed alternate regulatory or effector functions.

IL-21 inhibits protective and humoral immunity to L. sigmodontis

Our data suggests that, in contrast to their loss of ability to produce classical Th2 cytokines (IL-4, IL-5) [19], Th2 cells gain IL-21 production as they become hypo-responsive (Fig 6F). This switch from a Th2 response towards an IL-21 response indicated that IL-21 is playing a prominent role in regulating immunity to L. sigmodontis infection during chronicity. To confirm IL-21 expression at the protein level, we used flow cytometry to assess IL-21 production by CD4+ T cells at d 20 and 60 of L. sigmodontis infection. In agreement with the gene array data, PleC CD4+ T cells did not produce IL-21 during the active Th2 phase of infection (d 20 pi), but did produce IL-21 protein at d 60 pi, during the hypo-responsive phase of infection (Fig 7A & 7B).

Fig 7. IL-21R blockade increases resistance to L. sigmodontis.

Fig 7

(A) Representative flow staining showing IL-21 production by PleC CD4+ T cells from naïve, and L. sigmodontis infected mice. (B) IL-21 protein production by PleC CD4+ T cells from naïve and L. sigmodontis infected mice at d 20 and d 60 pi. Graph shows combined data from two independent experiments. Symbols represent individual animals and bars show means. ***Significant effect between groups (p<0.001, ANOVA using combined data from two independent experiments), # p<0.05 (Tukey HSD). (C–J) L. sigmodontis infected BALB/c mice were treated with a neutralising anti-IL-21R or rat IgG antibody between d 45 and 60 pi, and autopsied at d 60. Panels show combined data from two independent experiments with 5–6 mice per group. Symbols represent individual animals and bars show means. **p < 0.01, *** p < 0.001 (ANOVA using combined data from two independent experiments), # p < 0.05 (Tukeys HSD). (C) Number of Mf/ml of blood, and number of adult L. sigmodontis parasites recovered from the pleural cavity. (D) Number of PleC CD4+GATA-3+ T cells. (E) Percentage of PleC CD4+GATA-3+ T cells producing IL-4 or IL-5 protein. (F) Percentage of PNA+CD19+ B cells within the tLN. (G) Serum levels of L. sigmodontis specific IgM and IgG1. (H) Total number of PNA+CD19+ B cells within the tLN. (I) Total number of CD4+Bcl6+CXCR5+ Tfh cells within the tLN. (J) Total number of CD19+IgM-IgD- class-switched B cells within the tLN.

To test the role of IL-21 during the hypo-responsive phase of L. sigmodontis infection, IL-21 activity was neutralised in WT BALB/c mice between days 45 and 60 of L. sigmodontis infection using an anti-IL-21R blocking antibody. IL-21R blockade increased resistance to L. sigmodontis infection to a similar extent as PD-1 blockade as it significantly reduced the levels of blood microfilaria without impacting the number of adult parasites recovered (Fig 7C) [19].

To determine whether IL-21 is involved in the maintenance of Th2 cell hypo-responsiveness, we assessed whether IL-21 blockade enhanced Th2 cell expansion or ability to produce cytokines. IL-21R blockade did not increase the number of CD4+GATA-3+ Th2 cells within the PleC (Fig 7D) or recover the ability of CD4+GATA-3+ Th2 cells to produce IL-4 or IL-5 protein (Fig 7E). This indicates that IL-21 does not maintain Th2 cell-intrinsic hypo-responsiveness, and that the increase in resistance was not due to restoration of Th2 cell function.

As IL-21 is typically associated with promoting humoral immunity [40], we assessed whether it regulated Tfh and B cell responses to L. sigmodontis. IL-21R blockade significantly increased the percentage of CD19+PNA+ germinal centre B cells (Fig 7F), as well as serum levels of L. sigmodontis specific IgG1 (Fig 7G). Although this did not translate to increased total numbers of CD19+PNA+ germinal centre B cells (Fig 7H), it suggests that IL-21 inhibits L. sigmodontis specific B cell responses. IL-21 blockade had no effect on the expansion of CD4+Bcl6+CXCR5hi Tfh cells (Fig 7I), and did not change the numbers of CD19+IgM-IgD- class-switched B cells (Fig 7J). Thus, IL-21 inhibits protective and humoral immunity to L. sigmodontis, but is not involved in the maintenance of Th2 cell-intrinsic hyporesponsiveness.

Discussion

T cell-intrinsic regulation, such as anergy, adaptive tolerance, exhaustion, and plasticity, is an important way in which immune responses are controlled. Whilst Th2 cell-intrinsic regulation is known to develop during helminth infections [11,1619], and controls both resistance to infection and pathology [16,19], its phenotype is not well described. In this study we demonstrate that during L. sigmodontis infection the development of Th2 cell-intrinsic hypo-responsiveness associates with a switch from a classical IL-4+IL-5+IL-13+ Th2 cell phenotype at d 20 pi, to a non-classical Egr2+cMaf+Blimp-1+IL-4loIL-5loIL-13loIL-21+T-bet+IFN-γ+ Th2 cell phenotype at d 60 pi. The d 60 hypo-responsive IL-4gfp+ Th2 population had a divergent transcriptional profile to classical Th2 cells isolated at d 20 of L. sigmodontis infection or from mice acutely infected with N. brasiliensis. Transfer studies demonstrated that the hypo-responsive Th2 cells retained their dysfunctional phenotype in the absence of antigen and active infection. This indicates that Th2 cell-intrinsic hypo-responsiveness represents a distinct and stable state of Th2 cell differentiation.

Identifying shared traits of dysfunctional Th cells in diverse chronic settings is beneficial for the development of novel therapeutics. Our analysis is the first to compare Th2 cell-intrinsic dysfunction to known forms of T cell tolerance and unresponsiveness to find shared regulatory mechanisms. Hypo-responsive Th2 cells had dissimilar overall transcriptional profiles to exhausted, anergic, and tolerised CD4+ T cells, indicating the development of a novel form of T cell dysfunction. In particular, showing an inverse gene expression pattern to exhausted CD4+ T cells. As Th2 cell-intrinsic dysfunction is PD-1 dependent [19], this indicates that PD-1 co-inhibition leads to non-exhaustion forms of T cell dysfunction in Type 2 settings. Dysfunctional Th2 cells shared specific characteristics with these forms of T cell dysfunction, and compared to active d 20 Th2 cells had a more similar expression profile to anergic T cells. Hypo-responsive Th2 cells upregulated Blimp-1 and c-Maf transcription factors, which associate with inhibition of proliferation and increased IL-10 production respectively [4143]. They also switched on the transcription factor Egr2, which plays a central role in the induction of T cell anergy [27,35], and upregulated expression of key genes and proteins associated with tolerance, including Lag3, Tigit, Havcr2, Icos, Il10, IL-21, c-Maf, Nfil3, Ahr, Furin and Ncf1 [36,39,44]. Of these, Furin is involved in the activation of TGF-β from its latent form [39], and TGF-β was predicted to be an upstream regulator of the hypo-responsive Th2 cells suggesting a role in generating or maintaining the hypo-responsive state. Post-transcriptional silencing is a mechanism of shutting down cytokine production in anergic T cells [45], and L. sigmodontis-elicited hypo-responsive Th2 cells retained expression of mRNA for Il4, Il5, Il13, and Il10 indicating that their downregulation is controlled at the translational level. Furthermore, Th2 cells from L. sigmodontis infected mice had reduced expression of translation factors, although their expression did not directly correlate with the onset of hypo-responsiveness.

A tolerised Th2 cell phenotype is consistent with published literature showing that human T cells display characteristics of anergy during helminth infections [1315]. PBMC from human filariasis patients upregulate anergy-associated ubiquitin ligases, including Nedd4 [15], while Th2 dysfunction in murine schistosomiasis is dependent upon GRAIL [16]. Both of these factors were increased in Th2 cells during L. sigmodontis infection, although their expression did not directly correlate with the development of hypo-responsiveness. Egr2 and GRAIL are linked with the development of an anergic-like T cell phenotype in Type 2 (Fasciola hepatica) and Type 1 (Trypanosoma cruzi and Mycobacterium tuberculosis) infection settings [17,46,47]. Thus, Th2 cell-intrinsic hypo-responsiveness shares characteristics with T cell dysfunction in humans and other infection settings. It also may share some common regulatory mechanisms with anergic and tolerised CD4+ T cells, despite indications that they are discrete phenotypes.

Anergy and tolerance can stimulate tolerised-memory, and there is recent evidence that exhausted T cells develop memory [810,48]. L. sigmodontis-elicited hypo-responsive Th2 cells retained their dysfunctional phenotype for 2 weeks upon transfer to a naïve host, indicating that the phenotype is stable in the absence of antigen and extrinsic parasite and host immune regulatory factors. Whilst this is too short a time span to draw firm conclusions, it raises the potential for tolerised Th2 memory cells. The development of a tolerised Th2 memory response may be one element explaining why protective immunity to helminths takes so long to develop [49], and has implications for vaccination of previously infected individuals if vaccine-induced responses have to outcompete pre-established dysfunctional Th2 cells. It could also have beneficial applications, as the ability to induce long-term tolerance in committed allergen-specific Th2 cells would represent an ideal approach for the treatment of allergies.

Despite their dysfunctional Th2 phenotype, hypo-responsive Th2 cells were not functionally inert and upregulated expression of soluble regulatory factors including IL-21, IFN-γ, Fgl2, Cd38, Spp1, Areg, Metrnl, Lgals3, and Csf1. Fgl2 has immune suppressive functions, inhibiting immunity to Echinococcus multilocularis [50], and is associated with suppression by Treg and inhibitory Th2 cells [51,52]. The hypo-responsive Th2 cells upregulated Lag3 mRNA, and Egr2 and Lag3 co-expression defines a population of IL-10+Foxp3- Treg cells [53]. They also switched on IL-21 and c-Maf protein, which play central roles in the differentiation of Tr1 cells [42,43]. Thus, similar to F. hepatica anergised T cells that are inhibitory [18], hypo-responsive Th2 cells may have a regulatory phenotype.

IL-21 has previously been shown to promote Th2, Tfh, and B cell responses, counteract T cell exhaustion, and inhibit Foxp3+ Treg function [40,43]. In contrast, blockade of the IL-21R during the hypo-responsive phase of L. sigmodontis infection increased resistance to infection and enhanced parasite-specific B cell and antibody responses. This indicates a novel role for IL-21 in suppressing Type 2 immune responses and humoral immunity during chronicity. The increased resistance was not associated with a reversal of Th2 cell dysfunction, indicating that IL-21 does not maintain Th2 cell intrinsic hypo-responsiveness, but rather has a downstream regulatory role. Whilst IL-21 can also be produced by Tfh cells, NK cells, and Foxp3+ Tregs, this suggests that L. sigmodontis-elicited hypo-responsive Th2 cells may represent a type of Foxp3- Treg cell, potentially inhibiting protective Type 2 immunity via IL-21.

Mixed Th1/Th2 responses are seen in human filariasis patients [54], and IL-5 and IFN-γ synergise to kill L. sigmodontis parasites [55]. The development of T-bet+IFN-γ+ Th2 cells during L. sigmodontis infection indicates that the IFN-γ response may be partly due to the development of Th2/Th1 hybrid cells. Importantly, the dysfunctional Th2 phenotype was not due to a Th2 to Th1 switch as the loss of Th2 cytokine producing cells was greater than the gain in those producing IFN-γ. The hypo-responsive Th2 population also retains a dominant, albeit muted, Th2 cytokine signature upon in vitro antigenic stimulation [19,20]. IL-21 inhibits T-bet and Th1 differentiation [43], suggesting that the IL-21 secreting Th2 cells and Th2/Th1 hybrids are distinct. Thus, the d 60 hypo-responsive Th2 population may be comprised of different subpopulations, including Th2/Th1 hybrid cells and dysfunctional Th2 cells, which have opposing protective and inhibitory roles respectively.

Th2 cells show heterogeneity between different Type 2 infectious and allergic settings [32], indicating that Th2 cell phenotype is context dependent. Our study shows that Th2 cell phenotype is also heterogeneous over time, and that Th2 cells can switch from a classical Th2 phenotype towards a transcriptionally distinct intrinsically hypo-responsive phenotype that is stably retained in the absence of antigen. We hypothesise that the hypo-responsive Th2 population elicited during L. sigmodontis infection comprises at least two different non-classical Th2 cell variants; a dysfunctional Egr2+cMaf+IL-21+IL-4loIL-5loIL-10loIL-13lo population with similarities to tolerance and regulatory T cells, and an IFN-γ+T-bet+IL-4-IL-5- Th2/Th1 hybrid population that retains mRNA for Gata3, Il4 and Il5. Further studies are now needed to resolve these different subpopulations, and determine whether they represent forms of tolerance, alternative Th2 effector cells, or regulatory Th2 cells. This ability of Th2 cells to change phenotype over time has important implications for understanding the outcomes of helminth infections and allergies. It also raises the prospect of new approaches for therapeutically manipulating committed Th2 cells towards different fates, for example to tolerise allergen-specific Th2 cells.

Supporting information

S1 Fig. Representative flow plots showing sorting strategy and purities for the tLN and spleen.

(PDF)

S2 Fig. Transcription factors that are differentially expressed between d 20 and d 60 PleC IL-4gfp+ Th2 cells during L. sigmodontis infection.

(PDF)

S3 Fig. Transcription factors that are expressed in common by d20 and d60 PleC IL-4gfp+ Th2 cells during L. sigmodontis infection.

(PDF)

S4 Fig. IFN-γ is an upstream regulator of PleC d 60 IL-4gfp+ Th2 cells.

(PDF)

S5 Fig. Comparison of gene expression in peptide-induced tolerance and Th2 cell-intrinsic hypo-responsiveness.

(PDF)

S6 Fig. TGF-β is an upstream regulator of core candidate genes.

(PDF)

S1 Table. Table showing KEGG pathways enriched in PleC and/or tLN IL-4gfp+ Th2 cells at d 60 pi, but not at d 20 pi.

(DOCX)

S2 Table. Table showing fold-change in expression of genes significantly upregulated in anergic T cells that was used to perform the unbiased clustering represent by the heat map in Fig 5D.

(XLSX)

S3 Table. Table showing fold-change in expression of genes significantly upregulated in T cells following peptide-elicited tolerance that was used to perform the unbiased clustering represented in the heat map in S5 Fig.

(XLSX)

S4 Table. Table of loci that significantly differ between d 20 and 60 of L. sigmodontis infection in PleC IL-4gfp+ Th2 cells.

(XLSX)

S5 Table. Table of loci that significantly differ between d 20 and 60 of L. sigmodontis infection in tLN IL-4gfp+ Th2 cells.

(XLSX)

S6 Table. Table of core candidate loci associated with Th2 cell-intrinsic hypo-responsiveness.

(XLSX)

S7 Table. Table showing subset of the core candidate loci that have significantly different expression in tLN IL-4gfp+ Th2 cells between d20 and d60.

(XLSX)

Acknowledgments

We would like to thank Alison Fulton for experimental support and maintaining the L. sigmodontis lifecycle, Martin Waterfall and Central Bioresearch Services for excellent technical support, Rebecca Brownlie, Marta Trüb, Sharon Campbell and Carlos Minutti for their helping hands on big experimental days, Robert Salmond, Judith Allen, Dominik Rückerl, Rose Zamoyska and Rick Maizels for helpful discussions, and Margo Chase-Topping for advice on statistical analysis.

Data Availability

All gene array data is available in Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/), accession number GSE114308. All other data are contained within the paper and its Supporting Information.

Funding Statement

This work was supported by the MRC UK (https://mrc.ukri.org) grant number MR/K020196/1, and the Wellcome Trust (https://wellcome.ac.uk/home) grant number 095831. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Allen JE, Maizels RM (2011) Diversity and dialogue in immunity to helminths. Nat Rev Immunol 11: 375–388. 10.1038/nri2992 [DOI] [PubMed] [Google Scholar]
  • 2.Hotez PJ, Brindley PJ, Bethony JM, King CH, Pearce EJ, et al. (2008) Helminth infections: the great neglected tropical diseases. J Clin Invest 118: 1311–1321. 10.1172/JCI34261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Maizels RM, Yazdanbakhsh M (2003) Immune regulation by helminth parasites: cellular and molecular mechanisms. Nat Rev Immunol 3: 733–744. 10.1038/nri1183 [DOI] [PubMed] [Google Scholar]
  • 4.Platts-Mills T, Vaughan J, Squillace S, Woodfolk J, Sporik R (2001) Sensitisation, asthma, and a modified Th2 response in children exposed to cat allergen: a population-based cross-sectional study. Lancet 357: 752–756. 10.1016/S0140-6736(00)04168-4 [DOI] [PubMed] [Google Scholar]
  • 5.Schwartz RH (2003) T cell anergy. Ann Rev Immunol 21: 305–334. [DOI] [PubMed] [Google Scholar]
  • 6.Wherry EJ, Kurachi M (2015) Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 15: 486–499. 10.1038/nri3862 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wherry EJ (2011) T cell exhaustion. Nat Immunol 12: 492–499. 10.1038/ni.2035 [DOI] [PubMed] [Google Scholar]
  • 8.Schietinger A, Greenberg PD (2014) Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol 35: 51–60. 10.1016/j.it.2013.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Schietinger A, Delrow JJ, Basom RS, Blattman JN, Greenberg PD (2012) Rescued tolerant CD8 T cells are preprogrammed to reestablish the tolerant state. Science 335: 723–727. 10.1126/science.1214277 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Reading JL, Galvez-Cancino F, Swanton C, Lladser A, Peggs KS, et al. (2018) The function and dysfunction of memory CD8(+) T cells in tumor immunity. Immunol Rev 283: 194–212. 10.1111/imr.12657 [DOI] [PubMed] [Google Scholar]
  • 11.Taylor MD, van der Werf N, Maizels RM (2012) T cells in helminth infection: the regulators and the regulated. Trends Immunol 33: 181–189. 10.1016/j.it.2012.01.001 [DOI] [PubMed] [Google Scholar]
  • 12.Maizels RM, McSorley HJ (2016) Regulation of the host immune system by helminth parasites. J Allergy Clin Immunol 138: 666–675. 10.1016/j.jaci.2016.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Borkow G, Leng Q, Weisman Z, Stein M, Galai N, et al. (2000) Chronic immune activation associated with intestinal helminth infections results in impaired signal transduction and anergy. J Clin Invest 106: 1053–1060. 10.1172/JCI10182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Leng Q, Bentwich Z, Borkow G (2006) Increased TGF-beta, Cbl-b and CTLA-4 levels and immunosuppression in association with chronic immune activation. Int Immunol 18: 637–644. 10.1093/intimm/dxh375 [DOI] [PubMed] [Google Scholar]
  • 15.Babu S, Blauvelt CP, Kumaraswami V, Nutman TB (2006) Regulatory networks induced by live parasites impair both Th1 and Th2 pathways in patent lymphatic filariasis: implications for parasite persistence. J Immunol 176: 3248–3256. 10.4049/jimmunol.176.5.3248 [DOI] [PubMed] [Google Scholar]
  • 16.Taylor JJ, Krawczyk CM, Mohrs M, Pearce EJ (2009) Th2 cell hyporesponsiveness during chronic murine schistosomiasis is cell intrinsic and linked to GRAIL expression. J Clin Invest 119: 1019–1028. 10.1172/JCI36534 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Aldridge A, O'Neill SM (2016) Fasciola hepatica tegumental antigens induce anergic-like T cells via dendritic cells in a mannose receptor-dependent manner. Eur J Immunol 46: 1180–1192. 10.1002/eji.201545905 [DOI] [PubMed] [Google Scholar]
  • 18.Sachdev D, Gough KC, Flynn RJ (2017) The Chronic Stages of Bovine Fasciola hepatica Are Dominated by CD4 T-Cell Exhaustion. Front Immunol 8: 1002 10.3389/fimmu.2017.01002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.van der Werf N, Redpath SA, Azuma M, Yagita H, Taylor MD (2013) Th2 cell-intrinsic hypo-responsiveness determines susceptibility to helminth infection. PLoS Pathog 9: e1003215 10.1371/journal.ppat.1003215 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Taylor MD, LeGoff L, Harris A, Malone E, Allen JE, et al. (2005) Removal of regulatory T cell activity reverses hyporesponsiveness and leads to filarial parasite clearance in vivo. J Immunol 174: 4924–4933. 10.4049/jimmunol.174.8.4924 [DOI] [PubMed] [Google Scholar]
  • 21.Campbell SM, Knipper JA, Ruckerl D, Finlay CM, Logan N, et al. (2018) Myeloid cell recruitment versus local proliferation differentiates susceptibility from resistance to filarial infection. Elife 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Petit G, Diagne M, Marechal P, Owen D, Taylor D, et al. (1992) Maturation of the filaria Litomosoides sigmodontis in BALB/c mice; comparative susceptibility of nine other inbred strains. Ann Parasitol Hum Comp 67: 144–150. 10.1051/parasite/1992675144 [DOI] [PubMed] [Google Scholar]
  • 23.Sutherland TE, Logan N, Ruckerl D, Humbles AA, Allan SM, et al. (2014) Chitinase-like proteins promote IL-17-mediated neutrophilia in a tradeoff between nematode killing and host damage. Nat Immunol 15: 1116–1125. 10.1038/ni.3023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kauffmann A, Huber W (2010) Microarray data quality control improves the detection of differentially expressed genes. Genomics 95: 138–142. 10.1016/j.ygeno.2010.01.003 [DOI] [PubMed] [Google Scholar]
  • 25.Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, et al. (2003) Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4: 249–264. 10.1093/biostatistics/4.2.249 [DOI] [PubMed] [Google Scholar]
  • 26.Crawford A, Angelosanto JM, Kao C, Doering TA, Odorizzi PM, et al. (2014) Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection. Immunity 40: 289–302. 10.1016/j.immuni.2014.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zheng Y, Zha Y, Spaapen RM, Mathew R, Barr K, et al. (2013) Egr2-dependent gene expression profiling and ChIP-Seq reveal novel biologic targets in T cell anergy. Mol Immunol 55: 283–291. 10.1016/j.molimm.2013.03.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Mohrs M, Shinkai K, Mohrs K, Locksley RM (2001) Analysis of type 2 immunity in vivo with a bicistronic IL-4 reporter. Immunity 15: 303–311. 10.1016/s1074-7613(01)00186-8 [DOI] [PubMed] [Google Scholar]
  • 29.Mohrs K, Wakil AE, Killeen N, Locksley RM, Mohrs M (2005) A two-step process for cytokine production revealed by IL-4 dual-reporter mice. Immunity 23: 419–429. 10.1016/j.immuni.2005.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.King IL, Mohrs M (2009) IL-4-producing CD4+ T cells in reactive lymph nodes during helminth infection are T follicular helper cells. J Exp Med 206: 1001–1007. 10.1084/jem.20090313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zaretsky AG, Taylor JJ, King IL, Marshall FA, Mohrs M, et al. (2009) T follicular helper cells differentiate from Th2 cells in response to helminth antigens. J Exp Med 206: 991–999. 10.1084/jem.20090303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Okoye IS, Czieso S, Ktistaki E, Roderick K, Coomes SM, et al. (2014) Transcriptomics identified a critical role for Th2 cell-intrinsic miR-155 in mediating allergy and antihelminth immunity. Proc Natl Acad Sci 111: E3081–3090. 10.1073/pnas.1406322111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Peine M, Rausch S, Helmstetter C, Frohlich A, Hegazy AN, et al. (2013) Stable T-bet(+)GATA-3(+) Th1/Th2 hybrid cells arise in vivo, can develop directly from naive precursors, and limit immunopathologic inflammation. PLoS Biol 11: e1001633 10.1371/journal.pbio.1001633 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, et al. (2006) Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439: 682–687. 10.1038/nature04444 [DOI] [PubMed] [Google Scholar]
  • 35.Zheng Y, Zha Y, Driessens G, Locke F, Gajewski TF (2012) Transcriptional regulator early growth response gene 2 (Egr2) is required for T cell anergy in vitro and in vivo. J Exp Med 209: 2157–2163. 10.1084/jem.20120342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Burton BR, Britton GJ, Fang H, Verhagen J, Smithers B, et al. (2014) Sequential transcriptional changes dictate safe and effective antigen-specific immunotherapy. Nat Commun 5: 4741 10.1038/ncomms5741 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ulges A, Klein M, Reuter S, Gerlitzki B, Hoffmann M, et al. (2015) Protein kinase CK2 enables regulatory T cells to suppress excessive TH2 responses in vivo. Nat Immunol 16: 267–275. 10.1038/ni.3083 [DOI] [PubMed] [Google Scholar]
  • 38.Hartmann W, Marsland BJ, Otto B, Urny J, Fleischer B, et al. (2011) A novel and divergent role of granzyme A and B in resistance to helminth infection. J Immunol 186: 2472–2481. 10.4049/jimmunol.0902157 [DOI] [PubMed] [Google Scholar]
  • 39.Pesu M, Watford WT, Wei L, Xu L, Fuss I, et al. (2008) T-cell-expressed proprotein convertase furin is essential for maintenance of peripheral immune tolerance. Nature 455: 246–250. 10.1038/nature07210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tangye SG (2015) Advances in IL-21 biology—enhancing our understanding of human disease. Curr Opin Immunol 34: 107–115. 10.1016/j.coi.2015.02.010 [DOI] [PubMed] [Google Scholar]
  • 41.Poholek AC, Jankovic D, Villarino AV, Petermann F, Hettinga A, et al. (2016) IL-10 induces a STAT3-dependent autoregulatory loop in TH2 cells that promotes Blimp-1 restriction of cell expansion via antagonism of STAT5 target genes. Sci Immunol 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pot C, Jin H, Awasthi A, Liu SM, Lai CY, et al. (2009) Cutting edge: IL-27 induces the transcription factor c-Maf, cytokine IL-21, and the costimulatory receptor ICOS that coordinately act together to promote differentiation of IL-10-producing Tr1 cells. J Immunol 183: 797–801. 10.4049/jimmunol.0901233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Tian Y, Zajac AJ (2016) IL-21 and T Cell Differentiation: Consider the Context. Trends Immunol 37: 557–568. 10.1016/j.it.2016.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hultqvist M, Backlund J, Bauer K, Gelderman KA, Holmdahl R (2007) Lack of reactive oxygen species breaks T cell tolerance to collagen type II and allows development of arthritis in mice. J Immunol 179: 1431–1437. 10.4049/jimmunol.179.3.1431 [DOI] [PubMed] [Google Scholar]
  • 45.Villarino AV, Katzman SD, Gallo E, Miller O, Jiang S, et al. (2011) Posttranscriptional silencing of effector cytokine mRNA underlies the anergic phenotype of self-reactive T cells. Immunity 34: 50–60. 10.1016/j.immuni.2010.12.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Sande OJ, Karim AF, Li Q, Ding X, Harding CV, et al. (2016) Mannose-Capped Lipoarabinomannan from Mycobacterium tuberculosis Induces CD4+ T Cell Anergy via GRAIL. J Immunol 196: 691–702. 10.4049/jimmunol.1500710 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Stempin CC, Rojas Marquez JD, Ana Y, Cerban FM (2017) GRAIL and Otubain-1 are Related to T Cell Hyporesponsiveness during Trypanosoma cruzi Infection. PLoS Negl Trop Dis 11: e0005307 10.1371/journal.pntd.0005307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Utzschneider DT, Legat A, Fuertes Marraco SA, Carrie L, Luescher I, et al. (2013) T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat Immunol 14: 603–610. 10.1038/ni.2606 [DOI] [PubMed] [Google Scholar]
  • 49.Yazdanbakhsh M, Sacks DL (2010) Why does immunity to parasites take so long to develop? Nat Rev Immunol 10: 80–81. 10.1038/nri2673 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang J, Vuitton DA, Muller N, Hemphill A, Spiliotis M, et al. (2015) Deletion of Fibrinogen-like Protein 2 (FGL-2), a Novel CD4+ CD25+ Treg Effector Molecule, Leads to Improved Control of Echinococcus multilocularis Infection in Mice. PLoS Negl Trop Dis 9: e0003755 10.1371/journal.pntd.0003755 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wang L, Yang C, Xu M, Hu M, Wang X, et al. (2014) The role of soluble fibrinogen-like protein 2 in transplantation: protection or damage. Transplantation 97: 1201–1206. 10.1097/TP.0000000000000116 [DOI] [PubMed] [Google Scholar]
  • 52.Altin JA, Goodnow CC, Cook MC (2012) IL-10+ CTLA-4+ Th2 inhibitory cells form in a Foxp3-independent, IL-2-dependent manner from Th2 effectors during chronic inflammation. J Immunol 188: 5478–5488. 10.4049/jimmunol.1102994 [DOI] [PubMed] [Google Scholar]
  • 53.Okamura T, Fujio K, Sumitomo S, Yamamoto K (2012) Roles of LAG3 and EGR2 in regulatory T cells. Ann Rheum Dis 71 Suppl 2: i96–100. 10.1136/annrheumdis-2011-200588 [DOI] [PubMed] [Google Scholar]
  • 54.Sartono E, Kruize YC, Kurniawan A, Maizels RM, Yazdanbakhsh M (1997) Depression of antigen-specific interleukin-5 and interferon-gamma responses in human lymphatic filariasis as a function of clinical status and age. J Infect Dis 175: 1276–1280. 10.1086/593701 [DOI] [PubMed] [Google Scholar]
  • 55.Saeftel M, Arndt M, Specht S, Volkmann L, Hoerauf A (2003) Synergism of gamma interferon and interleukin-5 in the control of murine filariasis. Infect Immun 71: 6978–6985. 10.1128/IAI.71.12.6978-6985.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0007908.r001

Decision Letter 0

Timothy G Geary, Benjamin L Makepeace

Transfer Alert

This paper was transferred from another journal. As a result, its full editorial history (including decision letters, peer reviews and author responses) may not be present.

16 Sep 2019

Dear Dr. Taylor:

Thank you very much for submitting your manuscript "Th2 cell-intrinsic dysfunction during chronic helminth infection is distinct from exhaustion and is maintained in the absence of antigen" (PNTD-D-19-01151) for review by PLOS Neglected Tropical Diseases. Your manuscript was fully evaluated at the editorial level and by independent peer reviewers. The reviewers appreciated the attention to an important topic but identified some aspects of the manuscript that should be improved.

We therefore ask you to modify the manuscript according to the review recommendations before we can consider your manuscript for acceptance. Your revisions should address the specific points made by each reviewer.

In addition, when you are ready to resubmit, please be prepared to provide the following:

(1) A letter containing a detailed list of your responses to the review comments and a description of the changes you have made in the manuscript.

(2) Two versions of the manuscript: one with either highlights or tracked changes denoting where the text has been changed (uploaded as a "Revised Article with Changes Highlighted" file ); the other a clean version (uploaded as the article file).

(3) If available, a striking still image (a new image if one is available or an existing one from within your manuscript). If your manuscript is accepted for publication, this image may be featured on our website. Images should ideally be high resolution, eye-catching, single panel images; where one is available, please use 'add file' at the time of resubmission and select 'striking image' as the file type.

Please provide a short caption, including credits, uploaded as a separate "Other" file. If your image is from someone other than yourself, please ensure that the artist has read and agreed to the terms and conditions of the Creative Commons Attribution License at http://journals.plos.org/plosntds/s/content-license (NOTE: we cannot publish copyrighted images).

(4) Appropriate Figure Files

Please remove all name and figure # text from your figure files upon submitting your revision. Please also take this time to check that your figures are of high resolution, which will improve both the editorial review process and help expedite your manuscript's publication should it be accepted. Please note that figures must have been originally created at 300dpi or higher. Do not manually increase the resolution of your files. For instructions on how to properly obtain high quality images, please review our Figure Guidelines, with examples at: http://journals.plos.org/plosntds/s/figures

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

We hope to receive your revised manuscript by Nov 15 2019 11:59PM. If you anticipate any delay in its return, we ask that you let us know the expected resubmission date by replying to this email.

To submit your revised files, please log in to https://www.editorialmanager.com/pntd/

If you have any questions or concerns while you make these revisions, please let us know.

Sincerely,

Benjamin L Makepeace

Associate Editor

PLOS Neglected Tropical Diseases

Timothy Geary

Deputy Editor

PLOS Neglected Tropical Diseases

***********************

Reviewer's Responses to Questions

Key Review Criteria Required for Acceptance?

As you describe the new analyses required for acceptance, please consider the following:

Methods

-Are the objectives of the study clearly articulated with a clear testable hypothesis stated?

-Is the study design appropriate to address the stated objectives?

-Is the population clearly described and appropriate for the hypothesis being tested?

-Is the sample size sufficient to ensure adequate power to address the hypothesis being tested?

-Were correct statistical analysis used to support conclusions?

-Are there concerns about ethical or regulatory requirements being met?

Reviewer #1: This is a very well written manuscript that clearly states the hypothesis and the objectives of the study. The study design is well chosen and uses several state-of-the-art methods to address the objectives. The number of replicates used for the microarray analysis is sufficient and all immunological experiments were repeated with a sufficient number of animals. There are no concerns regarding the statisctial analysis used, ethics or regulatory requirements.

Reviewer #2: Objectives are clear and design is appropriate to address the stated objectives.

Statistical analyses were correct. Study was IACUC approved.

Reviewer #3: The objectives of the study are well articulated. There is no clear testable hypothesis stated but this does not seem appropriate for the type of work reported. The overall goal of understanding the characteristics of hypo-responsive T cells during chronic worm infection is clear. In terms of sample size, the numbers of animals used are appropriate and statistical analyses are correct to support the conclusion. There is no concern about ethical or regulatory requirements being met.

--------------------

Results

-Does the analysis presented match the analysis plan?

-Are the results clearly and completely presented?

-Are the figures (Tables, Images) of sufficient quality for clarity?

Reviewer #1: Overall, the results are clearly and completely presented and the figures and tables are of good quality.

However, there are a few minor points that have to be addressed:

- Line 557: Reference of Fig. 7A is misplaced here.

- Legends for Fig. 6 and 7 have to be corrected. Descriptions of some of the data shown in those figures is missing/replaced in the legends.

- Heatmaps shown in Fig. 5D and S6: Tables that summarize the gene expression changes indicated in the heatmaps of Fig. 5D and S6 should be provided as suppl. data.

- Fig. 1D/Line 311-314: The bar indicating the statistically significant difference in Fig. 1D seems misplaced and not in accordance to the description given in the results section.

- Line 322: This should state 2 weeks, as the infection took place 2 weeks after the cell transfer.

- Please expand the legend of Fig. S6 to explain the experimental design used for the peptide induced tolerance.

Reviewer #2: Analysis matches the analysis plan.

Results are clearly and completely presented (except for critiques provided below).

The figures are of sufficient quality.

Reviewer #3: The results are extremely well presented and the results section has a strong narrative that makes it easy to understand the logic to the experiments. All figures are of sufficient quality for clarity.

--------------------

Conclusions

-Are the conclusions supported by the data presented?

-Are the limitations of analysis clearly described?

-Do the authors discuss how these data can be helpful to advance our understanding of the topic under study?

-Is public health relevance addressed?

Reviewer #1: With exception of the impact on the humoral immune responses the conclusions are supported by the presented data and the limitations of the study are presented in the discussion.

In detail, I have following questions and concerns regarding the interpretation of tha data:

- Fig. 7C: What is your average frequency of Mf positive animals during L.s. infection? Do you always observe a microfilaremia in almost all infected animals?

- Fig. 7D, E: Can the authors exclude that a lack of changes in the number of CD4+GATA3+ and IL-4 and IL-5 producing CD4+GATA3+ T cells by α-IL-21R blockage is not missed due to a belated blockade starting at 45dpi? Does 45dpi present a time point of acute or hyporesponsive infection & T cell function?

- Fig. 7G: Increase in L.s.-specific IgG1 in the α-IL-21R treated L.s. infected animals is very heterogeneous, which may be due to the pooling of 2 experiments. Was the increase in specific IgG1 in the depleted group consistent in both depletion experiments? Given the presented data, the statement that IL-21 inhibits L.s.-specific B cell responses/humoral responses is not well supported and should be confirmed or restated throughout the manuscript (e.g. l. 594, l. 686-688, abstract).

- Are there any indications that epigenetic changes may occur? The possible contribution of epigenetic changes should be discussed.

- Other potential sources of IL-21 and their possible contribution should be mentioned in the discussion

Reviewer #2: Yes, the conclusions are supported by the data. The authors do not overstate their findings. Limitations are described. Potential relevance is discussed.

Reviewer #3: The conclusions are supported by the data presented. There are definite limitations to the data presented, in particular the inability of the group to co-stain IL-4gfp with transcription factors that make it difficult to definitively define the phenotype of the hyporesponsive cells. However, these limitations are well discussed. The authors clearly discuss how the data may advance our understanding of T cell hypo-responsiveness and in particular discuss how this may be relevant to the publich health concerns of filarial nematode infection.

--------------------

Editorial and Data Presentation Modifications?

Use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity. If the only modifications needed are minor and/or editorial, you may wish to recommend “Minor Revision” or “Accept”.

Reviewer #1: (No Response)

Reviewer #2: Minor revision. No additional experiments needed. See notes below.

Reviewer #3: (No Response)

--------------------

Summary and General Comments

Use this section to provide overall comments, discuss strengths/weaknesses of the study, novelty, significance, general execution and scholarship. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. If requesting major revision, please articulate the new experiments that are needed.

Reviewer #1: This is a very well presented study that investigated the fate of Th2 cells during acute and chronic helminth infection. Interestingly, Th2 cells developed during chronic filarial infection a transcriptional profile that partially overlaps with anergic and tolerized T cells and differs from Th2 cells isolated during acute filarial or intestinal helminth infection. Furthermore, the hypo-responsive T cell phenotype was stable after transfer into naïve mice. While losing their ability to produce IL-4 and IL-5 proteins, the hypo-responsive T cells produced IL-21, which is suggested to support the development of microfilaremia.

The authors used to different models of helminth infections, microarray analysis and a thorough bioinformatic analysis of their gene expression data, which was in part confirmed by flow cytometry. Thus, the study represents a very robust work that further enhances our understanding of the immunomodulation of helminths and especially the development of hypo-responsive T cells.

Reviewer #2: SUMMARY/OVERVIEW

In this paper the investigators study changes in the transcriptome of dysfunctional Th2 cells that arise during chronic mouse infection with the filarial nematode Litomosoides sigmodontis.

Key findings include: showing that dysfunctional Th2 cells:

1) remain dysfunctional stably when transferred into a non-infected mouse

2) have a post-transciptional block for production of Th2 cytokines

3) have a divergent transcriptional profile when compared to classical Th2 cells that are isolated before hyporesponsiveness occurs in Litomosoides sigmodontis or after acute Nippostrongylus infection

4) upregulate Blimp-1 expression but have a transcriptional profile that is dissimilar from exhausted CD4 T cells

5) have a transcriptional profile that has some overlap with anergic/tolerized CD4 T cells, including upregulation of Egr2 and c-Maf

6) gain the ability to produce IL-21, which seems to be involved in a novel regulatory role of B cell antibody production during filarial infection.

The authors also found that there are many parasite-specfic differences in CD4 T cell transcriptional changes.

The results provide insights into how Th2 cells become dysfunctional in the context of chronic filarial infection. A strength is the careful comparison to transcriptional profiles associated with T cell exhaustion and T cell anergy, which clearly shows that what is occurring in Th2 cells during filarial infection is somewhat novel from either of these mechanisms of T cell dysfunction. Overall the methods and statistical analyses appear robust, and I suspect the article would be of interest to many researchers that study immune regulation during chronic helminth infection.

MAJOR CRITIQUES

1. Figure 7h I is supposed to be the total # of CD19+PNA+ germinal center B cells, but the figure is an exact copy of 7J, which is the # of CD19 Igm-IgD- class-switched B cells. Please correct.

MINOR CRITIQUES

1. In the results section, the authors state that d 60 CD4+IL-4gfp+ Th2 cells had “significantly impaired ability to produce IL-5 protein compared to d 20 CD4+IL-4gfp+ Th2 cells” in unchallenged mice. However, when looking at the datapoints in the figure, it doesn’t really appear as though there would be a statistically significant difference between those two groups. Additionally, the authors put a large bar with two asterisks above it across the entire figure. The authors in the results, however, appear to be comparing d20 to d60 in unchallenged and then in challenged mice….suggesting that perhaps they intended to include two bars in the figure (one over the first two groups, one over the final two groups).

2. In discussions of figure 3 in the text the authors switch from Tbx21 (which is in the figures) to using Tbet in the written results. This is ok, but a bit confusing to the reader…perhaps clarify with a parenthetical that Tbet is the same as Tbx21.

3. At times it is difficult to follow the logic for which genes are being assessed as a group. This was especially true for the results section in Figure 6. The authors may want to break up the results section covering Figure 6 into more paragraphs and provide a sentence or two explaining the groupings prior to discussing each individual figure in Figure 6.

4. The results section “Th2 cell-intrinsic hypo-responsiveness has similarities with T cell anergy and tolerance” has a concluding sentence “However, the overall transcriptional profile Th2 cell-intrinsic hyporesponsiveness was dissimilar to T cell anergy and tolerance” that contradicts the title. Would suggest simply changing the concluding sentence to something along the lines of “there is some overlap.”

5. Figure 7F y axis delete (x10exp5) from the axis title as it is percentage of B cells, not total numbers, being expressed.

Reviewer #3: In this manuscript by Knipper et al., the authors use an experimental filarial nematode infection (Litomosoides sigmodontis) to investigate the characteristics of hyporesponsive Th2 cells in this setting. Although, the authors do not directly study human filarial infection these findings may have relevance to human filarial disease as a similar hypo-responsive phenotype of T cells has also been described. Importantly, the data suggests that in chronic filarial infection Th2 cells switch from an active IL-4+IL-5+ Th2 phenotype to a modified and dysfunctional phenotype that is distinct from that described in other settings of T cell hyporesponsiveness (e.g. T cell exhaustion). These modified T cells are identified by their IL-21+Egr2+c-Maf+Blimp-1+IL-4loIL-5loTbet+IFN-gamma+ state. These may provide new therapeutic targets in filarial infection.

Overall, this is a novel and interesting study that provides new insight into T cell hyporesponsiveness during filarial nematode infection. A particular strength of the study is that there is excellent integration of microarray data with ex vivo flow cytometry data to define the global characteristics of hypo-responsive Th2 cells. Moreover, based on these findings the authors identify IL-21 as a cytokine involved in augmenting blood microfilarial levels at the chronic stage of L. sigmodontis infection.

One limitation to the study is the incapacity of the group to co-stain IL-4-gfp with transcription factors making it difficult to definitively demonstrate that the IL-4-gfp +ve cells (i.e. Th2 cells) are the ones that also co-express T-bet. However, this limitation is well discussed in the manuscript.

--------------------

PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: Yes: Edward Mitre, M.D. and Emilie Goguet, Ph.D.

Reviewer #3: Yes: Dr. John R. Grainger

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0007908.r002

Decision Letter 1

Timothy G Geary, Benjamin L Makepeace

6 Nov 2019

Dear Dr. Taylor,

We are pleased to inform you that your manuscript, "Th2 cell-intrinsic dysfunction during chronic helminth infection is distinct from exhaustion and is maintained in the absence of antigen", has been editorially accepted for publication at PLOS Neglected Tropical Diseases.

Before your manuscript can be formally accepted and sent to production you will need to complete our formatting changes, which you will receive in a follow up email. Please note: your manuscript will not be scheduled for publication until you have made the required changes.

IMPORTANT NOTES

* Copyediting and Author Proofs: To ensure prompt publication, your manuscript will NOT be subject to detailed copyediting and you will NOT receive a typeset proof for review. The corresponding author will have one final opportunity to correct any errors when sent the requests mentioned above. Please review this version of your manuscript for any errors.

* If you or your institution will be preparing press materials for this manuscript, please inform our press team in advance at plosntds@plos.org. If you need to know your paper's publication date for media purposes, you must coordinate with our press team, and your manuscript will remain under a strict press embargo until the publication date and time. PLOS NTDs may choose to issue a press release for your article. If there is anything that the journal should know, please get in touch.

*Now that your manuscript has been provisionally accepted, please log into EM and update your profile. Go to http://www.editorialmanager.com/pntd, log in, and click on the "Update My Information" link at the top of the page. Please update your user information to ensure an efficient production and billing process.

*Note to LaTeX users only - Our staff will ask you to upload a TEX file in addition to the PDF before the paper can be sent to typesetting, so please carefully review our Latex Guidelines [http://www.plosntds.org/static/latexGuidelines.action] in the meantime.

Thank you again for supporting open-access publishing; we are looking forward to publishing your work in PLOS Neglected Tropical Diseases.

Best regards,

Benjamin L Makepeace

Associate Editor

PLOS Neglected Tropical Diseases

Timothy Geary

Deputy Editor

PLOS Neglected Tropical Diseases

***********************************************************

PLoS Negl Trop Dis. doi: 10.1371/journal.pntd.0007908.r003

Acceptance letter

Timothy G Geary, Benjamin L Makepeace

4 Dec 2019

Dear Dr. Taylor,

We are delighted to inform you that your manuscript, "Helminth-induced Th2 cell dysfunction is distinct from exhaustion and is maintained in the absence of antigen," has been formally accepted for publication in PLOS Neglected Tropical Diseases.

We have now passed your article onto the PLOS Production Department who will complete the rest of the publication process. All authors will receive a confirmation email upon publication.

The corresponding author will soon be receiving a typeset proof for review, to ensure errors have not been introduced during production. Please review the PDF proof of your manuscript carefully, as this is the last chance to correct any scientific or type-setting errors. Please note that major changes, or those which affect the scientific understanding of the work, will likely cause delays to the publication date of your manuscript. Note: Proofs for Front Matter articles (Editorial, Viewpoint, Symposium, Review, etc...) are generated on a different schedule and may not be made available as quickly.

Soon after your final files are uploaded, the early version of your manuscript will be published online unless you opted out of this process. The date of the early version will be your article's publication date. The final article will be published to the same URL, and all versions of the paper will be accessible to readers.

Thank you again for supporting open-access publishing; we are looking forward to publishing your work in PLOS Neglected Tropical Diseases.

Best regards,

Serap Aksoy

Editor-in-Chief

PLOS Neglected Tropical Diseases

Shaden Kamhawi

Editor-in-Chief

PLOS Neglected Tropical Diseases

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Representative flow plots showing sorting strategy and purities for the tLN and spleen.

    (PDF)

    S2 Fig. Transcription factors that are differentially expressed between d 20 and d 60 PleC IL-4gfp+ Th2 cells during L. sigmodontis infection.

    (PDF)

    S3 Fig. Transcription factors that are expressed in common by d20 and d60 PleC IL-4gfp+ Th2 cells during L. sigmodontis infection.

    (PDF)

    S4 Fig. IFN-γ is an upstream regulator of PleC d 60 IL-4gfp+ Th2 cells.

    (PDF)

    S5 Fig. Comparison of gene expression in peptide-induced tolerance and Th2 cell-intrinsic hypo-responsiveness.

    (PDF)

    S6 Fig. TGF-β is an upstream regulator of core candidate genes.

    (PDF)

    S1 Table. Table showing KEGG pathways enriched in PleC and/or tLN IL-4gfp+ Th2 cells at d 60 pi, but not at d 20 pi.

    (DOCX)

    S2 Table. Table showing fold-change in expression of genes significantly upregulated in anergic T cells that was used to perform the unbiased clustering represent by the heat map in Fig 5D.

    (XLSX)

    S3 Table. Table showing fold-change in expression of genes significantly upregulated in T cells following peptide-elicited tolerance that was used to perform the unbiased clustering represented in the heat map in S5 Fig.

    (XLSX)

    S4 Table. Table of loci that significantly differ between d 20 and 60 of L. sigmodontis infection in PleC IL-4gfp+ Th2 cells.

    (XLSX)

    S5 Table. Table of loci that significantly differ between d 20 and 60 of L. sigmodontis infection in tLN IL-4gfp+ Th2 cells.

    (XLSX)

    S6 Table. Table of core candidate loci associated with Th2 cell-intrinsic hypo-responsiveness.

    (XLSX)

    S7 Table. Table showing subset of the core candidate loci that have significantly different expression in tLN IL-4gfp+ Th2 cells between d20 and d60.

    (XLSX)

    Data Availability Statement

    All gene array data is available in Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/), accession number GSE114308. All other data are contained within the paper and its Supporting Information.


    Articles from PLoS Neglected Tropical Diseases are provided here courtesy of PLOS

    RESOURCES