Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 15.
Published in final edited form as: Nat Immunol. 2019 Jul 15;20(8):1059–1070. doi: 10.1038/s41590-019-0418-x

Altered differentiation is central to HIV-specific CD4+ T cell dysfunction in progressive disease

Antigoni Morou 1,2, Elsa Brunet-Ratnasingham 1,2, Mathieu Dubé 1,3, Roxanne Charlebois 1, Eloi Mercier 4, Sam Darko 5, Nathalie Brassard 1, Krystelle Nganou-Makamdop 5, Sahaana Arumugam 5, Gabrielle Gendron-Lepage 1,2, Lifei Yang 6, Julia Niessl 1,2,3, Amy E Baxter 1,2,3,#, James M Billingsley 7, Premeela A Rajakumar 7, François Lefebvre 4, R Paul Johnson 7, Cécile Tremblay 1,2, Jean-Pierre Routy 8, Richard T Wyatt 3,6, Andrés Finzi 1,2, Daniel C Douek 5, Daniel E Kaufmann 1,2,3,*
PMCID: PMC6642691  NIHMSID: NIHMS1528611  PMID: 31308541

Abstract

Dysfunction of virus-specific CD4+ T cells in chronic human infections is poorly understood. We performed genome-wide transcriptional analyses and functional assays of CD4+ T cells specific for human immunodeficiency virus (HIV) from HIV-infected people prior and after initiation of antiretroviral therapy (ART). A follicular helper T cell (TFH cell)–like profile characterized HIV-specific CD4+ T cells in viraemic infection. HIV-specific CD4+ T cells from people spontaneously controlling the virus (elite controllers) robustly expressed genes associated with the TH1, TH17 and TH22 subsets of helper T cells. Viral suppression by ART resulted in a distinct transcriptional landscape, with a reduction in the expression of genes associated with TFH cells but persistently low expression of genes associated with TH1, TH17 and TH22 cells compared to the elite controller profile. Thus, altered differentiation is central to the impairment of HIV-specific CD4+ T cells and involves both gain of function and loss of function.


CD4+ T cell responses are critical for durable control of viral replication in chronic infections1, 2. Virus-specific CD4+ T cell immunity is of particular interest for HIV infection, which is characterized by functional impairment and destruction of CD4+ T cells. While classical models divided CD4+ T cells into distinct lineages, studies have demonstrated the importance of CD4+ T cell plasticity3. Persistent antigen and inflammatory signals cause impairment of antigen-specific responses, a state called immune exhaustion. Virus-specific CD8+ T cell exhaustion has been extensively investigated4 and represents a bona fide cell differentiation program. These studies highlighted the relevance of genome-wide transcriptional studies to understand T cell impairment5. Compared to CD8+ T cells, less is known on CD4+ T cell dysfunction. Murine LCMV-specific CD4+ T cells in chronic infection, while exhibiting some characteristics shared with CD8+ T cells, also present distinct features6, 7, including loss of a TH1-signature5 and skewing towards a T follicular helper (TFH) phenotype8, 9.

Whether findings in mice can be extrapolated to human HIV infection is unclear. Some features of virus-specific CD4+ T cells are shared between both infections: upregulation of co-inhibitory receptors are found in HIV progressors with ongoing viremia (chronic progressors, CP) and chronic LCMV infection5, 10. Rare subjects who spontaneously suppress HIV (elite controllers, EC) frequently exhibit robust virus-specific TH1 responses11 and strong proliferative capacity, similarly to mice infected with the acute strain of LCMV. However, HIV and LCMV are distinct viruses and there are notable differences between species in terms of T cell differentiation mechanisms, such as TFH generation12. An issue of critical clinical relevance is the lack of restoration of effective anti-HIV immunity after suppressive antiretroviral therapy (ART): viral rebound is the rule after cessation of therapy. Whether persistent HIV-specific CD4+ T cell dysfunction on ART contribute to this failed response is an important, yet unresolved, question. The paucity of experimental tools capable of identifying highly heterogeneous antigen-specific CD4+ T cells has hampered the study of HIV-specific CD4+ T cell help. Intracellular cytokine assays (ICS) are of limited sensitivity for many non-TH1 effector functions, and the use of HLA Class II tetramers in humans is constrained by availability, requirement for pre-defined epitopes and genetic diversity.

To determine key pathways and molecules that link HIV-specific T cell help to viral control, we here performed genome-wide transcriptional analyses and functional assays of HIV-specific CD4+ T cells from HIV-infected humans with diverse viral loads prior to ART initiation and followed a subgroup of them longitudinally after viral suppression on therapy.

RESULTS

Links between HIV-specific CD4 transcriptome profiles and viremia

To define molecular features that discriminate HIV-specific CD4+ T cells in progressive vs. controlled infection, we performed a cross-sectional study of 38 chronically infected people who were untreated at the time of sampling. These included “elite controllers” (EC, HIV plasma viral load <50 vRNA copies/ml), “viremic controllers” (VC, viral load between 50 and 5,000 copies/ml) and “chronic progressors” (CP, viral load ≥5,000 copies/ml) (Participant characteristics: Supplementary Table 1).

We utilized an activation induced marker (AIM) assay to identify CD4+ T cells specific for the Gag protein (hereafter termed HIV-specific CD4+ T cells). Ex vivo stimulated HIV-specific CD4+ T cells were identified by the co-upregulation of CD40L and CD69 on their surface after a 9-h stimulation with an HIV Gag peptide pool13 (Fig. 1a, Supplementary Fig. 1a). Combining two markers enhanced detection of HIV-specific CD4+ T cells by reducing background compared to CD40L alone (Supplementary Fig. 1b,c). This AIM assay overcomes limitations of cytokine-based techniques for detection of virus-specific cells, allows live-cell sorting and captures a broader Ag-specific CD4+ T cell population (Supplementary Fig. 1d). There was no significant difference in the magnitude of HIV-specific CD4+ T cell responses amongst cohorts or correlation with viremia or CD4 count (Fig. 1b, Supplementary Fig. 1e,f).

Figure 1. Deep coverage transcriptome analysis of HIV-specific CD4+ T cells from untreated HIV-infected people with distinct disease status.

Figure 1.

(a) Representative flow cytometry plots from an EC of detection of HIV-specific CD4+ T cells by an activation-induced marker (AIM) assay based on co-upregulation of CD40L and CD69 9h after stimulation with an HIV Gag peptide pool. (b) Quantification of (a). Comparison of net frequency (background of No Antigen condition subtracted) of CD69+CD40L+ Gag-specific CD4+ T cells amongst the CP, VC and EC groups by Kruskal-Wallis test. Horizontal lines and bars represent median and +/−interquartile range, respectively (n= 11CPs, 11 VCs, 13 ECs). (c) Paired volcano plot comparisons depicting differentially expressed genes (DEG) as measured by microarray analysis of sorted HIV-specific CD4+ T cells in the three groups. Red, pink and blue dots represent DEG with log2|FC| >1 and padj<0.05, log2|FC|>1 or padj<0.05, respectively. A two-sided moderated t-test followed by Benjamini-Hochberg method correction was applied. (n= 11 CPs, 9 VCs, 12 ECs). (d) Euler diagrams of DEG between each pairwise comparison of the three groups of participants (log2FC> 1, p<0.05); numbers represent DEGs. (e) Top significant enriched Gene Ontology terms from the curated C5 MSigDB via two-sided CAMERA analysis. Blue, Red and white color denote negative, positive or not significant (as defined by FDR<0.05 by Benjamini-Hochberg method), respectively.

We reasoned that the AIM assay could reveal the transcriptome modulation in response to cognate antigen-mediated TCR signaling, reflecting the functional state of CD4+ T cells. We thus performed microarray analysis of sorted ex vivo stimulated HIV-specific CD4+ T cells. Pairwise comparisons between cohorts (Fig. 1c) revealed sets of differentially expressed genes (DEGs) between all three cohorts, with extensive dissimilarities between CPs and ECs. Fewer transcriptional differences were detected between the VC and EC groups. The majority of DEGs between VCs and ECs were also observed in the CPs versus ECs comparison (Fig. 1d). CPs and VCs showed the fewest DEGs, suggesting that viremia strongly impacts the transcriptional landscape of HIV-specific CD4+ T cells.

We next used CAMERA (Correlation Adjusted Mean Rank gene set test)14 enrichment analysis to investigate molecular mechanisms underlying HIV-specific T help alterations associated with disease status. We compared the identified DEGs to the Molecular Signatures Database (MSigDB) C5 collection of curated gene sets, which comprises the Gene Ontology (GO) terms associated with biological processes, molecular functions and cellular components (Fig. 1e). The “Positive regulation of T helper cell differentiation” signature was negatively associated with viremia. Consistently, the “Positive regulation of CD4+ αβ T cell activation” and “Regulation of leukocyte migration” gene sets were significantly underrepresented in CP compared to both VCs and ECs, contrasting with upregulation of the MHC class II protein complex in CPs. Expression of genes associated with protein transport and/or processing was reduced in CPs. The pathways enriched with viremia included processes associated with chromatin modification, mitotic cell cycle, DNA packaging, chromosome segregation, response to stress, and apoptosis.

This approach identifies potential pathways associated with HIV-specific CD4+ T cell dysfunction, and points at a central role for altered T cell differentiation, regulation and activation in this impairment.

HIV-specific CD4 polarization is associated with disease status

Given the potential role for altered T cell differentiation in HIV-specific CD4+ T cell impairment, we determined features of CD4+ T cell differentiation associated with disease status. We compared by CAMERA our sets of DEGs with human T helper polarization signatures from the C7 collection MSigDB and publicly available microarray datasets. While the different conditions used in the generation of these signatures (e.g activated vs unactivated cells) may complicate the interpretation of such associations, we observed notable differences amongst cohorts. Compared to CPs, ECs and VCs displayed enriched TH1 and TH17 signatures15, 16, 17 (Fig. 2a,b,e,f; Supplementary Fig. 2a; Supplementary Tables 24). Cardinal genes associated with TH17 cell differentiation and function were upregulated in ECs (Fig. 2f). In viremic people (CPs, VCs), we noted a striking enrichment of a TFH signature18 (Fig. 2c,g, Supplementary Fig. 2a) compared to ECs. There was congruent upregulation of several key TFH-associated genes compared to EC with the notable exception of Bcl6 that was instead upregulated in ECs (Fig. 2g, Supplementary Table 5). Although we did not observe a statistically significant enrichment of an LCMV-specific CD4+ T cell exhaustion signature5 in CPs compared to ECs (Fig. 2d) (which might be due at least in part to the methodological differences), several of the DEGs enriched in CP were well-known exhaustion-associated genes (Fig. 2h, Supplementary Table 6). Several Interferon-stimulated genes (ISGs) upregulated in the LCMV model of exhaustion, such as IFH1, IFIT3 and IFI44, were upregulated in ECs.

Figure 2. HIV-specific CD4+ T cell polarization is associated with level of viral control.

Figure 2.

(a-d) Barcode plots of enriched Thelper polarization and exhaustion gene sets by CAMERA in CP (n=11) vs EC (n=12) people. Red and blue lines denote positive and negative enrichment of CP genes vs. EC, respectively. (A) TH1 signature: GSE59295 (TH1 vs TH2), (B) TH17 signature: GSE49703 (TH17 vs TH1), (C) TFH signature: GSE50391 (CXCR5high CD45RO vs CXCR5-tonsil samples), (d) LCMV exhaustion signature GSE41866 (LCMV Clone 13 D30 vs Armstrong D30-Exhausted vs Memory). Two sided p-value by CAMERA and FDR by Benjamini-Hochberg method. The arrows beneath the barcode plots depict the orientation of the enrichment between CP and EC. (e-h) Heatmaps of mRNA expression of the top 50 significant genes of each CD4+ T cell signature in CPs and ECs. Red and blue font of gene names denote upregulated and downregulated mRNA expression in CPs vs ECs, respectively. (i) PCA representation of CPs (n=11), VCs (n=9) and ECs (n=12) based on transcriptional expression of 90 genes associated with CD4+ T cell differentiation, effector functions and exhaustion. Transcriptional expression (-ΔCt values) was evaluated by qRT-PCR (Fluidigm). The top 35 contributing genes are displayed and color-coded according to their contribution. Bigger dots represent the mean PC coordinates for each group. The numbers in parenthesis stand for the percentage of variance that the PC accounts for. (j) Correlation of PC1 values for each individual with viral load by two-tailed Spearman correlation (n= 11CPs, 9 VCs, 12 ECs).

We next used qRT-PCR to further quantify levels of key DEGs identified by microarrays and selected additional CD4+ T cell genes. Principal component analysis discriminated CPs from ECs, whereas there was more overlap between CPs and VCs (Fig 2i). PC1 values significantly inversely correlated with viremia, showing that changes in the CD4+ T cell profile relate to antigen load (Fig 2j). The qRT-PCR data were consistent with the microarray results (Supplementary Table 7). They confirmed that genes associated with TFH functions and B cell help (CXCL13, SLAMF6, CD84) and exhaustion (TIGIT, CD200, LAG3, TOX) were enriched in CPs. This contrasted with genes related to TH17 and TH22 functions (IL17F, IL22) and the interferon-induced chemokines (CXCL9, CXCL10, CXCL11), which were enriched in ECs (Fig 2i). Expression of selected transcription factors was consistent with these observations (Supplementary Fig 2b): ECs expressed significantly more TBX21 and AHR, genes respectively coding for TH1 and TH22 master transcription factors, and there was in ECs a trend for enhanced expression of RORC, a master transcription factor of TH17 differentiation. The behavior of the master TFH transcription factor BCL6 contrasted to most TFH associated genes: BCL6 was significantly upregulated in ECs compared to CPs in contrast to MAF, another transcription factor critical for TFH differentiation, which was highly expressed in CPs. Helios (IKZF2), a transcription factor overexpressed in LCMV-specific CD4+ T cells in chronic infection, was also upregulated in CPs compared to ECs. Examining correlations between gene expression revealed positive and negative associations of genes, many not anticipated by classical dichotomic models of TH differentiation. These results are consistent with a complex spectrum of differentiation and functional profiles of HIV-specific CD4+ T cells (Supplementary Fig 2c,d).

These data show higher expression of a TFH-like signature in HIV-specific CD4+ T cells in progressive infection, contrasting with enriched TH1 and TH17 signatures in subjects achieving spontaneous viral control.

Upregulation of classical TFH-associated genes in CXCR5neg HIV-specific CD4 of CPs

We next investigated whether the enrichment of the TFH signature in HIV-specific CD4+ T cells of CPs compared to ECs was due to a higher fraction of HIV-specific circulating TFH cells (cTFH). We used the CD69/CD40L AIM assay to assess the percentages of HIV-specific CXCR5+CD45RA memory CD4+ T cells (CXCR5mem) in both cohorts (Fig. 3a and Supplementary Fig. 3a,b). As PD-1 expression on HIV-specific CD4+ T cells correlates with viremia19, PD-1 was omitted as a TFH marker to avoid bias towards CPs. We observed only a trend for higher fractions of HIV-specific CXCR5mem in CPs than ECs, with considerable overlap between groups (Fig. 3b). In contrast, single subject GSEA (ssGSEA) scores showed a significant enrichment of the TFH signature in CPs compared to ECs (Fig. 3c), consistent with the cohort-based CAMERA analyses of Fig. 2c,g. The TFH signature enrichment by ssGSEA correlated directly with viremia (Fig. 3d). These data suggest that differences in percentage of CXCR5mem alone are not sufficient to explain the markedly enriched TFH signature observed in HIV-specific CD4+ T cells of CPs.

Figure 3. An atypical TFH-like transcriptional signature in CXCR5neg HIV-specific CD4+ T cells discriminates chronic progressors from elite controllers.

Figure 3.

(a) Representative flow cytometry plots from a CP illustrating the enrichment of cTFH (CXCR5+ CD45RA-) in CD69+CD40L+ Gag-specific CD4+ T cells vs. total population in a CP subject, assessed 9h after stimulation with a Gag peptide pool. (b,c) Quantification from (a). Comparison between CPs and ECs of (b) the percentage of Gag-specific CXCR5mem assessed by flow cytometry (two-sided Mann-Whitney test) (n=10 CPs and 10 ECs); and (c) the enrichment score of a TFH tonsil signature (GSE50391) by ssGSEA (two-sided Mann-Whitney test) (nCP=11, nEC=12). Bars represent the median +/−interquartile range. (d) Correlation of single-sample enrichment score for the TFH tonsil signature with viral load by two-tailed Spearman test (n= 11 CPs, 9 VCs, 12 ECs). (e) Volcano plots depicting differential gene expression between CP and EC, amongst their live-sorted Gag-specific CXCR5mem or CXCR5neg subsets, by microarray analysis. Red and pink dots represent DEG with log2|FC|>1 and padj<0.05 or log2|FC| >1, respectively, and the number of DEGs are displayed in the small tables. Two-sided moderated t-test followed by Benjamini-Hochberg correction method (padj) (n= 5 CPs and 3 ECs). (f) CAMERA enrichment analysis of the tonsil TFH signature of CXCR5mem or CXCR5neg Gag-specific CD4 T cells of CPs compared to ECs (n= 5 CPs and 3 ECs). Two sided p-value by CAMERA and FDR by Benjamini-Hochberg method. (g) Comparison of expression of TFH surface markers on CXCR5mem and CXCR5neg HIV-specific CD4+ T cells of CPs and ECs by flow cytometry (n= 10 CPs and 10 ECs). (h) mRNA expression of TFH-related surface markers or (i) transcription factors by qRT-PCR (Fluidigm) (n= 6 CPs and 6 ECs) on sorted CXCR5mem and CXCR5neg HIV-specific CD4+ T cells of CPs and ECs. (g,h,i) Two-tailed Mann-Whitney test or for paired comparisons by two-tailed Wilcoxon matched-pairs signed-ranked test were used to verify significance. Only p˂0.05 are displayed for clarity. Bars represent the median +/− interquartile range.

To define the cell subsets that contributed to the TFH transcriptional signature observed in CPs, we applied microarray analysis to paired live-sorted subsets of CXCR5mem and CXCR5neg HIV-specific CD4+ T cells from CPs and ECs (Fig.3e; gating strategy Supplementary Fig. 3a). CXCR5 mRNA expression was consistent with protein expression (Fig. 3h). Differences in gene expression between CPs and ECs were more significant in the CXCR5neg than in the CXCR5mem subsets of HIV specific CD4+ T cells (Fig. 3e). CPs displayed a marked enrichment of the TFH signature not only in the CXCR5mem compartment, but also in the CXCR5neg HIV-specific CD4+ T cell subset (Fig. 3f). We confirmed expression of several markers classically associated with TFH cells in CXCR5mem and CXCR5neg HIV-specific CD4+ T cells at the mRNA level by qRT-PCR and at the protein level by flow cytometry. Consistent with the microarray results, upregulation of the TCR co-receptors PD1, TIGIT, CD200 and ICOS on CXCR5neg HIV-specific CD4+ T cells clearly differentiated CPs from ECs (Fig. 3g and Supplementary Fig. 3c). Surface markers SLAMF6 and CD84 (both associated with help to B cells) and BTLA (highly expressed in TFH cells; Fig. 3h) were significantly higher in CXCR5neg CD4+ T cells of CPs compared to ECs, while the mRNA expression of BCL6 and TCF7 did not differ in CXCR5neg CD4+ T cells between the two groups. Transcriptional expression of PRDM1 and MAF was significantly higher in CXCR5neg CD4+ T cells of CPs compared to ECs (Fig. 3i).

In contrast to ECs, CXCR5 expression in CPs did not clearly delineate cells with a TFH transcriptional profile: ICOS, SLAMF6, BTLA, CD84, TIGIT, CD200 and the TFH-associated transcription factor cMAF mRNA levels in CXCR5neg cells were equal or even superior to their CXCR5mem autologous counterparts (Fig. 3ghi, Supplementary Fig. 3c). These data demonstrate the atypical upregulation of “TFH-like” signature in the CXCR5neg HIV-specific CD4+ T cells of CPs compared to ECs.

Robust expression of TFH cytokines in CXCR5neg HIV-specific CD4 of CP

We next sought to elucidate if the enriched TFH signature in CXCR5mem and CXCR5neg HIV-specific CD4+ T cells of CPs compared to ECs is congruent with the enhancement of some TFH functions and determine the phenotype of these effector cells. We assessed mRNA expression of the TFH cytokines CXCL13 and IL-21 at single-cell resolution using a sensitive flow cytometric RNA Fluorescent In Situ Hybridization (RNA Flow FISH), as previously described20, 21. This assay is superior to standard ICS for detection of IL-21-producing cells20. There were increased frequencies of CXCL13 mRNA+ and IL21 mRNA+ HIV-specific CD4+ T cells in CPs compared to ECs (Fig. 4a,b). This was a distinct feature of HIV-specific responses, as the expression after stimulation with the superantigen SEB was similar in both cohorts (Fig. 4c) and CMV pp65-specific CD4+ T cells expressed less CXCL13 and IL21 than HIV-specific CD4+ T cells in paired comparisons within the same humans (Supplementary Fig. 4a). As CD40L is not expressed on all Ag-specific CD4+ T cells upon activation, we confirmed that the differential expression of CXCL13 and IL21 between CPs and ECs hold true without combining the RNA Flow FISH and AIM assays and without pre-gating on CD40L+ cells (Supplementary Fig. S4bi). Largely distinct CD4+ T cell populations produced CXCL13 and IL21 mRNAs at the time point examined (Fig. 4d,e). We confirmed the increased CXCL13 production in CPs compared to ECs at the protein level, by measuring CXCL13 concentration by ELISA in the supernatant of CD8-depleted PMBCs after 48h stimulation with HIV Gag (Supplementary Fig. 5a). CXCL13+ and IL21+ cells expressed high levels of CXCR5 protein to compared to total HIV-specific CD4+ T cells in ECs. In contrast, the levels of CXCR5 expression on CXCL13+ and IL21+ cell were comparable to the total population in CPs (Fig 4f). This gain of TFH-like functions by CXCR5neg HIV-specific CD4+ T cell in CPs was further apparent when we compared the ratios of IL21+ and CXCL13+ responses between the CXCR5mem and CXCR5neg subsets (Fig. 4g). As the inhibitory receptors PD-1 and TIGIT are markers of TFH cells12 and may also be associated with exhaustion, we examined their expression on CXCL13 mRNA+ and IL21 mRNA+ HIV-specific CD4+ T cells. The majority of these cytokine-expressing cells were PD-1+/TIGIT+, particularly in CPs (Fig. 4h,i, Supplementary Fig. 5b,c.). Thus, the expression pattern of the inhibitory co-receptors PD-1 and TIGIT was consistent with TFH skewing and may not imply exhaustion of these responses.

Figure 4. TFH cytokine expression by CXCR5neg HIV-specific CD4+ T cells of chronic progressors.

Figure 4.

(a) Representative RNA flow FISH plots from an CP of CXCL13 and IL21 mRNA in CD69+CD40L+ CD4+ T cells after a 15h stimulation with a Gag peptide pool. Numbers represent the frequency of mRNA+ cells among (red) CD69+CD40L+ and (grey) total CD4+ T cells (n= 6 CPs and 5 ECs) (b,c) Quantification of (a). Comparison of frequencies of CXCL13 or IL21 mRNA+ CD4+ T cells between CPs (n=6) and ECs (n=5) obtained as in a after Gag or SEB stimulation. Bars represent median frequencies +/−interquartile range (two-tailed Mann-Whitney test, **p<0.01). (d,e) RNA flow FISH analysis of co-expression of CXCL13 and IL21 mRNA by Gag-specific CD4+ T cells with (d) representative plots from CP and EC people and (e) related statistical analysis by permutation test (10000 permutations) in SPICE (n= 6 CPs and 5 ECs). Pie slices represent median frequency of CXCL13/IL21 mRNA+ subpopulations. (f) Representative histograms from CP and EC people of CXCR5 expression on CXCL13 or IL21 mRNA+ HIV-specific CD4+ T cells overlaid on total HIV-specific CD4+ T cells (grey). Numbers represent the percentages of CXCR5+ cells within each cytokine+ population (n= 6 CPs and 5 ECs). (g) Statistical analysis from (f) on 6 CPs and 5 ECs of the ratios between the CXCR5mem to the CXCR5neg subsets of frequencies of cytokine or chemokine mRNA+ HIV-specific CD4+ T cells (CXCL13, IL21, IL2 and IFNγ) as assessed by RNA flow cytometry. Bars represent median frequencies +/−interquartile range (two-tailed Mann-Whitney test). (h,i) Co-expression patterns of PD-1 and TIGIT (surface protein Ab labeling) on CXCL13 mRNA+ or IL21 mRNA+ Gag-specific CD4+ T cells identified by RNA flow FISH (n= 6 CPs and 5 ECs). (h) Representative examples of CP and EC people; cytokine/chemokine mRNA+ HIV-specific CD4+ T cells (red dots for CPs, blue dots for ECs) are overlaid on total HIV-specific CD4+ T cell subpopulations (grey dots). Numbers represent the percentages of CXCL13 RNA+ or IL21 mRNA+ HIV-specific CD4+ T cells located in each quadrant. (i) SPICE analysis of PD-1 and TIGIT expression from (h) on CXCL13 mRNA+ or IL21 mRNA+ HIV-specific CD4+ T cells. (*p<0.05, **p<0.01 by permutation test, 10000 permutations). Slices represent median frequency of PD-1 and TIGIT subpopulations. (j) Comparison of p24-specific antibodies levels in plasma between CPs and ECs as assessed by ELISA. (two-tailed Mann-Whitney test (n= 12 CPs and 13). Bars represent medians +/− interquartile range. (k,l,m) Correlation between p24-specific antibodies levels measured by ELISA and frequencies of k) total HIV-specific CD4+ T cells; l) CXCR5mem HIV-specific CD4+ T cells and m) CXCR5neg HIV-specific CD4+ T cells assessed by the CD69/CD40L AIM assay after a 9h stimulation with a Gag peptide pool (two-tailed Spearman test, n=12 CPs and 13 ECs).

Finally, we explored possible links between HIV-specific CD4+ T cell help and antibody (Ab) responses. We assessed plasma levels of p24-specific Abs by ELISA and Biolayer Interferometry (BLI) binding analysis. The results obtained by the two methods were highly correlated (Supplementary Fig. 5d). Although p24-specific Ab concentrations in the CP and EC groups were overall similar (Fig. 4j), we observed a significant correlation between the magnitude of HIV-specific CD4+ T cell responses and p24-specific Abs, which was maintained when only the CP group was considered (Fig 4k). There was no significant association between CXCR5mem responses and p24-specific antibodies (Fig. 4l). However, there was a significant correlation between CXCR5neg HIV-specific CD4+ T cell frequencies and Ab responses in CPs, but not ECs (Fig. 4m), which might suggest that the CXCR5neg subset has T helper function to B cells in vivo.

These data demonstrate that dysregulated CXCR5neg HIV-specific CD4+ T cells in progressive infection gain functions and phenotypic features classically attributed to TFH cells.

Mucosal immunity-related cytokines in HIV-specific CD4 of ECs

As we observed an enriched TH17 signature in HIV-specific CD4+ T cells in ECs compared to CPs, we characterized the expression of IL-17F and IL-22 at the single-cell level. RNA Flow FISH enabled superior detection of these cytokines in HIV-specific CD4+ T cells compared to ICS (Fig. 5a,b). We observed more robust IL17F and IL22 HIV-specific CD4+ T cell responses in ECs compared to CPs (Fig 5c). In contrast, IL17F and IL22 expression patterns in SEB- or CMV activated CD4+ T cells did not differ between the groups (Fig. 5d, Supplementary Fig. S4a). We confirmed the strong IL-17F, IL-22 and IL-17A secretion by HIV-specific CD4+ T cells in ECs at the protein level, in contrast to low or undetectable production in CPs (Fig. 5e, Supplementary Fig. 5e,f,g).

Figure 5. Increased expression of cytokines related to mucosal immunity in HIV-specific CD4+ T cells of elite controllers compared to chronic progressors.

Figure 5.

(a,b). Representative flow cytometry plots depicting detection of IL22 and IL17F in CD69+CD40L+ CD4+ T cells in one EC after stimulation with a Gag peptide pool by delayed ICS for cytokine protein and RNA flow FISH for cytokine mRNA, respectively. Numbers represent frequencies of mRNA+ HIV-specific CD4+ T cells among (blue) CD69+CD40L+ and (grey) total CD4+ T cells (n= 8 CPs and 8 ECs). (c,d) Statistical comparison from (a) of frequencies of cytokine mRNA+ antigen-specific CD4+ T cells between CPs and EC after (c) a Gag peptide pool (n= 8 CPs and 8 ECs) or (d) SEB stimulation (n=6 CPs and 8 ECs) (two-tailed Mann-Whitney test). (e) Statistical comparison of IL-22 and IL-17F protein levels detected by Luminex beads array in the supernatant of CD8-depleted PBMCs 48h after stimulation with a Gag peptide pool (n= 8 CPs and 8 ECs). (c,d,e) Bars represent median with +/−interquartile range. *p<0.05, **p<0.01 by two-tailed Mann-Whitney test. (f) Representative flow cytometry plots of co-expression patterns of IL22 mRNA and IL17F mRNA in HIV-specific CD4+ T cells in CP and EC people (n=6 CPs and ^ ECs). (g) Statistical analysis from (f) by SPICE (*p<0.05 by permutation test, 10000 permutations). Slices represent median frequency of IL22/IL17F mRNA+ subpopulations of HIV-specific CD4+ T cells (n= 6 CPs and 6 ECs). (h,i) Comparison of the frequencies of CCR6 and CXCR3 expression on IL22 mRNA+ and IL17F mRNA+ HIV-specific CD4+ T cells compared to total HIV-specific CD4+ T cells in CPs (n=6) and ECs (n=6) by two-tailed Mann-Whitney test and by two-tailed Wilcoxon matched-pairs signed-ranked test. Only p˂0.05 are displayed for clarity. Bars represent median with interquartile range. (j,k) Correlation of (j) IL22 and (k) IL-17F mRNA levels assessed by qRT-PCR on sorted HIV Gag-specific CD4+ T cells and CD4+ T cell activation measured by HLA-DR and CD38 co-expression (two-tailed Spearman test; n= 10 CPs, 6 VCs, 12 ECs). (l,m) Statistical comparisons of microbial translocation in plasma in cohorts of CP, EC, and uninfected control donors (UD) (l) quantitation of bacterial RNA reads in plasma (transcripts per million, TPM); (m) bacterial RNA species diversity (Shannon entropy score) (Kruskal Wallis test; n= 10 CPs, 8 ECs and 6 UDs). Bars represent median +/−interquartile range. (n,o) Correlation between bacterial RNA species diversity in plasma and frequencies of (n) HIV-specific IL22+ CD4+ T cells and (o) HIV-specific IL17+ CD4+ T cells (two-tailed Spearman test; n=10 CPs and 8 ECs). (p,q) Correlation between abundance of Proteobacteria translocation and frequencies of (p) HIV-specific IL22+ CD4+ T cells and (q) HIV-specific IL17+ CD4+ T cells (two-tailed Spearman test; n=10 CPs and 8 ECs).

The IL17F mRNA+ and IL22 mRNA+ subsets constituted two partially overlapping cell populations (Fig. 5f,g). In both CPs and ECs, these cells preferentially expressed CCR6, a chemokine receptor critical for recruitment of TH17 and TH22 cells to mucosal tissues (Fig. 5h). Expression of CXCR3, a marker of TH1 cells also expressed on TH1-TH17 cells of mixed properties that are expanded in some inflammatory diseases17, was low on IL17F and IL22 mRNA+ of both CPs and ECs (Fig 5i, Supplementary Fig. 5h). Performing the RNA Flow FISH without the AIM assay and CD40L+ pre-gating showed again contrasting patterns of IL17F and IL22 expression between CPs and ECs (Supplementary Figure S4bi).

Systemic translocation of microbial products play a critical role in immune activation related to HIV pathogenesis22. Loss of gut mucosal integrity and TH17 cell depletion have been associated with immune activation and disease progression23. We thus investigated possible links between HIV-specific TH17 and TH22 responses and systemic immune activation. We observed an inverse correlation between IL22 mRNA expression by HIV-specific CD4+ T cells and co-expression of the activation markers HLA-DR and CD38 on total CD4+ and CD8+ T cells (Fig. 5j, Supplementary Fig. 5i). We identified similar associations for IL17F (Fig. 5k, Supplementary Fig. 5j). We next investigated the relationship between preservation of IL17+ and/or IL22+ HIV-specific CD4+ T cells and microbial translocation by deep sequencing of cell-free RNA fragments in plasma — a reflection of the part of the body microbiome that can access the circulation, the majority of which resides in the gut. The total levels of bacterial RNA reads in plasma did not significantly differ between the groups of CPs, ECs and uninfected control donors (UDs) (Fig. 5l). However, we observed a lower plasma bacterial RNA species diversity in CPs compared to the EC and UD groups (Fig. 5m). These observations may reflect low gut microbiome diversity, a state associated with systemic inflammation in several diseases24 (Supplementary Fig. 5k,l). IL22 and IL17F transcription in HIV-specific CD4+ T cells was significantly correlated with bacterial RNA species diversity (Fig. 5n,o). CPs and ECs qualitatively differed in the bacterial RNA profile, forming largely distinct clusters (Supplementary Fig. S5m). IL22 and IL17F mRNA levels in HIV-specific CD4+ T cells inversely correlated with the prevalence of Proteobacteria, a phylum of Gram-negative bacteria that is increased in the gut microbiome in HIV-infected people25 and has been proposed as a marker of dysbiosis26 (Fig. 5p,q).

Thus, robust IL22+ and IL17F+ HIV-specific CD4+ T cells can be detected in the blood of ECs, while these responses are severely impaired in CPs. Associations of this deficiency with systemic immune activation, reduced plasma bacterial RNA species diversity with a skewing towards greater Proteobacteria abundance suggest that these findings in blood reflect perturbed gut homeostasis and loss of protective mucosal immunity.

ART results in a distinct transcriptome of HIV-specific CD4

Finally, we sought to determine whether the observed HIV-specific CD4+ T cell profiles were the consequences of ongoing viral antigen exposure, or the results of durable cell-fate decision programs that would persist after viral suppression on ART. We longitudinally followed a subgroup of 8 CPs and analyzed a second sample obtained while on ART (participant characteristics: Supplementary Table 8). Pairwise comparisons between the EC, CP pre-ART (CPpre) and CP post-ART (CPpost) cohorts (Fig 6a,b) showed sets of DEGs between all three groups, with fewer differences in the CPpre-CPpost and CPpost-EC than the CPpre-EC comparison, as may be expected. We next focused on selected GO terms and gene signatures identified as differentially expressed between CPs and ECs in Figs 1 and 2. While initiation of ART in CP modulated the activity of several apoptotic pathways regulating apoptosis and gene expression towards the levels observed in EC, differences in the “Positive regulation of T helper cell differentiation”, “Positive regulation of CD4+ αβ T cell activation” and “Regulation of leukocyte migration” persisted (Fig 6c). We therefore examined how ART affected Thelper polarization signatures. Expression of the TFH signature in HIV-specific CD4+ T cells was markedly reduced by ART, to levels similar to that found in EC (Fig 6d), while expression of the TH1, TH17 and LCMV exhaustion signatures was little affected by therapy: the differences clearly persisted between CPpost and EC (Fig 6e,f, Supplementary Fig. S6a). Expression of selected individual genes of interest was also consistent with these findings when expression levels in EC were considered as references (Fig 6g, Supplementary Fig. S6b and Supplementary Tables 9 and 10). While the results should be considered as trends given the limited size of the cohorts, expression of some genes appeared corrected by ART (e.g, CXCL13), whereas others are not restored at all (e.g. IL22).

Figure 6. Distinct transcriptional imprint with reduction in TFH-associated gene expression but poor correction of TH1, TH17 and TH22 gene levels after suppression of viremia by ART.

Figure 6.

(a) Paired volcano plot comparisons depicting DEGs as measured by microarray analysis of sorted HIV-specific CD4+ T cells in 8 CPs before and after ART and 12 ECs. Red, pink and blue dots represent DEG with log2|FC| >1 and padj<0.05, log2FC>1 or padj<0.05, respectively. Moderated two-sided t test followed by Benjamini-Hochberg method. (b) Euler diagrams of DEG between each pairwise comparison of the three groups of participants (log2|FC|> 1, p<0.05); numbers represent DEGs. (c) Significant enriched Gene Ontology terms from the curated C5 MSigDB via CAMERA analysis. Blue, Red and white color denote negative, positive or not significant (as defined by FDR<0.05, respectively. Two sided p-value by CAMERA and FDR by Benjamini-Hochberg method. (d-f) Barcode plots of enriched Thelper polarization gene sets by CAMERA in paired comparisons between the three groups (n= 8 CPpre 8 CPpost and 12 EC). Red and blue lines denote positive and negative enrichment, respectively. TH1 signature: GSE59295 (TH1 vs TH2), TH17 signature: GSE49703 (TH17 vs TH1), TFH signature: GSE50391 (CXCR5high CD45RO vs CXCR5-tonsil samples). Two sided p-value by CAMERA and FDR by Benjamini-Hochberg method. (g) Differential expression of genes associated with CD4 T cell differentiation and function in HIV-specific CD4+ T cells of CPs before and after treatment compared to ECs. Red and purple denote logarithmic fold change for comparisons CPpre. vs EC and CPpost vs EC, respectively, by microarray analysis in 8 CP before/after ART and 12 ECs.

These data demonstrate that suppressive ART results in a transcriptional landscape of HIV-specific CD4+ T cells distinct from the profiles of both untreated CPs and ECs. We identified gene modules susceptible to manipulation of antigen load, while others were unresponsive to antiviral therapy alone.

Discussion

The molecular features of virus-specific CD4+ T cells associated with pathogen containment are poorly understood in chronic human infections. We used genome-wide transcriptional analyses following ex vivo stimulation with HIV antigen to uncover differences in HIV-specific CD4+ T cell differentiation and function in chronically infected humans with various viremia levels. An atypical enrichment of a TFH-like signature in CXCR5neg HIV-specific CD4+ T cells characterized progressive infection. In contrast, elite control of viral replication was associated with robust HIV-specific TH22 and TH17 responses, which correlated negatively with immune activation and correlated positively with a greater diversity of bacterial RNA species in plasma and lower Proteobacteria abundance. None of these differences were detected in SEB or CMVpp65-reactive cells, revealing antigen-specific TCR signaling-driven processes. Suppressive ART in chronic progressors elicited a distinct HIV-specific CD4+ T cell transcriptome, revealing in comparison to elite controllers, the presence of viremia sensitive (e.g, TFH signature) and viremia unresponsive (e.g, TH1 and TH17 and TH22 signatures) gene modules. These data suggest that HIV-specific CD4+ T cell dysregulation does not represent a mere loss of Thelper functions, as classically defined for immune exhaustion, but also an adaptation to environmental cues combined with persistent changes, likely resulting from durable cell-fate decision programs.

At first sight, the marked TFH signature18 identified in CPs contrasts with the murine LCMV model, in which TFH responses in chronic infection are critical for delayed viral control8. The extraordinary ability of HIV to generate escape mutations and elude autologous Ab neutralization contribute to these opposite associations between TFH differentiation and disease outcome. A study showed that TFH cells in chronic LCMV infection are capable of supporting generation of neutralizing Ab effective against the contemporaneous virus9. In contrast, the immune system lags behind the evolution of autologous HIV strains27. Kinetics of viral evolution in HIV infection may thus overcome the ability of adaptive TFH skewing to contribute to delayed viral control. While technical limitations have to date hampered investigation of HIV-specific GC TFH in lymph nodes, robust expansion of total GC TFH populations was demonstrated in viremic HIV-infected people and SIV-infected macaques, albeit with qualitative defects28, 29, 30, 31.

Genes classically associated with TFH cells that are enriched in the CXCR5neg HIV-specific CD4+ T cells of CPs include IL21 and CXCL13. HIV p24-specific Abs titers in plasma correlated with frequencies of CXCR5neg HIV-specific CD4+ T cells, suggesting that these responses may provide help to B cells in vivo. Studies in murine models have shown that CXCR5−/− CD4+ T cells are able to take over some B helper functions32, 33, resulting in the generation of low-affinity binding antibodies9. Acquisition of TFH-like properties by CXCR5neg CD4+ T cells has also been reported in rheumatoid arthritis34 and cancer35 and combines B help function with migration to inflamed tissues. These analogies suggest shared pathways of CD4+ T cell differentiation between diverse diseases associated with chronic inflammation.

Damage to the gut mucosa with associated microbial translocation22 and increased inflammatory properties of the enteric microbiota36 are major drivers of HIV-associated chronic inflammation. We observed that HIV-specific TH17 and TH22 responses were abundant in ECs and rare in CPs. IL-17F and IL-17A are closely related cytokines recruiting neutrophils to sites of inflammation. IL-22 promotes intestinal barrier function37, and supports complement-mediated elimination of pathobionts38. Lack of HIV-specific TH17 and TH22 responses was associated with CD4+ and CD8+ T cell activation and a reduced diversity of translocated bacterial RNA in plasma, with overrepresentation of Proteobacteria. These results are consistent with studies in SIV infection showing preferential translocation of Proteobacteria compared to other phylums from the gut lumen into tissues, resulting in enhanced immune activation39. Our data do not establish causal links between features of HIV-specific CD4+ T cells and microbial translocation. It is more likely that lack of HIV-specific TH-17/TH22 responses and altered translocated microbiome both result from persistent insults to the gastrointestinal tract. The paucity of HIV-specific IL17+ and IL22+ CD4+ T cells observed in CPs may result from high susceptibility of TH17 and TH22 subsets to HIV infection37, 40 and/or alteration of CD4+ T cell differentiation41. Together with the analysis of bacterial RNA fragments in plasma, these CD4+ T cell data suggest that studies of peripheral blood can provide useful insight into the systemic consequences of perturbed gut homeostasis. Probiotic and IL-21 therapy in ART-treated SIV-infected macaques was associated with enhanced polyfunctional TH17 expansion and reduced markers of microbial translocation and dysbiosis compared to ART alone42. It will be important to determine if such interventions restore HIV-specific CD4+ T cell profiles.

This work raises questions that will need to be addressed in further studies. AIM assays require activation to enable cell identification. While we believe that analyzing the transcriptional profile modulated by TCR signaling is highly relevant for analysis of T cell responses, it will be important to investigate other populations of HIV-specific CD4+ T cells that do not co-upregulate CD69 and CD40L, for example by other AIM markers or by tetramers. Experiments in animal models will be required to define the ontogeny of CXCR5neg CD4+ T cells with TFH-like features and whether they transitioned through a bona fide TFH stage during their development. Longitudinal studies from the time of acute infection may identify events in the genesis or persistence of HIV-specific CD4+ T cell dysregulation. Investigations of human gut and lymph node biopsies, while challenging, would define the relationships between features of HIV-specific CD4+ T cells in blood and other anatomical compartments.

In summary, this work identifies extensive alterations in HIV-specific CD4+ T cell transcriptional profiles and demonstrates that skewed lineage differentiation is a key feature of altered CD4+ T cell help in the setting of high viral load, in which atypical gain of function is combined with classical exhaustion features. The data also reveal distinct features of HIV-specific CD4+ T cells after successful ART. The identified molecular features may therefore help direct selective interventions to improve immune function. Given the paucity of transcriptome datasets available on pathogen-specific CD4+ T cells in humans, our results will also provide useful benchmark comparisons for other infectious diseases and vaccine-induced responses.

ONLINE METHODS

Participants and Samples

Leukaphereses were obtained from study participants at the Montreal General Hospital, Montreal, Canada; at the Centre Hospitalier de l’Université de Montréal (CHUM) in Montreal, Canada; and at the Massachusetts General Hospital (MGH) in Boston, USA. The study was approved by the respective IRBs, written informed consent obtained from all participants prior to enrolment and PBMC samples obtained by leukapheresis. Samples were collected between years 2013 and 2018. Subject characteristics are summarized in Supplementary Table 1. “Chronic progressors” (CP) were defined as people with a viral load of more than 5,000 vRNA copies per ml of plasma; “viremic controllers” (VC) were defined as people with more than 50 and less than 5,000 vRNA per ml plasma; and “elite controllers” (EC) were defined as subjects who spontaneously controlled viremia to below 50 RNA copies per ml plasma in the absence of therapy. CPs, VCs, and ECs.were either treatment naive or untreated for at least 3 months. Eight CPs were followed longitudinally and a second sample was obtained at least 6 months after initiation of ART therapy and suppression of viral load below 50 copies mL (Supplementary Table 8). PBMC were isolated by the Ficoll density gradient method and stored in liquid nitrogen.

Antibodies

For details of antibodies used, see Supplementary Tables 1120 and descriptions of use below. In all cases, antibodies are monoclonal and raised in mice unless otherwise stated. All antibodies are commercially available, were validated by the suppliers and titrated using biological and/or isotype controls.

CD40L/CD69 AIM assay for live-cell sorting of HIV Gag-specific CD4+ T cells

Cryopreserved PBMCs were thawed and rested at 37°C for 3 hours. Cells were aliquoted into wells of a 24-well plate, at a total of 15 × 106 cells per well in RPMI medium supplemented with 10% Human AB (HAB) serum and PenStrep. For each assay, we used a no exogenous stimulation (No Ag) condition as control and an HIV-specific stimulation with an overlapping peptide pool corresponding to HIV Gag (JPT, PM-HIV-Gag ULTRA), or HCMVA pp65 (JPT, PM-PP65) at a final concentration of 0.5μg/ml/peptide. A CD40 blocking antibody (Miltenyi Biotec, clone HB14) was added to each well 15 minutes prior to stimulation at a final concentration of 0.5 μg/ml. Following a 9 hr stimulation, the cells were stained for 30 min at 4°C before live cell sorting on a FACS Aria cell sorter (BD BioSciences) equipped for handling of biohazardous material. Two panels of antibodies were used to isolate live cells subsets according to combinations of surface markers. In the first panel for isolation of total CD69+CD40L+ Gag-specific CD4+ T cells, PBMCs were stained as indicated in Supplementary Table 11. The gating strategy for cell sorting is illustrated in Supplemental Figure 1. In the second panel used for isolation of CXCR5mem and CXCR5-subsets of CD69+CD40L+ Gag-specific CD4+ T cells, PBMCs were stained as indicated in Supplementary Table 12. The gating strategy for cell sorting is illustrated in Supplementary Fig. 3. The FACS Aria was operated at 70 pounds per square inch with a 70-μm nozzle. For all populations, 5,000 cells were collected directly into RLT lysis buffer (Qiagen).

Phenotyping of Gag-specific CD4+ T cells identified by the CD69/CD40L AIM assay.

PBMCs were thawed and plated in 24-well plates at 10 × 106/ml in RPMI + 10% HAB Serum (HAB) and rested for 3 hours at 37°C. Prior to stimulation, CD40 blocking antibody was added directly into culture at a final concentration of 0.5μg/ml, as well as antibodies against CXCR5, CCR6 and CXCR3. Cells were incubated for 15 min, then left unstimulated or stimulated either with an overlapping peptide pool corresponding to HIV Gag (JPT, PM-HIV-Gag ULTRA), at a final concentration of 0.5μg/ml/peptide, or with Staphylococcal Enterotoxin B (SEB) as a positive control (1 μg/mL). Cells were stimulated for 9h, collected and stained for 30 min at 4°C with the antibodies in Supplementary Table 13. Cells were washed, fixed for 20 min in 2% paraformaldehyde, washed, and resuspended in 1% FCS/PBS for flow acquisition on a 5-laser LSRII (BD BioSciences).

Phenotyping of immune activation markers

PBMCs were thawed and rested overnight at 37°C. Next day, cells were washed and stained with viability dye (20 min at 4 C°) and surface markers (20 min at 4 C°) (Supplementary Table 14). Fluorescence minus one staining was used to define the cutoff for positivity. Cells were washed, fixed for 1h in 2% paraformaldehyde, washed, and resuspended in 1% FCS/PBS for flow acquisition on a 5-laser LSRII (BD BioSciences).

Standard intracellular cytokine staining (ICS) for cytokine measurements

PBMCs were incubated for 6 hours with an HIV Gag peptide pool at a concentration of 0.5μg/ml/peptide in the presence of 2.5 μg/ml brefeldin A (BD GolgiPlug) and (0.3μl/ml) monensin (BD GolgiStop). For delayed ICS assays, 2.5 μg/ml brefeldin A (BD GolgiPlug) and (0.3μl/ml) monensin (BD GolgiStop) were added 9h after stimulation with the Gag peptide pool and cells were cultured for 12h. Unstimulated cells were used as negative control and SEB (0.5) μg/ml as positive control. Cells were then stained with viability dye (20 min at 4 C°), surface markers (20 min at 4 C°), fixed with Fixation Solution (eBioscience), and stained for intracellular proteins in permeabilization 1X buffer (eBioscience) (30 min at 4 C°) as described in Supplementary Tables 15 and 16. Cells were acquired on an LSR II (BD Biosciences).

Gene Expression Analysis

Whole Transcriptome Analysis by microarrays

Total RNA was purified using the RNeasy Plus Micro Kit (Qiagen). RNA integrity was assessed using a 2100 Bioanalyzer (Agilent Technologies). Sense-strand cDNA was synthesized from total RNA using a fixed volume of 3μL. Fragmentation and labeling were performed to produce ss-DNA with the Applied Biosystems™ GeneChip® WT Pico Terminal Labeling Kit according to manufacturer’s instructions (Applied Biosystems™). After fragmentation and labeling, 5 μg DNA target was hybridized on GeneChip® Clariom™ D, human (ThermoFisher Scientific) and incubated at 450C in the Genechip® Hybridization oven 640 (Affymetrix) for 17 hours at 60 rpm. GeneChips were then washed in a GeneChips® Fluidics Station 450 (Affymetrix) using Applied Biosystems™ Hybridization Wash and Stain kit according to the manufacturer’s instructions (Applied Biosystems™). The microarrays were finally scanned on a GeneChip® scanner 3000 (Affymetrix).

Validation of transcriptional expression by Fluidigm

Total RNA was purified using the RNeasy Plus Micro Kit (Qiagen). cDNA was synthesized using all RNA available (or 1–5 ng) with the High-Capacity Reverse Transcription Kit with RNase Inhibitor (Life Technologies) (25° C for 10 min, 37° C for 120 min, 85° C for 5 min). cDNA equivalent to 1000 sorted cells was subjected to gene-specific preamplification using Taqman Preamp MasterMix (Applied Biosystems) and 96 pooled TaqMan Assays (Applied Biosystems) (Supplementary Tables 2123) at final concentration 0.2X (95°C for 10 min, followed by 16 cycles of 95°C for 15 s and 60°C for 4 min). The preamplified cDNA was diluted 5-fold in DNA suspension buffer (Teknova) and was mixed with TaqMan Universal PCR Master mix (Life Technologies) and 20X GE sample loading reagent (Fluidigm). 20X Taqman assays were diluted 1:1 with 2X assay loading buffer (Fluidigm). Taqman assays mixtures were loaded onto a primed 96.96 Dynamic Array chip (Fluidigm). The chip was loaded into the IFC Controller, where each sample was mixed with each assay in every possible combination. The chip was transferred in a Biomark (Fluidigm) for real-time PCR amplification and fluorescence acquisition using single probe (FAM-MGB, reference: ROX) settings and the default hot-start protocol with 40 cycles. Cycle thresholds (Ct) were calculated using the Fluidigm BioMark software v1.4.2.

Combined cytokine/chemokine mRNA Flow FISH and protein staining assays

PBMCs were either left unstimulated or were stimulated with an HIV Gag peptide pool (JPT) or SEB for 15 hours after 15h min incubation with a CD40 blocking antibody (Miltenyi Biotec, clone HB14) at a final concentration of 0.5 μg/ml. For experiments for evaluation of mRNA expression without CD40 blocking antibody PBMCs were stimulated for 12h. After stimulation, cells were stained with a viability dye (20 min, 4°C, Fixable LiveDead, eBioscience) and then surface markers were labelled (30 min 4°C) with antibodies (See panels in Supplementary Tables 1720). Samples were next subjected to the PrimeFlow RNA assay (Affymetrix/eBioscience) for specific mRNA detection as per manufacturer’s instructions. All buffers and fixation reagents were provided with the kit, with the exception of flow cytometry staining buffer (2% FBS/PBS). After fixation and permeabilization, cytokine/chemokine mRNAs were labeled with sets of probe pairs, as listed in Supplementary Tables 1720. The probes were diluted 1:5 in diluent and hybridized to the target mRNA for 2 hr at 40 °C. Samples were washed to remove excess probes and stored overnight in the presence of RNAsin. Signal amplification was achieved as previously described20 by sequential 1.5 hr, 40 °C incubations with the pre-amplification and amplification mix. Amplified mRNA was labeled with fluorescently-tagged probes for 1 hr at 40 °C. A negative control probe and a positive control probe (against house-keeping gene RPL13A) were included in each experiment. Gates were set on the unstimulated control where appropriate or scrambled probes. Samples were acquired on an LSRII (BD Bioscience) or a BD LSRFortessa™ (experiments without CD40 blocking antibody). Analysis was performed using FlowJo (Treestar, V10) and Spice 5.35 software.

Measurements of cytokine secretion by beads arrays and ELISA

One million CD8 T cell-depleted PBMCs (Dynabeads CD8, StemCell) were incubated with an HIV Gag peptide pool (1μg/ml/peptide) or left unstimulated and incubated at 37°C in RPMI supplemented with 10% HAB and PenStrep. Cytokine secretion in supernatants was measured 48 hours after stimulation on a Luminex beads array platform, as previously described19, 20. Measurements were performed in duplicates using the Human High Sensitivity Cytokine Premixed Kit B (R&D Systems) for IL-2, IFN-γ, TNF-α, IL-17A, IL-22 and IL-31 or for IL-17F on a Bio-plex 200 array system (Bio-Rad Laboratories) per manufacturer’ s instructions. CXCL13 was measured separately with the Human CXCL13 Quantikine ELISA Kit (R&D Systems), as per manufacturer’s instructions.

Measurement of CD4i p24-specific antibodies by ELISA

The anti-p24 ELISA was performed as follows: 96 well plates (Thermo Scientific Nunc, FluoroNunc/LumiNunc, MaxiSorp Surface) were coated with 0.1μg/ml of recombinant p24 (NIH #12028) or BSA (Bioshop #ALB001.100) in PBS over night at 4°C. Plates were blocked for 90 min at room temperature (RT) with blocking buffer (TBS, Tween 0.1%, BSA 2%) and then washed 4 times with washing buffer (TBS, Tween 0.1%). Dilutions of human sera (1/1000, 1/3000, 1/6000) or rabbit anti-HIV p24 antiserum (NIH #4250) in washing buffer containing 0,1% of BSA were incubated for 2 h at RT. Plates were washed 4 times with washing buffer before incubation for 90 min at RT with HRP-conjugated secondary Abs: goat anti-human IgG HRP (Invitrogen #31413) or anti IgG rabbit HRP (Invitrogen #31462). Plates were then washed 4 times with washing buffer before revealing with standard ECL (Perkin Elmer #NEL105001EA) with a TriStar luminometer (LB 941, Berthold Technologies).

Plasma microbiome sequencing

Circulating total RNA and DNA were isolated from frozen plasma using RNAzolBD (MRCgene) according to the manufacturer’s recommendations. After RNA fragmentation and reverse transcription using random hexamers, obtained transcripts as well as the isolated plasma DNA were individually incorporated into barcoded cDNA libraries based on the Illumina TruSeq platform. Libraries were validated by microelectrophoresis, quantified, pooled and clustered on Illumina patterned flow cells. Clustered flow cells were sequenced on an Illumina HiSeq 4000 in 75-base paired reads.

Gene expression data analysis

Microarrays:

The preprocessing of the microarray data was done in R43 v3.3.3 using the oligo package44 v1.38.0. Specifically, the CEL files were loaded in R and the rma function was applied for background subtraction, normalization and summarization. Quality control was performed by inspecting various diagnostic plots of the intensity distribution and correlation structure of control and regular probes. After review of the QC plots, 3 outliers were removed. Combat (sva package 3.30.1) has been applied for batch normalization of samples using 4 replicates as reference for the normalization. The normalized data were used for subsequent subanalyses CPpre vs EC, CPpre vs CPpost, CPpost vs EC.

Differential expression:

Differential analysis of gene expression was done by the R package limma45. Using the lmFit function, a linear model was fit to each gene separately. Moderated t-tests were performed between each group. P-values were adjusted using Benjamini-Hochberg method for multiple test correction.

CAMERA

TH1 signature (GSE59295; TH1 vs TH2) and TH17 signature (GSE49703; TH17 vs TH1) were available in C7 collection of MSigDB. The TFH signature (GSE50391; CXCR5high CD45RO vs CXCR5-tonsil samples) and LCMV exhaustion signature (GSE41866; LCMV Clone 13 D30 vs Armstrong D30-Exhausted vs Memory) were created using the GEO2R online tool (https://www.ncbi.nlm.nih.gov/geo/geo2r/). For each signature, a gene set test was performed using the camera function from the limma package, accounting for inter-gene correlation.

Venn diagrams were created using the online tool Eulerr.co, (R package version 4.1.0, https://cran.r-project.org/package=eulerr)46. Unsupervised hierarchical clustering was performed using the in heatmap2 function in R43 on relative signal intensity values applying the Euclidean distance metric and complete linkage clustering method. Barcode plots were generated with the barcodeplot function from the limma package.

qRT-PCR results

Analysis of the qRT-PCR data obtained on the microfluidic platform was carried out using GenEx software (MultiD Analyses, URL: http://www.multid.se). Five endogenous control genes were included in the Fluidigm run and the stability of endogenous control genes across all experimental samples was evaluated applying the NormFinder algorithm47 in GenEx. The mean expression of the most stable endogenous control genes was used for normalization and calculation of -ΔCt values. Principal component analysis, biplots and correlograms were created using the prcomp, fviz_pca_biplot and corrplot functions, respectively, in R programming language.

Statistical analyses of flow cytometry, beads array and qRT-PCR results

Statistical analyses were performed using Prism V6.0 (GraphPad) using non-parametric tests. Two-group comparisons were performed using the Mann-Whitney test and pairwise comparisons were performed using the Wilcoxon matched-pair test. For comparisons between three groups, Kruskal-Wallis or Friedman one-way ANOVA with Dunn’s post-test was used. Permutation test (10000 permutations) was applied for pie-chart comparison using the SPICE software. For correlations, Spearman’s R correlation coefficient was applied. Statistical tests were two-sided and p<0.05 were considered significant.

Bioinformatic analysis of deep sequencing results of cell-free RNA fragments in plasma

Raw, paired-end Illumina reads were first trimmed for low quality bases and adaptor contamination with Trimmomatic48. As a first pass to remove host-derived sequences, trimmed reads were first aligned against the HG38 genome with Bowtie 249 and then aligned to HG38 transcriptome with the splice aware aligner, STAR50. Unaligned paired-end reads were then assembled into contigs using Trinity51. Trinity contigs were then aligned against HG38 again to further remove host-derived sequences with BLAST+52. Remaining contigs were then aligned against the NCBI nt database53 and quantified with Salmon54. Taxonomic information was assigned for each contig and TPM values were collapsed at the species level for each sample for downstream analysis. TPM values for each sample were collapsed on the Genus taxonomic level and Morisita-Horn Dissimilarity indexes were calculated on a pairwise basis using the R package, vegan55. The pairwise dissimilarity matrix was then clustered and the heatmap was produced with gplots56.

Supplementary Material

1
2
3

ACKNOWLEDGEMENTS

We thank Josée Girouard, the clinical staff at the McGill University Health Centre in Montreal, the Ragon/MGH clinical and technical staff and all study participants for their invaluable role in this project; Dr Bruce Walker for providing clinical samples; Dr Dominique Gauchat and the CRCHUM Flow Cytometry Platform for technical assistance; Nathalie Hamel and McGill University And Génome Québec Innovation Centre for microarray analysis; Drs Jeremy Boss and Shane Crotty for their input on this manuscript. The following reagent was obtained through the NIH AIDS Reagent Program, Division of AIDS, NIAID, NIH: HIV-1 IIIB p24 Recombinant Protein from ImmunoDX, LLC. This study was supported by the National Institutes of Health grants HL092565 (D.E.K), AI100663 CHAVI-ID (D.E.K, R.T.W; Consortium PI: D. Burton), OD011103 (R.P.J) and OD011132 (R.P.J); the Canadian Institutes for Health Research grants #137694 (D.E.K), #152977 (D.E.K), MOP-93770 (C.T) and foundation #352417 (A.F); the Canada Foundation for Innovation Program Leader grant #31756 (D.E.K); the FRQS AIDS and Infectious Diseases Network. This work was funded in part by the Intramural Program of the National Institutes of Health (D.C.D, S.D). D.E.K and C.T are supported by Senior Research Scholar Awards of the Quebec Health Research Fund (FRQS). J.P.R is the holder of Louis Lowenstein Chair in Hematology & Oncology, McGill University. A.F. is a Canada Research Chair on Retroviral Entry.

Data availability

Microarray data generated during the current study were deposited in the Gene Expression Omnibus (GEO) public depository with the accession number GSE128297 for the SuperSeries, and the following accession numbers for the Subseries; GSE129872 HIV-specific CD4+ T cells samples from CPs, VCs and ECs), GSE128280 (CXCR5mem and CXCR5neg HIV-specific CD4+ T cells from CPs and ECs), GSE128296 (HIV-specific CD4+ T cells samples from CPs before/after ART and ECs). mRNA expression data by high-throughput qRT-PCR are available in Supplementary Material. All the datasets that support the findings of this study are available from the corresponding author on reasonable request.

Footnotes

Life Sciences Reporting Summary.

Further information on research design is available in the Life Sciences Reporting Summary linked to this article.

Competing interests

The authors declare no competing interests

REFERENCES

  • 1.Laidlaw BJ, Craft JE & Kaech SM The multifaceted role of CD4(+) T cells in CD8(+) T cell memory. Nat Rev Immunol 16, 102–111 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Swain SL, McKinstry KK & Strutt TM Expanding roles for CD4(+) T cells in immunity to viruses. Nat Rev Immunol 12, 136–148 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.O’Shea JJ & Paul WE Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science 327, 1098–1102 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wherry EJ & Kurachi M Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 15, 486–499 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Crawford A et al. Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection. Immunity 40, 289–302 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Morou A, Palmer BE & Kaufmann DE Distinctive features of CD4+ T cell dysfunction in chronic viral infections. Curr Opin HIV AIDS 9, 446–451 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Vella LA, Herati RS & Wherry EJ CD4(+) T Cell Differentiation in Chronic Viral Infections: The Tfh Perspective. Trends Mol Med 23, 1072–1087 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Fahey LM et al. Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells. J Exp Med 208, 987–999 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Greczmiel U et al. Sustained T follicular helper cell response is essential for control of chronic viral infection. Sci Immunol 2 (2017). [DOI] [PubMed] [Google Scholar]
  • 10.Kaufmann DE et al. Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat Immunol 8, 1246–1254 (2007). [DOI] [PubMed] [Google Scholar]
  • 11.Ferre AL et al. HIV controllers with HLA-DRB1*13 and HLA-DQB1*06 alleles have strong, polyfunctional mucosal CD4+ T-cell responses. J Virol 84, 11020–11029 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Crotty S T follicular helper cell differentiation, function, and roles in disease. Immunity 41, 529–542 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Reiss S et al. Comparative analysis of activation induced marker (AIM) assays for sensitive identification of antigen-specific CD4 T cells. PLoS One 12, e0186998 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wu D & Smyth GK Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res 40, e133 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ono C et al. Fluorescently activated cell sorting followed by microarray profiling of helper T cell subtypes from human peripheral blood. PLoS One 9, e111405 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhang W et al. Effector CD4+ T cell expression signatures and immune-mediated disease associated genes. PLoS One 7, e38510 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ramesh R et al. Pro-inflammatory human Th17 cells selectively express P-glycoprotein and are refractory to glucocorticoids. J Exp Med 211, 89–104 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Locci M et al. Human circulating PD-1+CXCR3-CXCR5+ memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody responses. Immunity 39, 758–769 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Porichis F et al. Responsiveness of HIV-specific CD4 T cells to PD-1 blockade. Blood 118, 965–974 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Porichis F et al. High-throughput detection of miRNAs and gene-specific mRNA at the single-cell level by flow cytometry. Nat Commun 5, 5641 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Baxter AE et al. Multiparametric characterization of rare HIV-infected cells using an RNA-flow FISH technique. Nat Protoc 12, 2029–2049 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Brenchley JM et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med 12, 1365–1371 (2006). [DOI] [PubMed] [Google Scholar]
  • 23.Favre D et al. Critical loss of the balance between Th17 and T regulatory cell populations in pathogenic SIV infection. PLoS Pathog 5, e1000295 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Levy M, Kolodziejczyk AA, Thaiss CA & Elinav E Dysbiosis and the immune system. Nat Rev Immunol 17, 219–232 (2017). [DOI] [PubMed] [Google Scholar]
  • 25.Vujkovic-Cvijin I et al. Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism. Sci Transl Med 5, 193ra191 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Shin NR, Whon TW & Bae JW Proteobacteria: microbial signature of dysbiosis in gut microbiota. Trends Biotechnol 33, 496–503 (2015). [DOI] [PubMed] [Google Scholar]
  • 27.Richman DD, Wrin T, Little SJ & Petropoulos CJ Rapid evolution of the neutralizing antibody response to HIV type 1 infection. Proc Natl Acad Sci U S A 100, 4144–4149 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Petrovas C et al. CD4 T follicular helper cell dynamics during SIV infection. J Clin Invest 122, 3281–3294 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cubas RA et al. Inadequate T follicular cell help impairs B cell immunity during HIV infection. Nat Med 19, 494–499 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lindqvist M et al. Expansion of HIV-specific T follicular helper cells in chronic HIV infection. J Clin Invest 122, 3271–3280 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Perreau M et al. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J Exp Med 210, 143–156 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Miyauchi K et al. Protective neutralizing influenza antibody response in the absence of T follicular helper cells. Nat Immunol 17, 1447–1458 (2016). [DOI] [PubMed] [Google Scholar]
  • 33.Kahan SM & Zajac AJ Late arising T follicular helper cells cultivate the B cell crop during chronic infections. Sci Immunol 2 (2017). [DOI] [PubMed] [Google Scholar]
  • 34.Rao DA et al. Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 542, 110–114 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gu-Trantien C et al. CD4(+) follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest 123, 2873–2892 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Neff CP et al. Fecal Microbiota Composition Drives Immune Activation in HIV-infected Individuals. EBioMedicine 30, 192–202 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kim CJ et al. A role for mucosal IL-22 production and Th22 cells in HIV-associated mucosal immunopathogenesis. Mucosal Immunol 5, 670–680 (2012). [DOI] [PubMed] [Google Scholar]
  • 38.Hasegawa M et al. Interleukin-22 regulates the complement system to promote resistance against pathobionts after pathogen-induced intestinal damage. Immunity 41, 620–632 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Klase Z et al. Dysbiotic bacteria translocate in progressive SIV infection. Mucosal Immunol 8, 1009–1020 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.El Hed A et al. Susceptibility of human Th17 cells to human immunodeficiency virus and their perturbation during infection. J Infect Dis 201, 843–854 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Favre D et al. Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 alters the balance of TH17 to regulatory T cells in HIV disease. Sci Transl Med 2, 32ra36 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ortiz AM et al. IL-21 and probiotic therapy improve Th17 frequencies, microbial translocation, and microbiome in ARV-treated, SIV-infected macaques. Mucosal Immunol 9, 458–467 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Team RCR: A language and environment for statistical computing. In: Computing, R.F.f.S., editor. Vienna, Austria; 2014. [Google Scholar]
  • 44.Carvalho BS & Irizarry RA A framework for oligonucleotide microarray preprocessing. Bioinformatics 26, 2363–2367 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ritchie ME et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43, e47 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Larsson J eulerr: Area-Proportional Euler and Venn Diagrams with Ellipses. R package version 4.1.0. 2018.
  • 47.Andersen CL, Jensen JL & Orntoft TF Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res 64, 5245–5250 (2004). [DOI] [PubMed] [Google Scholar]
  • 48.Bolger AM, Lohse M & Usadel B Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Langmead B & Salzberg SL Fast gapped-read alignment with Bowtie 2. Nat Methods 9, 357–359 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dobin A et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Grabherr MG et al. Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat Biotechnol 29, 644–652 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Altschul SF, Gish W, Miller W, Myers EW & Lipman DJ Basic local alignment search tool. J Mol Biol 215, 403–410 (1990). [DOI] [PubMed] [Google Scholar]
  • 53.O’Leary NA et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res 44, D733–745 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Patro R, Duggal G, Love MI, Irizarry RA & Kingsford C Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods 14, 417–419 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Oksanen J et al. vegan: Community Ecology Package. R package version 2.4–3.; 2017.
  • 56.Warnes G & al, e. gplots: Various R programming tools for plotting data. 2005.

Associated Data

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

Supplementary Materials

1
2
3

RESOURCES