Abstract
T follicular helper cells are the main CD4+ T cells specialized in supporting B-cell responses, but their role in driving transfusion-associated alloimmunization is not fully characterized. Reports of T follicular helper subsets displaying various markers and functional activities underscore the need for better characterization/identification of markers with defined functions. Here we show that a previously unidentified subset of human circulating T follicular helper cells expressing TIGIT, the T-cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory domains, exhibit strong B-cell help functions. Compared to the subset lacking the receptor, T follicular helper cells expressing this receptor up-regulated co-stimulatory molecules and produced higher levels of interleukins (IL-21 and IL-4) critical for promoting B-cell activation/differentiation. Furthermore, this subset was more efficient at inducing the differentiation of B cells into plasmablasts and promoting immunoglobulin G production. Blocking antibodies abrogated the B-cell help properties of receptor-expressing T follicular helper cells, consistent with the key role of this molecule in T follicular helper-associated responses. Importantly, in chronically transfused patients with sickle cell anemia, we identified functional differences of this subset between alloimmunized and non-alloimmunized patients. Altogether, these studies suggest that expression of the T-cell immunoreceptor with Ig and immunoreceptor tyro-sine-based inhibitory domains not only represents a novel circulating T follicular helper biomarker, but is also functional and promotes strong B-cell help and ensuing immunoglobulin G production. These findings open the way to defining new diagnostic and therapeutic strategies in modulating humoral responses in alloimmunization, and possibly vaccination, autoimmunity and immune deficiencies.Introduction
T follicular helper (TFH) cells have emerged as the main effector CD4 T cells specialized in supporting B-cell responses to generate the initial wave of antibody response as well as in promoting B-cell differentiation into high affinity antibody-producing cells and long-lasting IgG antibody.1 TFH cells express chemokine (C-X-C motif) receptor 5 (CXCR5),2–4 which allows their migration into B-cell follicles in response to its ligand CXCL13. Bcl-6 is the main lineage-associated transcription factor driving TFH differentiation.5,6 Interleukin (IL)-21 is the canonical TFH-associated cytokine driving B-cell help,5,7,8 although TFH cells can also secrete additional cytokines promoting growth, differentiation and class-switching of B cells, such as IL-4.9,10 TFH cells also express several key co-stimulatory molecules specialized in providing B-cell help, including inducible T-cell co-stimulator (ICOS),11,12 required for T-cell proliferation and T/B-cell interactions as well as CD40 lig-and (CD40L), a potent activator of B cells, inducing their activation and differentiation.1
Transfusion therapy remains an important treatment modality for patients with sickle cell disease (SCD). Despite its therapeutic benefits, 20–60% patients with SCD develop alloantibodies with specificities against disparate antigens on transfused red blood cells, causing complications ranging from life-threatening hemolytic transfusion reactions, to logistical problems in finding compatible red cells for transfusion.13 Given their key role in providing help to B cells and driving antibody responses, TFH cells are likely to be critical in alloimmunization biology. In a recent study of a cohort of transfused patients with SCD studied by Vingert et al.,14 despite a higher frequency of TFH cells in non-alloimmunized patients, only the alloimmunized group displayed antigen-specific TFH cells expressing IL-21, suggesting that TFH cells may play a role in alloimmunization.
Exciting new studies indicate that circulating TFH-related cells (cTFH), ranging from 5–25% of CD4 memory cells, exist in the peripheral blood in humans and mice, and can promote antibody production.7,12,15–20 These blood TFH cells (CD45RA) express similar markers as lymphoid TFH cells (including CXCR5, ICOS, CD40L and IL-21),21 but do not express the Bcl-6 protein.7,12,15–17,22 Importantly, cTFH levels correlate with auto-antibodies and levels of protective antibodies following vaccination.12,16,20,23,24 Several cTFH subsets have been reported, each displaying drastically different functions.25 For example, cTFH subsets harboring TH1-, TH2- and TH17-like effector functions have been identified in humans, with TH2- and TH17-like cTFH being more efficient in inducing B cells to produce IgG than TH1-like cTFH.12,21 Furthermore, recent studies have identified lymphoid Foxp3 TFH cells [named follicular regulatory T (TFR) cells] that can suppress germinal center reactions,26–28 also detectable in the periphery.29,30 In addition, similar to lymphoid TFH, cTFH cells can express significant levels of surface programmed cell death-1 (PD-1), 31 although the function of PD-1 on TFH cells remains controversial since it is associated with both promotion16,17 and inhibition of B-cell responses.18,32–34 Taken together, these reports underscore the need for better characterization of markers for cTFH cells displaying defined functions not only in steady state but also in diseases. Remarkably, PD-1 has been described as a member of the growing family of inhibitory receptors also referred to as immune checkpoints, responsible for aborting T-cell responses.35 Interestingly, another member of the immune checkpoint family, TIGIT (T-cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory domains), was reported to be over-expressed on both tonsillar and cTFH cells,17 and was shown to be involved in interactions between T cells and follicular dendritic cells to regulate B-cell responses.36,37 However, the functional activity of TIGIT on TFH cells, including cTFH cells, has not been studied to date.
In this study, we took the approach of using TIGIT and PD-1 to characterize the phenotype and function of circulating TFH subsets and to investigate whether expression of these molecules on cTFH cells modulated their functions in healthy volunteer donors and in a group of chronically transfused SCD patients with or without alloantibodies.
Methods
Human samples
All studies were approved by the Institutional Review Boards of the New York Blood Center (NYBC). De-identified fresh leukopaks were obtained from healthy donors at the NYBC. For SCD patients’ samples, blood was obtained solely from discard apheresis waste bags obtained during erythrocytapheresis procedures at the Children’s Hospital of Philadelphia (see Online Supplementary Material for details).
T-cell studies
Freshly-sorted CD4 T-cell subsets and autologous naïve or memory B cells were used (see Online Supplementary Material for details). Blocking antibodies for TIGIT38 and PD-134,39 were pre-incubated with sorted T cells before being co-cultured with autologous B cells.
Results
PD-1+ cTFH cells co-express TIGIT and represent a limited fraction of TIGIT+ cTFH cells
In healthy donors, a large subset of cTFH cells, as defined by CD4CD45RACXCR5 T cells in peripheral blood, express TIGIT: 6.3%±0.8 of all CD4 T cells (Figure 1A,B) or 47%±3.2 of cTFH (Figure 1A,C). Strikingly, we also found that >92% PD-1-expressing cTFH cells co-express TIGIT and that cTFH cells expressing PD-1, but lacking TIGIT (PD-1/TIGIT) were barely detectable (<0.042%±0.008 of CD4 T cells or 2.0%±0.4 of cTFH). Within the cTFH subset, the PD-1/TIGIT cTFH population represented a significantly lower frequency of cTFH (8.6%±0.9) as compared to PD-1/TIGIT cTFH cells (32.5%±1.6, P=0.0001; Figure 1).
Expression of ICOS, CD40L and IL-21 by TIGIT+ cTFH cells
We next tested whether TIGIT cTFH cells were functionally different from cTFH cells lacking TIGIT (PD-1/TIGIT). To this end, PD-1/TIGIT and PD-1/TIGIT CXCR5 cTFH populations from a small number of healthy donors (n=3 or 4) were sorted and their ability to express TFH-associated co-stimulatory markers and cytokines following stimulation was compared to those of sorted autologous PD-1/TIGIT subsets (gating strategy shown in Online Supplementary Figure S1). As a control, sort-purified autologous CXCR5 non-cTFH cells expressing TIGIT (“TIGIT+”) or not (“TIGIT-”) were also tested.
We first monitored expression of co-stimulatory molecules CD40L and ICOS, both specialized in providing B-cell help, on T-cell subsets before or after stimulation by immunostaining. TIGIT cTFH cells significantly over-expressed ICOS (>2-fold) prior to stimulation i.e. at resting state (Figure 2A,C; P=0.001 as compared to TIGIT cTFH cells or CXCR5 non-TFH cells; n=3). Although not statistically significant, CD40L was also over-expressed (>2-fold) by TIGIT cTFH cells (Figure 2B,D). After stimulation by phorbol myristate acetate/ionomycin, the expression of both ICOS and CD40L was still significantly higher on TIGIT cTFH cells than on TIGIT cTFH cells (Figure 2E,F) or CXCR5 non-TFH cells. Altogether, these data suggest that TIGIT cTFH cells are well-equipped to provide B-cell help, as they display stable intrinsic functions both at resting and activated states.
Next the production of TFH-associated cytokine IL-21 by TIGIT cTFH cells was assessed upon stimulation with phorbol myristate acetate/ionomycin. Significantly, TIGIT cTFH cells expressed more IL-21 (n=4, PD1/TIGIT: 14.4%±2.4 and PD1/TIGIT: 17.8%±3.2) than TIGIT cTFH cells (6.5%±2.1) or CXCR5 non-TFH cells (Figure 2G and Online Supplementary Figure S2A). We also tested expression of interferon gamma (IFNγ) and IL-4 (Online Supplementary Figure S2B,C, respectively). In contrast, TIGIT cTFH cells produced less IFNγ than TIGIT cTFH cells (Online Supplementary Figure S2B) and overall, fewer CXCR5 cTFH cells expressed IFNγ as compared to CXCR5 non-TFH cells. Within cTFH cells, IL-4 production was also measured, but no significant differences were found between subsets (Online Supplementary Figure S2C). Given the role of IL-21 in driving antibody responses, these data suggest that cTFH cells could represent an efficient subset in promoting humoral responses.
TIGIT + cTFH cells constitute a novel subset
Several CXCR5 cTFH subsets with distinct capabilities in supporting B-cell activation and antibody production have been characterized, including three subsets distinguished based on their co-expression of TH1, TH2 or TH17 features and differential expression of chemokine receptors, namely CXCR3 and CCR6.7,12 We therefore compared TIGIT cTFH cells to CXCR3/CCR6 (type 1-like), CXCR3/CCR6(type 2-like) and CXCR3/CCR6 (type 17-like) cTFH subsets. These subsets all contained significant levels (from 24% to 54% of total cTFH cells) of at least two out of the three subsets, specifically PD-1/TIGIT (“DP”) PD-1/TIGIT (“SP”) and/or PD-1/TIGIT (“DN”) cells (Figure 3A). Furthermore, PD-1/TIGIT, PD1/TIGIT and PD-1/TIGIT subsets each displayed type 1, 2 and 17-like cTFH cells (Figure 3B). Each subset contained between 10% and 61% of every type 1, 2 and 17-like subsets within total cTFH cells, demonstrating that TIGIT cTFH subset(s) is(are) separate/independent, at least phenotypically, of these three previously described subsets.
Together these results establish TIGIT cTFH cells as a phenotypically distinct subset potentially efficient at promoting humoral responses.
TIGIT functionally drives TFH function/B-cell help
To further characterize TIGIT cTFH cells and determine whether TIGIT was functionally responsible for their ability to promote B-cell help, sorted PD-1/TIGIT, PD-1/TIGIT and PD-1/TIGIT subsets from healthy donors were co-cultured with autologous CD27 naïve or CD27 memory B cells in the presence of SEB to facilitate T/B-cell interactions. At day 7, TIGIT cTFH cells (both PD-1 and PD-1) expressed 1.5- to 3-fold higher levels of both ICOS and CD40L compared to TIGIT cTFH cells when co-cultured with CD27 naïve or CD27 memory B cells (Online Supplementary Figure S3).
T-cell subsets were also assessed at day 7 for cytokine expression upon re-stimulation using phorbol myristate acetate/ionomycin to assess non-specific T-cell function. Both PD-1 and PD-1 TIGIT cTFH cells expressed more IL-21 than TIGIT cTFH cells when co-cultured with naïve or memory B cells (Figure 4A). As expected, non-TFH cells produced less IL-21 than cTFH cells, including TIGIT cTFH cells (Figure 4A). Strikingly, TIGIT cTFH cells, independently of their PD-1 expression, also produced less IFNγ than TIGIT cTFH cells when co-cultured with naïve or memory B cells (Figure 4B). Furthermore, TIGIT cTFH cells, regardless of PD-1 expression, produced higher levels of IL-4 than TIGIT cT cells, although the increase was only significant in co-cultures with naïve B cells (Figure 4C). Of note, both CXCR5 and CXCR5 cells produced similar amounts of IL-4 (Figure 4C). Hence, within cTFH cells, TIGIT expression increases IL-21 and IL-4 production, but inhibits IFNγ production.
The ability of sorted cTFH subsets to drive B cells to differentiate into CD19/CD38 plasmablasts was also determined on day 7 by immunostaining. Naïve and memory B cells differentiated into plasmablasts more efficiently when co-cultured with TIGIT cTFH cells than with TIGIT cTFH cells (Figure 5A,B and Online Supplementary Figure S4). Accordingly, more IgG was produced by both naïve (>1.3-to 1.7-fold) and memory B cells (>3-fold) co-cultured with TIGIT cTFH cells than with TIGIT cTFH cells (Figure 5C and Online Supplementary Figure S4).
To further confirm that TIGIT expression strengthens B-cell help function, blocking antibodies against TIGIT or PD-1 were added to T/B-cell co-cultures. Relative to isotype control, up-regulation of ICOS by TIGIT cTFH subsets expressing PD-1 or not, co-cultured with memory B cells were significantly inhibited by anti-TIGIT (22% to 30%, Figure 6A). Of note, the anti-TIGIT antibody clone used (MBSA43) has previously been reported to block TIGIT signaling,38 further supporting the belief that this decrease results from TIGIT blockade. However, PD-1/TIGIT cells were not inhibited by anti-PD-1 (Figure 6A), despite the known blocking function of this antibody.34,39 As expected, PD-1 blockade did not affect PD-1/TIGIT cell responses. To a lesser extent, TIGIT cTFH cells co-cultured with naïve B cells also displayed decreased levels of ICOS (23% to 28% inhibition compared to isotype control) when TIGIT, but not PD-1, was blocked (Figure 6A). No significant inhibition of CD40L was observed using the same antibodies (data not shown). Furthermore, IL-21 expression by TIGIT cTFH cells was significantly inhibited by TIGIT blockade (49% to 66% inhibition compared to isotype control), but not by anti-PD-1 (Figure 6B). On the other hand, IFNγ and IL-4 expression was not affected by TIGIT blockade (data not shown). Regarding B-cell responses, a trend toward lower plasmablast differentiation (between 26% to 65% inhibition) was observed in co-cultures with TIGIT cTFH cells following TIGIT blockade, but not using an anti-PD-1 antibody (Figure 6C). IgG production (between 39% to 59% inhibition) was also decreased by anti-TIGIT in naïve and memory cell co-cultures with TIGIT cTFH cells (Figure 6D). There was a trend towards lower IgG responses with anti-PD-1 in co-cultures with TIGIT cTFH cells; however, the inhibition may not be specific as the same effect was also observed in co-cultures with TIGIT cTFH cells lacking PD-1 (Figure 6D).
TIGIT expression by cTFH in patients with sickle cell disease
To assess whether TIGIT-expressing CXCR5 cTFH cells were different in SCD patients between those who were alloimmunized (with a history and/or currently detectable red blood cell-specific allo-antibodies) and those who were not alloimmunized (had never produced any auto- or allo-antibodies), we analyzed a group of SCD patients on a chronic transfusion protocol (n=21). The frequency of CD4 cells was lower in SCD patients than in healthy donors (data not shown), consistent with a previous report.40 The percentage of total cTFH cells within CD4 T cells was also lower in samples from chronically transfused SCD patients (7.8%±1.1% versus 11.2%±1.2% in healthy donors). Since peripheral blood mononuclear cells from healthy donors were obtained from leukocyte-enriched peripheral blood whereas peripheral blood mononuclear cells from SCD patients were derived from their transfusion exchange waste bags, we restricted all subsequent analysis to comparing data from alloimmunized versus non-alloimmunized SCD patients in order to control for potential confounding effects related to blood collection and transfusion. No significant difference in the percentage of total cTFH cells within CD4 T cells was detected between samples from non-alloimmunized (n=6) or alloimmunized patients currently expressing antibodies (“active”; n=5) or not (“non-active”; n=10) (Figure 7A). As seen in healthy donors, the majority of PD-1 cTFH cells co-expressed TIGIT in this patient population, but no significant difference in the percentage of TIGIT cTFH subsets between alloimmunized or non-alloimmunized patients were detected (Figure 7B). Additionally, levels of TIGIT expression per cTFH cell did not differ between patient groups or healthy donors (data not shown).
TIGIT-dependence of cTFH-mediated allo-responses in patients with sickle cell disease
To further investigate TIGIT-dependent functions in samples from chronically transfused patients with SCD, we performed 7-day cTFH subset/B-cell co-culture experiments using sorted PD-1/TIGIT, PD-1/TIGIT or PD-1/TIGIT cTFH subsets from 16 SCD patients. For these analyses, alloimmunized patients without (n=6) or with (n=4) detectable alloantibodies were grouped as alloimmunized due to the limited number of patients studied. Relative to the PD-1/TIGIT cTFH subset, there was a trend toward higher levels of ICOS (Figure 7C) and CD40L (Figure 7D) on TIGIT cTFH cells co-cultured with autologous B cells from alloimmunized as compared to non-alloimmunized patients. Strikingly, and consistent with the observed co-stimulatory molecule expression pattern, TIGIT cTFH cells relative to the PD-1/TIGIT cTFH subset from alloimmunized patients also produced significantly more IL-21 (>2.5-fold increase) upon re-stimulation (Figure 7E) as compared to non-alloimmunized patients. As in healthy donors, IFNγ production by TIGIT cTFH cells, relative to PD-1/TIGITcTFH, was also lower, but no significant difference was detected between the two groups of patients (Figure 7F).
B-cell differentiation into CD19/CD38 plasmablasts was determined as for healthy donors at day 7. Poor plasmablast differentiation, particularly from naïve B cells, was observed for several patients. For patients whose memory B cells differentiated into plasmablasts, their global IgG production was determined by enzyme-linked immunosorbent assay. A trend toward higher IgG levels was detected in TIGIT cTFH co-cultures from alloimmunized versus non-alloimmunized patients (Figure 7G), although significance was not reached, likely due to the limited numbers analyzed. Consistent with their red cell alloimmunization state, red blood cell-specific IgG was detected in the B-cell-TIGIT cTFH co-culture supernatants in four of ten alloimmunized patients [adjusted mean fluorescence intensity (MFI): 7, 14, 122, 193] and in one of six of non-alloimmunized patients, although the latter was only weakly reactive (adjusted MFI: 5, Online Supplementary Figure S5).
Altogether these results suggest that TIGIT cTFH cells from chronically transfused alloimmunized patients with SCD are more potent in providing B-cell help than those from non-alloimmunized patients.
Discussion
The present study identifies TIGIT-expressing circulating TFH cells as a novel subset displaying heightened B-cell help functions. Specifically, we found that TIGIT cTFH cells expressed higher levels of ICOS and CD40L co-stimulatory molecules associated with B-cell help, as compared to TIGIT cTFH cells at steady state as well as following activation (Figure 2A–F and Online Supplementary Figure S3). Upon stimulation, they also secrete substantially more IL-21, and to some extent IL-4, than TIGIT cTFH cells (Figures 2G and 4A,C). Both cytokines, especially IL-21, play major roles in promoting B-cell activation/differentiation and antibody production. Accordingly, TIGIT cTFH cells induced enhanced differentiation into plasmablasts from both naïve and memory B cells relative to the TIGIT cTFH subset (Figure 5A,B and Online Supplementary Figure S4). B-cells co-cultured with TIGIT cTFH cells also produced significantly higher levels of IgG than when co-cultured with other T-cell subsets (Figure 5C and Online Supplementary Figure S4), further emphasizing the key role of TIGIT cTFH cells in supporting B-cell responses and antibody production. Of note, the higher levels of IgG produced by memory as compared to naïve B cells in TIGIT cTFH cell co-cultures may reflect differences in interactions between TIGIT cTFH cells and naïve versus memory B cells. Indeed, TIGIT binds its high affinity receptor CD155 on target cells.36,41 CD155 is expressed by both memory and naïve B cells, albeit at slightly higher levels on memory B cells (Online Supplementary Figure S6). This may explain the differential IgG production of the B-cell subsets upon co-culture with identical TIGIT-expressing cTFH cells, but needs further investigation.
Anti-TIGIT blocking experiments indicated a functional role for TIGIT on cTFH cells. Indeed, TIGIT blockade in co-cultures, using naïve or memory B cells, decreased TIGIT cTFH cells’ ability to up-regulate ICOS (Figure 6A) and to produce IL-21 (Figure 6B) and also prevented B cells from secreting IgG (Figure 6D). It remains to be determined whether TIGIT signaling improves cTFH cells’ ability to stimulate B cells (e.g. through IL-21 secretion) or whether TIGIT co-stimulation to B cells directly induces B-cell maturation/differentiation.
CXCR5 cTFH subsets exhibiting distinct functions have been characterized, including three subsets based on co-expression of type 1, 2 or 17 features and differential expression of CXCR3 and/or CCR6.7,12 None of the cTFH subsets defined based on TIGIT (and PD-1) expression entirely falls into one of the type 1, 2 or 17 cTFH subsets or vice versa (Figure 3), demonstrating that TIGIT cTFH subsets are distinct from these three previously described subsets. Surprisingly, PD-1/TIGIT cTFH cells contained around 50% of CXCR3/CCR6 type 1 TFH cells, formerly reported to secrete IFNγ In our assays, and consistent with data obtained in non-TFH cells,42–45 TIGIT/PD-1 cTFH cells produced very low amounts of IFNγ, reinforcing the idea that these subsets are substantially different. Nonetheless, whether TIGIT/PD1 cTFH subsets can differentially modulate isotype switching as described for type 1, 2 and 17-like cTFH cells, remains unknown. Taken together, these results show that TIGIT cTFH cells represent a phenotypically and potentially functionally distinct subset from those described previously, although a direct comparison of the activities of TIGIT cTFH and these three cTFH subsets or other previously-described TFH subsets7,12,15–20 remains to be performed.
In our study, PD-1 cells represented about 10% of cTFH cells; others have found this population to be about 30% of cTFH in humans.16,17 This discrepancy could be due to different anti-PD-1 clones used for detection and isolation of the PD-1 cell population [MIH4 (us) versus eBioJ105 (others)]. PD-1 human cTFH cells were previously shown to drive heightened B-cell help responses,16,17 although PD-1 expression on TFH cells has also been associated with inhibition of B-cell responses.18,32–34 Remarkably, we found no difference in B-cell help function between TIGIT cTFH cells expressing PD-1 or not (Figures 2, 4, and 5 and Online Supplementary Figure S2), and virtually all PD-1 cTFH cells co-expressed TIGIT (Figure 1). Furthermore, unlike other experimental systems,18,32–34 no notable effects of PD-1 blockade on TFH cells were detected in our system using purified cell populations. Together, these results raise the possibility that PD-1 expression on human cTFH cells might represent a non-functional biomarker and that the function of these cells might be due to their concomitant TIGIT expression. This hypothesis is also compatible with the results obtained with blocking antibodies: while TIGIT blockade affected B-cell help, PD-1 blockade had no specific effect (Figure 6). To date, TIGIT has mainly been studied for its role in inhibiting type 1 responses, e.g. IFNγ and IL-2 production or cytotoxicity.37,41,46–48 Our data indicate that TIGIT also drives TFH responses. This raises the possibility that other immune “checkpoints”, especially those over-expressed by TFH cells such as BTLA49 may also strengthen TFH responses. While representing a provocative hypothesis, this could reflect an evolutionary mechanism by which TIGIT and potentially other immune checkpoints are not only involved in reducing chronic inflammation, but also in switching lymphocyte responses from cellular toward potentially more adaptive humoral responses.
Our study of chronically transfused SCD patients suggests that alloimmunized patients have TIGIT cTFH cells with more robust B-cell help functions than non-alloimmunized patients (Figure 7C–G, Online Supplementary Figure S5). We should note that patients’ samples were from apheresis waste bags whereas leukocyte-enriched preparations, rather than whole blood, were used for the healthy donor studies. Direct comparison of the TIGIT cTFH compartment between SCD patients and healthy donors was not, therefore, possible; such studies would require whole blood sampling, a challenging undertaking given the anemic state of the patients being studied. Instead, we compared the functional characteristics of TIGIT cTFH populations in alloimmunized versus non-alloimmunized SCD patients. Since all the samples from patients with SCD were taken from teenagers or young adults (15–30 years of age) who had been on a chronic red cell exchange program for 2 or more years and had, therefore, been heavily exposed to allogeneic transfusions, the comparison of alloimmunized versus non-alloimmunized patients’ cells is especially interesting. Relative to TIGIT cTFH cells, IL-21 and CD40L expression by TIGIT cTFH cells from non-alloimmunized patients was lower (whereas it was higher on TIGIT cTFH cells from alloimmunized patients). Defective TIGIT signaling in non-alloimmunized patients and/or an exacerbated TIGIT-mediated responses in alloimmunized patients may be responsible for such functional differences and needs to be further investigated. Additionally, the presence of regulatory Foxp3/CXCR5 follicular TFR26–30 may also participate in modulating cTFH functions. Our preliminary data suggest that alloimmunized patients with currently detectable alloantibodies have lower frequencies of circulating TFR than alloimmunized patients whose alloantibodies were undetectable at the time of drawing blood or healthy donors (manuscript in preparation). Unraveling mechanisms responsible for different functional activities of TIGIT cTFH cells between groups of SCD patients without or with alloantibodies would represent a key step in identifying predictive bio-markers as well as developing therapeutic strategies to limit SCD alloimmunization.
In conclusion, we have identified TIGIT as a novel marker of cTFH with ability to drive potent B-cell help responses These findings have the potential to influence therapeutic strategies, not only regarding alloimmunization, but also in vaccination as well as in autoimmunity and immune deficiencies in which the role of TFH cells, particularly cTFH cells, is being increasingly appreciated.21
Acknowledgments
We thank the members of the Laboratory of Complement Biology, Drs. Yunfeng Lui, Lifeng Liu (NYBC) and Ms. Meredith Spadaccia for technical help and discussions. We are grateful to Randy Velliquette, Christine Lomas-Francis, and Dr. Connie Westhoff of the Immunohematology Laboratory of NYBC for help in selecting appropriate RBC panel cells for RBC-specific anti-IgG detection in culture supernatant studies. This work was in part supported by an NIH grant R01HL121415 (to KY), an American Heart Association grant, 14GRNT20480109 (to KY) and the Badgeley Charitable Trust toward the purchase of reagents.
Footnotes
- ↵2 Current address: Chinese Academy of Medical Sciences, Beijing, China
- The online version of this article has a Supplementary Appendix.
- Authorship and DisclosuresInformation on authorship, contributions, and financial & other disclosures was provided by the authors and is available with the online version of this article at www.haematologica.org.
- Received June 23, 2015.
- Accepted August 3, 2015.
References
- Crotty S. Follicular helper CD4 T cells (TFH). Ann Rev Immunol. 2011; 29:621-663. PubMedhttps://doi.org/10.1146/annurev-immunol-031210-101400Google Scholar
- Breitfeld D, Ohl L, Kremmer E. Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med. 2000; 192(11):1545-1552. PubMedhttps://doi.org/10.1084/jem.192.11.1545Google Scholar
- Kim CH, Rott LS, Clark-Lewis I, Campbell DJ, Wu L, Butcher EC. Subspecialization of CXCR5+ T cells: B helper activity is focused in a germinal center-localized subset of CXCR5+ T cells. J Exp Med. 2001; 193(12):1373-1381. PubMedhttps://doi.org/10.1084/jem.193.12.1373Google Scholar
- Schaerli P, Willimann K, Lang AB, Lipp M, Loetscher P, Moser B. CXC chemokine receptor 5 expression defines follicular homing T cells with B cell helper function. J Exp Med. 2000; 192(11):1553-1562. PubMedhttps://doi.org/10.1084/jem.192.11.1553Google Scholar
- Chtanova T, Tangye SG, Newton R. T follicular helper cells express a distinctive transcriptional profile, reflecting their role as non-Th1/Th2 effector cells that provide help for B cells. J Immunol. 2004; 173(1):68-78. PubMedhttps://doi.org/10.4049/jimmunol.173.1.68Google Scholar
- Liu X, Nurieva RI, Dong C. Transcriptional regulation of follicular T-helper (Tfh) cells. Immunol Rev. 2013; 252(1):139-145. PubMedhttps://doi.org/10.1111/imr.12040Google Scholar
- Bentebibel SE, Lopez S, Obermoser G. Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination. Sci Transl Med. 2013; 5(176):176ra32. PubMedhttps://doi.org/10.1126/scitranslmed.3005191Google Scholar
- Bryant VL, Ma CS, Avery DT. Cytokine-mediated regulation of human B cell differentiation into Ig-secreting cells: predominant role of IL-21 produced by CXCR5+ T follicular helper cells. J Immunol. 2007; 179(12):8180-8190. PubMedhttps://doi.org/10.4049/jimmunol.179.12.8180Google Scholar
- Reinhardt RL, Liang HE, Locksley RM. Cytokine-secreting follicular T cells shape the antibody repertoire. Nat Immunol. 2009; 10(4):385-393. PubMedhttps://doi.org/10.1038/ni.1715Google Scholar
- Yusuf I, Kageyama R, Monticelli L. Germinal center T follicular helper cell IL-4 production is dependent on signaling lymphocytic activation molecule receptor (CD150). J Immunol. 2010; 185(1):190-202. PubMedhttps://doi.org/10.4049/jimmunol.0903505Google Scholar
- Choi YS, Kageyama R, Eto D. ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity. 2011; 34(6):932-946. PubMedhttps://doi.org/10.1016/j.immuni.2011.03.023Google Scholar
- Morita R, Schmitt N, Bentebibel SE. Human blood CXCR5(+)CD4(+) T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity. 2011; 34(1):108-121. PubMedhttps://doi.org/10.1016/j.immuni.2010.12.012Google Scholar
- Yazdanbakhsh K, Ware RE, Noizat-Pirenne F. Red blood cell alloimmunization in sickle cell disease: pathophysiology, risk factors, and transfusion management. Blood. 2012; 120(3):528-537. PubMedhttps://doi.org/10.1182/blood-2011-11-327361Google Scholar
- Vingert B, Tamagne M, Habibi A. Phenotypic differences of CD4(+) T cells in response to red blood cell immunization in transfused sickle cell disease patients. Eur J Immunol. 2015; 45(6):1868-1879. PubMedhttps://doi.org/10.1002/eji.201445187Google Scholar
- Chevalier N, Jarrossay D, Ho E. CXCR5 expressing human central memory CD4 T cells and their relevance for humoral immune responses. J Immunol. 2011; 186(10):5556-5568. PubMedhttps://doi.org/10.4049/jimmunol.1002828Google Scholar
- He J, Tsai LM, Leong YA. Circulating precursor CCR7(lo)PD-1(hi) CXCR5(+) CD4(+) T cells indicate Tfh cell activity and promote antibody responses upon antigen reexposure. Immunity. 2013; 39(4):770-781. PubMedhttps://doi.org/10.1016/j.immuni.2013.09.007Google Scholar
- Locci M, Havenar-Daughton C, Landais E. Human circulating PD-(+)1CXCR3(−)CXCR5(+) memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody responses. Immunity. 2013; 39(4):758-769. PubMedhttps://doi.org/10.1016/j.immuni.2013.08.031Google Scholar
- Sage PT, Francisco LM, Carman CV, Sharpe AH. The receptor PD-1 controls follicular regulatory T cells in the lymph nodes and blood. Nat Immunol. 2013; 14(2):152-161. PubMedhttps://doi.org/10.1038/ni.2496Google Scholar
- Saito R, Onodera H, Tago H. Altered expression of chemokine receptor CXCR5 on T cells of myasthenia gravis patients. J Neuroimmunol. 2005; 170(1–2):172-178. PubMedhttps://doi.org/10.1016/j.jneuroim.2005.09.001Google Scholar
- Simpson N, Gatenby PA, Wilson A. Expansion of circulating T cells resembling follicular helper T cells is a fixed phenotype that identifies a subset of severe systemic lupus erythematosus. Arthritis Rheum. 2010; 62(1):234-244. PubMedhttps://doi.org/10.1002/art.25032Google Scholar
- Schmitt N, Bentebibel SE, Ueno H. Phenotype and functions of memory Tfh cells in human blood. Trends Immunol. 2014; 35(9):436-442. PubMedhttps://doi.org/10.1016/j.it.2014.06.002Google Scholar
- Kitano M, Moriyama S, Ando Y. Bcl6 protein expression shapes pre-germinal center B cell dynamics and follicular helper T cell heterogeneity. Immunity. 2011; 34(6):961-972. PubMedhttps://doi.org/10.1016/j.immuni.2011.03.025Google Scholar
- Pallikkuth S, Parmigiani A, Silva SY. Impaired peripheral blood T-follicular helper cell function in HIV-infected nonresponders to the 2009 H1N1/09 vaccine. Blood. 2012; 120(5):985-993. PubMedhttps://doi.org/10.1182/blood-2011-12-396648Google Scholar
- Zhu C, Ma J, Liu Y. Increased frequency of follicular helper T cells in patients with autoimmune thyroid disease. J Clin Endocrinol Metab. 2012; 97(3):943-950. PubMedhttps://doi.org/10.1210/jc.2011-2003Google Scholar
- Ma CS, Deenick EK. Human T follicular helper (Tfh) cells and disease. Immunol Cell Biol. 2014; 92(1):64-71. https://doi.org/10.1038/icb.2013.55Google Scholar
- Chung Y, Tanaka S, Chu F. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat Med. 2011; 17(8):983-988. PubMedhttps://doi.org/10.1038/nm.2426Google Scholar
- Linterman MA, Pierson W, Lee SK. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. 2011; 17(8):975-982. PubMedhttps://doi.org/10.1038/nm.2425Google Scholar
- Wollenberg I, Agua-Doce A, Hernandez A. Regulation of the germinal center reaction by Foxp3+ follicular regulatory T cells. J Immunol. 2011; 187(9):4553-4560. PubMedhttps://doi.org/10.4049/jimmunol.1101328Google Scholar
- Sage PT, Alvarez D, Godec J, von Andrian UH, Sharpe AH. Circulating T follicular regulatory and helper cells have memory-like properties. J Clin Invest. 2014; 124(12):5191-5204. PubMedhttps://doi.org/10.1172/JCI76861Google Scholar
- Wang L, Qiu J, Yu L, Hu X, Zhao P, Jiang Y. Increased numbers of CD5+CD19+ CD1dhighIL-10+ Bregs, CD4+Foxp3+ Tregs, CD4+CXCR5+Foxp3+ follicular regulatory T (TFR) cells in CHB or CHC patients. J Transl Med. 2014; 12:251. PubMedhttps://doi.org/10.1186/s12967-014-0251-9Google Scholar
- Cubas RA, Mudd JC, Savoye AL. Inadequate T follicular cell help impairs B cell immunity during HIV infection. Nat Med. 2013; 19(4):494-499. PubMedhttps://doi.org/10.1038/nm.3109Google Scholar
- Hams E, McCarron MJ, Amu S. Blockade of B7-H1 (programmed death ligand 1) enhances humoral immunity by positively regulating the generation of T follicular helper cells. J Immunol. 2011; 186(10):5648-5655. PubMedhttps://doi.org/10.4049/jimmunol.1003161Google Scholar
- Liu T, Lu X, Zhao C, Fu X, Zhao T, Xu W. PD-1 deficiency enhances humoral immunity of malaria infection treatment vaccine. Infect Immun. 2015; 83(5):2011-2017. PubMedhttps://doi.org/10.1128/IAI.02621-14Google Scholar
- Velu V, Titanji K, Zhu B. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature. 2009; 458(7235):206-210. PubMedhttps://doi.org/10.1038/nature07662Google Scholar
- Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012; 12(4):252-264. PubMedhttps://doi.org/10.1038/nrc3239Google Scholar
- Boles KS, Vermi W, Facchetti F. A novel molecular interaction for the adhesion of follicular CD4 T cells to follicular DC. Eur J Immunol. 2009; 39(3):695-703. PubMedhttps://doi.org/10.1002/eji.200839116Google Scholar
- Joller N, Hafler JP, Brynedal B. Cutting edge: TIGIT has T cell-intrinsic inhibitory functions. J Immunol. 2011; 186(3):1338-1342. PubMedhttps://doi.org/10.4049/jimmunol.1003081Google Scholar
- Lozano E, Dominguez-Villar M, Kuchroo V, Hafler DA. The TIGIT/CD226 axis regulates human T cell function. J Immunol. 2012; 188(8):3869-3875. PubMedhttps://doi.org/10.4049/jimmunol.1103627Google Scholar
- Seung E, Dudek TE, Allen TM, Freeman GJ, Luster AD, Tager AM. PD-1 blockade in chronically HIV-1-infected humanized mice suppresses viral loads. PLoS One. 2013; 8(10):e77780. PubMedhttps://doi.org/10.1371/journal.pone.0077780Google Scholar
- Kaaba SA, al Harbi SA. Reduced levels of CD2+ cells and T-cell subsets in patients with sickle cell anaemia. Immunol Lett. 1993; 37(1):77-81. PubMedhttps://doi.org/10.1016/0165-2478(93)90135-OGoogle Scholar
- Yu X, Harden K, Gonzalez LC. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol. 2009; 10(1):48-57. PubMedhttps://doi.org/10.1038/ni.1674Google Scholar
- Barber DL, Wherry EJ, Masopust D. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006; 439(7077):682-687. PubMedhttps://doi.org/10.1038/nature04444Google Scholar
- Benson DM, Bakan CE, Mishra A. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010; 116(13):2286-2294. PubMedhttps://doi.org/10.1182/blood-2010-02-271874Google Scholar
- Hamid O, Robert C, Daud A. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013; 369(2):134-144. PubMedhttps://doi.org/10.1056/NEJMoa1305133Google Scholar
- Wherry EJ, Ha SJ, Kaech SM. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity. 2007; 27(4):670-684. PubMedhttps://doi.org/10.1016/j.immuni.2007.09.006Google Scholar
- Levin SD, Taft DW, Brandt CS. Vstm3 is a member of the CD28 family and an important modulator of T-cell function. Eur J Immunol. 2011; 41(4):902-915. PubMedhttps://doi.org/10.1002/eji.201041136Google Scholar
- Lozano E, Dominguez-Villar M, Kuchroo V, Hafler DA. The TIGIT/CD226 axis regulates human T cell function. J Immunol. 2012; 188(8):3869-3875. PubMedhttps://doi.org/10.4049/jimmunol.1103627Google Scholar
- Stanietsky N, Simic H, Arapovic J. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci USA. 2009; 106(42):17858-17863. PubMedhttps://doi.org/10.1073/pnas.0903474106Google Scholar
- M’Hidi H, Thibult ML, Chetaille B. High expression of the inhibitory receptor BTLA in T-follicular helper cells and in B-cell small lymphocytic lymphoma/chronic lymphocytic leukemia. Am J Clin Pathol. 2009; 132(4):589-596. PubMedhttps://doi.org/10.1309/AJCPPHKGYYGGL39CGoogle Scholar