Next Article in Journal
Emerging Strategies to Combat β-Lactamase Producing ESKAPE Pathogens
Next Article in Special Issue
Resident Memory T Cells and Their Role within the Liver
Previous Article in Journal
Optimization of the Biocatalysis for D-DIBOA Synthesis Using a Quick and Sensitive New Spectrophotometric Quantification Method
Previous Article in Special Issue
Needle in a Haystack: The Naïve Repertoire as a Source of T Cell Receptors for Adoptive Therapy with Engineered T Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tfh Cells in Health and Immunity: Potential Targets for Systems Biology Approaches to Vaccination

1
The Kirby Institute, UNSW Sydney, Sydney 2052, Australia
2
St Vincent’s Clinical School, UNSW Sydney, Sydney 2052, Australia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(22), 8524; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21228524
Submission received: 16 October 2020 / Revised: 9 November 2020 / Accepted: 10 November 2020 / Published: 12 November 2020
(This article belongs to the Special Issue Recent Advances in T Cell Immunity)

Abstract

:
T follicular helper (Tfh) cells are a specialised subset of CD4+ T cells that play a significant role in the adaptive immune response, providing critical help to B cells within the germinal centres (GC) of secondary lymphoid organs. The B cell receptors of GC B cells undergo multiple rounds of somatic hypermutation and affinity maturation within the GC response, a process dependent on cognate interactions with Tfh cells. B cells that receive sufficient help from Tfh cells form antibody-producing long-lived plasma and memory B cells that provide the basis of decades of effective and efficient protection and are considered the gold standard in correlates of protection post-vaccination. However, the T cell response to vaccination has been understudied, and over the last 10 years, exponential improvements in the technological underpinnings of sampling techniques, experimental and analytical tools have allowed multidisciplinary characterisation of the role of T cells and the immune system as a whole. Of particular interest to the field of vaccinology are GCs and Tfh cells, representing a unique target for improving immunisation strategies. Here, we discuss recent insights into the unique journey of Tfh cells from thymus to lymph node during differentiation and their role in the production of high-quality antibody responses as well as their journey back to the periphery as a population of memory cells. Further, we explore their function in health and disease and the power of next-generation sequencing techniques to uncover their potential as modulators of vaccine-induced immunity.

1. Introduction

The adaptive immune response relies on the orchestration of a complex network of interactions between antigen, numerous cell types and the signalling and effector molecules they produce. The ability to generate immunological memory is dependent on T cells, and the diversity of the T cell receptor (TCR) repertoire plays a significant role in maintaining the delicate balance between the ability to recognise antigen with extraordinary specificity and the ability to continue detecting an immense range of potential pathogens. Ideally, T cell responses would always establish sterilising immunity, but realistically, the obstacles of antigenic variability, pathogen evasion strategies and T cell exhaustion mean total elimination of the pathogen may not be possible.
Vaccination is undoubtedly one of the most successful interventions for reducing the burden of infection-related disease on communities, and it is responsible for significant decreases in disease prevalence, morbidity, and mortality globally [1]. Establishing immune memory to prevent the spread of pathogens throughout communities is the ultimate goal of vaccination. This is achieved during the germinal centre (GC) reaction, where the primary function is to establish and maintain memory cell populations that can mediate rapid recall responses and prolonged production of antibodies [2]. Of particular interest in the GC reaction are the interactions between Tfh cells and GC B cells and how the relationship between these cell populations could be leveraged to improve immune responses to vaccination.
The study of the differentiation and functioning of Tfh cells has demonstrated that dysregulation in these processes can drive inadequate immune responses and can be linked to autoimmunity. It is therefore crucial to understand the development and complex functioning of Tfh cells to potentially prevent or counteract their dysregulation and for generation of rational vaccine design. In this review, we discuss the differentiation of naïve T cells into fully mature Tfh cells, the complex interactions between Tfh and B cells in the GC and the role of circulating Tfh (c-Tfh) cells in the periphery during health and disease. In addition, we discuss the contribution of next-generation sequencing techniques to our understanding of other immune cell biology and the potential future applications of these technologies in uncovering insights into Tfh cell responses to infection and vaccination.

2. Role of the T Cell Receptor

The ability to maintain immune homeostasis, control continuous, multiple insults to the immune system and generate immunological memory is dependent on T cells. Defence against numerous pathogens, allergens and tumours can be attributed to the generation of a highly diverse repertoire of TCRs during cell development in the thymus [3,4]. Each T cell possesses a TCR that enables response to an antigen with extraordinary specificity whilst retaining enough heterogeneity as a population to support recognition of the immense diversity of antigens. The TCR expressed on a cell surface also plays a role in self-tolerance, cell lineage and cell fate decisions. Each TCR has a unique antigen-binding site formed by three complementarity-determining region (CDR) loops, which engage with the peptide–major histocompatibility complex (pMHC) molecule and determine the antigen(s) bound to the surface of antigen-presenting cells (APCs) that the T cell will respond to [5,6]. In addition to the combinatorial diversity achieved through pairing of different variable (V), diversity (D) and joining (J) genes, the enormous diversity of the TCR repertoire is further enhanced by the variation in the amino acid sequence at the primary antigen-binding site, termed the CDR3 [7,8,9]. Although only a small set of genes encode for the TCR, recombination of non-contiguous sequences and deletion and insertion of nucleotides increase the number of potential TCRs to between 1015 and 1020 clonotypes [10,11]. However, the realistic possible diversity of the repertoire is limited by the ~1012 T cells within the human body [12]. One study utilised high-throughput sequencing to determine that the thymic repertoire of αβ T cells ranged from 40 to 70 × 106 and 60 to 100 × 106 unique β and α sequences, respectively [13]. Therefore, the ability of one TCR to recognise and effectively respond to more than one antigen, a concept termed cross-reactivity, is essential to our ability to mount immune responses against the large potential diversity of pathogens [14]. Characteristics of the repertoire, including TCR affinity, TCR avidity, clonality and breadth, all play substantial roles in our ability to identify and respond to the immense diversity of often highly variable antigens (reviewed in [4]). Therefore, although it is evident that specificity and diversity can be achieved, the balance between TCR frequency, cross-reactivity and diversity is essential to optimum immune surveillance.

3. Lymphocyte Development in the Thymus

The stages of differentiation and functional maturation of thymocytes are marked by rearrangement of the TCR genes, expression of the TCR and expression of cell surface proteins. Interaction with the thymic stroma initiates commitment to and differentiation along the T cell lineage pathway. The initial phase of thymocyte differentiation is termed the double-negative (DN) phase due to the absence of expression of the T cell coreceptors CD4 and CD8 (Figure 1A). Naïve DN1 thymocytes express Notch, Kit and CD44, but as they progress towards mature thymocytes, surface expression of the IL-2Rα chain, CD25, is initiated, at which stage they are called DN2 cells [15]. During the DN2 phase, sustained Notch expression is required for rearrangement of the β-chain locus of the TCR, from the Diversity β (Dβ) to the Joining β (Jβ) region, to begin [16]. As expression of CD44 and Kit are reduced, Vβ to DβJβ rearrangements occur, and cells progress into the DN3 phase [17]. Productive expression of a rearranged β chain paired with a surrogate pre-T cell receptor α chain to form the pre-T cell receptor (pre-TCR) is essential to progression through the DN3 phase [18,19,20]. If cells fail to produce successful rearrangements of the β locus, they do not progress and eventually undergo apoptosis. DN3 thymocytes then lose CD25 expression and progress to the final DN phase, DN4, where the pre-TCR induces ligand-independent dimerisation, causing the cell to begin to proliferate and express both CD4 and CD8 on their cell surface as cells enter the double-positive phase of differentiation (Figure 1B) [17].
Primary and secondary recombination of Vα to Jα gene segments occurs during this double-positive phase, estimated to take an average of five rounds of recombination per allele until positive selection or cell death occurs [21,22,23]. During this double-positive phase, thymocytes undergo positive selection, where ~98% of thymocytes are eliminated [24,25]. Negative selection in the thymic medulla ensures that T cells with high self-pMHC reactivity undergo apoptosis, eliminating risk of autoimmunity [26,27]. Paradoxically, weak self-pMHC recognition is a requirement for development of self-tolerance in the thymic cortex and ultimately T cell survival [28]. T cells with low self-pMHC recognition are rescued, and T cells with no affinity of self-pMHC undergo apoptosis, a process termed positive selection [29]. The remaining ~2% of thymocytes selected as both functionally competent and self-tolerant progress to the final stages of differentiation, downregulating either CD4 or CD8 and becoming single-positive T cells that exit the thymus as naïve T cells (Figure 1C). Naïve T cells migrate to the periphery and subsequently traffic to lymphoid tissues throughout the body.

4. T Cell Entry into the Lymph Nodes

Lymphocyte trafficking and entry into the lymph nodes (LN) is made possible by an extensive network of lymphatic capillaries and vessels that infiltrate tissues within the body [30]. Lymphocytes contained in the lymph drain from interstitial spaces into the afferent lymphatic vessels (LVs), where their migration into the draining LN is mediated by interactions of C-C motif chemokine ligand 21 (CCL21) expression on vessels and C-C chemokine receptor type 7 (CCR7) expressed on T cells [31,32]. Lymphatic endothelial cells that line the LVs secrete sphingosine 1-phosphate (S1P), which contributes to T cell LN migration via S1P receptor 1 binding [33,34,35]. In contrast, lymphocytes in the blood drain into LNs via high endothelial venules (HEVs), a process initiated by CD62L (L-selectin), which facilitates rolling and tethering of lymphocytes to the walls of venules [36]. Further, migration of naïve T cells through HEVs is mediated by a complex adhesion cascade facilitated by interaction between CCL21 and CCR7 [37]. Once inside the LN, the fibroblastic reticular cell (FRC) network creates a scaffold-like architecture and a CCL21 gradient that influences dendritic cells (DCs) and T cells intranodal positioning and migration through the LN [38,39].

5. Tfh Cell Migration through the Lymph Nodes

The structural components and major cell migration pathways are schematically summarised in Figure 2. Lymph and its cellular components traffic through the subcapsular sinus and medullary sinuses of lymph nodes. Here, T cells survey subcapsular macrophages and dendritic cells in the T cell zone for cognate antigen (Figure 2), which, if found, will promote entry into the LN parenchyma [40]. Solubility and size of an antigen can impact the stimulation process as different size/forms of antigen, such as soluble or particulate, are directed to different microanatomical sites within the LN (reviewed in [41]). For example, soluble antigens are taken up and sequestered by lymphatic endothelial cells. Antigens of low molecular weight are channelled via the conduit system surrounded by FRCs, ultimately delivering antigens to follicular DCs in the B cell follicle, allowing cognate B cell sampling of antigen [42,43]. Immune complexes and particles traverse the subcapsular sinus via help from subcapsular macrophages, where they then interact with FDCs and B cells [40,44]. If a cognate antigen is not identified via TCR–pMHC-II interactions, T cells will transmigrate to the medullary sinuses and exit the LN via the efferent LVs [45]. The innermost section of the LN, termed the medulla, is the site where the positioning of DCs determines interaction with antigen and subsequent contact with T cells [46]. Once T cells have encountered antigen, migration towards the paracortex or the T cell zone begins, and CCR7 expression is downregulated, accompanied by C-X-C chemokine receptor 5 (CXCR5) upregulation (Figure 2) [47]. This promotes homing to the B cell follicle of the LN cortex, formation of the GC and differentiation of Tfh and GC B cells. Following stimulation, memory cells formed during the GC reaction home to particular sites within the lymph node, where they await reactivation. One such population is memory Tfh cells, which preferentially migrate to the T–B cell border in the LN cortex via upregulation of CXCR5 [48]. It is here that, upon reactivation, these long-lived memory Tfh cells provide help to B cells, and a subset further differentiate into effector Tfh cells by Bcl6 upregulation [48]. Importantly, the lymphatic flow of the subcapsular sinus plays an essential role in secondary immune responses as an efficient transport mechanism for follicular memory T cells to migrate out of the GC and survey APCs, enabling proliferation and necessary rapid responses to antigen re-encounter [49].

6. T Cell Egress from the Lymph Node

Similar to the role of S1P in the recruitment and trafficking of cells into the LN, a S1P gradient created by low concentrations in the LN and high concentrations in the lymph causes the transmigration of T cells into the lymphatic sinuses and exit via the efferent LVs [50]. The major homeostatic cellular component of efferent lymph is CD4+ T cells; however, antigen presence and stimulation have been shown to influence efferent lymph composition. Following antigen stimulation, the LN will undergo three transitory states until returning to homeostasis. Initially, lymphocyte output is decreased as the LN enters quiescence. This is followed by an intermediary increase in lymphocyte levels to above resting levels in the lymph and finally an ultimate reduction in lymphocyte output to homeostatic levels [51].

7. Stages of Tfh Cell Differentiation

Tfh cell differentiation is a multistage and complex process involving many signalling pathways [52]. Unlike the differentiation pathways of other specialised CD4+ T cells, Tfh cell differentiation accommodates significant heterogeneity within the population [53]. Initially, naïve CD4+ T cells are primed by DCs in the T cell area of the LN, resulting in downregulation of CCR7 and increased expression of CXCR5, which is essential for Tfh cell homing to the B cell follicle (Table 1) [54,55,56]. It has been shown that during this early phase of Tfh differentiation, interleukin (IL)-6-mediated induction of the transcription factors signal transducer and activator of transcription (STAT) 1 and STAT3 is required for Bcl6 induction and downregulation of the IL-2Rα, respectively, limiting Th1 differentiation and promoting Tfh differentiation [57,58,59]. IL-21-dependent expression of inducible costimulator ligand (ICOSL) by B cells and its essential interactions with ICOS are well established as regulators of humoral immunity [60]; however, IL-21 can also regulate Tfh differentiation through the transcription factor c-Maf [61]. Regulation of the expression of cytokines, including IL-21 and IL4, that promote B cell and Tfh cell differentiation and proliferation is the major role of c-Maf expressed by Tfh cells [62,63]. Interestingly, dendritic cell production of IL-12 induces sustained expression of ICOS and has been demonstrated to be essential for early expression of, and closely correlated to, levels of CXCR5 expression [64,65,66]. Upregulation of CXCR5 is accompanied by downregulation of CCR7 and the transcription factor Blimp-1, mediated by upregulation of KLF2 [67,68]. Also critical to cell fate and function is higher TCR affinity, resulting in longer TCR–MHC-II dwell time [69,70,71]. Immature Tfh cells subsequently follow a gradient of CXCL13, the chemokine ligand for CXCR5, to migrate towards the T–B cell border [72,73]. This is where they interact with B cells and commit to the Tfh lineage.
The later phases of Tfh differentiation highlight the symbiotic relationship between Tfh cells and B cells, which are essential to the survival of maturing Tfh cells, providing additional signals aiding in the maintenance of Bcl6 expression and their development into functional B cell helpers. The T–B cell border within the LN is an important site where signalling between signalling lymphocytic activation molecule (SLAM) family receptors expressed on T and B cells occurs [74]. Interactions between SLAM expressed on B cells and SLAM-associated protein (SAP) expressed on T cells are necessary for forming stable T–B cell conjugates [75]. Signalling interactions between CD40L expressed by Tfh cells and CD40 expressed on B cells are essential to the formation and maintenance of GCs as well as promoting class switching of antibodies, inhibiting plasma cell differentiation and providing crucial GC B cell survival signals [76,77,78,79]. Therefore, both GC formation and T cell-dependent antibody responses are contingent on these interactions [74,75]. Furthermore, studies have demonstrated that for persistent T cell migration to the T–B cell border, normal Tfh cell development and optimal germinal centre responses as well as engagement of ICOS via ICOSL on follicular bystander B cells is essential [80]. Blockade of ICOSL function ultimately results in Tfh cell and GC formation inhibition [64,81]. This is further supported by findings that signalling via ICOS promotes the expansion of Tfh cell populations and overexpression results in spontaneous Tfh cell and GC development [82,83]. Deenick and colleagues (2010) suggested that Tfh cell development is more heavily influenced by the role of ICOS/ICOSL interactions in the positioning of B cells to provide plentiful sources of antigen rather than co-stimulation [84]. Therefore, ICOS/ICOSL interactions play an important role in T and B cell positioning within the follicle as well as co-stimulation.
Full differentiation of Tfh cells occurs within the GC, the site where their primary role in the development of an immune response is performed [52,85]. GC Tfh cells are characterised by their high expression of CXCR5 and low CCR7, high ICOS, high PD-1 and high IL-21 expression [86]. In addition, SAP is highly expressed on GC Tfh, where it is essential in sustaining long-lasting T–B cell adhesion [87,88]. SAP plays a critical role in preventing robust inhibitory signalling through SLAMF6 by outcompeting Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) binding [89]. When SAP is bound to SLAMF6, adhesion and development of Tfh cell and B cell responses are supported [90,91]. The importance of these interactions is supported by the observations of severely reduced humoral immunity in X-linked lymphoproliferative patients caused by mutations in the gene SH2DLA that encodes SAP [92,93,94].
During the primary immune response, Tfh cells were found to locate to two anatomically distinct compartments of the LN, the follicle mantle (FM) and the GC, within the cortex [95]. FM Tfh and GC Tfh were found to not only be spatially separated but also represented molecularly distinct subpopulations with little migratory crossover [95]. GC Tfh cells expressed higher levels of genes associated with Tfh cell differentiation and proliferation and B cell class switching [95]. FM Tfh cells expressed high amounts of genes associated with temporospatial guidance, cell adhesion and immune regulation [95]. Interestingly, the GC has been described as an open structure in secondary immune responses [86], where migration of Tfh cells between neighbouring GCs and the FM demonstrated a heterogeneous distribution of these subpopulations and therefore greater diversity of Tfh cell help [95], hypothesised to ultimately improve recall responses. Finally, the migration of Tfh into the subcapsular sinus to survey APCs provides an opportunity for antigen-experienced Tfh to egress from the LN and enter circulation, contributing to the c-Tfh cell population.

8. The GC Response and Tfh Cell Function in the Immune Response

Effective humoral immunity is often mediated by sterilising or broadly neutralising antibodies (bAbs), which are produced by memory B cells during the germinal centre reaction [97,98]. The GC forms when antigen is presented by DCs, promoting differentiation and expansion of Tfh cells. GCs are also the site where activated B cells capture and process antigen for presentation on MHC class II complexes [99]. After Tfh cells recognise cognate peptide, further CD4+ T cell differentiation into Tfh cells and B cell differentiation is re-enforced and promoted [100]. Once these initial T–B cell interactions occur, B cells will either differentiate into short-lived antibody-secreting cells (ASCs), or they will enter the GC reaction and undergo further rounds of selection, differentiation and proliferation [97] (Figure 2).
The GC comprises two functionally distinct compartments (Figure 2): the light zone (LZ) and the dark zone (DZ). In the DZ, B cells undergo multiple iterations of proliferation and somatic hypermutation to produce a heterogeneous B cell population with diverse B cell receptor (BCR) sequences [101]. B cells then exit the DZ and migrate into the LZ, where they compete for antigen bound to the surface of DCs [102,103]. Here, Tfh cells selectively provide help to B cells with high-affinity BCRs due to their ability to internalise and therefore present more antigen to Tfh cells [104,105,106]. After interacting with Tfh cells in the LZ, B cells have three potential fates: (1) differentiate into memory B cells and exit the GC [107], (2) differentiate into long-lived plasma cells and thus exit the GC [108], or (3) re-enter the DZ for further rounds of somatic hypermutation and selection [109]. Many studies have reported this bidirectional movement of B cells between LZ and DZ within the GC [110,111] and suggest that the strength of the interaction between Tfh cells and B cells directly determines B cell fate [97,112]. Interestingly, one study has reported that the proportion of Tfh cell help provided to GC B cells directly translates to the degree of mutations in the B cell receptor, and thus the number of cell divisions, that a given GC B cell will undergo in a single round of selection [113]. Therefore, the GC reaction, preferential support of high-affinity B cells and subsequent production of diverse B cell repertoires are all dependent on help from Tfh cells, although perhaps not to an equal degree as antigen, ultimately impacting on the quality of the immune response.

9. Utilisation of c-Tfh Cells to Study Disease States

Tfh cells are empirically defined by their ability to migrate into the GC of secondary lymphoid tissue and the aid they provide to B cells within these anatomically protected sites during the immune response. This establishes an inherent, yet significant, obstacle to studying their role in host responses to infection, vaccination and autoimmunity. Coupled with confounding ethical concerns with accessing healthy human lymph nodes, many studies have utilised animal models, human tonsillar tissue or a circulating, phenotypically similar counterpart, c-Tfh, to highlight the importance of Tfh cell assistance and functioning in the generation of effective immunity. Several studies have utilised CD4+ CXCR5+ ICOS+ c-Tfh as a surrogate biomarker of humoral immunity under the hypothesis that these cells are memory Tfh that egress from the LN following antigen encounter. This hypothesis has been supported by the detection of tetanus-toxoid-specific c-Tfh cells in peripheral blood even years after receiving booster vaccination [114]. Importantly, more recent studies have established the clonal relationship between GC Tfh and their memory c-Tfh counterparts by TCR repertoire sequencing of matched human blood and tonsillar samples [115,116]. Perhaps the most definitive evidence that GC Tfh exit the LN and enter the periphery to form c-Tfh was provided by an interesting study that demonstrated efferent lymph collected from the thoracic duct in humans was enriched for Tfh cells, and treatment with the S1PR1 modulator, fingolimod (FTY720), prevented LN egress and reduced c-Tfh cell numbers and frequency in peripheral blood [117].
Aberrant Tfh cell development and functioning can drive autoimmunity and have been linked with inadequate immune responses [118,119]. However, due to the difficulty accessing bona fide GC Tfh, the use of c-Tfh as a surrogate biomarker has become standard across studies of autoimmunity, immunodeficiencies, allergy and malignancies. In the case of systemic lupus erythematosus (SLE), increases in the frequency of activated c-Tfh positively correlated with serum autoantibody titres and disease severity [120,121,122,123]. Furthermore, elegant studies performed in mice demonstrated that excessive Tfh cell responses and subsequent GC formation were sufficient to cause lupus-like autoimmunity [124,125].
Tfh cell dysregulation leading to impaired B cell responses and ultimately humoral immune responses have also been reported in human immunodeficiency virus/simian immunodeficiency virus (HIV/SIV) [126]. Studies have shown that expanded Tfh cell populations harbour higher levels of HIV or SIV DNA compared to other CD4+ T cell subsets, can effectively support productive HIV infection and form a major viral reservoir [127,128,129]. Additionally, expansion of HIV-specific GC Tfh cells and c-Tfh cells have been observed in chronic infection and are associated with increased IL-21 secretion and polyclonal hypergammaglobulinemia [130]. HIV-specific c-Tfh demonstrated a Th-1-like phenotype and functionality, which had a significant positive association with the size of the translation-competent viral reservoir [131]. Interestingly, Th-1-like c-Tfh have been shown to largely lack the ability to help naïve or memory B cells [132]. Conversely, IL-21-producing c-Tfh were observed to have greater helper capacity to induce B cell maturation and class switching in HIV elite controllers and were associated with antigen-specific B cells in HIV progressors [133]. However, significant impairment of c-Tfh cell functioning in chronic HIV infection leads to reduced B cell responses, a phenomenon mirrored in secondary lymphoid organs [134,135].
The presence of Tfh is required for IgE production and development of an allergic response [136,137]. Increased numbers of c-Tfh were observed in atopic dermatitis patients, which correlated with activated memory B cells, IgE production and disease severity [138,139]. In allergic rhinitis and asthma, levels of c-Tfh cells with a Tfh2 phenotype are also elevated, correlating with total IgE levels and therefore contributing to a pro-inflammatory milieu in the latter [140]. Further supporting the role of Tfh in allergy, mice with a mutated IL-6R were unable to expand Tfh cell populations, resulting in significantly reduced IgG1 and IgE responses to house dust mite sensitisation when compared to wild-type mice [141]. Therefore, it is evident that the accumulation of c-Tfh has been linked to increased IgE production and proallergic function. Interestingly, these results demonstrate that Tfh dysregulation can lead to impaired B cell responses and humoral immunity across many pathological modalities.

10. Role of Tfh Cells in Vaccination

Multiple studies have demonstrated the emergence of a transient population of PD-1+ICOS+ c-Tfh cells following vaccination with inactivated influenza vaccine [142,143,144,145]. Inactivated influenza vaccine-induced c-Tfh cells were phenotypically similar to their GC counterparts, expressing various activation markers, including PD-1, CD38, ICOS and Ki67 [143,144,146]. Levels of c-Tfh cells correlated with increased Ab serum titres and presence of ASCs and corresponded to peak GC responses in mice and plasmablast response in humans [142,143,144,145,147]. Subsequent studies also demonstrated a positive correlation between activated c-Tfh and an increased affinity of inactivated influenza vaccine-induced Abs and magnitude of memory B cell responses [145,148]. c-Tfh occurrence correlated with active Tfh cell differentiation in secondary lymphoid organs of mice [147], which was mirrored in a vaccination study with an MF59-adjuvanted inactivated H5N1 vaccine [149] and supported by observations of an association between diminished c-Tfh and reduction in vaccine-specific Ab levels in serum of older people in response to vaccination [150]. Secondary immunisation elicited a predominantly plasmablast response, with the induction of few secondary GC responses in mice [151] and in rhesus macaques, although this is potentially attributable to the short interval between immunisations [152]. Further, rhesus macaques that produced higher levels of neutralising Abs had larger GC responses, and antigen-specific CD4+ T cells isolated from peripheral blood were enriched for Tfh-associated genes, including IL-21, ICOS and CD40L [152]. In a subsequent study, GC B cell and antigen-specific CD4+ T cell numbers in the draining LN increased significantly by day 7 post primary vaccination, and this response was 10-fold larger when immunised subcutaneously compared to intramuscularly [153]. However, there was no significant difference in serum IgG titres between the two immunisation protocols, suggesting the magnitude of response may not be directly proportionate to quality of response.
Two critical studies in Tfh cell immunology were published in 2017 that utilised TCR β chain repertoire sequencing to characterise the c-Tfh response to vaccination and establish the clonal relationship to GC Tfh [116,143]. The initial clonal-activated c-Tfh cell response correlated with plasmablast responses and vaccine-specific serum IgG. This was observed following successive seasonal influenza vaccinations and was clonally related to tonsillar GC Tfh [116,143]. Interestingly, between vaccinations, these c-Tfh clones were found in the “inactivated” c-Tfh repertoire and were seen to expand into activated c-Tfh upon antigen re-encounter [143]. These observations were mirrored in an antigen-specific c-Tfh population [116]. A subsequent study observed a transcriptional and clonal relationship between c-Tfh and lymph node GC Tfh [154]. Interestingly, they demonstrated that the combination adjuvant glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE) promoted the emergence of public TCR β sequences as well as long-lived Ab responses [154]. Further to establishing that GC Tfh and c-Tfh are clonally convergent, it has been shown that although circulating non-Tfh cells share few TCR β clones with GC Tfh, they are clonally distinct from c-Tfh cells [115]. A population of Tfh cells isolated from human thoracic duct lymph were shown to be phenotypically and transcriptionally similar to GC Tfh [117]. Utilisation of next-generation sequencing by these recent studies have allowed critical analysis of c-Tfh cell kinetics in response to vaccination but also validation of the hypothesis that c-Tfh cells represent a circulating memory pool critical in aiding B cells during immune responses.

11. The Future Role of Systems Biology Approaches in Characterising Tfh Cells

Next-generation sequencing technologies have played an increasingly important role in understanding Tfh cell biology in recent years [115,116,117,143,154] by enabling researchers to obtain data sets that are sufficiently representative of immune cell populations. Moreover, the recent progress that has been made using such technologies to characterise other immune cell populations and their responses to infection and vaccination indicate strong potential for gaining similar insights for Tfh cells from future studies.
High-throughput sequencing technologies allow for the large-scale “bulk” sequencing required to capture data for large and/or highly diverse populations of cells. Investigations of the enormously diverse immune receptor repertoires of T cells and B cells have flourished with this technology, yielding valuable immunological insights. For example, the immune receptor repertoires of cells involved in responses to infection or vaccination to pathogens such as Cytomegalovirus (CMV), Epstein-Barr Virus (EBV), Tuberculosis (TB) and influenza have been well characterised, enabling the identification of immune receptor “signatures” in T cells [155,156,157] and B cells [158]. Furthermore, it has been demonstrated that such features may allow prediction of the antigen specificity of cells using machine learning approaches [155,156,157]. Bulk TCR repertoire sequencing has also revealed broad heterogeneity of CD4+ T cell functionality in response to microbial infection and vaccination [159], and transcriptomics have demonstrated that T cells defined as the same clone by their TCR sequence can represent phenotypically distinct subsets [160]. Another application of TCR deep sequencing is as a predictor of disease outcome or state (reviewed in [161]); for example, TCRβ oligoclonality is linked to clinical relapse in juvenile idiopathic arthritis [162], and EBV-cross-reactive TCR clonotypes are enriched in cerebrospinal fluid from multiple sclerosis patients [163].
Single-cell sequencing technologies have also experienced rapid development and growth in application to immunological studies, with increasing capability to generate multi-omic data by measuring the proteome, genome, transcriptome, methylome and spatial expression of single cells [164]. Single-cell RNA sequencing (scRNA-seq) techniques have, to date, been the most popular and have been deployed to analyse the network of cell types, the cytokines and chemokines they produce, their transcriptomic profiles and their involvement in complex immune responses [165]. Explorations of immune responses to vaccination have provided valuable insights by assessing for proteomic and transcriptomic predictors of improved vaccination response. Notably, researchers have identified certain gene expression profiles, including the B cell growth factor tumour necrosis factor receptor superfamily member 17 (TNFRS17) and eukaryotic translation factor 2 alpha kinase 4, that respectively predicted neutralising antibody titres and antigen-specific CD8 T cell responses to yellow fever vaccination [166]. Similar utilisation of systems biology has been applied to groups of interest, such as older adults, where one study validated the identification of genes, including CAMK4, previously associated with influenza vaccine responses [167]. New genes of interest that were not previously associated with responses to influenza vaccination and previously uncharacterised genes were also linked to vaccine-induced immunity [167]. Gene signatures identified as possible regulators of vaccine-induced immunity have been described across successive years and multiple cell populations post influenza vaccination [168,169,170], measles vaccination [171] and meningococcal vaccination [172]. The use of single-cell RNA-seq enabled identification of molecular signatures that underpin relationships between Th1 and Tfh cells and showed each cell population differentiated from a highly proliferative single precursor cell during malaria infection in mice [173].
Despite the incredible contribution of next-generation sequencing techniques to the understanding of immune responses to infection and vaccination, the discovery of new relationships between cell types and the identification of correlates of protection, there are a number of challenges associated with these technologies [165]. Thus, ongoing development of innovative experimental technologies is focussed not only on increasing throughput and resolution and decreasing costs but also on improving data quality and reproducibility. Also crucial to addressing these challenges is the development of novel computational methods to process, analyse and interpret experimental data. Computational methods utilising machine learning and artificial intelligence approaches is another rapid-growth research area with enormous implications for improving predictive power and developing integrated models of immune responses to infection. Such studies rely on application of these methods to large-scale data sets and thus greatly benefit from recent growth in the number and size of data repositories that facilitate sharing of data. Collectively, these technological advances, coupled with increasing research resources, are allowing researchers to pursue systems biology approaches that are expected to lead to a better understanding of not only Tfh cells but the complex immune system as a whole and how it may best be manipulated to achieve positive health outcomes.

12. Concluding Remarks

Over the past decade, great efforts to characterise the roles and responsibilities of Tfh cells in immunity and infection have yielded interesting and powerful insights, establishing them as key players in immune responses to vaccination in both secondary lymphoid organs and in the periphery and as potential correlates of protection. Despite these significant advances in understanding of the role of Tfh cells in health and immunity, particularly in the field of vaccinology, there are still areas to be explored. Validation of the clonal relationship present between the two hugely heterogeneous populations of cTfh and secondary lymphoid organ (SLO)-derived Tfh is essential to the utilisation of cTfh as a surrogate biomarker in future studies. Could the adoption of scRNA-seq technologies provide powerful insight into the transcriptional factors and signalling pathways that regulate T cell egress from the LN and contribute to our understanding of the relationship between LN Tfh and their circulating counterparts? Ultimately, validation of a clonal relationship will address significant ethical and practical obstacles in accessing and studying SLO-derived Tfh. Given the substantial heterogeneity within the Tfh population, is it possible to identify subpopulations with greater helper functionality or certain gene expression profiles that could be leveraged to improve immune responses to vaccination? Are the Tfh cell responses studied using vaccination as a model truly indicative of their role during the course of natural infection? Finally, there remains a unique opportunity to harness emerging tools to characterise the Tfh cell response to vaccination on a transcriptomic and proteomic level. The ability to identify gene signatures associated with improved vaccine-induced immunity in Tfh cells could be a very powerful tool in rational vaccine design.

Author Contributions

All authors have made significant and intellectual contribution to the review and approve it for publication. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a UNSW RTP Scholarship (RSAP1000) (H.L.), NHMRC Career Development Fellowship 1067590 (V.V.) and NHMRC Program grant 1149990 (A.K. and C.M.M.).

Acknowledgments

We would like to thank Trevor Dougherty for his outstanding administrative support.

Conflicts of Interest

The authors have no conflicts of interest to disclose.

References

  1. Centers for Disease Control and Prevention. Control of infectious diseases. MMWR Morb. Mortal Wkly. Rep. 1999, 48, 621–629. [Google Scholar]
  2. Sarkander, J.; Hojyo, S.; Tokoyoda, K. Vaccination to gain humoral immune memory. Clin. Transl. Immunology 2016, 5, e120. [Google Scholar] [CrossRef]
  3. Chen, H.; Ndhlovu, Z.M.; Liu, D.; Porter, L.C.; Fang, J.W.; Darko, S.; Brockman, M.A.; Miura, T.; Brumme, Z.L.; Schneidewind, A.; et al. TCR clonotypes modulate the protective effect of HLA class I molecules in HIV-1 infection. Nat. Immunol. 2012, 13, 691–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Davenport, M.P.; Price, D.A.; McMichael, A.J. The T cell repertoire in infection and vaccination: Implications for control of persistent viruses. Curr. Opin. Immunol. 2007, 19, 294–300. [Google Scholar] [CrossRef] [PubMed]
  5. Piepenbrink, K.H.; Blevins, S.J.; Scott, D.R.; Baker, B.M. The basis for limited specificity and MHC restriction in a T cell receptor interface. Nat. Commun. 2013, 4, 1948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Vrisekoop, N.; Monteiro, J.P.; Mandl, J.N.; Germain, R.N. Revisiting thymic positive selection and the mature T cell repertoire for antigen. Immunity 2014, 41, 181–190. [Google Scholar] [CrossRef] [Green Version]
  7. Rudolph, M.G.; Stanfield, R.L.; Wilson, I.A. How TCRs bind MHCs, peptides, and coreceptors. Annu. Rev. Immunol. 2006, 24, 419–466. [Google Scholar] [CrossRef]
  8. Freeman, J.D.; Warren, R.L.; Webb, J.R.; Nelson, B.H.; Holt, R.A. Profiling the T-cell receptor beta-chain repertoire by massively parallel sequencing. Genome Res. 2009, 19, 1817–1824. [Google Scholar] [CrossRef] [Green Version]
  9. Borg, N.A.; Ely, L.K.; Beddoe, T.; Macdonald, W.A.; Reid, H.H.; Clements, C.S.; Purcell, A.W.; Kjer-Nielsen, L.; Miles, J.J.; Burrows, S.R.; et al. The CDR3 regions of an immunodominant T cell receptor dictate the ‘energetic landscape’ of peptide-MHC recognition. Nat. Immunol. 2005, 6, 171–180. [Google Scholar] [CrossRef]
  10. Davis, M.M.; Bjorkman, P.J. T-cell antigen receptor genes and T-cell recognition. Nature 1988, 334, 395–402. [Google Scholar] [CrossRef]
  11. Lieber, M.R. Site-specific recombination in the immune system. FASEB J. 1991, 5, 2934–2944. [Google Scholar] [CrossRef] [PubMed]
  12. Arstila, T.P.; Casrouge, A.; Baron, V.; Even, J.; Kanellopoulos, J.; Kourilsky, P. A direct estimate of the human alphabeta T cell receptor diversity. Science 1999, 286, 958–961. [Google Scholar] [CrossRef]
  13. Vanhanen, R.; Heikkila, N.; Aggarwal, K.; Hamm, D.; Tarkkila, H.; Patila, T.; Jokiranta, T.S.; Saramaki, J.; Arstila, T.P. T cell receptor diversity in the human thymus. Mol. Immunol. 2016, 76, 116–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sewell, A.K. Why must T cells be cross-reactive? Nat. Rev. Immunol. 2012, 12, 669–677. [Google Scholar] [CrossRef] [PubMed]
  15. Ciofani, M.; Knowles, G.C.; Wiest, D.L.; von Boehmer, H.; Zuniga-Pflucker, J.C. Stage-specific and differential notch dependency at the alphabeta and gammadelta T lineage bifurcation. Immunity 2006, 25, 105–116. [Google Scholar] [CrossRef] [Green Version]
  16. Wolfer, A.; Wilson, A.; Nemir, M.; MacDonald, H.R.; Radtke, F. Inactivation of Notch1 impairs VDJbeta rearrangement and allows pre-TCR-independent survival of early alpha beta Lineage Thymocytes. Immunity 2002, 16, 869–879. [Google Scholar] [CrossRef] [Green Version]
  17. Krangel, M.S. Mechanics of T cell receptor gene rearrangement. Curr. Opin. Immunol. 2009, 21, 133–139. [Google Scholar] [CrossRef] [Green Version]
  18. Dudley, E.C.; Petrie, H.T.; Shah, L.M.; Owen, M.J.; Hayday, A.C. T cell receptor beta chain gene rearrangement and selection during thymocyte development in adult mice. Immunity 1994, 1, 83–93. [Google Scholar] [CrossRef]
  19. Mallick, C.A.; Dudley, E.C.; Viney, J.L.; Owen, M.J.; Hayday, A.C. Rearrangement and diversity of T cell receptor beta chain genes in thymocytes: A critical role for the beta chain in development. Cell 1993, 73, 513–519. [Google Scholar] [CrossRef]
  20. Capone, M.; Hockett, R.D., Jr.; Zlotnik, A. Kinetics of T cell receptor beta, gamma, and delta rearrangements during adult thymic development: T cell receptor rearrangements are present in CD44(+)CD25(+) Pro-T thymocytes. Proc. Natl. Acad. Sci. USA 1998, 95, 12522–12527. [Google Scholar] [CrossRef] [Green Version]
  21. Warmflash, A.; Dinner, A.R. A model for TCR gene segment use. J. Immunol. 2006, 177, 3857–3864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wang, F.; Huang, C.Y.; Kanagawa, O. Rapid deletion of rearranged T cell antigen receptor (TCR) Valpha-Jalpha segment by secondary rearrangement in the thymus: Role of continuous rearrangement of TCR alpha chain gene and positive selection in the T cell repertoire formation. Proc. Natl. Acad. Sci. USA 1998, 95, 11834–11839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Guo, J.; Hawwari, A.; Li, H.; Sun, Z.; Mahanta, S.K.; Littman, D.R.; Krangel, M.S.; He, Y.W. Regulation of the TCRalpha repertoire by the survival window of CD4(+)CD8(+) thymocytes. Nat. Immunol. 2002, 3, 469–476. [Google Scholar] [CrossRef] [PubMed]
  24. Shortman, K.; Egerton, M.; Spangrude, G.J.; Scollay, R. The generation and fate of thymocytes. Semin. Immunol. 1990, 2, 3–12. [Google Scholar]
  25. Surh, C.D.; Sprent, J. T-cell apoptosis detected in situ during positive and negative selection in the thymus. Nature 1994, 372, 100–103. [Google Scholar] [CrossRef]
  26. Palmer, E. Negative selection—Clearing out the bad apples from the T-cell repertoire. Nat. Rev. Immunol. 2003, 3, 383–391. [Google Scholar] [CrossRef]
  27. Daley, S.R.; Hu, D.Y.; Goodnow, C.C. Helios marks strongly autoreactive CD4+ T cells in two major waves of thymic deletion distinguished by induction of PD-1 or NF-kappaB. J. Exp. Med. 2013, 210, 269–285. [Google Scholar] [CrossRef] [Green Version]
  28. Kondo, K.; Ohigashi, I.; Takahama, Y. Thymus machinery for T-cell selection. Int. Immunol. 2019, 31, 119–125. [Google Scholar] [CrossRef]
  29. Klein, L.; Kyewski, B.; Allen, P.M.; Hogquist, K.A. Positive and negative selection of the T cell repertoire: What thymocytes see (and don’t see). Nat. Rev. Immunol. 2014, 14, 377–391. [Google Scholar] [CrossRef] [Green Version]
  30. Cueni, L.N.; Detmar, M. The lymphatic system in health and disease. Lymphat. Res. Biol. 2008, 6, 109–122. [Google Scholar] [CrossRef]
  31. Forster, R.; Braun, A.; Worbs, T. Lymph node homing of T cells and dendritic cells via afferent lymphatics. Trends Immunol. 2012, 33, 271–280. [Google Scholar] [CrossRef] [PubMed]
  32. Vaahtomeri, K.; Brown, M.; Hauschild, R.; De Vries, I.; Leithner, A.F.; Mehling, M.; Kaufmann, W.A.; Sixt, M. Locally Triggered Release of the Chemokine CCL21 Promotes Dendritic Cell Transmigration across Lymphatic Endothelia. Cell Rep. 2017, 19, 902–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Fang, V.; Chaluvadi, V.S.; Ramos-Perez, W.D.; Mendoza, A.; Baeyens, A.; Rivera, R.; Chun, J.; Cammer, M.; Schwab, S.R. Gradients of the signaling lipid S1P in lymph nodes position natural killer cells and regulate their interferon-gamma response. Nat. Immunol. 2017, 18, 15–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Vigl, B.; Aebischer, D.; Nitschke, M.; Iolyeva, M.; Rothlin, T.; Antsiferova, O.; Halin, C. Tissue inflammation modulates gene expression of lymphatic endothelial cells and dendritic cell migration in a stimulus-dependent manner. Blood 2011, 118, 205–215. [Google Scholar] [CrossRef]
  35. Mendoza, A.; Fang, V.; Chen, C.; Serasinghe, M.; Verma, A.; Muller, J.; Chaluvadi, V.S.; Dustin, M.L.; Hla, T.; Elemento, O.; et al. Lymphatic endothelial S1P promotes mitochondrial function and survival in naive T cells. Nature 2017, 546, 158–161. [Google Scholar] [CrossRef]
  36. Rosen, S.D. Ligands for L-selectin: Homing, inflammation, and beyond. Annu. Rev. Immunol. 2004, 22, 129–156. [Google Scholar] [CrossRef]
  37. Girard, J.P.; Moussion, C.; Forster, R. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat. Rev. Immunol. 2012, 12, 762–773. [Google Scholar] [CrossRef]
  38. Novkovic, M.; Onder, L.; Cupovic, J.; Abe, J.; Bomze, D.; Cremasco, V.; Scandella, E.; Stein, J.V.; Bocharov, G.; Turley, S.J.; et al. Topological Small-World Organization of the Fibroblastic Reticular Cell Network Determines Lymph Node Functionality. PLoS Biol. 2016, 14, e1002515. [Google Scholar] [CrossRef] [Green Version]
  39. Braun, A.; Worbs, T.; Moschovakis, G.L.; Halle, S.; Hoffmann, K.; Bolter, J.; Munk, A.; Forster, R. Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration. Nat. Immunol. 2011, 12, 879–887. [Google Scholar] [CrossRef]
  40. Phan, T.G.; Green, J.A.; Gray, E.E.; Xu, Y.; Cyster, J.G. Immune complex relay by subcapsular sinus macrophages and noncognate B cells drives antibody affinity maturation. Nat. Immunol. 2009, 10, 786–793. [Google Scholar] [CrossRef]
  41. Hirosue, S.; Dubrot, J. Modes of Antigen Presentation by Lymph Node Stromal Cells and Their Immunological Implications. Front. Immunol. 2015, 6, 446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Harwood, N.E.; Batista, F.D. The antigen expressway: Follicular conduits carry antigen to B cells. Immunity 2009, 30, 177–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Sixt, M.; Kanazawa, N.; Selg, M.; Samson, T.; Roos, G.; Reinhardt, D.P.; Pabst, R.; Lutz, M.B.; Sorokin, L. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 2005, 22, 19–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Phan, T.G.; Grigorova, I.; Okada, T.; Cyster, J.G. Subcapsular encounter and complement-dependent transport of immune complexes by lymph node B cells. Nat. Immunol. 2007, 8, 992–1000. [Google Scholar] [CrossRef] [PubMed]
  45. Tomura, M.; Yoshida, N.; Tanaka, J.; Karasawa, S.; Miwa, Y.; Miyawaki, A.; Kanagawa, O. Monitoring cellular movement in vivo with photoconvertible fluorescence protein “Kaede” transgenic mice. Proc. Natl. Acad. Sci. USA 2008, 105, 10871–10876. [Google Scholar] [CrossRef] [Green Version]
  46. Gerner, M.Y.; Casey, K.A.; Kastenmuller, W.; Germain, R.N. Dendritic cell and antigen dispersal landscapes regulate T cell immunity. J. Exp. Med. 2017, 214, 3105–3122. [Google Scholar] [CrossRef] [Green Version]
  47. Fazilleau, N.; Eisenbraun, M.D.; Malherbe, L.; Ebright, J.N.; Pogue-Caley, R.R.; McHeyzer-Williams, L.J.; McHeyzer-Williams, M.G. Lymphoid reservoirs of antigen-specific memory T helper cells. Nat. Immunol. 2007, 8, 753–761. [Google Scholar] [CrossRef]
  48. Ise, W.; Inoue, T.; McLachlan, J.B.; Kometani, K.; Kubo, M.; Okada, T.; Kurosaki, T. Memory B cells contribute to rapid Bcl6 expression by memory follicular helper T cells. Proc. Natl. Acad. Sci. USA 2014, 111, 11792–11797. [Google Scholar] [CrossRef] [Green Version]
  49. Gerner, M.Y.; Torabi-Parizi, P.; Germain, R.N. Strategically localized dendritic cells promote rapid T cell responses to lymph-borne particulate antigens. Immunity 2015, 42, 172–185. [Google Scholar] [CrossRef] [Green Version]
  50. Cyster, J.G.; Schwab, S.R. Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs. Annu Rev. Immunol. 2012, 30, 69–94. [Google Scholar] [CrossRef]
  51. Hunter, M.C.; Teijeira, A.; Halin, C. T Cell Trafficking through Lymphatic Vessels. Front. Immunol. 2016, 7, 613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Crotty, S. T follicular helper cell differentiation, function, and roles in disease. Immunity 2014, 41, 529–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Trub, M.; Barr, T.A.; Morrison, V.L.; Brown, S.; Caserta, S.; Rixon, J.; Ivens, A.; Gray, D. Heterogeneity of Phenotype and Function Reflects the Multistage Development of T Follicular Helper Cells. Front. Immunol. 2017, 8, 489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Goenka, R.; Barnett, L.G.; Silver, J.S.; O’Neill, P.J.; Hunter, C.A.; Cancro, M.P.; Laufer, T.M. Cutting edge: Dendritic cell-restricted antigen presentation initiates the follicular helper T cell program but cannot complete ultimate effector differentiation. J. Immunol. 2011, 187, 1091–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Schaerli, P.; Willimann, K.; Lang, A.B.; 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, 1553–1562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Hardtke, S.; Ohl, L.; Forster, R. Balanced expression of CXCR5 and CCR7 on follicular T helper cells determines their transient positioning to lymph node follicles and is essential for efficient B-cell help. Blood 2005, 106, 1924–1931. [Google Scholar] [CrossRef] [Green Version]
  57. Choi, Y.S.; Eto, D.; Yang, J.A.; Lao, C.; Crotty, S. Cutting edge: STAT1 is required for IL-6-mediated Bcl6 induction for early follicular helper cell differentiation. J. Immunol. 2013, 190, 3049–3053. [Google Scholar] [CrossRef] [Green Version]
  58. Johnston, R.J.; Poholek, A.C.; DiToro, D.; Yusuf, I.; Eto, D.; Barnett, B.; Dent, A.L.; Craft, J.; Crotty, S. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science 2009, 325, 1006–1010. [Google Scholar] [CrossRef] [Green Version]
  59. Hatzi, K.; Nance, J.P.; Kroenke, M.A.; Bothwell, M.; Haddad, E.K.; Melnick, A.; Crotty, S. BCL6 orchestrates Tfh cell differentiation via multiple distinct mechanisms. J. Exp. Med. 2015, 212, 539–553. [Google Scholar] [CrossRef] [Green Version]
  60. Nurieva, R.I.; Chung, Y.; Hwang, D.; Yang, X.O.; Kang, H.S.; Ma, L.; Wang, Y.H.; Watowich, S.S.; Jetten, A.M.; Tian, Q.; et al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity 2008, 29, 138–149. [Google Scholar] [CrossRef] [Green Version]
  61. Hiramatsu, Y.; Suto, A.; Kashiwakuma, D.; Kanari, H.; Kagami, S.; Ikeda, K.; Hirose, K.; Watanabe, N.; Grusby, M.J.; Iwamoto, I.; et al. c-Maf activates the promoter and enhancer of the IL-21 gene, and TGF-beta inhibits c-Maf-induced IL-21 production in CD4+ T cells. J. Leukoc. Biol. 2010, 87, 703–712. [Google Scholar] [CrossRef] [PubMed]
  62. Kroenke, M.A.; Eto, D.; Locci, M.; Cho, M.; Davidson, T.; Haddad, E.K.; Crotty, S. Bcl6 and Maf cooperate to instruct human follicular helper CD4 T cell differentiation. J. Immunol. 2012, 188, 3734–3744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Andris, F.; Denanglaire, S.; Anciaux, M.; Hercor, M.; Hussein, H.; Leo, O. The Transcription Factor c-Maf Promotes the Differentiation of Follicular Helper T Cells. Front. Immunol. 2017, 8, 480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Choi, Y.S.; Kageyama, R.; Eto, D.; Escobar, T.C.; Johnston, R.J.; Monticelli, L.; Lao, C.; Crotty, S. ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity 2011, 34, 932–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ma, C.S.; Suryani, S.; Avery, D.T.; Chan, A.; Nanan, R.; Santner-Nanan, B.; Deenick, E.K.; Tangye, S.G. Early commitment of naive human CD4(+) T cells to the T follicular helper (T(FH)) cell lineage is induced by IL-12. Immunol. Cell Biol. 2009, 87, 590–600. [Google Scholar] [CrossRef] [PubMed]
  66. Schmitt, N.; Morita, R.; Bourdery, L.; Bentebibel, S.E.; Zurawski, S.M.; Banchereau, J.; Ueno, H. Human dendritic cells induce the differentiation of interleukin-21-producing T follicular helper-like cells through interleukin-12. Immunity 2009, 31, 158–169. [Google Scholar] [CrossRef] [Green Version]
  67. Weber, J.P.; Fuhrmann, F.; Feist, R.K.; Lahmann, A.; Al Baz, M.S.; Gentz, L.J.; Vu Van, D.; Mages, H.W.; Haftmann, C.; Riedel, R.; et al. ICOS maintains the T follicular helper cell phenotype by down-regulating Kruppel-like factor 2. J. Exp. Med. 2015, 212, 217–233. [Google Scholar] [CrossRef] [Green Version]
  68. Lee, J.Y.; Skon, C.N.; Lee, Y.J.; Oh, S.; Taylor, J.J.; Malhotra, D.; Jenkins, M.K.; Rosenfeld, M.G.; Hogquist, K.A.; Jameson, S.C. The transcription factor KLF2 restrains CD4(+) T follicular helper cell differentiation. Immunity 2015, 42, 252–264. [Google Scholar] [CrossRef] [Green Version]
  69. Stoll, S.; Delon, J.; Brotz, T.M.; Germain, R.N. Dynamic imaging of T cell-dendritic cell interactions in lymph nodes. Science 2002, 296, 1873–1876. [Google Scholar] [CrossRef]
  70. Celli, S.; Lemaitre, F.; Bousso, P. Real-time manipulation of T cell-dendritic cell interactions in vivo reveals the importance of prolonged contacts for CD4+ T cell activation. Immunity 2007, 27, 625–634. [Google Scholar] [CrossRef] [Green Version]
  71. Fazilleau, N.; McHeyzer-Williams, L.J.; Rosen, H.; McHeyzer-Williams, M.G. The function of follicular helper T cells is regulated by the strength of T cell antigen receptor binding. Nat. Immunol. 2009, 10, 375–384. [Google Scholar] [CrossRef] [PubMed]
  72. Haynes, N.M.; Allen, C.D.; Lesley, R.; Ansel, K.M.; Killeen, N.; Cyster, J.G. Role of CXCR5 and CCR7 in follicular Th cell positioning and appearance of a programmed cell death gene-1high germinal center-associated subpopulation. J. Immunol. 2007, 179, 5099–5108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Chen, X.; Ma, W.; Zhang, T.; Wu, L.; Qi, H. Phenotypic Tfh development promoted by CXCR5-controlled re-localization and IL-6 from radiation-resistant cells. Protein Cell 2015, 6, 825–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kamperschroer, C.; Roberts, D.M.; Zhang, Y.; Weng, N.P.; Swain, S.L. SAP enables T cells to help B cells by a mechanism distinct from Th cell programming or CD40 ligand regulation. J. Immunol. 2008, 181, 3994–4003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Qi, H.; Cannons, J.L.; Klauschen, F.; Schwartzberg, P.L.; Germain, R.N. SAP-controlled T-B cell interactions underlie germinal centre formation. Nature 2008, 455, 764–769. [Google Scholar] [CrossRef]
  76. Foy, T.M.; Laman, J.D.; Ledbetter, J.A.; Aruffo, A.; Claassen, E.; Noelle, R.J. gp39-CD40 interactions are essential for germinal center formation and the development of B cell memory. J. Exp. Med. 1994, 180, 157–163. [Google Scholar] [CrossRef] [Green Version]
  77. Randall, T.D.; Heath, A.W.; Santos-Argumedo, L.; Howard, M.C.; Weissman, I.L.; Lund, F.E. Arrest of B lymphocyte terminal differentiation by CD40 signaling: Mechanism for lack of antibody-secreting cells in germinal centers. Immunity 1998, 8, 733–742. [Google Scholar] [CrossRef] [Green Version]
  78. Choe, J.; Kim, H.S.; Zhang, X.; Armitage, R.J.; Choi, Y.S. Cellular and molecular factors that regulate the differentiation and apoptosis of germinal center B cells. Anti-Ig down-regulates Fas expression of CD40 ligand-stimulated germinal center B cells and inhibits Fas-mediated apoptosis. J. Immunol. 1996, 157, 1006–1016. [Google Scholar]
  79. Watanabe, M.; Fujihara, C.; Radtke, A.J.; Chiang, Y.J.; Bhatia, S.; Germain, R.N.; Hodes, R.J. Co-stimulatory function in primary germinal center responses: CD40 and B7 are required on distinct antigen-presenting cells. J. Exp. Med. 2017, 214, 2795–2810. [Google Scholar] [CrossRef]
  80. Xu, H.; Li, X.; Liu, D.; Li, J.; Zhang, X.; Chen, X.; Hou, S.; Peng, L.; Xu, C.; Liu, W.; et al. Follicular T-helper cell recruitment governed by bystander B cells and ICOS-driven motility. Nature 2013, 496, 523–527. [Google Scholar] [CrossRef]
  81. Akiba, H.; Takeda, K.; Kojima, Y.; Usui, Y.; Harada, N.; Yamazaki, T.; Ma, J.; Tezuka, K.; Yagita, H.; Okumura, K. The role of ICOS in the CXCR5+ follicular B helper T cell maintenance in vivo. J. Immunol. 2005, 175, 2340–2348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Burmeister, Y.; Lischke, T.; Dahler, A.C.; Mages, H.W.; Lam, K.P.; Coyle, A.J.; Kroczek, R.A.; Hutloff, A. ICOS controls the pool size of effector-memory and regulatory T cells. J. Immunol. 2008, 180, 774–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Vinuesa, C.G.; Cook, M.C.; Angelucci, C.; Athanasopoulos, V.; Rui, L.; Hill, K.M.; Yu, D.; Domaschenz, H.; Whittle, B.; Lambe, T.; et al. A RING-type ubiquitin ligase family member required to repress follicular helper T cells and autoimmunity. Nature 2005, 435, 452–458. [Google Scholar] [CrossRef] [PubMed]
  84. Deenick, E.K.; Chan, A.; Ma, C.S.; Gatto, D.; Schwartzberg, P.L.; Brink, R.; Tangye, S.G. Follicular helper T cell differentiation requires continuous antigen presentation that is independent of unique B cell signaling. Immunity 2010, 33, 241–253. [Google Scholar] [CrossRef] [Green Version]
  85. Qi, H.; Chen, X.; Chu, C.; Lu, P.; Xu, H.; Yan, J. Follicular T-helper cells: Controlled localization and cellular interactions. Immunol. Cell Biol. 2014, 92, 28–33. [Google Scholar] [CrossRef]
  86. Shulman, Z.; Gitlin, A.D.; Targ, S.; Jankovic, M.; Pasqual, G.; Nussenzweig, M.C.; Victora, G.D. T follicular helper cell dynamics in germinal centers. Science 2013, 341, 673–677. [Google Scholar] [CrossRef] [Green Version]
  87. Yusuf, I.; Kageyama, R.; Monticelli, L.; Johnston, R.J.; Ditoro, D.; Hansen, K.; Barnett, B.; Crotty, S. Germinal center T follicular helper cell IL-4 production is dependent on signaling lymphocytic activation molecule receptor (CD150). J. Immunol. 2010, 185, 190–202. [Google Scholar] [CrossRef] [Green Version]
  88. Cannons, J.L.; Qi, H.; Lu, K.T.; Dutta, M.; Gomez-Rodriguez, J.; Cheng, J.; Wakeland, E.K.; Germain, R.N.; Schwartzberg, P.L. Optimal germinal center responses require a multistage T cell:B cell adhesion process involving integrins, SLAM-associated protein, and CD84. Immunity 2010, 32, 253–265. [Google Scholar] [CrossRef] [Green Version]
  89. Kageyama, R.; Cannons, J.L.; Zhao, F.; Yusuf, I.; Lao, C.; Locci, M.; Schwartzberg, P.L.; Crotty, S. The receptor Ly108 functions as a SAP adaptor-dependent on-off switch for T cell help to B cells and NKT cell development. Immunity 2012, 36, 986–1002. [Google Scholar] [CrossRef] [Green Version]
  90. Morra, M.; Barrington, R.A.; Abadia-Molina, A.C.; Okamoto, S.; Julien, A.; Gullo, C.; Kalsy, A.; Edwards, M.J.; Chen, G.; Spolski, R.; et al. Defective B cell responses in the absence of SH2D1A. Proc. Natl. Acad. Sci. USA 2005, 102, 4819–4823. [Google Scholar] [CrossRef] [Green Version]
  91. McCausland, M.M.; Yusuf, I.; Tran, H.; Ono, N.; Yanagi, Y.; Crotty, S. SAP regulation of follicular helper CD4 T cell development and humoral immunity is independent of SLAM and Fyn kinase. J. Immunol. 2007, 178, 817–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Coffey, A.J.; Brooksbank, R.A.; Brandau, O.; Oohashi, T.; Howell, G.R.; Bye, J.M.; Cahn, A.P.; Durham, J.; Heath, P.; Wray, P.; et al. Host response to EBV infection in X-linked lymphoproliferative disease results from mutations in an SH2-domain encoding gene. Nat. Genet. 1998, 20, 129–135. [Google Scholar] [CrossRef] [PubMed]
  93. Ma, C.S.; Deenick, E.K. Human T follicular helper (Tfh) cells and disease. Immunol. Cell Biol. 2014, 92, 64–71. [Google Scholar] [CrossRef] [PubMed]
  94. Nichols, K.E.; Harkin, D.P.; Levitz, S.; Krainer, M.; Kolquist, K.A.; Genovese, C.; Bernard, A.; Ferguson, M.; Zuo, L.; Snyder, E.; et al. Inactivating mutations in an SH2 domain-encoding gene in X-linked lymphoproliferative syndrome. Proc. Natl. Acad. Sci. USA 1998, 95, 13765–13770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Suan, D.; Nguyen, A.; Moran, I.; Bourne, K.; Hermes, J.R.; Arshi, M.; Hampton, H.R.; Tomura, M.; Miwa, Y.; Kelleher, A.D.; et al. T follicular helper cells have distinct modes of migration and molecular signatures in naive and memory immune responses. Immunity 2015, 42, 704–718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Nurieva, R.I.; Chung, Y.; Martinez, G.J.; Yang, X.O.; Tanaka, S.; Matskevitch, T.D.; Wang, Y.H.; Dong, C. Bcl6 mediates the development of T follicular helper cells. Science 2009, 325, 1001–1005. [Google Scholar] [CrossRef] [Green Version]
  97. Inoue, T.; Moran, I.; Shinnakasu, R.; Phan, T.G.; Kurosaki, T. Generation of memory B cells and their reactivation. Immunol. Rev. 2018, 283, 138–149. [Google Scholar] [CrossRef]
  98. Shinnakasu, R.; Kurosaki, T. Regulation of memory B and plasma cell differentiation. Curr. Opin. Immunol. 2017, 45, 126–131. [Google Scholar] [CrossRef]
  99. Koutsakos, M.; Nguyen, T.H.O.; Kedzierska, K. With a Little Help from T Follicular Helper Friends: Humoral Immunity to Influenza Vaccination. J. Immunol. 2019, 202, 360–367. [Google Scholar] [CrossRef] [Green Version]
  100. Cattoretti, G.; Angelin-Duclos, C.; Shaknovich, R.; Zhou, H.; Wang, D.; Alobeid, B. PRDM1/Blimp-1 is expressed in human B-lymphocytes committed to the plasma cell lineage. J. Pathol. 2005, 206, 76–86. [Google Scholar] [CrossRef]
  101. Rogerson, B.; Hackett, J., Jr.; Peters, A.; Haasch, D.; Storb, U. Mutation pattern of immunoglobulin transgenes is compatible with a model of somatic hypermutation in which targeting of the mutator is linked to the direction of DNA replication. EMBO J. 1991, 10, 4331–4341. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, Y.J.; Xu, J.; de Bouteiller, O.; Parham, C.L.; Grouard, G.; Djossou, O.; de Saint-Vis, B.; Lebecque, S.; Banchereau, J.; Moore, K.W. Follicular dendritic cells specifically express the long CR2/CD21 isoform. J. Exp. Med. 1997, 185, 165–170. [Google Scholar] [CrossRef] [PubMed]
  103. Qin, D.; Wu, J.; Vora, K.A.; Ravetch, J.V.; Szakal, A.K.; Manser, T.; Tew, J.G. Fc gamma receptor IIB on follicular dendritic cells regulates the B cell recall response. J. Immunol. 2000, 164, 6268–6275. [Google Scholar] [CrossRef] [PubMed]
  104. Good-Jacobson, K.L.; Szumilas, C.G.; Chen, L.; Sharpe, A.H.; Tomayko, M.M.; Shlomchik, M.J. PD-1 regulates germinal center B cell survival and the formation and affinity of long-lived plasma cells. Nat. Immunol. 2010, 11, 535–542. [Google Scholar] [CrossRef] [PubMed]
  105. Victora, G.D.; Nussenzweig, M.C. Germinal centers. Annu Rev. Immunol. 2012, 30, 429–457. [Google Scholar] [CrossRef] [PubMed]
  106. Shulman, Z.; Gitlin, A.D.; Weinstein, J.S.; Lainez, B.; Esplugues, E.; Flavell, R.A.; Craft, J.E.; Nussenzweig, M.C. Dynamic signaling by T follicular helper cells during germinal center B cell selection. Science 2014, 345, 1058–1062. [Google Scholar] [CrossRef] [Green Version]
  107. Blink, E.J.; Light, A.; Kallies, A.; Nutt, S.L.; Hodgkin, P.D.; Tarlinton, D.M. Early appearance of germinal center-derived memory B cells and plasma cells in blood after primary immunization. J. Exp. Med. 2005, 201, 545–554. [Google Scholar] [CrossRef] [Green Version]
  108. Phan, T.G.; Paus, D.; Chan, T.D.; Turner, M.L.; Nutt, S.L.; Basten, A.; Brink, R. High affinity germinal center B cells are actively selected into the plasma cell compartment. J. Exp. Med. 2006, 203, 2419–2424. [Google Scholar] [CrossRef]
  109. Heesters, B.A.; Myers, R.C.; Carroll, M.C. Follicular dendritic cells: Dynamic antigen libraries. Nat. Rev. Immunol. 2014, 14, 495–504. [Google Scholar] [CrossRef]
  110. Allen, C.D.; Okada, T.; Tang, H.L.; Cyster, J.G. Imaging of germinal center selection events during affinity maturation. Science 2007, 315, 528–531. [Google Scholar] [CrossRef]
  111. Schwickert, T.A.; Lindquist, R.L.; Shakhar, G.; Livshits, G.; Skokos, D.; Kosco-Vilbois, M.H.; Dustin, M.L.; Nussenzweig, M.C. In Vivo imaging of germinal centres reveals a dynamic open structure. Nature 2007, 446, 83–87. [Google Scholar] [CrossRef] [PubMed]
  112. Hams, E.; McCarron, M.J.; Amu, S.; Yagita, H.; Azuma, M.; Chen, L.; Fallon, P.G. 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, 5648–5655. [Google Scholar] [CrossRef] [PubMed]
  113. Gitlin, A.D.; Shulman, Z.; Nussenzweig, M.C. Clonal selection in the germinal centre by regulated proliferation and hypermutation. Nature 2014, 509, 637–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Rivino, L.; Messi, M.; Jarrossay, D.; Lanzavecchia, A.; Sallusto, F.; Geginat, J. Chemokine receptor expression identifies Pre-T helper (Th)1, Pre-Th2, and nonpolarized cells among human CD4+ central memory T cells. J. Exp. Med. 2004, 200, 725–735. [Google Scholar] [CrossRef] [PubMed]
  115. Brenna, E.; Davydov, A.N.; Ladell, K.; McLaren, J.E.; Bonaiuti, P.; Metsger, M.; Ramsden, J.D.; Gilbert, S.C.; Lambe, T.; Price, D.A.; et al. CD4(+) T Follicular Helper Cells in Human Tonsils and Blood Are Clonally Convergent but Divergent from Non-Tfh CD4(+) Cells. Cell Rep. 2020, 30, 137–152.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Heit, A.; Schmitz, F.; Gerdts, S.; Flach, B.; Moore, M.S.; Perkins, J.A.; Robins, H.S.; Aderem, A.; Spearman, P.; Tomaras, G.D.; et al. Vaccination establishes clonal relatives of germinal center T cells in the blood of humans. J. Exp. Med. 2017, 214, 2139–2152. [Google Scholar] [CrossRef] [Green Version]
  117. Vella, L.A.; Buggert, M.; Manne, S.; Herati, R.S.; Sayin, I.; Kuri-Cervantes, L.; Bukh Brody, I.; O’Boyle, K.C.; Kaprielian, H.; Giles, J.R.; et al. T follicular helper cells in human efferent lymph retain lymphoid characteristics. J. Clin. Investig. 2019, 129, 3185–3200. [Google Scholar] [CrossRef] [Green Version]
  118. Blanco, P.; Ueno, H.; Schmitt, N. T follicular helper (Tfh) cells in lupus: Activation and involvement in SLE pathogenesis. Eur J. Immunol. 2016, 46, 281–290. [Google Scholar] [CrossRef]
  119. Tangye, S.G.; Ma, C.S.; Brink, R.; Deenick, E.K. The good, the bad and the ugly—TFH cells in human health and disease. Nat. Rev. Immunol. 2013, 13, 412–426. [Google Scholar] [CrossRef]
  120. Oli, A.N.; Obialor, W.O.; Ifeanyichukwu, M.O.; Odimegwu, D.C.; Okoyeh, J.N.; Emechebe, G.O.; Adejumo, S.A.; Ibeanu, G.C. Immunoinformatics and Vaccine Development: An Overview. Immunotargets Ther. 2020, 9, 13–30. [Google Scholar] [CrossRef] [Green Version]
  121. Simpson, N.; Gatenby, P.A.; Wilson, A.; Malik, S.; Fulcher, D.A.; Tangye, S.G.; Manku, H.; Vyse, T.J.; Roncador, G.; Huttley, G.A.; et al. 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, 234–244. [Google Scholar] [CrossRef] [PubMed]
  122. Le Coz, C.; Joublin, A.; Pasquali, J.L.; Korganow, A.S.; Dumortier, H.; Monneaux, F. Circulating TFH subset distribution is strongly affected in lupus patients with an active disease. PLoS ONE 2013, 8, e75319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Vieth, B.; Parekh, S.; Ziegenhain, C.; Enard, W.; Hellmann, I. A systematic evaluation of single cell RNA-seq analysis pipelines. Nat. Commun. 2019, 10, 4667. [Google Scholar] [CrossRef] [Green Version]
  124. Vogel, K.U.; Edelmann, S.L.; Jeltsch, K.M.; Bertossi, A.; Heger, K.; Heinz, G.A.; Zoller, J.; Warth, S.C.; Hoefig, K.P.; Lohs, C.; et al. Roquin paralogs 1 and 2 redundantly repress the Icos and Ox40 costimulator mRNAs and control follicular helper T cell differentiation. Immunity 2013, 38, 655–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Pratama, A.; Ramiscal, R.R.; Silva, D.G.; Das, S.K.; Athanasopoulos, V.; Fitch, J.; Botelho, N.K.; Chang, P.P.; Hu, X.; Hogan, J.J.; et al. Roquin-2 shares functions with its paralog Roquin-1 in the repression of mRNAs controlling T follicular helper cells and systemic inflammation. Immunity 2013, 38, 669–680. [Google Scholar] [CrossRef] [Green Version]
  126. Cubas, R.A.; Mudd, J.C.; Savoye, A.L.; Perreau, M.; van Grevenynghe, J.; Metcalf, T.; Connick, E.; Meditz, A.; Freeman, G.J.; Abesada-Terk, G., Jr.; et al. Inadequate T follicular cell help impairs B cell immunity during HIV infection. Nat. Med. 2013, 19, 494–499. [Google Scholar] [CrossRef] [Green Version]
  127. Xu, Y.; Weatherall, C.; Bailey, M.; Alcantara, S.; De Rose, R.; Estaquier, J.; Wilson, K.; Suzuki, K.; Corbeil, J.; Cooper, D.A.; et al. Simian immunodeficiency virus infects follicular helper CD4 T cells in lymphoid tissues during pathogenic infection of pigtail macaques. J. Virol. 2013, 87, 3760–3773. [Google Scholar] [CrossRef] [Green Version]
  128. Perreau, M.; Savoye, A.L.; De Crignis, E.; Corpataux, J.M.; Cubas, R.; Haddad, E.K.; De Leval, L.; Graziosi, C.; Pantaleo, G. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J. Exp. Med. 2013, 210, 143–156. [Google Scholar] [CrossRef]
  129. Zaunders, J.; Xu, Y.; Kent, S.J.; Koelsch, K.K.; Kelleher, A.D. Divergent Expression of CXCR5 and CCR5 on CD4(+) T Cells and the Paradoxical Accumulation of T Follicular Helper Cells during HIV Infection. Front. Immunol. 2017, 8, 495. [Google Scholar] [CrossRef] [Green Version]
  130. Lindqvist, M.; van Lunzen, J.; Soghoian, D.Z.; Kuhl, B.D.; Ranasinghe, S.; Kranias, G.; Flanders, M.D.; Cutler, S.; Yudanin, N.; Muller, M.I.; et al. Expansion of HIV-specific T follicular helper cells in chronic HIV infection. J. Clin. Investig. 2012, 122, 3271–3280. [Google Scholar] [CrossRef]
  131. Niessl, J.; Baxter, A.E.; Morou, A.; Brunet-Ratnasingham, E.; Sannier, G.; Gendron-Lepage, G.; Richard, J.; Delgado, G.G.; Brassard, N.; Turcotte, I.; et al. Persistent expansion and Th1-like skewing of HIV-specific circulating T follicular helper cells during antiretroviral therapy. EBioMedicine 2020, 54, 102727. [Google Scholar] [CrossRef] [PubMed]
  132. Ueno, H. Human Circulating T Follicular Helper Cell Subsets in Health and Disease. J. Clin. Immunol. 2016, 36 (Suppl. 1), 34–39. [Google Scholar] [CrossRef]
  133. Buranapraditkun, S.; Pissani, F.; Teigler, J.E.; Schultz, B.T.; Alter, G.; Marovich, M.; Robb, M.L.; Eller, M.A.; Martin, J.; Deeks, S.; et al. Preservation of Peripheral T Follicular Helper Cell Function in HIV Controllers. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Cubas, R.; van Grevenynghe, J.; Wills, S.; Kardava, L.; Santich, B.H.; Buckner, C.M.; Muir, R.; Tardif, V.; Nichols, C.; Procopio, F.; et al. Reversible Reprogramming of Circulating Memory T Follicular Helper Cell Function during Chronic HIV Infection. J. Immunol. 2015, 195, 5625–5636. [Google Scholar] [CrossRef] [PubMed]
  135. Colineau, L.; Rouers, A.; Yamamoto, T.; Xu, Y.; Urrutia, A.; Pham, H.P.; Cardinaud, S.; Samri, A.; Dorgham, K.; Coulon, P.G.; et al. HIV-Infected Spleens Present Altered Follicular Helper T Cell (Tfh) Subsets and Skewed B Cell Maturation. PLoS ONE 2015, 10, e0140978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Qin, L.; Waseem, T.C.; Sahoo, A.; Bieerkehazhi, S.; Zhou, H.; Galkina, E.V.; Nurieva, R. Insights Into the Molecular Mechanisms of T Follicular Helper-Mediated Immunity and Pathology. Front. Immunol. 2018, 9, 1884. [Google Scholar] [CrossRef] [PubMed]
  137. Kobayashi, T.; Iijima, K.; Dent, A.L.; Kita, H. Follicular helper T cells mediate IgE antibody response to airborne allergens. J. Allergy Clin. Immunol. 2017, 139, 300–313.e7. [Google Scholar] [CrossRef] [Green Version]
  138. Szabo, K.; Gaspar, K.; Dajnoki, Z.; Papp, G.; Fabos, B.; Szegedi, A.; Zeher, M. Expansion of circulating follicular T helper cells associates with disease severity in childhood atopic dermatitis. Immunol. Lett. 2017, 189, 101–108. [Google Scholar] [CrossRef] [Green Version]
  139. Czarnowicki, T.; Esaki, H.; Gonzalez, J.; Renert-Yuval, Y.; Brunner, P.; Oliva, M.; Estrada, Y.; Xu, H.; Zheng, X.; Talasila, S.; et al. Alterations in B-cell subsets in pediatric patients with early atopic dermatitis. J. Allergy Clin. Immunol. 2017, 140, 134–144.e9. [Google Scholar] [CrossRef] [Green Version]
  140. Gong, F.; Qian, C.; Zhu, H.; Zhu, J.; Pan, Y.; Dong, Q.; Jiang, D. Circulating follicular T-helper cell subset distribution in patients with asthma. Allergy Asthma Proc. 2016, 37, 154–161. [Google Scholar] [CrossRef]
  141. Noble, A.; Zhao, J. Follicular helper T cells are responsible for IgE responses to Der p 1 following house dust mite sensitization in mice. Clin. Exp. Allergy 2016, 46, 1075–1082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Bentebibel, S.E.; Lopez, S.; Obermoser, G.; Schmitt, N.; Mueller, C.; Harrod, C.; Flano, E.; Mejias, A.; Albrecht, R.A.; Blankenship, D.; et al. Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination. Sci. Transl. Med. 2013, 5, 176ra132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Herati, R.S.; Muselman, A.; Vella, L.; Bengsch, B.; Parkhouse, K.; Del Alcazar, D.; Kotzin, J.; Doyle, S.A.; Tebas, P.; Hensley, S.E.; et al. Successive annual influenza vaccination induces a recurrent oligoclonotypic memory response in circulating T follicular helper cells. Sci. Immunol. 2017, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Herati, R.S.; Reuter, M.A.; Dolfi, D.V.; Mansfield, K.D.; Aung, H.; Badwan, O.Z.; Kurupati, R.K.; Kannan, S.; Ertl, H.; Schmader, K.E.; et al. Circulating CXCR5+PD-1+ response predicts influenza vaccine antibody responses in young adults but not elderly adults. J. Immunol. 2014, 193, 3528–3537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Koutsakos, M.; Wheatley, A.K.; Loh, L.; Clemens, E.B.; Sant, S.; Nussing, S.; Fox, A.; Chung, A.W.; Laurie, K.L.; Hurt, A.C.; et al. Circulating TFH cells, serological memory, and tissue compartmentalization shape human influenza-specific B cell immunity. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Pilkinton, M.A.; Nicholas, K.J.; Warren, C.M.; Smith, R.M.; Yoder, S.M.; Talbot, H.K.; Kalams, S.A. Greater activation of peripheral T follicular helper cells following high dose influenza vaccine in older adults forecasts seroconversion. Vaccine 2017, 35, 329–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. He, J.; Tsai, L.M.; Leong, Y.A.; Hu, X.; Ma, C.S.; Chevalier, N.; Sun, X.; Vandenberg, K.; Rockman, S.; Ding, Y.; et al. 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, 770–781. [Google Scholar] [CrossRef] [Green Version]
  148. Bentebibel, S.E.; Khurana, S.; Schmitt, N.; Kurup, P.; Mueller, C.; Obermoser, G.; Palucka, A.K.; Albrecht, R.A.; Garcia-Sastre, A.; Golding, H.; et al. ICOS(+)PD-1(+)CXCR3(+) T follicular helper cells contribute to the generation of high-avidity antibodies following influenza vaccination. Sci. Rep. 2016, 6, 26494. [Google Scholar] [CrossRef]
  149. Spensieri, F.; Borgogni, E.; Zedda, L.; Bardelli, M.; Buricchi, F.; Volpini, G.; Fragapane, E.; Tavarini, S.; Finco, O.; Rappuoli, R.; et al. Human circulating influenza-CD4+ ICOS1+IL-21+ T cells expand after vaccination, exert helper function, and predict antibody responses. Proc. Natl. Acad. Sci. USA 2013, 110, 14330–14335. [Google Scholar] [CrossRef] [Green Version]
  150. Stebegg, M.; Bignon, A.; Hill, D.L.; Silva-Cayetano, A.; Krueger, C.; Vanderleyden, I.; Innocentin, S.; Boon, L.; Wang, J.; Zand, M.S.; et al. Rejuvenating conventional dendritic cells and T follicular helper cell formation after vaccination. eLife 2020, 9. [Google Scholar] [CrossRef]
  151. Pape, K.A.; Taylor, J.J.; Maul, R.W.; Gearhart, P.J.; Jenkins, M.K. Different B cell populations mediate early and late memory during an endogenous immune response. Science 2011, 331, 1203–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Havenar-Daughton, C.; Carnathan, D.G.; Torrents de la Pena, A.; Pauthner, M.; Briney, B.; Reiss, S.M.; Wood, J.S.; Kaushik, K.; van Gils, M.J.; Rosales, S.L.; et al. Direct Probing of Germinal Center Responses Reveals Immunological Features and Bottlenecks for Neutralizing Antibody Responses to HIV Env Trimer. Cell Rep. 2016, 17, 2195–2209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Havenar-Daughton, C.; Carnathan, D.G.; Boopathy, A.V.; Upadhyay, A.A.; Murrell, B.; Reiss, S.M.; Enemuo, C.A.; Gebru, E.H.; Choe, Y.; Dhadvai, P.; et al. Rapid Germinal Center and Antibody Responses in Non-human Primates after a Single Nanoparticle Vaccine Immunization. Cell Rep. 2019, 29, 1756–1766 e1758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Hill, D.L.; Pierson, W.; Bolland, D.J.; Mkindi, C.; Carr, E.J.; Wang, J.; Houard, S.; Wingett, S.W.; Audran, R.; Wallin, E.F.; et al. The adjuvant GLA-SE promotes human Tfh cell expansion and emergence of public TCRbeta clonotypes. J. Exp. Med. 2019, 216, 1857–1873. [Google Scholar] [CrossRef] [PubMed]
  155. Dash, P.; Fiore-Gartland, A.J.; Hertz, T.; Wang, G.C.; Sharma, S.; Souquette, A.; Crawford, J.C.; Clemens, E.B.; Nguyen, T.H.O.; Kedzierska, K.; et al. Quantifiable predictive features define epitope-specific T cell receptor repertoires. Nature 2017, 547, 89–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Emerson, R.O.; DeWitt, W.S.; Vignali, M.; Gravley, J.; Hu, J.K.; Osborne, E.J.; Desmarais, C.; Klinger, M.; Carlson, C.S.; Hansen, J.A.; et al. Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire. Nat. Genet. 2017, 49, 659–665. [Google Scholar] [CrossRef]
  157. Glanville, J.; Huang, H.; Nau, A.; Hatton, O.; Wagar, L.E.; Rubelt, F.; Ji, X.; Han, A.; Krams, S.M.; Pettus, C.; et al. Identifying specificity groups in the T cell receptor repertoire. Nature 2017, 547, 94–98. [Google Scholar] [CrossRef]
  158. Jackson, K.J.; Liu, Y.; Roskin, K.M.; Glanville, J.; Hoh, R.A.; Seo, K.; Marshall, E.L.; Gurley, T.C.; Moody, M.A.; Haynes, B.F.; et al. Human responses to influenza vaccination show seroconversion signatures and convergent antibody rearrangements. Cell Host Microbe 2014, 16, 105–114. [Google Scholar] [CrossRef] [Green Version]
  159. Becattini, S.; Latorre, D.; Mele, F.; Foglierini, M.; De Gregorio, C.; Cassotta, A.; Fernandez, B.; Kelderman, S.; Schumacher, T.N.; Corti, D.; et al. T cell immunity. Functional heterogeneity of human memory CD4(+) T cell clones primed by pathogens or vaccines. Science 2015, 347, 400–406. [Google Scholar] [CrossRef]
  160. Han, A.; Glanville, J.; Hansmann, L.; Davis, M.M. Linking T-cell receptor sequence to functional phenotype at the single-cell level. Nat. Biotechnol. 2014, 32, 684–692. [Google Scholar] [CrossRef]
  161. Attaf, M.; Huseby, E.; Sewell, A.K. alphabeta T cell receptors as predictors of health and disease. Cell Mol. Immunol. 2015, 12, 391–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Wu, Q.; Pesenacker, A.M.; Stansfield, A.; King, D.; Barge, D.; Foster, H.E.; Abinun, M.; Wedderburn, L.R. Immunological characteristics and T-cell receptor clonal diversity in children with systemic juvenile idiopathic arthritis undergoing T-cell-depleted autologous stem cell transplantation. Immunology 2014, 142, 227–236. [Google Scholar] [CrossRef] [PubMed]
  163. Lossius, A.; Johansen, J.N.; Vartdal, F.; Robins, H.; Jurate Saltyte, B.; Holmoy, T.; Olweus, J. High-throughput sequencing of TCR repertoires in multiple sclerosis reveals intrathecal enrichment of EBV-reactive CD8+ T cells. Eur. J. Immunol. 2014, 44, 3439–3452. [Google Scholar] [CrossRef] [PubMed]
  164. Gomes, T.; Teichmann, S.A.; Talavera-Lopez, C. Immunology Driven by Large-Scale Single-Cell Sequencing. Trends Immunol. 2019, 40, 1011–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Davis, M.M.; Tato, C.M.; Furman, D. Systems immunology: Just getting started. Nat. Immunol. 2017, 18, 725–732. [Google Scholar] [CrossRef] [PubMed]
  166. Querec, T.D.; Akondy, R.S.; Lee, E.K.; Cao, W.; Nakaya, H.I.; Teuwen, D.; Pirani, A.; Gernert, K.; Deng, J.; Marzolf, B.; et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat. Immunol. 2009, 10, 116–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Voigt, E.A.; Grill, D.E.; Zimmermann, M.T.; Simon, W.L.; Ovsyannikova, I.G.; Kennedy, R.B.; Poland, G.A. Transcriptomic signatures of cellular and humoral immune responses in older adults after seasonal influenza vaccination identified by data-driven clustering. Sci. Rep. 2018, 8, 739. [Google Scholar] [CrossRef] [Green Version]
  168. Nakaya, H.I.; Hagan, T.; Duraisingham, S.S.; Lee, E.K.; Kwissa, M.; Rouphael, N.; Frasca, D.; Gersten, M.; Mehta, A.K.; Gaujoux, R.; et al. Systems Analysis of Immunity to Influenza Vaccination across Multiple Years and in Diverse Populations Reveals Shared Molecular Signatures. Immunity 2015, 43, 1186–1198. [Google Scholar] [CrossRef] [Green Version]
  169. Zimmermann, M.T.; Oberg, A.L.; Grill, D.E.; Ovsyannikova, I.G.; Haralambieva, I.H.; Kennedy, R.B.; Poland, G.A. System-Wide Associations between DNA-Methylation, Gene Expression, and Humoral Immune Response to Influenza Vaccination. PLoS ONE 2016, 11, e0152034. [Google Scholar] [CrossRef]
  170. Mizukami, T.; Momose, H.; Kuramitsu, M.; Takizawa, K.; Araki, K.; Furuhata, K.; Ishii, K.J.; Hamaguchi, I.; Yamaguchi, K. System vaccinology for the evaluation of influenza vaccine safety by multiplex gene detection of novel biomarkers in a preclinical study and batch release test. PLoS ONE 2014, 9, e101835. [Google Scholar] [CrossRef]
  171. Haralambieva, I.H.; Zimmermann, M.T.; Ovsyannikova, I.G.; Grill, D.E.; Oberg, A.L.; Kennedy, R.B.; Poland, G.A. Whole Transcriptome Profiling Identifies CD93 and Other Plasma Cell Survival Factor Genes Associated with Measles-Specific Antibody Response after Vaccination. PLoS ONE 2016, 11, e0160970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Li, S.; Rouphael, N.; Duraisingham, S.; Romero-Steiner, S.; Presnell, S.; Davis, C.; Schmidt, D.S.; Johnson, S.E.; Milton, A.; Rajam, G.; et al. Molecular signatures of antibody responses derived from a systems biology study of five human vaccines. Nat. Immunol. 2014, 15, 195–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Lonnberg, T.; Svensson, V.; James, K.R.; Fernandez-Ruiz, D.; Sebina, I.; Montandon, R.; Soon, M.S.; Fogg, L.G.; Nair, A.S.; Liligeto, U.; et al. Single-cell RNA-seq and computational analysis using temporal mixture modelling resolves Th1/Tfh fate bifurcation in malaria. Sci. Immunol. 2017, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. T cell differentiation in the thymus. (A) Immature thymocytes in the thymic cortex upregulate Notch, Kit and CD44 to begin differentiation in double-negative (DN) phase 1. As CD25 expression is upregulated, thymocytes progress into DN phase 2, where T cell receptor (TCR) β-chain rearrangement begins. Rearrangement of the β-chain locus of the TCR, from the Diversity β (Dβ) to the Joining β (Jβ) region, begins, and as expression of CD44 and Kit are reduced, Vβ to DβJβ rearrangements occur and cells progress into the DN3 phase. Only cells that successfully produce a reproductive β-chain survive and progress into DN phase 4, where the β-chain is paired with a surrogate pre-α chain to form the pre-TCR. (B) During the DN4 phase, expression of CD4 and CD8 are triggered, progressing cells into the double-positive (DP) phase of differentiation. It is here that α-chain rearrangement begins, and recombination of Vα to Jα gene segments continues until cell death or positive selection occurs. During the DP phase, cells produce a functional TCR and undergo positive selection so that ultimately only functionally competent and self-tolerant cells remain. (C) Thymocytes then migrate into the thymic medulla, where downregulation of Notch and either CD4 or CD8 occurs. This is the final phase of differentiation, termed the single-positive phase, where naïve T cells expressing either CD4 or CD8 and a functional TCR can exit the thymus and traffic to the periphery (created with BioRender.com).
Figure 1. T cell differentiation in the thymus. (A) Immature thymocytes in the thymic cortex upregulate Notch, Kit and CD44 to begin differentiation in double-negative (DN) phase 1. As CD25 expression is upregulated, thymocytes progress into DN phase 2, where T cell receptor (TCR) β-chain rearrangement begins. Rearrangement of the β-chain locus of the TCR, from the Diversity β (Dβ) to the Joining β (Jβ) region, begins, and as expression of CD44 and Kit are reduced, Vβ to DβJβ rearrangements occur and cells progress into the DN3 phase. Only cells that successfully produce a reproductive β-chain survive and progress into DN phase 4, where the β-chain is paired with a surrogate pre-α chain to form the pre-TCR. (B) During the DN4 phase, expression of CD4 and CD8 are triggered, progressing cells into the double-positive (DP) phase of differentiation. It is here that α-chain rearrangement begins, and recombination of Vα to Jα gene segments continues until cell death or positive selection occurs. During the DP phase, cells produce a functional TCR and undergo positive selection so that ultimately only functionally competent and self-tolerant cells remain. (C) Thymocytes then migrate into the thymic medulla, where downregulation of Notch and either CD4 or CD8 occurs. This is the final phase of differentiation, termed the single-positive phase, where naïve T cells expressing either CD4 or CD8 and a functional TCR can exit the thymus and traffic to the periphery (created with BioRender.com).
Ijms 21 08524 g001
Figure 2. Tfh circulation in the lymph node (LN). Naïve T cells enter the LN either through afferent lymphatic vessels, or from the blood via the high endothelial venules (HEV). A complex adhesion cascade driven by interactions of CCR7 and CCL21 facilitate naïve T cell migration through the medulla and to the paracortex (T cell zone). Here, dendritic cells (DCs) prime naïve T cells, resulting in CCR7 downregulation and CXCR5 upregulation. CXCR5 expression promotes Tfh cell differentiation and migration to the border of the B cell follicle via a CXCL13 gradient. It is here, at the T–B cell border, that cognate interactions involving critical signalling through ICOS/ICOSL, CD40L/CD40, TCR/MHC-II and SAP/SLAM support further Tfh cell polarisation and differentiation, B cell help and germinal centre (GC) formation. The GC reaction begins with Tfh and B cell interactions, where B cells will either differentiate into short-lived antigen-secreting cells or enter the GC dark zone (DZ) to undergo multiple rounds of somatic hypermutation (SHM), selection and proliferation. B cells then exit the DZ and migrate into the light zone (LZ) of the GC, where they compete for antigen presented by follicular dendritic cells (FDC). Tfh cells then selectively provide help to B cells possessing high-affinity B cell receptors (BCRs). After these Tfh–B cell interactions, B cells either differentiate into memory B cells, differentiate into long-lived plasma cells or re-enter the DZ for further rounds of SHM and selection (created with BioRender.com).
Figure 2. Tfh circulation in the lymph node (LN). Naïve T cells enter the LN either through afferent lymphatic vessels, or from the blood via the high endothelial venules (HEV). A complex adhesion cascade driven by interactions of CCR7 and CCL21 facilitate naïve T cell migration through the medulla and to the paracortex (T cell zone). Here, dendritic cells (DCs) prime naïve T cells, resulting in CCR7 downregulation and CXCR5 upregulation. CXCR5 expression promotes Tfh cell differentiation and migration to the border of the B cell follicle via a CXCL13 gradient. It is here, at the T–B cell border, that cognate interactions involving critical signalling through ICOS/ICOSL, CD40L/CD40, TCR/MHC-II and SAP/SLAM support further Tfh cell polarisation and differentiation, B cell help and germinal centre (GC) formation. The GC reaction begins with Tfh and B cell interactions, where B cells will either differentiate into short-lived antigen-secreting cells or enter the GC dark zone (DZ) to undergo multiple rounds of somatic hypermutation (SHM), selection and proliferation. B cells then exit the DZ and migrate into the light zone (LZ) of the GC, where they compete for antigen presented by follicular dendritic cells (FDC). Tfh cells then selectively provide help to B cells possessing high-affinity B cell receptors (BCRs). After these Tfh–B cell interactions, B cells either differentiate into memory B cells, differentiate into long-lived plasma cells or re-enter the DZ for further rounds of SHM and selection (created with BioRender.com).
Ijms 21 08524 g002
Table 1. Key Tfh cell receptors and ligands.
Table 1. Key Tfh cell receptors and ligands.
Receptor/LigandCorresponding GeneReciprocal Receptor/LigandFunction
CCR7CCR7CCL21Promotes naïve T cell homing to the T cell zone (paracortex) of the LN [31,47,54,56].
CXCR5CXCR5CXCL13Essential for T and B cell homing to the B cell follicle, within the cortex of the LN [55,72,73].
Bcl6BCL6-Master transcription factor for Tfh cell lineage. Limits Th1 differentiation through Blimp-1 pathway and supports Tfh cell differentiation [96].
IL-21IL21IL-21RRegulates ICOSL expression on B cells essential for Tfh and B cell interactions through ICOS/ICOSL signalling [60].
Regulates Tfh differentiation through c-Maf promotion of IL-21 production [61].
ICOSICOSICOSLInteracts with ICOSL expressed on B cells, influencing Tfh and B cell positioning within the LN [84].
Necessary for persistent T cell migration to the T–B cell border of the B cell follicle and optimal germinal centre responses [80].
TCR-MCH-IIT cell receptor that binds with peptide–MHC-II complexes.
Higher TCR affinity results in longer TCR–MHC-II interactions, ultimately influencing cell fate decisions [69,70,71].
SAPSH2D1ASLAMF6 and SLAMF5Necessary for forming stable T–B cell conjugates via SLAMF6 binding within the GC [75].
Supports Tfh cell adhesion and development and B cell responses when bound to SLAMF6 [90,91].
Essential for optimal GC responses by SLAMF5 binding [88].
CD40LCD40LGCD40Essential for formation and maintenance of the GC, promotes antibody class switching and provides crucial survival signals to GC B cells [76,77,78,79].
EBI2GPR1837α,25-dihydroxycholesterol (7α,25-OHC)Involved in temporospatial guidance of B cells and Tfh cells through the LN follicle mantle and GC [95].
S1PR1S1PR1S1PBinds to S1P secreted by lymphatic vessels, facilitating T cell migration into the LN [33,34,35].
High concentrations of S1P in efferent lymph promotes Tfh cell egress from the LN via S1PR1 binding [50].
S1PR2S1PR2S1PInvolved in guidance and retention of Tfh cells within the GC via repelling them from S1P-rich lymph [95].
CD62LSELLGlyCAM-1Facilitates adhesion and rolling of T cells along high endothelial venules as they migrate into the LN [36].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Law, H.; Venturi, V.; Kelleher, A.; Munier, C.M.L. Tfh Cells in Health and Immunity: Potential Targets for Systems Biology Approaches to Vaccination. Int. J. Mol. Sci. 2020, 21, 8524. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21228524

AMA Style

Law H, Venturi V, Kelleher A, Munier CML. Tfh Cells in Health and Immunity: Potential Targets for Systems Biology Approaches to Vaccination. International Journal of Molecular Sciences. 2020; 21(22):8524. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21228524

Chicago/Turabian Style

Law, Hannah, Vanessa Venturi, Anthony Kelleher, and C. Mee Ling Munier. 2020. "Tfh Cells in Health and Immunity: Potential Targets for Systems Biology Approaches to Vaccination" International Journal of Molecular Sciences 21, no. 22: 8524. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21228524

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop