Abstract
Purpose of review
T follicular helper (Tfh) cells play a critical role as providers of B-cell help and dysfunction in Tfh/B-cell interactions can lead to autoimmunity or immunodeficiency. These observations have generated a great deal of interest in understanding how these cells are affected during HIV infection and how their functional changes might affect antibody responses.
Recent findings
Recent studies have shown that HIV/SIV infection affects both Tfh-cell frequency and function and suggest that Tfh-cell perturbations might contribute to the relative inefficiency of HIV-infected individuals to generate broadly neutralizing antibodies (bNAbs).
Summary
The present review will highlight these recent findings addressing the role of Tfh cells in HIV infection as well as the impact HIV infection has on Tfh and circulating memory Tfh (cTfh) cell frequency and function.
Keywords: Tfh cells, HIV, B cells and dysfunction
Introduction
T follicular helper (Tfh) cells are a specialized subset of CD4 T cells that provides help to B cells in germinal centers (GCs) [1]. These cells are necessary for GC formation, immunoglobulin (Ig) class-switch recombination (CSR), somatic hypermutation (SHM) and differentiation of B cells into long-lived memory B cells and plasma cells [2–10]. GCs are specialized structures within B-cell follicles where B cells undergo SHM of their Ig variable (V) region genes to achieve higher affinity against a corresponding antigen conferring greater protective efficacy and CSR of their constant Ig domain (Fc) which provides different effector potentials [11–20]. Due to their critical role as providers of B-cell help, there has been a great deal of interest in understanding how these cells are affected during HIV infection and how their functional changes alter antibody responses.
Tfh-cell differentiation is driven by the transcription factor Bcl-6 [21, 22], although IRF4, BATF, MAF and the recently described Ascl2 are also involved in Tfh-cell differentiation and function [1, 23–26]. Tfh cells are characterized by their high level of CXCR5, ICOS and PD-1 [6–9] (described tissue resident Tfh cell subsets are summarized in Table 1) expression and produce a variety of cytokines including IL-4, IL-10 and IL-21 that are critical for the survival and differentiation of GC B cells into long-lived memory B cells and plasma cells [2–10]. Tfh/B-cell interactions are therefore essential for the generation of efficient neutralizing and non-neutralizing antibody responses [9, 10, 15, 20, 21, 41, 42] and have attracted a lot of recent interest.
Table 1.
Phenotype | Model | Tissue | References |
---|---|---|---|
Circulating Tfh-like cell subsets | |||
CD45RO+CXCR5+ | Human | Blood | [27] Bossaller et al. |
CXCR5+ with increased ICOS/PD-1 expression in SLE patients | Human | Blood | [28] Simpson et al. |
CD45RA−CXCR5+ subdivided into CXCR3−CCR6+, CXCR3−CCR6− and CXCR3+CCR6− cells | Human | Blood | [29] Morita et al. |
CD45RA−CCR7+CXCR5+ | Human | Blood | [30] Chevalier et al. |
CD45RA−CXCR5+ | Human | Blood | [31] Pallikkuth et al. |
CD45RO+CXCR5+CXCR3−PD-1+ | Human | Blood | [32] Locci et al. |
CD27hiCD45ROhiCCR7hiCXCR5hiCCR6hiPD-1hi | Human | Blood | [33] Boswell et al. |
Tissue resident Tfh cell subsets | |||
CD45RA−CXCR5+PD-1hi | Human | LN | [34] Perreau et al. |
CXCR5+PD-1hi | Human | LN | [35] Linqvist et al. |
CCR7lowPD-1hiICOShi | Macaques | LN | [36] Petrovas et al. |
CD3+PD-1hi (in situ) | Macaques | LN | [37] Hong et al. |
CD45RA−PD-1hiCD127low | Macaques | Spleen and LN | [38] Xu et al. |
CD45RA−CXCR5+CD57+ | Human | Tonsils | [39] Kim et al. |
CXCR5+ICOS+ | Human | Tonsils | [8] Breitfeld et al. |
CD45RA−CXCR5hi | Human | Tonsils and LN | [40] Cubas et al. |
The frequency and function of Tfh cells are tightly controlled under physiological conditions and their dysregulation can lead to various immune perturbations such as autoimmunity and immunodeficiency [1, 43–46]. In particular, recent studies have shown that HIV/SIV infection affects both Tfh-cell frequency and function [34–36, 40] and suggest that Tfh-cell perturbations might contribute to the relative inefficiency of HIV-infected individuals to generate bNAbs [32, 40]. Finally, the recently described population of blood circulating memory Tfh cells (cTfh) might mirror at least in part GC Tfh-cell perturbations [32]. In this review, we will highlight the recent findings that addressed the role of Tfh cells in HIV infection as well as the impact HIV infection has on Tfh and cTfh-cell frequency and function.
Role of Tfh cells in HIV infection
Lymphoid organs are the primary anatomic compartments for HIV/SIV replication and spreading [47–51] and in situ hybridization clearly indicated that HIV/SIV RNA was associated with GCs [37, 50, 52,]. In viremic HIV-1 infected individuals, Tfh cells were shown to contain the highest percentage of CD4 T cells harboring HIV DNA and were the most efficient in supporting productive infection in vitro [34]. Replication competent HIV was also readily isolated from Tfh cells in subjects with high and low viremia (<2000 HIV RNA copies) [34]. In addition, the frequency of Tfh cells was found to correlate with plasma viremia suggesting that Tfh cells might also be one of the primary sources of circulating virus or the primary target for HIV infection [34].
Interestingly, recent studies have also shown a relative expansion of Tfh cells during the viremic phase of both HIV and SIV infection [34–37]. These observations are not surprising as Tfh cells likely expand in response to cognate antigen, but this is in contrast with their increased susceptibility to SIV [36, 38] and HIV [34] infection. Indeed, HIV-infected CD4 T cells can be killed by either direct viral cytopathic effects or by HIV-specific CD8 T cells [53, 54]. Although the precise mechanism by which Tfh cells could withstand HIV-mediated depletion is unknown, HIV and SIV-specific CD8 T cells appeared to locate outside GCs [37, 55], which may in turn facilitate HIV/SIV-infected Tfh-cell accumulation in the follicles. Recently, a population of regulatory Qa-1-restricted CD8 T cells has been shown to localize in GCs and dampen Tfh cell development in mice [39]. However, their presence in human GCs and their role in targeting Tfh cells have not been investigated.
HIV-1 infected activated CD4 T cells escaping cytotoxic CD8 T cells as well as viral cytopathic effects can enter a quiescent state and thereby represent a major source of latently infected cells [56, 57] and a major obstacle for HIV eradication [56–59]. Indeed, estimates for the half-life of the HIV latent reservoir in the blood indicated that it might take as long as 70 years to completely eradicate the latent reservoir in the presence of fully suppressive ART [60]. Pioneer studies demonstrated that latently infected cells are relatively rare with a frequency of about 1 in 106 resting CD4 T cells with no significant difference observed between blood and lymph nodes [56, 61]. These observations led to the conclusion that cells from peripheral blood could be appropriately used to study the HIV latent reservoir. Using this strategy, Chomont et al. have identified central memory (CM; defined by the CD45RA−CCR7+CD27+) and transitional memory (TM; CD45RA−CCR7−CD27+) CD4 T cells as major cellular compartments of the latent HIV-1 reservoir in blood [62]. However, lymphoid organs contain about 98% of the total body lymphocytes [56] which are phenotypically and functionally distinct from CD4 T-cell populations circulating in the blood [6]. Therefore, studying HIV-1 latently infected LN memory CD4 T-cell populations might enable the identification of new cellular compartments that may contribute to the latent reservoir and help in the discovery of new targets for HIV-1 eradication. In this context, Yukl et al., are currently investigating the cellular subtype distribution of HIV-1 DNA contained in blood and lymphoid tissues collected from ART treated HIV-infected individuals and showed that effector memory (EM; CD45RO+CCR7−CD27−) CD4 T cells generally contained a higher level of HIV DNA than other CD4 T-cell populations in LNs (CROI 2014, #137). While Tfh cells might be enriched in this EM compartment [6, 8], further investigation and use of additional markers will be required to highlight the specific role of Tfh cells in the HIV latent reservoir.
Taken together, while recent studies have clearly demonstrated the preponderant role of Tfh cells during the viremic phase of HIV/SIV [34, 36] infection, the identification of latently infected CD4 T-cell reservoirs in the blood and lymph nodes (LNs) and their relationship to each other remain to be established and will contribute to efforts in finding targets for a potential cure.
Tfh cell functional alterations during HIV infection
While, HIV-1 infection elicits robust antibody responses to envelope proteins (gp120 and gp41) [63, 64], only 10–30% of HIV-infected individuals generate potent bNAbs [65–69]. This relative inefficiency to generate bNAbs is partially attributed to HIV-1 structure, which harbors numerous mechanisms of humoral evasion like sequence variable loops [70], glycosylation [71] and conformational masking of receptor-binding sites [72]) and by the fact that HIV-infected individuals develop lymphadenopathies associated with damages to follicular structures likely affecting B-cell differentiation and affinity maturation [73, 74]. Notably, the aforementioned morphological modifications have also been associated with Ig and B-cell abnormalities during the course of HIV infection [73]. Numerous B-cell perturbations appear during HIV infection and although many of these defects are improved by ART, B-cell responses to HIV specific antigens are diminished [75–81].
Due to their critical requirement in GC B-cell responses, several groups have investigated the impact of HIV/SIV infection on Tfh-cell frequency and function [34–37]. These studies have shown an expansion of Tfh cells in HIV/SIV-infected individuals that correlated positively with the frequency of GC B-cells and antibody production [34–37]. However, despite an increase in the frequency of these cells, we have recently shown that Tfh cells from HIV-infected individuals are functionally impaired and cannot provide adequate B-cell help [40]. This appeared to be partly due to increased PD-L1 expression by GC B cells leading to PD-1 triggering on Tfh cells which can affect Tfh cell proliferation, activation, ICOS expression and IL-21 cytokine secretion. Indeed blocking PD-1 signaling enhanced antibody production and exogenous IL-21 could rescue antibody responses and plasma cell generation in vitro [40]. It therefore appears that Tfh-cell function is affected in HIV infected LNs and might arise due to microenvironmental signals leading to an aberrant expression of inhibitory molecules.
The recent identification of follicular regulatory T (Tfr) cells that can migrate into follicles and restrain Tfh-cell differentiation represents another level of regulation in lymphoid tissues which could affect Tfh-cell function and B-cell responses during HIV infection [82–86]. Their mechanism of action is unknown but studies in mice indicated that in the absence of PD-1 and PD-L1 these cells expanded and inhibited Tfh-cell function [87]. In a PD-1/PD-L1 deregulated environment such as the one in HIV-infected LNs, the function of these cells might be reduced leading to an expansion of Tfh cells.
A better understanding of Tfh/GC B-cell interactions will have important implications for the generation of robust HIV-specific B cell responses and for the generation of de novo humoral responses to infections or vaccination. Clearly, the amount of data currently available is very limited and further investigation on the impact of HIV infection on Tfh-mediated B-cell help is required. A better understanding of the mechanisms that are affected by HIV infection leading to defective Tfh-cell signaling and B-cell responses could provide a critical framework for the development of novel therapeutics and vaccines and could also shed some light on the mechanisms responsible for the failure in the majority of HIV-infected individuals to produce bNAbs and their diminished responses to immunization [88–90].
Circulating memory Tfh cells
The fate and commitment of Tfh cells is still incompletely understood [91–95]. Indeed, using a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection, Hale et al., showed that both LCMV-specific memory CD4 T cells with Tfh and Th1 commitment were generated [91]. By using an IL-21 reporter mouse system, Lüthje et al., showed that Tfh cells gave rise to Tfh-like memory cells that could differentiate into conventional effector T helper cells or Tfh cells upon antigen recall [93]. Additional studies showed that Tfh cells could revert in long-lived memory Tfh cells expressing low levels of Bcl6, CXCR5 and PD-1 expression in the absence of antigen [92, 95]. Choi et al. also confirmed the capacity of Tfh cells to form memory and showed that Tfh cells shared phenotypic characteristics with memory precursor CD8 T cells [96]. Overall, these studies demonstrated that at least in mice, a fraction of Tfh cells have the capacity to become memory cells and can promote GC formation and antibody production more effectively than non-Tfh cells upon antigenic challenge [91–95].
Initial evidence for memory cTfh cells in humans came from studies of human malignancies [27, 28, 97]. A more comprehensive study by Morita et al. further defined a population of memory Tfh-like cells with an enhanced capacity to provide naïve and memory B-cell help [29]. This study subdivided circulating Tfh-like cells into three subsets (Th1, Th2 and Th17-like) based on their expression or the lack of CXCR3 and CCR6 and demonstrated that both the Th2 and Th17-like cells (CXCR3−CCR6− and CXCR3−CCR6+ respectively) efficiently helped naïve B cells and differentially modulated isotype switching while Th1-like subset (CXCR3+) did not provide adequate help to naïve B cells (described circulating Tfh-like cell subsets are summarized in Table 1). The presence of blood central memory CD4 T cells expressing CXCR5 and CXCL13 and efficiently induced plasma cell differentiation and antibody secretion further supported the existence of cTfh cells [30].
Studies of memory cTfh cells in the context of HIV infection have been scarce but they have suggested that the capacity of these cells to provide B-cell help might be compromised [31–33, 98]. Indeed, Pallikkuth et al. assessed H1N1/09 influenza responses induced following vaccination in HIV-uninfected and HIV infected individuals under ART and showed that half of HIV-infected vaccinees did not respond to the immunization. Based on this observation, the authors assessed cTfh-cell capacity to produce IL-21, CXCL-13 and express ICOS, and concluded that cTfh cells from non-responders were functionally impaired (reduced IL-21 secretion and ICOS expression) [31].
A more recent study identified a population of memory CD4+CXCR5+ T cells (CCR6hiPD-1hi) that produced IL-21 and supported B cell differentiation [33]. This study found that the frequency of these cells was decreased in treatment naïve HIV infected individuals but was recovered after antiretroviral therapy. Additionally, this study also found decreased antibody production and a reduced frequency of IL-2, IL-17 and IL-21 producing CD4+CXCR5+CCR6hiPD-1hi cells in HIV infected when compared to uninfected controls. The relationship between these circulating Tfh-like cells and those previously described by other studies [29, 30, 32,] is unknown. However, analysis of Tfh-associated genes showed that these cells more closely resembled non-Tfh memory cells as opposed to GC Tfh cells [33].
In addition, Locci et al. showed that memory CXCR5+CXCR3−PD-1+ CD4+ T cells were closely related to GC Tfh cells based on gene expression and cytokine profiles and their capacity to provide B-cell help [32]. Interestingly, the frequency of these cells positively correlated with the development of bNAbs in HIV-infected subjects [32], suggesting that the frequency of cTfh cells could be indicative of enhanced and probably efficient GC responses. In contrast, Boswell et al. showed that the frequencies of cTfh cells (defined by CXCR5+CCR6hiPD-1hi expression) in untreated HIV infected did not correlate with the frequencies of memory B cells or with Ig neutralizing activity [33]. This lack of concordance between the two studies likely arises from differences in the patient samples studied as well as the markers of cTfh used.
Taken together, the use of cTfh-cell frequency and function as an indirect marker of GC Tfh cells or ongoing GC reactions represents a very interesting tool, but will require additional investigation in order to improve their definition and harmonize their assessment.
Conclusion
The past few years have witnessed considerable findings with regards to Tfh cells and HIV pathogenesis. However, it is clear that intense investigation will be needed in order to better understand the mechanisms in charge of both Tfh and cTfh cell dysfunctions during the course of HIV infection and to determine the role of these cells during HIV infection. The progressive characterization of Tfh and cTfh cells will also be highly useful to elucidate the impact of HIV infection on the generation of efficient HIV-specific B-cell responses. Interestingly, as recently suggested [32], cTfh cells might be valuable indicators of vaccine-induced B-cell responses [31, 32, 98, 99].
Tfh cells serve as the major CD4 compartment for HIV-1 infection, replication and production. Yet, their role in establishing and forming part of the latent reservoir remains to be elucidated.
While Tfh-cell frequencies are increased during the viremic phase of HIV infection, Tfh cells are functionally impaired and cannot provide adequate B-cell help.
The presence of blood circulating memory Tfh-like cells has recently been described. Their actual relationship to GC Tfh cells still requires further investigation but a population of these cells has been shown to be closely related phenotypically and functionally to GC Tfh cells and their frequency correlated with the development of potent bNAbs in HIV-infected individuals.
Recent studies have suggested that the function of circulating memory Tfh cells might be impaired in HIV infected subjects but this requires further investigation.
Acknowledgments
This work was funded in part by the Swiss National Science Foundation and by amfAR 10867155-RKVA and NIH-1RO1AI106482-01AI.
Footnotes
The authors declare no competing financial interests.
References
- 1.Crotty S. Follicular helper CD4 T cells (TFH) Annu Rev Immunol. 2011;29:621–663. doi: 10.1146/annurev-immunol-031210-101400. [DOI] [PubMed] [Google Scholar]
- 2.Victora GD, Nussenzweig MC. Germinal centers. Annu Rev Immunol. 2012;30:429–457. doi: 10.1146/annurev-immunol-020711-075032. [DOI] [PubMed] [Google Scholar]
- 3.Rolf J, Fairfax K, Turner M. Signaling pathways in T follicular helper cells. J Immunol. 2010;184:6563–6568. doi: 10.4049/jimmunol.1000202. [DOI] [PubMed] [Google Scholar]
- 4.Victora GD, Schwickert TA, Fooksman DR, Kamphorst AO, Meyer-Hermann M, Dustin ML, Nussenzweig MC. Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell. 2010;143:592–605. doi: 10.1016/j.cell.2010.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zotos D, Coquet JM, Zhang Y, Light A, D’Costa K, Kallies A, Corcoran LM, Godfrey DI, Toellner KM, Smyth MJ, et al. IL-21 regulates germinal center B cell differentiation and proliferation through a B cell-intrinsic mechanism. J Exp Med. 2010;207:365–378. doi: 10.1084/jem.20091777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kim CH, Rott LS, Clark-Lewis I, Campbell DJ, Wu L, Butcher EC. Subspecialization of CXCR5+ T cells: B helper activity is focused in a germinal center-localized subset of CXCR5+ T cells. J Exp Med. 2001;193:1373–1381. doi: 10.1084/jem.193.12.1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Schaerli P, Willimann K, Lang AB, Lipp M, Loetscher P, Moser B. CXC chemokine receptor 5 expression defines follicular homing T cells with B cell helper function. J Exp Med. 2000;192:1553–1562. doi: 10.1084/jem.192.11.1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Breitfeld D, Ohl L, Kremmer E, Ellwart J, Sallusto F, Lipp M, Forster R. Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med. 2000;192:1545–1552. doi: 10.1084/jem.192.11.1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Fazilleau N, Mark L, McHeyzer-Williams LJ, McHeyzer-Williams MG. Follicular helper T cells: lineage and location. Immunity. 2009;30:324–335. doi: 10.1016/j.immuni.2009.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Fazilleau N, McHeyzer-Williams LJ, Rosen H, McHeyzer-Williams MG. The function of follicular helper T cells is regulated by the strength of T cell antigen receptor binding. Nat Immunol. 2009;10:375–384. doi: 10.1038/ni.1704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Good-Jacobson KL, Shlomchik MJ. Plasticity and heterogeneity in the generation of memory B cells and long-lived plasma cells: the influence of germinal center interactions and dynamics. J Immunol. 2010;185:3117–3125. doi: 10.4049/jimmunol.1001155. [DOI] [PubMed] [Google Scholar]
- 12.Goodnow CC, Vinuesa CG, Randall KL, Mackay F, Brink R. Control systems and decision making for antibody production. Nat Immunol. 2010;11:681–688. doi: 10.1038/ni.1900. [DOI] [PubMed] [Google Scholar]
- 13.McHeyzer-Williams M, Okitsu S, Wang N, McHeyzer-Williams L. Molecular programming of B cell memory. Nat Rev Immunol. 2012;12:24–34. doi: 10.1038/nri3128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tangye SG, Tarlinton DM. Memory B cells: effectors of long-lived immune responses. Eur J Immunol. 2009;39:2065–2075. doi: 10.1002/eji.200939531. [DOI] [PubMed] [Google Scholar]
- 15.Ackerman ME, Alter G. Opportunities to exploit non-neutralizing HIV-specific antibody activity. Curr HIV Res. 2013;11:365–377. doi: 10.2174/1570162x113116660058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Forthal D, Hope TJ, Alter G. New paradigms for functional HIV-specific nonneutralizing antibodies. Curr Opin HIV AIDS. 2013;8:393–401. doi: 10.1097/COH.0b013e328363d486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lai JI, Licht AF, Dugast AS, Suscovich T, Choi I, Bailey-Kellogg C, Alter G, Ackerman ME. Divergent antibody subclass and specificity profiles but not protective HLA-B alleles are associated with variable antibody effector function among HIV-1 controllers. J Virol. 2014;88:2799–2809. doi: 10.1128/JVI.03130-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Excler JL, Tomaras GD, Russell ND. Novel directions in HIV-1 vaccines revealed from clinical trials. Curr Opin HIV AIDS. 2013;8:421–431. doi: 10.1097/COH.0b013e3283632c26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hessell AJ, Hangartner L, Hunter M, Havenith CE, Beurskens FJ, Bakker JM, Lanigan CM, Landucci G, Forthal DN, Parren PW, et al. Fc receptor but not complement binding is important in antibody protection against HIV. Nature. 2007;449:101–104. doi: 10.1038/nature06106. [DOI] [PubMed] [Google Scholar]
- 20.Ackerman ME, Crispin M, Yu X, Baruah K, Boesch AW, Harvey DJ, Dugast AS, Heizen EL, Ercan A, Choi I, et al. Natural variation in Fc glycosylation of HIV-specific antibodies impacts antiviral activity. J Clin Invest. 2013;123:2183–2192. doi: 10.1172/JCI65708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chtanova T, Tangye SG, Newton R, Frank N, Hodge MR, Rolph MS, Mackay CR. T follicular helper cells express a distinctive transcriptional profile, reflecting their role as non-Th1/Th2 effector cells that provide help for B cells. J Immunol. 2004;173:68–78. doi: 10.4049/jimmunol.173.1.68. [DOI] [PubMed] [Google Scholar]
- 22.Johnston RJ, Poholek AC, DiToro D, Yusuf I, Eto D, Barnett B, Dent AL, Craft J, Crotty S. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science. 2009;325:1006–1010. doi: 10.1126/science.1175870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ma CS, Deenick EK, Batten M, Tangye SG. The origins, function, and regulation of T follicular helper cells. J Exp Med. 2012;209:1241–1253. doi: 10.1084/jem.20120994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nutt SL, Tarlinton DM. Germinal center B and follicular helper T cells: siblings, cousins or just good friends? Nat Immunol. 2011;12:472–477. doi: 10.1038/ni.2019. [DOI] [PubMed] [Google Scholar]
- 25.Liu X, Chen X, Zhong B, Wang A, Wang X, Chu F, Nurieva RI, Yan X, Chen P, van der Flier LG, et al. Transcription factor achaete-scute homologue 2 initiates follicular T-helper-cell development. Nature. 2014;507:513–518. doi: 10.1038/nature12910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yu D, Rao S, Tsai LM, Lee SK, He Y, Sutcliffe EL, Srivastava M, Linterman M, Zheng L, Simpson N, et al. The transcriptional repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity. 2009;31:457–468. doi: 10.1016/j.immuni.2009.07.002. [DOI] [PubMed] [Google Scholar]
- 27.Bossaller L, Burger J, Draeger R, Grimbacher B, Knoth R, Plebani A, Durandy A, Baumann U, Schlesier M, Welcher AA, et al. ICOS deficiency is associated with a severe reduction of CXCR5+CD4 germinal center Th cells. J Immunol. 2006;177:4927–4932. doi: 10.4049/jimmunol.177.7.4927. [DOI] [PubMed] [Google Scholar]
- 28.Simpson N, Gatenby PA, Wilson A, Malik S, Fulcher DA, Tangye SG, Manku H, Vyse TJ, Roncador G, Huttley GA, 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. doi: 10.1002/art.25032. [DOI] [PubMed] [Google Scholar]
- 29.Morita R, Schmitt N, Bentebibel SE, Ranganathan R, Bourdery L, Zurawski G, Foucat E, Dullaers M, Oh S, Sabzghabaei N, et al. Human blood CXCR5(+)CD4(+) T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity. 2011;34:108–121. doi: 10.1016/j.immuni.2010.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chevalier N, Jarrossay D, Ho E, Avery DT, Ma CS, Yu D, Sallusto F, Tangye SG, Mackay CR. CXCR5 expressing human central memory CD4 T cells and their relevance for humoral immune responses. J Immunol. 2011;186:5556–5568. doi: 10.4049/jimmunol.1002828. [DOI] [PubMed] [Google Scholar]
- 31.Pallikkuth S, Parmigiani A, Silva SY, George VK, Fischl M, Pahwa R, Pahwa S. Impaired peripheral blood T-follicular helper cell function in HIV-infected nonresponders to the 2009 H1N1/09 vaccine. Blood. 2012;120:985–993. doi: 10.1182/blood-2011-12-396648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32*.Locci M, Havenar-Daughton C, Landais E, Wu J, Kroenke MA, Arlehamn CL, Su LF, Cubas R, Davis MM, Sette A, et al. Human Circulating PD-1(+)CXCR3(−)CXCR5(+) Memory Tfh Cells Are Highly Functional and Correlate with Broadly Neutralizing HIV Antibody Responses. Immunity. 2013;39:758–769. doi: 10.1016/j.immuni.2013.08.031. This study correlates the frequency of blood circulating memory Tfh cells with the development of potent broadly neuatrlizing antibodies against HIV. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Boswell KL, Paris R, Boritz E, Ambrozak D, Yamamoto T, Darko S, Wloka K, Wheatley A, Narpala S, McDermott A, et al. Loss of circulating CD4 T cells with B cell helper function during chronic HIV infection. PLoS Pathog. 2014;10:e1003853. doi: 10.1371/journal.ppat.1003853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34*.Perreau M, Savoye AL, De Crignis E, Corpataux JM, Cubas R, Haddad EK, 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. doi: 10.1084/jem.20121932. This study demonstrates that Tfh-cell population contained the highest percentage of CD4 T cells harboring HIV DNA and was the most efficient in supporting productive infection in vitro. Furthermore, only the percentage of Tfh cells correlated with plasma viremia levels. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lindqvist M, van Lunzen J, Soghoian DZ, Kuhl BD, Ranasinghe S, Kranias G, Flanders MD, Cutler S, Yudanin N, Muller MI, et al. Expansion of HIV-specific T follicular helper cells in chronic HIV infection. J Clin Invest. 2012;122:3271–3280. doi: 10.1172/JCI64314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Petrovas C, Yamamoto T, Gerner MY, Boswell KL, Wloka K, Smith EC, Ambrozak DR, Sandler NG, Timmer KJ, Sun X, et al. CD4 T follicular helper cell dynamics during SIV infection. J Clin Invest. 2012;122:3281–3294. doi: 10.1172/JCI63039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hong JJ, Amancha PK, Rogers K, Ansari AA, Villinger F. Spatial alterations between CD4(+) T follicular helper, B, and CD8(+) T cells during simian immunodeficiency virus infection: T/B cell homeostasis, activation, and potential mechanism for viral escape. J Immunol. 2012;188:3247–3256. doi: 10.4049/jimmunol.1103138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Xu Y, Weatherall C, Bailey M, Alcantara S, De Rose R, Estaquier J, Wilson K, Suzuki K, Corbeil J, Cooper DA, 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. doi: 10.1128/JVI.02497-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kim HJ, Verbinnen B, Tang X, Lu L, Cantor H. Inhibition of follicular T-helper cells by CD8(+) regulatory T cells is essential for self tolerance. Nature. 2010;467:328–332. doi: 10.1038/nature09370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40*.Cubas RA, Mudd JC, Savoye AL, Perreau M, van Grevenynghe J, Metcalf T, Connick E, Meditz A, Freeman GJ, Abesada-Terk G, Jr, et al. Inadequate T follicular cell help impairs B cell immunity during HIV infection. Nat Med. 2013;19:494–499. doi: 10.1038/nm.3109. This study demonstrates that the function of T follicular helper cells is abnormal in HIV-infected individuals. These results may help to partly explain why B-cell and antibody responses are impaired in HIV infection. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ozaki K, Spolski R, Feng CG, Qi CF, Cheng J, Sher A, Morse HC, 3rd, Liu C, Schwartzberg PL, Leonard WJ. A critical role for IL-21 in regulating immunoglobulin production. Science. 2002;298:1630–1634. doi: 10.1126/science.1077002. [DOI] [PubMed] [Google Scholar]
- 42.Avery DT, Deenick EK, Ma CS, Suryani S, Simpson N, Chew GY, Chan TD, Palendira U, Bustamante J, Boisson-Dupuis S, et al. B cell-intrinsic signaling through IL-21 receptor and STAT3 is required for establishing long-lived antibody responses in humans. J Exp Med. 2010;207:155–171. doi: 10.1084/jem.20091706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tangye SG, Ma CS, Brink R, Deenick EK. The good, the bad and the ugly - TFH cells in human health and disease. Nat Rev Immunol. 2013;13:412–426. doi: 10.1038/nri3447. [DOI] [PubMed] [Google Scholar]
- 44.King C, Tangye SG, Mackay CR. T follicular helper (TFH) cells in normal and dysregulated immune responses. Annu Rev Immunol. 2008;26:741–766. doi: 10.1146/annurev.immunol.26.021607.090344. [DOI] [PubMed] [Google Scholar]
- 45.Ma CS, Deenick EK. Human T follicular helper (Tfh) cells and disease. Immunol Cell Biol. 2014;92:64–71. doi: 10.1038/icb.2013.55. [DOI] [PubMed] [Google Scholar]
- 46.Pratama A, Vinuesa CG. Control of TFH cell numbers: why and how? Immunol Cell Biol. 2014;92:40–48. doi: 10.1038/icb.2013.69. [DOI] [PubMed] [Google Scholar]
- 47.Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beilman GJ, Nguyen PL, Khoruts A, Larson M, Haase AT, et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med. 2004;200:749–759. doi: 10.1084/jem.20040874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Embretson J, Zupancic M, Ribas JL, Burke A, Racz P, Tenner-Racz K, Haase AT. Massive covert infection of helper T lymphocytes and macrophages by HIV during the incubation period of AIDS. Nature. 1993;362:359–362. doi: 10.1038/362359a0. [DOI] [PubMed] [Google Scholar]
- 49.Pantaleo G, Graziosi C, Butini L, Pizzo PA, Schnittman SM, Kotler DP, Fauci AS. Lymphoid organs function as major reservoirs for human immunodeficiency virus. Proc Natl Acad Sci U S A. 1991;88:9838–9842. doi: 10.1073/pnas.88.21.9838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Pantaleo G, Graziosi C, Demarest JF, Butini L, Montroni M, Fox CH, Orenstein JM, Kotler DP, Fauci AS. HIV infection is active and progressive in lymphoid tissue during the clinically latent stage of disease. Nature. 1993;362:355–358. doi: 10.1038/362355a0. [DOI] [PubMed] [Google Scholar]
- 51.Chakrabarti L, Isola P, Cumont MC, Claessens-Maire MA, Hurtrel M, Montagnier L, Hurtrel B. Early stages of simian immunodeficiency virus infection in lymph nodes. Evidence for high viral load and successive populations of target cells. Am J Pathol. 1994;144:1226–1237. [PMC free article] [PubMed] [Google Scholar]
- 52.Hufert FT, van Lunzen J, Janossy G, Bertram S, Schmitz J, Haller O, Racz P, von Laer D. Germinal centre CD4+ T cells are an important site of HIV replication in vivo. AIDS. 1997;11:849–857. doi: 10.1097/00002030-199707000-00003. [DOI] [PubMed] [Google Scholar]
- 53.Borrow P, Lewicki H, Hahn BH, Shaw GM, Oldstone MB. Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J Virol. 1994;68:6103–6110. doi: 10.1128/jvi.68.9.6103-6110.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, Farthing C, Ho DD. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol. 1994;68:4650–4655. doi: 10.1128/jvi.68.7.4650-4655.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Connick E, Mattila T, Folkvord JM, Schlichtemeier R, Meditz AL, Ray MG, McCarter MD, Mawhinney S, Hage A, White C, et al. CTL fail to accumulate at sites of HIV-1 replication in lymphoid tissue. J Immunol. 2007;178:6975–6983. doi: 10.4049/jimmunol.178.11.6975. [DOI] [PubMed] [Google Scholar]
- 56.Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, Hermankova M, Chadwick K, Margolick J, Quinn TC, et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature. 1997;387:183–188. doi: 10.1038/387183a0. [DOI] [PubMed] [Google Scholar]
- 57.Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, Baseler M, Lloyd AL, Nowak MA, Fauci AS. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci U S A. 1997;94:13193–13197. doi: 10.1073/pnas.94.24.13193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R, et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science. 1997;278:1295–1300. doi: 10.1126/science.278.5341.1295. [DOI] [PubMed] [Google Scholar]
- 59.Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, Spina CA, Richman DD. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science. 1997;278:1291–1295. doi: 10.1126/science.278.5341.1291. [DOI] [PubMed] [Google Scholar]
- 60.Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, Kovacs C, Gange SJ, Siliciano RF. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med. 2003;9:727–728. doi: 10.1038/nm880. [DOI] [PubMed] [Google Scholar]
- 61.Chun TW, Finzi D, Margolick J, Chadwick K, Schwartz D, Siliciano RF. In vivo fate of HIV-1-infected T cells: quantitative analysis of the transition to stable latency. Nat Med. 1995;1:1284–1290. doi: 10.1038/nm1295-1284. [DOI] [PubMed] [Google Scholar]
- 62.Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, Boucher G, Boulassel MR, Ghattas G, Brenchley JM, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15:893–900. doi: 10.1038/nm.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Mascola JR, Montefiori DC. The role of antibodies in HIV vaccines. Annu Rev Immunol. 2010;28:413–444. doi: 10.1146/annurev-immunol-030409-101256. [DOI] [PubMed] [Google Scholar]
- 64.Pantophlet R, Burton DR. GP120: target for neutralizing HIV-1 antibodies. Annu Rev Immunol. 2006;24:739–769. doi: 10.1146/annurev.immunol.24.021605.090557. [DOI] [PubMed] [Google Scholar]
- 65.Kwong PD, Mascola JR. Human antibodies that neutralize HIV-1: identification, structures, and B cell ontogenies. Immunity. 2012;37:412–425. doi: 10.1016/j.immuni.2012.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Doria-Rose NA, Klein RM, Manion MM, O’Dell S, Phogat A, Chakrabarti B, Hallahan CW, Migueles SA, Wrammert J, Ahmed R, et al. Frequency and phenotype of human immunodeficiency virus envelope-specific B cells from patients with broadly cross-neutralizing antibodies. J Virol. 2009;83:188–199. doi: 10.1128/JVI.01583-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Gray ES, Madiga MC, Hermanus T, Moore PL, Wibmer CK, Tumba NL, Werner L, Mlisana K, Sibeko S, Williamson C, et al. The neutralization breadth of HIV-1 develops incrementally over four years and is associated with CD4+ T cell decline and high viral load during acute infection. J Virol. 2011;85:4828–4840. doi: 10.1128/JVI.00198-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Gray ES, Moody MA, Wibmer CK, Chen X, Marshall D, Amos J, Moore PL, Foulger A, Yu JS, Lambson B, et al. Isolation of a monoclonal antibody that targets the alpha-2 helix of gp120 and represents the initial autologous neutralizing-antibody response in an HIV-1 subtype C-infected individual. J Virol. 2011;85:7719–7729. doi: 10.1128/JVI.00563-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Mikell I, Sather DN, Kalams SA, Altfeld M, Alter G, Stamatatos L. Characteristics of the earliest cross-neutralizing antibody response to HIV-1. PLoS Pathog. 2011;7:e1001251. doi: 10.1371/journal.ppat.1001251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Starcich BR, Hahn BH, Shaw GM, McNeely PD, Modrow S, Wolf H, Parks ES, Parks WP, Josephs SF, Gallo RC, et al. Identification and characterization of conserved and variable regions in the envelope gene of HTLV-III/LAV, the retrovirus of AIDS. Cell. 1986;45:637–648. doi: 10.1016/0092-8674(86)90778-6. [DOI] [PubMed] [Google Scholar]
- 71.Wyatt R, Kwong PD, Desjardins E, Sweet RW, Robinson J, Hendrickson WA, Sodroski JG. The antigenic structure of the HIV gp120 envelope glycoprotein. Nature. 1998;393:705–711. doi: 10.1038/31514. [DOI] [PubMed] [Google Scholar]
- 72.Kwong PD, Doyle ML, Casper DJ, Cicala C, Leavitt SA, Majeed S, Steenbeke TD, Venturi M, Chaiken I, Fung M, et al. HIV-1 evades antibody-mediated neutralization through conformational masking of receptor-binding sites. Nature. 2002;420:678–682. doi: 10.1038/nature01188. [DOI] [PubMed] [Google Scholar]
- 73.Lane HC, Masur H, Edgar LC, Whalen G, Rook AH, Fauci AS. Abnormalities of B-cell activation and immunoregulation in patients with the acquired immunodeficiency syndrome. N Engl J Med. 1983;309:453–458. doi: 10.1056/NEJM198308253090803. [DOI] [PubMed] [Google Scholar]
- 74.Vago L, Antonacci MC, Cristina S, Parravicini C, Lazzarin A, Moroni M, Negri C, Uberti-Foppa C, Musicco M, Costanzi G. Morphogenesis, evolution and prognostic significance of lymphatic tissue lesions in HIV infection. Appl Pathol. 1989;7:298–309. [PubMed] [Google Scholar]
- 75.De Milito A. B lymphocyte dysfunctions in HIV infection. Curr HIV Res. 2004;2:11–21. doi: 10.2174/1570162043485068. [DOI] [PubMed] [Google Scholar]
- 76.Cagigi A, Nilsson A, De Milito A, Chiodi F. B cell immunopathology during HIV-1 infection: lessons to learn for HIV-1 vaccine design. Vaccine. 2008;26:3016–3025. doi: 10.1016/j.vaccine.2007.11.063. [DOI] [PubMed] [Google Scholar]
- 77.Moir S, Fauci AS. B cells in HIV infection and disease. Nat Rev Immunol. 2009;9:235–245. doi: 10.1038/nri2524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Moir S, Buckner CM, Ho J, Wang W, Chen J, Waldner AJ, Posada JG, Kardava L, O’Shea MA, Kottilil S, et al. B cells in early and chronic HIV infection: evidence for preservation of immune function associated with early initiation of antiretroviral therapy. Blood. 2010;116:5571–5579. doi: 10.1182/blood-2010-05-285528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.van Grevenynghe J, Cubas RA, Noto A, DaFonseca S, He Z, Peretz Y, Filali-Mouhim A, Dupuy FP, Procopio FA, Chomont N, et al. Loss of memory B cells during chronic HIV infection is driven by Foxo3a- and TRAIL-mediated apoptosis. J Clin Invest. 2011;121:3877–3888. doi: 10.1172/JCI59211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Morris L, Binley JM, Clas BA, Bonhoeffer S, Astill TP, Kost R, Hurley A, Cao Y, Markowitz M, Ho DD, et al. HIV-1 antigen-specific and -nonspecific B cell responses are sensitive to combination antiretroviral therapy. J Exp Med. 1998;188:233–245. doi: 10.1084/jem.188.2.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O’Shea MA, Roby G, Kottilil S, Arthos J, Proschan MA, et al. Evidence for HIV-associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV-infected viremic individuals. J Exp Med. 2008;205:1797–1805. doi: 10.1084/jem.20072683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wollenberg I, Agua-Doce A, Hernandez A, Almeida C, Oliveira VG, Faro J, Graca L. Regulation of the germinal center reaction by Foxp3+ follicular regulatory T cells. J Immunol. 2011;187:4553–4560. doi: 10.4049/jimmunol.1101328. [DOI] [PubMed] [Google Scholar]
- 83.Chung Y, Tanaka S, Chu F, Nurieva RI, Martinez GJ, Rawal S, Wang YH, Lim H, Reynolds JM, Zhou XH, et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat Med. 2011;17:983–988. doi: 10.1038/nm.2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lim HW, Hillsamer P, Kim CH. Regulatory T cells can migrate to follicles upon T cell activation and suppress GC-Th cells and GC-Th cell-driven B cell responses. J Clin Invest. 2004;114:1640–1649. doi: 10.1172/JCI22325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Tsuji M, Komatsu N, Kawamoto S, Suzuki K, Kanagawa O, Honjo T, Hori S, Fagarasan S. Preferential generation of follicular B helper T cells from Foxp3+ T cells in gut Peyer’s patches. Science. 2009;323:1488–1492. doi: 10.1126/science.1169152. [DOI] [PubMed] [Google Scholar]
- 86.Linterman MA, Pierson W, Lee SK, Kallies A, Kawamoto S, Rayner TF, Srivastava M, Divekar DP, Beaton L, Hogan JJ, et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. 2011;17:975–982. doi: 10.1038/nm.2425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sage PT, Francisco LM, Carman CV, Sharpe AH. The receptor PD-1 controls follicular regulatory T cells in the lymph nodes and blood. Nat Immunol. 2013;14:152–161. doi: 10.1038/ni.2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.De Milito A, Nilsson A, Titanji K, Thorstensson R, Reizenstein E, Narita M, Grutzmeier S, Sonnerborg A, Chiodi F. Mechanisms of hypergammaglobulinemia and impaired antigen-specific humoral immunity in HIV-1 infection. Blood. 2004;103:2180–2186. doi: 10.1182/blood-2003-07-2375. [DOI] [PubMed] [Google Scholar]
- 89.Titanji K, De Milito A, Cagigi A, Thorstensson R, Grutzmeier S, Atlas A, Hejdeman B, Kroon FP, Lopalco L, Nilsson A, et al. Loss of memory B cells impairs maintenance of long-term serologic memory during HIV-1 infection. Blood. 2006;108:1580–1587. doi: 10.1182/blood-2005-11-013383. [DOI] [PubMed] [Google Scholar]
- 90.Hart M, Steel A, Clark SA, Moyle G, Nelson M, Henderson DC, Wilson R, Gotch F, Gazzard B, Kelleher P. Loss of discrete memory B cell subsets is associated with impaired immunization responses in HIV-1 infection and may be a risk factor for invasive pneumococcal disease. J Immunol. 2007;178:8212–8220. doi: 10.4049/jimmunol.178.12.8212. [DOI] [PubMed] [Google Scholar]
- 91.Hale JS, Youngblood B, Latner DR, Mohammed AU, Ye L, Akondy RS, Wu T, Iyer SS, Ahmed R. Distinct memory CD4+ T cells with commitment to T follicular helper- and T helper 1-cell lineages are generated after acute viral infection. Immunity. 2013;38:805–817. doi: 10.1016/j.immuni.2013.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Liu X, Yan X, Zhong B, Nurieva RI, Wang A, Wang X, Martin-Orozco N, Wang Y, Chang SH, Esplugues E, et al. Bcl6 expression specifies the T follicular helper cell program in vivo. J Exp Med. 2012;209:1841–1852. S1841–1824. doi: 10.1084/jem.20120219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Luthje K, Kallies A, Shimohakamada Y, Belz GT, Light A, Tarlinton DM, Nutt SL. The development and fate of follicular helper T cells defined by an IL-21 reporter mouse. Nat Immunol. 2012;13:491–498. doi: 10.1038/ni.2261. [DOI] [PubMed] [Google Scholar]
- 94.Pepper M, Pagan AJ, Igyarto BZ, Taylor JJ, Jenkins MK. Opposing signals from the Bcl6 transcription factor and the interleukin-2 receptor generate T helper 1 central and effector memory cells. Immunity. 2011;35:583–595. doi: 10.1016/j.immuni.2011.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Weber JP, Fuhrmann F, Hutloff A. T-follicular helper cells survive as long-term memory cells. Eur J Immunol. 2012;42:1981–1988. doi: 10.1002/eji.201242540. [DOI] [PubMed] [Google Scholar]
- 96.Choi YS, Yang JA, Yusuf I, Johnston RJ, Greenbaum J, Peters B, Crotty S. Bcl6 expressing follicular helper CD4 T cells are fate committed early and have the capacity to form memory. J Immunol. 2013;190:4014–4026. doi: 10.4049/jimmunol.1202963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Ravirajan CT, Rahman MA, Papadaki L, Griffiths MH, Kalsi J, Martin AC, Ehrenstein MR, Latchman DS, Isenberg DA. Genetic, structural and functional properties of an IgG DNA-binding monoclonal antibody from a lupus patient with nephritis. Eur J Immunol. 1998;28:339–350. doi: 10.1002/(SICI)1521-4141(199801)28:01<339::AID-IMMU339>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
- 98.Pallikkuth S, Pilakka Kanthikeel S, Silva SY, Fischl M, Pahwa R, Pahwa S. Upregulation of IL-21 receptor on B cells and IL-21 secretion distinguishes novel 2009 H1N1 vaccine responders from nonresponders among HIV-infected persons on combination antiretroviral therapy. J Immunol. 2011;186:6173–6181. doi: 10.4049/jimmunol.1100264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Bentebibel SE, Lopez S, Obermoser G, Schmitt N, Mueller C, Harrod C, Flano E, Mejias A, Albrecht RA, Blankenship D, et al. Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination. Sci Transl Med. 2013;5:176ra132. doi: 10.1126/scitranslmed.3005191. [DOI] [PMC free article] [PubMed] [Google Scholar]