Abstract
Interleukin (IL)-21 is a member of a family of cytokines which includes IL-2, IL-4, IL-7, IL-9 and IL-15 all of which utilize a common γ chain in their individual receptor complexes for delivering intracellular signals in their target cells. IL-21 is produced by CD4+ T cells, in particular T follicular helper cells, and is critically important in the regulation and maintenance of T cells and B cells in innate and adaptive immunity. Effects of IL-21 are pleiotropic, owing to the broad cellular distribution of the IL-21 receptor and it plays a critical role in T cell-dependent and independent human B cell differentiation for generating humoral immune responses. This chapter reviews current knowledge about the importance of IL-21 and IL-21 receptor interaction in human B cell responses, immune defects of B cells and IL-21 in HIV infection and the potential applicability of IL-21 in vaccines/immunotherapeutic approaches to augment relevant immune responses.
Keywords: IL-21 and IL-21 receptor, IL-21 and B cells, B cells and HIV, vaccines and IL-21
Introduction
Cytokines play a pivotal role in modulating immune responses. Agonist as well as antagonist therapies directed at cytokines, cytokine receptors or signaling pathways are being intensely investigated with some entering the clinical arena. Interleukin (IL)-21 is a member of the family of cytokines IL-2, IL-4, IL-7, IL-9, IL-15 that all utilize CD132, the common γ chain (γc), in their receptor complexes for delivering intracellular signals in their target cells.1,2 Among these, IL-21 has the most pleiotropic activities because the IL-21 receptor (IL-21R) is expressed on a broad range of cells.1–3 Production of IL-21 however is restricted predominantly to CD4 T cells, mainly to T follicular helper cells (TFH) and to some extent Th17 and other cells.1,4–7 Advances in the knowledge about the biology of IL-21 and its immunomodulatory activity have stimulated interest in this cytokine in different clinical and experimental settings.
There is now evidence for a critical role of IL-21 in protection against several virus infections.8–14 For example, in chronic lymphocytic choriomeningitis virus infection of mice, higher levels of IL-21 produced by virus-specific CD4 T cells were demonstrated to be essential for sustaining antiviral effector functions of CD8 T cells and preventing them from immune exhaustion.8,9,13 In chronic hepatitis B virus infected patients, increase in serum concentrations of IL-21 was associated with activation of the antiviral immune responses that could control hepatitis B virus replication.10 During acute hepatitis B virus infection, IL-21 mRNA levels were increased in patients who cleared the virus compared with patients who did not.12 In hepatitis C virus infection as well, a recent study has demonstrated a possible role of IL-21 in distinguishing patients with hepatitis C virus clearance from those with hepatitis C virus persistence.11. In patients infected with the human immunodeficiency virus (HIV), existing data suggest that IL-21 could play an important role in modulating T and B cell function, thereby influencing the immunodeficiency associated with it.15–21 IL-21 has been investigated as a therapeutic agent in humans for various malignancies [reviewed in3,22–24]. In rhesus macaques infected with simian immune deficiency virus (SIV),18 IL-21 was shown to have a good safety record and to be biologically active, resulting in improved immune responses. The true potential of IL-21 as a therapeutic or vaccine adjuvant for HIV-infected individuals has not yet been established.
The hallmark of HIV infection is progressive decline of CD4 T cells25–27 with the virus infecting CD4 T cells, macrophages and dendritic cells. HIV-infected patients have widespread immune dysfunction that includes immunosuppression in concert with increased inflammation and immune activation;28 this is because the virus has both direct and indirect effects that impact infected- as well as uninfected cells. Advances in antiretroviral therapy (ART) have vastly improved the lifespan of HIV-infected individuals, but it is intriguing that even with suppression of virus to below detectable levels in plasma, immune deficits can persist to varying degrees. These effects are attributed to ongoing low level virus replication in tissues as well as to gut associated microbial translocation, resulting in immune activation, inflammation and B cell related immune deficits. Thus, even though B cells are not infected, patients with chronic HIV infection manifest abnormal distribution of B cell subsets, loss of memory B cells and impaired humoral immune responses,29 and [reviewed in30,31]. Institution of ART early in the course of infection can to a large extent preserve the T and B cell immune system and reduce the persistent immune dysfunction that is noted when treatment is started later.29
Emerging data suggests that IL-21 is associated with preservation of virus control and immunity in different stages of HIV infection.15,20,21 Although correlates of immune protection in HIV infection are not well understood, cytotoxic T cells32–34 and more recently, natural killer (NK) cells have been shown to play important roles in virologic control.35,36 Treatment of HIV-infected patient cells ex vivo with IL-21 and administration of IL-21 to SIV-infected rhesus macaques can augment the expression of the cytotoxic molecules perforin and granzyme B in CD8 T cells18,19 and NK cells18,37 without significant cellular proliferation or activation. Recent data suggests that IL-21 also plays an important role in preserving B cell immunity in HIV-infected patients.16,17 Many aspects in the life of B cells are regulated by IL-21, such as proliferation, differentiation, class switching or death, depending upon the type of antigenic stimulation and accessory signals.38–42 IL-21-elicited signaling has been identified as the most important among all the γc cytokines for long-lived humoral immunity.42 In this review we summarize the biology of IL-21 and its receptor (IL-21R), their role in modulating human B cell responses and their relevance to HIV infection.
I. Interleukin-21 and IL-21R expressing cells
The IL-21 polypeptide is 131 amino acids in length and has 57% amino acid sequence homology to its murine counterpart.1,2 In humans, the IL21 gene is encoded on chromosome 4. This cytokine was first identified by Parrish-Novak et al. from the culture supernatants of activated human CD3 T cells.1 Subsequently, it was established that IL-21 was secreted predominantly by T cells, in particular by the CD4 TFH and Th17 subsets, as well as by NK T cells [reviewed in.3,43]. The human IL21R gene is located adjacent to IL4RA gene on chromosome 16. It encodes a 538 amino acid protein and its amino acid sequence is most closely related to IL-2Rβ. Human IL-21R shares 62% amino acid sequence similarity with its murine counterpart, encoded on chromosome 7.1,44 The distinct IL-21Rα chain couples with the γc to form the cytokine receptor complex.1,44 IL-21R is expressed on a broad range of cells [reviewed by,3,45], predominantly in cells of lymphoid tissues including spleen, thymus and lymph nodes. It is constitutively expressed on B cells, CD4 T cells, NK cells, macrophages, monocytes, dendritic cells as well as hematopoietic and non- hematopoietic cells such as fibroblasts, keratinocytes and intestinal epithelial cells, and less often in cells from lung and small intestine.3,44 This broad range of expression of IL-21R explains the pleiotropic effect of IL-21.
Interestingly, the level of expression of IL-21R is highest on B cells.44,46 In human B cell subsets, IL-21R is constitutively expressed on naïve B cells and germinal center (GC) B cells, with memory B cells expressing it at low levels; recent reports show that plasma cells (PCs) also express IL-21R.46,47 The surface expression of IL-21R increases on human memory B cells following their activation, eg with anti-CD40 mAb.46 IL-21 upregulates its own receptor expression on B cells by itself, or in combination with anti-CD40 stimulation.48 Thus, the development and activation-dependent regulation of IL-21R expression on the surface of B cells suggests that IL-21 has important functions in B cell development and consequently on the immune response.
II. IL-21 signaling on B cells and its relevance to human B cell development
Like other γc-dependent cytokines, the binding of IL-21 to its receptor activates the Janus family tyrosine kinases (JAK), JAK1 and JAK3 which, in turn, activate signal transducer and activator of transcription (STAT)3, and to a weaker and more transient degree, STAT1, STAT4 and STAT5.44,49,50 Current views on IL-21/IL-21R signaling in B cells are depicted in Figure 1. Results from in vitro studies using human B cell lines indicate that binding of IL-21 to IL-21R leads to strong STAT3 activation50,51 as early as 5 minutes after binding and lasts for up to 6 days.50 In B cells, IL-21 upregulates the master plasma cell transcription factor B lymphocyte induced maturation protein 1 (BLIMP-1), a transcriptional repressor that is important for plasma cell differentiation.6,39,42,52,53 BLIMP-1 is encoded by positive regulatory domain zinc finger protein 1 gene and its expression is regulated in a STAT3-dependent manner. In combination with CD40 ligation, IL-21 promotes class switch recombination, plasma cell differentiation, and immunoglobulin (Ig) production from B cells through the induction of activation-induced cytidine deaminase (AID), an essential factor for somatic hypermutation and class switch recombination. In primary human B cells as well, IL-21 induces activation of STAT3 which in turn triggers expression of BLIMP-1 and promotes Ig production.54 In addition to JAK/STAT, IL-21 also activates extracellular signal-regulated protein kinases 1/2 that are mitogen-activated protein kinases and PI3K pathways in other cell types like neoplastic cells, epithelial cells, and monocytes.55,56 These signal transduction pathways eventually modulate the transcription program within the activated cell, thus regulating its phenotype, function and fate.
Figure 1. Role of IL-21 in B cell differentiation and function.
CD4 T cells provide a fundamental help to B cell differentiation and function through a complex network of signaling pathways. The conjunct activation of CD40 and IL-21R by interaction with CD40 ligand and IL-21 secreted by CD4 T cells activates the transcription factors NF-kB and STAT3, that orchestrate the transcription of target genes responsible for B cell proliferation and differentiation into antibody-secreting plasma cells or memory B cells. These genes include: PRDM1, encoding for the transcription repressor Blimp-1, and AID. Concurrently, they promote class switch recombination and somatic hypermutation. IL-21 also induces the expression of its own receptor in activated B cells.
The significant role of IL-21/IL-21R induced B cell signaling in human B cell development and memory generation is exemplified in specific primary immunodeficiency disorders. In patients with severe combined immunodeficiency due to IL2RG/JAK3 deficiency, the B cells exhibit defective γc-dependent cytokine signaling that results in impaired B-cell responses, humoral dysfunction and significantly reduced memory B cells. This defect is not corrected if there is poor donor B-cell engraftment after hematopoietic cell transplantation, and the ex-vivo stimulation with CD40L/IL-21 induces B-cell proliferation, plasmablast differentiation, and antibody secretion only in patients with donor B cell engraftment.42 These data indicate the importance of IL-21-mediated signaling in developing long-lived humoral immunity and antibody responses.
The critical role of STAT3 downstream of IL-21 is demonstrated in patients with STAT3 mutations who exhibit dramatically reduced numbers of functional, antigen specific memory B cells with inability of IL-21 to induce naive B cells to differentiate into plasma cells. In contrast to STAT3, STAT1 deficiency has no effect on memory B cell formation in vivo or on IL-21-induced immunoglobulin secretion in vitro indicating that STAT3 plays a critical role in generating effector B cells from naive precursors in humans.52 Specifically, levels of circulating anti-tetanus IgG were found to be reduced in STAT3 mutant patients and in vitro recall experiments showed that both the overall quantity and quality of antigen specific B cells was lost in these individuals. In addition, mutations in STAT3 largely impaired IL-21-mediated B cell expansion and plasma cell differentiation with failure to upregulate BLIMP-1 in response to IL-21 and CD40L while upregulation of AID was unaffected.52 In a more recent study STAT3-deficient CD4 T cells were shown to have defective expression of IL-21 with diminished B-cell helper activity in vitro. These results indicate the important role of STAT3 in IL-21-mediated differentiation of both, T and B cells.57 Overall, these studies suggest that IL-21-mediated induction of AID and class switching is STAT3-independent, while STAT3 is critical for IL-21-induced B cell expansion, differentiation, and/ or survival and support the concept that IL-21 is indispensible for the generation and maintenance of human B cell memory.
III. Role of IL-21 in human B cell development and plasma cell differentiation
IL-21 plays a critical role in B cell fate decisions and differentiation and influences the expression of many functionally important B-cell surface molecules. Direct stimulation of B cells with IL-21 alone induces upregulation of CD19, CD21, CD10, CD20, CD22, CD95, CD9, CD86, and the IL-21R, but not CD23 or CD2758 with induction of AID without initiating somatic hypermutation.39 In contrast, stimulation of B cells with anti-CD40 alone markedly increases expression of both CD23 and CD2758 with minimal proliferation.1,6,39 The most potent and unique B cell stimulatory effects are seen with IL-21 and CD40 costimulation, unlike other B cell stimulatory cytokines like IL-2 or IL-10. The IL-21/CD40 costimulation induces expansion of both naïve and memory B cells isolated from either cord blood or peripheral blood, with inhibition of B cell death in cord blood together with augmentation of proliferation, class switch recombination, plasma cell differentiation, and Ig production. IL-21 induces IgG3 class switch in cord blood B cells and both IgG1 and IgG3 class switch in peripheral blood or splenic naïve B cells.39,53 In addition to IgG production, IL-21 is also capable of inducing IgA production from naïve cord blood or peripheral blood B cells38,39 but not IgE.39,53 CD40 and IL-21 costimulation also induces extensive proliferation of B cell subsets isolated from various anatomical sites such as human spleen or tonsil.46,59 These findings imply that IL-21 has a profound impact in direct T cell-driven stimulation of B cells.
Further support for the unique role of IL-21 in T cell-driven B cell responses was derived from experiments showing that blocking endogenously produced IL-21 following CD4 T cell activation was sufficient to significantly inhibit B cell expansion, plasma cell differentiation, and antibody production,6,60 whereas, blockade of other cytokines, specifically, IL-2, IL-4, or IL-10, did not have this effect. In addition to IgG, IgM production was also significantly reduced when IL-21 was blocked in this experimental system.60 Blockade of IL-21 production inhibited IgM production from naïve B cells and IgG production from both naïve and memory B cells, whereas IgM production by pre-switched B cells was less affected by IL-21 blockade.60 These studies indicate that IL-21 is able to costimulate plasma cell differentiation more effectively in post-switched B cells, at later stages of B cell differentiation.6,60 It was shown that purified TFH cells from tonsils induced a marked increase of Ig production by tonsillar PC, and this effect was impaired when endogenous IL-21 production was blocked, indicating the efficiency of IL-21 in inducing immunoglobulin production in differentiated PC.47 These studies strongly support the importance of IL-21 in effective plasma cell differentiation of naïve B cells and post-switched memory B cells which is a prerequisite for an effective humoral immune response.
IV. IL-21, TFH cells and Germinal Center (GC) reaction in antigen induced Ab formation
The most critical site for the generation of T dependent antibody responses is the GC within lymphoid follicles in secondary lymphoid organs (reviewed in61) wherein interaction of antigen-primed B cells with TFH drives the B cells to undergo proliferation and differentiation, with Ig isotype switching, somatic hypermutation and secretion of high affinity antibodies.43,62 The GC TFH cells are a specialized effector CD4 T helper cell subset [63,64 and reviewed65], characterized by presence of the transcriptional regulator Bcl-6, surface expression of CXCR5, ICOS and programmed cell death 1 (PD-1) molecules and secretion of IL-21.42,66–68 Following pathogen invasion or vaccination, the antigen-activated specialized CD4 T cells and B cells are recruited into the lymphoid organs and into the GC where the GC reaction is activated. Experiments conducted ex-vivo demonstrated that IL-21 is capable of driving plasma cell differentiation and IgM, IgG, and IgA antibody production from human splenic GC B cells.6 Experimental evidence indicates that initial GC formation occurs within 6 days of primary immunization, when rapidly proliferating B cells begin to appear as foci in the B cell follicles of lymph nodes and spleen. These foci later increase rapidly in size and differentiate into the mature GC reaction.61,69 Within the GC, B cells move constantly and engage in short, dynamic interactions with both TFH and antigen that enhance B cell affinity and eventually the quality of immune response.61 TFH-derived IL-21 orchestrates GC development and maintenance and many aspects of B cell differentiation and function, including development of memory B cells and long-lived plasma cells.39,43,46,53,62 Thus TFH cells have emerged as key facilitators of Ab responses and are now being studied for their role in induction of Ab against HIV and SIV in systems aimed to inform vaccine design.
Although the role of TFH-B cell interaction for B cell differentiation and function was initially described for TFH cells residing within GC, the importance of TFH cells at other sites is becoming evident.43,70,71 While some GC TFH undergo apoptosis, recent data suggests that they are also capable of migrating out from the GC72,73 to interact with B cells at the border of the T cell-B cell zone in the lymph nodes to generate a heightened memory cell response or to migrate into the peripheral circulation.71 Human CXCR5+ memory CD4 T cells in peripheral circulation share functional properties with the GC TFH cells,63,64,74–78 such as the ability to secrete IL-21 and to induce autologous naïve and memory B cell subsets to produce immunoglobulins.16,79 These peripheral CXCR5+ CD4 T cells are absent in circulation of patients with ICOS deficiency80 who also lack GC. Thus, these cells, termed peripheral TFH (pTFH), are easily accessible in peripheral blood and may serve as important biomarkers for assessing TFH function. Our findings related to pTFH following influenza vaccination in HIV-infected patients and healthy donors support this concept, and are discussed later in this review.
V. HIV infection and consequences on GC, TFH, and IL-21 responses
It has been shown that CD57+ CD4 GC T cells are one of the sites of HIV infection and replication that may play a pivotal role in the pathogenesis of HIV infection.81 In rhesus macaques, direct SIV infection of TFH has been demonstrated.82 In a recent study,83 SIV-infected rhesus macaques were followed for changes in TFH and B cells in situ as the animals progressed from uninfected, to the acute and chronic stages of infection.83 Progression of SIV infection was accompanied by an increased numbers of follicles containing GCs, as well as by increased frequency of PD-1-expressing TFH cells that correlated with accumulation of activated Ki-67+ B cells within GCs. If preferential infection of TFH is occurring in lymph nodes, these data suggest that the accumulation of TFH could initially stimulate GC formation and B cell proliferation. The absence of CD8 T cells in the GC further suggests that GC TFH could serve as reservoirs for HIV. We contend that HIV-infected TFH may not function efficiently and thus their helper function for B cells and Ab production could be impaired. These possibilities need to be evaluated.
Additional mechanisms contributing to poor Ab responses may relate to impaired B-T cell interaction. Chronic progressive HIV infection deeply alters lymph node architecture, with increased collagen deposition and interstitial fibrosis, resulting in perturbed cellular distribution and abnormal cytokine milieu [reviewed84]. Consequently, alterations in the B-CD4 T cell interactions are to be expected. Under physiological conditions, the formation of a GC is mediated by changes in the expression of the chemokine receptors CXCR4, CXCR5 and CCR7 in B cells, together with changes in the local concentration of the chemokines that bind to these receptors, such as CXCL12, CXCL13, and CCL19/21.85–87 HIV infection is associated with alteration of the levels of these soluble mediators and their receptors, including increased levels of circulating CXCL12 and CXCL13, and decreased CXCR5 expression on B cells.88 HIV-associated changes in the chemokines and their respective receptors that coordinate the GC formation and B-T cell crosstalk could impact the generation of Ab responses. Thus, together with CD4 T cell depletion and impaired TFH function, altered lymph node architecture could contribute to alterations in GC formation and contribute to impaired B cell derived Ab responses in HIV-infected individuals.
Recent studies have addressed the role of IL-21 and peripheral T cells in HIV infection and disease progression.20,21,89,90 In general these studies indicate that IL-21 plays a beneficial role in arming CD8 T cells. In a study of primary infection it was shown that IL-21-mediated T helper function is established with subsequent acquisition of CD8 T cell multifunctional traits, but this quality is rapidly lost in chronic, progressive infection.20 In elite controllers in this study, not only CD4 T cells but also CD8 T cells secreted IL-21 in response to HIV-specific stimulation and both were more polyfunctional in elite controllers compared to viremic controllers and rapid progressors. Others have also found that elevated frequencies of HIV-specific IL-21-producing CD4 T cells correlate with relative viral control, probably by promoting CD8 T cell maintenance and function.21 In general, current data suggests that loss of ability to produce IL-21 accompanies disease progression. In cross-sectional and longitudinal studies in different groups of HIV-infected patients, plasma levels of IL-21 were lower in HIV-infected individuals compared to uninfected healthy control subjects. Moreover, the levels of IL-21 correlated with CD4 T cell counts, suggesting a predominant role for CD4 T cells as a source of IL-21 or a role of IL-21 in promoting CD4 T cell survival.89,90 It has also been suggested that HIV-infected individuals have greater circulating IL-21-producing CD4 T cells compared to uninfected individuals.21 One reason for differences in results is that antigen-specific IL-21 secreting cells are generally in extremely low frequency in circulation and data derived from PMA activated cells may not reflect deficiencies of antigen specific cells. The question of IL-21 secreting peripheral T cells in HIV infection has not been conclusively established, and further studies are awaited.
Study of the role of TFH cells in HIV infection in the context of B cell function and HIV antibody responses as well as Ab responses to non-HIV vaccines has been limited. HIV-specific IL-21-producing CD4 T cells detected in blood during untreated acute and chronic HIV infection have been reported to express CXCR5 and to have an effector memory phenotype.21 A study by our group indicates an important role for peripheral TFH cells in influenza vaccine-induced antibody responses of HIV-infected patients.16 As IL-21 is produced mainly from CD4 cells, in particular from TFH,6 reconstitution of this subset of CD4 T cells following ART in HIV-infected patients may be critically important for restoring B cell function. Due to the fact that TFH-derived IL-21 orchestrates many aspects of B cell differentiation and function,39,43,46,53,62 it will be of interest to see how the overall depletion of CD4 T cells after HIV infection affects the TFH compartment of cells and how these changes affect the IL-21 levels and B cell mediated immune response during HIV infection. We have explored these questions in HIV-infected and uninfected volunteers following influenza H1N1/09 vaccination discussed below.16,17,91
VI. B cells in HIV infection
It is well known that in addition to the defects in the cell-mediated arm of immune responses, HIV infection impairs the humoral arm of the immune system. The failure of the immune system to control the virus in infected individuals is a great challenge to the treatment of HIV infection and points to the need for the development of a successful HIV vaccine. Although CD4 T cells are the primary target for HIV, the B cell compartment is also profoundly affected by HIV infection.92 B cell abnormalities, that are both phenotypic and functional, can be ascribed to three mechanisms: a direct virus-induced B cell activation, bystander B cell polyclonal activation triggered by the abnormal milieu of cytokines, HIV proteins, other soluble mediators, and impaired cross-talk between B and CD4 T helper cells. These impairments lead to apparent loss of humoral immune control over HIV and also to other pathogens.
HIV does not productively infect B cells, but replication-competent HIV virions can be isolated from the surface of B cells obtained from HIV-infected individuals.93 These virions have been shown to adhere to B cells because the complement and antibodies that opsonize such virions can be bound by CD21, that is expressed on B cell membrane.94 As a consequence of this interaction, it has been suggested that triggering of CD21 by bound virions could alter physiological B cell responses,95,96 thus contributing to B cell dysfunction during the course of HIV infection. Several HIV proteins have also been reported to directly interact with B cells, resulting in their hyperactivation, altered functionality and increased propensity to cell death. Among them, Tat, released by infected CD4 T cells, has been shown to induce surface expression of Fas (CD95) in B cells,97 that is associated with increased B cell apoptosis upon engagement with Fas ligand.98 Paradoxically, while HIV protein Nef impairs immunoglobulin class switch by repressing AID expression,99 Env promotes AID expression. This latter reaction leads to chronic polyclonal B cell activation and accumulation of nonprotective IgG and IgA.100
Several members of γc-sharing cytokines101,102 which are important in B cell homeostasis, differentiation and function undergo alterations in HIV-infected patients. For example, reduction of physiological levels of IL-2 and IL-4, that provide important proliferation and survival signals to B cells103–106 may negatively impact B cell homeostasis and survival. Higher levels of IL-7,107,108 a cytokine that promotes the proliferation of B cell precursors109 may be responsible for elevated levels of circulating B cell precursors in HIV-infected individuals.110 As discussed earlier, ability of CD4 T cells to produce IL-21, is also affected during the course of HIV infection,15,20,21,89 with consequences on B cells. Other than γc-sharing cytokines, many other soluble mediators are altered during the course of HIV infection that may affect the B cell compartment [reviewed in30,31]. Microbial translocation, that is the presence of bacteria-derived products such as lipopolysaccharide in the bloodstream due to compromised integrity of the gastrointestinal mucosa, has been suggested as a leading cause of immune activation that drives HIV disease progression.111,112 B cells express toll-like receptor 4 (TLR4)113 and CD180,114 two receptors that can sense lipopolysaccharide. Thus, increased microbial translocation could contribute to B cell hyperactivation both directly, stimulating TLR4 and CD180 on B cells, and indirectly, possibly by inducing the production of pro-inflammatory cytokines TNF and IFNα.111
Overall, HIV infection affects the B cell compartment profoundly, leading to B cell functional defects in the face of intense polyclonal B-cell activation manifested as increased circulating levels of immunoglobulins and autoantibodies,115 increased incidence of B cell malignancies,116 expansion of B-cell areas within lymphoid tissues [reviewed in117], and accumulation of terminally differentiated B cells with high expression of activation markers and inhibitory surface molecules.92,118–120 Profound changes in B cell subpopulations occur in HIV-infected persons, with accumulation of terminally-differentiated B cells, that are characterized by loss of CD20 and CD21, that are markers of resting B cells, and an increased expression of the plasmacytoid-associated markers CD27 and CD38. Consequently, HIV-infected individuals have higher levels of circulating CD20−CD21loCD27++CD38+++ plasma cells121 as compared to uninfected controls. Concurrently, a progressive depletion of peripheral CD27+ memory B cells, that already occurs during the acute phase of HIV infection,122,123 is also observed in chronic HIV-infected subjects.124 Immature/transitional B cells (CD20+CD10+CD27−) are also perturbed in HIV disease, being about three times more represented in HIV-infected subjects than in healthy controls,110 probably as a consequence of the elevated circulating levels of IL-7. Reduction of viremia with ART does not revert the loss of memory B cells,17,119,122,125–127 possibly due to residual disruption in IL-2 signaling that is required for memory B cell survival,106 while other B cell subsets tend to normalize.128,129 Several inhibitory receptors that negatively regulate B cell function are over-expressed in B cells of HIV-infected individuals. These inhibitory receptors have been suggested to play a role in the exhaustion and dysfunction of B cells, in particular the tissue-like memory B cells.120 These inhibitory receptors include: Fc receptor-like-4 (FcRL4); FcγRIIB (CD32b), a low-affinity IgG receptor; CD22 (Siglec-2), a sialic acid-binding Ig-like lectin; CD85j and CD85d, members of the leukocyte Ig-like receptor family; CD22; CD72; leukocyte-associated Ig-like receptor 1 (LAIR-1); PD-1; and cytotoxic T-lymphocyte antigen 4 (CTLA4).121 B cells from HIV-infected subjects are also characterized by increased expression of activation markers that are specific for the B cell compartment (including CD80 and CD86),130 or that are also seen in CD4 and CD8 T cells (such as the proliferation marker Ki-67 and the surface molecule CD38).30 Even though B cell activation markers are reduced by ART, a chronic state of B cell hyperactivation may persist for several years.131 More recent investigations suggest that early initiation of ART within the first year of HIV infection is associated with better preservation of humoral function.29
The changes in B cell phenotype and differentiation subsets observed in HIV-infected individuals are unmistakably associated with altered effector function.92 The observation that HIV-infected subjects accumulate immature/transitional and terminally differentiated B cells can be used to explain the overall poor B cell responses that have been reported both in vivo and ex vivo, including hyperimmunoglobulinemia, loss of responsiveness to stimuli and increased susceptibility to apoptosis.119,121,132 Similarly, the loss of memory B cells in HIV infection is associated with reduced serological memory against both T cell-dependent (such as influenza virus, measles virus and tetanus toxoid) and T cell-independent antigens (such as polysaccharide pneumococcal vaccine) and are not normalized by ART administration.127,133–135 The decreased B cell responses to vaccine immunogens have important consequences especially for HIV-infected pediatric patients,136 and strategies such as re-vaccination after ART-induced virus suppression have been adopted to overcome persistent memory B cell deficits.
VII. Anti-HIV Ab in HIV infection
Studies of natural history of HIV-infected individuals showed approximately 10–30% of HIV-infected individuals develop broadly neutralizing antibodies (bNAbs), though it usually takes three or more years post infection for such antibodies to develop. But the antibodies with outstanding breadth and potency develop only in 1% of infected individuals and they are called “elite neutralizers”.137 Monoclonal bNAbs isolated from these individuals provide more precise clues to the vulnerable regions on HIV Env that can then be targeted for vaccine design. Many bNAbs to HIV-1 gp140 have been isolated including those that bind to gp120 (b12, 2G12, PG9/PG16, HJ16 and VRC01) and others that are specific for gp41 (2F5, 4E10 and Z13),138–140, reviewed in.141–143
One of the major challenges in the HIV-1 vaccine research is the limited development of bNAbs against Env antigen. This is mainly due to the conserved nature of viral Env epitopes that are poorly immunogenic because of their complex chemical nature. Moreover, Env glycans are antigenically similar to host carbohydrates and homology of Env proteins with self proteins might induce polyreactive antibodies and lead to alterations in tolerance mechanisms. In addition, selection of virus escape mutations will lead to transient neutralizing effect of the Abs to the virus [reviewed in144–146]. Thus vaccine trials in non-human primates and humans with HIV-1 Env antigens have been unsuccessful. For the induction of protective antibodies, the focus is now directed to define ways to induce bNAbs systemically and mucosally and to understand the reason for the short lived nature of HIV-1 Env antibodies, based on a thorough understanding the B cell regulatory and tolerance pathways during humoral immune responses against HIV. Future studies need to identify traits of relevant bNAbs for their functions and in vivo regulation, understanding the ontogeny of bNAbs with specific focus towards defining the precursor B cell subsets from which bNAbs originate including identifying the checkpoints of bNAbs, and understanding how somatic hypermutation and affinity maturation impact acquisition of bNAbs functional properties. Interestingly, nearly all bNAbs that target gp120 are heavily hypermutated, and this property appears to be important for the neutralizing activity which is lost with reversion to the germline.
Design of optimal immunogens targeted at naive and memory B cell populations that can respond to HIV-1 Envelope and in this context the role of modulation of B cell function to enhance the specificity of Ab response may be an interesting approach. Recently, Mouquet et al. reported a novel and promising approach using a clade B YU-2 gp140 trimeric antigen and single-cell antibody cloning methods to obtain 189 new anti-gp140 Abs representing 51 independent B cell clones from the IgG memory B cells of 3 patients infected with HIV-1 clade A or B viruses and exhibiting broad neutralizing serologic activity.147 Development of highly functional antigen-specific memory B cell is an essential requirement for these types of approaches. It is possible that during HIV infection defective B and TFH cell function could be a significant contributing factor to prevent the development of antigen-specific memory B cells and prevent desired immune responses.
While the gold standard for HIV Ab in vaccine research has been the elicitation of bNAbs, data from the RV144 trial (reviewed in143,148) points to the role of non-neutralizing Ab with antibody dependent cellular cytotoxicity capability in affording protection from HIV acquisition. Thus efforts are being directed at not only eliciting bNAb but also to explore all other potential mechanisms of Ab mediated and cellular protection from HIV.
We have recently demonstrated cumulative defects in B cell intrinsic and extrinsic factors, IL-21/IL-21R and peripheral blood TFH cell dysfunction in the development of a H1N1/09 vaccine induced immune responses in a group of ART treated, virologically controlled HIV-infected patients.16,17,91 These studies underscore the fact that an important path to induction of long-lived systemic and mucosal antibody responses in HIV-infected individuals lies in the understanding of, and optimizing, the B-TFH cell interactions during a vaccine-induced antibody response. The role of IL-21 in elucidation of HIV Ab needs to be investigated further as a component in HIV vaccine design, owing to its potent effects on B cells; pre-clinical studies in non-human primates are showing promise as discussed below.
VIII. IL-21 and B cells in HIV infection
As discussed earlier, B cell subsets and function are disrupted in HIV infection, resulting from excessive B cell activation and impaired survival.31,106 The altered distribution includes accumulation of immature transitional B cells, activated mature cells, tissue-like exhausted and short lived plasmablasts with reduced naïve and resting memory B cells. Following effective ART, the B cell subsets do not completely revert to normal, and the CD27+ resting memory B cells remain significantly decreased in comparison to healthy uninfected donors, despite virologic control and CD4 T cell recovery.126,133 Studies of IL-21 and IL-21R in the context of B cells in HIV infection have been limited. Increased IL-21R expression has been described in B cells from HIV-1-infected patients compared to control subjects, with the highest levels in non-treated patients and an inverse correlation between IL-21R expression and percentages of circulating resting memory B cells.149 Levels of plasma soluble CD14, a marker of monocyte/macrophage activation that is increased in association with microbial translocation were correlated with high IL-21R expression. The authors of this study attributed B cell defects to microbial translocation, as TLR activation of B cells in vitro resulted in IL-21R up-regulation, and IL-21R+ memory B cells were more susceptible to spontaneous apoptosis. Lower survival of memory B cells from HIV-infected subjects has also been attributed to the disrupted IL-2 signaling leading to increased transcriptional activity of Foxo3a and increased expression of its pro-apoptotic target TRAIL. In a study by Van Grevenynghe et al,106 IL-21 could prevent the apoptosis by increasing phosphorylation of Foxo3a, which in turn downregulated the transcription of its pro-apoptotic target BimEL. These results support the concept that IL-21 could be beneficial in preserving the memory B cell compartment in HIV-infected people and thereby benefit the overall humoral immune responses.89,90
In a study of 2009/H1N1 influenza vaccine recipients our data was supportive of the role of IL-21 in promoting vaccine-induced B cell responses in a cohort of HIV-infected subjects.16,17,91 At 4 weeks post vaccination, only 50% patients developed vaccine-specific Ab titers of >1:40 and 4 fold increase in Ab titers and were designated as H1N1 vaccine responders, while the rest of the patients with H1N1 antibody titers of ≤ 1:20 units were designated as H1N1 vaccine non-responders. Healthy controls who were also given the same vaccine and were all found to be responders. Pre-vaccination frequencies of B cells, CD4 and CD8 T cell counts, and mean age were statistically equivalent between the patient groups. Several interesting observations were made in this study (summarized in Table 1). Vaccine responders manifested expansion of plasmablasts and spontaneous Ab secreting cells and at 1 week post vaccination, with development of memory B cells that could secrete Ab following ex-vivo antigen stimulation at 4 wks post vaccination. Plasma IL-21 and IL-21 secretion by PBMC following antigen stimulation increased in responders at 4 wks together with induction of IL-21R on total- and memory B cells. The upregulation of IL-21/IL-21R in the vaccine responders corresponded with development of plasmablasts and memory B cells, suggesting that responsiveness to IL-21 was important for the Ab response. Patients who did not respond to the H1N1/09 vaccine failed to develop these vaccine-induced characteristic B cells changes. These observations are consistent with a recent report showing the importance of IL-21 in developing humoral immune responses in humans with severe combined immunodeficiency mentioned earlier.42 In addition to the IL-21/IL-21R pathway, certain innate immune factors which influence the B cell development and differentiation were also defective in the H1N1/09 vaccine non-responder patient group.91
Table 1.
Immunological changes in B and peripheral TFH cells associated with successful humoral response to seasonal influenza vaccine1
| Cells | Changes in vaccine responders |
|---|---|
| B cells | In vivo up-regulation of IL-21R upon vaccination |
| In vivo development of spontaneous Ab secreting plasmablasts 7 days post-vaccination | |
| in vivo generation of memory B cells 28 days post-vaccination | |
| pTFH cells | In vivo expansion after vaccination |
| In vivo development of Ag-specific memory pTFH cells post-vaccination | |
| Ex vivo IL-21 production upon antigen stimulation | |
| Induction of Ag-specific IgG Ab production in co-cultures with autologous B cells | |
| CXCL13 secretion upon antigen stimulation ex vivo |
Mechanisms underlying a successful influenza vaccine-induced Ab response were evaluated in a cohort of HIV-infected persons on combination antiretroviral therapy who received a single dose of nonadjuvanted H1N1/09 influenza vaccine during the 2009 H1N1 epidemic. Peripheral TFH (pTFH) cells 1 from individuals who responded to the influenza vaccination expanded. IL-21 and CXCL13 secretion was increased, together with the ability of pTFH cells to promote antigen-specific IgG secretion when co-cultured with autologous B cells. Moreover, IL-21R expression on B cells was increased upon vaccination, indicating the importance of the IL-21/IL-21R pathway in both the pTFH and the B cell compartment. Ab production and generation of memory B cells was observed in response to the vaccine. These changes in B and pTFH compartments were not observed in HIV-infected individuals who did not respond to the H1N1/09 vaccine. The table reports the changes in both B and peripheral TFH cell compartments that were associated with responsiveness to the vaccine.
Based on the data discussed here and prior evidence for the role of IL-21 in promoting T-dependent B cell proliferation, isotype switching, differentiation of B cells into plasma cells and development of memory B cells,38,39,52,150 IL-21 merits continued attention in the future for non-HIV and HIV vaccine responses. Since IL-21 production is mainly stimulated by activation of TFH cells as discussed above, it is important to determine if the B cell defects in HIV-infected people are secondary to a deficiency in the CD4 TFH cell compartment and can be reversed by IL-21. In the H1N1/09 vaccine responders we found expansion of pTFH with secretion of IL-21. Moreover we could demonstrate that the antigen-stimulated purified pTFH cells could selectively help autologous antigen-stimulated B cells to secrete H1N1 Ab in vaccine responders whereas non-pTFH (CXCR5− CD4 T cells) had poor helper function. Thus the novel H1N1 influenza vaccine was able to induce development of efficient IL-21 secreting pTFH, memory B cells and plasmablasts in about half of the patients, although all including vaccine non-responders had quantitative CD4 T cell reconstitution and virus control with ART. These findings point out that we still need to understand the basis of immune deficits that persist despite otherwise successful ART which leads to increases in CD4 cells and virologic control. Though not investigated, it is possible that the function of pTFH in vaccine non-responders is compromised because of higher HIV proviral burden and residual low-level virus replication.
Whether IL-21 can directly improve the survival and function of immune cells including B cells was investigated in chronically SIV-infected rhesus macaques by administering rMamu-IL-21 to them. We found a small but significant increase in the frequencies of CD27+ memory B cells with upregulation of IL-21R on both CD27+ memory and CD27− naïve B cells, together with an increase in anti-SIV antibodies in the serum of the IL-21-treated animals,18 providing proof of a biologic activity of IL-21 on B cell populations. In addition to peripheral blood, peripheral lymph nodes also showed an increase in CD27+ memory B cells with increased expression of IL-21R, illustrating the potential of IL-21 to increase B cell memory at different anatomical sites. It is interesting to note that administration of IL-21 was able to increase the memory B cells and anti-SIV Abs despite an advanced chronic phase of SIV infection, highlighting its immunotherapeutic properties in SIV infection.18 At this time IL-21 has not been evaluated as an adjuvant for its B cell stimulating properties in HIV/SIV vaccine strategies to prevent infection. The importance of induction of mucosal and systemic HIV/SIV Ab with vaccines is being increasingly emphasized as discussed earlier based on encouraging results in humans and rhesus macaques with vaccines utilizing boosting with envelope protein.151.
IX. IL-21/IL-21R involvement in human inflammatory diseases and cancer
Because IL-21 exerts regulatory effects on lymphoid and myeloid cells, the role of IL-21 has been investigated in certain human immune-mediated diseases such as those involving the gastrointestinal tract, skin, rheumatoid arthritis and systemic lupus erythematosus and malignancies (Reviewed3, 48). IL-21 is over-expressed in the gut of patients with Crohn's disease and ulcerative colitis, the two major forms of inflammatory bowel disease. High IL-21 production is also seen in other Th1-cell-associated gastrointestinal diseases, such as Helicobacter pylori-related gastritis and celiac disease. IL-21 and IL-21R exhibit increased expression in skin lesions of patients with atopic dermatitis, and IL-21R expression is increased in epidermal keratinocytes of patients with systemic sclerosis. The synovial macrophages and fibroblasts express high IL-21 and IL-21R in patients with rheumatoid arthritis. Interestingly, reduced IL-21R expression in peripheral blood B cells of patients with lupus has been associated with nephritis and high titers of autoantibody (Reviewed3, 48).
Recent studies have highlighted the potential of IL-21 as an immunotherapeutic agent for hematological malignancies based on its ability to induce apoptosis of leukemic cells. Importantly, the ability of IL-21 to enhance CD8+ T and NK cell cytolytic activity have led to clinical trials in patients with renal cell carcinoma and metastatic melanoma, with evidence for improved anti-tumor CD8 T cell responses. IL-21 was well-tolerated, with mild adverse side effects and showed some efficacy in tumor control or regression.152–155 There is a need for a better understanding of how and when IL-21 exerts its effects optimally in vivo. This knowledge could lead to future studies moving IL-21 into the therapeutic setting
Conclusions
IL-21 is an important cytokine that regulates both humoral and cellular immune responses in health and disease states. In HIV infection a beneficial role has been ascribed to IL-21 based on studies in animal models and humans with viral infections, including HIV/SIV infected states and with the influenza H1N1/09 vaccine in HIV-infected and uninfected individuals. In chronically SIV-infected rhesus macaques rMamuIL-21 alone has shown promising effects on humoral immune responses. Importantly, IL-21 also favorably influenced cellular immunity in these animals without inducing immune activation that adversely influences disease progression, thus pointing to its potential applicability in preventative or therapeutic strategies for HIV. Strategies incorporating IL-21 are favored on the basis of IL-21’s B, T and NK stimulating activities. Besides its direct effects on these subsets, another mechanism by which IL-21 could augment immune responses is through inhibition of regulatory T cell activity, by inhibiting the transforming growth factor beta-mediated differentiation of naive CD4 T cells into regulatory T cells, or as recently demonstrated, through inhibiting IL-2, which is necessary for Treg growth and survival.156 Studies of cancer, autoimmune and infectious disease models suggest that administration or blocking of IL-21 may be an attractive therapeutic strategy in infectious and non-infectious disease settings.
Defects in the humoral arm of the immune system during HIV infection are prominent as antibody titers against antigens previously encountered through vaccination or natural infection are low in some patients even among those who are stable on ART. Contributing factors could involve intrinsic B cell defects including altered expression of molecules involved in the B-cell homing process, abnormal T cell activation, impaired TFH function and dysfunctional interaction between T and B cells in the GC of lymphoid tissues. The significant association of IL-21 and IL-21R with H1N1/09 vaccine-mediated immune response in HIV-infected individuals (Table 1) indicates that alterations in IL-21/IL-21R function can contribute to defects in the overall immune response to vaccine antigens. Defects in IL-21 signaling within T and B cells would also impair the immune response against T dependent and independent vaccine responses. Incorporation of more than one costimulatory molecules involved in T-B interaction may be the most effective approach for optimal vaccine induced humoral immune responses. The pleiotropic actions of IL-21 on multiple lineages of cells indicate that modulation of IL-21 and IL-21R signaling could be harnessed for therapeutic effects. Continued research is needed to understand the IL-21/IL-21R driven optimal B cell responses and factors that interfere with them.
Current knowledge of the biologic activity of IL-21, promising data in viral infections in mice, interesting findings in cells of HIV-infected patients, and in-vivo trials in humans and non-human primates all point to a favorable profile for IL-21, but as yet a definitive role of IL-21 in the prevention or treatment of HIV infection has not been established. Knowledge gained from studies on the effect of IL-21 in HIV infection, autoimmunity and immunity to tumors will advance the field for the development of IL-21-based adjuvants and therapeutics for prevention or treatment that could potentially be used in the fight against HIV.
Acknowledgements
This work was supported by NIH grant A1077501 to SP. We acknowledge support from the Miami Center for AIDS Research (CFAR) at the University of Miami Miller School of Medicine which is funded by a grant (P30AI073961) from the National Institutes of Health (NIH). The CFAR program at the NIH includes the following co-funding and participating Institutes and Centers: NIAID, NCI, NICHD, NHLBI, NIDA, NIMH, NIA, FIC, and OAR.
Abbreviations used
- IL-21R
IL-21 receptor
- TFH
T follicular helper cells
- NK
natural killer
- HIV
human immunodeficiency virus
- SIV
simian immunodeficiency virus
- Ab
antibody
- ART
antiretroviral therapy
- GC
germinal center
- PCs
plasma cells
- JAK
Janus family tyrosine kinases
- STAT
signal transducer and activator of transcription
- AID
activation-induced cytidine deaminase
- pTFH
peripheral TFH
- BLIMP-1
B lymphocyte induced maturation protein 1
- bNAbs
broadly neutralizing antibodies
Footnotes
The authors have no financial conflicts of interest.
References
- 1.Parrish-Novak J, Dillon SR, Nelson A, Hammond A, Sprecher C, Gross JA, Johnston J, Madden K, Xu W, West J, Schrader S, Burkhead S, Heipel M, Brandt C, Kuijper JL, Kramer J, Conklin D, Presnell SR, Berry J, Shiota F, Bort S, Hambly K, Mudri S, Clegg C, Moore M, Grant FJ, Lofton-Day C, Gilbert T, Rayond F, Ching A, Yao L, Smith D, Webster P, Whitmore T, Maurer M, Kaushansky K, Holly RD, Foster D. Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature. 2000;408:57–63. doi: 10.1038/35040504. [DOI] [PubMed] [Google Scholar]
- 2.Parrish-Novak J, Foster DC, Holly RD, Clegg CH. Interleukin-21 and the IL-21 receptor: novel effectors of NK and T cell responses. J Leukoc Biol. 2002;72:856–863. [PubMed] [Google Scholar]
- 3.Spolski R, Leonard WJ. Interleukin-21: basic biology and implications for cancer and autoimmunity. Annu Rev Immunol. 2008;26:57–79. doi: 10.1146/annurev.immunol.26.021607.090316. [DOI] [PubMed] [Google Scholar]
- 4.Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L, Wang YH, Watowich SS, Jetten AM, Tian Q, Dong C. 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. doi: 10.1016/j.immuni.2008.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wei L, Laurence A, Elias KM, O'Shea JJ. IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner. J Bil Chem. 282:34605–34610. doi: 10.1074/jbc.M705100200. 200730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bryant VL, Ma CS, Avery DT, Li Y, Good KL, Corcoran LM, de Waal Malefyt R, Tangye SG. Cytokine-mediated regulation of human B cell differentiation into Ig-secreting cells: predominant role of IL-21 produced by CXCR5+ T follicular helper cells. J Immunol. 2007;179:8180–8190. doi: 10.4049/jimmunol.179.12.8180. [DOI] [PubMed] [Google Scholar]
- 7.Coquet JM, Kyparissoudis K, Pellicci DG, Besra G, Berzins SP, Smyth MJ, Godfrey DI. IL-21 is produced by NKT cells and modulates NKT cell activation and cytokine production. J Immunol. 2007;178:2827–2834. doi: 10.4049/jimmunol.178.5.2827. [DOI] [PubMed] [Google Scholar]
- 8.Elsaesser H, Sauer K, Brooks DG. IL-21 is required to control chronic viral infection. Science. 2009;324:1569–1572. doi: 10.1126/science.1174182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Fröhlich A, Kisielow J, Schmitz I, Freigang S, Shamshiev AT, Weber J, Marsland BJ, Oxenius A, Kopf M. IL-21R on T cells is critical for sustained functionality and control of chronic viral infection. Science. 2009;324:1576–1580. doi: 10.1126/science.1172815. [DOI] [PubMed] [Google Scholar]
- 10.Ma SW, Huang X, Li YY, Tang LB, Sun XF, Jiang XT, Zhang YX, Sun J, Liu ZH, Abbott WG, Dong YH, Naoumov NV, Hou JL. High serum IL-21 levels after 12 weeks of antiviral therapy predict HBeAg seroconversion in chronic hepatitis B. J Hepatol. 2012;56:775–781. doi: 10.1016/j.jhep.2011.10.020. [DOI] [PubMed] [Google Scholar]
- 11.Pawlak K, Mysliwiec M, Pawlak D. Interleukin-21 in hemodialyzed patients: association with the etiology of chronic kidney disease and the seropositivity against hepatitis C virus infection. Clin Biochem. 2011;44:1416–1420. doi: 10.1016/j.clinbiochem.2011.09.003. [DOI] [PubMed] [Google Scholar]
- 12.Publicover J, Goodsell A, Nishimura S, Vilarinho S, Wang ZE, Avanesyan L, Spolski R, Leonard WJ, Cooper S, Baron JL. IL-21 is pivotal in determining age-dependent effectiveness of immune responses in a mouse model of human hepatitis B. J Clin Invest. 2011;121:1154–1162. doi: 10.1172/JCI44198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yi JS, Du M, Zajac AJ. A vital role for interleukin-21 in the control of a chronic viral infection. Science. 2009;324:1572–1576. doi: 10.1126/science.1175194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yi JS, Ingram JT, Zajac AJ. IL-21 deficiency influences CD8 T cell quality and recall responses following an acute viral infection. J Immunol. 2010;185:4835–4845. doi: 10.4049/jimmunol.1001032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Chevalier MF, Jülg B, Pyo A, Flanders M, Ranasinghe S, Soghoian DZ, Kwon DS, Rychert J, Lian J, Muller MI, Cutler S, McAndrew E, Jessen H, Pereyra F, Rosenberg ES, Altfeld M, Walker BD, Streeck H. HIV-1-specific interleukin-21+ CD4+ T cell responses contribute to durable viral control through the modulation of HIV-specific CD8+ T cell function. J Virol. 2011;85:733–741. doi: 10.1128/JVI.02030-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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 non-responders to the 2009 H1N1/09 vaccine. Blood. 2012 doi: 10.1182/blood-2011-12-396648. [Epub ahead of print, PMID:22692510]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.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]
- 18.Pallikkuth S, Rogers K, Villinger F, Dosterii M, Vaccari M, Franchini G, Pahwa R, Pahwa S. Interleukin-21 administration to rhesus macaques chronically infected with simian immunodeficiency virus increases cytotoxic effector molecules in T cells and NK cells and enhances B cell function without increasing immune activation or viral replication. Vaccine. 2011;29:9229–9238. doi: 10.1016/j.vaccine.2011.09.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.White L, Krishnan S, Strbo N, Liu H, Kolber MA, Lichtenheld MG, Pahwa RN, Pahwa S. Differential effects of IL-21 and IL-15 on perforin expression, lysosomal degranulation, and proliferation in CD8 T cells of patients with human immunodeficiency virus-1 (HIV) Blood. 2007;109:3873–3880. doi: 10.1182/blood-2006-09-045278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Williams LD, Bansal A, Sabbaj S, Heath SL, Song W, Tang J, Zajac AJ, Goepfert PA. Interleukin-21-Producing HIV-1-Specific CD8 T Cells Are Preferentially Seen in Elite Controllers. J Virol. 2011;85:2316–2324. doi: 10.1128/JVI.01476-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yue FY, Lo C, Sakhdari A, Lee EY, Kovacs CM, Benko E, Liu J, Song H, Jones RB, Sheth P, Chege D, Kaul R, Ostrowski MA. HIV-specific IL-21 producing CD4+ T cells are induced in acute and chronic progressive HIV infection and are associated with relative viral control. J Immunol. 2010;185:498–506. doi: 10.4049/jimmunol.0903915. [DOI] [PubMed] [Google Scholar]
- 22.Ma J, Ma D, Ji C. The role of IL-21 in hematological malignancies. Cytokine. 2011;56:133–139. doi: 10.1016/j.cyto.2011.07.011. [DOI] [PubMed] [Google Scholar]
- 23.Sarra M, Franze E, Pallone F, Monteleone G. Targeting interleukin-21 in inflammatory diseases. Expert Opin Ther Targets. 2011;15:695–702. doi: 10.1517/14728222.2011.561319. [DOI] [PubMed] [Google Scholar]
- 24.Sondergaard H, Skak K. IL-21: roles in immunopathology and cancer therapy. Tissue Antigens. 2009;74:467–479. doi: 10.1111/j.1399-0039.2009.01382.x. [DOI] [PubMed] [Google Scholar]
- 25.Lane HC, Depper JM, Greene WC, Whalen G, Waldmann TA, Fauci AS. Qualitative analysis of immune function in patients with the acquired immunodeficiency syndrome. Evidence for a selective defect in soluble antigen recognition. N Engl J Med. 1985;313:79–84. doi: 10.1056/NEJM198507113130204. [DOI] [PubMed] [Google Scholar]
- 26.Bowen DL, Lane HC, Fauci AS. Immunopathogenesis of the acquired immunodeficiency syndrome. Ann Intern Med. 1985;103:704–709. doi: 10.7326/0003-4819-103-5-704. [DOI] [PubMed] [Google Scholar]
- 27.Rosenberg ZF, Fauci AS. The immunopathogenesis of HIV infection. Adv Immunol. 1989;47:377–431. doi: 10.1016/s0065-2776(08)60665-3. [DOI] [PubMed] [Google Scholar]
- 28.Giorgi JV, Hultin LE, McKeating JA, Johnson TD, Owens B, Jacobson LP, Shih R, Lewis J, Wiley DJ, Phair JP, Wolinsky SM, Detels R. Shorter survival in advanced human immunodeficiency virus type 1 infection is more closely associated with T lymphocyte activation than with plasma virus burden or virus chemokine coreceptor usage. J Infect Dis. 1999;179:859–870. doi: 10.1086/314660. [DOI] [PubMed] [Google Scholar]
- 29.Moir S, Buckner CM, Ho J, Wang W, Chen J, Waldner AJ, Posada JG, Kardava L, O'Shea MA, Kottilil S, Chun TW, Proschan MA, Fauci AS. 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]
- 30.Moir S, Fauci AS. Pathogenic mechanisms of B-lymphocyte dysfunction in HIV disease. J Allergy Clin Immunol. 2008;122:12–19. doi: 10.1016/j.jaci.2008.04.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.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]
- 32.Migueles SA, Osborne CM, Royce C, Compton AA, Joshi RP, Weeks KA, Rood JE, Berkley AM, Sacha JB, Cogliano-Shutta NA, Lloyd M, Roby G, Kwan R, McLaughlin M, Stallings S, Rehm C, O'Shea MA, Mican J, Packard BZ, Komoriya A, Palmer S, Wiegand AP, Maldarelli F, Coffin JM, Mellors JW, Hallahan CW, Follman DA, Connors M. Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control. Immunity. 2008;19(29):1009–1021. doi: 10.1016/j.immuni.2008.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Schmitz JE, Kuroda MJ, Santra S, Sasseville VG, Simon MA, Lifton MA, Racz P, Tenner-Racz K, Dalesandro M, Scallon BJ, Ghrayeb J, Forman MA, Montefiori DC, Rieber EP, Letvin NL, Reimann KA. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science. 1999 Feb 5;283(5403):857–860.35. doi: 10.1126/science.283.5403.857. [DOI] [PubMed] [Google Scholar]
- 34.Hersperger AR, Pereyra F, Nason M, Demers K, Sheth P, Shin LY, Kovacs CM, Rodriguez B, Sieg SF, Teixeira-Johnson L, Gudonis D, Goepfert PA, Lederman MM, Frank I, Makedonas G, Kaul R, Walker BD, Betts MR. Perforin expression directly ex vivo by HIV-specific CD8 T-cells is a correlate of HIV elite control. PLoS Pathog. 2010;6 doi: 10.1371/journal.ppat.1000917. e1000917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Alter G, Altfeld M. NK cells in HIV-1 infection: evidence for their role in the control of HIV-1 infection. J Intern Med. 2009;265:29–42. doi: 10.1111/j.1365-2796.2008.02045.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tiemessen CT, Shalekoff S, Meddows-Taylor S, Schramm DB, Papathanasopoulos MA, Gray GE, Sherman GG, Coovadia AH, Kuhn L. Natural killer cells that respond to human immunodeficiency virus type 1 (HIV-1) peptides are associated with control of HIV-1 infection. J Infect Dis. 2010;202:1444–1453. doi: 10.1086/656535. 38. [DOI] [PMC free article] [PubMed] [Google Scholar]; Strbo N, de Armas L, Liu H, Kolber MA, Lichtenheld M, Pahwa S. IL-21 augments natural killer effector functions in chronically HIV-infected individuals. AIDS. 2008 Aug 20;22(13):1551–1560. doi: 10.1097/QAD.0b013e3283089367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Strbo N, de Armas L, Liu H, Kolber MA, Lichtenheld M, Pahwa S. IL-21 augments natural killer effector functions in chronically HIV-infected individuals. AIDS. 2008;22:1551–1560. doi: 10.1097/QAD.0b013e3283089367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Avery DT, Bryant VL, Ma CS, de Waal Malefyt R, Tangye SG. IL-21-induced isotype switching to IgG and IgA by human naive B cells is differentially regulated by IL-4. J Immunol. 2008;181:1767–1779. doi: 10.4049/jimmunol.181.3.1767. [DOI] [PubMed] [Google Scholar]
- 39.Ettinger R, Sims GP, Fairhurst AM, Robbins R, da Silva YS, Spolski R, Leonard WJ, Lipsky PE. IL-21 induces differentiation of human naive and memory B cells into antibody-secreting plasma cells. J Immunol. 2005;175:7867–7879. doi: 10.4049/jimmunol.175.12.7867. [DOI] [PubMed] [Google Scholar]
- 40.Jin H, Carrio R, Yu A, Malek TR. Distinct activation signals determine whether IL-21 induces B cell costimulation, growth arrest, or Bim-dependent apoptosis. J Immunol. 2004;173:657–665. doi: 10.4049/jimmunol.173.1.657. [DOI] [PubMed] [Google Scholar]
- 41.Mehta DS, Wurster AL, Whitters MJ, Young DA, Collins M, Grusby MJ. IL-21 induces the apoptosis of resting and activated primary B cells. J Immunol. 2003;170:4111–4118. doi: 10.4049/jimmunol.170.8.4111. [DOI] [PubMed] [Google Scholar]
- 42.Recher M, Berglund LJ, Avery DT, Cowan MJ, Gennery AR, Smart J, Peake J, Wong M, Pai SY, Baxi S, Walter JE, Palendira U, Tangye GA, Rice M, Brothers S, Al-Herz W, Oettgen H, Eibel H, Puck JM, Cattaneo F, Ziegler JB, Giliani S, Tangye SG, Notarangelo LD. IL-21 is the primary common gamma chain-binding cytokine required for human B-cell differentiation in vivo. Blood. 2011;118:6824–6835. doi: 10.1182/blood-2011-06-362533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.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]
- 44.Ozaki K, Kikly K, Michalovich D, Young PR, Leonard WJ. Cloning of a type I cytokine receptor most related to the IL-2 receptor beta chain. Proc Natl Acad Sci U S A. 2000;97:11439–11444. doi: 10.1073/pnas.200360997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mehta DS, Wurster AL, Grusby MJ. Biology of IL-21 and the IL-21 receptor. Immunol Rev. 2004;202:84–95. doi: 10.1111/j.0105-2896.2004.00201.x. [DOI] [PubMed] [Google Scholar]
- 46.Good KL, Bryant VL, Tangye SG. Kinetics of human B cell behavior and amplification of proliferative responses following stimulation with IL-21. J Immunol. 2006;177:5236–5247. doi: 10.4049/jimmunol.177.8.5236. [DOI] [PubMed] [Google Scholar]
- 47.Rodriguez-Bayona B, Ramos-Amaya A, Bernal J, Campos-Caro A, Brieva JA. Cutting edge: IL-21 derived from human follicular helper T cells acts as a survival factor for secondary lymphoid organ, but not for bone marrow, plasma cells. J Immunol. 2012;188:1578–1581. doi: 10.4049/jimmunol.1102786. [DOI] [PubMed] [Google Scholar]
- 48.Ettinger R, Kuchen S, Lipsky PE. The role of IL-21 in regulating B-cell function in health and disease. Immunol Rev. 2008;223:60–86. doi: 10.1111/j.1600-065X.2008.00631.x. [DOI] [PubMed] [Google Scholar]
- 49.Asao H, Okuyama C, Kumaki S, Ishii N, Tsuchiya S, Foster D, Sugamura K. Cutting edge: the common gamma-chain is an indispensable subunit of the IL-21 receptor complex. J Immunol. 2001;167:1–5. doi: 10.4049/jimmunol.167.1.1. [DOI] [PubMed] [Google Scholar]
- 50.Konforte D, Paige CJ. Identification of cellular intermediates and molecular pathways induced by IL-21 in human B cells. J Immunol. 2006;177:8381–8392. doi: 10.4049/jimmunol.177.12.8381. [DOI] [PubMed] [Google Scholar]
- 51.de Totero D, Meazza R, Zupo S, Cutrona G, Matis S, Colombo M, Balleari E, Pierri I, Fabbi M, Capaia M, Azzarone B, Gobbi M, Ferrarini M, Ferrini S. Interleukin-21 receptor (IL-21R) is up-regulated by CD40 triggering and mediates proapoptotic signals in chronic lymphocytic leukemia B cells. Blood. 2006;107:3708–3715. doi: 10.1182/blood-2005-09-3535. [DOI] [PubMed] [Google Scholar]
- 52.Avery DT, Deenick EK, Ma CS, Suryani S, Simpson N, Chew GY, Chan TD, Palendira U, Bustamante J, Boisson-Dupuis S, Choo S, Bleasel KE, Peake J, King C, French MA, Engelhard D, Al-Hajjar S, Al-Muhsen S, Magdorf K, Roesler J, Arkwright PD, Hissaria P, Riminton DS, Wong M, Brink R, Fulcher DA, Casanova JL, Cook MC, Tangye SG. 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]
- 53.Pene J, Gauchat JF, Lecart S, Drouet E, Guglielmi P, Boulay V, Delwail A, Foster D, Lecron JC, Yssel H. Cutting edge: IL-21 is a switch factor for the production of IgG1 and IgG3 by human B cells. J Immunol. 2004;172:5154–5157. doi: 10.4049/jimmunol.172.9.5154. [DOI] [PubMed] [Google Scholar]
- 54.Diehl SA, Schmidlin H, Nagasawa M, van Haren SD, Kwakkenbos MJ, Yasuda E, Beaumont T, Scheeren FA, Spits H. STAT3-mediated up-regulation of BLIMP1 Is coordinated with BCL6 down-regulation to control human plasma cell differentiation. J Immunol. 2008;180:4805–4815. doi: 10.4049/jimmunol.180.7.4805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Fuqua CF, Akomeah R, Price JO, Adunyah SE. Involvement of ERK-1/2 in IL-21-induced cytokine production in leukemia cells and human monocytes. Cytokine. 2008;44:101–107. doi: 10.1016/j.cyto.2008.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Habib T, Senadheera S, Weinberg K, Kaushansky K. The common gamma chain (gamma c) is a required signaling component of the IL-21 receptor and supports IL-21-induced cell proliferation via JAK3. Biochemistry. 2002;41:8725–8731. doi: 10.1021/bi0202023. [DOI] [PubMed] [Google Scholar]
- 57.Ma CS, Avery DT, Chan A, Batten M, Bustamante J, Boisson-Dupuis S, Arkwright PD, Kreins AY, Averbuch D, Engelhard D, Magdorf K, Kilic SS, Minegishi Y, Nonoyama S, French MA, Choo S, Smart JM, Peake J, Wong M, Gray P, Cook MC, Fulcher DA, Casanova JL, Deenick EK, Tangye SG. Functional STAT3 deficiency compromises the generation of human T follicular helper cells. Blood. 2012;119:3997–4008. doi: 10.1182/blood-2011-11-392985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chen S, Sims GP, Chen XX, Gu YY, Lipsky PE. Modulatory effects of 1,25-dihydroxyvitamin D3 on human B cell differentiation. J Immunol. 2007;179:1634–1647. doi: 10.4049/jimmunol.179.3.1634. [DOI] [PubMed] [Google Scholar]
- 59.Ettinger R, Sims GP, Robbins R, Withers D, Fischer RT, Grammer AC, Kuchen S, Lipsky PE. IL-21 and BAFF/BLyS synergize in stimulating plasma cell differentiation from a unique population of human splenic memory B cells. J Immunol. 2007;178:2872–2882. doi: 10.4049/jimmunol.178.5.2872. [DOI] [PubMed] [Google Scholar]
- 60.Kuchen S, Robbins R, Sims GP, Sheng C, Phillips TM, Lipsky PE, Ettinger R. Essential role of IL-21 in B cell activation, expansion, and plasma cell generation during CD4+ T cell-B cell collaboration. J Immunol. 2007;179:5886–5896. doi: 10.4049/jimmunol.179.9.5886. [DOI] [PubMed] [Google Scholar]
- 61.Victora GD, Nussenzweig MC. Germinal centers. Annu Rev Immunol. 2012;30:429–457. doi: 10.1146/annurev-immunol-020711-075032. [DOI] [PubMed] [Google Scholar]
- 62.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]
- 63.Schaerli P, Loetscher P, Moser B. Cutting edge: induction of follicular homing precedes effector Th cell development. J Immunol. 2001;167:6082–6086. doi: 10.4049/jimmunol.167.11.6082. [DOI] [PubMed] [Google Scholar]
- 64.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]
- 65.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]
- 66.Eto D, Lao C, DiToro D, Barnett B, Escobar TC, Kageyama R, Yusuf I, Crotty S. IL-21 and IL-6 are critical for different aspects of B cell immunity and redundantly induce optimal follicular helper CD4 T cell (Tfh) differentiation. PLoS One. 2011;6:e17739. doi: 10.1371/journal.pone.0017739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Linterman MA, Beaton L, Yu D, Ramiscal RR, Srivastava M, Hogan JJ, Verma NK, Smyth MJ, Rigby RJ, Vinuesa CG. IL-21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses. J Exp Med. 2010;207:353–363. doi: 10.1084/jem.20091738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.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]
- 69.Jacob J, Kassir R, Kelsoe G. In situ studies of the primary immune response to (4-hydroxy-3-nitrophenyl)acetyl. I. The architecture and dynamics of responding cell populations. J Exp Med. 1991;173:1165–1175. doi: 10.1084/jem.173.5.1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Haynes NM, Allen CD, Lesley R, Ansel KM, Killeen N, Cyster JG. 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. doi: 10.4049/jimmunol.179.8.5099. [DOI] [PubMed] [Google Scholar]
- 71.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. doi: 10.1182/blood-2004-11-4494. [DOI] [PubMed] [Google Scholar]
- 72.Fazilleau N, Eisenbraun MD, Malherbe L, Ebright JN, Pogue-Caley RR, McHeyzer-Williams LJ, McHeyzer-Williams MG. Lymphoid reservoirs of antigen-specific memory T helper cells. Nat Immunol. 2007;8:753–761. doi: 10.1038/ni1472. [DOI] [PubMed] [Google Scholar]
- 73.Rasheed AU, Rahn HP, Sallusto F, Lipp M, Muller G. Follicular B helper T cell activity is confined to CXCR5(hi)ICOS(hi) CD4 T cells and is independent of CD57 expression. Eur J Immunol. 2006;36:1892–1903. doi: 10.1002/eji.200636136. [DOI] [PubMed] [Google Scholar]
- 74.Forster R, Emrich T, Kremmer E, Lipp M. Expression of the G-protein--coupled receptor BLR1 defines mature, recirculating B cells and a subset of T-helper memory cells. Blood. 1994;84:830–840. [PubMed] [Google Scholar]
- 75.Forster R, Mattis AE, Kremmer E, Wolf E, Brem G, Lipp M. A putative chemokine receptor, BLR1, directs B cell migration to defined lymphoid organs and specific anatomic compartments of the spleen. Cell. 1996;87:1037–1047. doi: 10.1016/s0092-8674(00)81798-5. [DOI] [PubMed] [Google Scholar]
- 76.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]
- 77.Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–763. doi: 10.1146/annurev.immunol.22.012703.104702. [DOI] [PubMed] [Google Scholar]
- 78.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]
- 79.Morita R, Schmitt N, Bentebibel SE, Ranganathan R, Bourdery L, Zurawski G, Foucat E, Dullaers M, Oh S, Sabzghabaei N, Lavecchio EM, Punaro M, Pascual V, Banchereau J, Ueno H. 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]
- 80.Bossaller L, Burger J, Draeger R, Grimbacher B, Knoth R, Plebani A, Durandy A, Baumann U, Schlesier M, Welcher AA, Peter HH, Warnatz K. 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]
- 81.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]
- 82.Klatt NR, Vinton CL, Lynch RM, Canary LA, Ho J, Darrah PA, Estes JD, Seder RA, Moir SL, Brenchley JM. SIV infection of rhesus macaques results in dysfunctional T- and B-cell responses to neo and recall Leishmania major vaccination. Blood. 2011;118:5803–5812. doi: 10.1182/blood-2011-07-365874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.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]
- 84.van Grevenynghe J, Halwani R, Chomont N, Ancuta P, Peretz Y, Tanel A, Procopio FA, shi Y, Said EA, Haddad EK, Sekaly RP. Lymph node architecture collapse and consequent modulation of FOXO3a pathway on memory T- and B-cells during HIV infection. Semin Immunol. 2008;20:196–203. doi: 10.1016/j.smim.2008.07.008. [DOI] [PubMed] [Google Scholar]
- 85.Okada T, Ngo VN, Ekland EH, Forster R, Lipp M, Littman DR, Cyster JG. Chemokine requirements for B cell entry to lymph nodes and Peyer's patches. J Exp Med. 2002;196:65–75. doi: 10.1084/jem.20020201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Allen CD, Ansel KM, Low C, Lesley R, Tamamura H, Fujii N, Cyster JG. Germinal center dark and light zone organization is mediated by CXCR4 and CXCR5. Nat Immunol. 2004;5:943–952. doi: 10.1038/ni1100. [DOI] [PubMed] [Google Scholar]
- 87.Allen CD, Okada T, Tang HL, Cyster JG. Imaging of germinal center selection events during affinity maturation. Science. 2007;315:528–531. doi: 10.1126/science.1136736. [DOI] [PubMed] [Google Scholar]
- 88.Cagigi A, Mowafi F, Phuong Dang LV, Tenner-Racz K, Atlas A, Grutzmeier S, Racz P, Chiodi F, Nilsson A. Altered expression of the receptor-ligand pair CXCR5/CXCL13 in B cells during chronic HIV-1 infection. Blood. 2008;112:4401–4410. doi: 10.1182/blood-2008-02-140426. [DOI] [PubMed] [Google Scholar]
- 89.Iannello A, Boulassel MR, Samarani S, Debbeche O, Tremblay C, Toma E, Routy JP, Ahmad A. Dynamics and consequences of IL-21 production in HIV-infected individuals: a longitudinal and cross-sectional study. J Immunol. 2010;184:114–126. doi: 10.4049/jimmunol.0901967. [DOI] [PubMed] [Google Scholar]
- 90.Iannello A, Tremblay C, Routy JP, Boulassel MR, Toma E, Ahmad A. Decreased levels of circulating IL-21 in HIV-infected AIDS patients: correlation with CD4+ T-cell counts. Viral Immunol. 2008;21:385–388. doi: 10.1089/vim.2008.0025. [DOI] [PubMed] [Google Scholar]
- 91.Pallikkuth S, Kanthikeel SP, Silva SY, Fischl M, Pahwa R, Pahwa S. Innate immune defects correlate with failure of antibody responses to H1N1/09 vaccine in HIV-infected patients. J Allergy Clin Immunol. 2011 Dec;128(6):1279–1285. doi: 10.1016/j.jaci.2011.05.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.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]
- 93.Moir S, Malaspina A, Li Y, Chun TW, Lowe T, Adelsberger J, Baseler M, Ehler LA, Liu S, Davey RT, Jr, Mican JA, Fauci AS. B cells of HIV-1-infected patients bind virions through CD21-complement interactions and transmit infectious virus to activated T cells. J Exp Med. 2000;192:637–646. doi: 10.1084/jem.192.5.637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Stoiber H, Speth C, Dierich MP. Role of complement in the control of HIV dynamics and pathogenesis. Vaccine. 2003;21(Suppl 2):S77–S82. doi: 10.1016/s0264-410x(03)00203-2. [DOI] [PubMed] [Google Scholar]
- 95.Dorner T, Radbruch A. Antibodies and B cell memory in viral immunity. Immunity. 2007;27:384–392. doi: 10.1016/j.immuni.2007.09.002. [DOI] [PubMed] [Google Scholar]
- 96.Rickert RC. Regulation of B lymphocyte activation by complement C3 and the B cell coreceptor complex. Curr Opin Immunol. 2005;17:237–243. doi: 10.1016/j.coi.2005.03.001. [DOI] [PubMed] [Google Scholar]
- 97.Huang L, Li CJ, Pardee AB. Human immunodeficiency virus type 1 TAT protein activates B lymphocytes. Biochem Biophys Res Commun. 1997;237:461–464. doi: 10.1006/bbrc.1997.7162. [DOI] [PubMed] [Google Scholar]
- 98.Moir S, Malaspina A, Pickeral OK, Donoghue ET, Vasquez J, Miller NJ, Krishnan SR, Planta MA, Turney JF, Justement JS, Kottilil S, Dybul M, Mican JM, Kovacs C, Chun TW, Birse CE, Fauci AS. Decreased survival of B cells of HIV-viremic patients mediated by altered expression of receptors of the TNF superfamily. J Exp Med. 2004;200:587–599. [PubMed] [Google Scholar]
- 99.Qiao X, He B, Chiu A, Knowles DM, Chadburn A, Cerutti A. Human immunodeficiency virus 1 Nef suppresses CD40-dependent immunoglobulin class switching in bystander B cells. Nat Immunol. 2006;7:302–310. doi: 10.1038/ni1302. [DOI] [PubMed] [Google Scholar]
- 100.He B, Qiao X, Klasse PJ, Chiu A, Chadburn A, Knowles DM, Moore JP, Cerutti A. HIV-1 envelope triggers polyclonal Ig class switch recombination through a CD40-independent mechanism involving BAFF and C-type lectin receptors. J Immunol. 2006;176:3931–3941. doi: 10.4049/jimmunol.176.7.3931. [DOI] [PubMed] [Google Scholar]
- 101.Biancotto A, Grivel JC, Iglehart SJ, Vanpouille C, Lisco A, Sieg SF, Debernardo R, Garate K, Rodriguez B, Margolis LB, Lederman MM. Abnormal activation and cytokine spectra in lymph nodes of people chronically infected with HIV-1. Blood. 2007;109:4272–4279. doi: 10.1182/blood-2006-11-055764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Pahwa S. Role of common gamma chain utilizing cytokines for immune reconstitution in HIV infection. Immunol Res. 2007;38:373–386. doi: 10.1007/s12026-007-0036-9. [DOI] [PubMed] [Google Scholar]
- 103.Sousa AE, Chaves AF, Doroana M, Antunes F, Victorino RM. Bulk cytokine production versus frequency of cytokine-producing cells in HIV1 infection before and during HAART. Clin Immunol. 2000;97:162–170. doi: 10.1006/clim.2000.4930. [DOI] [PubMed] [Google Scholar]
- 104.Younes SA, Yassine-Diab B, Dumont AR, Boulassel MR, Grossman Z, Routy JP, Sekaly RP. HIV-1 viremia prevents the establishment of interleukin 2-producing HIV-specific memory CD4+ T cells endowed with proliferative capacity. J Exp Med. 2003;198:1909–1922. doi: 10.1084/jem.20031598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Choe J, Kim HS, Armitage RJ, Choi YS. The functional role of B cell antigen receptor stimulation and IL-4 in the generation of human memory B cells from germinal center B cells. J Immunol. 1997;159:3757–3766. [PubMed] [Google Scholar]
- 106.van Grevenynghe J, Cubas RA, Noto A, DaFonseca S, He Z, Peretz Y, Filali-Mouhim A, Dupuy FP, Procopio FA, Chomont N, Balderas RS, Said EA, Boulassel MR, Tremblay CL, Routy JP, Sékaly RP, Haddad EK. 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]
- 107.Fry TJ, Connick E, Falloon J, Lederman MM, Liewehr DJ, Spritzler J, Steinberg SM, Wood LV, Yarchoan R, Zuckerman J, Landay A, Mackall CL. A potential role for interleukin-7 in T-cell homeostasis. Blood. 2001;97:2983–2990. doi: 10.1182/blood.v97.10.2983. [DOI] [PubMed] [Google Scholar]
- 108.Napolitano LA, Grant RM, Deeks SG, Schmidt D, De Rosa SC, Herzenberg LA, Herndier BG, Andersson J, McCune JM. Increased production of IL-7 accompanies HIV-1-mediated T-cell depletion: implications for T-cell homeostasis. Nat Med. 2001;7:73–79. doi: 10.1038/83381. [DOI] [PubMed] [Google Scholar]
- 109.LeBien TW, Tedder TF. B lymphocytes: how they develop and function. Blood. 2008;112:1570–1580. doi: 10.1182/blood-2008-02-078071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Malaspina A, Moir S, Ho J, Wang W, Howell ML, O'Shea MA, Roby GA, Rehm CA, Mican JM, Chun TW, Fauci AS. Appearance of immature/transitional B cells in HIV-infected individuals with advanced disease: correlation with increased IL-7. Proc Natl Acad Sci U S A. 2006;103:2262–2267. doi: 10.1073/pnas.0511094103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, Kazzaz Z, Bornstein E, Lambotte O, Altmann D, Blazar BR, Rodriguez B, Teixeira-Johnson L, Landay A, Martin JN, Hecht FM, Picker LJ, Lederman MM, Deeks SG, Douek DC. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
- 112.Pilakka-Kanthikeel S, Huang S, Fenton T, Borkowsky W, Cunningham CK, Pahwa S. Increased gut microbial translocation in HIV-infected children persists in virologic responders and virologic failures after antiretroviral therapy. Pediatr Infect Dis J. 2012;31:583–591. doi: 10.1097/INF.0b013e31824da0f5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003;21:335–376. doi: 10.1146/annurev.immunol.21.120601.141126. [DOI] [PubMed] [Google Scholar]
- 114.Miyake K, Yamashita Y, Ogata M, Sudo T, Kimoto M. RP105, a novel B cell surface molecule implicated in B cell activation, is a member of the leucine-rich repeat protein family. J Immunol. 1995;154:3333–3340. [PubMed] [Google Scholar]
- 115.Ng VL. B-lymphocytes and autoantibody profiles in HIV disease. Clin Rev Allergy Immunol. 1996;14:367–384. doi: 10.1007/BF02771753. [DOI] [PubMed] [Google Scholar]
- 116.Martinez-Maza O, Breen EC. B-cell activation and lymphoma in patients with HIV. Curr Opin Oncol. 2002;14:528–532. doi: 10.1097/00001622-200209000-00009. [DOI] [PubMed] [Google Scholar]
- 117.Pantaleo G, Graziosi C, Demarest JF, Cohen OJ, Vaccarezza M, Gantt K, Muro-Cacho C, Fauci AS. Role of lymphoid organs in the pathogenesis of human immunodeficiency virus (HIV) infection. Immunol Rev. 1994;140:105–130. doi: 10.1111/j.1600-065x.1994.tb00867.x. [DOI] [PubMed] [Google Scholar]
- 118.Shirai A, Cosentino M, Leitman-Klinman SF, Klinman DM. Human immunodeficiency virus infection induces both polyclonal and virus-specific B cell activation. J Clin Invest. 1992;89:561–566. doi: 10.1172/JCI115621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Moir S, Malaspina A, Ogwaro KM, Donoghue ET, Hallahan CW, Ehler LA, Liu S, Adelsberger J, Lapointe R, Hwu P, Baseler M, Orenstein JM, Chun TW, Mican JA, Fauci AS. HIV-1 induces phenotypic and functional perturbations of B cells in chronically infected individuals. Proc Natl Acad Sci U S A. 2001;98:10362–10367. doi: 10.1073/pnas.181347898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kardava L, Moir S, Wang W, Ho J, Buckner CM, Posada JG, O'Shea MA, Roby G, Chen J, Sohn HW, Chun TW, Pierce SK, Fauci AS. Attenuation of HIV-associated human B cell exhaustion by siRNA downregulation of inhibitory receptors. J Clin Invest. 2011;121:2614–2624. doi: 10.1172/JCI45685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O'Shea MA, Roby G, Kottilil S, Arthos J, Proschan MA, Chun TW, Fauci AS. 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]
- 122.Titanji K, Chiodi F, Bellocco R, Schepis D, Osorio L, Tassandin C, Tambussi G, Grutzmeier S, Lopalco L, De Milito A. Primary HIV-1 infection sets the stage for important B lymphocyte dysfunctions. AIDS. 2005;19:1947–1955. doi: 10.1097/01.aids.0000191231.54170.89. [DOI] [PubMed] [Google Scholar]
- 123.Levesque MC, Moody MA, Hwang KK, Marshall DJ, Whitesides JF, Amos JD, Gurley TC, Allgood S, Haynes BB, Vandergrift NA, Plonk S, Parker DC, Cohen MS, Tomaras GD, Goepfert PA, Shaw GM, Schmitz JE, Eron JJ, Shaheen NJ, Hicks CB, Liao HX, Markowitz M, Kelsoe G, Margolis DM, Haynes BF. Polyclonal B cell differentiation and loss of gastrointestinal tract germinal centers in the earliest stages of HIV-1 infection. PLoS Med. 2009;6 doi: 10.1371/journal.pmed.1000107. e1000107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.De Milito A, Morch C, Sonnerborg A, Chiodi F. Loss of memory (CD27) B lymphocytes in HIV-1 infection. AIDS. 2001;15:957–964. doi: 10.1097/00002030-200105250-00003. [DOI] [PubMed] [Google Scholar]
- 125.Chong Y, Ikematsu H, Kikuchi K, Yamamoto M, Murata M, Nishimura M, Nabeshima S, Kashiwagi S, Hayashi J. Selective CD27+ (memory) B cell reduction and characteristic B cell alteration in drug-naive and HAART-treated HIV type 1-infected patients. AIDS Res Hum Retroviruses. 2004;20:219–226. doi: 10.1089/088922204773004941. [DOI] [PubMed] [Google Scholar]
- 126.Moir S, Malaspina A, Ho J, Wang W, Dipoto AC, O'Shea MA, Roby G, Mican JM, Kottilil S, Chun TW, Proschan MA, Fauci AS. Normalization of B cell counts and subpopulations after antiretroviral therapy in chronic HIV disease. J Infect Dis. 2008;197:572–579. doi: 10.1086/526789. [DOI] [PubMed] [Google Scholar]
- 127.Titanji K, De Milito A, Cagigi A, Thorstensson R, Grutzmeier S, Atlas A, Hejdeman B, Kroon FP, Lopalco L, Nilsson A, Chiodi F. 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]
- 128.Notermans DW, de Jong JJ, Goudsmit J, Bakker M, Roos MT, Nijholt L, Cremers J, Hellings JA, Danner SA, de Ronde A. Potent antiretroviral therapy initiates normalization of hypergammaglobulinemia and a decline in HIV type 1-specific antibody responses. AIDS Res Hum Retroviruses. 2001;17:1003–1008. doi: 10.1089/088922201300343681. [DOI] [PubMed] [Google Scholar]
- 129.Jacobson MA, Khayam-Bashi H, Martin JN, Black D, Ng V. Effect of long-term highly active antiretroviral therapy in restoring HIV-induced abnormal B-lymphocyte function. J Acquir Immune Defic Syndr. 2002;31:472–477. doi: 10.1097/00126334-200212150-00003. [DOI] [PubMed] [Google Scholar]
- 130.Malaspina A, Moir S, Kottilil S, Hallahan CW, Ehler LA, Liu S, Planta MA, Chun TW, Fauci AS. Deleterious effect of HIV-1 plasma viremia on B cell costimulatory function. J Immunol. 2003;170:5965–5972. doi: 10.4049/jimmunol.170.12.5965. [DOI] [PubMed] [Google Scholar]
- 131.Regidor DL, Detels R, Breen EC, Widney DP, Jacobson LP, Palella F, Rinaldo CR, Bream JH, Martínez-Maza O. Effect of highly active antiretroviral therapy on biomarkers of B-lymphocyte activation and inflammation. AIDS. 2011;25:303–314. doi: 10.1097/QAD.0b013e32834273ad. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Ho J, Moir S, Malaspina A, Howell ML, Wang W, DiPoto AC, O'Shea MA, Roby GA, Kwan R, Mican JM, Chun TW, Fauci AS. Two overrepresented B cell populations in HIV-infected individuals undergo apoptosis by different mechanisms. Proc Natl Acad Sci U S A. 2006;103:19436–19441. doi: 10.1073/pnas.0609515103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Malaspina A, Moir S, Orsega SM, Vasquez J, Miller NJ, Donoghue ET, Kottilil S, Gezmu M, Follmann D, Vodeiko GM, Levandowski RA, Mican JM, Fauci AS. Compromised B cell responses to influenza vaccination in HIV-infected individuals. J Infect Dis. 2005;191:1442–1450. doi: 10.1086/429298. [DOI] [PubMed] [Google Scholar]
- 134.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]
- 135.Tejiokem MC, Gouandjika I, Beniguel L, Zanga MC, Tene G, Gody JC, Njamkepo E, Kfutwah A, Penda I, Bilong C, Rousset D, Pouillot R, Tangy F, Baril L. HIV-infected children living in Central Africa have low persistence of antibodies to vaccines used in the Expanded Program on Immunization. PLoS One. 2007;2:e1260. doi: 10.1371/journal.pone.0001260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Bliss SJ, O'Brien KL, Janoff EN, Cotton MF, Musoke P, Coovadia H, Levine OS. The evidence for using conjugate vaccines to protect HIV-infected children against pneumococcal disease. Lancet Infect Dis. 2008;8:67–80. doi: 10.1016/S1473-3099(07)70242-6. [DOI] [PubMed] [Google Scholar]
- 137.Simek MD, Rida W, Priddy FH, Pung P, Carrow E, Laufer DS, Lehrman JK, Boaz M, Tarragona-Fiol T, Miiro G, Birungi J, Pozniak A, McPhee DA, Manigart O, Karita E, Inwoley A, Jaoko W, Dehovitz J, Bekker LG, Pitisuttithum P, Paris R, Walker LM, Poignard P, Wrin T, Fast PE, Burton DR, Koff WC. Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. J Virol. 2009;83:7337–7348. doi: 10.1128/JVI.00110-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Falkowska E, Ramos A, Feng Y, Zhou T, Moquin S, Walker LM, Wu X, Seaman MS, Wrin T, Kwong PD, Wyatt RT, Mascola JR, Poignard P, Burton DR. PGV04, an HIV-1 gp120 CD4 binding site antibody, is broad and potent in neutralization but does not induce conformational changes characteristic of CD4. J Virol. 2012;86:4394–4403. doi: 10.1128/JVI.06973-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Lynch RM, Tran L, Louder MK, Schmidt SD, Cohen M, Dersimonian R, Euler Z, Gray ES, Abdool Karim S, Kirchherr J, Montefiori DC, Sibeko S, Soderberg K, Tomaras G, Yang ZY, Nabel GJ, Schuitemaker H, Morris L, Haynes BF, Mascola JR CHAVI 001 Clinical Team Members. The Development of CD4 Binding Site Antibodies during HIV-1 Infection. J Virol. 86:7588–7595. doi: 10.1128/JVI.00734-12. 2012l. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.McLellan JS, Pancera M, Carrico C, Gorman J, Julien JP, Khayat R, Louder R, Pejchal R, Sastry M, Dai K, O'Dell S, Patel N, Shahzad-ul-Hussan S, Yang Y, Zhang B, Zhou T, Zhu J, Boyington JC, Chuang GY, Diwanji D, Georgiev I, Kwon YD, Lee D, Louder MK, Moquin S, Schmidt SD, Yang ZY, Bonsignori M, Crump JA, Kapiga SH, Sam NE, Haynes BF, Burton DR, Koff WC, Walker LM, Phogat S, Wyatt R, Orwenyo J, Wang LX, Arthos J, Bewley CA, Mascola JR, Nabel GJ, Schief WR, Ward AB, Wilson IA, Kwong PD. Structure of HIV-1 gp120 V1/V2 domain with broadly neutralizing antibody PG9. Nature. 2011;480:336–343. doi: 10.1038/nature10696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Koff WC. HIV vaccine development: Challenges and opportunities towards solving the HIV vaccine-neutralizing antibody problem. Vaccine. 2012;30:4310–4315. doi: 10.1016/j.vaccine.2011.11.014. [DOI] [PubMed] [Google Scholar]
- 142.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]
- 143.Tomaras GD, Haynes BF. Strategies for eliciting HIV-1 inhibitory antibodies. Curr Opin HIV AIDS. 2010;5:421–427. doi: 10.1097/COH.0b013e32833d2d45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Verkoczy L, Kelsoe G, Moody MA, Haynes BF. Role of immune mechanisms in induction of HIV-1 broadly neutralizing antibodies. Curr Opin Immunol. 2011;23:383–390. doi: 10.1016/j.coi.2011.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Tomaras GD, Haynes BF. HIV-1-specific antibody responses during acute and chronic HIV-1 infection. Curr Opin HIV AIDS. 2009;4:373–379. doi: 10.1097/COH.0b013e32832f00c0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.McElrath MJ, Haynes BF. Induction of immunity to human immunodeficiency virus type-1 by vaccination. Immunity. 2010;33:542–554. doi: 10.1016/j.immuni.2010.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Mouquet H, Klein F, Scheid JF, Warncke M, Pietzsch J, Oliveira TY, Velinzon K, Seaman MS, Nussenzweig MC. Memory B cell antibodies to HIV-1 gp140 cloned from individuals infected with clade A and B viruses. PLoS One. 2011;6:e24078.. doi: 10.1371/journal.pone.0024078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Alter G, Moody MA. The humoral response to HIV-1: new insights, renewed focus. J Infect Dis. 2010;202(Suppl 2):S315–S322. doi: 10.1086/655654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Ruffin N, Lantto R, Pensieroso S, Sammicheli S, Hejdeman B, Rethi B, Chiodi F. Immune activation and increased IL-21R expression are associated with the loss of memory B cells during HIV-1 infection. J Intern Med. 2012 doi: 10.1111/j.1365-2796.2012.02550.x. [Epub ahead of print, PMID:22530560]. [DOI] [PubMed] [Google Scholar]
- 150.Borte S, Pan-Hammarstrom Q, Liu C, Sack U, Borte M, Wagner U, Graf D, Hammarström L. Interleukin-21 restores immunoglobulin production ex vivo in patients with common variable immunodeficiency and selective IgA deficiency. Blood. 2009;114:4089–4098. doi: 10.1182/blood-2009-02-207423. [DOI] [PubMed] [Google Scholar]
- 151.Brocca-Cofano E, McKinnon K, Demberg T, Venzon D, Hidajat R, Xiao P, Daltabuit-Test M, Patterson LJ, Robert-Guroff M. Vaccine-elicited SIV and HIV envelope-specific IgA and IgG memory B cells in rhesus macaque peripheral blood correlate with functional antibody responses and reduced viremia. Vaccine. 2011;29:3310–3319. doi: 10.1016/j.vaccine.2011.02.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Davis ID, Brady B, Kefford RF, Millward M, Cebon J, Skrumsager BK, Mouritzen U, Hansen LT, Skak K, Lundsgaard D, Frederiksen KS, Kristjansen PE, McArthur G. Clinical and biological efficacy of recombinant human interleukin-21 in patients with stage IV malignant melanoma without prior treatment: a phase IIa trial. Clin Cancer Res. 2009;15:2123–2129. doi: 10.1158/1078-0432.CCR-08-2663. 2009. [DOI] [PubMed] [Google Scholar]
- 153.Frederiksen KS, Lundsgaard D, Freeman JA, Hughes SD, Holm TL, Skrumsager BK, Petri A, Hansen LT, McArthur GA, Davis ID, Skak K. IL-21 induces in vivo immune activation of NK cells and CD8(+) T cells in patients with metastatic melanoma and renal cell carcinoma. Cancer Immunol Immunother. 2008;57:1439–1449. doi: 10.1007/s00262-008-0479-4. 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Thompson JA, Curti BD, Redman BG, Bhatia S, Weber JS, Agarwala SS, Sievers EL, Hughes SD, DeVries TA, Hausman DF. Phase I study of recombinant interleukin-21 in patients with metastatic melanoma and renal cell carcinoma. J Clin Oncol. 2008;26:2034–2039. doi: 10.1200/JCO.2007.14.5193. 2008. [DOI] [PubMed] [Google Scholar]
- 155.Schmidt H, Brown J, Mouritzen U, Selby P, Fode K, Svane IM, Cook GP, Mollerup DH, Geertsen PF. Safety and clinical effect of subcutaneous human interleukin-21 in patients with metastatic melanoma or renal cell carcinoma: a phase I trial. Clin Cancer Res. 2010;16:5312–5319. doi: 10.1158/1078-0432.CCR-10-1809. [DOI] [PubMed] [Google Scholar]
- 156.Attridge K, Wang CJ, Wardzinski L, Kenefeck R, Chamberlain JL, Manzotti C, Kopf M, Walker LS. IL-21 inhibits T cell IL-2 production and impairs Treg homeostasis. Blood. 2012;119:4656–4664. doi: 10.1182/blood-2011-10-388546. [DOI] [PubMed] [Google Scholar]

