Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Dec 1.
Published in final edited form as: Curr Opin Immunol. 2010 Oct 21;22(6):732–739. doi: 10.1016/j.coi.2010.09.010

Targeting BAFF in autoimmunity

Anne Davidson 1
PMCID: PMC2997938  NIHMSID: NIHMS243072  PMID: 20970975

Abstract

BAFF and APRIL are TNF-like cytokines that support survival and differentiation of B cells. The early appreciation that overexpression of BAFF leads to B cell expansion and a lupus-like syndrome in mice, and the demonstration that BAFF inhibition delays lupus onset in spontaneous mouse models of SLE and other autoimmune diseases has rapidly led to the development of strategies for inhibiting both BAFF and APRIL. The commercialization of this new class of drugs has proceeded in parallel with the continuing elucidation of the biology of the cytokines and their receptors. Recent studies have uncovered a role for BAFF in enhancing both innate and adaptive immune responses and in amplifying aberrant pathways that arise during inflammation. Two phase III studies of an anti-BAFF antibody have yielded positive, although modest, results in SLE and alternate inhibitors are being tested in a variety of autoimmune diseases in which BAFF may play a pathogenic role.

Introduction

BAFF and APRIL are expressed by many cell types including monocytes, DCs, neutrophils, stromal cells, activated T cells, B cells and B cell tumors, and epithelial cells. BAFF binds to three receptors, BAFF-R, TACI and BCMA that are expressed on B cells at different developmental stages whereas APRIL binds to TACI and BCMA and has a proteoglycan binding site that facilitates its aggregation on cell surfaces (Figure 1). Increased serum levels of BAFF and APRIL are found in several autoimmune diseases, and both cytokines can be elaborated in inflammatory sites. Comprehensive descriptions of BAFF and APRIL and their receptors including the consequences of their overexpression or deletion have recently been published [1,2].

Figure 1.

Figure 1

The BAFF/APRIL family and their receptors: BAFF and APRIL are cleaved by furin proteases to yield soluble homotrimers. BAFF and APRIL can also heterotrimerize. APRIL is expressed on the cell membrane when it is fused to the transmembrane and cytoplasmic portion of TWEAK (TWE-PRIL). BAFF is expressed on the cell membrane either as full length BAFF or as an alternatively spliced form missing 57bp (ΔBAFF) that is not cleaved. Other splice variants of various family members have been identified. Soluble BAFF can multimerize into a 20 trimer structure that is the preferential ligand for TACI. APRIL is multimerized by binding to proteoglycans. TACI can also bind to proteoglycans such as syndecan. Drugs that target the cytokines include belimumab that blocks soluble BAFF and atacicept that blocks both BAFF and APRIL.

Abbreviations: Abbreviations: APRIL, A proliferation inducing ligand; BAFF, B cell activating factor belonging to the TNF family; TACI, Transmembrane activator and calcium modulator ligand interactor; BCMA, B cell maturation antigen; BAFF-R, BAFF receptor; HSPG, heparan sulphate proteoglycan

Expression of the BAFF/APRIL receptors first becomes functional at the transitional B cell stage with BAFF-R being the predominant receptor on naïve and memory B cells, TACI the predominant receptor on marginal zone B cells and short-lived plasma cells and BCMA the predominant receptor on long-lived plasma cells. Each receptor activates its own set of signaling pathways with BAFF-R being the only BAFF receptor to activate the alternative NF-κB pathway (reviewed in [15]).

Selective antagonists of BAFF include a fully human anti-BAFF antibody that binds only soluble BAFF (belimumab - Human Genome Sciences) and other antibodies that block both soluble and membrane bound BAFF (K. Kikly, abstract 693, presented at American College of Rheumatology Meeting, Philadelphia, November 2009). A BAFF-R-Ig fusion protein is also under development, as is a depleting antibody to BAFF-R [6]. TACI-Ig is a non-selective antagonist of both BAFF and APRIL (atacicept – EMD, Serono - Figure 1).

Variant forms of BAFF and APRIL

BAFF and APRIL are Type II transmembrane proteins that are cleaved by furin proteases to yield soluble homotrimers. APRIL is also expressed on the cell membrane as a fusion protein consisting of the extracellular domain of APRIL and the transmembrane and cytoplasmic domain of TWEAK (TWE-PRIL). BAFF is extensively cleaved but it is also expressed on the cell membrane either as full length BAFF or as an alternatively spliced form missing 57bp (ΔBAFF) that is not cleaved and acts as an inhibitor [7]. The physiologic role of membrane BAFF is important to understand because some BAFF inhibitors target the membrane form whereas others do not. Recent reports suggest that reverse signaling through membrane BAFF may occur [8,9]; the physiologic significance of this observation remains to be determined.

A small proportion of soluble BAFF multimerizes into a 20 trimer structure. While BAFF-R is activated by BAFF trimers, signaling through TACI requires multimerized ligands [10] such as membrane BAFF, circulating BAFF 60-mer, or multimerized APRIL. APRIL is multimerized by binding to proteoglycans but a possible role for TWE-PRIL in APRIL-TACI interactions has not been excluded. Of note, TACI-Ig blocks the binding of BAFF to BAFF-R indicating that it inhibits the function of the trimeric form of BAFF; it is possible that binding of TACI to monomeric BAFF may occur although this is not sufficient to initiate signaling through TACI. BAFF and APRIL can heterotrimerize, but the level of these heterotrimers is low [11] and their physiologic significance is not known. Other splice variants of the cytokines and their receptors continue to be identified [12].

Further analysis of the functional activities of BAFF and APRIL and their variant forms as well as clarification of their roles in supporting each of the known B cell subsets during periods of inflammation will undoubtedly be forthcoming as the various inhibitors are tested in human diseases.

BAFF supports naïve B cell survival and influences B cell selection

The interaction of BAFF with BAFF-R cooperates with BCR signaling at the late transitional B cell stage in determining whether or not a B cell will compete efficiently for survival and absence of either BAFF or BAFF-R results in substantial loss of mature B cells (reviewed [2]). BCR signaling upregulates expression of BAFF-R and also generates p100, an essential substrate for the non-classical NF-κB signaling pathway used by BAFF-R [13]. This is dependent on the presence of Btk [14,15]. It is important to note however that signaling through the alternative NFκB pathway is not sufficient to maintain B cell survival if BCR-mediated activation of the PI3K/Akt pathway is inhibited [16]. Partially tolerized autoreactive B cells compete less well for BAFF because they have immature rafts and may have downregulated expression of the BCR; they therefore require a stronger BCR signal to generate sufficient p100. Deregulation of the TRAF3-NIK axis that is downstream of p100 releases the brake conferred by competition for BAFF; this may play an important role in lymphomagenesis [17]. The mechanisms of BAFF-R/BCR cross talk have recently been reviewed [18].

BAFF-R deficient mice have normal serum IgA levels despite profound B cell depletion, and their mesenteric lymph nodes contain normal numbers of IgA expressing germinal center B cells [19]; mucosal IgA responses preferentially depend on the interaction of APRIL with TACI [20]. Increased transitional B cells, decreased mature B cells and normal IgA levels have similarly been reported in two humans with complete BAFF-R deficiency [21]. Both BCR stimulation and CD40 ligation upregulate the expression of BAFF-R in follicular B cells and this compartment remains dependent on BAFF for its continued maintenance [22]. BAFF-R signals also regulate the expression of complement receptors on the B cell surface through a 7 amino acid intracytoplasmic site different from that required to support B cell survival [23].

While TACI does not play a major role in selection of the naïve B cell repertoire in adults, functional TACI mutations predispose to the development of common variable immunodeficiency. The most common TACI mutation A181E, in the transmembrane region, when expressed in mice, leads to deficiency of IgA and IgG1 and abnormal responses to T-independent antigens [24]. Another mutation, C104R, in the extracellular domain, is associated with hypogammaglobulinemia and a defect in memory B cell production [25,26]. TACI also facilitates class switching through a pathway involving the unique interaction of a conserved intracytoplasmic domain of TACI with the intermediary region domain of MyD88 [27]. TACI also has a negative regulatory role with respect to B cell activation; several mechanisms for this have been proposed [1]. The recent discovery that BAFF can propagate the expansion of a MZ-like subset of regulatory B cells that produce IL-10 [28] suggests another mechanism by which deficiency of TACI, that is highly expressed on the MZ subset, could lead to altered B cell homeostasis. The roles of TACI in maintaining B cell homeostasis on the one hand and in participating in immune responses on the other, could account for the seemingly contradictory observations that TACI deficiency is associated with B cell hyperactivity and autoimmunity (reviewed [1]) whereas inhibition of the TACI ligands is therapeutic in autoimmunity.

BAFF serum levels rise immediately following any period of B cell lymphopenia, subsiding again once B cell numbers return to normal. Since there are now several drugs available that deplete B cells it is important to determine whether an increase in availability of BAFF generates an autoreactive naïve B cell repertoire during B cell reconstitution. Studies in transgenic systems have shown that excess BAFF does not rescue very high affinity autoreactive B cells that are usually deleted in the bone marrow but alters the fate of autoreactive B cells with lower affinities that escape deletion in the bone marrow or that might otherwise have undergone receptor editing [29,30]. Kawabata et al used a mouse expressing an autoreactive class switched IgG2a transgene with specificity for dsDNA to demonstrate that B cell depletion results in selection of autoreactive B cells into the naïve repertoire during B cell reconstitution. Because these cells produce non-mutated but class switched antibodies that are capable of tissue penetration these authors were able to demonstrate that the germline encoded autoantibodies produced are of sufficient affinity to deposit in the kidneys [31].

The role of BAFF and APRIL during B cell activation

Myeloid dendritic cells, neutrophils and basophils release BAFF when stimulated by a variety of innate stimuli including cytokines (Type I IFN and TNFα) and Fc receptor ligation [3236]. During antigen activation BAFF upregulates TLR expression, promotes B cell survival and, in collaboration with cytokines, costimulatory signals or TLR signals promotes Ig class switching [37,38]. Activation of intracellular TLRs in B cells upregulates expression of BAFF receptors, particularly TACI [3941], and increases both BCR signaling and BAFF/APRIL sensitivity (Figure 2A). These findings help explain the dependence of T independent (type 2) responses on TACI. Since autoreactive B cells are more likely to internalize immune complexes containing autoantigens, particularly those containing nucleic acids, excess BAFF and APRIL may preferentially drive class switching of naïve autoreactive B cells. It is therefore conceivable that high levels of BAFF induced by viral infections or following B cell depletion will induce pathogenic autoimmunity in susceptible individuals by enhancing both selection and class switching of naïve autoreactive B cells. Studies of BAFF transgenic mice that express 50-100 fold increased serum BAFF levels, have indeed shown that the SLE-like autoimmunity that ensues over time is a consequence of expansion and class switching of autoreactive marginal zone and/or B1 B cells and is independent of CD4 T cells but dependent on signaling through MyD88 [40]. In contrast, our own studies have shown that the 3 fold increase in serum BAFF level induced by administration of a small dose of Type I IFN is not sufficient to induce the production of pathogenic class switched autoantibodies in T cell depleted NZB/W mice (Z Liu and A Davidson, manuscript in press).

Figure 2.

Figure 2

Amplification pathways mediated by BAFF during immune and inflammatory responses: A: During antigen activation BAFF upregulates TLR expression, promotes B cell survival and, in collaboration with cytokines, costimulatory signals or TLR signals promotes Ig class switching. Activation of intracellular TLRs in B cells upregulates expression of TACI and increases both BCR signaling and BAFF/APRIL sensitivity. (Loop 1). Immune complexes induce release of Type I IFN from plasmacytoid dendritic cells that contributes to activation of myeloid dendritic cells. Activated monocytes and dendritic cells express TACI and produce BAFF. BAFF supports survival and differentiation of monocytes and enhances cytokine and chemokine production from macrophages and dendritic cells (Loop 2). T cells may be directly stimulated by BAFF to produce inflammatory cytokines including IFNγ or, if IL-6 is also present, IL-17 (Loop 3). BAFF can also be produced by cytokine activated neutrophils and by basophils that have been activated by immune complexes (Loop 4). B: inflammatory cytokines can collaborate with BAFF in the induction of memory B cell differentiation to plasma cells thus generating more immune complexes.

N, neutrophils; Mon, monocytes; Ba, basophils; TLR, toll like receptor; IFN, interferon; BCR, B-cell receptor;

The germinal center environment contains low levels of BAFF and APRIL and germinal center B cells express only low levels of the receptors [42]. Germinal centers that support somatic mutation and class switching are initiated even if BAFF and APRIL are absent. Nevertheless both BAFF and BAFF-R are required to maintain optimal size and longevity of the germinal center response and the follicular dendritic cell network within the germinal center fails to fully mature when BAFF is absent, resulting in a decrease in magnitude of the antibody response (reviewed [43]). Whether increased serum levels of BAFF alter germinal center longevity or selection remains to be determined. As germinal B cells become either memory cells or plasma cells their differentiation program is associated with continued expression of BAFF-R and TACI (in the memory population) or with loss of BAFF-R and upregulation of BCMA (in plasma cells) [42]. Crosslinking of BCR and FcRIIB during humoral immune responses attenuates expression of BAFF-R thus blunting the effect of BAFF on cell survival [44].

BAFF and APRIL are not necessary for survival or reactivation of class switched memory B cells in vivo [4547] but unswitched IgM memory B cells are still partially BAFF dependent [46]. Similarly, in SLE patients exposed to belimumab for up to 3 years IgM+ memory B cells decreased by a median of 76% but there was no effect on class switched memory B cells [48]. Nevertheless, two studies in humans have shown that BAFF collaborates with inflammatory cytokines IL-21 or IL-17 in driving the differentiation of memory B cells to plasma cells, thus providing a role for BAFF receptor expression on these cells [49,50]. In humans treated with either belimumab or atacicept an increase in the absolute number of circulating memory cells is observed during the first 8 weeks of treatment ([51,52], W Stohl, abstract 1160 presented at ACR meeting, San Diego, 2005). Whether this is due to mobilization of cells or to proliferation of cells has not yet been investigated.

In normal adult mice long-lived plasma cells in both the spleen and bone marrow are dependent on either BAFF or APRIL, consistent with their preferential expression of BCMA [47]. Survival of the earliest plasma cells in the bone marrow appears to be more dependent on APRIL than on BAFF and the poor survival of plasma cells in neonates may be due to the attenuated expression of TACI on B cells [53] and of APRIL by neonatal bone marrow stromal cells [54]. Similarly, short-lived IgM producing plasma cells that predominantly express TACI [55], can be supported by either BAFF or APRIL [47] and are therefore depleted by TACI-Ig but not by selective BAFF inhibition [56,57]. However in several SLE prone strains TACI-Ig has no effect on long-lived bone marrow plasma cells showing that other survival factors can maintain plasma cell survival under inflammatory conditions [56,58]. Importantly, elimination of plasma cells is not required in order to achieve a therapeutic effect with BAFF blockade in mice [56,57].

In human SLE, belimumab, has little effect on plasma cells and only a very modest effect on circulating IgG levels, as expected [48,59]. Even atacicept has a more profound effect on serum levels of IgM and IgA than on IgG with only a 20% decrease in total serum IgG levels and little effect on antibody titers to recall antigens such as tetanus toxoid [51,52,60,61]. These observations in sum are consistent with the idea that fully differentiated bone marrow plasma cells in most humans are no longer entirely dependent on BAFF and APRIL. The reason why IgM-producing memory and plasma cells are more sensitive to BAFF/APRIL inhibition than are IgG-producing cells may involve signaling through the BCR as there are considerable differences in gene expression and an exaggerated calcium flux in IgG compared with IgM-bearing cells [62]. This may be due to differences in the rate of BCR clustering on the cell surface [63].

BAFF-R is expressed on some T cells; its possible role in T cell function has recently been reviewed [64]. Although T cell numbers are normal in BAFF deficient mice, T cells from SLE patients produce more IFNγ in response to BAFF than do T cells from normal individuals and monocytes from SLE patients produce more BAFF in response to IFNγ stimulation than do normal monocytes [65]. Mice with deficiency of lyn kinase, express a hyperactivated myeloid cell phenotype associated with elevated serum levels of BAFF. Adoptive transfer of BAFF-R deficient and wt T cells into these mice resulted in reduced activation and IFNγ production only in the BAFF-R deficient T cells [66].

BAFF also supports the survival of monocytes and enhances their differentiation into macrophages [67,68]. Human myeloid DCs stimulated with BAFF in vitro up-regulate co-stimulatory molecules, lose their phagocytic ability and produce inflammatory cytokines and chemokines including IL-1, IL-6, CCL2 and CCL5, inducing Th1 responses [67]. In a mouse model of arthritis, synovial dendritic cells transduced with an siRNA that silences BAFF, remain in an immature state and fail to produce the IL-6 required for the differentiation of T helper type 17 (Th17) cells [69].

These studies in sum, suggest an amplification loop in which BAFF acts on DCs to help them activate and recruit immune cells and directly enhances the proinflammatory activity of T cells; this in turn may elicit further production of BAFF (Figure 2A).

Murine studies of BAFF/APRIL inhibitors

BAFF or BAFF/APRIL inhibition have been used successfully in murine models of several autoimmune diseases including SLE, collagen induced arthritis, type I diabetes and multiple sclerosis. The studies in murine SLE have been instructive because several strains have been tested at both early and late stages of disease and both selective and non-selective inhibitors have been used [56,58]. In most SLE strains BAFF-R-Ig and TACI-Ig are equally effective at delaying disease onset but reversal of established disease is more difficult to achieve and depends on the amount of concomitant systemic inflammation [57]. Neither inhibitor prevents autoantibody formation or the deposition of autoantibodies in the kidneys however the rapid B cell depletion that accompanies BAFF inhibition results in shrinkage of secondary lymphoid organs and therefore a diminution in the total number of activated T cells and dendritic cells. This is associated with a decrease in production of the circulating inflammatory mediators that activate endothelial cells and enhance local inflammation. In the NZM2410 strain both BAFF-R-Ig and TACI-Ig are equally effective at reversing established nephritis, in part by decreasing the activation state of both circulating and renal mononuclear phagocytes. This appears to be an indirect effect of BAFF inhibition as it is sustained long after the treatment course is complete [57]. Similar results have been observed in the Lyn deficient mouse [66] although as mentioned above there is evidence in this strain that BAFF inhibition directly inhibits T cell activation, an effect that has not been observed in the other SLE strains. In contrast, in MRL/lpr mice, autoantibody producing plasma cells that are mostly generated in extrafollicular foci are highly dependent on BAFF and APRIL and serum IgG autoantibody levels plummet within 1–2 weeks of receiving TACI-Ig; this is associated with a marked decrease in renal immune complex deposition and improved survival. T cell activation and interstitial nephritis are not affected by TACI-Ig in this strain [70]. These studies highlight the heterogeneity of the responses to BAFF and BAFF/APRIL inhibition in multiple murine models of SLE and suggest that there may be subsets of humans that respond better to BAFF inhibition than others.

Human studies of BAFF/APRIL inhibitors

There are few published reports of clinical trials of either selective BAFF or non selective BAFF/APRIL inhibitors in human autoimmune diseases; most of these studies have been reported in abstract form only. Nevertheless some trends are emerging that can be reviewed here. The inhibitors have been used in three diseases, rheumatoid arthritis, SLE and multiple sclerosis.

Both belimumab and atacicept have been used for the treatment of rheumatoid arthritis [51,52]. In Phase II studies belimumab had a modest effect on disease activity whereas no effect was observed in Phase II studies with TACI-Ig. Thus these drugs have not moved forward to Phase III studies. In a phase II study, moderate, but not high or low doses, of a different anti-BAFF antibody (LY2127399; Eli Lilly) that blocks both soluble and membrane BAFF had beneficial effects in RA similar to that of TNF blockers (M Genovese, abstract 1923 presented at American College of Rheumatology Meeting, Philadelphia, 2009). This result raises interesting questions about the potential benefits of inhibiting the various BAFF forms that will need to be formally addressed if this drug continues to show efficacy in larger trials.

BAFF overexpression has been detected in the brains of mice and patients with multiple sclerosis [71] and TACI-Ig had a beneficial effect in a mouse model of MS [72]. However a phase II study of atacicept for MS had to be terminated because of disease worsening (www.clinicaltrials.gov). These findings stand in stark contrast to the beneficial effects of global B cell depletion in multiple sclerosis using anti-CD20 antibodies. IFNβ is standard treatment for MS and increases serum BAFF levels in MS patients [73]. Whether the negative effect of atacicept was due to a decrease in Type I IFN, or alterations in other cytokines such as IFNγ or IL-10 has not been determined. A clinical trial of LY2127399 began in 2009 and is ongoing.

The most exciting findings have been the positive results of two large Phase III studies of belimumab in SLE. Although a previous large Phase II study of this drug in SLE failed to meet its primary endpoints [59], the study prompted the development of a composite SLE responder index that was used as a primary endpoint in the subsequent Phase III studies [74]. In both Phase III studies, belimumab given together with standard of care therapy outperformed standard of care alone over a period of 52 weeks. This was associated with steroid sparing effects and a reduction in the number of severe flares (R.F. van Vollenhoven, abstract OP0068 presented at EULAR Congress, Rome 2010 and S. Navarra, abstract SAT0204 presented at EULAR Congress, Rome, 2010. By 76 weeks however, the difference between the two groups was no longer statistically significant although there was still some advantage to the belimumab group when other post-hoc analyses were performed (R. Furie, abstract presented at 9th International SLE Congress, Vancouver, 2010). Human Genome Sciences announced submission of a Biologics License Application for belimumab to the FDA in June 2010 that should be reviewed by the end of 2010. Mechanistic studies in humans have shown that, as predicted by the mouse physiology, and based on its selective inhibition of BAFF, belimumab depletes naïve and transitional B cells within the first 6 months of treatment and depletes IgM+ memory B cells and IgM producing plasma cells with delayed kinetics but has no effect on class switched memory B cells even after 2 years of treatment [48,59]. The effect of drug on T cell activation pathways and on monocytes remains to be determined. An important difference between the mouse and human/primate studies is that the kinetics of B cell depletion takes much longer in humans and is associated with delayed shrinkage of lymphoid organs [75]. This is consistent with the apparently delayed onset of action of belimumab.

Conclusions

It has been a little over a decade since the discovery of BAFF, the homologous molecule APRIL and the three BAFF/APRIL receptors followed by the rapid development of several therapeutic BAFF inhibitors, one of which, belimumab, has recently demonstrated efficacy in two large Phase III clinical trials for human SLE. While this has certainly been reason to celebrate, enthusiasm has been tempered by the modest improvement in disease activity measures conferred by belimumab over time. Many questions remain about the mechanisms of action of BAFF/APRIL inhibitors, including their effects on each B cell subset during periods of disease quiescence and flare, their effects on cell types other than B cells, and whether drugs with improved efficacy can be designed based on the function of the various forms of the cytokines and their receptors. Clinical questions that need to be answered include whether it will be possible to identify subsets of patients that respond better than others, what is the optimal length of time to treat patients, which other drugs might be synergistic with BAFF/APRIL inhibition, and which autoimmune diseases will be responsive to or exacerbated by this new class of drugs. Most importantly, because SLE is a disease in which damage accumulates over time, the long term advantage of a class of drugs that even modestly decreases the frequency of major flares and spares exposure to other toxic drugs needs to be determined using appropriately designed clinical trials.

Acknowledgements

The author acknowledges grant support from the NIH (R01 AR049938 and RO1 AI082037).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Mackay F, Schneider P. TACI, an enigmatic BAFF/APRIL receptor, with new unappreciated biochemical and biological properties. Cytokine Growth Factor Rev. 2008;19:263–276. doi: 10.1016/j.cytogfr.2008.04.006. [DOI] [PubMed] [Google Scholar]
  • 2.Mackay F, Schneider P. Cracking the BAFF code. Nat Rev Immunol. 2009;9:491–502. doi: 10.1038/nri2572. [DOI] [PubMed] [Google Scholar]
  • 3.Bossen C, Schneider P. BAFF, APRIL and their receptors: structure, function and signaling. Semin Immunol. 2006;18:263–275. doi: 10.1016/j.smim.2006.04.006. [DOI] [PubMed] [Google Scholar]
  • 4.Woodland RT, Fox CJ, Schmidt MR, Hammerman PS, Opferman JT, Korsmeyer SJ, Hilbert DM, Thompson CB. Multiple signaling pathways promote B lymphocyte stimulator dependent B-cell growth and survival. Blood. 2008;111:750–760. doi: 10.1182/blood-2007-03-077222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Khan WN. B cell receptor and BAFF receptor signaling regulation of B cell homeostasis. J Immunol. 2009;183:3561–3567. doi: 10.4049/jimmunol.0800933. [DOI] [PubMed] [Google Scholar]
  • 6.Lin WY, Gong Q, Seshasayee D, Lin Z, Ou Q, Ye S, Suto E, Shu J, Lee WP, Lee CW, et al. Anti-BR3 antibodies: a new class of B-cell immunotherapy combining cellular depletion and survival blockade. Blood. 2007;110:3959–3967. doi: 10.1182/blood-2007-04-088088. [DOI] [PubMed] [Google Scholar]
  • 7.Gavin AL, Duong B, Skog P, Ait-Azzouzene D, Greaves DR, Scott ML, Nemazee D. {Delta}BAFF, a Splice Isoform of BAFF, Opposes Full-Length BAFF Activity In Vivo in Transgenic Mouse Models. J Immunol. 2005;175:319–328. doi: 10.4049/jimmunol.175.1.319. [DOI] [PubMed] [Google Scholar]
  • 8.Diaz-de-Durana Y, Mantchev GT, Bram RJ, Franco A. TACI-BLyS signaling via B-cell-dendritic cell cooperation is required for naive CD8+ T-cell priming in vivo. Blood. 2006;107:594–601. doi: 10.1182/blood-2004-12-4708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Jeon ST, Kim WJ, Lee SM, Lee MY, Park SB, Lee SH, Kim IS, Suk K, Choi BK, Choi EM, et al. Reverse signaling through BAFF differentially regulates the expression of inflammatory mediators and cytoskeletal movements in THP-1 cells. Immunol Cell Biol. 2010;88:148–156. doi: 10.1038/icb.2009.75. * This paper describes the pro-inflammatory effects of reverse signaling through membrane BAFF in a macrophage cell line
  • 10. Bossen C, Cachero TG, Tardivel A, Ingold K, Willen L, Dobles M, Scott ML, Maquelin A, Belnoue E, Siegrist CA, et al. TACI, unlike BAFF-R, is solely activated by oligomeric BAFF and APRIL to support survival of activated B cells and plasmablasts. Blood. 2008;111:1004–1012. doi: 10.1182/blood-2007-09-110874. ** This paper shows that recognition of BAFF and APRIL by TACI requires multimerization of the ligands.
  • 11.Dillon SR, Harder B, Lewis KB, Moore MD, Liu H, Bukowski TR, Hamacher NB, Lantry MM, Maurer M, Krejsa CM, et al. B-lymphocyte stimulator/a proliferation-inducing ligand heterotrimers are elevated in the sera of patients with autoimmune disease and are neutralized by atacicept and B-cell maturation antigen-immunoglobulin. Arthritis Res Ther. 2010;12:R48. doi: 10.1186/ar2959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Smirnova AS, Andrade-Oliveira V, Gerbase-Delima M. Identification of new splice variants of the genes BAFF and BCMA. Mol Immunol. 2008;45:1179–1183. doi: 10.1016/j.molimm.2007.07.028. [DOI] [PubMed] [Google Scholar]
  • 13. Stadanlick JE, Kaileh M, Karnell FG, Scholz JL, Miller JP, Quinn WJ, 3rd, Brezski RJ, Treml LS, Jordan KA, Monroe JG, et al. Tonic B cell antigen receptor signals supply an NF-kappaB substrate for prosurvival BLyS signaling. Nat Immunol. 2008;9:1379–1387. doi: 10.1038/ni.1666. ** This paper shows that BCR signals and BAFF_R signals cooperate in mediating B cell survival as BCR signals provide the substrate p100 that is required for productive signalling through the alternative NFκB pathway.
  • 14.Castro I, Wright JA, Damdinsuren B, Hoek KL, Carlesso G, Shinners NP, Gerstein RM, Woodland RT, Sen R, Khan WN. B cell receptor-mediated sustained c-Rel activation facilitates late transitional B cell survival through control of B cell activating factor receptor and NF-kappaB2. J Immunol. 2009;182:7729–7737. doi: 10.4049/jimmunol.0803281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hoek KL, Carlesso G, Clark ES, Khan WN. Absence of mature peripheral B cell populations in mice with concomitant defects in B cell receptor and BAFF-R signaling. J Immunol. 2009;183:5630–5643. doi: 10.4049/jimmunol.0901100. [DOI] [PubMed] [Google Scholar]
  • 16.Srinivasan L, Sasaki Y, Calado DP, Zhang B, Paik JH, DePinho RA, Kutok JL, Kearney JF, Otipoby KL, Rajewsky K. PI3 kinase signals BCR-dependent mature B cell survival. Cell. 2009;139:573–586. doi: 10.1016/j.cell.2009.08.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sasaki Y, Calado DP, Derudder E, Zhang B, Shimizu Y, Mackay F, Nishikawa S, Rajewsky K, Schmidt-Supprian M. NIK overexpression amplifies, whereas ablation of its TRAF3-binding domain replaces BAFF:BAFF-R-mediated survival signals in B cells. Proc Natl Acad Sci U S A. 2008;105:10883–10888. doi: 10.1073/pnas.0805186105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Mackay F, Figgett WA, Saulep D, Lepage M, Hibbs ML. B-cell stage and context-dependent requirements for survival signals from BAFF and the B-cell receptor. Immunol Rev. 2010;237:205–225. doi: 10.1111/j.1600-065X.2010.00944.x. * This is an excellent review of BAFF signaling pathways.
  • 19.Sasaki Y, Casola S, Kutok JL, Rajewsky K, Schmidt-Supprian M. TNF Family Member B Cell-Activating Factor (BAFF) Receptor-Dependent and -Independent Roles for BAFF in B Cell Physiology. J Immunol. 2004;173:2245–2252. doi: 10.4049/jimmunol.173.4.2245. [DOI] [PubMed] [Google Scholar]
  • 20.He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J, Shan M, Chadburn A, Villanacci V, Plebani A, et al. Intestinal bacteria trigger T cell-independent immunoglobulin A(2) class switching by inducing epithelial-cell secretion of the cytokine APRIL. Immunity. 2007;26:812–826. doi: 10.1016/j.immuni.2007.04.014. [DOI] [PubMed] [Google Scholar]
  • 21.Warnatz K, Salzer U, Rizzi M, Fischer B, Gutenberger S, Bohm J, Kienzler AK, Pan-Hammarstrom Q, Hammarstrom L, Rakhmanov M, et al. B-cell activating factor receptor deficiency is associated with an adult-onset antibody deficiency syndrome in humans. Proc Natl Acad Sci U S A. 2009;106:13945–13950. doi: 10.1073/pnas.0903543106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Swee LK, Tardivel A, Schneider P, Rolink A. Rescue of the mature B cell compartment in BAFF-deficient mice by treatment with recombinant Fc-BAFF. Immunol Lett. 2010 doi: 10.1016/j.imlet.2010.03.007. * This paper confirms that mature B cells are dependent on BAFF throughout their lifespan.
  • 23.Mayne CG, Amanna IJ, Hayes CE. Murine BAFF-receptor residues 168–175 are essential for optimal CD21/35 expression but dispensable for B cell survival. Mol Immunol. 2009;47:590–599. doi: 10.1016/j.molimm.2009.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lee JJ, Rauter I, Garibyan L, Ozcan E, Sannikova T, Dillon SR, Cruz AC, Siegel RM, Bram R, Jabara H, et al. The murine equivalent of the A181E TACI mutation associated with common variable immunodeficiency severely impairs B-cell function. Blood. 2009;114:2254–2262. doi: 10.1182/blood-2008-11-189720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wood PM. Primary antibody deficiency syndromes. Curr Opin Hematol. 2010;17:356–361. doi: 10.1097/MOH.0b013e328338f69e. [DOI] [PubMed] [Google Scholar]
  • 26.Salzer U, Bacchelli C, Buckridge S, Pan-Hammarstrom Q, Jennings S, Lougaris V, Bergbreiter A, Hagena T, Birmelin J, Plebani A, et al. Relevance of biallelic versus monoallelic TNFRSF13B mutations in distinguishing disease-causing from risk-increasing TNFRSF13B variants in antibody deficiency syndromes. Blood. 2009;113:1967–1976. doi: 10.1182/blood-2008-02-141937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.He B, Santamaria R, Xu W, Cols M, Chen K, Puga I, Shan M, Xiong H, Bussel JB, Chiu A, et al. The transmembrane activator TACI triggers immunoglobulin class switching by activating B cells through the adaptor MyD88. Nat Immunol. 2010;11:836–845. doi: 10.1038/ni.1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yang M, Sun L, Wang S, Ko KH, Xu H, Zheng BJ, Cao X, Lu L. Novel function of B cell-activating factor in the induction of IL-10-producing regulatory B cells. J Immunol. 2010;184:3321–3325. doi: 10.4049/jimmunol.0902551. [DOI] [PubMed] [Google Scholar]
  • 29.Thien M, Phan TG, Gardam S, Amesbury M, Basten A, Mackay F, Brink R. Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches. Immunity. 2004;20:785–798. doi: 10.1016/j.immuni.2004.05.010. [DOI] [PubMed] [Google Scholar]
  • 30.Ota M, Duong BH, Torkamani A, Doyle CM, Gavin AL, Ota T, Nemazee D. Regulation of the B Cell Receptor Repertoire and Self-Reactivity by BAFF. J Immunol. 185:4128–4136. doi: 10.4049/jimmunol.1002176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kawabata D, Venkatesh J, Ramanujam M, Davidson A, Grimaldi CM, Diamond B. Enhanced selection of high affinity DNA-reactive B cells following cyclophosphamide treatment in mice. PLoS One. 2010;5:e8418. doi: 10.1371/journal.pone.0008418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Scapini P, Bazzoni F, Cassatella MA. Regulation of B-cell-activating factor (BAFF)/B lymphocyte stimulator (BLyS) expression in human neutrophils. Immunol Lett. 2008;116:1–6. doi: 10.1016/j.imlet.2007.11.009. [DOI] [PubMed] [Google Scholar]
  • 33.Li X, Su K, Ji C, Szalai AJ, Wu J, Zhang Y, Zhou T, Kimberly RP, Edberg JC. Immune opsonins modulate BLyS/BAFF release in a receptor-specific fashion. J Immunol. 2008;181:1012–1018. doi: 10.4049/jimmunol.181.2.1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Boule MW, Broughton C, Mackay F, Akira S, Marshak-Rothstein A, Rifkin IR. Toll-like Receptor 9-Dependent and -Independent Dendritic Cell Activation by Chromatin-Immunoglobulin G Complexes. J Exp Med. 2004;199:1631–1640. doi: 10.1084/jem.20031942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Charles N, Hardwick D, Daugas E, Illei GG, Rivera J. Basophils and the T helper 2 environment can promote the development of lupus nephritis. Nat Med. 2010;16:701–707. doi: 10.1038/nm.2159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Chen K, Xu W, Wilson M, He B, Miller NW, Bengten E, Edholm ES, Santini PA, Rath P, Chiu A, et al. Immunoglobulin D enhances immune surveillance by activating antimicrobial, proinflammatory and B cell-stimulating programs in basophils. Nat Immunol. 2009;10:889–898. doi: 10.1038/ni.1748. * This paper is the first to show that basophils stimulated by immune complexes release BAFF.
  • 37.Xu W, Santini PA, Matthews AJ, Chiu A, Plebani A, He B, Chen K, Cerutti A. Viral double-stranded RNA triggers Ig class switching by activating upper respiratory mucosa B cells through an innate TLR3 pathway involving BAFF. J Immunol. 2008;181:276–287. doi: 10.4049/jimmunol.181.1.276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.He B, Qiao X, Cerutti A. CpG DNA induces IgG class switch DNA recombination by activating human B cells through an innate pathway that requires TLR9 and cooperates with IL-10. J Immunol. 2004;173:4479–4491. doi: 10.4049/jimmunol.173.7.4479. [DOI] [PubMed] [Google Scholar]
  • 39.Treml LS, Carlesso G, Hoek KL, Stadanlick JE, Kambayashi T, Bram RJ, Cancro MP, Khan WN. TLR stimulation modifies BLyS receptor expression in follicular and marginal zone B cells. J Immunol. 2007;178:7531–7539. doi: 10.4049/jimmunol.178.12.7531. [DOI] [PubMed] [Google Scholar]
  • 40.Groom JR, Fletcher CA, Walters SN, Grey ST, Watt SV, Sweet MJ, Smyth MJ, Mackay CR, Mackay F. BAFF and MyD88 signals promote a lupuslike disease independent of T cells. J Exp Med. 2007;204:1959–1971. doi: 10.1084/jem.20062567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Good KL, Avery DT, Tangye SG. Resting human memory B cells are intrinsically programmed for enhanced survival and responsiveness to diverse stimuli compared to naive B cells. J Immunol. 2009;182:890–901. doi: 10.4049/jimmunol.182.2.890. [DOI] [PubMed] [Google Scholar]
  • 42.Darce JR, Arendt BK, Wu X, Jelinek DF. Regulated Expression of BAFF-Binding Receptors during Human B Cell Differentiation. J Immunol. 2007;179:7276–7286. doi: 10.4049/jimmunol.179.11.7276. [DOI] [PubMed] [Google Scholar]
  • 43.Kalled SL. Impact of the BAFF/BR3 axis on B cell survival, germinal center maintenance and antibody production. Semin Immunol. 2006;18:290–296. doi: 10.1016/j.smim.2006.06.002. [DOI] [PubMed] [Google Scholar]
  • 44.Crowley JE, Stadanlick JE, Cambier JC, Cancro MP. FcgammaRIIB signals inhibit BLyS signaling and BCR-mediated BLyS receptor up-regulation. Blood. 2009;113:1464–1473. doi: 10.1182/blood-2008-02-138651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ramanujam M, Wang X, Huang W, Schiffer L, Grimaldi C, Akkerman A, Diamond B, Madaio MP, Davidson A. Mechanism of action of transmembrane activator and calcium modulator ligand interactor-Ig in murine systemic lupus erythematosus. J Immunol. 2004;173:3524–3534. doi: 10.4049/jimmunol.173.5.3524. [DOI] [PubMed] [Google Scholar]
  • 46. Scholz JL, Crowley JE, Tomayko MM, Steinel N, O'Neill PJ, Quinn WJ, 3rd, Goenka R, Miller JP, Cho YH, Long V, et al. BLyS inhibition eliminates primary B cells but leaves natural and acquired humoral immunity intact. Proc Natl Acad Sci U S A. 2008;105:15517–15522. doi: 10.1073/pnas.0807841105. * This paper shows that IgM memory cells are more dependent on BAFF and APRIL than are IgG memory cells.
  • 47. Benson MJ, Dillon SR, Castigli E, Geha RS, Xu S, Lam KP, Noelle RJ. Cutting edge: the dependence of plasma cells and independence of memory B cells on BAFF and APRIL. J Immunol. 2008;180:3655–3659. doi: 10.4049/jimmunol.180.6.3655. * This paper shows that memory cells are independent of BAFF and APRIL whereas long-lived plasma cells need either BAFF or APRIL to survive.
  • 48.Jacobi AM, Huang W, Wang T, Freimuth W, Sanz I, Furie R, Mackay M, Aranow C, Diamond B, Davidson A. Effect of long-term belimumab treatment on b cells in systemic lupus erythematosus: Extension of a phase II, double-blind, placebo-controlled, dose-ranging study. Arthritis Rheum. 2009;62:201–210. doi: 10.1002/art.27189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Doreau A, Belot A, Bastid J, Riche B, Trescol-Biemont MC, Ranchin B, Fabien N, Cochat P, Pouteil-Noble C, Trolliet P, et al. Interleukin 17 acts in synergy with B cell-activating factor to influence B cell biology and the pathophysiology of systemic lupus erythematosus. Nat Immunol. 2009;10:778–785. doi: 10.1038/ni.1741. * This paper shows that BAFF collaborates with IL-17 in inducing memory B cell differentiation to plasma cells in lupus patients.
  • 50.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]
  • 51.Pena-Rossi C, Nasonov E, Stanislav M, Yakusevich V, Ershova O, Lomareva N, Saunders H, Hill J, Nestorov I. An exploratory dose-escalating study investigating the safety, tolerability, pharmacokinetics and pharmacodynamics of intravenous atacicept in patients with systemic lupus erythematosus. Lupus. 2009;18:547–555. doi: 10.1177/0961203309102803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tak PP, Thurlings RM, Rossier C, Nestorov I, Dimic A, Mircetic V, Rischmueller M, Nasonov E, Shmidt E, Emery P, et al. Atacicept in patients with rheumatoid arthritis: results of a multicenter, phase Ib, double-blind, placebo-controlled, dose-escalating, single- and repeated-dose study. Arthritis Rheum. 2008;58:61–72. doi: 10.1002/art.23178. [DOI] [PubMed] [Google Scholar]
  • 53.Kanswal S, Katsenelson N, Selvapandiyan A, Bram RJ, Akkoyunlu M. Deficient TACI expression on B lymphocytes of newborn mice leads to defective Ig secretion in response to BAFF or APRIL. J Immunol. 2008;181:976–990. doi: 10.4049/jimmunol.181.2.976. [DOI] [PubMed] [Google Scholar]
  • 54.Belnoue E, Pihlgren M, McGaha TL, Tougne C, Rochat AF, Bossen C, Schneider P, Huard B, Lambert PH, Siegrist CA. APRIL is critical for plasmablast survival in the bone marrow and poorly expressed by early-life bone marrow stromal cells. Blood. 2008;111:2755–2764. doi: 10.1182/blood-2007-09-110858. [DOI] [PubMed] [Google Scholar]
  • 55.Hondowicz BD, Alexander ST, Quinn WJ, 3rd, Pagan AJ, Metzgar MH, Cancro MP, Erikson J. The role of BLyS/BLyS receptors in anti-chromatin B cell regulation. Int Immunol. 2007;19:465–475. doi: 10.1093/intimm/dxm011. [DOI] [PubMed] [Google Scholar]
  • 56.Ramanujam M, Wang X, Huang W, Liu Z, Schiffer L, Tao H, Frank D, Rice J, Diamond B, Yu KO, et al. Similarities and differences between selective and nonselective BAFF blockade in murine SLE. J Clin Invest. 2006;116:724–734. doi: 10.1172/JCI26385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Ramanujam M, Bethunaickan R, Huang W, Tao H, Madaio MP, Davidson A. Selective blockade of BAFF for the prevention and treatment of systemic lupus erythematosus nephritis in NZM2410 mice. Arthritis Rheum. 2010;62:1457–1468. doi: 10.1002/art.27368. * This paper shows that BAFF inhibition prevents the activation of circulating monocytes and renal macrophages in a lupus model.
  • 58.Kahn P, Ramanujam M, Bethunaickan R, Huang W, Tao H, Madaio MP, Factor SM, Davidson A. Prevention of murine antiphospholipid syndrome by BAFF blockade. Arthritis Rheum. 2008;58:2824–2834. doi: 10.1002/art.23764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Wallace DJ, Stohl W, Furie RA, Lisse JR, McKay JD, Merrill JT, Petri MA, Ginzler EM, Chatham WW, McCune WJ, et al. A phase II, randomized, double-blind, placebo-controlled, dose-ranging study of belimumab in patients with active systemic lupus erythematosus. Arthritis Rheum. 2009;61:1168–1178. doi: 10.1002/art.24699. ** The first published human Phase II clinical trial of a BAFF inhibitor in lupus.
  • 60.Dall'Era M, Chakravarty E, Wallace D, Genovese M, Weisman M, Kavanaugh A, Kalunian K, Dhar P, Vincent E, Pena-Rossi C, et al. Reduced B lymphocyte and immunoglobulin levels after atacicept treatment in patients with systemic lupus erythematosus: results of a multicenter, phase Ib, double-blind, placebo-controlled, dose-escalating trial. Arthritis Rheum. 2007;56:4142–4150. doi: 10.1002/art.23047. [DOI] [PubMed] [Google Scholar]
  • 61.Nestorov I, Munafo A, Papasouliotis O, Visich J. Pharmacokinetics and biological activity of atacicept in patients with rheumatoid arthritis. J Clin Pharmacol. 2008;48:406–417. doi: 10.1177/0091270008315312. [DOI] [PubMed] [Google Scholar]
  • 62.Horikawa K, Martin SW, Pogue SL, Silver K, Peng K, Takatsu K, Goodnow CC. Enhancement and suppression of signaling by the conserved tail of IgG memory-type B cell antigen receptors. J Exp Med. 2007;204:759–769. doi: 10.1084/jem.20061923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Liu W, Meckel T, Tolar P, Sohn HW, Pierce SK. Intrinsic properties of immunoglobulin IgG1 isotype-switched B cell receptors promote microclustering and the initiation of signaling. Immunity. 2010;32:778–789. doi: 10.1016/j.immuni.2010.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mackay F, Leung H. The role of the BAFF/APRIL system on T cell function. Semin Immunol. 2006;18:284–289. doi: 10.1016/j.smim.2006.04.005. [DOI] [PubMed] [Google Scholar]
  • 65. Harigai M, Kawamoto M, Hara M, Kubota T, Kamatani N, Miyasaka N. Excessive production of IFN-gamma in patients with systemic lupus erythematosus and its contribution to induction of B lymphocyte stimulator/B cell-activating factor/TNF ligand superfamily-13B. J Immunol. 2008;181:2211–2219. doi: 10.4049/jimmunol.181.3.2211. * This paper shows that T cells and monocytes from SLE patients are more sensitive to the activating effects of BAFF than are T cells and monocytes from normal individuals.
  • 66.Scapini P, Hu Y, Chu CL, Migone TS, Defranco AL, Cassatella MA, Lowell CA. Myeloid cells, BAFF, and IFN-{gamma} establish an inflammatory loop that exacerbates autoimmunity in Lyn-deficient mice. J Exp Med. 2010 doi: 10.1084/jem.20100086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chang SK, Arendt BK, Darce JR, Wu X, Jelinek DF. A role for BLyS in the activation of innate immune cells. Blood. 2006;108:2687–2694. doi: 10.1182/blood-2005-12-017319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Chang SK, Mihalcik SA, Jelinek DF. B lymphocyte stimulator regulates adaptive immune responses by directly promoting dendritic cell maturation. J Immunol. 2008;180:7394–7403. doi: 10.4049/jimmunol.180.11.7394. * This paper shows that BAFF can activate human monocytes
  • 69. Lai Kwan Lam Q, King Hung Ko O, Zheng BJ, Lu L. Local BAFF gene silencing suppresses Th17-cell generation and ameliorates autoimmune arthritis. Proc Natl Acad Sci U S A. 2008;105:14993–14998. doi: 10.1073/pnas.0806044105. ** This paper shows that local silencing of BAFF in the synovium results in deactivation of dendritic cells and prevention of local TH17 differentiation
  • 70.Liu W, Szalai A, Zhao L, Liu D, Martin F, Kimberly RP, Zhou T, Carter RH. Control of spontaneous B lymphocyte autoimmunity with adenovirus-encoded soluble TACI. Arthritis Rheum. 2004;50:1884–1896. doi: 10.1002/art.20290. [DOI] [PubMed] [Google Scholar]
  • 71.Thangarajh M, Gomes A, Masterman T, Hillert J, Hjelmstrom P. Expression of B-cell-activating factor of the TNF family (BAFF) and its receptors in multiple sclerosis. J Neuroimmunol. 2004;152:183–190. doi: 10.1016/j.jneuroim.2004.03.017. [DOI] [PubMed] [Google Scholar]
  • 72.Huntington ND, Tomioka R, Clavarino C, Chow AM, Linares D, Mana P, Rossjohn J, Cachero TG, Qian F, Kalled SL, et al. A BAFF antagonist suppresses experimental autoimmune encephalomyelitis by targeting cell-mediated and humoral immune responses. Int Immunol. 2006;18:1473–1485. doi: 10.1093/intimm/dxl080. [DOI] [PubMed] [Google Scholar]
  • 73.Krumbholz M, Faber H, Steinmeyer F, Hoffmann LA, Kumpfel T, Pellkofer H, Derfuss T, Ionescu C, Starck M, Hafner C, et al. Interferon-beta increases BAFF levels in multiple sclerosis: implications for B cell autoimmunity. Brain. 2008;131:1455–1463. doi: 10.1093/brain/awn077. [DOI] [PubMed] [Google Scholar]
  • 74.Furie RA, Petri MA, Wallace DJ, Ginzler EM, Merrill JT, Stohl W, Chatham WW, Strand V, Weinstein A, Chevrier MR, et al. Novel evidence-based systemic lupus erythematosus responder index. Arthritis Rheum. 2009;61:1143–1151. doi: 10.1002/art.24698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Halpern WG, Lappin P, Zanardi T, Cai W, Corcoran M, Zhong J, Baker KP. Chronic administration of belimumab, a BLyS antagonist, decreases tissue and peripheral blood B-lymphocyte populations in cynomolgus monkeys: pharmacokinetic, pharmacodynamic, and toxicologic effects. Toxicol Sci. 2006;91:586–599. doi: 10.1093/toxsci/kfj148. [DOI] [PubMed] [Google Scholar]

RESOURCES