Sie sind auf Seite 1von 18

|

Received: 31 October 2018    Accepted: 28 November 2018

DOI: 10.1111/imr.12731

INVITED REVIEW

Germinal center B cell initiation, GC maturation, and the


coevolution of its stromal cell niches

Ann M. Haberman1,2  | David G. Gonzalez1,3 | Patrick Wong1 | 


Ting-ting Zhang1 | Steven M. Kerfoot4

1
Department of Immunobiology, Yale
University, New Haven, Connecticut Summary
2
Department of Laboratory Medicine, Yale Throughout the developing GC response, B cell survival and fate choices made at the
University, New Haven, Connecticut
single cell level are dependent on signals received largely through interactions with
3
Department of Genetics, Yale University,
other cells, often with cognate T cells. The type of signals that a given B cell can en-
New Haven, Connecticut
4
Department of Microbiology and counter is dictated by its location within tissue microarchitecture. The focus of this
Immunology, Western University, London, review is on the initiation and evolution of the GC response at the earliest time
ON, Canada
points. Here, we review the key factors influencing the progression of GC B cell
Correspondence ­differentiation that are both stage and context dependent. Finally, we describe the
Ann M. Haberman, Department of
Immunobiology, Yale University, New Haven, coevolution of niches within and surrounding the GC that influence the outcome of
Connecticut. the GC response.
Email: ann.haberman@yale.edu

KEYWORDS
Funding information
Multiple Sclerosis Society of Canada; B cells, cell differentiation, cytokines, lineage commitment/specification, stromal cells,
National Institute of Arthritis and transcription factors
Musculoskeletal and Skin Diseases, Grant/
Award Number: P30AR053495; National
Institute of Allergy and Infectious Diseases,
Grant/Award Number: R01AI080850 and
R21AI101704

1 | I NTRO D U C TI O N the same time, antigen-­specific B cells become activated after encoun-
tering antigen in its intact, unprocessed form and experiencing B c­ ell
Effective immune responses to pathogens and vaccines critically receptor (BCR) ligation. B cells that have internalized antigen bound to
depend on the generation of high-­affinity antibodies. The steady in- its specific BCR can then also present processed components in the
crease in antibody affinity during adaptive immune responses, known context of surface MHC II. A series of coordinate interactions between
as affinity maturation, requires the formation of an organized cluster of activated T and B cells specific for the same (cognate) antigen are the
proliferating B and T lymphocytes in germinal centers (GC). Germinal foundation for B cell activation throughout the GC response, both
center responses are initiated within secondary lymphoid tissues during the initial pre-­GC phase and within the mature GC.
that are inherently organized, where antigen-­inexperienced T and B The GCs that emerge within B cell follicles of secondary lym-
cells are separated into T-­cell zones and B cell follicles. CD4+ T cells phoid tissue are highly organized structures, within which GC B cells
expressing T-­cell receptors (TCRs) specific for newly arriving antigen compete to acquire additional antigen and engage follicular helper
are activated to both proliferate and differentiate through interactions T cells. With contemporary thinking, GC B cells expressing BCR
with dendritic cells (DCs) that have internalized antigen and presented with higher affinities are selected to further proliferate, while less
proteolytically degraded fragments on surface MHC II molecules. At fit B cells expressing BCRs with weaker affinities may not receive
sufficient survival or instructive signals and undergo apoptotic cell
death. In addition to survival, T/B interactions also drive differenti-
This article is part of a series of reviews covering Germinal Center and Extrafollicular
Immune Responses appearing in Volume 288 of Immunological Reviews. ation of GC B cells into one of two lineages that are fundamental to

10  |  wileyonlinelibrary.com/journal/imr
© 2019 John Wiley & Sons A/S. Immunological Reviews. 2019;288:10–27.
Published by John Wiley & Sons Ltd
HABERMAN et al. |
      11

durable immunity; long-­lived high-­affinity plasma cells and memory


B cells.1-4 In this way, GCs mold, amplify, and add persistence to the GC
most effective B cells of the immune response. Follicle
Throughout the developing GC response, B cell survival and fate Interfollicular zone
choices made at the single cell level are dependent on signals received Sub-SCS
largely through interactions with other cells, often with cognate T T/B border
cells. The type of signals that a given B cell can encounter is dictated T cell zone
by its location within tissue microarchitecture. A major component FDC
of B cell activation and differentiation in the GC response involves
F I G U R E   1   Lymph node tissue compartments influencing the
sequential changes in migration patterns that move the cell from one
developmental stages of germinal center B cells
specialized zone to the next. The focus of this review is on the initia-
tion and evolution of the GC response at the earliest time points. We'll
review the key factors influencing the progression of GC B cell differ- express lower levels of a variety of surface markers, including BCRs,
entiation that are both stage and context dependent. The review will CD40, and CD86, but a higher level of Bcl6 and CXCR4 compared
finish with a discussion of the niches within and surrounding the GC to their LZ counterparts.32-34 Many LZ-­resident GC B cells, aka cen-
that may influence the outcome of the GC response. trocytes, are smaller in size and are typically CD86hi, CD83hi, and
CXCR4lo.33 Although these phenotypic differences have come to be
viewed as surrogate markers of GC B cell location, it is important
2 | L A N D M A R K S A N D U N I Q U E FE AT U R E S
to note that B cells residing within each zone are far from homoge-
O F E S TA B LI S H E D G C S
neous. GCs are consistently oriented within B cell follicles (Figure 1).
The LZ is reproducibly more proximal to the subcapsular sinus of
2.1 | Tissue compartments of mature GCs
lymph nodes or the marginal sinus enveloping the white pulp of
To better appreciate the similarities and disparities of the initial GC spleens. The DZ balloons beneath the FDC network, extending to-
response, we begin with an overview of the fundamentals of fully ward the border of the follicle with the T-­cell zone.
mature GCs. As the newly established GC expands, non-­responding
B cells are peripherally displaced and surround the emerging GC,
2.2 | Selection of higher-­affinity variants, interzonal
forming what is known as the follicular mantle. Mature GCs have
migration, and cyclic reprogramming
a complex architecture that is comprised of two prominent subdo-
mains, the light zone (LZ) and dark zone (DZ), originally named for During their extensive clonal expansion, B cells with higher-­affinity
their relative appearance in hematoxylin/eosin-­stained tissue sec- BCR variants are selectively enriched.35-38 GC B cells express high
tions.5-9 The LZ is distinguished by the presence of the fine pro- levels of activation induced cytidine deaminase that drives a unique
cesses of a stromal cell subset, follicular dendritic cells (FDCs), which process of somatic hypermutation (SHM) of immunoglobulin (Ig)
entirely comprise the stromal cell matrix in this region and form a genes that can introduce amino acid substitutions in the antibodies
8
reticular network that GC B cells infiltrate. Within primary follicles produced.39,40 Because SHM is introduced during DNA replication, it
FDCs are a key source of CXCL13, a chemokine that is requisite for was thought to occur in the DZ where mitotic cells are more evident,
the correct organization of GCs into compartmentalized DZ and especially in tonsils.32,33,41,42 Selection for higher-­affinity B cells has
10 been proposed to result from competition for the acquisition of anti-
LZs. FDCs retain deposits of activated complement components
and antigen-­antibody complexes via the low-­affinity Fc receptor for gen.6,43,44 As FDCs are the most evident site of antigen accumulation
IgG (FcγRIIb) and the complement receptors CD21 and CD35.11-15 within follicles, and have the ability to promote the survival of GC
FDCs can serve as long-­term depots of undegraded Ag,11,13,15-17 and B cells that are prone to apoptosis, the selection process is thought
for this reason they have classically been considered an important to occur within the LZ compartment.45-47 Early thinking on GC B ­cell
site of antigen presentation to B cells. Intrafollicular CD4+ T-­helper selection envisioned that a key element involved the crosslinking
cells known as T follicular helpers (Tfh) are predominantly found of BCRs while in contact with antigen-­laden FDCs.48-50 However,
within the LZ, sometimes in great numbers, but are also present a recent study by Luo et al51,52 indicates that some aspects of ca-
5,18,19 nonical BCR signaling are rewired in Bcl6hi GC B cells, such that
throughout the follicle mantle. Tfh are critical to the formation
and maintenance of a productive GC, largely due to their provision BCR crosslinking per se cannot fully instruct the extensive self-­
20-26 renewal observed within mature GCs. Other studies strongly sup-
of IL-­21, IL-­4, and CD40. At the peak of the response, Tfh can be
distinguished from other cell subsets by a high level of expression of port a model in which positive selection requires GC B cells to form
the molecules PD-­1, ICOS, and the chemokine receptor CXCR5. 27-30 immunological synapses with Tfh cells53-55 and reviewed in.56,57 B
The DZ contains a higher density of proliferating cells and har- cells with higher affinity acquire more protein antigen, resulting in
bors another stromal cell type that produces CXCL12, the ligand increased surface presentation of MHC II-­antigen complexes, and
for CXCR4, and lacks the distinctive CD35 expression of FDCs.31,32 a greater capacity to stimulate Tfh cells with a cognate specific-
The majority of DZ-­resident B cells, also referred to as centroblasts, ity.33,58-62 It is important to note that BCR signaling during antigen
|
12       HABERMAN et al.

acquisition and Tfh cell engagement need not be mutually exclusive response, some of these markers are also expressed by developing
and could be synergistic. In support of this, the combination of BCR plasmablasts.89,90 A further challenge to the study of early B cell ac-
and CD40 agonism more robustly invokes cMYC expression and fur- tivation events is the downregulation of surface BCR after crosslink-
ther cell division than either alone.52 Other models for affinity matu- ing, making antigen specificity hard to confirm in some cases. A
ration postulated forms of negative selection, which could operate superior indicator of this lineage commitment emerged when the
48,63-66
in concert with positive selection. requisite role of Bcl6 was demonstrated, empowering investigations
To reconcile a perceived need for sequential events to occur in into the early steps in this pathway that were previously enigmatic.
spatially separated regions of the GC, the “cyclic re-­entry” model Expression of the transcriptional repressor Bcl6 is required for both
was proposed.67,68 With a contemporary view of this model, DZ-­ Tfh and GC B cell differentiation, a process that involves shifts in
resident B cells that have exited cell cycle lose the transcriptional the transcription of other influential TFs.91-95 In order to more easily
profile typical of centroblasts and with lower CXCR4 levels subse- identify and track the behavior of antigen-­specific T and B cells, we
quently migrate to the LZ.10,32,33,69 Intravital imaging studies of GC and others have developed strains of mice that ubiquitously express
B cell migration patterns are consistent with infrequent movement fluorescent proteins such as GFP or RFP and harbor transgenic or
59,70-72
between compartments. Within the LZ, the recent immigrants knockin BCR or TCR alleles. With the transfer of such fluorescently
compete for access to antigen, potentially experience BCR crosslink- tagged cells, specific cells can be interrogated while remaining ag-
ing during antigen acquisition, and then compete for contacts with nostic on all other phenotypic aspects during their activation and as
Tfh cells that selectively provide factors such as CD40L, IL-­21, and they undergo significant change during subsequent differentiation.
52,73,74
IL-­4. Foxo1, cMyc, and AP4 are among the transcription fac- The creation of a productive GC involves step-­wise changes to
tors known to play a critical role in the transition of such positively the transcriptional profiles and phenotype of responding lympho-
selected centrocytes to the centroblast transcriptional program, al- cytes, often including shifts in their chemotactic tendencies. Prior
though expression of these transcription factors (TFs) can be short to activation, antigen-­naïve B and T cells are segregated into B cell
34,75-78
lived. Thus, the LZ is currently seen as the compartment in follicles and T-­cell zones (Figure 1). This is the result of migration
which both self-­renewal and differentiation toward long-­lived effec- patterns not dictated by structural or anatomical limits, but rather
tor lineages are initiated, the latter evidenced by the expression of by localized expression of chemotactic agents drawing in B and
antibody secreting cell (ASC)-­associated TFs by B cells within the LZ T cells to their respective zones based on their expression of recep-
55,79-82
harboring the majority of Tfh cells. Self-­renewing GC B cells tors for those agents. By virtue of B cell expression of CXCR5, they
undergo a centrocyte to centroblast transition that is characterized are attracted to its ligand chemokine CXCL13 that is produced by
by a reciprocal increase in CXCR4, a decrease in costimulatory mol- FDCs centrally positioned in follicles96,97 and by marginal reticular
ecules, initiation of cell division, and their return to the DZ to com- cells (MRCs) that line the subcaspular sinus (SCS) of LNs and splenic
33
plete cell division there. Within the DZ, centroblasts reduce their marginal sinus (SMS).98-100 T-­cell localization to the T-­cell zone is me-
surface BCR and recycle existing MHC II complexes, and in this way diated by their expression of CCR7, the receptor for CCL19/21.101,102
have an opportunity to better express their newly mutated BCR that An additional chemoattractant within lymphoid tissue includes the
can be more fully tested when antigen is next encountered.83,84 compound EBI2L that is enzymatically modified by stroma lining the
The bulk of research on GCs has interrogated mature GCs. SCS and SMS. EBI2L is also found at the cortical sinuses descending
However, recent studies have implications on its initiation and de- through the interfollicular (IF) regions, at the border between T-­cell
velopment. The focus of the remainder of this review is on the pro- zones and follicles (T-­B border), as well as at the bridging channels
gression of the GC response at the earliest time points, both of the positioned between follicles in spleens (Figure 1).103-109 The com-
responding B cells and within the evolving stromal compartments binatorial expression of even just these three chemoattractants by
that coordinate the context of their cellular interactions. stromal cell populations establishes multiple distinct architectural
domains within which lymphocytes with similar tropisms can aggre-
gate (Figure 1).
3 |  T- ­B C E LL CO NTAC T S D U R I N G G C With an influx of inflammatory material via lymph to lymph nodes,
I N ITI ATI O N or via blood in the spleen, B and T cells specific for any newly arriving
antigens become activated and adjust their migratory propensities.
Exploration of the earliest stages of GC development had long been Within lymph nodes, naïve B cells within follicles first encounter
hampered by the seemingly inscrutable developmental steps that their cognate antigen delivered via lymphatic vessels, cortical si-
preceded the appearance of the classical surface markers of GC B nuses, or at the surface of SCS lining macrophages.110-116 Exposure
cells. Markers typically ascribed to GC B cells include high levels of to pathogen-­associated molecular motifs and BCR crosslinking result
CD95 (Fas), the GL7 epitope, a loss of IgD, and an increased ability to in increased levels of CCR7, CD86, and MHC II, as well as receptivity
85-87
bind peanut agglutinin (PNA). B cells within established murine to CD40 signaling.117,118 With this increase in CCR7, activated B cells
GCs have uniformly low levels of CD38 in mice, while curiously ex- rapidly relocate to the periphery of the follicle119-121 and are retained
88
actly the opposite pattern is seen with human GC B cells. However, in part by further expression of EBI2.103,104 Activated CD4+ T cells
it is important to note that during the early phases of an immune also adjust their chemotactic propensities by an increase in their
HABERMAN et al. |
      13

expression of CXCR5 and EBI2.96,122 As a result, recently activated is true even under conditions in which activation and cell con-
+ + + +
CD4 T cells and B cells are similarly CXCR5 , CCR7 , and EBI2 and tacts are occurring with rapidity. The delay in the appearance
both migrate to and congregate at the T-­B border and interfollic- of GC B cells had been a long-­s tanding paradox. Multiple mod-
ular regions within LNs or the analogous bridging channels within els have proposed mechanisms that may influence the differen-
spleens. After engaging T cells that share their cognate specificity, tiation of activated B cells to either the GC or ASC lineages. In
activated B cells commit to one of several potential fates; they either vitro studies have suggested that B cell ­f ate decisions could be
re-­enter the follicle and commit to the formation of a new GC, or stochastic and B cell intrinsic.136-138 Alternatively, the initial BCR
they differentiate outside of GCs into low-­affinity short-­lived ASCs, signal strength139,140 or competition for antigen and its presen-
123-125
early memory B cells, or other effector B cell lineages. It is at tation to cognate Th cells141 may be deciding factors. However,
these locations that elevated Bcl6 protein levels are first apparent the delay in GC B cell development could also be influenced by
in both responding B and T cells.126,127 Once Bcl6-­mediated tran- other potential constraints. For example, Tfh cells require mul-
scriptional repression is fully enforced in nascent GC B cells and Tfh tiple days to complete their effector differentiation, but once
cells, they lose expression of chemokine receptors such as EBI2 and complete might be capable of instructing GC B cell differentia-
CCR7 that retained them at the follicle periphery, resulting in revised tion expeditiously. A teleologic explanation for the delay in GC B
chemotactic patterns that instead drive them to the follicle interior cell formation might instead relate to the need for multiple line-
shortly thereafter.108,127-130 age fates to be created from every antigen-­s pecific B cell after
Importantly, the requirements for commitment to the intrafollic- its initial activation. Perhaps multiple rounds of proliferation are
ular GC pathway differ for T and B cells. It is clear that T-­cell differ- key to ensuring that an investment in the GC response only oc-
entiation to the perifollicular Tfh state can be driven initially by their curs after the production of early memory and short-­t erm ASC B
interactions with DCs.131 Bcl6 expression by a subset of responding cells. Here, we review the results of recent studies that suggest
T cells is detectable very early, within 1-­2 days of antigen encoun- additional mechanisms and provide some insights and into these
126,132,133
ter. Interestingly, typical Tfh markers CXCR5 and PD-­1 are aspects of B cell differentiation.
quickly upregulated on many responding T cells, but after 3 days of
interactions with B cells at the follicle periphery are then only main-
4.2 | Impact of CD40 signaling
tained on a population that ultimately gains entry to the follicle and
continues to interact with B cells.126,132-135 In contrast, full activation B ­cell lineage commitment is governed by opposing transcriptional
of B cells to commit to the GC pathway depends on those B cells networks that can be mutually antagonistic. Bcl6 acts as a transcrip-
interacting with cognate T cells. tional repressor of the TFs IRF4 and BLIMP1, and whereas it is es-
Over the course of several days, cognate B and T cells form sta- sential for GC B c­ ell formation,93,127,142-144 when overexpressed it
ble immunological synapses, but repeatedly engage new partners can prevent ASC formation.145-147 Reciprocally, high levels of IRF4
during this protracted and formative time period, all the while re- or induction of BLIMP1 represses transcription of Bcl6 but facilitates
maining at the follicle periphery. Bcl6 expression by B cells is not ASC differentiation.148,149 To further confound our understand-
detectable until after the emergence of Bcl6+ Th cells. Thus, B cell ing of transcriptional regulation, a transient and low expression of
commitment to the GC pathway, re-­entry to the follicle, and the co- IRF4 promotes the transcription of Bcl6 and is required for proper
alescence to form a proliferative foci there requires several days of GC B cell progression,150 despite the fact that it directly binds to
ongoing serial interactions with perifollicular Tfh. Interestingly, Tfh and represses the Bcl6 promoter when at higher levels.92,151 Thus,
126
entry into the follicle proceeds that of GC B cells, suggesting that IRF4 is required for both ASC and GC development, although ASC-­
the availability of Tfh support within the follicle is a prerequisite for associated genes are promoted when IRF4 is at sustained or high
the survival of newly arriving GC B cells. Early, pre-­GC cognate in- levels of expression.79,150,152-155
teractions that occur at the follicle periphery are therefore critical to Tfh cells express the surface costimulatory molecule CD40L
establishing the GC and determining immune outcome. and supply the cytokines IL-­4 and IL-­21 to GC B cells, all of which
are critical to the formation of fully mature and productive GCs. 20-
26
The combined expression of IL-­4 and IL-­21 is unique to Tfh cells,
4 | PRO G R E S S I V E G C B C E LL
however, all Th-­cell populations defined to date are thought to ex-
D I FFE R E NTI ATI O N H A S D I S TI N C TLY
press CD40L, and very early in an immune response.156,157 Signaling
R EG U L ATE D S TAG E S
via CD40 ligation invokes the non-­canonical NF-­kB pathway and
uniquely the nuclear translocation of heterodimeric RelB/p52.158-160
4.1 | Germinal center B cells are developmentally
Although CD40 signaling is required for GC B cell development,1,161
delayed
their formation can also be antagonized by chronic CD40 signaling
Shortly after BCR signaling in vitro and in vivo, activated B cells that instead promotes ASC development.151,162-165 ASC formation
are well poised to engage Th cells and respond to CD40 liga- during chronic CD40 signaling is in part mediated by IRF4, a TF
tion.117 However, many cell divisions are required before Bcl6 whose nuclear translocation is promoted by CD40 signaling.151,166
26
protein expression becomes evident in responding B cells. This These seemingly conflicting roles of CD40 pose a conundrum: CD40
|
14       HABERMAN et al.

and CD40L are required for GC B cell formation, but continued It remains unclear whether the ineffective repression of Bcl6 target
CD40 signaling discourages it. genes in GC B cell precursors is the result of subthreshold levels of
Some insight into the paradoxical roles of CD40 was garnered by Bcl6, or alternatively the absence of other corepressors that would
our recent work that examined the temporal relationship between otherwise enable effective transcriptional repression.170,172,173
26
CD40 signaling and TF shifts during GC B cell differentiation. In Although CD40 signaling is required for this first step in pre-­GC B
this study, B cells were first observed to express intermediate levels cell development, CD40 signaling must cease at the immediately sub-
of Bcl6 within the interfollicular regions of LNs. Intriguingly, these B sequent stage to allow for that incomplete stage of differentiation to
cells also were found at their first emergence to co-­express RelB and further progress.26 The intermediate differentiative stage of pre-­GC
IRF4 at intermediate levels, factors known to be mutually antagonis- B cells is characterized by mutually antagonistic TFs (IRF4int Bcl6int)
92,151
tic to Bcl6. Consistent with the presence of nuclear RelB and and is followed shortly thereafter by the appearance of mature GC
IRF4, CD40 signaling was required for this very first step in Bcl6 pro- B cells that are Bcl6hi with low levels of IRF4 and RelB and a more
tein production. At this early stage, Bcl6int GC precursors retained classical phenotype (CD38lo PNAhi).26 Interestingly, the shift from the
some plasticity—the infusion of either anti-­CD40 or additional an- immature pre-­GC B ­cell state to the more mature Bcl6hi state involves
tigen in vivo at this incomplete stage of development could still di- both transcriptional and post-­transcriptional regulation,26 the latter
26
vert them toward Bcl6lo IRF4hi ASCs. However, it is important to a form of regulation that can be mediated by micro-­RNAs.162,174,175
note that Bcl6lo IRF4hi ASC can be seen prior to the detection of Importantly, disrupting T-­B interactions or inhibiting CD40 signaling
Bcl6int pre-­GC B cells, 26 an observation that is consistent with the in vivo during this early transitional stage encourages GC B c­ ell mat-
co-­existence of an alternative pathway for extrafollicular ASC for- uration. However, when CD40 agonism is prolonged or additional
mation that is CD40 dependent but does not involve Bcl6. soluble antigen is introduced, GC precursors are instead expanded
Although CD40 signaling may establish a transcriptional land- and shunted away from the intrafollicular GC pathway.26,165 Based on
scape that is initially compatible with intermediate expression levels these results, a model of initial GC B cell differentiation has been pro-
of Bcl6, the presence of RelB and the antagonistic TF it promotes, posed to be a multistaged process completed by a transient diminution
IRF4, is incompatible with a complete repression of Bcl6 target genes in the receipt of T-­cell help within its immediate microenvironment
within pre-­GC B cells. 26 Consistent with this, pre-­GC B cells display (Figure 2). Here, it is envisioned that GC B cell differentiation is a two-­
a partial GC B cell phenotype (CD38int, PNAint). 26 Bcl6 is a member stage process involving first a T-­cell contact and CD40-­dependent
of the BTB-­POZ zinc finger family of TFs that forms a homodimer phase that generates RelB+ IRF4+ Bcl6int GC precursors and second,
167,168
capable of binding the corepressors SMRT, NCOR, and BCOR. a maturation phase immediately following that leads to the Bcl6hi in-
Bcl6 controls GC B cell differentiation by regulating cell cycle genes, trafollicular state that is prevented by further CD40 signaling. Thus,
repressing terminal differentiation factors, and suppressing some the impact of CD40 signaling may be stage specific in that it promotes
91,169
signaling pathways, including some aspects of BCR signaling. GC B cell precursor formation, but diverts precursor progression once
Repressed target genes in dark zone GC B cells in mice include cd38, formed. It is intriguing that the shifting transcriptional profiles of GC
irf4, and prdm1 (encoding BLIMP1).92,143,144 Notably, the extent of re- B cell precursors are staged over the course of multiple cell divisions.
pression of potential Bcl6 target genes is heavily influenced by core- More research is required to understand how the sequential introduc-
91,147,170,171
pressors that are themselves independently regulated. tion and subsequent removal of TFs is advantageous to the initial for-
Therefore, Bcl6 expression per se does not ensure target repression. mation of GC B cells.

Short-lived ASC
signalin D40
C
g
+

Further
IRF4 +
RelB+

IRF4 + GC lineage
nuclear RelB+
N?

Bcl6 + Bcl6 ++

CD40 signaling

IL-21
CD40 refractory
phase F I G U R E   2   Temporal and context-­
dependent shifts in transcription
IRF4int RelB+ factor composition during GC B cell
FDC Centrocyte Centroblast Plasma cell
Bcl6int PreGC B cell development
HABERMAN et al. |
      15

It remains unclear whether a transient abstinence from CD40L


4.3 | Role of IL-­21 and IL-­4 in GC B ­cell
stimulation plays a similar role in established GCs. The transition
maturation and intrafollicular development
between the classical centrocyte phenotype to that of an idealized
centroblast may involve progressive changes as well. LZ B cells experi- Similar to CD40L, the Tfh-­derived cytokines IL-­4 and IL-­21 play dis-
encing CD40L stimulation during engagement with Tfh cells gain cMyc tinct roles at discreet phases of GC B cell development. Although
expression, which subsequently promotes expression of AP4, a TF that many Tfh cells found within mature GCs can produce both IL-­4 and
is maintained within B cells by IL-­21 signaling.78 Subsequent loss of AP4 IL-­21, the cytokine expression profiles of Tfh cells can shift during
requires further cell division(s) with the DZ, a process that is needed to their development and can differ at distinct locations within lymphoid
eventually exit cell cycle.78 The potential impact of additional CD40 tissue.21-23,181,182 Earlier in adaptive immune responses, perifollicu-
signaling by DZ-­resident B cells, and whether it could occur in the ab- lar Tfh predominantly expresses IL-­21 and less IL-­4,23,182 while the
sence of IL-­21, remains unclear. As there are far fewer Tfh cells within reverse is true within the distal LZ of fully mature GCs where Tfh
the DZ than the contiguous LZ, this compartment offers a relative cells are more likely to produce IL-­4 than IL-­21.23 CXCR5+ iNK T cells
dearth of T-­cell–derived CD40L. The sheltered spaces within the DZ positioned at follicular boundaries are also capable of providing IL-­21
compartment appear to provide other advantages as well to newly and/or IL-­4 and promoting the GC lineage development of B cells that
transitioning centroblasts to GC dark zones. A study by Bannard et al32 are presenting glycolipid antigens, but the resulting GCs are short-­
indicates that GC B cells unable to escape the Tfh-­rich environment of lived and unproductive without the added participation of intrafol-
the LZ have a competitive disadvantage over time, suggesting that in licular Tfh cells responding to protein antigens.183-186 Importantly, the
the long run the cyclic movement between zones ultimately empowers delivery of IL-­4 and/or IL-­21 by iNK T cells is insufficient for the for-
a selective advantage in a way that is not yet understood. Although mation of sustainable GCs when CD40 signaling is absent.185 Other
some analogies can be made between the initial development of GC innate cell types including eosinophils and basophils are also known
B cells at the follicle periphery and the cyclic reprogramming of mature to produce IL-­4 and can function as antigen-­presenting cells to Th2
GCs, there are two major differences—(a) newly arriving Bcl6hi centro- cells,187-190 but they have not been observed within intrafollicular
cytes that are seeking antigen and Tfh help are already experiencing GCs188 and it remains unclear whether such innate cell types have
some level of transcriptional repression by Bcl6, and (b) a reservoir of the ability to provide IL-­4 in a directed fashion discreetly to antigen-­
antigen is accessible within the FDC network. More research is needed specific B cells.
to untangle this complex series of events. Both IL-­4 and IL-­21 are critical to GC B cell formation. Although
How might pre-­G C B cells transiently avoid CD40 signaling B cells that are unable to signal via IL-­21R or IL-­4Rs can still develop
and resolve their mixed state of mutually antagonistic TFs? One GCs in vivo, the stunted GCs that do form are significantly reduced in
means by which RelB+ pre-­G C B cells could lose that status is by size.182,191-194 In vitro, both IL-­21 and IL-­4 can enhance the transcrip-
an asymmetric cell division (ACD) that results in one daughter tion and/or translation of Bcl6 in B cells.195,196 Both cytokines are also
receiving less RelB. Consistent with this, we have observed an known to promote ASC formation.22,197-201 In the case of IL-­21, an in-
additional cell division of pre-­G C B cells correlates with reduced ability to signal via its receptor has a direct effect on B cells, resulting
RelB levels and a rise in Bcl6. 26 In this scenario, pre-­G C B cells in lower Bcl6 levels among the more limited number of GC B cells that
could undergo an ACD in which Bcl6 is unequally partitioned to form.182,191,192
one of the emerging daughters and RelB to the other, resulting in IL-­4R and IL-­21R complexes activate distinct signaling pathways.
two distinct cell fates. ACDs involving an unequal distribution of Like other cytokine receptors employing the common gamma chain,
Bcl6 have been reported among GC B cells, although its role in GC both IL-­21R and IL-­4R signal via the Janus kinase molecules JAK1 and
maintenance has been questioned. 176,177
Alternatively, engage- JAK3.202-205 However, these cytokine receptors activate different
ment with perifollicular cognate T cells could be attenuated by a signal transducer and activator of transcription (STAT) molecules.
subthreshold level of antigen presentation. Multiple cell divisions Whereas IL-­21 predominantly activates STAT3 and STAT1, and to a
could lead to a reduced capacity to present antigen, particularly lesser extent STAT5,206-209, IL-­4, and IL-­13 uniquely and requisitely
under conditions when antigen is limiting within that local micro- activate STAT6210,211.
environment. This would be consistent with previous reports of The results of a recent study we performed suggest that B cell
progressively shorter T-­B interaction lengths with non-­infectious signaling by these cytokines may influence GC B cells at different
immunogens. 59,126,127 In further support of this, conditions that stages.182 These experiments employed the cell transfer of antigen-­
instead create persistent pathogen-­d erived antigen promote ex- specific B cells that were either deficient in IL-­21R, STAT6, or both in
trafollicular ASCs and impair GC responses.178 Moreover, antigen order to assess the impact of IL-­21 and IL-­4/IL-­13 to B cells intrinsi-
that has been internalized after BCR capping has been reported cally. We found that the initial transition from the perifollicular pre-­GC
to be asymmetrically distributed between daughters after B cell B cell state to the intrafollicular stage is reduced in IL-­21R−/− B cells,
division, resulting in two daughters who differ in their ability to but unaffected in STAT6−/− B cells, suggesting that IL-­21 has a greater
179,180
engage cognate T cells. However, once GC B cells have mi- influence on differentiation at the time of pre-­GC B cell re-­entry to the
grated into the follicle interior, they again have access to a rich follicle interior.182 Importantly, in the absence of both IL-­21 and IL-­4
source of antigen within the FDC network. signaling pathways, Bcl6+ B cells can still form, however, they fail to
|
16       HABERMAN et al.

establish intrafollicular GCs.182 These findings support the idea that the response of IL-­4-­deficient mice to helminth infection and ob-
GC B cell development undergoes a step-­wise progression in which served a preponderance of DZ phenotype GC B cells that could be
the initial steps toward this lineage can occur in the absence of IL-­4 corrected by the introduction of IL-­4-­sufficient cells in BM chime-
and IL-­21, but is dependent upon CD40L. The Bcl6-­expressing B cells ras.194 Together, these intriguing studies raise more questions than
that form without IL-­21R or STAT6 signaling can persist transiently in they answer. Can these signals be received independently during se-
vivo, but their expansion is limited, indicating that GC B cell develop- quential cell contacts with Tfh cells that differ in their cytokine pro-
ment can be arrested at this stage. This could functionally provide a file? Are DZ centroblasts engaging Th cells within that compartment?
key checkpoint in this lineage pathway that would allow for the ini- Although more work is needed to address these and other questions,
tial creation of GC precursors, without committing to their furthered it is clear that the programming of GC B cells capable of responding
proliferation. In this way, a diverse repertoire could be potentiated to both cytokines is different from that obtained with one but not
without a concomitant clonal expansion of antigen-­reactive B cells the other.
for which there are not yet cognate Tfh cells capable of sustaining a
mature GC.
The results of this study further suggest that proper GC form 5 | E S TA B LI S H M E NT O F
and function within follicles is highly reliant on both cytokines.182 I NTR A FO LLI CU L A R G C S A N D TH E
IL-­21 and IL-­4 play non-­redundant roles in the subsequent matura- CO E VO LU TI O N O F FO LLI C U L A R S TRO M A L
tion and self-­renewal of GC B cells within follicles. Smaller numbers C E LL N I C H E S
of intrafollicular GC B cells form in the absence of either of these
signaling pathways, but with aberrant phenotypic features.182 When The cellular composition of tissue compartments effectively creates
GCs mature in the absence of either IL-­21 or STAT6 signaling, the microclimates that can be quite different. The cell types residing in
efficiency in the transition between the centroblast and centrocyte each compartment, together with the cytokines and chemokines
182,197 −/−
states is compromised and self-­renewal is reduced. IL-­21R they secrete, likely shape the nature of lymphocyte activation, pro-
B cells can form CXCR4hi centroblasts but they overexpress some liferation, and differentiation.10,110 Differences in local accessory
features that are more typical of centrocytes such as CD40 and cells ensure that B cell experiences within the molecular milieu of
CD86.182 Moreover, IL-­
21R−/− B cells are predominantly located one compartment will be different from those experienced in an-
within the FDC network, further suggesting that IL-­21 signaling is other compartment. For example, the composition of the stromal cell
required to properly instruct the centocyte to centroblast transition. population, their activation status, and any molecular products could
By contrast, IL-­4-­dependent signaling appears to have a greater con- potentially create a microenvironment that fosters modes of B cell
tribution to the proper formation of the centrocyte profile. Although growth or effector lineages that are distinct from those occurring
−/−
STAT6 GC B cells have a broadly normal spatial distribution within elsewhere. Although stromal communities may delineate the bounda-
their smaller GCs, albeit with a smaller percentage of centrocytes, ries of neighborhoods defined by characteristic chemokine compo-
the CXCR4lo centrocytes that form aberrantly underexpress CD86. sitions, they are themselves capable of change under inflammatory
Together, these results suggest that newly generated centroblasts conditions.212 Here, we review the progression of events that occur
formed without prior IL-­4 exposure might be compromised in their within the follicle as Tfh and GC B cells arrive, stromal cell popula-
ability to recapitulate the complete centrocyte program. This is con- tions evolve, and the unique compartments of the mature GC are
sistent with a study by Turqueti-­Neves and coworkers that examined established.

F I G U R E   3   Stages of GC maturation and associated tissue niches. (A) Schematic representation of the stages of germinal center (GC)
maturation. (B-­E) Immunofluorescence staining of popliteal lymph node (B) and spleen (C-­E) from C57BL/6 mice that received an adoptive
transfer of OVA-­specific T cells and antigen-­specific B cells enriched by immunomagnetic purification (StemCell Technologies). T cells
specific for OVA were obtained from spleens of OT-­II mice while either B1-­8+ Jκ−/− or MD4 mice served as donors for anti-­NP-­or anti-­HEL-­
specific B cells, respectively. Recipients were immunized after transfer with 50 μg of NP-­OVA/CFA in the footpad (lymph node, panels
B), NP-­OVA/alum injected i.p. (spleen, panels C, E), or HEL-­OVA/alum (spleen, panel D). (B) Popliteal lymph node histology time-­course
demonstrating GC maturation after immunization. Stages of maturation represented include the unimmunized follicle (d0/naïve), maturation
of the FDC network and light zone expansion (day 4), centroblast increase, zonal segregation and follicle swelling (day 5), and a mature GC
(day 10). Sections were stained to highlight CD4+ T cells (blue), IgD+ non-­responding B cells within the follicular mantel (green), and CD35+
FDC network (red). (C) Histology showing a maturing GC (day 6) stained to highlight IgD+ naïve B cells (grayscale), PNA+ GC cells (green),
and the distribution of sonic hedgehog (SHH, red) in relation to the FDC network stained with CD35 (blue). (D) Representative histology of
splenic GCs demonstrating initial differentiative events shortly after the arrival of Ag-­specific B cells to the follicle interior (day 3). Sections
were stained to highlight GFP+ Ag-­specific B cells (green), CD35 FDCs (blue), and Bcl6 (red) to identify GC committed B cells, and IgMa to
assess the level of cytosolic immunoglobulin. Arrows point to B cells within the FDC network that are cIg hi (consistent with ASC effector
lineage), Bcl6hi (GC lineage), and Bcl6neg cIg lo (of an undefined alternative fate). (E) Histology showing GFP+ Ag-­specific B cell interaction
with dendritic cells (DC) at the T-­B border (day 6). Section was stained for CD4+ T cells (blue), B220+ B cells (red), GFP+ Ag-­specific B cells
(green), and CD11c+ DC cells (grayscale). Higher-­magnification images of the T-­B border demonstrate the ability of GC B cells, DCs, and T
cells at the T-­zone border to directly engage each other
HABERMAN et al. |
      17

(A) Stages of GC maturation

(B) Intrafollicular GC expansion


CD4 IgD CD35

d0 d4 d5 d10
(C) FDC network differences (D) ASC within nascent GCs
IgD PNA CD35 SHH CD35
GFP
Ig
Bcl6 CD35 Ig Bcl6

d3.5 Ig
(E) DZ-T zone interface

d6 CD4 GFP IgD CD11c

dendritic than is typically observed within mature GCs (Figure 3A


5.1 | Early intrafollicular events and FDC maturation
and B). Immunization promotes the furthered development of FDCs
Follicular dendritic cells are derived from a stromal cell subset that from perivascular-­associated stromal cells, which then protrude long
uniquely differentiates within B cell follicles and are the primary dendritic extensions forming an extensive network of dendrites.9
97,213,214
source of CXCL13 in this zone of primary lymphoid tissues. Immunization can also elicit the formation of FDC-­like cells from the
Within lymphoid tissue of naïve mice, FDCs can be identified as MRCs that descend from the SCS of LNs or marginal sinus lining cells
uniquely expressing the long variant of the complement receptor of spleens, and hence contribute to the CD35+ CXCL13-­producing
14
CR1 (CD35) within follicles. They are centrally and sparsely dis- dendritic network closer to the outer follicle sinus.98,99 Both a nu-
tributed within primary B cell follicles and are substantially less merical increase and furthered differentiation occurs with these
|
18       HABERMAN et al.

MRC-­derived FDCs, known to uniquely produce RANK-­L within the vitro and reduces Fas-­induced apoptosis,224 providing further evidence
follicle.99 that within the GC LZ both FDC-­derived and Tfh-­derived factors can
After Tfh and pre-­GC B cells have further differentiated at the promote protection from the apoptosis-­prone state of GC B cells.215,229
interfollicular zone and T-­B border, they are observed to migrate
directly from that region toward the follicle interior, with the mi-
5.2 | B ­cell differentiation within the newly
gration of disengaged cognate T cells occurring independently and
formed LZ
preceding that of GC B cells.126 Responsiveness to CXCL13, down-
regulation of EBI2 and CCR7 all promote this initial intrafollicular Studies that have examined the formation of BM-­resident long-­
movement.10,103,104,129 Once within the follicle, Bcl6hi B cells are pri- lived plasma cells have observed that the plasma cells that per-
marily found in the outer follicle, the upper follicle more distal to the sist are predominantly generated after the peak of the GC
T-­cell zone, and appear there typically 3-­4 days postimmunization in response.125,230 However, there is evidence of B cell differentia-
126,127
non-­infectious experimental systems. tion toward this effector lineage within the FDC network, even
FDCs undergo a form of activation after immunization that is at this very early stage. In addition to Bcl6+ GC B cells, antigen-­
influenced by TLRs and NF-­kB signaling pathways, as well as im- specific B cells with large amounts of cytosolic immunoglobulin
mune complex binding. 215-218 This activation results in an increase (cIg) can be seen centrally placed in follicles and adjacent to FDCs
in the expression of numerous proteins that can play a role in GC (Figure 3D).126 It remains unclear whether the interactions with
B biology, including Fc receptor FcγRIIb, complement receptors accessory cells within the still evolving LZ microenvironment are
215,217-219
CD21/35, ICAM1, and VCAM1. FcγRIIb together with the sufficient to instruct the transcriptional program that is needed
complement receptors CD21/CD35 mediate the bulk of the antigen for longevity in plasma cells. 231
retention by the binding to proteins in immune complexes or tagged At this same place and time, within the emerging GC LZ, re-
with activated complement components. 220 After immunization, sponding B cells lacking both Bcl6 and cIg can also be observed,
FDCs also increase their production of Mfge8 that will promote the suggestive of an alternative lineage fate such as a GC-­d erived
phagocytosis of emerging apoptotic B cells by tingible body macro- memory B cell. Although the lineage fate of such Bcl6neg antigen-­
phages.7,221 FDC activation and dendrite extension is similar in tim- specific B cells remains unclear, and whether they have the ca-
ing to the arrival of Tfh and GC B cells to the follicle interior. Notably, pacity to persist long term remains unknown, these observations
Tfh cells when first arriving to the follicle interior within LNs can nevertheless suggest that not all GC B cell progeny are committed
first be found primarily in contact with the centrally located and un- to Bcl6 expression and self-­renewal in this arena even during this
derdeveloped CD35+ FDCs, but are also found in the follicular space stage of LZ expansion. Although Tfh cells are already in residence
above nearer the SCS (Figure 3B).126,127 Although coincident, there at this point in time, it is intriguing that the first events are not
is only in vitro evidence to suggest that Tfh cells can influence FDC entirely devoted to centroblast formation. Given that the produc-
activation222; there is no evidence to date in vivo. However, the TLR-­ tion of long-­lived memory B cells is the predominant effector cell
dependent activation of FDCs suggests that some aspects of their output of younger GCs that persists,125,232 it is tempting to spec-
furthered differentiation can take place after exposure to adjuvant ulate that the unique conditions of this stage may contribute to
216
alone. that lineage-­f ate preference. There are multiple differences within
As the GC matures, FDC network differences become more ap- newly formed GCs (days 3-­5 with most immunization regimes): (a)
parent. ICAM, FCγRIIb expression,223 and sonic hedgehog (SHH) ex- FDCs have more recently undergone activation, (b) antigen depos-
pression (Figure 3C)224 by FDCs are more evident within the GC LZ, its may be less likely initially to be in complexes with high-­affinity
and in particular at the LZ-­DZ interface, than is observed with the antibody, possibly enabling more processes reliant on activated
FDCs within the FM more proximal to the SCS. Given the positioning complement, 220,233 (c) newly arrived Tfh cells are more prone to
of MRC-­derived FDCs closer to the SCS of LNs, it may be that their produce IL-­21 than IL-­4 than at later stages in GC maturation, 23
99
contribution to the FDC network found within the FM is larger. The and (d) LZ-­resident B cells have just transitioned from the perifol-
functional differences between these FDC zones suggest that the na- licular pre-­G C program and could therefore theoretically not have
ture of the cellular interactions with lymphocytes within these distinct yet obtained the centrocyte program or higher affinities that are
domains could differ. For example, SHH found within GC LZs is known typical of a late time point. 55,234
to influence self-­renewal and cell-­fate determination in other tissue
compartments, including as hematopoietic niches, hair follicles, and
5.3 | B ­cell expansion and effector differentiation at
thymocyte selection.225-228 In support of the idea that SHH could in-
peripheral sinuses
fluence LZ centrocytes, it has been reported that a subset of GC B cells
express both of the required components of the Hedgehog (Hh) recep- When colonization of the FDC network by GC B cells has just begun,
224
tor, Ptc and Smo. Interestingly, expression of Smo can be increased B cell differentiation toward other non-­GC lineages is also apparent
with CD40 or BCR signaling,224 suggesting that successful engagement by histology at other peripheral arenas. Strikingly, at this very early
with stimulatory antigen and/or Tfh cells would empower hedgehog stage of the immune response, expanded populations of antigen-­
signaling. Hedgehog signaling in GC B cells promotes their viability in specific B cells form immediately under the SCS of LNs126,127 and
HABERMAN et al. |
      19

at splenic marginal sinuses. 235,236 This phenomena is short lived and CXCL12-­producing reticular cells (CRC) also have a reticular morphol-
quickly diminishes as the GC response progresses. During this early ogy, although the dendritic extensions lack the high levels of CD35 ex-
phase, responding B cells at the SCS were seen to exit follicles via the pression and complement binding that are distinctive for FDCs.31,32,242
subcapsular lumen of LNs during intravital imaging, but importantly, Interestingly, in some but not all GCs, there are DZ areas that are not
the reverse was not true—antigen-­specific B cells near the SCS were filled by a CRC network.242 The DZ stromal cells without CRC features
not observed to enter the follicle via that route, strongly suggest- are as yet uncharacterized, but are found closer to the DZ/LZ inter-
ing that proliferative B cells at the SCS do not contribute to the face and at this position could potentially shelter GC B cells from the
GC reaction.126 In this study, Bcl6 protein was not apparent within influences of both CRC and FDCs. In this regard, the distribution of
B cells at that location as assessed histologically. This is in contrast freshly minted centroblasts is intriguing. The emerging centroblasts
to the results of another histologic study that discerned a temporal typically form both a collection immediately under the FDC network
relationship of first a predominance of Ki67+ GL7+ B cells adjacent to as well as a loose column that stretches toward the T-­zone border
the marginal sinus, followed by an appearance of aggregated GL7+ (Figure 3B).141,182 This is consistent with chemotaxis toward CXCL12
B cells in follicle interiors. Here, GL7 expression was presumed to be as the primary drive for movement out of the FDC zone.10
a marker uniquely expressed by GC destined B cells. Although GL7 CRC can differ phenotypically and morphologically within the
expression appears restricted to B cells within GCs at mature time- DZ, perhaps relating to differences in either their derivation or mode
points,87 it is important to note that it is also known to be expressed of activation. Although fibroblastic reticular cells (FRCs) similar to
by LPS-­stimulated B cells in vitro, including by B cells forming ASC89 T-­zone FRC are only peripherally present at the edge of the B cell
+ 237 235
as well as by IgM early memory B cells. In the Coffey et al follicle in naïve LNs, after immunization the expanding GC swells
study, Bcl6 levels were assessed by qRT PCR of mRNA. However, the follicle, causing both IgD+ and GC B cells to shift past the large
Bcl6 can be transcribed with little translation to protein in some cell cortical lymphatic sinuses present at the naïve border and into the
types.162 Thus, the difference in these studies vis a vis GC B cell for- T-­zone FRC-­rich area.100,105,106 Results presented by Mionnet et al100
mation might therefore relate to the well-­known post-­transcriptional are consistent with the idea that at least some DZ stroma may de-
regulation of Bcl6. 238-240 rive from T-­zone FRC cells. In this way, new microenvironments are
There is some evidence of ASC development among activated created by evolving stromal cells thereby forming a novel venue for
B cells at LN SCS. Some SCS-­adjacent B cells have copious amounts events that might not have otherwise been possible, in this case an
126
of cIg light chain, consistent with commitment to a ASC lineage. avenue bringing DZ GC B cells in close contact with the T-­zone bor-
However, in another study, B cells adjacent to splenic marginal sinus der. Like other T-­zone FRC, the stromal network newly infiltrated by
lacked the CD138 expression that is characteristic of late-­
stage the expanding follicle also exhibits a collagen dense core typical of
plasma(blast) cell formation and had not undergone class switch re- FRC-­ensheathed lymph conduits, 243,244 a feature that is less apparent
combination to IgG. 235 Together the results of these studies remain among the more centrally positioned CRC within mature DZs.32,242
consistent with the idea that a subset of sinus lining B cells are in the Given that CRCs are also found within primary non-­reactive B cell
process of differentiating to IgM ASCs. In addition to the SCS, ASC follicles, 242 it seems possible that CXCL12-­expressing stroma within
cell formation can also be seen at the T-­B border, and among the re- DZ compartments can derive from more than one cell type.
126,241
maining cognate B and T cells within the interfollicular region. In addition to the blasting centroblasts, GC DZs also harbor fol-
Consistent with this, prdm1-­expressing ASC B cells have also been licular T cells as well as DCs (Figure 3B). Not all follicular T cells are
observed to move from deeper perifollicular locations toward med- located in the LZ. They can also be found within the center of DZs as
ullary sinuses.90 The lineage fates of the non-­A SC B cells at these well as at the outer edges of DZs adjacent to the FM, albeit in lower
alternative locations remain unclear, but given the absence of Bcl6 densities (Figure 3B). 21,245 Although little is known about how these
+
protein and prior reports of very early IgM memory B cells that are CD4+ T cells and DC subsets might differ from their counterparts
237
GC independent, it is likely that a subset is destined to become at the T-­B border and GC LZ, there is evidence to suggest that Tfh
early memory B cells or alternative B cell effectors.123,125 cells producing IFNγ or IL-­21 can localize to the DZ outer edge within
the follicle. 21,23 Follicular regulatory T cells (Tfr) have also been ob-
served within DZs. 245 Such DZ-­resident T cells are in a unique posi-
5.4 | Zonal segregation and DZ evolution
tion to influence centroblast behavior, either favorably as they might
Depending on the immunogen and mode of immunization, the emer- engage Tfh cells within the LZ, or in the case of DZ Tfr cells influenc-
gence of a discernable DZ may not occur for another day or two. This ing centroblast self-­renewal or differentiation to long-­lived memory
zonal segregation requires, by definition, the presence of GC B cells or plasma cells. 246-248
outside of the FDC network, but as described below there are sev-
eral other unique features of the DZ compartment that evolve dur-
5.5 | Follicular Mantle
ing the course of the response. One key element of the centroblast
program is the increased expression of CXCR4, which enables centro- As the GC expands, GC B cells form a large egg-­shaped aggregate that
blast migration to CXL12-­producing stroma initially found most proxi- displaces non-­responding B cells, causing them to form the follicu-
mal to the basement of the follicle, near the T-­B border.10,33,242 These lar mantle. Intravital imaging has demonstrated that non-­responding
|
20       HABERMAN et al.

B cells are not excluded by a physical barrier and can move freely GC B cells to interact with cell types that are not present in other GC
70,72
through GCs. Although early stages of GC development show compartments.
extensive admixing with IgD+ non-­cognate B cells (Figure 3B), this is This arena of mature GCs is poorly understood but clearly has
uncommon within mature GCs. The follicular mantle harbors more the potential to be an influential venue for self-­renewing cen-
than just non-­responding B cells. In addition to the sinus-­proximal troblasts as well as GC-­d erived long-­term plasma cells. Maturing
FDCs described above, a distinct subset of Tfh cells can also be found plasmablasts can be found at this location, 82,251,262 and in a re-
within FMs. The follicular mantle surrounding mature GCs harbors Tfh cent study found to be in association with CD157hi podoplanin+
cells that are phenotypically and functionally distinct, with a lower FRCs as early as 6 days postimmunization. 241 Similar to the CRC
22,122
expression of IL-­4. After the resolution of a primary response, cells within the DZ, the reported CD157hi FRC at the DZ base of
both memory B cells and memory Tfh cells can be found within FMs, a young GCs also express CXCL12, but appear to lack dendritic ex-
positioning that is influenced by their EBI2 expression.122,249,250 tensions into the DZ. 241,242 An intriguing analysis of LN stromal
Theoretically, this compartment could influence the differen- cells by single-­cell RNAseq further suggests that FRC at the T-­zone
tiative state of antigen-­specific B cells that are leaving the GC via interface with B cell follicles is heterogenous, and may include
this route. As cells within this arena are less likely to be in active both CCL19lo and CCL19hi subsets, as well as activated subsets
cell cycle, this environment may be more conducive to gaining a responding to inflammation.109
quiescent state, sheltered from the cellular interactions that pro- There are several features to this unique niche that have the po-
mote a centroblast program and their self-­renewal. It is import- tential to influence centroblasts and other DZ-­resident cell types at
ant to note that this is not the predominant route of exit for the this DZ-­T-­zone border. Here, there are opportunities for direct con-
prdm1-­expresing ASCs leaving GCs, which largely leave via the tact with perifollicular Th cell and DC subsets (Figure 3E), as well as
GC outer zone through the DZ base due to their downregulation Tregs.245 There is also the potential for fresh antigen delivery via in-
82,90,251-253
of CXCR5 and increased responsiveness to CXCL12. coming local lymph within adjacent sinuses.106,243,263 Scavenging APCs
However, the FM could be the preferred exit route of GC-­d erived could theoretically participate in antigen acquisition from local sinuses,
memory B cells, as has been shown for Tfh memory cells during similar to DCs probing lymphatic sinuses or FRC junctures,244,264 and
122
secondary responses. CCR6 expression has been identified as provide antigen processing and presentation to perifollicular Tfh cell
a distinctive feature of memory B cells, 254,255 including those ob- present at the DZ-­T border. In this regard it is interesting to note that
256
served to be completing their differentiation within GC LZs. A some DCs have a specialized capacity to retain and present antigens in
ligand for CCR6, CCL20, is known to be produced by some ep- native form without degradation, theoretically permitting BCR cross-
ithelial cell types and importantly by lymphatic endothelial cells linking of centroblasts in direct contact with DCs presenting incom-
lining the subcapsular sinus of LNs near B cell follicles, the latter ing lymph-­borne antigen.115,265 Thus, at this niche, DZ GC B cells may
shown to be key to the localization of IL7Ra+ CCR6hi innate-­like have access to antigen to process and present, as well as APCs and
lymphocytes. 257,258 FM non-­responding B cells are also CCR6hi, in T-­cell help, which in toto could be sufficient for any of the proposed
sharp contrast to CCR6lo GC B cells. However, naive B cells lack mechanisms for GC B cell selection.33,43,45,59 Consistent with this
responsiveness to CCL20 despite expressing the receptor for this niche playing a role in DZ B cell dynamics, movement to and from the
chemokine, unlike CCR6hi memory B cells which chemotax well DZ-­T-­zone border has been observed by intravital imaging.70 Future
255
to CCL20. Although memory B cells have been reported to be work in this arena is very much needed to gain insight into this poorly
positioned within lymphoid tissues at marginal zones, perifollicular understood niche.
zones, and some follicles after the resolution of response, 255,259,260
two studies have reported a predominant FM location for Ki67-­
memory B cells in spleens and LNs. 249,250 Although the above 6 | CO N C LU S I O N
studies support the idea that memory B cells primarily exit via fol-
licle adjacent sinuses, it is worth noting that CCR6hi memory B GC development is a complex series of events that requires that
255
cells are also responsive to CXCL12. cognate B and T cells periodically engage during discreet stages in
their differentiation and at distinct locations within tissue. While GC
B- cell development begins at the periphery of B cell follicles, this
5.6 | The unique niche of the DZ-­T zone border in
line of differentiation is progressive and is characterized by step-­
mature GCs
wise changes in transcription and phenotype over the course of
The follicular mantle is thin to non-­existent at the true base of the DZ multiple cell divisions. GC B cell development can be paused at an
in mature GCs. In cross sections of mature GCs that are central and intermediate stage of differentiation when conditions are not quite
en face, IgD+ B cells can be infrequent or absent at the base of the right, permitting the refinement of the immune response when an-
DZ. This DZ-­T zone interface is commonly observed in splenic and LN tigen is in excess, or when the differentiation of regional activated
GCs when canvassed in their entirety via serial sections (Figure 3E, T cells has not sufficiently progressed. Under conditions when re-
Haberman lab unpublished data).32,82,242,261 The DZ-­T zone interface cently activated CD40L+ CD4+ T cells are present, but IL-­21 and IL-­4
can be quite extensive in larger GCs and could theoretically allow DZ are lacking, pre-­GC B cells will transiently persist at perifollicular
HABERMAN et al. |
      21

locations. This is in part regulated by a stage-­specific effect of CD40 formation and the development of B cell memory. J Exp Med.
signaling; while an early phase in GC B cell development is depend- 1994;180:157‐163.
2. Linton PJ, Lo D, Lai L, Thorbecke GJ, Klinman NR. Among naive
ent on CD40 signaling, the subsequent phase is refractory to CD40
precursor cell subpopulations only progenitors of memory B cells
agonism and likely involves a transient abstention from T-­cell help originate germinal centers. Eur J Immunol. 1992;22:1293‐1297.
at the periphery of B cell follicles. When IL-­21 or IL-­4 signaling also 3. Linton PL, Decker DJ, Klinman NR. Primary antibody-­forming cells
occurs, then GC B cell maturation is allowed to progress to the next and secondary B cells are generated from separate precursor cell
subpopulations. Cell. 1989;59:1049‐1059.
stage that empowers follicular entry, but only if CD40 agonism is
4. Tarlinton DM, Smith KG. Dissecting affinity maturation: a model
transiently avoided. The migration of Tfh cells into the follicle inte- explaining selection of antibody-­forming cells and memory B cells
rior before GC B cell arrival might therefore help at multiple stages in the germinal centre. Immunol Today. 2000;21:436‐441.
and locations by: (a) vacating the perifollicular sites and thereby re- 5. MacLennan IC. Germinal centers. Annu Rev Immunol.
1994;12:117‐139.
ducing the chance of CD40L stimulation, and (b) entering into the
6. Liu YJ, Johnson GD, Gordon J, MacLennan IC. Germinal cen-
follicle in advance, immediately ensuring that centrocyte and centro- tres in T-­ cell-­
dependent antibody responses. Immunol Today.
blast instruction occurs properly and sustainably, events that are re- 1992;13:17‐21.
liant on both IL-­21 and IL-­4. Thus, early B and T cognate interactions 7. Camacho SA, Kosco-Vilbois MH, Berek C. The dynamic structure
of the germinal center. Immunol Today. 1998;19:511‐514.
at the follicle periphery change in quality as they co-­evolve, critically
8. Liu YJ, Grouard G, de Bouteiller O, Banchereau J. Follicular den-
influencing when and how intrafollicular GCs are established.
dritic cells and germinal centers. Int Rev Cytol. 1996;166:139‐179.
All these early events are temporally and spatially choreographed 9. Krautler NJ, Kana V, Kranich J, et al. Follicular dendritic cells emerge
by the serial movements of cognate B and T cells to different and dis- from ubiquitous perivascular precursors. Cell. 2012;150:194‐206.
creet tissue locales. The stroma at these tissue destinations are them- 10. Allen CD, Ansel KM, Low C, et al. Germinal center dark and light
zone organization is mediated by CXCR4 and CXCR5. Nat Immunol.
selves influenced by the evolving immune response in their arena.
2004;5:943‐952.
Together these interactions help to ensure that activated B cells re- 11. Szakal AK, Kosco MH, Tew JG. Microanatomy of lymphoid tissue
ceive the correct signals for their stage of GC B cell differentiation during humoral immune responses: structure function relation-
and coordinate the timing and extent of that differentiation. Finally, ships. Annu Rev Immunol. 1989;7:91‐109.
12. Qin D, Wu J, Vora KA, et  al. Fc gamma receptor IIB on follicu-
we conclude that it is advantageous for GC B cell differentiation to
lar dendritic cells regulates the B cell recall response. J Immunol.
be staged and slowly progressive in that this creates checkpoints that 2000;164:6268‐6275.
allow for the initial creation of GC precursors, without committing to 13. Carroll MC. The role of complement and complement recep-
an energy-­intensive, and potentially misdirected and unproductive GC. tors in induction and regulation of immunity. Annu Rev Immunol.
1998;16:545‐568.
14. Liu YJ, Xu J, de Bouteiller O, et al. Follicular dendritic cells
AC K N OW L E D G E M E N T S specifically express the long CR2/CD21 isoform. J Exp Med.
1997;185:165‐170.
We thank Jaymin Patel for skillful technical assistance with IF his- 15. Heesters BA, Chatterjee P, Kim YA, et  al. Endocytosis and recy-
tology. This work was supported by National Institutes of Health cling of immune complexes by follicular dendritic cells enhances B
cell antigen binding and activation. Immunity. 2013;38:1164‐1175.
NIAID grants R01AI080850 and R21AI101704 and NIAMS
16. Mandel TE, Phipps RP, Abbot A, Tew JG. The follicular dendritic
Rheumatic Diseases Research Core Centers grant P30AR053495. cell: long term antigen retention during immunity. Immunol Rev.
T. Z. was supported by a fellowship by the Canadian Institutes of 1980;53:29‐59.
Health Research. S. M. K. is funded by an Operating Grant from the 17. Klaus GG, Humphrey JH, Kunkl A, Dongworth DW. The follicular
dendritic cell: its role in antigen presentation in the generation of
Canadian Institutes of Health Research and was the recipient of
immunological memory. Immunol Rev. 1980;53:3‐28.
the Garrett Herman endMS Research and Training Network Career 18. King C, Tangye SG, Mackay CR. T follicular helper (TFH) cells in
Development Award from the Multiple Sclerosis Society of Canada. normal and dysregulated immune responses. Annu Rev Immunol.
2008;26:741‐766.
19. Arnold CN, Campbell DJ, Lipp M, Butcher EC. The germinal center
C O N FL I C T O F I N T E R E S T response is impaired in the absence of T cell-­expressed CXCR5.
Eur J Immunol. 2007;37:100‐109.
The authors certify that there is no actual or potential conflict of 20. Foy TM, Aruffo A, Bajorath J, Buhlmann JE, Noelle RJ. Immune
interest in relation to this article. regulation by CD40 and its ligand GP39. Annu Rev Immunol.
1996;14:591‐617.
21. Reinhardt RL, Liang HE, Locksley RM. Cytokine-­ secreting fol-
ORCID licular T cells shape the antibody repertoire. Nat Immunol.
2009;10:385‐393.
Ann M. Haberman  http://orcid.org/0000-0001-5168-8229 22. Yusuf I, Kageyama R, Monticelli L, et  al. Germinal center T fol-
licular helper cell IL-­ 4 production is dependent on signaling
lymphocytic activation molecule receptor (CD150). J Immunol.
2010;185:190‐202.
REFERENCES
23. Weinstein JS, Herman EI, Lainez B, et al. TFH cells progres-
1. Foy TM, Laman JD, Ledbetter JA, Aruffo A, Claassen E, Noelle sively differentiate to regulate the germinal center response. Nat
RJ. gp39-­
CD40 interactions are essential for germinal center Immunol. 2016;17:1197‐1205.
|
22       HABERMAN et al.

24. Ramiscal RR, Vinuesa CG. T-­cell subsets in the germinal center. 44. Hauser AE, Kerfoot SM, Haberman AM. Cellular choreography
Immunol Rev. 2013;252:146‐155. in the germinal center: new visions from in vivo imaging. Semin
25. Crotty S. A brief history of T cell help to B cells. Nat Rev Immunol. Immunopathol. 2010;32:239‐255.
2015;15:185‐189. 45. Liu YJ, Joshua DE, Williams GT, Smith CA, Gordon J, MacLennan
26. Zhang TT, Gonzalez DG, Cote CM, et al. Germinal center B cell IC. Mechanism of antigen-­driven selection in germinal centres.
development has distinctly regulated stages completed by disen- Nature. 1989;342:929‐931.
gagement from T cell help. Elife. 2017;6:e19552. 46. Wu J, Qin D, Burton GF, Szakal AK, Tew JG. Follicular dendritic
27. Schaerli P, Willimann K, Lang AB, Lipp M, Loetscher P, Moser cell-­derived antigen and accessory activity in initiation of memory
B. CXC chemokine receptor 5 expression defines follicu- IgG responses in vitro. J Immunol. 1996;157:3404‐3411.
lar homing T cells with B cell helper function. J Exp Med. 47. Yoon SO, Zhang X, Berner P, Blom B, Choi YS. Notch ligands ex-
2000;192:1553‐1562. pressed by follicular dendritic cells protect germinal center B cells
28. Kim CH, Rott LS, Clark-Lewis I, Campbell DJ, Wu L, Butcher EC. from apoptosis. J Immunol. 2009;183:352‐358.
Subspecialization of CXCR5+ T cells: B helper activity is focused 48. Liu YJ, de Bouteiller O, Fugier-Vivier I. Mechanisms of selec-
in a germinal center-­localized subset of CXCR5+ T cells. J Exp Med. tion and differentiation in germinal centers. Curr Opin Immunol.
2001;193:1373‐1381. 1997;9:256‐262.
29. Breitfeld D, Ohl L, Kremmer E, et al. Follicular B helper T cells ex- 49. Kosco MH, Pflugfelder E, Gray D. Follicular dendritic cell-­
press CXC chemokine receptor 5, localize to B cell follicles, and sup- dependent adhesion and proliferation of B cells in vitro. J Immunol.
port immunoglobulin production. J Exp Med. 2000;192:1545‐1552. 1992;148:2331‐2339.
30. Yu D, Batten M, Mackay CR, King C. Lineage specification and 50. Qin D, Wu J, Carroll MC, Burton GF, Szakal AK, Tew JG. Evidence
heterogeneity of T follicular helper cells. Curr Opin Immunol. for an important interaction between a complement-­d erived
2009;21:619‐625. CD21 ligand on follicular dendritic cells and CD21 on B cells in
31. Hardie DL, Johnson GD, Khan M, MacLennan IC. Quantitative anal- the initiation of IgG responses. J Immunol. 1998;161:4549‐4554.
ysis of molecules which distinguish functional compartments 51. Khalil AM, Cambier JC, Shlomchik MJ. B cell receptor signal trans-
within germinal centers. Eur J Immunol. 1993;23:997‐1004. duction in the GC is short-­circuited by high phosphatase activity.
32. Bannard O, Horton RM, Allen CD, An J, Nagasawa T, Cyster JG. Science. 2012;336:1178‐1181.
Germinal center centroblasts transition to a centrocyte phenotype 52. Luo W, Weisel F, Shlomchik MJ. B cell receptor and CD40 signal-
according to a timed program and depend on the dark zone for ing are rewired for synergistic induction of the c-­Myc transcrip-
effective selection. Immunity. 2013;39:912‐924. tion factor in germinal center B cells. Immunity. 2018;48:313‐326
33. Victora GD, Schwickert TA, Fooksman DR, et al. Germinal center e5.
dynamics revealed by multiphoton microscopy with a photoacti- 53. Casamayor-Palleja M, Gulbranson-Judge A, MacLennan IC.
vatable fluorescent reporter. Cell. 2010;143:592‐605. T cells in the selection of germinal center B cells. Chem Immunol.
34. Calado DP, Sasaki Y, Godinho SA, et  al. The cell-­c ycle regulator 1997;67:27‐44.
c-­Myc is essential for the formation and maintenance of germinal 54. Casamayor-Palleja M, Khan M, MacLennan IC. A subset of CD4+
centers. Nat Immunol. 2012;13:1092‐1100. memory T cells contains preformed CD40 ligand that is rapidly but
35. Jacob J, Kassir R, Kelsoe G. In situ studies of the primary im- transiently expressed on their surface after activation through the
mune response to (4-­ hydroxy-­3-­
nitrophenyl)acetyl. I. The archi- T cell receptor complex. J Exp Med. 1995;181:1293‐1301.
tecture and dynamics of responding cell populations. J Exp Med. 55. Ise W, Fujii K, Shiroguchi K, et al. T follicular helper cell-­germinal
1991;173:1165‐1175. center B cell interaction strength regulates entry into plasma cell
36. Jacob J, Kelsoe G, Rajewsky K, Weiss U. Intraclonal gen- or recycling germinal center cell fate. Immunity. 2018;48:702‐715
eration of antibody mutants in germinal centres. Nature. e4.
1991;354:389‐392. 56. Mesin L, Ersching J, Victora GD. Germinal center B cell dynamics.
37. Dal Porto JM, Haberman AM, Kelsoe G, Shlomchik MJ. Very low Immunity. 2016;45:471‐482.
affinity B cells form germinal centers, become memory B cells, and 57. Papa I, Vinuesa CG. Synaptic interactions in germinal centers.
participate in secondary immune responses when higher affinity Front Immunol. 2018;9:1858.
competition is reduced. J Exp Med. 2002;195:1215‐1221. 58. Walker LS, Gulbranson-Judge A, Flynn S, Brocker T, Lane PJ. Co-­
38. Tarlinton D. Germinal centers: form and function. Curr Opin stimulation and selection for T-­cell help for germinal centres: the
Immunol. 1998;10:245‐251. role of CD28 and OX40. Immunol Today. 2000;21:333‐337.
39. Muramatsu M, Kinoshita K, Fagarasan S, Yamada S, Shinkai Y, 59. Allen CD, Okada T, Tang HL, Cyster JG. Imaging of germinal
Honjo T. Class switch recombination and hypermutation require center selection events during affinity maturation. Science.
activation-­induced cytidine deaminase (AID), a potential RNA ed- 2007;315:528‐531.
iting enzyme. Cell. 2000;102:553‐563. 60. Meyer-Hermann ME, Maini PK, Iber D. An analysis of B cell
40. Cattoretti G, Buttner M, Shaknovich R, Kremmer E, Alobeid B, selection mechanisms in germinal centers. Math Med Biol.
Niedobitek G. Nuclear and cytoplasmic AID in extrafollicular and 2006;23:255‐277.
germinal center B cells. Blood. 2006;107:3967‐3975. 61. Liu D, Xu H, Shih C, et al. T-­B-­cell entanglement and ICOSL-­driven
41. Rogerson B, Hackett J Jr, Peters A, Haasch D, Storb U. Mutation feed-­ forward regulation of germinal centre reaction. Nature.
pattern of immunoglobulin transgenes is compatible with a 2014;517:214‐218.
model of somatic hypermutation in which targeting of the 62. Gitlin AD, Shulman Z, Nussenzweig MC. Clonal selection in the
mutator is linked to the direction of DNA replication. EMBO J. germinal centre by regulated proliferation and hypermutation.
1991;10:4331‐4341. Nature. 2014;509:637‐640.
42. Neuberger MS, Di Noia JM, Beale RC, Williams GT, Yang Z, Rada 63. Hennino A, Berard M, Krammer PH, Defrance T. FLICE-­inhibitory
C. Somatic hypermutation at A.T pairs: polymerase error versus protein is a key regulator of germinal center B cell apoptosis. J Exp
dUTP incorporation. Nat Rev Immunol. 2005;5:171‐178. Med. 2001;193:447‐458.
43. Haberman AM, Shlomchik MJ. Reassessing the function of immune-­ 64. Takahashi Y, Ohta H, Takemori T. Fas is required for clonal selec-
complex retention by follicular dendritic cells. Nat Rev Immunol. tion in germinal centers and the subsequent establishment of the
2003;3:757‐764. memory B cell repertoire. Immunity. 2001;14:181‐192.
HABERMAN et al. |
      23

65. Han S, Zheng B, Dal Porto J, Kelsoe G. In situ studies of the pri- 86. Smith KG, Nossal GJ, Tarlinton DM. FAS is highly expressed in the
mary immune response to (4-­ hydroxy-­ 3-­
nitrophenyl)acetyl. IV. germinal center but is not required for regulation of the B-­cell re-
Affinity-­dependent, antigen-­driven B cell apoptosis in germinal sponse to antigen. Proc Natl Acad Sci USA. 1995;92:11628‐11632.
centers as a mechanism for maintaining self-­tolerance. J Exp Med. 87. Cervenak L, Magyar A, Boja R, Laszlo G. Differential expression of
1995;182:1635‐1644. GL7 activation antigen on bone marrow B cell subpopulations and
66. Hao Z, Duncan GS, Seagal J, et al. Fas receptor expression in peripheral B cells. Immunol Lett. 2001;78:89‐96.
germinal-­center B cells is essential for T and B lymphocyte homeo- 88. Oliver AM, Martin F, Kearney JF. Mouse CD38 is down-­regulated
stasis. Immunity. 2008;29:615‐627. on germinal center B cells and mature plasma cells. J Immunol.
67. Kepler TB, Perelson AS. Cyclic re-­entry of germinal center 1997;158:1108‐1115.
B cells and the efficiency of affinity maturation. Immunol Today. 89. Laszlo G, Hathcock KS, Dickler HB, Hodes RJ. Characterization of
1993;14:412‐415. a novel cell-­surface molecule expressed on subpopulations of acti-
68. Oprea M, Perelson AS. Somatic mutation leads to efficient affin- vated T and B cells. J Immunol. 1993;150:5252‐5262.
ity maturation when centrocytes recycle back to centroblasts. J 90. Fooksman DR, Schwickert TA, Victora GD, Dustin ML, Nussenzweig
Immunol. 1997;158:5155‐5162. MC, Skokos D. Development and migration of plasma cells in the
69. Okada T, Cyster JG. B cell migration and interactions in the early mouse lymph node. Immunity. 2010;33:118‐127.
phase of antibody responses. Curr Opin Immunol. 2006;18:278‐285. 91. Bunting KL, Melnick AM. New effector functions and regulatory
70. Hauser AE, Junt T, Mempel TR, et al. Definition of germinal-­center mechanisms of BCL6 in normal and malignant lymphocytes. Curr
B cell migration in vivo reveals predominant intrazonal circulation Opin Immunol. 2013;25:339‐346.
patterns. Immunity. 2007;26:655‐667. 92. Basso K, Saito M, Sumazin P, et  al. Integrated biochemical and
71. Hauser AE, Shlomchik MJ, Haberman AM. In vivo imaging stud- computational approach identifies BCL6 direct target genes con-
ies shed light on germinal-­centre development. Nat Rev Immunol. trolling multiple pathways in normal germinal center B cells. Blood.
2007;7:499‐504. 2010;115:975‐984.
72. Schwickert TA, Lindquist RL, Shakhar G, et al. In vivo imaging 93. Dent AL, Shaffer AL, Yu X, Allman D, Staudt LM. Control of in-
of germinal centres reveals a dynamic open structure. Nature. flammation, cytokine expression, and germinal center formation
2007;446:83‐87. by BCL-­6. Science. 1997;276:589‐592.
73. Goodnow CC, Vinuesa CG, Randall KL, Mackay F, Brink R. Control 94. Ye BH, Cattoretti G, Shen Q, et al. The BCL-­6 proto-­oncogene con-
systems and decision making for antibody production. Nat trols germinal-­centre formation and Th2-­type inflammation. Nat
Immunol. 2010;11:681‐688. Genet. 1997;16:161‐170.
74. Bannard O, Cyster JG. Germinal centers: programmed for affin- 95. Choi YS, Yang JA, Crotty S. Dynamic regulation of BcI6 in follicular
ity maturation and antibody diversification. Curr Opin Immunol. helper CD4 T (Tfh) cells. Curr Opin Immunol. 2013;25:366‐372.
2017;45:21‐30. 96. Ansel KM, McHeyzer-Williams LJ, Ngo VN, McHeyzer-Williams
75. Dominguez-Sola D, Victora GD, Ying CY, et al. The proto-­oncogene MG, Cyster JG. In vivo-­ activated CD4 T cells upregulate CXC
MYC is required for selection in the germinal center and cyclic re- chemokine receptor 5 and reprogram their response to lymphoid
entry. Nat Immunol. 2012;13:1083‐1091. chemokines. J Exp Med. 1999;190:1123‐1134.
76. Sander S, Chu VT, Yasuda T, et al. PI3 kinase and FOXO1 transcrip- 97. Cyster JG, Ansel KM, Reif K, et al. Follicular stromal cells and lym-
tion factor activity differentially control B cells in the germinal phocyte homing to follicles. Immunol Rev. 2000;176:181‐193.
center light and dark zones. Immunity. 2015;43:1075‐1086. 98. Katakai T, Suto H, Sugai M, et al. Organizer-­like reticular stromal
77. Dominguez-Sola D, Kung J, Holmes AB, et  al. The FOXO1 tran- cell layer common to adult secondary lymphoid organs. J Immunol.
scription factor instructs the Germinal Center Dark Zone Program. 2008;181:6189‐6200.
Immunity. 2015;43:1064‐1074. 99. Jarjour M, Jorquera A, Mondor I, et al. Fate mapping reveals origin
78. Chou C, Verbaro DJ, Tonc E, et al. The transcription factor AP4 me- and dynamics of lymph node follicular dendritic cells. J Exp Med.
diates resolution of chronic viral infection through amplification of 2014;211:1109‐1122.
germinal center B cell responses. Immunity. 2016;45:570‐582. 100. Mionnet C, Mondor I, Jorquera A, et al. Identification of a new
79. Cattoretti G, Shaknovich R, Smith PM, Jack HM, Murty VV, stromal cell type involved in the regulation of inflamed B cell folli-
Alobeid B. Stages of germinal center transit are defined by cles. PLoS Biol. 2013;11:e1001672.
B cell transcription factor coexpression and relative abun- 101. Forster R, Schubel A, Breitfeld D, et al. CCR7 coordinates the pri-
dance. J Immunol. 2006;177:6930‐6939. mary immune response by establishing functional microenviron-
80. Cattoretti G, Angelin-Duclos C, Shaknovich R, Zhou H, Wang D, ments in secondary lymphoid organs. Cell. 1999;99:23‐33.
Alobeid B. PRDM1/Blimp-­1 is expressed in human B-­lymphocytes 102. Luther SA, Tang HL, Hyman PL, Farr AG, Cyster JG. Coexpression
committed to the plasma cell lineage. J Pathol. 2005;206:76‐86. of the chemokines ELC and SLC by T zone stromal cells and dele-
81. Liu YJ, Banchereau J. Regulation of B-­cell commitment to plasma tion of the ELC gene in the plt/plt mouse. Proc Natl Acad Sci USA.
cells or to memory B cells. Semin Immunol. 1997;9:235‐240. 2000;97:12694‐12699.
82. Krautler NJ, Suan D, Butt D, et al. Differentiation of germinal cen- 103. Gatto D, Paus D, Basten A, Mackay CR, Brink R. Guidance of B cells
ter B cells into plasma cells is initiated by high-­affinity antigen and by the orphan G protein-­coupled receptor EBI2 shapes humoral
completed by Tfh cells. J Exp Med. 2017;214:1259‐1267. immune responses. Immunity. 2009;31:259‐269.
83. MacLennan IC, Liu YJ, Johnson GD. Maturation and dispersal 104. Pereira JP, Kelly LM, Xu Y, Cyster JG. EBI2 mediates B cell
of B-­ cell clones during T cell-­ dependent antibody responses. segregation between the outer and centre follicle. Nature.
Immunol Rev. 1992;126:143‐161. 2009;460:1122‐1126.
84. Bannard O, McGowan SJ, Ersching J, et al. Ubiquitin-­m ediated 105. Sinha RK, Park C, Hwang IY, Davis MD, Kehrl JH. B lymphocytes
fluctuations in MHC class II facilitate efficient germinal center exit lymph nodes through cortical lymphatic sinusoids by a mech-
B cell responses. J Exp Med. 2016;213:993‐1009. anism independent of sphingosine-­1-­phosphate-­mediated chemo-
85. Martinez-Valdez H, Guret C, de Bouteiller O, Fugier I, Banchereau taxis. Immunity. 2009;30:434‐446.
J, Liu YJ. Human germinal center B cells express the apoptosis-­ 106. Grigorova IL, Panteleev M, Cyster JG. Lymph node cortical sinus
inducing genes Fas, c-­myc, P53, and Bax but not the survival gene organization and relationship to lymphocyte egress dynamics and
bcl-­2. J Exp Med. 1996;183:971‐977. antigen exposure. Proc Natl Acad Sci USA. 2010;107:20447‐20452.
|
24       HABERMAN et al.

107. Yi T, Cyster JG. EBI2-­mediated bridging channel positioning sup- 1 29. Haynes NM, Allen CD, Lesley R, Ansel KM, Killeen N,
ports splenic dendritic cell homeostasis and particulate antigen Cyster JG. Role of CXCR5 and CCR7 in follicular Th cell po-
capture. Elife. 2013;2:e00757. sitioning and appearance of a programmed cell death gene-­
108. Kelly LM, Pereira JP, Yi T, Xu Y, Cyster JG. EBI2 guides se- 1high germinal center-­a ssociated subpopulation. J Immunol.
rial movements of activated B cells and ligand activity is de- 2007;179:5099‐5108.
tectable in lymphoid and nonlymphoid tissues. J Immunol. 130. Pereira JP, Kelly LM, Cyster JG. Finding the right niche: B-­cell mi-
2011;187:3026‐3032. gration in the early phases of T-­dependent antibody responses. Int
109. Rodda LB, Lu E, Bennett ML, et al. Single-­cell RNA sequencing of Immunol. 2010;22:413‐419.
lymph node stromal cells reveals niche-­associated heterogeneity. 131. Goenka R, Barnett LG, Silver JS, et al. Cutting edge: dendritic cell-­
Immunity. 2018;48:1014‐1028 e6. restricted antigen presentation initiates the follicular helper T cell
110. Batista FD, Harwood NE. The who, how and where of antigen pre- program but cannot complete ultimate effector differentiation. J
sentation to B cells. Nat Rev Immunol. 2009;9:15‐27. Immunol. 2011;187:1091‐1095.
111. Harwood NE, Batista FD. The antigen expressway: follicular con- 132. Choi YS, Kageyama R, Eto D, et al. ICOS receptor instructs T follic-
duits carry antigen to B cells. Immunity. 2009;30:177‐179. ular helper cell versus effector cell differentiation via induction of
112. Heesters BA, van der Poel CE, Das A, Carroll MC. Antigen presen- the transcriptional repressor Bcl6. Immunity. 2011;34:932‐946.
tation to B cells. Trends Immunol. 2016;37:844‐854. 133. Baumjohann D, Okada T, Ansel KM. Cutting edge: distinct waves
113. Junt T, Moseman EA, Iannacone M, et al. Subcapsular sinus mac- of BCL6 expression during T follicular helper cell development. J
rophages in lymph nodes clear lymph-­borne viruses and present Immunol. 2011;187:2089‐2092.
them to antiviral B cells. Nature. 2007;450:110‐114. 134. Deenick EK, Chan A, Ma CS, et al. Follicular helper T cell differen-
114. Carrasco YR, Batista FD. B cells acquire particulate antigen tiation requires continuous antigen presentation that is indepen-
in a macrophage-­rich area at the boundary between the folli- dent of unique B cell signaling. Immunity. 2010;33:241‐253.
cle and the subcapsular sinus of the lymph node. Immunity. 135. Poholek AC, Hansen K, Hernandez SG, et  al. In vivo regula-
2007;27:160‐171. tion of Bcl6 and T follicular helper cell development. J Immunol.
115. Qi H, Egen JG, Huang AY, Germain RN. Extrafollicular activation 2010;185:313‐326.
of lymph node B cells by antigen-­bearing dendritic cells. Science. 136. Hasbold J, Corcoran LM, Tarlinton DM, Tangye SG, Hodgkin PD.
2006;312:1672‐1676. Evidence from the generation of immunoglobulin G-­secreting cells
116. Wykes M, Pombo A, Jenkins C, MacPherson GG. Dendritic cells that stochastic mechanisms regulate lymphocyte differentiation.
interact directly with naive B lymphocytes to transfer antigen Nat Immunol. 2004;5:55‐63.
and initiate class switching in a primary T-­dependent response. J 137. Hasbold J, Lyons AB, Kehry MR, Hodgkin PD. Cell division number
Immunol. 1998;161:1313‐1319. regulates IgG1 and IgE switching of B cells following stimulation by
117. Damdinsuren B, Zhang Y, Khalil A, et  al. Single round of antigen CD40 ligand and IL-­4. Eur J Immunol. 1998;28:1040‐1051.
receptor signaling programs naive B cells to receive T cell help. 138. Hodgkin PD, Lee JH, Lyons AB. B cell differentiation and iso-
Immunity. 2010;32:355‐366. type switching is related to division cycle number. J Exp Med.
118. Turner JS, Marthi M, Benet ZL, Grigorova I. Transiently antigen-­ 1996;184:277‐281.
primed B cells return to naive-­like state in absence of T-­cell help. 139. Paus D, Phan TG, Chan TD, Gardam S, Basten A, Brink R. Antigen
Nat Commun. 2017;8:15072. recognition strength regulates the choice between extrafollicular
119. Garside P, Ingulli E, Merica RR, Johnson JG, Noelle RJ, Jenkins MK. plasma cell and germinal center B cell differentiation. J Exp Med.
Visualization of specific B and T lymphocyte interactions in the 2006;203:1081‐1091.
lymph node. Science. 1998;281:96‐99. 140. O'Connor BP, Vogel LA, Zhang W, et  al. Imprinting the fate of
120. Okada T, Miller MJ, Parker I, et  al. Antigen-­engaged B cells un- antigen-­reactive B cells through the affinity of the B cell receptor.
dergo chemotaxis toward the T zone and form motile conjugates J Immunol. 2006;177:7723‐7732.
with helper T cells. PLoS Biol. 2005;3:e150. 141. Schwickert TA, Victora GD, Fooksman DR, et al. A dynamic T cell-­
121. Reif K, Ekland EH, Ohl L, et  al. Balanced responsiveness to che- limited checkpoint regulates affinity-­dependent B cell entry into
moattractants from adjacent zones determines B-­cell position. the germinal center. J Exp Med. 2011;208:1243‐1252.
Nature. 2002;416:94‐99. 142. Huang CX, Gonzalez DG, Cote CM, et al. The BCL6 RD2 domain
122. Suan D, Nguyen A, Moran I, et al. T follicular helper cells have dis- governs commitment of activated B cells to form germinal centers.
tinct modes of migration and molecular signatures in naive and Cell Rep. 2014;8:1497‐1508.
memory immune responses. Immunity. 2015;42:704‐718. 143. Ci W, Polo JM, Cerchietti L, et  al. The BCL6 transcriptional pro-
123. Lund FE, Randall TD. Effector and regulatory B cells: modulators of gram features repression of multiple oncogenes in primary B cells
CD4+ T cell immunity. Nat Rev Immunol. 2010;10:236‐247. and is deregulated in DLBCL. Blood. 2009;113:5536‐5548.
124. Shi W, Liao Y, Willis SN, et  al. Transcriptional profiling of mouse 144. Johnston RJ, Poholek AC, DiToro D, et  al. Bcl6 and Blimp-­1 are
B cell terminal differentiation defines a signature for antibody-­ reciprocal and antagonistic regulators of T follicular helper cell dif-
secreting plasma cells. Nat Immunol. 2015;16:663‐673. ferentiation. Science. 2009;325:1006‐1010.
125. Weisel F, Shlomchik M. Memory B cells of mice and humans. Annu 145. Tunyaplin C, Shaffer AL, Angelin-Duclos CD, Yu X, Staudt LM,
Rev Immunol. 2017;35:255‐284. Calame KL. Direct repression of prdm1 by Bcl-­6 inhibits plasma-
126. Kerfoot SM, Yaari G, Patel JR, et  al. Germinal center B cell and cytic differentiation. J Immunol. 2004;173:1158‐1165.
T follicular helper cell development initiates in the interfollicular 146. Crotty S, Johnston RJ, Schoenberger SP. Effectors and memo-
zone. Immunity. 2011;34:947‐960. ries: Bcl-­6 and Blimp-­1 in T and B lymphocyte differentiation. Nat
127. Kitano M, Moriyama S, Ando Y, et  al. Bcl6 protein expression Immunol. 2010;11:114‐120.
shapes pre-­germinal center B cell dynamics and follicular helper T 147. Basso K, Dalla-Favera R. Roles of BCL6 in normal and transformed
cell heterogeneity. Immunity. 2011;34:961‐972. germinal center B cells. Immunol Rev. 2012;247:172‐183.
128. Shaffer AL, Yu X, He Y, Boldrick J, Chan EP, Staudt LM. BCL-­6 re- 148. Shaffer AL, Lin KI, Kuo TC, et al. Blimp-­1 orchestrates plasma cell
presses genes that function in lymphocyte differentiation, inflam- differentiation by extinguishing the mature B cell gene expression
mation, and cell cycle control. Immunity. 2000;13:199‐212. program. Immunity. 2002;17:51‐62.
HABERMAN et al. |
      25

149. Nutt SL, Hodgkin PD, Tarlinton DM, Corcoran LM. The gen- 169. Basso K, Dalla-Favera R. BCL6: master regulator of the germinal
eration of antibody-­ secreting plasma cells. Nat Rev Immunol. center reaction and key oncogene in B cell lymphomagenesis. Adv
2015;15:160‐171. Immunol. 2010;105:193‐210.
150. Ochiai K, Maienschein-Cline M, Simonetti G, et al. Transcriptional 170. Hatzi K, Jiang YW, Huang CX, et al. A hybrid mechanism of action
regulation of germinal center B and plasma cell fates by dynamical for BCL6 in B cells defined by formation of functionally distinct
control of IRF4. Immunity. 2013;38:918‐929. complexes at enhancers and promoters. Cell Rep. 2013;4:578‐588.
151. Saito M, Gao J, Basso K, et  al. A signaling pathway mediating 171. Hatzi K, Nance JP, Kroenke MA, et  al. BCL6 orchestrates Tfh
downregulation of BCL6 in germinal center B cells is blocked cell differentiation via multiple distinct mechanisms. J Exp Med.
by BCL6 gene alterations in B cell lymphoma. Cancer Cell. 2015;212:539‐553.
2007;12:280‐292. 172. Beguelin W, Popovic R, Teater M, et al. EZH2 is required for ger-
152. Kwon H, Thierry-Mieg D, Thierry-Mieg J, et  al. Analysis of minal center formation and somatic EZH2 mutations promote lym-
interleukin-­21-­induced Prdm1 gene regulation reveals functional phoid transformation. Cancer Cell. 2013;23:677‐692.
cooperation of STAT3 and IRF4 transcription factors. Immunity. 173. Velichutina I, Shaknovich R, Geng H, et al. EZH2-­mediated epigen-
2009;31:941‐952. etic silencing in germinal center B cells contributes to proliferation
153. Klein U, Casola S, Cattoretti G, et al. Transcription factor IRF4 con- and lymphomagenesis. Blood. 2010;116:5247‐5255.
trols plasma cell differentiation and class-­switch recombination. 174. Belver L, Papavasiliou FN, Ramiro AR. MicroRNA control of
Nat Immunol. 2006;7:773‐782. lymphocyte differentiation and function. Curr Opin Immunol.
154. Sciammas R, Shaffer AL, Schatz JH, Zhao H, Staudt LM, Singh H. 2011;23:368‐373.
Graded expression of interferon regulatory factor-­4 coordinates 175. O'Connell RM, Rao DS, Chaudhuri AA, Baltimore D. Physiological
isotype switching with plasma cell differentiation. Immunity. and pathological roles for microRNAs in the immune system. Nat
2006;25:225‐236. Rev Immunol. 2010;10:111‐122.
155. Willis SN, Good-Jacobson KL, Curtis J, et al. Transcription factor 176. Barnett BE, Ciocca ML, Goenka R, et al. Asymmetric B cell division
IRF4 regulates germinal center cell formation through a B cell-­ in the germinal center reaction. Science. 2012;335:342‐344.
intrinsic mechanism. J Immunol. 2014;192:3200‐3206. 177. Hawkins ED, Oliaro J, Kallies A, et  al. Regulation of asymmetric
156. Koguchi Y, Buenafe AC, Thauland TJ, et  al. Preformed CD40L is cell division and polarity by Scribble is not required for humoral
stored in Th1, Th2, Th17, and T follicular helper cells as well as immunity. Nat Commun. 2013;4:1801.
CD4+ 8-­ thymocytes and invariant NKT cells but not in Treg cells. 178. Di Niro R, Lee SJ, Vander Heiden JA, et  al. Salmonella infection
PLoS ONE. 2012;7:e31296. drives promiscuous B cell activation followed by extrafollicular af-
157. Rothoeft T, Balkow S, Krummen M, et al. Structure and duration of finity maturation. Immunity. 2015;43:120‐131.
contact between dendritic cells and T cells are controlled by T cell 179. Thaunat O, Granja AG, Barral P, et al. Asymmetric segregation of
activation state. Eur J Immunol. 2006;36:3105‐3117. polarized antigen on B cell division shapes presentation capacity.
158. Fusco AJ, Savinova OV, Talwar R, Kearns JD, Hoffmann A, Ghosh Science. 2012;335:475‐479.
G. Stabilization of RelB requires multidomain interactions with 180. Thaunat O, Batista FD. Daughter B cells are not created equal:
p100/p52. J Biol Chem. 2008;283:12324‐12332. asymmetric segregation of antigen during B cell division. Cell Cycle.
159. Vallabhapurapu S, Karin M. Regulation and function of NF-­kappaB 2012;11:2219‐2220.
transcription factors in the immune system. Annu Rev Immunol. 181. Luthje K, Kallies A, Shimohakamada Y, et al. The development and
2009;27:693‐733. fate of follicular helper T cells defined by an IL-­21 reporter mouse.
160. Homig-Holzel C, Hojer C, Rastelli J, et al. Constitutive CD40
Nat Immunol. 2012;13:491‐U93.
signaling in B cells selectively activates the noncanonical NF-­ 182. Gonzalez DG, Cote CM, Patel J, et al. Non-­redundant roles of IL-­21
kappa B pathway and promotes lymphomagenesis. J Exp Med. and IL-­4 in the phased initiation of germinal center B cells and sub-
2008;205:1317‐1329. sequent self-­renewal transitions. J Immunol 2018;201:3569‐3579.
161. Xu J, Foy TM, Laman JD, et al. Mice deficient for the CD40 ligand. 183. Chang PP, Barral P, Fitch J, et al. Identification of Bcl-­6-­dependent
Immunity. 1994;1:423‐431. follicular helper NKT cells that provide cognate help for B cell re-
162. Allman D, Jain A, Dent A, et al. BCL-­6 expression during B-­cell ac- sponses. Nat Immunol. 2012;13:35‐43.
tivation. Blood. 1996;87:5257‐5268. 184. King IL, Fortier A, Tighe M, et al. Invariant natural killer T cells di-
163. Bolduc A, Long AE, Stapler D, et al. Constitutive CD40L expres- rect B cell responses to cognate lipid antigen in an IL-­21-­dependent
sion on B cells prematurely terminates germinal center response manner. Nat Immunol. 2012;13:44‐50.
and leads to augmented plasma cell production in T cell areas. J 185. Tonti E, Fedeli M, Napolitano A, et al. Follicular helper NKT cells
Immunol. 2010;185:220‐230. induce limited B cell responses and germinal center formation in
164. Kishi Y, Aiba Y, Higuchi T, et al. Augmented antibody response with the absence of CD4(+) T cell help. J Immunol. 2012;188:3217‐3222.
premature germinal center regression in CD40L transgenic mice. J 186. Gaya M, Barral P, Burbage M, et al. Initiation of antiviral B cell im-
Immunol. 2010;185:211‐219. munity relies on innate signals from spatially positioned NKT cells.
165. Erickson LD, Durell BG, Vogel LA, et al. Short-­circuiting long-­lived Cell. 2018;172:517‐533 e20.
humoral immunity by the heightened engagement of CD40. J Clin 187. Voehringer D, Reese TA, Huang X, Shinkai K, Locksley RM. Type
Invest. 2002;109:613‐620. 2 immunity is controlled by IL-­4/IL-­13 expression in hematopoi-
166. Grumont RJ, Gerondakis S. Rel induces interferon regulatory fac- etic non-­eosinophil cells of the innate immune system. J Exp Med.
tor 4 (IRF-­4) expression in lymphocytes: modulation of interferon-­ 2006;203:1435‐1446.
regulated gene expression by rel/nuclear factor kappaB. J Exp Med. 188. Sokol CL, Barton GM, Farr AG, Medzhitov R. A mechanism for
2000;191:1281‐1292. the initiation of allergen-­induced T helper type 2 responses. Nat
167. Ghetu AF, Corcoran CM, Cerchietti L, Bardwell VJ, Melnick A, Immunol. 2008;9:310‐318.
Prive GG. Structure of a BCOR corepressor peptide in complex 189. Perrigoue JG, Saenz SA, Siracusa MC, et  al. MHC class II-­
with the BCL6 BTB domain dimer. Mol Cell. 2008;29:384‐391. dependent basophil-­CD4+ T cell interactions promote T(H)2
168. Ahmad KF, Melnick A, Lax S, et  al. Mechanism of SMRT core- cytokine-­dependent immunity. Nat Immunol. 2009;10:697‐705.
pressor recruitment by the BCL6 BTB domain. Mol Cell. 190. Gessner A, Mohrs K, Mohrs M. Mast cells, basophils, and eosino-
2003;12:1551‐1564. phils acquire constitutive IL-­4 and IL-­13 transcripts during lineage
|
26       HABERMAN et al.

differentiation that are sufficient for rapid cytokine production. J 211. Paul WE. History of interleukin-­4. Cytokine. 2015;75:3‐7.
Immunol. 2005;174:1063‐1072. 12. Dasoveanu DC, Shipman WD, Chia JJ, Chyou S, Lu TT. Regulation
2
191. Linterman MA, Beaton L, Yu D, et al. IL-­21 acts directly on B cells of lymph node vascular-­stromal compartment by dendritic cells.
to regulate Bcl-­6 expression and germinal center responses. J Exp Trends Immunol. 2016;37:764‐777.
Med. 2010;207:353‐363. 213. Kapasi ZF, Qin D, Kerr WG, et  al. Follicular dendritic cell
192. Zotos D, Coquet JM, Zhang Y, et al. IL-­21 regulates germinal cen- (FDC) precursors in primary lymphoid tissues. J Immunol.
ter B cell differentiation and proliferation through a B cell-­intrinsic 1998;160:1078‐1084.
mechanism. J Exp Med. 2010;207:365‐378. 214. Balogh P, Aydar Y, Tew JG, Szakal AK. Ontogeny of the follicular
193. Cunningham AF, Serre K, Toellner KM, et  al. Pinpointing IL-­4-­ dendritic cell phenotype and function in the postnatal murine
independent acquisition and IL-­4-­influenced maintenance of Th2 spleen. Cell Immunol. 2001;214:45‐53.
activity by CD4 T cells. Eur J Immunol. 2004;34:686‐694. 215. Garin A, Meyer-Hermann M, Contie M, et al. Toll-­like receptor 4
194. Turqueti-Neves A, Otte M, Prazeres da Costa O, et  al. B-­cell-­ signaling by follicular dendritic cells is pivotal for germinal center
intrinsic STAT6 signaling controls germinal center formation. Eur J onset and affinity maturation. Immunity. 2010;33:84‐95.
Immunol. 2014;44:2130‐2138. 216. El Shikh ME, El Sayed RM, Wu Y, Szakal AK, Tew JG. TLR4 on follic-
195. Chevrier S, Kratina T, Emslie D, Tarlinton DM, Corcoran LM. IL4 ular dendritic cells: an activation pathway that promotes accessory
and IL21 cooperate to induce the high Bcl6 protein level required activity. J Immunol. 2007;179:4444‐4450.
for germinal center formation. Immunol Cell Biol. 2017;95:925‐932. 217. Victoratos P, Lagnel J, Tzima S, et  al. FDC-­specific functions of
196. Arguni E, Arima M, Tsuruoka N, Sakamoto A, Hatano M, Tokuhisa p55TNFR and IKK2 in the development of FDC networks and of
T. JunD/AP-­1 and STAT3 are the major enhancer molecules antibody responses. Immunity. 2006;24:65‐77.
for high Bcl6 expression in germinal center B cells. Int Immunol. 218. El Shikh ME, El Sayed R, Szakal AK, Tew JG. Follicular dendritic cell
2006;18:1079‐1089. (FDC)-­FcgammaRIIB engagement via immune complexes induces
197. Ozaki K, Spolski R, Feng CG, et al. A critical role for IL-­21 in regu- the activated FDC phenotype associated with secondary follicle
lating immunoglobulin production. Science. 2002;298:1630‐1634. development. Eur J Immunol. 2006;36:2715‐2724.
198. Ozaki K, Spolski R, Ettinger R, et al. Regulation of B cell differentia- 219. Balogh P, Aydar Y, Tew JG, Szakal AK. Appearance and phenotype
tion and plasma cell generation by IL-­21, a novel inducer of Blimp-­1 of murine follicular dendritic cells expressing VCAM-­1. Anat Rec.
and Bcl-­6. J Immunol. 2004;173:5361‐5371. 2002;268:160‐168.
199. Kuchen S, Robbins R, Sims GP, et al. Essential role of IL-­21 in B cell 220. Heesters BA, Myers RC, Carroll MC. Follicular dendritic cells: dy-
activation, expansion, and plasma cell generation during CD4(+) T namic antigen libraries. Nat Rev Immunol. 2014;14:495‐504.
cell-­B cell collaboration. J Immunol. 2007;179:5886‐5896. 221. Kranich J, Krautler NJ, Heinen E, et  al. Follicular dendritic cells
200. Bryant VL, Ma CS, Avery DT, et al. Cytokine-­mediated regulation control engulfment of apoptotic bodies by secreting Mfge8. J Exp
of human B cell differentiation into Ig-­secreting cells: predomi- Med. 2008;205:1293‐1302.
nant role of IL-­21 produced by CXCR5+ T follicular helper cells. J 222. Kim HS, Zhang X, Choi YS. Activation and proliferation of follicu-
Immunol. 2007;179:8180‐8190. lar dendritic cell-­like cells by activated T lymphocytes. J Immunol.
201. Defrance T, Vanbervliet B, Pene J, Banchereau J. Human recom- 1994;153:2951‐2961.
binant Il-­4 induces activated lymphocytes-­B to produce IgG and 2 23. Yoshida K, van den Berg TK, Dijkstra CD. Two functionally
IgM. J Immunol. 1988;141:2000‐2005. different follicular dendritic cells in secondary lymphoid
202. Russell SM, Keegan AD, Harada N, et  al. Interleukin-­2 receptor follicles of mouse spleen, as revealed by CR1/2 and FcR
gamma chain: a functional component of the interleukin-­4 recep- gamma II-­ m ediated immune-­ c omplex trapping. Immunology.
tor. Science. 1993;262:1880‐1883. 1993;80:34‐39.
203. Habib T, Senadheera S, Weinberg K, Kaushansky K. The common 224. Sacedon R, Diez B, Nunez V, et al. Sonic hedgehog is produced by
gamma chain (gamma c) is a required signaling component of the follicular dendritic cells and protects germinal center B cells from
IL-­21 receptor and supports IL-­21-­induced cell proliferation via apoptosis. J Immunol. 2005;174:1456‐1461.
JAK3. Biochemistry. 2002;41:8725‐8731. 225. Crompton T, Outram SV, Hager-Theodorides AL. Sonic hedgehog
204. Spolski R, Leonard WJ. Interleukin-­21: a double-­edged sword with signalling in T-­cell development and activation. Nat Rev Immunol.
therapeutic potential. Nat Rev Drug Discov. 2014;13:379‐395. 2007;7:726‐735.
205. Asao H, Okuyama C, Kumaki S, et al. Cutting edge: the common 226. Trowbridge JJ, Scott MP, Bhatia M. Hedgehog modulates cell cycle
gamma-­chain is an indispensable subunit of the IL-­21 receptor regulators in stem cells to control hematopoietic regeneration.
complex. J Immunol. 2001;167:1‐5. Proc Natl Acad Sci USA. 2006;103:14134‐14139.
206. Zeng R, Spolski R, Casas E, Zhu W, Levy DE, Leonard WJ.
227. Campbell C, Risueno RM, Salati S, Guezguez B, Bhatia M. Signal
The molecular basis of IL-­ 21-­mediated proliferation. Blood. control of hematopoietic stem cell fate: Wnt, Notch, and Hedgehog
2007;109:4135‐4142. as the usual suspects. Curr Opin Hematol. 2008;15:319‐325.
207. Avery DT, Deenick EK, Ma CS, et  al. B cell-­intrinsic signal- 228. St-Jacques B, Dassule HR, Karavanova I, et  al. Sonic hedge-
ing through IL-­ 21 receptor and STAT3 is required for estab- hog signaling is essential for hair development. Curr Biol.
lishing long-­ lived antibody responses in humans. J Exp Med. 1998;8:1058‐1068.
2010;207:155‐171. 229. Allen CD, Cyster JG. Follicular dendritic cell networks of primary
208. Deenick EK, Avery DT, Chan A, et  al. Naive and memory human follicles and germinal centers: phenotype and function. Semin
B cells have distinct requirements for STAT3 activation to dif- Immunol. 2008;20:14‐25.
ferentiate into antibody-­ secreting plasma cells. J Exp Med. 230. Smith KG, Light A, Nossal GJ, Tarlinton DM. The extent of affin-
2013;210:2739‐2753. ity maturation differs between the memory and antibody-­forming
209. Mehta DS, Wurster AL, Grusby MJ. Biology of IL-­21 and the IL-­21 cell compartments in the primary immune response. EMBO J.
receptor. Immunol Rev. 2004;202:84‐95. 1997;16:2996‐3006.
210. Quelle FW, Shimoda K, Thierfelder W, et  al. Cloning of murine 231. Peperzak V, Vikstrom I, Walker J, et al. Mcl-­1 is essential for the
Stat6 and human Stat6, Stat proteins that are tyrosine phosphor- survival of plasma cells. Nat Immunol. 2013;14:290‐297.
ylated in responses to IL-­4 and IL-­3 but are not required for mito- 232. Weisel FJ, Zuccarino-Catania GV, Chikina M, Shlomchik

genesis. Mol Cell Biol. 1995;15:3336‐3343. MJ. A temporal switch in the germinal center determines
HABERMAN et al. |
      27

differential output of memory B and plasma cells. Immunity. 251. Roth K, Oehme L, Zehentmeier S, Zhang Y, Niesner R, Hauser AE.
2016;44:116‐130. Tracking plasma cell differentiation and survival. Cytometry A.
233. Zhang Y, Garcia-Ibanez L, Toellner KM. Regulation of germinal cen- 2014;85:15‐24.
ter B-­cell differentiation. Immunol Rev. 2016;270:8‐19. 252. Hauser AE, Debes GF, Arce S, et al. Chemotactic responsiveness
234. Shinnakasu R, Inoue T, Kometani K, et  al. Regulated selection of toward ligands for CXCR3 and CXCR4 is regulated on plasma
germinal-­center cells into the memory B cell compartment. Nat blasts during the time course of a memory immune response. J
Immunol. 2016;17:861‐869. Immunol. 2002;169:1277‐1282.
235. Coffey F, Alabyev B, Manser T. Initial clonal expansion of germinal 253. Hargreaves DC, Hyman PL, Lu TT, et al. A coordinated change in
center B cells takes place at the perimeter of follicles. Immunity. chemokine responsiveness guides plasma cell movements. J Exp
2009;30:599‐609. Med. 2001;194:45‐56.
236. Nikbakht N, Shen S, Manser T. Cutting edge: macrophages are 254. Bhattacharya D, Cheah MT, Franco CB, et al. Transcriptional pro-
required for localization of antigen-­activated B cells to the fol- filing of antigen-­dependent murine B cell differentiation and mem-
licular perimeter and the subsequent germinal center response. J ory formation. J Immunol. 2007;179:6808‐6819.
Immunol. 2013;190:4923‐4927. 255. Elgueta R, Marks E, Nowak E, et al. CCR6-­dependent positioning
237. Taylor JJ, Pape KA, Jenkins MK. A germinal center-­independent of memory B cells is essential for their ability to mount a recall
pathway generates unswitched memory B cells early in the pri- response to antigen. J Immunol. 2015;194:505‐513.
mary response. J Exp Med. 2012;209:597‐606. 256. Suan D, Krautler NJ, Maag JLV, et al. CCR6 defines memory B cell
238. Duan S, Cermak L, Pagan JK, et  al. FBXO11 targets BCL6 for precursors in mouse and human germinal centers, revealing light-­
degradation and is inactivated in diffuse large B-­cell lymphomas. zone location and predominant low antigen affinity. Immunity.
Nature. 2012;481:90‐93. 2017;47:1142‐1153 e4.
239. Saito Y, Liang G, Egger G, et al. Specific activation of microR- 257. Piqueras B, Connolly J, Freitas H, Palucka AK, Banchereau J. Upon
NA-­127 with downregulation of the proto-­oncogene BCL6 by viral exposure, myeloid and plasmacytoid dendritic cells produce 3
chromatin-­ modifying drugs in human cancer cells. Cancer Cell. waves of distinct chemokines to recruit immune effectors. Blood.
2006;9:435‐443. 2006;107:2613‐2618.
240. Takahashi H, Kanno T, Nakayamada S, et al. TGF-­beta and retinoic 258. Zhang Y, Roth TL, Gray EE, et  al. Migratory and adhesive cues
acid induce the microRNA miR-­10a, which targets Bcl-­6 and con- controlling innate-­like lymphocyte surveillance of the pathogen-­
strains the plasticity of helper T cells. Nat Immunol. 2012;13:587‐595. exposed surface of the lymph node. Elife. 2016;5:e18156.
241. Zhang Y, Tech L, George LA, et al. Plasma cell output from germinal 259. Liu YJ, Oldfield S, MacLennan IC. Memory B cells in T cell-­
centers is regulated by signals from Tfh and stromal cells. J Exp dependent antibody responses colonize the splenic marginal
Med. 2018;215:1227‐1243. zones. Eur J Immunol. 1988;18:355‐362.
242. Rodda LB, Bannard O, Ludewig B, Nagasawa T, Cyster JG. 260. Anderson SM, Tomayko MM, Ahuja A, Haberman AM,

Phenotypic and morphological properties of germinal cen- Shlomchik MJ. New markers for murine memory B cells
ter dark zone Cxcl12-­ expressing reticular cells. J Immunol. that define mutated and unmutated subsets. J Exp Med.
2015;195:4781‐4791. 2007;204:2103‐2114.
243. Gretz JE, Anderson AO, Shaw S. Cords, channels, corridors and 261. Liu YJ, Zhang J, Lane PJ, Chan EY, MacLennan IC. Sites of spe-
conduits: critical architectural elements facilitating cell inter- cific B cell activation in primary and secondary responses to T
actions in the lymph node cortex. Immunol Rev. 1997;156:11‐24. cell-­dependent and T cell-­independent antigens. Eur J Immunol.
244. Sixt M, Kanazawa N, Selg M, et  al. The conduit system trans- 1991;21:2951‐2962.
ports soluble antigens from the afferent lymph to resident 262. Meyer-Hermann M, Mohr E, Pelletier N, Zhang Y, Victora GD,
dendritic cells in the T cell area of the lymph node. Immunity. Toellner KM. A theory of germinal center B cell selection, division,
2005;22:19‐29. and exit. Cell Rep. 2012;2:162‐174.
245. Sayin I, Radtke AJ, Vella LA, et al. Spatial distribution and function 263. Gonzalez SF, Degn SE, Pitcher LA, Woodruff M, Heesters BA,
of T follicular regulatory cells in human lymph nodes. J Exp Med. Carroll MC. Trafficking of B cell antigen in lymph nodes. Annu Rev
2018;215:1531‐1542. Immunol. 2011;29:215‐233.
246. Lim HW, Hillsamer P, Kim CH. Regulatory T cells can migrate to 264. Gerner MY, Torabi-Parizi P, Germain RN. Strategically localized
follicles upon T cell activation and suppress GC-­T h cells and GC-­ dendritic cells promote rapid T cell responses to lymph-­borne par-
Th cell-­driven B cell responses. J Clin Invest. 2004;114:1640‐1649. ticulate antigens. Immunity. 2015;42:172‐185.
247. Lim HW, Hillsamer P, Banham AH, Kim CH. Cutting edge: direct 265. Bergtold A, Desai DD, Gavhane A, Clynes R. Cell surface recycling
suppression of B cells by CD4+ CD25+ regulatory T cells. J Immunol. of internalized antigen permits dendritic cell priming of B cells.
2005;175:4180‐4183. Immunity. 2005;23:503‐514.
248. Sage PT, Ron-Harel N, Juneja VR, et al. Suppression by TFR cells
leads to durable and selective inhibition of B cell effector function.
Nat Immunol. 2016;17:1436‐1446. How to cite this article: Haberman AM, Gonzalez DG, Wong P,
249. Dogan I, Bertocci B, Vilmont V, et al. Multiple layers of B cell memory Zhang T, Kerfoot SM. Germinal center B ­cell initiation, GC
with different effector functions. Nat Immunol. 2009;10:1292‐1299.
maturation, and the coevolution of its stromal cell niches.
250. Moran I, Nguyen A, Khoo WH, et  al. Memory B cells are reac-
tivated in subcapsular proliferative foci of lymph nodes. Nat Immunol Rev. 2019;288:10–27. https://doi.org/10.1111/
Commun. 2018;9:3372. imr.12731

Das könnte Ihnen auch gefallen