kclpure.kcl.ac.uk · Web viewDiversification of the antibody repertoire is essential for the normal...

49
Transcriptional networks controlling B cell germinal center activities Timothy Recaldin 1,2 and David J. Fear 1,2 1 Division of Asthma, Allergy and Lung Biology, King’s College London, London, United Kingdom. 2 Medical Research Council & Asthma U.K. Centre in Allergic Mechanisms of Asthma, King’s College London, London, United Kingdom.

Transcript of kclpure.kcl.ac.uk · Web viewDiversification of the antibody repertoire is essential for the normal...

Transcriptional networks controlling B cell germinal center activities

Timothy Recaldin1,2 and David J. Fear1,2

1Division of Asthma, Allergy and Lung Biology, King’s College London, London, United

Kingdom.

2Medical Research Council & Asthma U.K. Centre in Allergic Mechanisms of Asthma,

King’s College London, London, United Kingdom.

Corresponding author: David J. Fear: Dept. Asthma, Allergy and Respiratory Science,

King’s College London, Guy’s Hospital, St. Thomas Street, London, SE19RT, United

Kingdom, [email protected]

Abstract

Diversification of the antibody repertoire is essential for the normal operation of the

vertebrate adaptive immune system. Following antigen encounter, B cells are

activated, rapidly proliferate, and undergo two diversification events; somatic

hypermutation (followed by selection), which enhances the affinity of the antibody for its

cognate antigen, and class switch recombination, which alters the effector functions of

the antibody to adapt the response to the challenge faced. B cells must then

differentiate into antibody-secreting plasma cells or long-lived memory B cells. These

activities take place in specialised immunological environments called germinal

centers, usually located in the secondary lymphoid organs. To successfully complete

the germinal center activities, a B cell adopts a transcriptional program that allows it to

migrate to specific sites within the germinal center, proliferate, modify its DNA

recombination and repair pathways, alter it apoptotic potential and finally undergo

terminal differentiation. To coordinate these processes, B cells employ a number of

“master regulator” transcription factors which mediate wholesale transcriptomic

changes. These master transcription factors are mutually antagonistic and form a

complex regulatory network to maintain distinct gene expression programs. Within this

network, multiple points of positive and negative feedback ensure the expression of the

“master regulators”, augmented by a number of “secondary” factors that reinforce these

networks and sense the progress of the immune response. In this review we will

discuss the different activities B cells must undertake to mount a successful T cell-

dependent immune response and describe how a regulatory network of transcription

factors controls these processes.

Introduction

In order for our bodies to mount a successful humoral immune response, B cells must

first encounter, and then be activated by, their cognate antigens. Following activation,

the B cell (or its progeny) diversifies the antibody it produces, increasing its affinity for

antigen and altering its effector function, thus tailoring the response to the

immunological challenge faced. The B cell then differentiates into either a specialised

“antibody-secreting cell” (plasmablast or plasma cell (PC)) to eliminate the challenge,

or a long-lived memory B cell, so that a more rapid (and specific) response can be

mounted upon reencountering the same foe. This sequence of events takes place

within a specialised immunological environment termed the germinal center (GC). First

described by Flemming in 1884 (1), the GC is a transient structure that forms within

secondary lymphoid organs such as the lymph nodes, spleen and Peyer’s patches, and

is essential for an efficient response to a T cell-dependent antigen. GC like structures

can also form within “local” ectopic tissues(2), promoting B cell diversification(3, 4),

where they play an important role in the pathology of chronic inflammation, infection,

autoimmunity and atopic disease.

In the GC, B cells undergo rapid clonal expansion (proliferation) and express the

genome mutator enzyme activation-induced cytidine deaminase (AID). AID is

indispensible for two key GC processes, somatic hypermutation (SHM) and class-

switch recombination (CSR). SHM introduces non-templated mutations in the

immunoglobulin (Ig) variable region, changing the antibody’s affinity for antigen

(reviewed in (5)). CSR alters the antibody effector functions by recombination of the

variable region (initially linked to the μ and δ heavy chain exons, encoding IgM and IgD

respectively) with one of the downstream constant region genes of the α, γ, or ε

isotypes, encoding IgA, IgG and IgE. Each antibody isotype initiates different

downstream immune reactions and thus adapts the response to the nature of the

challenge faced (6). Together these processes shape the antibody response,

producing GC B cells with high affinity, class-switched antibodies that can differentiate

into either antibody-secreting PCs or long-lived memory B cells.

Lymphocytes are the only vertebrate somatic cells that undergo such drastic and

potentially dangerous modifications of their genomic DNA as part of their “normal”

biology (including VDJ recombination that occurs during B and T cell ontogeny (7)). It is

critical that these events are tightly regulated; accumulation of dangerous “off-target”

mutations poses a significant threat to genome integrity and likely explains why B cells

are highly prone to transformation into a variety of leukemias/lymphomas(8).

Successful completion of the GC reaction therefore relies on careful regulation and

coordination of B cell movement, division, apoptosis, differentiation, DNA repair and

recombination. This is achieved through the activation (and repression) of multiple

transcriptional programs that interact in a series of complex regulatory networks,

discussed in this review.

Germinal center structure, formation and B cell movement

GCs are not permanent structures, but rather arise transiently within the lymphoid

tissue such as lymph nodes (and local tissues) in response to a T cell-dependent

antigen (Figure 1). Lymph nodes are composed of multiple lobules surrounded by

lymph-filled sinuses enclosed by a capsule (9). Naïve B and T cells from the circulation

continually cycle through the lymph node, residing within distinct areas of the lobule

where they can interact with specialised antigen presenting cells (APCs) to survey the

antigen environment. The outer layer (cortex) contains follicles of naïve B cells and

follicular dendritic cells (FDCs). These follicles are separated from the T cell zone

(paracortex), which contains naïve T cells and dendritic cells (DCs), by the

interfollicular zone (10, 11).

Circulating naïve B cells bear the chemokine receptor CXCR5 and are thus attracted to

the lymphoid follicles by the chemokine CXCL13, which is expressed by resident FDCs

and marginal reticular cells (12). Similarly, circulating naïve T cells expressing the

chemokine receptor CCR7 are recruited to the T cell zone by fibroblastic reticular cell

expression of the CCR7-ligands, CCL19 and CCL21, (13).

Once in the follicles, naïve B cells interact with antigen via their B cell receptor (BCR).

The antigen is usually displayed on the surface of FDCs, but can also be captured from

macrophages lining the subcapsular sinus (SCS) (14, 15). BCR binding to cognate

antigen activates the B cell, triggering internalisation of the BCR:antigen complex and

subsequent presentation of antigen on the cell surface in the context of MHC class II

molecules (16). BCR engagement also upregulates expression of the chemokine

receptor CCR7 which promotes B cell migration to the periphery of the T cell zone

where its ligands, CCL19 and CCL21, are abundantly expressed (17, 18). During this

phase of activation, the B cells continue to maintain expression of CXCR5; the balance

between CXCL13 expression in the follicles and CCL19/21 expression in the T cell

zone positions B cells at the border of the T cell zone (17). Meanwhile, naïve T cells in

the T cell zone encounter their cognate antigen, here presented by DCs, initiating

commitment towards a T follicular helper (TFH) cell phenotype (19). TFH cell commitment

is accompanied by CCR7 downregulation and CXCR5 upregulation, promoting TFH

migration to the T/B cell boundary where they can support B cell expansion(20, 21).

Two days after antigen encounter, activated B cells find their cognate TFH cells and

form long-lived interactions that result in full B cell activation and proliferation(22, 23).

At this time, a subset of activated B cells move away from the extrafollicular sites into

the SCS where they differentiate into short-lived plasmablasts. These cells secrete

IgM, providing immediate (albeit low specificity) protection to the individual (24). After 3

days, the activated TFH and B cells migrate into the centre of the follicle where the B

cells start to rapidly proliferate. By this time the B cells begin to express the “master

regulator” B cell lymphoma 6 protein (BCL6, see below), which drives the acquisition of

a “GC B cell” phenotype (25). Of note, BCL6 expression is also responsible for

commitment of T cells to the TFH fate (26), thus regulating both the T and B cell GC

transcriptional programs. The rapid proliferation of activated GC B cells within a

network of FDCs, pushes aside the resident follicular B cells to form the early GC over

days 5-6. By day 7 the rapid proliferation of GC B cells, coupled with the continued

influx of activated GC cells, results in the polarisation of the fully formed GC into two

distinct microenvironments, the so called dark and light zones.

In the dark zone, densely packed GC B cells, referred to as centroblasts, divide rapidly,

and undergo SHM. Centroblasts are retained in the dark zone by their expression of

the chemokine receptor CXCR4, the ligand of which, CXCL12, is abundantly expressed

by dark zone stromal cells(27). Downregulation of CXCR4 and upregulation of CD83

and CD86 allow the GC B cells to migrate from the dark zone into the light zone, a less

densely packed compartment populated with TFH cells, macrophages and FDCs (28). In

the light zone, B cells undergo a process of Darwinian selection whereby B cells

producing higher affinity antibodies compete for available antigen (29, 30) and/or T cell

help(31, 32), thus receiving survival signals via BCR binding. Selection promotes

centrocyte reentry into the dark zone for further rounds of mutation and selection.

(Reviewed in Victoria (33). Conversely, lower affinity B cells, receive no survival cues

and undergo apoptosis (34). In parallel, CSR drives apoptosis of undesirable B cell

clones through deletion of the Ig heavy chain in a process called locus suicide

recombination. This prevents BCR expression and thus eliminates the survival signals

the BCR transmits, inducing apoptosis (35). Having survived selection in the light zone,

GC B cells can do one of three things: They can re-enter the dark zone for additional

rounds of proliferation and SHM (36, 37). Alternatively, GC B cells can leave the GC

and differentiate into plasmablasts (precursors of antibody-secreting PCs) or they can

differentiate to form long-lived memory B cells to enable a rapid response upon

reencountering the same antigen (reviewed in (38, 39)).

Coordination of all the events described above are controlled by a number of “master

regulator” transcription factors (Figure 2). In the remainder of this review we will

discuss the role of these transcription factors and how their interacting regulatory

networks control the GC activities.

Master regulators of B cell identity

PAX5

Paired box protein 5 (PAX5) is the master regulator of B cell identity and is expressed

throughout B cell development (40), from pro-B cells (41) through to mature GC B cells

(42). PAX5 directly binds to thousands of DNA sites in B cells and functions by both

activating and repressing gene expression (40). During early B cell development PAX5

is absolutely required for the initial commitment of lymphoid progenitors to the B cell

fate (43) and V-DJ recombination of the Ig locus (44, 45). In mature B cells it regulates

the expression of genes critical to B cell identity, including components of the B cell

receptor (Ig heavy chain and CD79A), CD19, CD21, BLK, IRF4 and IRF8(40). In

addition, PAX5 further reinforces B cell identity by repressing the expression of lineage

inappropriate genes including FLT3, CCR2 and CD28, which are expressed in PCs

following PAX5 downregulation, and M-CSF receptor, NOTCH1, RAMP1, LMO2 and

CCL3, which are expressed in common lymphoid progenitors and myeloid cells (46).

As PAX5 promotes and maintains the expression of the B cell transcriptional program,

its downregulation is required for differentiation into committed Ig-secreting PCs (47)

(see master regulators of plasma cell identity). Critically, PAX5 directly represses the

expression of one of the master regulators of the PC program, XBP1, and its

downregulation is required for Ig secretion(48, 49).

BCL6

As mentioned earlier, B cell lymphoma 6 protein, BCL6, is essential for GC formation

(50) and is considered the master regulator of the GC, where it controls gene

expression programs in both GC B cells (25) and in TFH cells(26) (reviewed in Basso

and Dalla-Favera, 2012(51)). Within these cells BCL6 predominantly functions as a

transcriptional repressor, directly suppressing multiple genes involved in the DNA

damage sensing pathway, including TP53, ATR and CHEK1 and regulators of the cell

cycle, p21, p53 (52, 53); this establishes a transcriptional program that allows both the

rapid proliferation of cells and the tolerance of DNA damage essential to SHM. In

addition, BCL6 controls the migration of B cells into the follicle. To this end, BCL6-

deficient GC B cells fail to upregulate CXCR4 (25), the chemokine receptor responsible

for GC localisation into the dark zone, and fail to downregulate sphingosine-1

phosphate receptor type 1 (S1PR1) (54), which facilitates trafficking of B cells out of

the follicles (55). One of the critical functions of BCL6 appears to be the repression PC

differentiation, in this case mediated by repression of BLIMP1(56) (see below): Thus,

expression of BCL6 in the GC B cell not only activates GC B cell identity but

concomitantly blocks establishment of the PC program. Although BCL6 functions as a

transcriptional repressor, it also indirectly induces AID expression in GC B cells (50) by

inhibiting expression of miR-155 and miR-361, two negative regulators of AID (57).

Similarly, repression of Spi1, IRF8 and MYB is also relieved, all regulators of the GC

transcriptional program. Further supporting GC activities, BCL6 has been shown to

regulate B cell/TFH cell interactions, through an undetermined integrin dependent

mechanism(25).

BLIMP1 and XBP1: Master regulators of plasma cell identity

Two transcription factors, B lymphocyte-induced maturation protein 1 (BLIMP1, also

known as PR domain zinc finger protein 1, PRDM1) and X-box-binding protein 1

(XBP1) are essential for orchestrating PC differentiation (reviewed in (38). BLIMP1 is a

transcriptional repressor that, within the B cell lineage, is exclusively expressed in

antibody-secreting cells, being expressed at a low level in plasmablasts and high level

in mature PCs (58, 59). During PC commitment, BLIMP1 represses the expression of

the B cell specific regulators, PAX5, BCL6, ID3, cMYC and SPIB, (47, 60), thus

allowing expression of XBP1(47). However, although XBP1 appears to act downstream

of BLIMP1 in the regulatory network(61), BLIMP1 is necessary, but not sufficient for

XBP1 expression(47). Furthermore, it seems that BLIMP1 is not required for initiation

of the PC differentiation program since pre-plasma-blasts form in the absence of

BLIMP1(62).

XBP1 acts downstream of BLIMP1, and is a key regulator of PC development, but it is

not absolutely required for the formation of antibody-secreting cells (63). Rather, XBP1

appears to predominantly act to set up the cells to allow for the secretion of vast

quantities of Ig (64), inducing endoplasmic reticulum remodeling, activation of mTOR

(65) and autophagic pathways(66) and the induction of the unfolded protein

response(64). Although much is known regarding the interconnections that exist

between the regulatory networks of these B cell lineage master regulators, questions

still remain as to exactly what initiates each pathway.

Other factors implicated in regulating GC activities.

NF-B

The classical NF-B pathway consists of 3 subunits, cREL, RELA and p50, forming two

major heterodimers, cREL/p50 and RELA/p50 (67). Both cREL and RELA are induced

transiently upon BCR ligation, peaking 1-2 hours after stimulation (68). Here, they

induce expression of chemokines that promote B: T cell interaction, namely CCR7,

which is responsible for B cell movement to the periphery of the T cell zones (17), and

CCL3 and CCL4, two T cell chemoattractants (69).

In the GC, cREL and RELA have non-redundant roles. While NF-B expression is

generally absent in GC B cells, it is detected in a small subset of centrocytes in the light

zone. These cells likely represent positively-selected B cells being primed for cyclic

reentry into the dark zone. cREL-ablation in this context results in the collapse of the

GC. Unlike cREL, RELA-ablation has no impact on GC maintenance, but instead

results in a significant reduction in the prevalence of PCs and a subsequent loss in IgG

serum titres, through impaired upregulation of BLIMP1(67).

cMYC

cMYC is a global driver of cell growth and division. However, counter-intuitively, it is

actively suppressed in the rapidly dividing GC centroblasts by BCL6 (70, 71).

Therefore, dark zone GC B cells must proliferate in a MYC-independent manner. This

is, at least in part, facilitated by BCL6 interaction with the MYC-binding protein, MIZ1,

which together suppress transcription of the cell cycle arrest gene CDKN1A (72).

Despite its absence in centroblasts, cMYC still plays an essential role in the initiation

and functioning of the GC. It is transiently detected 2 hours after antigenic stimulation,

providing a boost in the population of antigen-responsive B cells prior to GC

commitment. Accordingly, cMYC ablation prior to immunisation prevents the

development of the GC (73).

cMYC is also detected in a subset of B cells localised to the light zone and is likely

involved in their re-entry into the dark zone. Consequently, GC B cells that have been

engineered to lose cMYC expression after GC formation, display a collapsed GC

response. The purpose of this transient, yet indispensible, bout of cMYC expression is

not clear but may represent a mechanism by which selected B cells are primed for

another wave of proliferative expansion in the dark zone (70).

IRF4

IRF4 is a member of the IRF (interferon regulatory factor) superfamily of transcription

factors that shows relatively weak DNA binding on its own. Therefore, in order to exert

its diverse functions it binds DNA cooperatively with a host of other transcription

factors, including IRF8, PU.1 and SPIB (74, 75). IRF4 plays an essential role in isotype

switching, with IRF4-deficient mice failing to induce AID expression and undergo CSR

when stimulated in vitro (76, 77). IRF4 may regulate AID expression through

cooperative binding with BATF, a transcription factor essential for AID expression(78),

through binding to AP-1-IRF composite element motifs (79).

IRF4 is rapidly induced upon BCR ligation (79, 80) and reported to be required for

BCL6-induction and entry into the GC reaction. However, it is not required for

maintenance of the GC as deletion at a later points does not impair the GC reaction

(76). In addition to establishing the GC reaction, IRF4-deficient mice also fail to make

mature PCs (76, 77). Sciammas et al(77) report that this defect is a result of failure to

induce BLIMP1 expression. However, Klein et al(76) suggest that the failure to induce

PC differentiation is independent of BLIMP1 expression (which they detect at similar

levels in IRF4 deficient B cells and wild type B cells), and instead attribute the

impairment to a loss in XBP1 expression.

The ability of IRF4 to initiate two distinct cell fate transitions, GC B cell and PC

differentiation, stems from its differing expression levels at these times. IRF4 is

expressed at low levels in naïve B cells but is upregulated during PC differentiation

(81). It is thought that the strength of the BCR signaling, as determined by the affinity of

the BCR for antigen, determines the level of IRF4 induction. This, in turn, determines

whether the GC B cell program or the PC differentiation program is initiated: Initial, low

concentrations of IRF4 activate AID and BCL6 expression. As the GC reaction

continues, Ig affinity increases, leading to increased BCR signaling and elevated IRF4

expression, favoring BLIMP1 expression(79, 82), BCL6 repression(83) and

extinguishment of the GC program. These divergent functions of IRF4 are mediated

through its ability to associate with different binding motifs. At lower concentrations

IRF4 cooperates with PU.1 and BATF, facilitating binding to ETS-IRF or AP-1-IRF

composite motifs and coordinating the GC program. At high concentrations, IRF4

favors binding to interferon sequence response elements (ISREs), shifting the cells

expression profile towards the PC program (79).

IRF8

IRF8 is another member of the IRF transcription factor superfamily, but unlike IRF4, is

abundantly expressed in centroblasts (84) and downregulated in centrocytes (85). IRF8

was initially proposed to positively regulate BCL6 and AID; IRF8 overexpression in

human B cells increased the abundance of BCL6 and AID transcripts, while siRNA-

mediated knockdown of IRF8 in a murine GC-derived B cell line had the opposite effect

(84). However, more recently, IRF8-deficient mice have been shown to display only

minor reductions in AID and BCL6 expression and have a normal antibody response

(86). Whilst the phenotype of IRF8-deficient B cells is relatively minor, knockout of both

IRF8 and its common binding partner PU.1, result in heightened PC differentiation and

class switch recombination (81). This mouse model showed that IRF8:PU.1 are

together able to help maintain the B cell program by promoting expression of PAX5 and

BCL6 and concurrently repressing BLIMP1.

BACH2

BACH2 is a basic leucine zipper transcription factor, which is abundantly expressed in

both developing and mature B cells but repressed in PCs. BACH2 expression is

mediated, at least in part, by PAX5 (87, 88) and is further augmented in the GC by

BCL6 (89). One of the main functions of BACH2 appears to be the repression of

BLIMP1, reinforcing the PAX5/BCL6 mediated block on PC differentiation. As such,

BACH2-deficient B cells show inappropriate BLIMP1 expression and subsequent

impairment of AID induction and therefore CSR (90). The inhibition of BLIMP1 is

mediated through BACH2’s interaction with the protein MAFK, facilitating binding of

BACH-2:MAFK to MAFK recognition elements in the BLIMP1 promoter, suppressing its

expression(53, 91). It is likely that high levels of IRF4, resulting from strong BCR

signaling, are required to outcompete the repressive effects of BACH2 expression and

induce BLIMP1 expression(92).

FRA1

FRA1 is an AP1 family member recently identified as playing an essential role in

suppressing premature PC differentiation. Fra1 is strongly induced following B cell

stimulation and Fra1-overexpressing B cells display reduced proliferation, increased

apoptosis and disrupted BLIMP1 upregulation. FRA1 has been shown to directly bind

the BLIMP1 promoter and repress BLIMP1 induction. Accordingly, FRA1-

overexpressing B cells show impaired antibody production in vivo, while FRA1-deficient

B cells secrete higher titres of antigen-specific antibodies (93).

ZBTB20

ZBTB20 is a factor that has recently emerged as being required for PC longevity

following immunisation with alum adjuvant (19, 94). ZBTB20 expression, which is

dependent on high IRF4, peaks in PCs and its overexpression in B cells accelerates

PC differentiation in vitro through augmentation of BLIMP1, IRF4 and XBP1 and

reduction of BCL6. In vivo, although ZBTB20-deficient mice show no obvious

impairment in PC induction a progressive reduction in antigen-specific antibody titers is

seen, suggesting impairment in the maintenance of long-lived antigen-specific PCs

(19). Interestingly, the ZBTB20-dependent survival-defect is overridden when an

immunogen is delivered in conjunction with TLR-activating adjuvants. This suggests

that different adjuvants can activate alternate survival programs in long-lived PCs and

has implications for vaccination strategies (94).

Coordination of GC expression programs

Over the last 10 years it has become increasingly apparent that the different B cell

expression programs, activated as the GC reaction proceeds, are controlled by a highly

coordinated regulatory network. Within this network, multiple points of positive and

negative feedback ensure the mutually antagonistic expression of the “master

regulators”, augmented by an ever-increasing number of “secondary” factors that

reinforce these networks and contribute towards “sensing” the progress of the GC

reaction (Figure 3). Initially, the B cell specific expression pattern is established by

PAX5, which not only regulates the expression of proteins critical to B cell function but

also drives the expression of IRF4 (at a low level), IRF8 and BACH2. Together, these

factors inhibit the expression of the master regulators of PC differentiation, BLIMP1 and

XBP1: PAX5 directly represses XBP1, while IRF8, in combination with PU.1 both

maintains PAX5 and inhibits BLIMP1. BLIMP1 is also actively suppressed by BACH2

and FRA1. Following activation of the B cell via BCR engagement, BCL6 is activated

by IRF4/PU.1. BCL6 not only controls the establishment of the GC fate, initiating the

diversification pathways and rapid proliferation of the B cells, but also further represses

BLIMP1.

Although much has been elucidated as to how these pathways repress B cell

differentiation into PCs, it is less clear how the “switch is flipped” towards favoring

terminal differentiation to PCs, essential for the final success of the GC reaction. As

SHM produces Igs of ever increasing affinity, BCR signal strength increases, in turn

increasing IRF4 expression. Increased IRF4 expression then starts to activate BLIMP1,

which in turn represses BCL6 and PAX5. This switch is further reinforced by the

activation of ZBTB20, which also enhances BLIMP1, IRF4 and XBP1 expression. Once

BLIMP1 accumulates, it represses multiple genes responsible for maintaining B cell

identity, as well as PAX5 and BCL6. This in turn allows the expression of genes

responsible for PC identity, driven in part by IRF4 and ZBTB20. Finally, suppression of

PAX5 relieves repression of XBP1, allowing establishment of the full secretory

program.

Although critical, the circuitry described above appears not to be the whole story. The

rapid proliferation of B cells is a necessary part of the GC response; but it now seems

likely that this process also plays an active role in determining cell fate. It has been

known for many years that a cell’s potential to undergo CSR is determined (at least in

part) by the number of divisions it has undergone (95, 96). Shortly after these findings,

it was shown that a B cell’s potential to undergo differentiation into an antibody

secreting cell was also dependent upon division number(97). Using ground breaking

imaging and tracking of single cell activities it has further been shown that fundamental

aspects of a B cells life (apoptosis, cell division, CSR and differentiation) are all

intimately linked to the number of times the cell has divided(98). Together, these data

suggest that B cells (and possibly all cells) posses some form of “division counting

mechanism”, that changes the cells potential to undertake the processes of cell

division, apoptosis and differentiation (reviewed extensively in Nutt et al., (38)).

Although the molecular mechanism underlying this phenomenon is not known, it likely

plays a major role in regulating the GC activities.

Much is now known about the molecular circuitry regulating the GC response and PC

differentiation, both of which are largely controlled by the expression of a small number

of “master regulators”. However, as yet no deterministic transcription factor for memory

B cells has been found. Given that longevity is a key feature of the memory response,

an alternative view is that memory B cells differentiate stochastically from GC B cells

and that a survival advantage is sufficient for memory B cell differentiation (99, 100)

(reviewed in Kurosaki et al., (39)).

Conclusions

The production of high affinity antibodies of specific isotypes by PCs and generation of

long-lived memory B cells is critical to human health, not only providing protection

against the initial challenge but also shaping the immune system to generate a more

specific response upon reexposure. Conversely, the dysregulation of these processes

underlie many diseases such as autoimmune disease, allergy, lymphoma and immune

dysfunction in aging. Knowledge of these processes have allowed the development of

monoclonal antibodies that are used as diagnostic tools and to treat many diseases

and will continue towards the development of ever more successful immunotherapies.

Although much has been discovered regarding the coordination of the GC response, a

number of fundamental questions remain unanswered: What drives the re-circulation of

B cells from the light zone back into the dark zone, prolonging the GC response; how is

CSR regulated to determine the final isotype of antibody produced, and what cues

drive the cells towards terminal differentiation into memory B cells and PCs? The

unraveling of these mechanisms will no doubt provide valuable insights into the

development of novel vaccine strategies for infectious disease and novel

immunotherapy strategies to treat disease.

Acknowledgments

The authors acknowledge financial support from the Department of Health via the

National Institute for Health Research (NIHR) comprehensive Biomedical Research

Centre award to Guy's & St Thomas' NHS Foundation Trust in partnership with King's

College London and King’s College Hospital NHS Foundation Trust. TR is supported

by a studentship awarded as part of the Medical Research Council & Asthma U.K

Centre in Allergic Mechanisms of Asthma.

References

1. Nieuwenhuis, P., and D. Opstelten. 1984. Functional anatomy of germinal centers.

Am. J. Anat. 170: 421–435.

2. Weyand, C. M., and J. J. Goronzy. 2003. Ectopic germinal center formation in

rheumatoid synovitis. Annals of the New York Academy of Sciences 987: 140–149.

3. Coker, H. A., S. R. Durham, and H. J. Gould. 2003. Local somatic hypermutation

and class switch recombination in the nasal mucosa of allergic rhinitis patients. J.

Immunol. 171: 5602–5610.

4. Takhar, P., L. Smurthwaite, H. A. Coker, D. J. Fear, G. K. Banfield, V. A. Carr, S. R.

Durham, and H. J. Gould. 2005. Allergen drives class switching to IgE in the nasal

mucosa in allergic rhinitis. J. Immunol. 174: 5024–5032.

5. Peled, J. U., F. L. Kuang, M. D. Iglesias-Ussel, S. Roa, S. L. Kalis, M. F. Goodman,

and M. D. Scharff. 2008. The biochemistry of somatic hypermutation. Annu. Rev.

Immunol. 26: 481–511.

6. Stavnezer, J., J. E. Guikema, and C. E. Schrader. 2008. Mechanism and regulation

of class switch recombination. Annu. Rev. Immunol. 26: 261–292.

7. Jung, D., C. Giallourakis, R. Mostoslavsky, and F. W. Alt. 2006. Mechanism and

Control of V(D)J Recombination at the Immunoglobulin Heavy Chain Locus. Annu.

Rev. Immunol.

8. Shaffer, A. L., III, R. M. Young, and L. M. Staudt. 2012. Pathogenesis of Human B

Cell Lymphomas *. Annu. Rev. Immunol. 30: 565–610.

9. Heesters, B. A., R. C. Myers, and M. C. Carroll. 2014. Follicular dendritic cells:

dynamic antigen libraries. Nature reviews. Immunology 14: 495–504.

10. Willard-Mack, C. 2006. Normal Structure, Function, and Histology of Lymph Nodes.

Toxicologic Path. 34: 409–424.

11. De Silva, N. S., and U. Klein. 2015. Dynamics of B cells in germinal centres. Nature

reviews. Immunology 15: 137–148.

12. Pereira, J. P., L. M. Kelly, and J. G. Cyster. 2010. Finding the right niche: B-cell

migration in the early phases of T-dependent antibody responses. International

Immunology 22: 413–419.

13. Link, A., T. K. Vogt, S. Favre, M. R. Britschgi, H. Acha-Orbea, B. Hinz, J. G. Cyster,

and S. A. Luther. 2007. Fibroblastic reticular cells in lymph nodes regulate the

homeostasis of naive T cells. Nature immunology 8: 1255–1265.

14. Batista, F. D., and N. E. Harwood. 2009. The who, how and where of antigen

presentation to B cells. Nature reviews. Immunology 9: 15–27.

15. Junt, T., E. A. Moseman, M. Iannacone, S. Massberg, P. A. Lang, M. Boes, K. Fink,

S. E. Henrickson, D. M. Shayakhmetov, N. C. Di Paolo, N. van Rooijen, T. R. Mempel,

S. P. Whelan, and U. H. von Andrian. 2007. Subcapsular sinus macrophages in lymph

nodes clear lymph-borne viruses and present them to antiviral B cells. nature 450:

110–114.

16. Crawford, A., M. MacLeod, T. Schumacher, L. Corlett, and D. Gray. 2006. Primary

T Cell Expansion and Differentiation In Vivo Requires Antigen Presentation by B Cells.

The Journal of immunology 176: 3498–3506.

17. Reif, K., E. H. Ekland, L. Ohl, H. Nakano, M. Lipp, R. Förster, and J. G. Cyster.

2002. Balanced responsiveness to chemoattractants from adjacent zones determines

B-cell position. nature 416: 94–99.

18. Okada, T., M. J. Miller, I. Parker, M. F. Krummel, M. Neighbors, S. B. Hartley, A.

O'Garra, M. D. Cahalan, and J. G. Cyster. 2005. Antigen-Engaged B Cells Undergo

Chemotaxis toward the T Zone and Form Motile Conjugates with Helper T Cells. PLoS

biology 3: e150.

19. Chevrier, S., D. Emslie, W. Shi, T. Kratina, C. Wellard, A. Karnowski, E. Erikci, G.

K. Smyth, K. Chowdhury, D. Tarlinton, and L. M. Corcoran. 2014. The BTB-ZF

transcription factor Zbtb20 is driven by Irf4 to promote plasma cell differentiation and

longevity. Journal of Experimental Medicine 209: 1241–1253.

20. Ansel, K. M., L. J. McHeyzer-Williams, V. N. Ngo, M. G. McHeyzer-Williams, and J.

G. Cyster. 1999. In vivo-activated CD4 T cells upregulate CXC chemokine receptor 5

and reprogram their response to lymphoid chemokines. The Journal of experimental

medicine 190: 1123–1134.

21. Hardtke, S. 2005. Balanced expression of CXCR5 and CCR7 on follicular T helper

cells determines their transient positioning to lymph node follicles and is essential for

efficient B-cell help. Blood 106: 1924–1931.

22. Qi, H., J. L. Cannons, F. Klauschen, P. L. Schwartzberg, and R. N. Germain. 2008.

SAP-controlled T–B cell interactions underlie germinal centre formation. nature 455:

764–769.

23. Kerfoot, S. M., G. Yaari, J. R. Patel, K. L. Johnson, D. G. Gonzalez, S. H.

Kleinstein, and A. M. Haberman. 2011. Germinal Center B Cell and T Follicular Helper

Cell Development Initiates in the Interfollicular Zone. Immunity 34: 947–960.

24. Jacob, J., and G. Kelsoe. 1992. In situ studies of the primary immune response to

(4-hydroxy-3-nitrophenyl)acetyl. II. A common clonal origin for periarteriolar lymphoid

sheath-associated foci and germinal centers. The Journal of experimental medicine

176: 679–687.

25. Kitano, M., S. Moriyama, Y. Ando, M. Hikida, Y. Mori, T. Kurosaki, and T. Okada.

2011. Bcl6 Protein Expression Shapes Pre-Germinal Center B Cell Dynamics and

FollicularHelper T Cell Heterogeneity. Immunity 34: 961–972.

26. Baumjohann, D., T. Okada, and K. M. Ansel. 2011. Cutting Edge: Distinct Waves of

BCL6 Expression during T Follicular Helper Cell Development. The Journal of

immunology 187: 2089–2092.

27. 2010. Stromal cell contributions to the homeostasis and functionality of the immune

system.

28. Allen, C. D. C., K. M. Ansel, C. Low, R. Lesley, H. Tamamura, N. Fujii, and J. G.

Cyster. 2004. Germinal center dark and light zone organization is mediated by CXCR4

and CXCR5. Nature immunology 5: 943–952.

29. Tarlinton, D. M., and K. G. Smith. 2000. Dissecting affinity maturation: a model

explaining selection of antibody-forming cells and memory B cells in the germinal

centre. Immunol. Today 21: 436–441.

30. Zhang, Y., M. Meyer-Hermann, L. A. George, M. T. Figge, M. Khan, M. Goodall, S.

P. Young, A. Reynolds, F. Falciani, A. Waisman, C. A. Notley, M. R. Ehrenstein, M.

Kosco-Vilbois, and K. M. Toellner. 2013. Germinal center B cells govern their own fate

via antibody feedback. Journal of Experimental Medicine 210: 457–464.

31. Batista, F. D., D. Iber, and M. S. Neuberger. 2001. B cells acquire antigen from

target cells after synapse formation. nature 411: 489–494.

32. Tarlinton, D. M. 2008. Evolution in miniature: selection, survival and distribution of

antigen reactive cells in the germinal centre. Immunology and Cell Biology 86: 133–

138.

33. Victora, G. D., and M. C. Nussenzweig. 2012. Germinal Centers. Annu. Rev.

Immunol. 30: 429–457.

34. Vikstrom, I., and D. M. Tarlinton. 2011. Molecular Immunology. Molecular

Immunology 48: 1301–1306.

35. Peron, S., B. Laffleur, N. Denis-Lagache, J. Cook-Moreau, A. Tinguely, L. Delpy, Y.

Denizot, E. Pinaud, and M. Cogne. 2012. AID-Driven Deletion Causes Immunoglobulin

Heavy Chain Locus Suicide Recombination in B Cells. Science 336: 931–934.

36. Schwickert, T. A., R. L. Lindquist, G. Shakhar, G. Livshits, D. Skokos, M. H. Kosco-

Vilbois, M. L. Dustin, and M. C. Nussenzweig. 2007. In vivo imaging of germinal

centres reveals a dynamic open structure. nature 446: 83–87.

37. Allen, C. D. C., T. Okada, H. L. Tang, and J. G. Cyster. 2007. Imaging of Germinal

Center Selection Events During Affinity Maturation. Science 315: 528–531.

38. Nutt, S. L., P. D. Hodgkin, D. M. Tarlinton, and L. M. Corcoran. 2015. The

generation of antibody-secretingplasma cells. Nature reviews. Immunology 15: 160–

171.

39. Kurosaki, T., K. Kometani, and W. Ise. 2015. Memory B cells. Nature reviews.

Immunology 15: 149–159.

40. Revilla-i-Domingo, R., I. Bilic, B. Vilagos, H. Tagoh, A. Ebert, I. M. Tamir, L.

Smeenk, J. Trupke, A. Sommer, M. Jaritz, and M. Busslinger. 2012. The B-cell identity

factor Pax5 regulates distinct transcriptional programmes in early and late B

lymphopoiesis. The EMBO Journal 31: 3130–3146.

41. Nutt, S. L., P. Urbánek, A. Rolink, and M. Busslinger. 1997. Essential functions of

Pax5 (BSAP) in pro-B cell development: difference between fetal and adult B

lymphopoiesis and reduced V-to-DJ recombination at the IgH locus. Genes &

development 11: 476–491.

42. Horcher, M., A. Souabni, and M. Busslinger. 2001. Pax5/BSAP maintains the

identity of B cells in late B lymphopoiesis. Immunity 14: 779–790.

43. Nutt, S. L., B. Heavey, A. G. Rolink, and M. Busslinger. 1999. Commitment to the

B-lymphoid lineage depends on the transcription factor Pax5. nature 401: 556–562.

44. Fuxa, M., J. Skok, A. Souabni, G. Salvagiotto, E. Roldan, and M. Busslinger. 2004.

Pax5 induces V-to-DJ rearrangements and locus contraction of the immunoglobulin

heavy-chain gene. Genes & development 18: 411–422.

45. Hsu, L. Y. 2004. Pax5 Activates Immunoglobulin Heavy Chain V to DJ

Rearrangement in Transgenic Thymocytes. Journal of Experimental Medicine 199:

825–830.

46. Delogu, A., A. Schebesta, Q. Sun, K. Aschenbrenner, T. Perlot, and M. Busslinger.

2006. Gene Repression by Pax5 in B Cells Is Essential for Blood Cell Homeostasis and

Is Reversed in Plasma Cells. Immunity 24: 269–281.

47. Lin, K. I., C. Angelin-Duclos, T. C. Kuo, and K. Calame. 2002. Blimp-1-Dependent

Repression of Pax-5 Is Required for Differentiation of B Cells to Immunoglobulin M-

Secreting Plasma Cells. Molecular and cellular biology 22: 4771–4780.

48. Nera, K.-P., P. Kohonen, E. Narvi, A. Peippo, L. Mustonen, P. Terho, K. Koskela,

J.-M. Buerstedde, and O. Lassila. 2006. Loss of Pax5 Promotes Plasma Cell

Differentiation. Immunity 24: 283–293.

49. Yasuda, T., F. Hayakawa, S. Kurahashi, K. Sugimoto, Y. Minami, A. Tomita, and T.

Naoe. 2012. B Cell Receptor-ERK1/2 Signal Cancels PAX5-Dependent Repression of

BLIMP1 through PAX5 Phosphorylation: A Mechanism of Antigen-Triggering Plasma

Cell Differentiation. The Journal of immunology 188: 6127–6134.

50. Basso, K., C. Schneider, Q. Shen, A. B. Holmes, M. Setty, C. Leslie, and R. Dalla-

Favera. 2012. BCL6 positively regulates AID and germinal center gene expression via

repression of miR-155. Journal of Experimental Medicine 209: 2455–2465.

51. Basso, K., and R. Dalla-Favera. 2012. Roles of BCL6 in normal and transformed

germinal center B cells. Immunological reviews 247: 172–183.

52. Basso, K., M. Saito, P. Sumazin, A. A. Margolin, K. Wang, W. K. Lim, Y. Kitagawa,

C. Schneider, M. J. Alvarez, A. Califano, and R. Dalla-Favera. 2010. Integrated

biochemical and computational approach identifies BCL6 direct target genes controlling

multiple pathways in normal germinal center B cells. Blood 115: 975–984.

53. Huang, C., H. Geng, I. Boss, L. Wang, and A. Melnick. 2014. Cooperative

transcriptional repression by BCL6 and BACH2 in germinal center B-cell differentiation.

Blood 123: 1012–1020.

54. Huang, C., D. G. Gonzalez, C. M. Cote, Y. Jiang, K. Hatzi, M. Teater, K. Dai, T.

Hla, A. M. Haberman, and A. Melnick. 2014. The BCL6 RD2 Domain Governs

Commitment of Activated B Cells to Form Germinal Centers. CELREP 8: 1497–1508.

55. Cinamon, G., M. Matloubian, M. J. Lesneski, Y. Xu, C. Low, T. Lu, R. L. Proia, and

J. G. Cyster. 2004. Sphingosine 1-phosphate receptor 1 promotes B cell localization in

the splenic marginal zone. Nature immunology 5: 713–720.

56. Tunyaplin, C., A. L. Shaffer, C. D. Angelin-Duclos, X. Yu, L. M. Staudt, and K. L.

Calame. 2004. Direct repression of prdm1 by Bcl-6 inhibits plasmacytic differentiation.

J. Immunol. 173: 1158–1165.

57. Teng, G., P. Hakimpour, P. Landgraf, A. Rice, T. Tuschl, R. Casellas, and F. N.

Papavasiliou. 2008. MicroRNA-155 is a negative regulator of activation-induced

cytidine deaminase. Immunity 28: 621–629.

58. Angelin-Duclos, C., G. Cattoretti, K. I. Lin, and K. Calame. 2000. Commitment of B

Lymphocytes to a Plasma Cell Fate Is Associated with Blimp-1 Expression In Vivo. The

Journal of immunology 165: 5462–5471.

59. Kallies, A. 2004. Plasma Cell Ontogeny Defined by Quantitative Changes in Blimp-

1 Expression. Journal of Experimental Medicine 200: 967–977.

60. Shaffer, A. L., K. I. Lin, T. C. Kuo, X. Yu, E. M. Hurt, A. Rosenwald, J. M. Giltnane,

L. Yang, H. Zhao, K. Calame, and L. M. Staudt. 2002. Blimp-1 orchestrates plasma cell

differentiation by extinguishing the mature B cell gene expression program. Immunity

17: 51–62.

61. Shaffer, A. L., M. Shapiro-Shelef, N. N. Iwakoshi, A.-H. Lee, S.-B. Qian, H. Zhao, X.

Yu, L. Yang, B. K. Tan, A. Rosenwald, E. M. Hurt, E. Petroulakis, N. Sonenberg, J. W.

Yewdell, K. Calame, L. H. Glimcher, and L. M. Staudt. 2004. XBP1, downstream of

Blimp-1, expands the secretory apparatus and other organelles, and increases protein

synthesis in plasma cell differentiation. Immunity 21: 81–93.

62. Kallies, A., J. Hasbold, K. Fairfax, C. Pridans, D. Emslie, B. S. McKenzie, A. M.

Lew, L. M. Corcoran, P. D. Hodgkin, D. M. Tarlinton, and S. L. Nutt. 2007. Initiation of

Plasma-Cell Differentiation Is Independent of the Transcription Factor Blimp-1.

Immunity 26: 555–566.

63. Todd, D. J., L. J. McHeyzer-Williams, C. Kowal, A. H. Lee, B. T. Volpe, B.

Diamond, M. G. McHeyzer-Williams, and L. H. Glimcher. 2009. XBP1 governs late

events in plasma cell differentiation and is not required for antigen-specific memory B

cell development. Journal of Experimental Medicine 206: 2151–2159.

64. Taubenheim, N., D. M. Tarlinton, S. Crawford, L. M. Corcoran, P. D. Hodgkin, and

S. L. Nutt. 2012. High Rate of Antibody Secretion Is not Integral to Plasma Cell

Differentiation as Revealed by XBP-1 Deficiency. The Journal of immunology 189:

3328–3338.

65. Benhamron, S., S. P. Pattanayak, M. Berger, and B. Tirosh. 2014. mTOR

Activation Promotes Plasma Cell Differentiation and Bypasses XBP-1 for

Immunoglobulin Secretion. Molecular and cellular biology 35: 153–166.

66. Pengo, N., M. Scolari, L. Oliva, E. Milan, F. Mainoldi, A. Raimondi, C. Fagioli, A.

Merlini, E. Mariani, E. Pasqualetto, U. Orfanelli, M. Ponzoni, R. Sitia, S. Casola, and S.

Cenci. 2013. Plasma cells require autophagy for sustainable immunoglobulin

production. Nature immunology 14: 298–305.

67. Heise, N., N. S. De Silva, K. Silva, A. Carette, G. Simonetti, M. Pasparakis, and U.

Klein. 2014. Germinal center B cell maintenance and differentiation are controlled by

distinct NF- B transcription factor subunits. Journal of Experimental Medicine 211:

2103–2118.

68. Damdinsuren, B., Y. Zhang, A. Khalil, W. H. Wood III, K. G. Becker, M. J.

Shlomchik, and R. Sen. 2010. Single Round of Antigen Receptor Signaling Programs

Naive B Cells to Receive T Cell Help. Immunity 32: 355–366.

69. Bystry, R. S., V. Aluvihare, K. A. Welch, M. Kallikourdis, and A. G. Betz. 2001. B

cells and professional APCs recruit regulatory T cells via CCL4. Nature immunology 2:

1126–1132.

70. Dominguez-Sola, D., G. D. Victora, C. Y. Ying, R. T. Phan, M. Saito, M. C.

Nussenzweig, and R. Dalla-Favera. 2012. The proto-oncogene MYC is required for

selection in the germinal center and cyclic reentry. Nature immunology 13: 1083–1091.

71. Klein, U., Y. Tu, G. A. Stolovitzky, J. L. Keller, J. Haddad, V. Miljkovic, G. Cattoretti,

A. Califano, and R. Dalla-Favera. 2003. Transcriptional analysis of the B cell germinal

center reaction. Proc. Natl. Acad. Sci. U.S.A. 100: 2639–2644.

72. Phan, R. T., M. Saito, K. Basso, H. Niu, and R. Dalla-Favera. 2005. BCL6 interacts

with the transcription factor Miz-1 to suppress the cyclin-dependent kinase inhibitor p21

and cell cycle arrest in germinal center B cells. Nature immunology 6: 1054–1060.

73. Calado, D. P., Y. Sasaki, S. A. Godinho, A. Pellerin, K. Köchert, B. P. Sleckman, I.

M. de Alborán, M. Janz, S. Rodig, and K. Rajewsky. 2012. The cell-cycle regulator c-

Myc is essential for the formation and maintenance of germinal centers. Nature

immunology 13: 1092–1100.

74. Lu, R. 2003. IRF-4,8 orchestrate the pre-B-to-B transition in lymphocyte

development. Genes & development 17: 1703–1708.

75. Rao, S., A. Matsumura, J. Yoon, and M. C. Simon. 1999. SPI-B activates

transcription via a unique proline, serine, and threonine domain and exhibits DNA

binding affinity differences from PU.1. J. Biol. Chem. 274: 11115–11124.

76. Klein, U., S. Casola, G. Cattoretti, Q. Shen, M. Lia, T. Mo, T. Ludwig, K. Rajewsky,

and R. Dalla-Favera. 2006. Transcription factor IRF4 controls plasma cell differentiation

and class-switch recombination. Nature immunology 7: 773–782.

77. Sciammas, R., A. L. Shaffer, J. H. Schatz, H. Zhao, L. M. Staudt, and H. Singh.

2006. Graded Expression of Interferon Regulatory Factor-4 Coordinates Isotype

Switching with Plasma Cell Differentiation. Immunity 25: 225–236.

78. Ise, W., M. Kohyama, B. U. Schraml, T. Zhang, B. Schwer, U. Basu, F. W. Alt, J.

Tang, E. M. Oltz, T. L. Murphy, and K. M. Murphy. 2011. The transcription factor BATF

controls the global regulators of class-switch recombination in both B cells and T cells.

Nature immunology 12: 536–543.

79. Ochiai, K., M. Maienschein-Cline, G. Simonetti, J. Chen, R. Rosenthal, R. Brink, A.

S. Chong, U. Klein, A. R. Dinner, H. Singh, and R. Sciammas. 2013. Transcriptional

Regulation of Germinal Center B and Plasma Cell Fates by Dynamical Control of IRF4.

Immunity 38: 918–929.

80. Matsuyama, T., A. Grossman, H. W. Mittrücker, D. P. Siderovski, F. Kiefer, T.

Kawakami, C. D. Richardson, T. Taniguchi, S. K. Yoshinaga, and T. W. Mak. 1995.

Molecular cloning of LSIRF, a lymphoid-specific member of the interferon regulatory

factor family that binds the interferon-stimulated response element (ISRE). Nucleic

acids research 23: 2127–2136.

81. Carotta, S., S. N. Willis, J. Hasbold, M. Inouye, S. H. M. Pang, D. Emslie, A. Light,

M. Chopin, W. Shi, H. Wang, H. C. Morse, D. M. Tarlinton, L. M. Corcoran, P. D.

Hodgkin, and S. L. Nutt. 2014. Transcription Factor IRF4 Regulates Germinal Center

Cell Formation through a B Cell-Intrinsic Mechanism. The Journal of immunology 192:

3200–3206.

82. Sciammas, R., Y. Li, A. Warmflash, Y. Song, A. R. Dinner, and H. Singh. 2011. An

incoherent regulatory network architecture that orchestrates B cell diversification in

response to antigen signaling. Molecular Systems Biology 7: 495–495.

83. Saito, M., J. Gao, K. Basso, Y. Kitagawa, P. M. Smith, G. Bhagat, A. Pernis, L.

Pasqualucci, and R. Dalla-Favera. 2007. A Signaling Pathway Mediating

Downregulation of BCL6 in Germinal Center B Cells Is Blocked by BCL6 Gene

Alterations in B Cell Lymphoma. Cancer Cell 12: 280–292.

84. Lee, C. H., M. Melchers, H. Wang, T. A. Torrey, R. Slota, C.-F. Qi, J. Y. Kim, P.

Lugar, H. J. Kong, L. Farrington, B. van der Zouwen, J. X. Zhou, V. Lougaris, P. E.

Lipsky, A. C. Grammer, and H. C. Morse. 2006. Regulation of the germinal center gene

program by interferon (IFN) regulatory factor 8/IFN consensus sequence-binding

protein. The Journal of experimental medicine 203: 63–72.

85. Cattoretti, G., R. Shaknovich, P. M. Smith, H. M. Jack, V. V. Murty, and B. Alobeid.

2006. Stages of Germinal Center Transit Are Defined by B Cell Transcription Factor

Coexpression and Relative Abundance. The Journal of immunology 177: 6930–6939.

86. Feng, J., H. Wang, D. M. Shin, M. Masiuk, C. F. Qi, and H. C. Morse. 2011. IFN

Regulatory Factor 8 Restricts the Size of the Marginal Zone and Follicular B Cell Pools.

The Journal of immunology 186: 1458–1466.

87. Schebesta, A., S. McManus, G. Salvagiotto, A. Delogu, G. A. Busslinger, and M.

Busslinger. 2007. Transcription Factor Pax5 Activates the Chromatin of Key Genes

Involved in B Cell Signaling, Adhesion, Migration, and Immune Function. Immunity 27:

49–63.

88. McManus, S., A. Ebert, G. Salvagiotto, J. Medvedovic, Q. Sun, I. Tamir, M. Jaritz,

H. Tagoh, and M. Busslinger. 2011. The transcription factor Pax5 regulates its target

genes by recruiting chromatin-modifying proteins in committed B cells. The EMBO

Journal 30: 2388–2404.

89. Alinikula, J., K.-P. Nera, S. Junttila, and O. Lassila. 2011. Alternate pathways for

Bcl6-mediated regulation of B cell to plasma cell differentiation. European journal of

immunology 41: 2404–2413.

90. Muto, A., K. Ochiai, Y. Kimura, A. Itoh-Nakadai, K. L. Calame, D. Ikebe, S. Tashiro,

and K. Igarashi. 2010. Bach2 represses plasma cell gene regulatory network in B cells

to promote antibody class switch. The EMBO Journal 29: 4048–4061.

91. Oyake, T., K. Itoh, H. Motohashi, N. Hayashi, H. Hoshino, M. Nishizawa, M.

Yamamoto, and K. Igarashi. 1996. Bach proteins belong to a novel family of BTB-basic

leucine zipper transcription factors that interact with MafK and regulate transcription

through the NF-E2 site. Molecular and Cellular Biology 16: 6083–6095.

92. Igarashi, K., K. Ochiai, A. Itoh-Nakadai, and A. Muto. 2014. Orchestration of

plasma cell differentiation by Bach2 and its gene regulatory network. Immunological

reviews 261: 116–125.

93. Grotsch, B., S. Brachs, C. Lang, J. Luther, A. Derer, U. Schlotzer-Schrehardt, A.

Bozec, S. Fillatreau, I. Berberich, E. Hobeika, M. Reth, E. F. Wagner, G. Schett, D.

Mielenz, and J. P. David. 2014. The AP-1 transcription factor Fra1 inhibits follicular B

cell differentiation into plasma cells. Journal of Experimental Medicine 211: 2199–2212.

94. Wang, Y., and D. Bhattacharya. 2014. Adjuvant-specific regulation of long-term

antibody responses by ZBTB20. Journal of Experimental Medicine 211: 841–856.

95. Hasbold, J., A. B. Lyons, M. R. Kehry, and P. D. Hodgkin. 1998. Cell division

number regulates IgG1 and IgE switching of B cells following stimulation by CD40

ligand and IL-4. European journal of immunology 28: 1040–1051.

96. Tangye, S. G., D. T. Avery, and P. D. Hodgkin. 2003. A division-linked mechanism

for the rapid generation of Ig-secreting cells from human memory B cells. J. Immunol.

170: 261–269.

97. Hasbold, J., L. M. Corcoran, D. M. Tarlinton, S. G. Tangye, and P. D. Hodgkin.

2003. Evidence from the generation of immunoglobulin G–secreting cells that

stochastic mechanisms regulate lymphocyte differentiation. Nature immunology 5: 55–

63.

98. Duffy, K. R., C. J. Wellard, J. F. Markham, J. H. S. Zhou, R. Holmberg, E. D.

Hawkins, J. Hasbold, M. R. Dowling, and P. D. Hodgkin. 2012. Activation-Induced B

Cell Fates Are Selected by Intracellular Stochastic Competition. Science 335: 338–341.

99. Fischer, S. F., P. Bouillet, K. O'Donnell, A. Light, D. M. Tarlinton, and A. Strasser.

2007. Proapoptotic BH3-only protein Bim is essential for developmentally programmed

death of germinal center-derived memory B cells and antibody-forming cells. Blood

110: 3978–3984.

100. Clybouw, C., S. Fischer, M. T. Auffredou, P. Hugues, C. Alexia, P. Bouillet, M.

Raphael, G. Leca, A. Strasser, D. M. Tarlinton, and A. Vazquez. 2011. Regulation of

memory B-cell survival by the BH3-only protein Puma. Blood 118: 4120–4128.

Figure legends

Figure 1. Germinal center structure, formation and B cell movement.

Panel A shows the anatomical structure of the lymph node prior to commencement of

an immune response. In addition, the location of the resident cell populations and

movement of cells/immunogens are indicated. Naïve T cells (light yellow) move into the

T cell zone (paracortex) from the circulation, where resident DCs (light green) collect

and display antigen (purple triangle). Circulating naïve B cells move into B cell follicles,

located within the interfollicular region (cortex), where they sample antigen displayed

by FDCs (dark green). The location of sub-capsular macrophages (purple) is also

shown.

Panel B shows the cell interactions and movements associated with the early stages of

the immune response. Initially (day 1 of the response) naïve T cells are primed (dark

orange cell) following recognition of their cognate antigens, presented by DCs in the T

cell cortex (light pink). The T cells then move to the interfollicular regions (dark pink),

where they mature into TFH cells. Similarly naïve B cells are activated (dark blue cell) by

their cognate antigen displayed on the surface of FDCs and move out of the follicle into

the interfollicular region. By day 2 of the response antigen-primed B cells find their

cognate TFH cells to form stable interactions and become fully activated.

Panel C shows the early stages of GC formation. Cognate B/T cells move back into the

B cell follicle where B cells acquire a GC fate, undergoing rounds of rapid proliferation.

This B cell clonal expansion starts to form the GC. By day 4-5 some activated B cells

move into the sub-capsular sinus (SCS – light green) and differentiate into short-lived

plasmablasts.

Panel D. The final architecture, cellular composition and cell movement within the

mature GC is shown with the dark zone composed of rapidly proliferating centroblasts

and light zone containing centrocytess undergoing affinity maturation by selection with

TFH and FDCs. Here, non-selected cells undergo apoptosis, while some cells move out

of the GC to differentiate into long-lived PCs.

Figure 2. Regulators of GC activities and B cell fate.

Figure 2 shows the expression of the critical transcription factors that control B cell fate

at different stages of B cell maturity, from naïve B cells (light blue), through activated

GC B cells (dark blue) to plasmablasts and plasma cells. At the transition between

each stage (indicated by thick arrows) the critical change in transcription factor

expression is shown (up/downregulation of factors is indicated by small arrows). In

addition the receptors responsible for B cell movement/localisation are also shown.

Figure 3. Regulatory network controlling the GC response.

The regulatory network that coordinates the GC response is illustrated at the three

main stages of B cell differentiation, from naïve B cell, through to activated GC B cell

and finally mature plasma cell. The “master regulators” expressed in each cell type are

shown in blue boxes whilst their critical target genes/pathways is given below. The

“secondary factors” that augment the master regulators are shown above. The

regulatory interactions that exist between each of the transcription factors are depicted

by either arrows (stimulatory) or flat-headed arrows (inhibitory). Each transcription

factor and its corresponding interactions is colour coded. The activation of XBP1

brought about by the relief of PAX5 repression is represented by a dashed line.