Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

nature reviews immunology https://doi.org/10.

1038/s41577-023-00897-3

Review article Check for updates

Memory B cells
Takeshi Inoue 1
& Tomohiro Kurosaki 1,2,3,4

Abstract Sections

Recent advances in studies of immune memory in mice and humans Introduction

have reinforced the concept that memory B cells play a critical role Memory B cells in protection
in protection against repeated infections, particularly from variant and pathology

viruses. Hence, insights into the development of high-quality memory Generation of memory B cells
B cells that can generate broadly neutralizing antibodies that bind Phenotypic heterogeneity
such variants are key for successful vaccine development. Here, we of memory B cells

review the cellular and molecular mechanisms by which memory B cells Reactivation of memory B cells
are generated and how these processes shape the antibody diversity Lessons from SARS-CoV-2
and breadth of memory B cells. Then, we discuss the mechanisms
Memory B cells, TFH cells
of memory B cell reactivation in the context of established immune and vaccines
memory; the contribution of antibody feedback to this process has Outlook
now begun to be reappreciated.

1
Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka,
Japan. 2Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan. 3Center for Infectious Disease
Education and Research, Osaka University, Osaka, Japan. 4Laboratory for Lymphocyte Differentiation, RIKEN
Center for Integrative Medical Sciences (IMS), Kanagawa, Japan. e-mail: kurosaki@ifrec.osaka-u.ac.jp

Nature Reviews Immunology


Review article

Introduction strain11. In addition, a recent study analysed individuals immunized


There is good evidence that the overall number of human memory with the 2018–2019 influenza seasonal vaccine12 and found that mem-
B cells (defined as canonical class-switched CD27+ B cells) is larger than ory B cells, formed in response to a previously encountered different
that of naive B cells in peripheral blood, particularly in aged individu- influenza strain, contained clones that were cross-reactive with the
als1,2. This, together with their remarkable stability (at least decades)3,4 new 2018–2019 strain. Upon vaccination with the 2018–2019 influ-
and their heightened in vitro activation kinetics compared with naive enza vaccine, some of these cross-reactive memory B cells became
cells5, has evoked the idea that memory B cells are the more direct pre- plasmablasts, whereas others entered GCs (Fig. 1a). Similarly, in the
cursors for plasma cells that exert both antibody-mediated protection mouse intestinal immune system, pre-existing cross-reactive IgA+
and pathology than naive B cells. Thus, it is important to identify the memory B cells appear to re-enter the GC in response to changes in the
unique properties of memory B cells and clarify how such properties microbiota, thereby reshaping their B cell receptors (BCRs) through
are acquired6. somatic mutations13,14 (Fig. 1b). Therefore, it is reasonable to conclude
Advances have been made in tracking antigen-experienced that pre-existing memory B cell clones can deal with relatively minor
memory B cells, and it has become apparent that at least two distinct structural changes, for instance in the influenza haemagglutinin (HA)
classes of memory B cells coexist — for example, germinal centre (GC)- stem region, by somatic mutations9, but not with major changes, for
dependent and GC-independent. Importantly, based on the results of instance strain-specific changes in the influenza HA head region15
adoptive transfer experiments, these subsets were found to possess (Fig. 1a). In the latter case, new naive B cell clones are recruited and
considerable intrinsic functional differences. In reviews published participate in the response, a phenomenon that has been observed in
in 2013 and 2015 (refs. 5,7), we and others summarized progress in humans as well as in mouse studies12.
the field and emphasized the importance of the interaction between Historically, studies of memory B cells have been carried out in the
memory T follicular helper cells (TFH cells) and memory B cells in recall context of infection or immunization. In autoimmune settings, there is
responses. In the approximately 10 years since then, our understanding also increasing evidence that memory B cells are the direct precursors
of memory B cells has progressed significantly, with great advances in of plasma cells that produce pathogenic antibodies.
their in vivo characterization and new insights into the mechanisms by
which they develop diversity, and the molecular details of their genera- Generation of memory B cells
tion and reactivation. This fundamental knowledge has mainly relied It has generally been assumed that the memory B cell compartment in
on mouse experimental systems and focused on IgG+ memory B cells; mice is exclusively derived from conventional B2 B cells and not from
but now, data from human studies have also been forthcoming. Here, B1 B cells, given that the remaining small subpopulation of B1 B cells is
by integrating such human data, we summarize the advances over the unable to proliferate in response to BCR-mediated stimulation in vitro16.
past decade in our understanding of memory B cell generation and However, recent data demonstrate that some B1 B cells also proliferate
reactivation. In this Review, unless otherwise indicated, we focus on in an antigen-dependent manner in vivo and can form a memory pool,
mouse studies. likely in a T cell-independent manner17. This conclusion was further
substantiated by ‘time-stamp’ lineage tracing experiments showing
Memory B cells in protection and pathology that B1 B cells contribute to intestinal IgA+ memory B cells that are
The memory compartment of the humoral immune system is composed specific for commensal microbiota14. Hence, clarifying how B1 B cells
of long-lived plasma cells (LLPCs) and memory B cells (Fig. 1a, left). form memory B cell and LLPC pools is one of the important unresolved
The functional dissection of these two compartments has allowed to issues in B cell biology. This is particularly relevant given that mouse
decipher the roles of these cells in protection from reinfection with B1-derived antibodies, such as anti-α-1,3-glucan IgA antibodies that
a particular pathogen. For example, in a mouse model of West Nile bind cockroach antigens, have been shown to have a protective role
virus infection, pre-existing protective antibodies secreted by LLPCs in allergic reactions18,19.
were shown to act as a first line of defence against reinfection with the We focus on the development of memory B cells from conven-
homologous virus, whereas memory B cells encoded antibodies that tional B2 B cells, which is dependent on interactions with T cells.
were capable of binding viral variants8. Consistent with this observa- Here, fate decisions of activated naive B cells take place at two distinct
tion, two recent lines of in vivo evidence from mouse models show that stages: at the interface between the B cell follicle and the T cell zone
memory B cells indeed act as defence against re-challenge with variants (T–B border; the pre-GC state), recently activated B cells either
of influenza virus and flavivirus9,10 and that pre-existing cross-reactive differentiate into plasma cells, enter the GC to become CG B cells
clones contained in the memory B cell, but not in the LLPC compart- or differentiate into GC-independent memory B cells20,21; and after
ment, are positively selected and directly differentiate into plasma the T cell–B cell interactions at the T–B border, within the GC itself, GC
cells after re-challenge with a variant virus (Fig. 1a, right). As both LLPC B cells continue the light zone–dark zone re-entry cycle or interrupt
and memory B cell compartments undergo somatic hypermutation this cycle to exit the GC, again differentiating into either plasma cells
(SHM) during the primary infection, these data suggest differences in or GC-derived memory B cells20 (Fig. 2). As isotype switching initiates
the selection mechanism between these two memory compartments early at the pre-GC stage22–24, switched IgG+ and IgA+ memory B cells
during GC responses. are also generated during the pre-GC stage, although the majority of
The importance of memory B cells in protection against variant pre-GC-derived memory B cells are IgM+.
virus infection has also been well substantiated in vaccination stud-
ies in humans. In the case of vaccination against the pandemic 2009 Interactions between TFH cells and B cells
H1N1 influenza strain, individuals who have pre-existing cross-reactive TFH cells are a specialized subset of CD4+ T cells that are required for the
antibodies (even at very low levels) were shown to generate antibodies activation and maintenance of pre-GC and GC B cells25. Importantly,
from the memory B cell compartment that were cross-reactive with and in contrast to non-TFH effector cells such as T helper 1 (TH1) cells,
both previously encountered viruses and the novel 2009 influenza TH2 cells or TH17 cells, which are destined to leave lymphoid tissues and

Nature Reviews Immunology


Review article

a
Memory B cell
(strain-specific)

Memory B cell
(cross-reactive)
Virus

Primary virus infection Variant virus reinfection


Variant
BCR Memory B cell virus
(strain-specific)
Memory B cell
GC B cell (cross-reactive, mature)
Naïve Light Memory B cell
B cell zone (cross-reactive)

Dark
zone Long-lived
plasma cell Antibody
Germinal centre

b Secretory IgA

GC B cell

GC

IgA+ memory B cell Memory B cell


(cross-reactive) (cross-reactive, mature)

Fig. 1 | GC dynamics and memory B cell selection. a, The humoral memory B cells against haemagglutinin (HA) antigen. Here, B cells can either bind the
compartment is composed of memory B cells and long-lived plasma cells immunodominant strain-specific epitopes to become strain-specific memory
(LLPCs). After primary virus infection, antigen-specific naive B cells enter B cells or the subdominant cross-reactive epitopes to become cross-reactive
the germinal centre (GC) reaction and differentiate into strain-specific memory B cells. b, In the mouse gut immune system, pre-existing IgA+ memory
or cross-reactive memory B cells and LLPCs. Upon variant virus infection, B cells can re-enter the GC reaction in response to a change in the composition
the cross-reactive memory B cells are selected to differentiate into plasma of the microbiota and accumulate somatic hypermutations (SHMs) over time
cells or recruited to the GC reaction for further affinity maturation. The figure to generate cross-reactive memory B cells. BCR, B cell receptor.
illustrates how influenza virus infection induces the generation of memory

traffic to sites of infection, TFH cells remain resident in lymph nodes and the presence of TFH cell-inductive signals such as IL-6 they upregulate
the spleen25 and their primary purpose is to help B cells. BCL-6, a key transcription factor for TFH cell identity28,29. This facili-
In response to immunization with virus-like particles, B cells can tates the maintenance and upregulation of the chemokine receptor
act as the dominant first ‘professional’ antigen-presenting cells to CXCR5 and repression of CCR7, allowing their migration to the T–B
initiate naive CD4+ T cell activation26. Similarly, memory TFH cells do border where they differentiate into non-GC TFH cells in response to
not require dendritic cells for their activation; instead, the antigen- T cell–B cell interactions (Fig. 2a). A key cytokine produced by TFH
presenting function of B cells is sufficient27. However, during a standard cells is IL-21, which promotes GC B cell proliferation, maintenance
protein-immunization regimen of naive mice, TFH cell differentia- and the generation of plasma cells30,31. The current hypothesis is that
tion begins with the interaction of naive CD4+ T cells with antigen- this basic TFH cell programme (BCL-6+IL-21+) can be modified by the
presenting dendritic cells in the T cell zone (‘pre-TFH cells’), where in cytokine milieu towards TFH1 cells (these produce IFNγ/IL-21, which is

Nature Reviews Immunology


Review article

a
Extrafollicular/pre-GC process GC process b

‘Trifurcation’ ‘Stepwise’

Plasma cell Plasma cell


Antibody

B cell follicle
Non-GC Non-GC
TFH cell Cytokine GC-dependent TFH cell
receptor memory B cell GC-independent
Cytokine
memory
GC-independent GC B cell GC TFH cell B cell
memory
B cell

FDC
CD40 CD40L Precursor for GC B cell
TCR MHC II GC/memory
BCR B cell
Light
zone
Pre-TFH cell B cell Dark
zone
Antigen

Naïve
B cell

T cell
CD25+ T cell zone
dendritic cell

Naïve
CD4+ cell

Fig. 2 | Generation of GC-independent and GC-dependent memory B cells. the affinity maturation of GC B cells by providing cytokines, such as IL-4 and IL-21.
a, Simultaneous trifurcation model. b, Stepwise differentiation model. Naive CD4+ In the simultaneous trifurcation model (panel a), non-GC TFH cells induce antigen-
T cells interact with antigen-presenting dendritic cells. These activated T cells activated B cells to differentiate into plasma cells, GC B cells or GC-independent
are thought to interact with the CD25+ dendritic cell subset before differentiation memory B cells simultaneously, whereas in a stepwise differentiation model
into pre-T follicular helper (pre-TFH) cells143 and further interacting with antigen- (panel b), plasma cell induction precedes the generation of a precursor for GC
engaged B cells to form TFH cells. In contrast to the highly proliferative non- B cells and GC-independent memory B cells. BCR, B cell receptor; FDC, follicular
germinal centre (non-GC) TFH cells, GC TFH cells are specialized to contribute to dendritic cell; TCR, T cell receptor.

important in immune responses against intracellular pathogens such border likely promotes the differentiation of non-GC TFH cells, eventu-
as viruses and promotes class switching in mice to IgG2a/c), TFH2 cells ally contributing to GC formation and maintenance (Fig. 2). Recent
(these produce IL-4/IL-21, which is important in immune responses detailed analysis of TFH cells in mice identified subsets of non-GC
against helminths and promotes class switching to IgG1 and IgE) and TFH cells (CD90hiS1PR2−) and GC TFH cells (CD90lowS1PR2+)36. The dif-
TFH17 cells (these produce IL-17/IL-21, which is important in immune ferentiation of non-GC TFH cells into GC TFH cells has been thought to
responses to extracellular pathogens and promotes class switching require sustained cognate interactions with GC B cells, and GC TFH
to IgA)32. However, a definitive role for TFH17 cells in the induction cells were found to be required for the efficient generation of GC
of IgA in the mouse gut immune system has not yet been demon- B cells37. Clearly, it will be important to clarify how these non-GC or GC
strated33. This model is largely based on results in mice. Circulating TFH cells become memory-type T cells that can be reactivated during
TFH cells from human blood can similarly be classified as circulating a secondary infection.
TFH1 cells (CXCR3hiCCR6low), circulating TFH2 cells (CXCR3lowCCR6low) The clonal level of TFH cell selection is thought to take place dur-
and circulating TFH17 cells (CXCR3lowCCR6hi). ing the GC reaction. In fact, there is evidence that TFH cells divide in
In order to interact with T cells at the T–B border, antigen- response to a peptide antigen to an extent proportional to their TCR
engaged B cells in the follicular zone upregulate CCR7 and EBI2, affinity, leading to the increased representation of high-affinity GC TFH
which act together to facilitate trafficking to the T–B border, where cells over time38. Thus, it is possible that such affinity-selected T cells
the B cells then interact with pre-TFH cells in an ICOS–ICOSL and with increased sensitivity to peptide–MHC during the GC reaction
peptide–MHC-dependent manner34,35. An additional signal such are likely to prolong the immune responses even in the presence of
as from ICOS–ICOSL interaction from cognate B cells at the T–B decreased amounts of antigen.

Nature Reviews Immunology


Review article

Pre-GC memory B cells during pre-GC responses, although less robustly44, qualitative and
With regards to activated B cell fates at the pre-GC selection phase, an quantitative changes in pre-GC memory B cells are likely to affect
elegant study tracing the fates of single antigen-specific naive B cells secondary immune responses.
with differing affinity for antigen provided significant insights39. After
stimulation with the protein antigen allophycocyanin with complete Diversity of GC B cells
Freund’s adjuvant, half of the B cells produced only one type of effector As many excellent reviews about the dynamics of GC responses have
cell among three possible fates (plasma cells, GC B cells and memory been published recently44–46, here, based on the recently emerged con-
B cells) (Fig. 2), suggesting, at least to some extent, a role for BCR affinity cept that GC-dependent memory B cells are more diversified relative to
in the subsequent T cell help-mediated selection of B cells. The general LLPCs10,47,48, we rather focus on how diversified GC B cells are generated
rule seems to be that higher or lower affinity predisposes the activated and differentiate into the memory B cell pool.
B cells to differentiate into plasma cells or memory B cells, respectively; Historically, the aim of studies of GCs has been to understand
positively selected and probably intermediate affinity B cells are more how they support maturation of B cells towards producing the highest
likely to enter the GC. This is consistent with a previous study showing affinity antibodies. The increase in the average affinity of GC B cells
that a decrease in BCR affinity blocks the generation of plasma cells but over time requires that B cells with higher affinity for antigen clonally
leaves intact the differentiation into GC B cells40. BCR affinity affects the expand, whereas those of lower affinity proportionally contract46. Such
strength of T cell help as B cells with higher affinity endocytose more positive selection of higher-affinity GC B cells has been thought to
antigen and present it to cognate T cells more efficiently. In this regard, take place through two (not necessarily mutually exclusive) potential
a report showing that T cell help is necessary to generate the immedi- mechanisms. One is that higher-affinity cells might receive stronger
ate precursors for GC cells but must cease in order to allow for GC BCR signals that favour their survival compared with lower-affinity
B cell progression41 supports the idea that there is an optimal window cells. The second is that higher-affinity cells could be more effective
of BCR affinity (presumably intermediate affinity) in the TFH cell–B cell in subsequently engaging GC TFH cells and receiving stronger survival
interaction that allows for GC differentiation. and proliferation signals through cognate T cell help. Although it is
However, given that microenvironmental states such as antigen hard to dissect the effects of the two signals (BCR versus T cell help
accessibility undergo dynamic spatiotemporal alterations, particularly driven), various studies suggest that the integration of these two signals
early in the immune response, other factors besides affinity should determines the positive selection in GCs45,49. In the case of T cell help
be considered in the determination of fate decisions. For instance, in for GC B cells, two types of signals, mediated by CD40L–CD40 and
the single-cell adoptive transfer experiments described above, about cytokine–cytokine receptor interactions, have been well appreciated
5–10% of clones gave rise to all three effector cell types39. Such multi- and both receptor/ligand interactions were previously thought to act
plicity of fates correlates well with a large clonal burst size. The high on a cognate (1:1) interaction basis. However, recent experiments show
bursting clones can be evoked by their high BCR affinity and/or by a that T cell-derived IL-21 may also act in a paracrine manner and function
B cell-extrinsic highly inflammatory milieu, where bystander TFH cells as a general GC growth and maintenance factor, rather than only as a
provide survival signals regardless of their BCR affinity. Hence, it is pos- cognate regulator50. In vitro imaging data also suggest this possibility
sible that even a high-affinity B cell clone can give rise to the memory for IL-4 (ref. 51). However, the caveat in these observations is that such
fate and that, on the other hand, even low-affinity B cells can join in the paracrine activity may only take place in an experimental environment
GC reaction, which can explain the fact that the unmutated ancestors where many TFH cells and GC B cells do not express the IL-21 receptor
of matured mutated broadly neutralizing BCRs to influenza virus or or may only occur in vitro.
HIVs can enter the GC, even though they show limited or, apparently, In addition to promoting the maturation of high-affinity anti-
even no antigen binding to the respective viruses42. bodies, emerging evidence has indicated that GC responses also sup-
Other types of experiments that involve the adoptive transfer port the development of a diverse population of antigen-specific GC
of naive B cells with fixed BCR affinity (high affinity for hapten NP; B cells52–54. The generation of diverse antigen-specific GC B cells is
B1-8hi) into recipient mice have provided new insights into fate deci- thought to be promoted in the following, at least, five ways45. First,
sion mechanisms at the pre-GC stage43 (Fig. 2b). Activated B cells as GC B cells cannot traffic between separate ‘GC islands’, clones are
were shown to initially give rise to plasma cells, and then many acti- confined to individual GCs and clonal evolution takes place in each
vated B cells exited the cell cycle, giving rise to precursors for GC and independent GC; these clones are not competed out by a stronger
memory B cells. This cell cycle exit is caused by limited antigen avail- clone residing in a neighbouring GC45,52. Second, some GCs undergo
ability. Hence, in such settings, the provision of antigen for a sufficient clonal bursting and are taken over by one or a few clones, whereas
amount of time, evoking high BCR signalling and subsequent high other GCs in the same lymphoid tissue do not undergo a clonal burst,
T cell help, facilitates plasma cell differentiation, whereas exposure thereby creating a diverse group of surviving clones52. Third, GC
to antigen for a relatively limited period causes the B cells to become B cell diversity may be promoted by antibody-mediated feedback, as
precursors for GC and memory B cells (GC/memory precursor). In dominant GC B cell clones with BCRs specific for particular antigen
this regard, rather than a simultaneous trifurcation model (Fig. 2a), epitopes give rise to plasma cells that secrete antibody which, in turn,
the current data appear to fit better with a stepwise model (Fig. 2b) masks these epitopes, thereby halting evolution of the correspond-
in which GC/memory precursors are generated before bifurcation of ing clone, and instead enhancing the selection of clones that bind
memory and GC B cells. Taken together, at least three determinants different epitopes55–58. Fourth, studies have shown that some naive
participate in fate decisions at the pre-GC phase in wild-type settings: B cells enter the GC at later stages during immune responses and that
affinity for antigen; duration of interaction with antigen and cognate these late participants possess very low affinity for the original immu-
T cells; and the provision of survival signals by the microenvironment. nogen54,59,60. Last, it is now clear that some B cells in the naive reper-
These factors are interrelated and further work is required to dissect toire have measurable autoreactivity (they are generally anergic) and
their individual and joint contributions. Given that SHM occurs even are able to enter the GC61,62. These GC B cells are initially selected for

Nature Reviews Immunology


Review article

a Affinity Mechanisms that promote diversity are particularly important


for allowing sufficient breadth in the B cell response to protect from
rapidly evolving pathogens such as HIV and influenza. Hence, this issue
Cytokine Cytokine requires further investigation.
receptor
MHC II
BCR TCR GC-dependent memory B cells
GC Antibody With regards to the mechanism of memory B cell generation in the GCs,
TFH cell
B cell and by combining older and current data, we have previously proposed
Memory
CD40 CD40L the affinity instruction model. In this model, a low-affinity BCR, which
B cell
means that the B cell will subsequently receive no or low T cell help
upon encountering antigen, is the primary determinant for memory
Plasma cell
B cell generation47,48,66–68. At any given time, GC B cells with no or low
Light affinity for antigen have generally been thought to undergo apoptosis.
zone However, those that do not die exit the GC as memory B cells, whereas
Dark
zone B cells with high affinity re-enter the dark zone for further proliferation
and SHM or exit the GC as plasmablasts (Fig. 3a). This model is further
supported by recent experiments in mice in which the availability of IL-4
and IL-21 in GCs was manipulated50,69. Follicular dendritic cells (FDCs)
b express the IL-4 receptor, which acts similar to a decoy, restricting IL-4
availability to GC B cells and augmenting memory B cell generation.
Conversely, increased IL-21 bioavailability reduces memory B cell gen-
eration. As an alternative to this affinity instruction model, Shlomchik
and colleagues proposed a temporal switch model, in which pre-GC and
early GC B cells tend to differentiate into memory B cells, whereas the
LLPCs are generated preferentially from the late GC reaction70. Given
Memory Memory GC B cell that memory B cells progressively accumulate SHMs with time, these
B cell precursor
two models are not mutually incompatible with each other46.
Bcl6low Bcl6int Bcl6hi
Recent detailed analyses using native proteins with multiple
Bcl2 Bcl2 epitopes as antigens have provided important insights into the memory
Hhex Survival
BCR BCR B cell selection mechanism. First, the selection of low-affinity B cells
EBI2 into the memory compartment takes place both in the late GC phase
Bach2hi
S1pr1 as well as in the early GC phase. Second, the germ-line BCR affinity of
c-Myc
Anti-proliferative
mTORC1 memory B cells is lower than that of LLPCs10,47. Finally, with regards to
clonal diversity, memory B cells have turned out to be more diverse than
Fig. 3 | Memory B cell generation from GCs. a, The affinity instruction model
suggests that memory B cell selection is primarily determined by the affinity of
LLPCs. The more diverse memory B cell population is, thus, most likely to
B cell receptors (BCRs) of germinal centre (GC) B cells for antigen. GC B cells with contain the usually non-immunodominant clones that cross-react with
low-affinity BCRs are more likely to enter memory B cell fate, whereas those different viral variants, such as those specific for the influenza HA stem
with high affinity re-enter the dark zone or exit the GC as plasma cells. b, Memory in the case of influenza-specific B cells, a prediction that has been veri-
B cell precursors are characterized by high BACH2 expression, which can fied experimentally8–10 (Fig. 1a, upper left). Collectively, the properties
mediate an anti-proliferative effect by restraining MYC and mTORC1 activation. of GC-dependent memory B cells as described above appear to fit well
These cells also start to downregulate BCL-6, which enables the induction of with the concept that one of the important functions of the memory B cell
BCL-2 expression and cell surface BCR expression, which together provide compartment is to deal with reinfection by variant pathogens.
survival signals, as well as the induction of EBI2 and S1PR1, which regulate B cell One of the unanswered questions for the lower-affinity model for
migration. Upregulation of HHEX also supports the repression of BCL-6 during selection towards memory B cells is why high-affinity cells are occasion-
the maturation of memory B cells from their precursor cells. TCR, T cell receptor;
ally included in the memory B cell compartment. Considering that the
TFH cell, T follicular helper cell.
numbers of GC TFH cells are much lower than light-zone GC B cells46,
some of the high-affinity light-zone GC B cells may not encounter TFH
cells because of their limited number, thereby allowing these B cells
mutations that diminish self-reactivity, in a process called redemp- to differentiate into memory B cells, even though they still retain the
tion, while preserving foreign antigen reactivity63. In some cases, this potential for being positively selected for GC or plasma cell fate71,72.
might induce a paradoxical decrease in overall affinity for the foreign GC B cells that recognize autoantigen are also thought to not interact
antigen, potentially enhancing the diversity of GC B cells. Given that with cognate TFH cells, because such cognate autoreactive T cells usually
human mature broadly neutralizing antibodies (bnAbs) against HIV undergo stringent negative selection. Hence, as in the case of the occa-
and influenza possess many mutations and that most of their germ-line sional selection of high-affinity cells into the memory compartment,
precursors are polyreactive and/or autoreactive11,64, B cells producing such autoreactive GC B cells might enter the memory pool as well.
such antibodies are likely to have undergone redemption during their Nevertheless, these polyreactive memory B cells have a disadvantage
maturation. In support of this mechanism, it was shown that facilitating for their persistence73.
redemption by using CTLA4-targeted checkpoint inhibitors increased The other possibility for the inclusion of high-affinity cells in the
bnAb responses against influenza virus65. memory B cell compartment is that, particularly in the late phase of

Nature Reviews Immunology


Review article

the GC reaction, antigen accessibility might be limited, for instance, how BCL-6 is downregulated in the process of differentiation towards
through antigen clearance or due to antigen masking by antibodies. memory B cell fate.
This may allow even high-affinity GC cells to differentiate into the
memory pool. Hence, we propose that, despite the importance of Phenotypic heterogeneity of memory B cells
affinity instruction, this instruction is not absolute, allowing a few Several distinct classes of memory B cells have been reported, even in a
high-affinity cells to enter the memory compartment. simple immunization regimen with a protein model antigen. Shlomchik
Another question concerns the molecular mechanisms that drive and others differentiated mouse memory B cell subsets according to
memory B cell differentiation and whether these are consistent with their expression patterns of CD80, PDL2 and CD73 (refs. 85–88). About
the affinity instruction model described above. Such studies had been 80% of the IgM-expressing memory B cells lacked CD80 and CD73 and,
hampered by the lack of a known master transcription factor for mem- upon transfer to naive mice, produced abundant GC B cells, whereas
ory B cell differentiation, but the recent characterization of memory CD80+CD73+ IgG1 memory B cells rapidly produced plasmablasts,
B precursors and the identification of key molecules by employing and later, only a small population of GC cells. The majority of the IgM-
CRISPR–Cas9 screening have started to clarify this issue66,68,74–76. For expressing memory B cells are generated in a GC-independent manner89
positively selected light-zone GC B cells to begin proliferating and to and the remaining 20% of CD80+CD73+ IgM memory B cells are likely
re-enter the dark zone, high levels of expression of the transcription derived from GC B cells. With regards to the mechanisms responsible
factor MYC and a high activity of the metabolic sensor mTORC1 are for such functional differences, given the evidence that switching of GC
required. Here, MYC activates cell division and mTORC1 is required for B cells to IgG1 strongly biases them towards the plasma cell and against
increasing biomass77,78. Unlike these cells, memory B precursor cells, the memory B cell fate73, these differences between CD80−CD73− and
which we have called pro-memory B cells, are in the G0 phase of the cell CD80+CD73+ memory B cell subsets might be caused by differences
cycle and express high levels of the transcription factor BACH2; this is in the signalling capabilities through IgM and IgG1 BCRs. However,
due to the low levels or absence of T cell help, which would otherwise nuclear transfer experiments also indicate another, not mutually exclu-
suppress BACH2 expression66,79. High BACH2 expression appears to be sive, possibility that cellular factors such as epigenetic changes due
required for differentiation into memory B cells, as BACH2-deficient to antigen experience contribute to these differences90. Thus, the key
GC B cells, which have constitutively active mTORC1 and high MYC driver that induces such functional differences among memory B cell
expression, are unable to generate memory B cells66. Hence, a high subsets remains to be clarified.
level of BACH2 expression contributes to memory B cell generation
through restraining MYC and mTORC1 pathways (Fig. 3b). Supporting
this conclusion is the observation that, when MYC function is perturbed Glossary
by the absence of its co-activator MIZ1, differentiation to memory
B cells is enhanced80.
The anti-proliferation programme, which is maintained at least Affinity maturation B1 and B2 B cells
partly by BACH2, is required but not sufficient for memory B cell gen- A process, as a result of somatic B cell populations that differ in
eration. A large portion of the light-zone GC B cells that fail to be posi- hypermutation (SHM) and B cell development, tissue localization and
tively selected undergo apoptosis. Only a small population of cells, the selection in the germinal centre (GC), by function. B2 B cells are the major
memory precursor B cells, express low levels of the transcription factor which B cells increase their affinity and and conventional B cell population
BCL-6 and high levels of BACH2, together with increased expression of avidity for the antigens. abundant in the spleen, lymph nodes
the anti-apoptotic regulator BCL-2 and cell surface expression of BCR81. and peripheral blood, which participate
As enforced downregulation of BCL-6 results in increased expression Age-associated B cells in the germinal centre (GC) reaction
of BCL-2 and the BCR, we speculate that downregulation of BCL-6 (ABCs). A subset of B cells expressing to produce high-affinity antibodies.
is a potential driver of memory B cell generation by, at least partly, CD11c and the transcription factor T-bet, B1 B cells are enriched in the peritoneal
provision of a BCL-2-mediated survival signal66. and lacking CD21 in mice. ABCs are and pleural cavities, which recognize
The importance of BCL-6 downregulation is further substantiated shown to increase during both ageing self-components and common
by experiments demonstrating the requirement for a key transcription and autoimmunity. bacterial antigens, and are the major
factor, HHEX, for memory B cell differentiation79. Here, ablation of Hhex source of natural IgM antibodies.
results in increased expression of BCL-6 and decreased expression of Antibody feedback
BCL-2, leading to cell death74. An immunological phenomenon in T follicular helper cells
The requirement for downregulation of BCL-6 might also explain which antibodies can either enhance (TFH cells). A subset of activated CD4+
the observation that memory precursor B cells reside on the edge or suppress the antigen-specific B cell T cells expressing the chemokine
of the GC light zone, because the G protein-coupled cell migratory responses. receptor CXCR5 and the transcription
receptors EBI2 and S1PR1, which control B cell positioning and facili- factor BCL-6. TFH cells provide help to
tate the movement to the edge of the GC light zone, are known to be Atypical memory B cells B cells for activation, germinal centre
directly repressed by BCL-6 (refs. 82,83) (Fig. 3b). Overall, as proposed A subset of memory B cells expressing (GC) formation and affinity maturation.
by Laidlaw and Cyster84, BACH2 and BCL-6 appear to be key participants inhibitory molecules and lacking
in modulating memory B cell differentiation. surface CD21 and CD27 in humans. Somatic hypermutation
Despite the progress with regards to the molecular mechanisms Atypical memory B cells often develop (SHM). A process by which B cells
of memory B cell generation, it is still unknown whether the small in individuals with chronic infection accumulate point mutations in their
portion of memory precursor B cells among the large pool of B cells or autoimmune disease, and are immunoglobulin genes, thus diversifying
that is undergoing apoptosis is selected stochastically or whether characterized by their reduced effector the specificity and affinity of their B cell
it receives an instructive signal. In addition, it needs to be clarified function. receptors (BCRs) for the antigens.

Nature Reviews Immunology


Review article

Atypical memory B cells More recently, analyses of data from humans with SARS-CoV-2
Atypical memory B cells were first identified as a B cell population infection and individuals who received COVID-19 mRNA vaccination
with unique morphology, functional capabilities and tissue localiza- have indeed identified, in both cases, CD21lowCD27− atypical memory
tion in humans91. They were called ‘atypical’ as they express inhibitory B cells corresponding to the CD11c+T-bet+ ABC subset in mice107. Given
receptors including FCRL4 and are relatively insensitive to in vitro that these atypical memory B cells are also generated in individuals
antigen stimulation. In humans, the non-plasmablast B cell population with other viral infections or with malaria, but not in those who receive
contains CD21+CD27+ ‘typical’ memory B cells and two poorly under- simple protein immunization, we speculate that B cells exposed to a
stood subsets: a population of CD21−CD27+ B cells, which contains a more inflammatory milieu (as is also the case for current COVID-19
plasmablast-like subset, and a CD21−CD27− ‘atypical’ memory B cell vaccines) are more prone to differentiate into atypical memory B cells.
population that contains a CD11c+ subset, which shares features with As for the inflammatory mediators, mouse studies indicated an
CD11chiCD21low age-associated B cells (ABCs) described in mice. important role for TLR7 and IL-21 as described above102,103; B cell specific
ABCs have been a focus of growing interest over the past decade, deletion of Tlr7 or Myd88 abolished the development of ABCs in aged
particularly in autoimmune settings92,93. In mice, the numbers of ABCs mice94, and IL-21 transgenic mice had spontaneous ABC formation108.
increase with age, as well as after chronic infection and in the context The function of ABCs has not been fully elucidated so far. Nevertheless,
of autoimmunity. Indeed, the depletion of ABCs in a mouse model of three potential functions have been proposed: ABCs may be exhausted
systemic lupus erythematosus (by putting diphtheria toxin receptor cells and non-functional, similar to CD8+ exhausted memory T cells; ABCs
(DTR) under the control of the CD11c promoter) abrogated the pro- may be able to differentiate in a less antigen-dependent manner com-
duction of autoantibodies, albeit with the caveat that this system can pared with conventional memory B cells; and ABCs may be specialized
potentially also deplete other activated B cells including GC B cells and for antigen presentation, their primary role being to activate T cells.
plasma cells94,95. Recent single-cell RNA-sequencing data from humans
demonstrate that the transcriptomic profiles of malaria-derived atypi- Reactivation of memory B cells
cal memory B cells and systemic lupus erythematosus-derived ABCs Experiments in which memory B cells are adoptively transferred into
are highly similar, although not identical96. Importantly, pseudotime naive recipient mice without memory lymphocytes or pre-existing
trajectory analysis of B cells in malaria infection shows that the dif- antibodies revealed differences in the functional capabilities of dif-
ferentiation pathway towards typical and atypical memory B cells ferent memory B cell subsets87,89,109,110. However, these experiments
becomes bifurcated as a consequence of their differentiation from did not necessarily reflect a natural secondary immunization/infec-
naive B cells, and that atypical memory B cells have higher expres- tion. In contrast to the naive state, mice that have experienced primary
sion of an IFNγ-driven gene signature than typical memory B cells97. immunization/infection harbour memory B cells, memory TFH cells and
Together, it is reasonable to conclude that, regardless whether reac- antigen-specific antibodies, all of which likely affect the subsequent
tive to non-self or self-antigen, ABCs can be categorized as a type of recall humoral responses.
memory B cell. The systematic analysis of human B cells across differ-
ent tissues also supports this conclusion by showing that the ABC-like In situ reactivation
CD11c+CD21−CD27− B cells in humans are memory B cells98. In situ re-immunization methods can model the natural situation of
With regards to the development of mouse ABCs, two groups memory B cell recall more closely than the adoptive transfer described
have identified ZEB2, which is not expressed by conventional memory above and can reveal the location of memory B cells and T cells before
B cells, as a key transcription factor for ABC generation99,100. ZEB2 reactivation, as well as their fates upon recall. In the lymph node, rest-
appears to affect cytokine receptor signalling pathways by transacti- ing memory B cells reside in a specialized niche in the subcapsular sinus
vating the genes encoding suppressor of cytokine signalling 5 (SOCS5) (SCS) region where they are closely associated with memory TFH cells
and the IL-21 receptor100. (Fig. 4). The SCS also contains a subset of CD169+ macrophages that
In addition to chronic infections, CD11c+CD21lowT-bet+ ABCs also display captured particulate antigens such as viruses and immune
appear following acute influenza and lymphocytic choriomeningitis complexes111,112. Once memory B cells bind antigens trapped by
virus (LCMV) infection in mice. In the case of LCMV infection101, mouse CD169+ macrophages, it is likely that these memory B cells present
CD11c+T-bet+ memory B cells, which correspond to ABCs, arise mainly them via MHC II molecules and swiftly activate memory TFH cells in a
during extrafollicular B cell responses (pre-GC phase) in the spleen, and cognate manner27, which is critical for secondary humoral responses
probably require non-GC TFH1 cells (IFNγ/IL-21 producing) for their devel- (Fig. 4). Hence, the close proximity of antigen-presenting CD169+
opment. Then, at the memory phase, these cells become localized at the macrophages, memory B cells and memory TFH cells is likely to allow
splenic marginal zone and express transcripts of plasma cell-associated for the generation of robust recall responses. Indeed, memory B cells
genes (such as BLIMP1, XBP1), suggesting that they are memory-type frequently make serial contacts with CD169+ macrophages via CCL21/
pre-plasma cells36, as initially proposed by McHeyzer-Williams and CCR7 interactions; CCL21 is expressed by stromal cells located close to
McHeyzer-Williams102. Indeed, although very few of these cells spon- the SCS and CCR7 is upregulated by memory B cells. The S1P–S1PR axis
taneously produce IgG in vitro, they rapidly differentiate into LCMV- is thought to direct the migration of memory B cells into the periphery,
specific plasma cells in response to stimulation with Toll-like receptor leading to their appearance in other lymphoid organs. On the other
7 (TLR7) ligand only103,104. In the case of the influenza infection model, hand, CCL21 directs memory B cells to remain in the original lymph
CD11c+T-bet+ memory B cells are generated in a GC-dependent manner node112. It has been reported that a subpopulation of memory B cells
and are located, similar to the LCMV acute infection model, in the spleen, does not recirculate and instead resides long-term in the draining
probably in the marginal zone105. Given the importance of marginal lymph node111,113. This might be explained by higher expression of CCR7
zone B cells for the initial recognition of blood-borne pathogens106, and lower expression of S1PR2. Similarly, a subpopulation of memory
these CD11c+T-bet+ memory B cells might play a more important role TFH cells was reported to reside long-term in the draining lymph node113.
for initiating protection against systemic rather than local infections. The continued residence of memory B cells and/or possibly memory

Nature Reviews Immunology


Review article

Antigen
Subcasular
sinus

Cytokine
receptor
Patrolling Cytokine
MHC II
TCR
Reactivated
SCS Subcapsular memory TFH cell
BCR
macrophage niche Reactivated
memory B cell CD40 CD40L GC entry
TCR
Plasma GC B cell
Memory differentiation
CCL21 Resting TFH cell Antibody
memory feedback
B cell

CCR7
Follicle

Fig. 4 | Memory B cell reactivation. At the memory state in the lymph node and to form GC-dependent memory B cells or differentiate into plasma cells.
before encountering antigens, resting memory B cells reside in a specialized Memory TFH cells are also activated in contact with SCS macrophages. Such
niche in the subcapsular sinus (SCS) region and are closely associated with close localization of SCS macrophages, memory B cells and memory TFH cells
memory T follicular helper (TFH) cells and SCS macrophages. CCL21 expressed may contribute to the robust recall responses. Pre-existing antibodies or those
by stromal cells near the SCS attracts memory B cells that express CCR7. After produced by boosted memory B cells can mask specific epitopes on antigen,
pathogens are captured by SCS macrophages, resting memory B cells are thereby impairing the activation of cognate memory B cells. BCR, B cell receptor;
activated in the subcapsular niche and can enter the germinal centre (GC) TCR, T cell receptor.

TFH cells in the draining lymph node may explain the observations that Characteristics of in vivo reactivated memory B cells
local antigen boosts facilitate recall GC responses better than distal Detailed clonal analysis of memory B cells in mice using homologous
ones114. Considering that many vaccines are administered as boosters, in situ influenza HA protein boosts clarified that a very strict bot-
clarifying what fractions of the populations of memory B cells and tleneck to clonal diversity operates at the time of recall of memory
T cells that are generated in a draining lymph node remain resident B cells to the secondary plasmablast response46. Of the hundreds of
versus entering circulation is an important unresolved issue. memory B cell clones that are generated during priming, only a few
Response to vaccine boosters or re-exposure to a pathogen has clones are activated and develop into secondary plasmablasts through
also been characterized in peripheral tissues. In the influenza infec- repeated recruitment of a few higher-affinity dominant clones to sec-
tion system, reinfection induces the formation of tertiary lymphoid ondary GCs119. Whereas most secondary plasmablasts are derived
structures (iBALT in the lung) where GC reactions can occur, thereby from memory B cells, secondary GCs in the prime-boost setting are
generating resident memory B cells as well as resident memory TFH cells predominantly composed of naive B cells, with only a minor memory
in the lung115. With regards to resident memory B cells, after primary B cell compartment. This leads to the formation of early secondary
influenza infection both antigen-specific CXCR3+CCR6+ B cells and GCs, which contain B cells that are just as diverse and poorly mutated
CXCR3−CCR6+ bystander resident memory B cells are generated116. as those in early primary-immunized GCs. Such recruitment of naive
This observation is not specific for influenza infection but has also B cells into secondary GCs was also observed in humans12, but the
been reported in SARS-CoV-2 infection117, and is likely to be a gen- ratio of memory B cells versus naive B cells in secondary GC reactions
eral phenomenon in lung infection. Although the exact function of is generally higher in humans compared with mice. This may reflect
bystander memory B cells is not clear at this point, the data suggest extensive histories of influenza antigen exposure in humans or pro-
that higher expression of the IgM Fc receptor, particularly by bystander found biological differences in the reactivation of memory B cells
CXCR3−CCR6+ resident memory B cells, might contribute to booster between humans and mice.
reactions through binding of IgM immune complexes. One important question is why there is such a paucity of memory
In the case of influenza reinfection, alveolar macrophages are B cell-derived B cells in recall GC responses in the mouse. Given that the
key initiators for humoral recall responses through the secretion of GC is the most important environment for generating BCR diversity and
IFNγ and CXCR3 ligands (CXCL9/CXCL10), which activates the recruit- antibody breadth, it is important to understand how memory B cells
ment of CXCR3+ resident memory B cells into foci of infected cells118. can re-enter GCs. Similar to the importance of precursor frequency and
Although it has not yet been clarified whether memory TFH cells are affinity of naive B cells for entering GCs, the numbers of memory B cells
required for the initiation of these recall responses, it is reasonable to generated by a single antigen encounter might be small120. In addition,
speculate that the resident memory B cells, as well as resident memory given that CD80+CD73+IgG+ memory B cells are prone to differentiate
TFH cells, are recruited close to alveolar macrophages, where these into plasma cells rather than enter the GC87, the numbers of functional
immune cells interact together, thereby exerting protective immunity. memory B cells that are capable of entering GCs might be even smaller.

Nature Reviews Immunology


Review article

Other possibilities should be considered, such as negative a two-dose or three-dose mRNA vaccine regimen, the distribution
feedback by antigen-specific antibodies121, as initially proposed by of epitopes recognized by memory B cells is shifted away from the
Smith in 1909 (ref. 122). Recent studies also indicate that antibody initial immunodominant RBD epitopes58,123. Thus, masking of these
feedback is particularly important in determining booster-mediated immunodominant epitopes by pre-generated antibodies diversifies
B cell fates. For instance, it was shown in adoptive transfer experiments immunogenicity of otherwise subdominant epitopes on the RBD. In
that antibodies produced by IgG+ memory B cells could inhibit the the case of mRNA vaccination, these second-layer antibodies target
formation of IgM+ memory B cell-derived secondary GCs110. Then, by RBD epitopes conserved between variants and acquire high-potency
using adoptive transfer of BCR knock-in memory or naive B cells, these neutralizing activity through GC reactions, together contributing to
donor B cells were only activated in naive mice, but not in immunized the development of functional breadth. However, in the case of HIV or
mice123, because of the existence of pre-existing masking antibodies influenza vaccination, the antibody feedback mechanism might halt
in the latter. Together with the data obtained from experiments in the maturation of memory B cells with potential bnAb BCRs11,123,139. For
which either antigen-specific or irrelevant antibodies were adminis- example, in the case of influenza vaccination and the response to the
tered124,125, antibodies either present prior to boosting or produced key conserved stem epitope, critical specific on-target neutralizing
acutely by boosted memory B cells are thought to compete with cog- antibodies coexist with many off-target non-neutralizing antibodies11.
nate naive as well as memory B cells for binding a specific epitope Hence, the efficient generation of off-target antibodies probably limits
through antibody-mediated epitope masking (Fig. 4, right). access to this conserved epitope by memory B cells with on-target
BCRs, thereby not allowing them to re-enter the GC. Eventually, this
Lessons from SARS-CoV-2 mechanism seems to operate by diverting immunity away from mak-
Many excellent reviews of the insights gained from SARS-CoV-2 infec- ing memory B cells with on-target bnAbs, and instead towards those
tion and COVID-19 vaccines with regards to B cell differentiation have with strain-specific ones.
been published126–128. Here, we focus on human memory B cells and,
in particular, how they acquire neutralizing antibody (nAb) breadth. Memory B cells, TFH cells and vaccines
Acquiring such a feature after only a single viral infection129 or after two How can we overcome the obstacles to bnAb development when design-
or three doses of mRNA vaccines encoding the ancestral SARS-CoV-2 ing vaccines? In the case of anti-stem influenza HA bnAbs, three poten-
spike protein would not be expected. tial obstacles exist with regard to B cell maturation: germ-line ancestors
Two doses of mRNA vaccine or one incidence of SARS-CoV-2 infec- of matured broadly neutralizing BCRs may have extremely low affinity
tion induces similar frequencies of IgG+ memory B cells that bind the for the native epitopes; naive B cells with these ancestor BCRs may be
spike receptor-binding domain (RBD), with comparable SHM levels. extremely rare; and these bnAbs are immunosubdominant. So, in the
Given that a substantial fraction of RBD+IgG+ memory B cells from competitive situation of a polyclonal naive B cell setting, knowing how
individuals who have been vaccinated with two doses of mRNA-based such rare naive B cells specific for the conserved epitope (such as the
COVID-19 vaccines can also bind to RBDs of variants of concern130,131, stem epitope in the case of influenza) are recruited into the immune
it appears that this vaccine regimen generated substantial numbers of response, and subsequent GC reactions, and induced to generate
affinity matured memory B cells132,133. However, the affinity maturation memory B cells is key to successful vaccine development. Moreover,
after a standard two-dose mRNA vaccine regimen is qualitatively poorer knowing how GC-dependent memory B cells are recruited again into GC
than that after a single SARS-CoV-2 infection. These differences may responses upon boosting with a second immunization is also important.
be related to the narrow window of time between dose 1 and dose 2 of Hence, one way to induce memory B cells for re-entering GC responses
the mRNA vaccines of usually 3 or 4 weeks, as experimental models would be to modify the immunogens, for example by introducing muta-
of vaccination against HIV have shown that extending the priming tions or glycan masking140,141, so that they are recognized by the desired
period can result in better nAb responses126,134. germ-line or intermediate BCRs, but not by non-neutralizing BCRs.
After mRNA vaccination, GCs were observed to persist for at least Given that limited TFH cell help at the T–B border is another bottle­
6 months in draining lymph nodes of healthy individuals after the sec- neck, increasing the number of antigen-specific TFH cells might allow
ond dose of COVID-19 mRNA vaccine135–137. GCs have been shown to play low-affinity B cells to be recruited into the immune responses and
a central role in the generation of nAb breadth of memory B cells132,133. subsequent GC reactions. Indeed, experiments using an HIV model
Indeed, nAb responses are substantially reduced in many individuals showed that higher levels of T cell help allow rare B cells that express
who are immunocompromised, such as recipients of kidney trans- the germ-line BCRs of mature bnAbs to enter the GC reaction142. Hence,
plants138. The lower GC responses in recipients of kidney transplant it is likely that with high quantities and qualities of TFH cells, rare B cells
could be due to weaker GC TFH cell responses, as the frequency of GC with germ-line BCRs of potential bnAbs could have a better chance of
TFH cells in these patients was severely reduced and associated with being recruited into GCs, wherein these BCRs become matured by SHM,
poor nAb titres. eventually generating high-quality bnAb-expressing memory B cells81.
Thus, the question arises of why memory B cells acquire nAb Finally, as discussed above, particularly during booster vaccina-
breadth much more easily in the case of SARS-CoV-2 infection and tion, and differing from SARS-CoV-2 infection, avoidance of antibody
COVID-19 mRNA vaccination compared with HIV or influenza infec- feedback may make sense for generating memory B cells that are
tion. Two types of studies have recently demonstrated the importance capable of producing bnAbs.
of antibody feedback in GC responses. First, in the context of mRNA
vaccination, the introduction of high-affinity epitope-specific mono- Outlook
clonal Abs affected memory B cell selection by two mechanisms: the In the past 10 years, the cellular and molecular mechanisms of memory
affinity threshold for the entry into the memory and GC compart- B cell function in polyclonal settings have been carefully analysed, and
ment was altered, and the epitopes targeted on RBD were changed the findings have broad implications for vaccinology and the treatment
by direct masking125. Second, we and others have shown that, after of autoimmunity. Although future directions and open questions have

Nature Reviews Immunology


Review article

been discussed throughout this Review, we emphasize the importance 23. Reboldi, A. et al. IgA production requires B cell interaction with subepithelial dendritic
cells in Peyer’s patches. Science 352, aaf4822 (2016).
of gaining a better fundamental understanding of the spatiotemporal 24. Toellner, K. M., Gulbranson-Judge, A., Taylor, D. R., Sze, D. M. & MacLennan, I. C.
aspects of memory B cell generation, maintenance and reactivation, Immunoglobulin switch transcript production in vivo related to the site and time of
and how memory B cell heterogeneity (such as conventional versus antigen-specific B cell activation. J. Exp. Med. 183, 2303–2312 (1996).
25. Crotty, S. T follicular helper cell biology: a decade of discovery and diseases. Immunity
atypical memory B cells) can be formed, as well as how other types of 50, 1132–1148 (2019).
cells, particularly TFH cells and FDCs, participate in these processes. 26. Hong, S. et al. B cells are the dominant antigen-presenting cells that activate naive CD4+
Recent technical advances in high-throughput single-cell sequencing T cells upon immunization with a virus-derived nanoparticle antigen. Immunity 49,
695–708.e4 (2018).
to explore both gene expression and immune repertoire profiles will 27. Ise, W. et al. Memory B cells contribute to rapid Bcl6 expression by memory follicular
allow us to uncover novel principles underlying clonal selection and helper T cells. Proc. Natl Acad. Sci. USA 111, 11792–11797 (2014).
the acquisition of different differentiation trajectories. The develop- 28. Choi, Y. S. et al. ICOS receptor instructs T follicular helper cell versus effector cell
differentiation via induction of the transcriptional repressor Bcl6. Immunity 34, 932–946
ment of technologies to investigate immunization-induced memory (2011).
B cells in higher primates shifts the studies to the human setting, which, 29. DiToro, D. et al. Differential IL-2 expression defines developmental fates of follicular
versus nonfollicular helper T cells. Science 361, eaao2933 (2018).
in turn, will allow us to engineer vaccines optimized for the generation
30. Zotos, D. et al. IL-21 regulates germinal center B cell differentiation and proliferation
of memory B cells or to target memory B cells for the treatment of through a B cell-intrinsic mechanism. J. Exp. Med. 207, 365–378 (2010).
autoimmune diseases and allergies in humans. In this respect, research 31. Linterman, M. A. et al. IL-21 acts directly on B cells to regulate Bcl-6 expression and
germinal center responses. J. Exp. Med. 207, 353–363 (2010).
into the functions of memory B cells in disease pathologies should be
32. Olatunde, A. C., Hale, J. S. & Lamb, T. J. Cytokine-skewed TFH cells: functional
a high-priority research area. consequences for B cell help. Trends Immunol. 42, 536–550 (2021).
33. Eisenbarth, S. C. et al. CD4+ T cells that help B cells—a proposal for uniform
nomenclature. Trends Immunol. 42, 658–669 (2021).
Published online: xx xx xxxx 34. Liu, D. et al. T-B-cell entanglement and ICOSL-driven feed-forward regulation of germinal
References centre reaction. Nature 517, 214–218 (2015).
1. Chappert, P. et al. Human anti-smallpox long-lived memory B cells are defined by 35. Crotty, S. T follicular helper cell differentiation, function, and roles in disease. Immunity
dynamic interactions in the splenic niche and long-lasting germinal center imprinting. 41, 529–542 (2014).
Immunity 55, 1872–1890.e9 (2022). 36. Tangye, S. G. & Warnatz, K. “Are you gonna go my way?” — Decisions at the TFH–B cell
2. Duggal, N. A., Niemiro, G., Harridge, S. D. R., Simpson, R. J. & Lord, J. M. Can physical interface. Immunity 55, 377–379 (2022).
activity ameliorate immunosenescence and thereby reduce age-related multi-morbidity? 37. Moriyama, S. et al. Sphingosine-1-phosphate receptor 2 is critical for follicular helper
Nat. Rev. Immunol. 19, 563–572 (2019). T cell retention in germinal centers. J. Exp. Med. 211, 1297–1305 (2014).
3. Amanna, I. J., Carlson, N. E. & Slifka, M. K. Duration of humoral immunity to common viral 38. Merkenschlager, J. et al. Dynamic regulation of TFH selection during the germinal centre
and vaccine antigens. N. Engl. J. Med. 357, 1903–1915 (2007). reaction. Nature 591, 458–463 (2021).
4. Crotty, S. et al. Cutting edge: long-term B cell memory in humans after smallpox 39. Taylor, J. J., Pape, K. A., Steach, H. R. & Jenkins, M. K. Humoral immunity. Apoptosis and
vaccination. J. Immunol. 171, 4969–4973 (2003). antigen affinity limit effector cell differentiation of a single naive B cell. Science 347,
5. Kurosaki, T., Kometani, K. & Ise, W. Memory B cells. Nat. Rev. Immunol. 15, 149–159 784–787 (2015).
(2015). This study shows that the differentiation fate of individual antigen-specific naive
6. Anderson, S. M., Tomayko, M. M. & Shlomchik, M. J. Intrinsic properties of human and B cells is influenced by BCR affinity for antigen.
murine memory B cells. Immunol. Rev. 211, 280–294 (2006). 40. Paus, D. et al. Antigen recognition strength regulates the choice between extrafollicular
7. Tarlinton, D. & Good-Jacobson, K. Diversity among memory B cells: origin, consequences, plasma cell and germinal center B cell differentiation. J. Exp. Med. 203, 1081–1091
and utility. Science 341, 1205–1211 (2013). (2006).
8. Purtha, W. E., Tedder, T. F., Johnson, S., Bhattacharya, D. & Diamond, M. S. Memory B cells, 41. Zhang, T. T. et al. Germinal center B cell development has distinctly regulated stages
but not long-lived plasma cells, possess antigen specificities for viral escape mutants. completed by disengagement from T cell help. eLife 6, e19552 (2017).
J. Exp. Med. 208, 2599–2606 (2011). 42. Lingwood, D. et al. Structural and genetic basis for development of broadly neutralizing
9. Leach, S. et al. Requirement for memory B-cell activation in protection from influenza antibodies. Nature 489, 566–570 (2012).
heterologous influenza virus reinfection. Int. Immunol. 31, 771–779 (2019). 43. Glaros, V. et al. Limited access to antigen drives generation of early B cell memory while
10. Wong, R. et al. Affinity-restricted memory B cells dominate recall responses to restraining the plasmablast response. Immunity 54, 2005–2023.e10 (2021).
heterologous flaviviruses. Immunity 53, 1078–1094.e7 (2020). 44. Young, C. & Brink, R. The unique biology of germinal center B cells. Immunity 54,
11. Andrews, S. F. et al. Immune history profoundly affects broadly protective B cell 1652–1664 (2021).
responses to influenza. Sci. Transl Med. 7, 316ra192 (2015). 45. Cyster, J. G. & Allen, C. D. C. B cell responses: cell interaction dynamics and decisions.
12. Turner, J. S. et al. Human germinal centres engage memory and naive B cells after Cell 177, 524–540 (2019).
influenza vaccination. Nature 586, 127–132 (2020). 46. Victora, G. D. & Nussenzweig, M. C. Germinal centers. Annu. Rev. Immunol. 40, 413–442
13. Lindner, C. et al. Diversification of memory B cells drives the continuous adaptation (2022).
of secretory antibodies to gut microbiota. Nat. Immunol. 16, 880–888 (2015). 47. Viant, C. et al. Antibody affinity shapes the choice between memory and germinal center
14. Vergani, S. et al. A self-sustaining layer of early-life-origin B cells drives steady-state B cell fates. Cell 183, 1298–1311.e11 (2020).
IgA responses in the adult gut. Immunity 55, 1829–1842.e6 (2022). 48. Shinnakasu, R. et al. Regulated selection of germinal-center cells into the memory B cell
15. Schiepers, A. et al. Molecular fate-mapping of serum antibody responses to repeat compartment. Nat. Immunol. 17, 861–869 (2016).
immunization. Nature 615, 482–489 (2023). 49. Shlomchik, M. J., Luo, W. & Weisel, F. Linking signaling and selection in the germinal
Together with Turner et al. (2020), this study suggests that boosting with immunogens center. Immunol. Rev. 288, 49–63 (2019).
of sufficient antigenic distance allows naive B cells to enter secondary GC responses. 50. Quast, I. et al. Interleukin-21, acting beyond the immunological synapse, independently
16. Morris, D. L. & Rothstein, T. L. Abnormal transcription factor induction through the controls T follicular helper and germinal center B cells. Immunity 55, 1414–1430.e15 (2022).
surface immunoglobulin M receptor of B-1 lymphocytes. J. Exp. Med. 177, 857–861 51. Huse, M., Lillemeier, B. F., Kuhns, M. S., Chen, D. S. & Davis, M. M. T cells use two
(1993). directionally distinct pathways for cytokine secretion. Nat. Immunol. 7, 247–255 (2006).
17. Savage, H. P. et al. TLR induces reorganization of the IgM–BCR complex regulating 52. Tas, J. M. et al. Visualizing antibody affinity maturation in germinal centers. Science 351,
murine B-1 cell responses to infections. eLife 8, e46997 (2019). 1048–1054 (2016).
18. Patel, P. S., King, R. G. & Kearney, J. F. Pulmonary α-1,3-glucan-specific IgA-secreting 53. Kuraoka, M. et al. Complex antigens drive permissive clonal selection in germinal
B cells suppress the development of cockroach allergy. J. Immunol. 197, 3175–3187 centers. Immunity 44, 542–552 (2016).
(2016). 54. Schwickert, T. A., Alabyev, B., Manser, T. & Nussenzweig, M. C. Germinal center
19. Macpherson, A. J. & Harris, N. L. Interactions between commensal intestinal bacteria and reutilization by newly activated B cells. J. Exp. Med. 206, 2907–2914 (2009).
the immune system. Nat. Rev. Immunol. 4, 478–485 (2004). 55. Zhang, Y. et al. Germinal center B cells govern their own fate via antibody feedback.
20. Taylor, J. J., Pape, K. A. & Jenkins, M. K. A germinal center-independent pathway J. Exp. Med. 210, 457–464 (2013).
generates unswitched memory B cells early in the primary response. J. Exp. Med. 209, 56. Toellner, K. M., Sze, D. M. & Zhang, Y. What are the primary limitations in B-cell
597–606 (2012). affinity maturation, and how much affinity maturation can we drive with vaccination?
21. Kaji, T. et al. Distinct cellular pathways select germline-encoded and somatically mutated A role for antibody feedback. Cold Spring Harb. Perspect. Biol. 10, a028795 (2018).
antibodies into immunological memory. J. Exp. Med. 209, 2079–2097 (2012). 57. Meyer-Hermann, M. Injection of antibodies against immunodominant epitopes tunes
22. Roco, J. A. et al. Class-switch recombination occurs infrequently in germinal centers. germinal centers to generate broadly neutralizing antibodies. Cell Rep. 29, 1066–1073.e5
Immunity 51, 337–350.e7 (2019). (2019).

Nature Reviews Immunology


Review article

58. Inoue, T. et al. Antibody feedback contributes to facilitating the development of 87. Zuccarino-Catania, G. V. et al. CD80 and PD-L2 define functionally distinct memory
Omicron-reactive memory B cells in SARS-CoV-2 mRNA vaccinees. J. Exp. Med. 220, B cell subsets that are independent of antibody isotype. Nat. Immunol. 15, 631–637
e20221786 (2023). (2014).
59. Hagglof, T. et al. Continuous germinal center invasion contributes to the diversity of the 88. Krishnamurty, A. T. et al. Somatically hypermutated plasmodium-specific IgM+ memory
immune response. Cell 186, 147–161.e15 (2023). B cells are rapid, plastic, early responders upon malaria rechallenge. Immunity 45,
60. de Carvalho, R. V. H. et al. Clonal replacement sustains long-lived germinal centers 402–414 (2016).
primed by respiratory viruses. Cell 186, 131–146.e13 (2023). 89. Viant, C. et al. Germinal center-dependent and -independent memory B cells produced
Together with Hagglof et al. (2023), this study shows that low-affinity naive B cells throughout the immune response. J. Exp. Med. 218, e20202489 (2021).
continuously invade ongoing GCs, progressively replacing the founder clones, 90. Kometani, K. et al. Repression of the transcription factor Bach2 contributes to
whereas rare long-lived founder clones generate highly mutated memory B cells that predisposition of IgG1 memory B cells toward plasma cell differentiation. Immunity 39,
can be recalled. 136–147 (2013).
61. Sabouri, Z. et al. Redemption of autoantibodies on anergic B cells by variable-region 91. Ehrhardt, G. R. et al. Expression of the immunoregulatory molecule FcRH4 defines
glycosylation and mutation away from self-reactivity. Proc. Natl Acad. Sci. USA 111, a distinctive tissue-based population of memory B cells. J. Exp. Med. 202, 783–791
E2567–E2575 (2014). (2005).
62. Reed, J. H., Jackson, J., Christ, D. & Goodnow, C. C. Clonal redemption of autoantibodies 92. Cancro, M. P. Age-associated B cells. Annu. Rev. Immunol. 38, 315–340 (2020).
by somatic hypermutation away from self-reactivity during human immunization. 93. Phalke, S. et al. Molecular mechanisms controlling age-associated B cells in
J. Exp. Med. 213, 1255–1265 (2016). autoimmunity. Immunol. Rev. 307, 79–100 (2022).
63. Burnett, D. L. et al. Germinal center antibody mutation trajectories are determined 94. Rubtsov, A. V. et al. Toll-like receptor 7 (TLR7)-driven accumulation of a novel CD11c+
by rapid self/foreign discrimination. Science 360, 223–226 (2018). B-cell population is important for the development of autoimmunity. Blood 118,
This study shows evidence that self-reactive BCR undergoes redemption by SHM 1305–1315 (2011).
during GC responses. 95. Gao, X. & Cockburn, I. A. The development and function of CD11c+ atypical B cells—insights
64. Bajic, G. et al. Influenza antigen engineering focuses immune responses to a from single cell analysis. Front. Immunol. 13, 979060 (2022).
subdominant but broadly protective viral epitope. Cell Host Microbe 25, 827–835.e6 96. Holla, P. et al. Shared transcriptional profiles of atypical B cells suggest common
(2019). drivers of expansion and function in malaria, HIV, and autoimmunity. Sci. Adv. 7,
65. Sangesland, M. et al. Allelic polymorphism controls autoreactivity and vaccine eabg8384 (2021).
elicitation of human broadly neutralizing antibodies against influenza virus. Immunity 55, 97. Ambegaonkar, A. A., Holla, P., Dizon, B. L., Sohn, H. & Pierce, S. K. Atypical B cells in
1693–1709.e8 (2022). chronic infectious diseases and systemic autoimmunity: puzzles with many missing
66. Inoue, T. et al. Exit from germinal center to become quiescent memory B cells pieces. Curr. Opin. Immunol. 77, 102227 (2022).
depends on metabolic reprograming and provision of a survival signal. J. Exp. 98. Dominguez Conde, C. et al. Cross-tissue immune cell analysis reveals tissue-specific
Med. 218, e20200866 (2021). features in humans. Science 376, eabl5197 (2022).
67. Inoue, T., Moran, I., Shinnakasu, R., Phan, T. G. & Kurosaki, T. Generation of memory B cells 99. Gao, X. et al. ZEB2 regulates the development of CD11c+ atypical B cells. Preprint at
and their reactivation. Immunol. Rev. 283, 138–149 (2018). bioRxiv https://doi.org/10.1101/2022.09.01.506173 (2022).
68. Suan, D. et al. CCR6 defines memory B cell precursors in mouse and human germinal 100. Gu, S. et al. The transcription factor Zeb2 drives differentiation of age-associated B cells.
centers, revealing light-zone location and predominant low antigen affinity. Immunity 47, Preprint at bioRxiv https://doi.org/10.1101/2021.07.24.453633 (2021).
1142–1153.e4 (2017). 101. Song, W. et al. Development of Tbet- and CD11c-expressing B cells in a viral infection
Together with Wong et al. (2020), Viant et al. (2020) and Shinnakasu et al. (2016), requires T follicular helper cells outside of germinal centers. Immunity 55, 290–307.e5
this work shows that the GC B cells with low-affinity BCR favour the memory B cell (2022).
differentiation fate. 102. McHeyzer-Williams, L. J. & McHeyzer-Williams, M. G. Antigen-specific memory B cell
69. Duan, L. et al. Follicular dendritic cells restrict interleukin-4 availability in germinal development. Annu. Rev. Immunol. 23, 487–513 (2005).
centers and foster memory B cell generation. Immunity 54, 2256–2272.e6 (2021). 103. Clingan, J. M. & Matloubian, M. B cell-intrinsic TLR7 signaling is required for optimal B cell
70. Weisel, F. J., Zuccarino-Catania, G. V., Chikina, M. & Shlomchik, M. J. A temporal switch responses during chronic viral infection. J. Immunol. 191, 810–818 (2013).
in the germinal center determines differential output of memory B and plasma cells. 104. Walsh, K. B. et al. Toll-like receptor 7 is required for effective adaptive immune
Immunity 44, 116–130 (2016). responses that prevent persistent virus infection. Cell Host Microbe 11, 643–653
71. Schwickert, T. A. et al. A dynamic T cell-limited checkpoint regulates affinity-dependent (2012).
B cell entry into the germinal center. J. Exp. Med. 208, 1243–1252 (2011). 105. Johnson, J. L. et al. The transcription factor T-bet resolves memory B cell subsets with
72. Mesin, L., Ersching, J. & Victora, G. D. Germinal center B cell dynamics. Immunity 45, distinct tissue distributions and antibody specificities in mice and humans. Immunity 52,
471–482 (2016). 842–855.e6 (2020).
73. Gitlin, A. D. et al. Independent roles of switching and hypermutation in the development 106. Cerutti, A., Cols, M. & Puga, I. Marginal zone B cells: virtues of innate-like antibody-
and persistence of B lymphocyte memory. Immunity 44, 769–781 (2016). producing lymphocytes. Nat. Rev. Immunol. 13, 118–132 (2013).
74. Laidlaw, B. J., Duan, L., Xu, Y., Vazquez, S. E. & Cyster, J. G. The transcription factor 107. McGrath, J. J. C., Li, L. & Wilson, P. C. Memory B cell diversity: insights for optimized
Hhex cooperates with the corepressor Tle3 to promote memory B cell development. vaccine design. Trends Immunol. 43, 343–354 (2022).
Nat. Immunol. 21, 1082–1093 (2020). 108. Naradikian, M. S. et al. Cutting Edge: IL-4, IL-21, and IFN-γ interact to govern T-bet and
75. Laidlaw, B. J. et al. The Eph-related tyrosine kinase ligand Ephrin-B1 marks germinal CD11c expression in TLR-activated B cells. J. Immunol. 197, 1023–1028 (2016).
center and memory precursor B cells. J. Exp. Med. 214, 639–649 (2017). 109. Dogan, I. et al. Multiple layers of B cell memory with different effector functions.
76. Wang, Y. et al. Germinal-center development of memory B cells driven by IL-9 from Nat. Immunol. 10, 1292–1299 (2009).
follicular helper T cells. Nat. Immunol. 18, 921–930 (2017). 110. Pape, K. A., Taylor, J. J., Maul, R. W., Gearhart, P. J. & Jenkins, M. K. Different B cell
77. Finkin, S., Hartweger, H., Oliveira, T. Y., Kara, E. E. & Nussenzweig, M. C. Protein amounts populations mediate early and late memory during an endogenous immune response.
of the MYC transcription factor determine germinal center B cell division capacity. Science 331, 1203–1207 (2011).
Immunity 51, 324–336.e5 (2019). 111. Moran, I. et al. Memory B cells are reactivated in subcapsular proliferative foci of lymph
78. Ersching, J. et al. Germinal center selection and affinity maturation require dynamic nodes. Nat. Commun. 9, 3372 (2018).
regulation of mTORC1 kinase. Immunity 46, 1045–1058.e6 (2017). 112. Zhang, Y. et al. Recycling of memory B cells between germinal center and lymph node
79. Chen, S. T., Oliveira, T. Y., Gazumyan, A., Cipolla, M. & Nussenzweig, M. C. B cell receptor subcapsular sinus supports affinity maturation to antigenic drift. Nat. Commun. 13, 2460
signaling in germinal centers prolongs survival and primes B cells for selection. (2022).
Immunity 56, 547–561.e7 (2023). 113. Fazilleau, N. et al. Lymphoid reservoirs of antigen-specific memory T helper cells.
80. Toboso-Navasa, A. et al. Restriction of memory B cell differentiation at the germinal Nat. Immunol. 8, 753–761 (2007).
center B cell positive selection stage. J. Exp. Med. 217, e20191933 (2020). 114. Kuraoka, M. et al. Recall of B cell memory depends on relative locations of prime and
81. Inoue, T., Shinnakasu, R. & Kurosaki, T. Generation of high quality memory B cells. boost immunization. Sci. Immunol. 7, eabn5311 (2022).
Front. Immunol. 12, 825813 (2021). 115. Tan, H. X. et al. Inducible bronchus-associated lymphoid tissues (iBALT) serve as sites of
82. Hatzi, K. et al. BCL6 orchestrates TFH cell differentiation via multiple distinct mechanisms. B cell selection and maturation following influenza infection in mice. Front. Immunol. 10,
J. Exp. Med. 212, 539–553 (2015). 611 (2019).
83. Huang, C. et al. The BCL6 RD2 domain governs commitment of activated B cells to form 116. Tan, H. X. et al. Lung-resident memory B cells established after pulmonary influenza
germinal centers. Cell Rep. 8, 1497–1508 (2014). infection display distinct transcriptional and phenotypic profiles. Sci. Immunol. 7,
84. Laidlaw, B. J. & Cyster, J. G. Transcriptional regulation of memory B cell differentiation. eabf5314 (2022).
Nat. Rev. Immunol. 21, 209–220 (2021). 117. Gregoire, C. et al. Viral infection engenders bona fide and bystander subsets of lung-
85. Anderson, S. M., Tomayko, M. M., Ahuja, A., Haberman, A. M. & Shlomchik, M. J. resident memory B cells through a permissive mechanism. Immunity 55, 1216–1233.e9
New markers for murine memory B cells that define mutated and unmutated subsets. (2022).
J. Exp. Med. 204, 2103–2114 (2007). 118. MacLean, A. J. et al. Secondary influenza challenge triggers resident memory B cell
86. Tomayko, M. M., Steinel, N. C., Anderson, S. M. & Shlomchik, M. J. Cutting edge: hierarchy migration and rapid relocation to boost antibody secretion at infected sites. Immunity
of maturity of murine memory B cell subsets. J. Immunol. 185, 7146–7150 (2010). 55, 718–733.e8 (2022).

Nature Reviews Immunology


Review article

119. Mesin, L. et al. Restricted clonality and limited germinal center reentry characterize 137. Mudd, P. A. et al. SARS-CoV-2 mRNA vaccination elicits a robust and persistent
memory B cell reactivation by boosting. Cell 180, 92–106.e11 (2020). T follicular helper cell response in humans. Cell 185, 603–613.e15 (2022).
This study shows that the secondary GCs during recall responses mostly consist of 138. Lederer, K. et al. Germinal center responses to SARS-CoV-2 mRNA vaccines in healthy
B cells with no prior GC experience, suggesting a clonality bottleneck for the antibody and immunocompromised individuals. Cell 185, 1008–1024.e15 (2022).
diversity. 139. Forsell, M. N. E., Kvastad, L., Sedimbi, S. K., Andersson, J. & Karlsson, M. C. I. Regulation
120. Abbott, R. K. et al. Precursor frequency and affinity determine B cell competitive fitness of subunit-specific germinal center B cell responses to the HIV-1 envelope glycoproteins
in germinal centers, tested with germline-targeting HIV vaccine immunogens. Immunity by antibody-mediated feedback. Front. Immunol. 8, 738 (2017).
48, 133–146.e6 (2018). 140. Duan, H. et al. Glycan masking focuses immune responses to the HIV-1 CD4-binding site
121. Heyman, B. Regulation of antibody responses via antibodies, complement, and Fc and enhances elicitation of VRC01-class precursor antibodies. Immunity 49, 301–311.e5
receptors. Annu. Rev. Immunol. 18, 709–737 (2000). (2018).
122. Smith, T. Active immunity produced by so called balanced or neutral mixtures of 141. Shinnakasu, R. et al. Glycan engineering of the SARS-CoV-2 receptor-binding domain
diphtheria toxin and antitoxin. J. Exp. Med. 11, 241–256 (1909). elicits cross-neutralizing antibodies for SARS-related viruses. J. Exp. Med. 218,
123. Tas, J. M. J. et al. Antibodies from primary humoral responses modulate the recruitment e20211003 (2021).
of naive B cells during secondary responses. Immunity 55, 1856–1871.e6 (2022). 142. Lee, J. H. et al. Modulating the quantity of HIV Env-specific CD4 T cell help promotes rare
124. McNamara, H. A. et al. Antibody feedback limits the expansion of B cell responses to B cell responses in germinal centers. J. Exp. Med. 218, e20201254 (2021).
malaria vaccination but drives diversification of the humoral response. Cell Host Microbe 143. Li, J., Lu, E., Yi, T. & Cyster, J. G. EBI2 augments TFH cell fate by promoting interaction with
28, 572–585.e7 (2020). IL-2-quenching dendritic cells. Nature 533, 110–114 (2016).
125. Schaefer-Babajew, D. et al. Antibody feedback regulates immune memory after
SARS-CoV-2 mRNA vaccination. Nature 613, 735–742 (2023). Acknowledgements
Together with Inoue et al. (2023) and Tas et al. (2022), this study suggests the The authors thank P. D. Burrows for critical reading of the manuscript. This work was
contribution of antibody feedback to B cell responses upon SARS-CoV-2 antigen supported, in part, by Japan Society for the Promotion of Science KAKENHI (JP21H02749 to
immunization in mice or mRNA vaccination in humans. T.I., JP22H00450 to T.K.), the Mochida Memorial Foundation for Medical and Pharmaceutical
126. Sette, A. & Crotty, S. Immunological memory to SARS-CoV-2 infection and COVID-19 Research (to T.I.) and the Chemo-Sero-Therapeutic Research Institute (to T.I.).
vaccines. Immunol. Rev. 310, 27–46 (2022).
127. Roltgen, K. & Boyd, S. D. Antibody and B cell responses to SARS-CoV-2 infection and Author contributions
vaccination. Cell Host Microbe 29, 1063–1075 (2021). T.K. contributed to conceptualization of the article. All authors contributed to writing and
128. Sette, A. & Crotty, S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 184, 861–880 editing of the manuscript.
(2021).
129. Rodda, L. B. et al. Functional SARS-CoV-2-specific immune memory persists after mild Competing interests
COVID-19. Cell 184, 169–183.e17 (2021). The authors declare no competing interests.
130. Goel, R. R. et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and
variants of concern. Science 374, abm0829 (2021). Additional information
131. Rodda, L. B. et al. Imprinted SARS-CoV-2-specific memory lymphocytes define hybrid Peer review information Nature Reviews Immunology thanks Jason Cyster and the other,
immunity. Cell 185, 1588–1601.e14 (2022). anonymous, reviewers for their contribution to the peer review of this work.
132. Wang, Z. et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants.
Nature 592, 616–622 (2021). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
133. Muecksch, F. et al. Increased memory B cell potency and breadth after a SARS-CoV-2 published maps and institutional affiliations.
mRNA boost. Nature 607, 128–134 (2022).
134. Cirelli, K. M. et al. Slow delivery immunization enhances HIV neutralizing antibody and Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this
germinal center responses via modulation of immunodominance. Cell 177, 1153–1171.e28 article under a publishing agreement with the author(s) or other rightsholder(s); author
(2019). self-archiving of the accepted manuscript version of this article is solely governed by the
135. Turner, J. S. et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre terms of such publishing agreement and applicable law.
responses. Nature 596, 109–113 (2021).
136. Kim, W. et al. Germinal centre-driven maturation of B cell response to mRNA vaccination.
Nature 604, 141–145 (2022). © Springer Nature Limited 2023

Nature Reviews Immunology

You might also like