You are on page 1of 4

Molecular pathogenesis of germinal center-derived B cell lymphomas

Summary
B cell lymphomas comprise a heterogeneous group of genetically, biologically, and clinically distinct neoplasms
that, in most cases, originate from the clonal expansion of B cells in the germinal center (GC). In recent years,
the advent of novel genomics technologies has revolutionized our understanding of the molecular
pathogenesis of lymphoid malignancies as a multistep process that requires the progressive accumulation of
multiple genetic and epigenetic alterations. A common theme that emerged from these studies is the ability of
lymphoma cells to co-opt the same biological programs and signal transduction networks that operate during
the normal GC reaction, and misuse them for their own survival advantage. This review summarizes recent
progress in the understanding of the genetic and epigenetic mechanisms that drive the malignant
transformation of GC B cells. These insights provide a conceptual framework for the identification of cellular
pathways that may be explored for precision medicine approaches.

INTRODUC TION
Among the plethora of genes and biological programs
The adaptive immune system has evolved to allow the
identified to date as targets of genetic alterations in these
effective recognition and response against a virtually
diseases, we will focus on those that are most frequently
unlimited number of invading pathogens. These
affected and have been functionally characterized in the
processes require the formation of germinal centers (GC),
context of the normal biology of the GC. We refer readers
a specialized microenvironment that ensures the
to other reviews for a comprehensive overview on the
generation and selection of cells producing antibodies with
interaction between the tumor cells and the
high affinity for the antigen.1–4 The GC reaction is thus
microenvironment, 11 the role of infectious agents,12 and
critical for the establishment of humoral immunity.
the clinical implications for mechanism-based therapeutic
However, the unique and complex events that are
approaches.13,14
associated with this reaction pose a significant risk to the
genome of GC B cells, which have to endure high THE D OUBLE-EDG ED S WORD O F T HE
replication stress while undergoing multiple DNA
breakage and recombination events required by the GC R E AC TION
antibody diversification processes of somatic
hypermutation (SHM) and class switch recombination Germinal centers are highly dynamic structures that form
(CSR).5 transiently in secondary lymphoid organs upon encounter
Additionally, GC B cells must have elevated epigenetic of a B cell with a foreign pathogen to enable the generation
plasticity in order to rapidly reprogram their transcriptional and selection of clones with high affinity antibodies.1,2,4,5
networks while recirculating between the GC dark zone In 2007, an elegant series of in vivo imaging studies
(DZ) and light zone (LZ) compartments to assume distinct demonstrated that, within these structures, B cells
functional states. Therefore, it does not come as a constantly recirculate between two anatomically separate
surprise that the majority of mature B cell lymphomas compartments15– 18: the DZ, which consists of rapidly
derive from the clonal expansion of a GC B cell, as proliferating cells (also known as centroblasts) capable of
documented by multiple evidences that include the dividing every 6-12 hours, and the LZ, which comprises
presence of somatically mutated immunoglobulin (IG) more quiescent cells termed centrocytes, admixed with
genes,6 the strong similarity of their gene expression resident accessory cells such as follicular dendritic cells
profiles with that of B cells in various phases of the GC (FDC), follicular helper T (Tfh) cells, and macrophages
reaction,7,8 and the frequent association with genetic (Figure 1).2,8 These two compartments correspond to
lesions—ie, chromosomal translocations and aberrant distinct functional states of the B lymphocytes.
SHM—that result from errors occurring during GC-specific Specifically, the DZ is the site where GC B cells undergo
DNA remodeling events.9,10 More recently, next- secondary diversification of their antibody genes through
generation sequencing studies and functional genomics the process of SHM,19 a DNA remodeling event initiated
analyses of lymphomas have refined this concept by by the activation induced cytidine deaminase (AID)
uncovering multiple genetic alterations, the consequence enzyme20,21 that introduces single nucleotide
of which is the dysregulation of epigenetic and substitutions, as well as small deletions and duplications,
transcriptional programs that operate during distinct in the variable region of the IG genes (IGV), the final goal
phases of the normal GC reaction, while facilitating being to increase the affinity of their B cell receptor (BCR)
evasion from immune recognition mechanisms. Indeed, for the antigen.22,23 DZ B cells then cease proliferation and
the common theme of these studies has been the move to the LZ, where they compete for signals delivered
realization that malignant cells co-opt the same signaling by a limited number of resident Tfh cells that trigger
pathways and regulatory networks that are normally positive selection.8 Based on affinity for the antigen, LZ B
utilized by GC B lymphocytes to sustain their own growth cells will either re-enter the DZ for iterative rounds of cell
and survival. This review will summarize current division and selection, which lead to affinity maturationat
knowledge about the genetics and pathogenesis of the the population level, exit the GC to become a memory B
most common GC-derived B-cell non-Hodgkin cell or a plasma cell, or be eliminated by the default GC
lymphomas, including Burkitt lymphoma (BL), follicular apoptotic program. The LZ is also the site where B cells
lymphoma (FL), and diffuse large B cell lymphoma undergo CSR, an intrachromosomal deletional
(DLBCL). recombination event that requires the activity of AID and
confers distinct effector functions to antibodies with
identical specificities.24,25 Ultimately, cells will terminate MEF2B and MEF2C
the cyclic DZ-LZ re-entry process and be selected for
survival and differentiation into long-lived memory B cells
The myocyte-specific enhancer factor 2B (MEF2B) and
and plasma cells.25,26
MEF2C proteins belong to an ancient family of
The two distinct phases of GC development reflect
transcription factors involved in the regulation of multiple
discrete transient states within the same B cell
developmental programs through interaction with specific
developmental stage, which can be recognized to some
transcriptional co-factors. 34 Recent studies have
extent in the transcriptional signature of the various
uncovered critical roles for these proteins in both normal
lymphoma subtypes, with BL being more similar to a DZ B
and malignant GC B cells, where MEF2B and MEF2C
cell, and FL and DLBCL being more related to a GC LZ
appear to play slightly distinct functions. In particular,
cell.8
MEF2C, which is expressed at all stages of mature B cell
TR ANSCRIPTION F AC TOR N E TWORKS differentiation, was shown to regulate the proliferation of
WIRING T HE G C GC B cells by functioning as a direct transcriptional
effector of BCR signaling via the p38 MAPK pathway.35
As mentioned, a common theme of recent genomic Consistently, loss of MEF2C function caused reduced
analyses has been the realization that most genetic immune responses to antigen and defective GC formation
alterations associated with B lymphoid malignancies due to impaired B-cell proliferation. 35 In contrast, MEF2B
converge on transcription factor networks that are is exquisitely expressed in the GC, where its transcription
normally used by the GC B cell to sense antigens and is rapidly induced after T-cell dependent antigen
assume distinct functional states during the GC reaction. activation, slightly before the upregulation of BCL6.36,37
This section focuses on selected proteins that, among This is in line with the ability of MEF2B to directly bind
those implicated in the control of GC physiology, are most promoter/enhancer sequences in the BCL6 locus and
commonly targeted by genetic lesions in lymphoma (see transactivate its expression.37,38 In addition, MEF2B is at
ref. 2 for a comprehensive review of the molecular the center of a broad transcriptional network that includes,
switches that coordinate the complex dynamics of the GC among other cis-regulatory elements, the full set of GC-
reaction). specific super-enhancers, indicating an important role in
instructing the GC reaction, and particularly the DZ.39 In
As a sequence specific DNA-binding transcription factor, this view, the modest effects on GC formation observed in
the MYC protein controls a broad range of cellular vivo upon single loss of Mef2b may suggest partially
programs, including proliferation, cell growth, energy redundant functions of Mef2b and Mef2c39, which can
metabolism, telomerase maintenance, differentiation, and dimerize with each other. In support of this hypothesis, co-
apoptosis,27 its target gene network being estimated to deletion of these two genes in GC B cells completely
include ~15% of all protein-coding genes as well as non- abrogated GC formation
coding RNAs.28 In addition, MYC was shown to control
DNA replication through mechanisms that are BCL6
independent of its transcriptional activity, a property that
may promote genomic instability by inducing replication Known as the “master regulator” of the GC reaction, BCL6
stress.29 is a potent transcriptional repressor that, within the mature
During the GC reaction, MYC expression is tightly B cell lineage, is selectively expressed in GC B cells40,41
regulated through a bimodal distribution pattern whereby and is absolutely required for GC development. The
the protein is first induced in the GC founding B cells, specific signals that drive upregulation of BCL6 following
shortly upon antigen binding to the BCR, but is then antigenic stimulation are in large part unknown; however,
transcriptionally repressed by BCL6 (and most likely other multiple transcription factors have been implicated in this
factors) in the DZ B cell population, before being re- process, including IRF4,42 IRF8,43 and MEF2B.37 BCL6
induced in a small subset of LZ B cells that are undergoing orchestrates the GC reaction by negatively modulating the
affinity-based positive selection (Figure 2).30 Given its expression of a diverse set of genes involved in multiple
critical role in almost all proliferating cells, the absence of biological programs, which it recognizes through specific
MYC expression in the GC DZ population has been a motifs in their promoter-proximal ordistal sequences,44
matter of confusion and represents a paradox that is still recruiting distinct co-repressor complexes.45,46
not completely understood.31 However, elegant in vivo Functions critical to the GC B cell phenotype and
studies have shed light into the stage-specific role of MYC controlled by BCL6 include T-cell mediated B cell
during the GC response. In particular, it has been activation,47,48 BCR and CD40 signaling, 49,50 the induction
demonstrated that upregulation of MYC expression in the of apoptosis (eg, by suppression of BCL2),51
LZ is required for antigen selected B cells to re-enter the the sensing and response to DNA damage (eg, by
cell cycle and recirculate to the DZ for additional rounds of suppression of TP53, ATR, CHEK1),52–54 several cytokine
proliferation, SHM and selection.30 This process, known and chemokine signaling pathways,47,48 Notch2
as “cyclic re-entry,” signaling,55 and plasma cell differentiation (via
is critical to sustain affinity maturation and to maintain the suppression of BLIMP1).56 Additionally, BCL6 regulates
GC reaction.8,30,32 Consistently, conditional deletion of gene transcription by repressing specific microRNAs (eg,
MYC early upon antigenic stimulation abrogates GC miR-155, which has inhibitory roles on AID).57,58 As such,
formation,32 and inhibition of its activity specifically in the BCL6 is critical to establish the hyper-proliferative status
LZ leads to dissolution of established GCs.30 Recent of DZ B cells, while allowing them to tolerate the DNA
elegant work suggests that, in LZ B cells, MYC is breaks associated with SHM and CSR without eliciting cell
induced through BCR and CD40 ligation via NF-κB and cycle arrest and apoptotic responses; moreover, BCL6
FOXO1 activation respectively prevents the transduction of receptor signals that could
lead to premature activation and differentiation prior to the
selection of high affinity B cell clones. Upon completion of
these processes, expression of BCL6 needs to be turned
off in order to license B cell exit from the GC; the
mechanisms responsible for this molecular switch have cells and involves the upregulation of IRF4, a known NF-
been partially elucidated and include at least two signals κB target and a master regulator of terminal B cell
that operate at the translational and transcriptional level differentiation.71,72 At high concentrations, IRF4 can also
respectively: (a) engagement of the BCR by the antigen repress BCL6,59 a mechanism required for termination of
acquired by FDCs, and (b) activation of the CD40 receptor the mature GC programme and induction of the other
by the CD40 ligand present on Tfh-cells. 49,50,59 By plasma cell master regulator, BLIMP1.73 BLIMP1, in turn,
relieving the expression of its target genes, maintains the plasma cell programme by suppressing
downregulation of BCL6 will restore the ability of the cell BCL6 through a negative feedback loop.56 Accordingly, a
to respond to microenvironmental cues that lead to small subpopulation of LZ B cells can be detected in the
termination of the GC reaction. The requirement of BCL6 GC LZ by immunofluorescence or immunohistochemistry
for the GC response was conclusively demonstrated by analysis, which displays nuclear translocation of the NFκB
the lack of GC formation and affinity maturation in Bcl6- subunit RELA and co-expression of IRF4 and BLIMP1, in
deficient mouse models,60,61 a phenotype also induced by the absence of BCL6.49,74 Consistently, conditional GC-
deletion of a highly interactive, GC-specific locus control specific deletion of RelA in mice blocks plasma cell
region upstream of Bcl6.62 differentiation, 75 analogous to the phenotype observed in
Irf4 or Prdm1-deficient mice
FOXO1
EPIGENETIC CONTROL OF THE GC
FOXO1 is a member of the Fox-O family of Forkhead
transcription factors that plays critical roles at defined The highly dynamic nature of the GC reaction requires the
stage transitions during B cell development.63 Within the rapid and coordinated switching between discrete
GC, FOXO1 is almost ubiquitously expressed in the DZ; transcriptional programs that contribute to cell fate
this pattern is in keeping with the low activity in this determination in response to signals delivered by the
compartment of PI3K-AKT signaling, a major negative microenvironment.76 GC B cells must therefore be able
regulator of FOXO1 that is only detectable in the GC LZ.64 to quickly remodel their epigenetic landscape. Indeed,
Indeed, FOXO1 is necessary for the establishment and recent studies showed that, within the GC
maintenance of GC polarity, as well as for the DZ B cell microenvironment, B cells undergo massive
phenotype.64,65 The role of FOXO1 in sustaining the DZ reorganization of the genomic architecture that encodes
program entails its ability to transactivate multiple genes the GC B cell transcriptome.62 This reprogramming
involved in functions typical of DZ B cells, such as the process requires the activity of dedicated
chemokine receptor CXCR4, cell proliferation, and histone/chromatin modifying enzymes that catalyze the
negative modulation of DNA repair, while silencing deposition of specific histone marks associated with open
signaling pathways characteristic of the GC LZ program or close chromatin. The identification of recurrent somatic
(eg, activation and differentiation), in part in cooperation mutations in the genes encoding for these very same
with BCL6.64,65 The importance of this dual activity is enzymes was a major, unanticipated finding of recent
demonstrated by the phenotype of GC-specific Foxo1 null whole exome sequencing efforts in GC B lymphoid
mouse models, which form apparently normal GCs malignancies.
entirely composed of LZ B cells and devoid of DZ gene
programmes.64,65 Intriguingly, Foxo1-negative GC B cells
displayed normal SHM but defective affinity maturation
EZH2
and CSR, suggesting that the AID-mediated SHM
machinery can be activated independently of the DZ Enhancer of zeste homologue 2 (EZH2) is a SET-domain
program.64,65 Histone methyltransferase that silences gene transcription
by trimethylating the lysine 27 residue of histone H3
E2A (TCF3) (H3K27me3) and recruiting the Polycomb Repressive
Complex-2 PRC2).77 Within the mature B cell lineage,
The E2A transcription factor plays an instructive role in EZH2 is expressed specifically in the GC, where it creates
normal B-cell development and GC formation by bivalent promoters that control the transcription of genes
regulating the transcription of several B-cell restricted involved in the negative regulation of cell cycle (CDKN1A)
genes, including those encoding the immunoglobulin and in terminal differentiation (IRF4, PRDM1).78,79 In
heavy and light chains, the negative BCR regulator SHP1 particular, recent studies defined a positive feedback loop
(a key contributor to the absence of BCR signaling in most by which EZH2 enables the characteristic proliferative
DZ GC B cells),66 and CCND3, which is required for the phenotype of GC B cells by suppressing CDKN1A and
proliferative expansion of GC B cells.67,68 Consistent with releasing the expression of E2F1.80 In vivo, loss of Ezh2
its pro-proliferative function, E2A is expressed at higher completely abrogated GC formation, establishing this
levels in DZ B cells8 and its deletion in vivo impaired the methyltransferase as a master regulator of the GC.78,79
expansion of GCs, although this gene appears to be
dispensable during the initial phases of antigen
CREBBP/EP300
activation.69
CREBBP and EP300 are ubiquitously expressed,
The NF-κB-IRF4-BLIMP1 axis pleiotropic regulators of gene expression that catalyze the
addition of acetyl groups to specific lysine residues in both
Activation of the NF-κB signaling cascade downstream of histone and non-histone proteins.81 Of particular
signals emanating from the BCR, CD40 receptor, and relevance to GC B cells, CREBBP has been shown to
TLR,70 represents acritical step in both the initiation and oppose the proto-oncogenic activity of BCL6 by two
the termination of the GC reaction (Figure 2).42,71,72 In mechanisms: (a) the direct acetylation of the BCL6
particular, biochemical evidence and in vivo mouse protein, which prevents the recruitment of HDACs and
models have allowed to reconstruct a critical circuit that is thus impairs its trans-repressor function82,83; and (b)
triggered by CD40:CD40L interaction in a subset of LZ B acetylation of H3K27 at the promoter/enhancer
sequences of virtually all BCL6 target genes, which
facilitates their transcription84,85 (Figure 3). Moreover,
CREBBP-mediated acetylation is an essential
requirement for the activation of the p53 tumor
suppressor, which is itself a target of BCL6.52,86 A
comprehensive characterization of the transcriptional
network modulated by CREBBP in the GC has been
recently obtained and revealed a preferential enrichment
in GC LZ signature genes,84,85 including components of
the BCR and CD40 signaling cascade, master regulators
of terminal differentiation, the CIITA transactivator,
multiple MHC-class II loci, and additional genes involved
in antigen presentation/processing84,85,87 (Figure 3).
CREBBP therefore appears to be particularly important in
the decision making of B cells that exit the GC.

KMT2D

The KMT2D gene (also known as MLL2 or MLL4) encodes


a member of the SET1 family of histone
methyltransferases that facilitates transcription by
predominantly mono-and di-methylating
the lysine at position 4 of histone H3 (H3K4), two
modifications that mark competent enhancers.88 The
genes bound by KMT2D in GC B cells and negatively
influenced in expression upon its deletion play key
roles in B-cell physiology, including the positive regulation
of apoptosis, CD40 signaling, and the control of cell
migration and proliferation. 89,90 Intriguingly, loss of Kmt2d
in CD19-Cre conditional knock-out
models, that is, before antigen encounter by naive B cells,
leads to a significant expansion of the GC B cell
population. These data suggest a role for this
methyltransferase in the commitment of mature B cells to
enter the GC reaction, as well as at later stages of GC
differentiation, and support current models of FL
pathogenesis inferring that KMT2D mutations, frequently
observed in FL and DLBCL, are early events in the history
of tumor evolution (see below).

You might also like