ArticlePDF AvailableLiterature Review

Multiple checkpoints keep follicular helper T cells under control to prevent autoimmunity

Authors:

Abstract and Figures

Follicular helper T (Tfh) cells select mutated B cells in germinal centres, which can then differentiate into long-lived high affinity memory B cells and plasma cells. Tfh cells are regulated by a unique molecular programme orchestrated by the transcriptional repressor Bcl6. This transcription factor turns down expression of multiple genes, including transcriptional regulators of other T helper lineages and a vast amount of microRNAs. This enables Tfh cells to express a suite of chemokine receptors, stimulatory ligands and cytokines that enable migration into B-cell follicles, and provision of effective help to B cells. Not surprisingly, dysregulation of this powerful helper subset can lead to a range of autoantibody-mediated diseases; indeed, aberrant accumulation of Tfh cells has been linked with systemic lupus erythematosus, Sjogren's disease and autoimmune arthritis. Here we dissect multiple checkpoints that operate throughout Tfh cell development and maturation to maintain immunological tolerance while mounting robust and long-lasting antibody responses.
Content may be subject to copyright.
REVIEW
Multiple checkpoints keep follicular helper T cells under
control to prevent autoimmunity
Di Yu
1
and Carola G Vinuesa
2
Follicular helper T (Tfh) cells select mutated B cells in germinal centres, which can then differentiate into long-lived high affinity
memory B cells and plasma cells. Tfh cells are regulated by a unique molecular programme orchestrated by the transcriptional
repressor Bcl6. This transcription factor turns down expression of multiple genes, including transcriptional regulators of other T helper
lineages and a vast amount of microRNAs. This enables Tfh cells to express a suite of chemokine receptors, stimulatory ligands and
cytokines that enable migration into B-cell follicles, and provision of effective help to B cells. Not surprisingly, dysregulation of this
powerful helper subset can lead to a range of autoantibody-mediated diseases; indeed, aberrant accumulation of Tfh cells has been
linked with systemic lupus erythematosus, Sjogren’s disease and autoimmune arthritis. Here we dissect multiple checkpoints that
operate throughout Tfh cell development and maturation to maintain immunological tolerance while mounting robust and long-lasting
antibody responses.
Cellular & Molecular Immunology (2010) 7, 198–203; doi:10.1038/cmi.2010.18; published online 5 April 2010
Keywords: autoimmunity; germinal centre; Tfh
INTRODUCTION
The enormously diversified repertoire of T-cell receptors (TCRs) cap-
able of recognising innumerable combinations of peptide–major his-
tocompatibility complex molecules, enable the adaptive immune
system to fight any invading pathogen. The trade-off of the random
process of receptor diversification is the unavoidable recognition of
self-antigens with potentially dangerous affinity. The adaptive immune
system faces the constant challenge of maintaining tolerance to self-
components of the body while mounting robust responses to foreign
pathogens. CD4
1
helper T (Th) cells orchestrate mammalian adaptive
immunity and are subjected to several tolerance checkpoints from their
earlier developmental stages in the thymus, through to their matura-
tion in the periphery and differentiation into effector and memory
subsets. The checkpoints that are common to all CD4
1
helper subsets
include: (i) clonal deletion of self-reactive T cells in the thymus; (ii)
biochemical tuning that limits activation downstream of chronic TCR
signalling from self-reactive receptors; (iii) limiting immunogenic cost-
imuli; (iv) control of the lifespan of the effector populations
1
and (v)
suppression of immune responses by regulatory T (Treg) cells.
2
Defects
in any of these tolerance mechanisms can lead to a wide range of
systemic and organ-specific autoimmune diseases. Further tolerance
mechanisms exist to control the accumulation and function of indi-
vidual CD4
1
helper subsets. Those that pertain to follicular helper T
(Tfh) cells have emerged as being critical to prevent autoantibody-
driven autoimmune diseases and are the subject of this review.
Tfh cells were originally identified as a subset of Th cells found to
reside in close proximity to follicular dendritic cells within germinal
centres.
3
Soon after, their phenotype was described to differ from that
of Th1 and Th2 cells.
4
It took more than 20 years to place Tfh cells in a
subset separate from other helper subsets: the recent identification of
Bcl6 as the transcriptional regulator of Tfh cells has firmly established
these cells as an independent lineage. This was supported by the
demonstration that Th1, Th2 and Th17 cells develop normally in
the absence of Bcl6 but Tfh cells were completely dependent on this
transcription factor.
5–7
The unique ability of Tfh cells to select
mutated high affinity B cells destined to live for decades in an indi-
vidual places them at a critical vulnerable spot for immunological
tolerance.
The process of somatic hypermutation typically occurs in germinal
centres—although it can also occur at extrafollicular sites in auto-
immune-prone mice—and targets immunoglobulin (Ig) variable
region genes of rapidly dividing germinal centre B cells (centroblasts).
8
This can lead to an increase in the affinity of the B-cell receptor for the
immunising antigen, but there is abundant evidence that this stoch-
astic process can also generate self-reactive specificities.
8
Furthermore,
once self-reactive B cells have been vaguely selected in germinal cen-
tres, their differentiated offspring can live and produce antibodies
unchecked, subject to virtually no further control. It has been long
known that most anti-double stranded DNA antibodies detected in
humans and in animal models of systemic lupus erythematosus (SLE)
are high-affinity IgG antibodies, which suggests that they may have
been generated in germinal centre reactions.
9
Thus, a tightly con-
trolled process of germinal centre B-cell selection by antigen-specific
Tfh cells is normally in place to ensure positive selection of those cells
1
Department of Immunology and Inflammation, Garvan Institute of Medical Research, Sydney, NSW, Australia and
2
Department of Immunology and Genetics, John Curtin School
of Medical Research, Canberra, ACT, Australia
Correspondence: Dr D Yu, Department of Immunology and Inflammation, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia.
E-mail: d.yu@garvan.org.au
Received 3 February 2010; accepted 28 February 2010
Cellular & Molecular Immunology (2010) 7, 198–203
ß
2010 CSI and USTC. All rights reserved 1672-7681/10 $32.00
www.nature.com/cmi
with the highest affinity towards foreign antigens while preventing
selection of cells that have become self-reactive. The randomness of
the mutation process, together with the longevity of post-germinal
centre memory B cells and plasma cells, and the abundance of exposed
nuclear antigens on the surface of numerous germinal centre cells
undergoing apoptosis, rank germinal reactions the highest in the risk
of triggering and maintaining autoantibody-driven autoimmunity.
8
While Tfh cells are to a large extent responsible for maintaining
tolerance in germinal centers, it is important to note this is not the
only mechanism operating at this stage of immune responses. Mutated
germinal centre B cells that fail to receive prosurvival signals are pro-
grammed to die by apoptosis, providing a mechanism that prevents
the production of high-affinity, long-lived self-reactive B cells.
10
A
critical checkpoint that can rapidly eliminate self-reactive germinal
centre B cells is the need to establish an immunological synapse with
follicular dendritic cells: germinal centre B cells that bind soluble
antigen without receiving integrin-mediated signals from follicular
dendritic cells die very rapidly—within 4–6 h.
11,12
In mice that lack
Dock8, required to accumulate intercellular adhesion molecule-1 in
the B-cell immunological synapse, germinal centres are short-lived.
13
This highlights the need for antigen to be presented in the form of
immune complexes, which will readily happen in the case of foreign
antigens, but not self-antigens. Germinal centre B cells then need to be
selected by Tfh cells.
What are the unique features of Tfh cells that ensure that not only
are they completely devoid of self-reactive specificities, but also can
ruthlessly eliminate all germinal centre B cells that have acquired
crossreactivity against self-antigens during the process of SHM? The
answer partly lies in: (i) the Tfh phenotype that facilitates follicular
homing and B-cell helper function depends on a sophisticated gene
expression programme controlled by Bcl6 that represses multiple tar-
get genes including several transcription factors and microRNAs
(miRNAs); (ii) strong TCR signalling, which in the presence of normal
thymic selection is likely to occur only in response to foreign antigens,
favours Tfh differentiation; (iii) Tfh development is dependent on the
ability to form highly stable cognate T–B cell interactions; (iv) Tfh cells
turn down production of potentially proinflammatory cytokines such
as interferon-c(IFN-c) and interleukin (IL)-17, and rely on the
actions of IL-21 and IL-4 that cooperate with CD40 ligand (CD40L)
to maintain germinal centres; (v) Tfh cells, like other T-helper subsets,
are also subject to Treg cell control, and (vi) Tfh cells and germinal
centre B cells turn on a proapoptotic programme that limits their
survival. When any of these features destined to promote self-tol-
erance and prevent autoimmunity, goes awry, Tfh cells can aberrantly
accumulate and/or aberrantly select self-reactive germinal centre B
cells. The key regulatory role of Tfh cells in germinal centres has placed
this population in the frontline of immunological tolerance. In this
review, we will summarise the emerging evidence for participation of
Tfh cells in autoimmune diseases and examine the known checkpoints
during Tfh development and maturation to prevent autoimmunity. It
is important to emphasise that this field is in its infancy, and there is
great need to increase our understanding of the molecular mechan-
isms that enable Tfh cell-mediated selection of high affinity and non-
self-reactive germinal centre B cells, and control Tfh differentiation
and lifespan.
DEVELOPMENT OF A SELF-REACTIVE TFH CELL
COMPARTMENT
In the thymus, TCRs that bind strongly to self-peptide–MHC com-
plexes trigger the death of thymocytes, a process known as negative
selection. When this process is defective due to defects in self-antigen
presentation, TCR signalling or apoptosis, autoreactive T cells reach
the periphery and cause systemic or organ specific autoimmunity.
1
Failed negative selection of self-reactive T cells resulting in a non-
self tolerant Tfh population has been shown to contribute to the
autoimmune arthritis of K/BxN mice. These mice develop an aggress-
ive form of arthritis that has many of the clinical, histological and
immunological features of rheumatoid arthritis in humans.
14
K/BxN
mice were originally generated by crossing KRN TCR-transgenic mice
on the C57BL6/J genetic background (K/B) with non-obese diabetic
mice. This TCR transgene is specific for a bovine ribonuclease peptide
(RNase 42–56) but was subsequently also found to recognise glucose-
6-phosphate isomerase (GPI), a ubiquitously expressed self-antigen
that is also present on the surface of inflamed joints, presented by the
MHC class II molecule I-A
g714
. KRN CD4
1
T cells escape clonal dele-
tion in the thymus and mature T cells appear in the periphery at 3
weeks of age, albeit in reduced numbers.
15
Primed KRN CD4
1
T cells
provide help to GPI-reactive B cells leading to the production of high
titres of anti-GPI antibodies.
14
Expression of the transgenic TCR is an
absolute requirement for the development of the arthritic phenotype
14
and adoptive transfer of CD4
1
T cells from healthy K/B donors into
mice of BxG7 (C57BL/6J 3B6.H2
g7
F1) genetic background was
sufficient to initiate germinal centre responses, produce anti-GPI
autoantibodies and develop arthritis.
16
Importantly, this model of autoimmune arthritis is autoantibody-
dependent: transfer of K/BxN serum or purified polyclonal GPI anti-
bodies are sufficient to trigger arthritis in Rag-1
2/2
recipient mice.
17
This corroborates that it is the B-cell helper activity of KRN Tfh cells
that triggers the autoimmune process. Furthermore, in the adoptive
transfer model described above, T cells deficient in CXC-motif che-
mokine receptor 5 (CXCR5) expression were precluded from entering
the follicles, an important step during Tfh-cell differentiation,
18
and
essentially lost the capability to induce germinal centre formation,
autoantibody production and arthritis development.
16
ABERRANT TFH ACCUMULATION
Although the signals that drive Tfh formation are not completely
understood, it has been shown that strong TCR signalling and
inducible costimulator (ICOS)-mediated costimulation favour Tfh
differentiation, which also requires a stable signalling lymphocyte
activation molecule-associated protein (SLAM-associated protein,
SAP)-mediated T–B interaction and is promoted by IL-21.
18,19
.
The pathogenic consequences of Tfh cell accumulation in the
absence of immunisation have been demonstrated in sanroque mice,
an N-ethyl-N-nitrosourea-induced strain bearing a homozygous point
mutation in the Roquin gene.
20
Sanroque mice develop a systemic
autoimmune syndrome with many features typical of SLE including
high-affinity anti-double stranded DNA antibodies, focal proliferative
glomerulonephritis with deposition of IgG-containing immune com-
plexes, anaemia and autoimmune thrombocytopenia as well as other
autoimmune manifestations such as lymphadenopathy, splenomeg-
aly, necrotising hepatitis and plasmacytosis.
20
Spontaneous germinal
centre formation is detected in sanroque mice as early as 4 weeks after
birth, accompanied by massive accumulation of CD4
1
T cells in ger-
minal centres. Sanroque CD4
1
cells express high amounts of CXCR5,
programme cell death-1 (PD-1), ICOS and IL-21, characteristic of a
Tfh phenotype.
20
Roquin was found to act T cell-autonomously to
cause Tfh cell accumulation.
21
Adoptively transferred Tfh cells from
sanroque mice into C57BL/6 mice enhanced germinal centre forma-
tion in wild-type recipient mice in the absence of immunisation.
Tfh cells in autoimmunity
D Yu and CG Vinuesa
199
Cellular & Molecular Immunology
Evidence for a causal role of Tfh dysregulation in the autoimmune
phenotype came from the demonstration that sanroque mice made
genetically deficient in SAP, completely abrogated Tfh cell accumula-
tion and formation of spontaneous germinal centers, prevented lupus-
like disease and end-organ damage.
21
Further evidence that sanroque
lupus is of Tfh/germinal center origin came from studies in which the
gene dose of Bcl6—the transcriptional regulator of both germinal
centre B and Tfh cells—was halved: lupus was significantly amelio-
rated in sanroque Bcl6
1/2
mice.
21
Taken together, these studies suggest
that aberrant positive selection by excessive and dysregulated Tfh cells
in sanroque mice is a key factor in the development of systemic auto-
immunity, suggesting that tight control of Tfh-cell numbers and func-
tion is a key checkpoint in the maintenance of immunological
tolerance.
We recently identified an increase of otherwise rare circulating Tfh-
like (cTfh) CXCR5
high
ICOS
high
PD-1
high
CD4
1
T cells in the blood of a
subset of SLE and Sjo
¨gren’s syndrome patients (20–30%). This
‘cTfh
high
’ phenotype was stable over time and closely correlated with
disease severity. Of note, a comparable cTfh population was seen in the
blood of sanroque mice, which correlated with the increase in resident
Tfh within secondary lymphoid organs.
22
Although cTfh cells from
SLE patients do not express high levels of the Tfh transcription factor
Bcl6,
22
the similarity in phenotype suggests that they might be either
Tfh progenitors capable of terminal differentiation into Tfh cells upon
entry into secondary lymphoid tissues, or they derive from Tfh cells
that have emigrating from germinal centres into the circulation. Thus,
excessive Tfh cell formation may be a common feature of at least a
proportion of patients with autoimmune diseases.
Roquin binds to Icos mRNA (unpublished observation) and regu-
lates its stability acting in concert with miRNAs.
23
Wild-type Roquin
represses ICOS post-transcriptionally, but this regulation is impaired
by the presence of mutant Roquin, leading to ICOS overexpression on
sanroque CD4
1
T cells, which contributes in part to the accumulation
of Tfh cells.
23
As the cellular and molecular mechanisms responsible
for maintaining tolerance are being deciphered, it is becoming clear
that spatiotemporal control of gene expression underpins most suc-
cessful tolerance mechanisms. miRNAs, as a group of short non-
coding RNAs of about 20–22 nucleotides modulating the stability
and translational efficiency of target mRNAs, present an important
new layer controlling gene expression to prevent different types of
polygenic disorders.
24
miRNAs target immune transcripts to fine-tune
gene expression and turn on negative feedback loops that maintain
the delicate balance between protective versus autoimmune res-
ponses.
25,26
Our recent description of Bcl6-mediated repression of
over 30 miRNAs, including miR-17–92 shown to repress CXCR5
expression,
7
underscores the role of this layer of regulation in Tfh
differentiation. Thus, it is likely that miRNA-regulated checkpoints
also operate to prevent autoimmunity of Tfh-germinal centre origin.
FAILED FOLLICULAR EXCLUSION OF UNSUITABLE TH CELLS
Each Th subset has a distinct cytokine production profile: Th1 cells are
potent IFN-cproducers, Th2 cells mainly secrete IL-4, IL-13 and IL-
15, and Th17 cells produce IL-17F, IL-22 and IL-21.
27
Tfh cells differ
from these subsets by their predominant production of two cytokines,
IL-21 and IL-4,
28–33
with only low amounts of IFN-cand IL-17 being
produced in the follicles.
7,34
We and others have shown that the tran-
scription repressor Bcl6 directs Tfh differentiation and suppresses the
differentiation programme to other Th lineages.
6,7,35
Bcl6 decreases
production of IFN-c, IL-4 and IL-17 probably through the direct
repression of the transcription factors T-box 21, GATA-binding
protein 3 and retinoic acid receptor-related orphan receptor gamma,
thymus-specific isoform.
6,7,35
Of interest, there is a striking negative correlation between express-
ion of CXCR5 and IL-17 in tonsil CD4
1
T cells, which are typically rich
in Th1, Th2, Th17 and Tfh cells.
7
This suggests that Th17 cells are
excluded from follicular entry and unlikely to provide help to germinal
centre B cells. Several lines of experimental evidence support this
notion. First, a pathogenic role of IL-17 in dysregulated antibody
responses and autoimmunity has been recently reported. SLE patients
display increased levels of both IL-17 and B cell-activating factor.
Strikingly, IL-17 alone or in combination with B cell-activating factor
that enhances human B-cell survival, promotes B-cell proliferation
and differentiation into antibody-secreting cells ex vivo.
36
This sug-
gests that exclusion of Th17 cells from B cell follicles would be import-
ant to prevent them from providing non-cognate survival and
differentiation signals to autoreactive germinal centre B cells and
lower the threshold for B-cell selection.
A second line of evidence supporting the idea IL-17 may subvert
tolerance mechanisms in germinal centres leading to autoimmunity
comes from studies of BDX2 mice. BDX2 mice were generated by
inbreeding the intercross progeny of C57BL/6J and DBA/2J mice for
more than 20 generations.
37
These mice show rising titres of autoan-
tibodies and circulating immune complexes and progressively develop
glomerulonephritis and erosive arthritis with age.
38
The pathogenic
autoantibodies developed in BDX2 mice are largely dependent on T-
cell help and are likely to be the products of germinal centres.
39
The
important, yet surprising observation is the accumulation of Th17
cells in BDX2 mice and the fact many of these Th17 cells localise within
germinal centres.
40
Introduction of two null alleles of IL-17R amelio-
rated the spontaneous formation of germinal centres in BDX2 mice,
whereas administration of exogenous IL-17 induced germinal centre
formation in wild-type mice and exaggerated spontaneous germinal
centre formation and autoantibody production in BDX2 mice.
40
It has
been suggested that excessive IL-17 upregulates the expression of regu-
lator of G-protein signalling 13 and/or 16 of B cells and causes reten-
tion of B cells in germinal centres, allowing them to undergo repeated
rounds of somatic hypermutation.
40
This together with a lower selec-
tion threshold due to the abundance of T cells and provision of
non-cognate survival signals would lead to an environment where
self-reactive B cells are aberrantly selected.
EXCESSIVE HELPER SIGNALS TO B CELLS
Tfh-derived helper signals such as CD40L and IL-21 not only sustain
germinal centres but also are critical to select mutant B cells and enable
them to terminally differentiate into long-lived high affinity memory
or plasma cells.
8
The question is: Do varying amounts of these helper
signals control germinal centre events and determine the threshold for
selection? There is indirect evidence to suggest that this is the case. In
humans, overexpression of CD40L
41,42
and IL-21
43
has been reported
in SLE patients, suggesting a link between excessive B helper signals
and autoimmunity. However, this does not necessarily mean that an
excess of these signals exclusively corrupts germinal centre selection,
since both CD40L and IL-21 play important roles in extrafollicular
antibody responses (see discussion below).
Overexpression of CD40L on T cells enhances thymocyte apoptosis,
which causes thymic atrophy and precludes investigating the func-
tional consequences of excessive expression of CD40L in the peri-
phery.
44
To circumvent this problem, CD40L was overexpressed on
B cells, and shown to provide autocrine helper signals that resulted in a
lupus-like autoimmune disease in mice with spontaneous production
Tfh cells in autoimmunity
D Yu and CG Vinuesa
200
Cellular & Molecular Immunology
of autoantibodies and development of glomerulonephritis with
immune-complex deposition.
45
IL-21 transgenic mice showed a dramatic increase in the number of
post-switch plasma cells and the titres of IgM and IgG1 in sera com-
pared to wild-type littermates.
46
Higher serum levels of IL-21 were
also detected in BSXB-Yaa mice, a mouse model of SLE with the
characteristic lymphadenopathy, splenomegaly, leukocytosis, hyper-
gammaglobulinemia and severe immune complex-mediated glomer-
ulonephritis.
46
Genetic depletion of IL-21R in these mice abrogated
the hypergammaglobulinemia, autoantibody production, renal dis-
ease and premature morbidity in these mice,
47
suggesting an essential
role of IL-21/IL-21R pathway in the pathogenesis of the autoimmune
disorder. It appears that both follicular and extrafollicular T cells are
important producers of IL-21 in these mice.
As mentioned above, the excessive B helper signals contributing to
autoimmunity do not act exclusively in germinal centres, but also
perturb extrafollicular B-cell responses. Arguably, the best example
is the demonstration of the pathogenic role of IL-21 in the MRL/
MpJ-Fas
lpr
(MRL
lpr
) mouse model of SLE, in which mutated auto-
reactive plasmablasts grow in extrafollicualr foci. In these mice, IL-21
contributes to the production of pathogenic autoantibodies.
48
An
extrafollicular population of ICOS
high
P-selectin glycoprotein ligand
1
low
CD4
1
helper T cells appears to be the primary source of CD40L
and IL-21 helper signals supporting extrafollicular development of
IgG plasmablasts.
49
The relationship between these extrafollicular
helper T cells and Tfh cells is still not fully resolved.
CONTROLLING THE LIFESPAN OF TFH CELLS
Most Tfh cells appear to be short-lived effectors with limited prolif-
erative potential once they enter germinal centres, probably due to
decreased expression of antiapoptotic Bcl2 and Bcl
xL
and increased
expression of other proapoptotic Bcl2 family members including
Bcl2-antagonist/killer 1, BH3 interacting domain death agonist and
Bcl2-associated agonist of cell death. Tfh cells also express high
amounts of the tumour-necrosis factor receptor superfamily, member
6 (Fas), and are more sensitive to cell death after TCR stimulation than
naı
¨ve or memory helper T cells.
50,51
Histological staining reveals that,
unlike interfollicular T cells, human tonsillar Tfh cells do not express
Bcl2,
52
which is consistent with the reported role for the Tfh-directing
transcription factor Bcl6 in repressing Bcl2 expression.
53
Over-
expression of Bcl2 in the entire hematopoietic compartment (Bcl2
transgene controlled by Vav gene regulatory sequences) instead of
selectively in B cells (Bcl2 transgene controlled by an IgH enhancer),
results in spontaneous germinal centre formation.
54
The germinal
centre hyperplasia was abrogated by CD4
1
T-cell depletion, suggest-
ing that excessive CD4
1
T cell longevity was the cause of this pheno-
type.
54
Although the entire peripheral T-cell compartment was
expanded in this mice by about five-fold,
54
it is likely that defective
Tfh cell apoptosis is a major contributor to the abnormal formation of
germinal centres.
TREG-MEDIATED SUPPRESSION OF TFH CELLS
Treg cells represent a small but bona fide population of T cells in
germinal centres.
55,56
These Treg cells are capable of suppressing the
effects of Tfh cells and may play a role in repressing Tfh function,
including IL-17 expression by Tfh cells, thus moderating the provi-
sion of help that Tfh cells can provide to B cells within the germinal
centres.
55,57
Of particular interest is the observation that Treg cells, an immu-
nosuppressive population, can differentiate into Tfh cells in the gut of
lymphopenic mice.
58
This conversion appears to only take place in
Peyer’s patches but not in spleen or lymph nodes of the same mice,
demonstrating the need of a unique microenvironment to foster Tfh-
cell differentiation from Treg cells.
58
However, it is possible that cer-
tain conditions or proinflammatory environments may promote Treg
to Tfh-cell conversion in secondary and tertiary lymphoid organs.
Figure 1 Cellular checkpoints that keep Tfh cells under control to maintain tolerance. Individual checkpoints are shown by numbered red crosses: (1) deletionof
immature T cells with high-affinity self-reactive T-cell receptors; (2) limiting extrinsic stimuli and intrinsic signals that promote Tfh differentiation; (3) exclusion of non-
Tfh effector populations from the follicles; (4) maintenance of the threshold for germinal centre B-cell selection; (5) short lifespan and proapoptotic nature of Tfh cells;
(6) suppression of Tfh cells by Treg cells. Tfh, follicular helper T; Treg, regulatory T.
Tfh cells in autoimmunity
D Yu and CG Vinuesa
201
Cellular & Molecular Immunology
Ectopic germinal centres are detected in 30–50% of joints from
patients with rheumatoid arthritis and the salivary glands from
patients with Sjo
¨gren’s syndrome.
59
Whether Treg to Tfh conversion
occurs at those sites or the reasons why it may fail to occur are still not
clear and will be an interesting subject of investigation.
CONCLUDING REMARKS
Tfh cells have recently acquired their own identity in the big Th cell
family. Growing evidence of their important role in maintaining ger-
minal centre tolerance and demonstration that autoimmunity can
arise when they are dysregulated have placed Tfh cells in the limelight
of the pathogenesis of autoantibody-driven autoimmune diseases
(Figure 1). There are still many unanswered questions regarding
Tfh-cell ontogeny, tolerisation, mechanism of action, factors that con-
trol their growth and survival, and their relationship with other helper
and regulatory subsets. The answers to these questions will hold the
key to unravelling how tolerance is maintained during the process of
negative and positive selection in germinal centres. Much of the
information available to date illuminating the checkpoints that pre-
vent autoimmunity of Tfh/germinal centre origin has been obtained
from autoimmune mouse models. The challenge ahead is to translate
these findings into information that is valuable to improve the dia-
gnosis and therapy of patients with autoimmune diseases. This will
need a much better understanding of human Tfh biology in health and
disease.
ACKNOWLEDGEMENTS
We wish to acknowledge constructive discussion with Nina Chevalier, Charles
R. Mackay, Jing He and Zhanguo Li. CGV is supported by a Viertel Senior
Medical Research Fellowship and NHMRC project and programme grants. DY
is supported by a Cancer Institute New South Wales Fellowship, an NHMRC
Fellowships and an NHMRC programme grant to CRM.
1 Goodnow CC, Sprent J, Fazekas de St Groth B, Vinue sa CG. Cellular and genetic
mechanisms of self tolerance and autoimmunity. Nature 2005; 435: 590
597.
2 Wing K, Sakaguchi S. Regulatory T cells exert checks and balances on self tolerance
and autoimmunity. Nat Immunol 2010; 11:7
13.
3 Stein H, Gerdes J, M ason DY. The normal and malignant germinal centre. Clin
Haematol 1982; 11: 531
559.
4 Velardi A, Mingari MC, Moretta L, Grossi CE. Functional analysis of cloned germinal
center CD4
1
cells with natural killer cell-related features. Divergence from typical T
helper cells. J Immunol 1986; 137: 2808
2813.
5 Johnston RJ, Poholek AC, DiToro D, Yusuf I, Eto D, Barnett B et al. Bcl6 and Blimp-1
are reciprocal and antagonistic regulators of T follicular helper cell differentiation.
Science 2009; 325: 1006
1010.
6 Nurieva RI, Chung Y, Martinez GJ, Yang XO, Tanaka S, Matskevitch TD et al. Bcl6
mediates the de velopment of T follicular helper cells. Science 2009; 325:
1001
1005.
7 Yu D, Rao S, Tsai LM, Lee SK, He Y, Sutcliffe EL et al. The transcriptional repressor
Bcl-6 directs T foll icular helper ce ll lineage comm itment. Immu nity 2009; 31:
457
468.
8 Vinuesa CG, Sanz I, Cook MC. Dysregul ation of germinal centre s in autoimmune
disease. Nat Rev Immunol 2009; 9: 845
857.
9 Radic MZ, Weigert M. Genetic and structural evidence for antigen selection of anti-
DNA antibodies. Annu Rev Immunol 1994; 12: 487
520.
10 Strasser A, Bouillet P. The control of apoptosis in lymphocyte selection. Immunol Rev
2003; 193:82
92.
11 Pulendran B, Kannourakis G, Nouri S, Smith KG, Nossal GJ. Soluble antigen can
cause enhanced apoptosis of germinal-centre B cells. Nature 1995; 375: 331
334.
12 Shokat KM, Goodnow CC. Antigen- induced B-ce ll death and elimination during
germinal-centre immune responses. Nature 1995; 375: 334
338.
13 Randall KL, Lambe T, Johnson A, Treanor B, Kucharska E, Domaschenz H et al. Dock8
mutations cripple B cell immunological synapses, germinal centers and long-lived
antibody production. Nat Immunol 2009; 10: 1283
1291.
14 Ditzel HJ. The K/BxN mouse: a model of human inflammatory arthritis. Trends Mol
Med 2004; 10:40
45.
15 Kouskoff V, Korganow AS, Duchatelle V, Degott C, Benoist C, Mathis D. Organ-specific
disease provoked by systemic autoimmunity. Cell 1996; 87: 811
822.
16 Victoratos P, Kollias G. Induction of autoantibody-mediated spontaneous arthritis
critically depends on follicular dendritic cells. Immunity 2009; 30: 130
142.
17 Korganow AS, Ji H, Mangialaio S, Duc hatelle V, Pelanda R, Martin T et al.From
systemic T cel l self-reac tivity to orga n-specific autoimmune disease via
immunoglobulins. Immunity 1999; 10: 451
461.
18 Yu D, Vinuesa CG. What makes Tfh cells special. Trends Immunol 2010; in press.
19 Yu D, Batten M, Mackay CR, King C. Lineage specification and heterogeneity of T
follicular helper cells. Curr Opin Immunol 2009; 21: 619
625.
20 Vinuesa CG, Cook MC, Angelucci C, Athanasopoulos V, Rui L, Hill KM et al. A RING-
type ubiquitin ligase family member required to repress follicular helper T cells and
autoimmunity. Nature 2005; 435: 452
458.
21 Linterman MA, Rigby RJ, Wong RK, Yu D, Brink R, Cannons JL et al. Follicular helper T
cells are required for systemic autoimmunity. J Exp Med 2009; 206: 561
576.
22 Simpson N, Gatenby PA, Wilson A, Malik S, Fulcher DA, Tangye SG et al. Expansion of
circulating T cells resembling follicular helper T cells is a fixed phenotype that
identifies a subset of severe systemic lupus erythematosus. Arthritis Rheum 2010;
62: 234
244.
23 Yu D, Tan AH, Hu X, Athanasopoulos V, Simpson N, Silva DG et al. Roquin represses
autoimmunity by l imiting inducible T-cell co-stimulator messenger RNA. Nat ure
2007; 450: 299
303.
24 Tsai LM, Yu D. M icroRNAs in c ommon diseases and potent ial therapeutic
applications. Clin Exp Pharmacol Physiol 2010; 37: 102
107.
25 Vinuesa CG, Ri gby RJ, Yu D. Logic and extent of miRNA -mediated c ontrol of
autoimmune gene expression. Int Rev Immunol 2009; 28: 112
138.
26 Luo X, Tsai LM, Shen N, Yu D. Evid ence for microRN A-mediated re gulation in
rheumatic diseases. Ann Rheum Dis 2010; 69(Suppl 1): i30
i36.
27 Zhu J, Paul WE. CD4 T cel ls: fates, func tions, and fau lts. Blood 2008; 112:
1557
1569.
28 Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L et al. Generation of T follicular
helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell
lineages. Immunity 2008; 29: 138
149.
29 Vogelzang A, McGuire HM, Yu D, Sprent J, Mackay CR, King C. A fundamental role for
interleukin-21 in the generation of T follicular helper cells . Immunity 2008; 29:
127
137.
30 Fazilleau N, McHeyzer-Williams LJ, Rosen H, McHeyzer-Williams MG. The function of
follicular helper T cells is regulated by the strength of T cell antigen receptor binding.
Nat Immunol 2009; 10: 375
384.
31 King IL, Mohrs M. I L-4-producing CD4
1
T cells in reactive lymph nodes during
helminth infection are T follicular helper cells. J Exp Med 2009; 206: 1001
1007.
32 Reinhardt RL, Liang HE, Locksley RM. Cytokine-secreting follicular T cells shape the
antibody repertoire. Nat Immunol 2009; 10: 385
393.
33 Zaretsky AG, Taylor JJ, King IL, Marshall FA, Mohrs M, Pearce EJ. T follicular helper
cells differentiate from Th2 cells in response to helminth antigens. J Exp Med 2009;
206: 991
999.
34 Ma CS, Suryani S, Avery DT, Chan A, Nanan R, S antner-Nan an B et al. Early
commitment of naive human CD4(1) T cells to the T follicular helper (T(FH)) cell
lineage is induced by IL-12. Immunol Cell Biol 2009; 87: 590
600.
35 Kusam S, Toney LM, Sato H, Dent AL. Inhibition of Th2 differentiation and GATA-3
expression by BCL-6. J Immunol 2003; 170: 2435
2441.
36 Doreau A, Belot A, Bastid J, Riche B, Trescol-Biemont MC, Ranchin B et al. Interleukin
17 acts in synergy wi th B cell-activa ting factor to influence B cell biolog y and
the pathophysiology of systemic lupus erythematosus. Nat Immunol 2009; 10:
778
785.
37 Taylor BA, Wnek C, Kotlus BS, Roemer N, MacTaggart T, Phillips SJ. Genotyping new
BXD recombinant inbred mouse strains and comparison of BXD and consensus maps.
Mamm Genome 1999; 10: 335
348.
38 Hsu HC, Zhou T, Kim H, Barnes S, Yang P, Wu Q et al. Production of a novel class of
polyreactive pathogenic autoantibodies in BXD2 mice causes glomerulonephritis and
arthritis. Arthritis Rheum 2006; 54: 343
355.
39 Hsu HC, Wu Y, Yang P, Wu Q, Job G, Chen J et al. Overexpression of activation-induced
cytidine deaminase in B cells is asso ciated with production of highly pathogenic
autoantibodies. J Immunol 2007; 178: 5357
5365.
40 Hsu HC, Yang P, Wang J, Wu Q, Myers R, Chen J et al. Interleukin 17-producing T
helper cells and interleukin 17 orchestrate autoreactive germinal center development
in autoimmune BXD2 mice. Nat Immunol 2008; 9: 166
175.
41 Desai-Mehta A, Lu L, Ramsey-Goldman R, Datta SK. Hyperexpression of CD40 ligand
by B and T cells in human lupus and its role in pathogenic autoantibody production. J
Clin Invest 1996; 97: 2063
2073.
42 Koshy M, Berger D, Crow MK. Increased expression of CD40 ligand on systemic lupus
erythematosus lymphocytes. J Clin Invest 1996; 98: 826
837.
43 Wong CK, Wong PT, Tam LS, Li EK, Chen DP, Lam CW. Elevated production of B cell
chemokine CXCL13 is correlated with systemic lupus erythematosus disease activity.
J Clin Immunol 2010; 30:45
52.
44 Clegg CH, Rulffes JT, Haugen HS, Hoggatt IH, Aruffo A, Durham SK et al. Thymus
dysfunction and chronic inflammatory disease in gp39 transgenic mice. Int Immunol
1997; 9: 1111
1122.
45 Higuchi T, Aiba Y, Nomura T, Matsuda J, Mochida K, Suzuki M et al. Cutting edge:
ectopic expression of CD40 ligand on B cells induces lupus-like autoimmune disease.
J Immunol 2002; 168:9
12.
46 Ozaki K, Spolski R, Ettinger R, Kim HP, Wang G, Qi CF et al. Regulation of B cell
differentiation and plasma cell generation by IL-21, a novel inducer of Blimp-1 and
Bcl-6. J Immunol 2004; 173: 5361
5371.
Tfh cells in autoimmunity
D Yu and CG Vinuesa
202
Cellular & Molecular Immunology
47 Bubier JA, Sproule TJ, Foreman O, Spolski R, Shaffer DJ, Morse HC 3rd et al. A critical
role for IL-21 receptor signaling in the pathogenesis of systemic lupus erythematosus
in BXSB-Yaa mice. Proc Natl Acad Sci USA 2009; 106: 1518
1523.
48 Herber D, Brown TP, Liang S, Young DA, Collins M, Dunussi-Joannopoulos K. IL-21
has a pathogenic role in a lupus-prone mouse model and its blockade with IL-21R.Fc
reduces disease progression. J Immunol 2007; 178: 3822
3830.
49 Odegard JM, Marks BR, DiPlacido LD, Poholek AC, Kono DH, Dong C et al. ICOS-
dependent extrafollicular helper T cells elicit IgG production via IL-21 in systemic
autoimmunity. J Exp Med 2008; 205: 2873
2886.
50 Johansson-Lindbom B, Ingvarsson S, Borrebaeck CA. Germinal center s regulate
human Th2 development. J Immunol 2003; 171: 1657
1666.
51 Marinova E, Han S, Zheng B. Human germinal center T cells are unique Th cells with
high propensity for apoptosis induction. Int Immunol 2006; 18: 1337
1345.
52 Schenka AA, Muller S, Fournie JJ, Capila F, Vassallo J, Delsol G et al. CD4
1
T cells
downregulate Bcl-2 in germinal centers. J Clin Immunol 2005; 25: 224
229.
53 Saito M, Novak U, Piovan E, Basso K, Sumazin P, Schneider C et al. BCL6 suppression
of BCL2 via Miz1 and its disruption in diffuse large B cell lymphoma. Proc Natl Acad
Sci USA 2009; 106: 11294
11299.
54 Egle A, Harris AW, Bath ML, O’Reilly L, Cory S. VavP-Bcl2 transgenic mice develop
follicular lymphoma preceded by germinal center hyperplasia . Blood 2004; 103:
2276
2283.
55 Lim HW, Hillsamer P, Kim CH. Regulatory T cells can migrate to follicles upon T cell
activation and suppress GC-Th cells and GC-Th cell-driven B cell responses. J Clin
Invest 2004; 114: 1640
1649.
56 Ito T, Hanabuchi S, Wang YH, Park WR, Arima K, Bover L et al. Two functional subsets
of FOXP3
1
regulatory T cells in human thymus and periphery. Immunity 2008; 28:
870
880.
57 Wu HY, Quintana FJ, Weiner HL. Nasal ant i-CD3 antibod y ameliorates lupus by
inducing an IL-10-secreting CD4
1
CD25
2
LAP
1
regulatory T cell and is associated
with down-regulation of IL-17
1
CD4
1
ICOS
1
CXCR5
1
follicular h elper T cells . J
Immunol 2008; 181: 6038
6050.
58 Tsuji M, Komatsu N, Kawamoto S, Suzuki K, Kanagawa O, Honjo T et al. Preferential
generation of follicular B helper T cells from Foxp3
1
T cells in gut Peyer’s patches.
Science 2009; 323: 1488
1492.
59 Aloisi F, Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory diseases. Nat
Rev Immunol 2006; 6: 205
217.
Tfh cells in autoimmunity
D Yu and CG Vinuesa
203
Cellular & Molecular Immunology
... CD4 + T helper cells (Th cells) are classified, mainly, in 5 sub-types: Th1, Th2, follicular T helper cells (Tfh), Th17 and Treg [162]. Each sub-type depends on specific transcription factors for its polarization and to exert its proper functions after specific cytokines and chemokines secretion. ...
Article
Full-text available
Heme oxygenase (HO) enzymes are responsible for the main oxidative step in heme degradation, generating equimolar amounts of free iron, biliverdin and carbon monoxide. HO-1 is induced as a crucial stress response protein, playing protective roles in physiologic and pathological conditions, due to its antioxidant, anti-apoptotic and anti-inflammatory effects. The mechanisms behind HO-1-mediated protection are being explored by different studies, affecting cell fate through multiple ways, such as reduction in intracellular levels of heme and ROS, transcriptional regulation, and through its byproducts generation. In this review we focus on the interplay between HO-1 and immune-related signaling pathways, which culminate in the activation of transcription factors important in immune responses and inflammation. We also discuss the dual interaction of HO-1 and inflammatory mediators that govern resolution and tissue damage. We highlight the dichotomy of HO-1 in innate and adaptive immune cells development and activation in different disease contexts. Finally, we address different known anti-inflammatory pharmaceuticals that are now being described to modulate HO-1, and the possible contribution of HO-1 in their anti-inflammatory effects.
... The homeostasis of T FH cells was initially suggested to occur under the control of apoptosis since human T FH cells express lower levels of Bcl-2 and higher Fas than non-T FH effector cells [42][43][44][45]. ...
Article
Follicular helper T (TFH) cells are a specialized subset of CD4⁺ T cells that support germinal centers in producing high-affinity antibody-secreting and memory B cells in mammals and birds. Therefore, mechanisms have evolved to control the life and death of TFH cells for balanced humoral immunity. Recent studies have collectively revealed at least two programmed cell death pathways, ferroptosis and pyroptosis, which govern TFH cell survival under diverse physiopathological conditions including immunization, infection, gut homeostasis, and autoimmunity. We review major recent advances in our understanding of the context-dependent regulation of TFH cell survival via cell death pathways that are closely connected with cellular metabolism. Such knowledge might be applied to inform new strategies aimed at modulating humoral immunity, potentially including enhancement of vaccine efficacies.
... Autoreactive antibody production is one of the hallmarks of SLE and plays a critical pathogenic role in LN [29]. T FH cells promote autoantibody-producing B cell differentiation in autoimmune diseases. ...
Article
Full-text available
Background Autoantibody production against endogenous cellular components is pathogenic feature of systemic lupus erythematosus (SLE). Follicular helper T (T FH ) cells aid in B cell differentiation into autoantibody-producing plasma cells (PCs). The IL-6 and IL-21 cytokine-mediated STAT3 signaling are crucial for the differentiation to T FH cells. Niclosamide is an anti-helminthic drug used to treat parasitic infections but also exhibits a therapeutic effect on autoimmune diseases due to its potential immune regulatory effects. In this study, we examined whether niclosamide treatment could relieve lupus-like autoimmunity by modulating the differentiation of T FH cells in two murine models of lupus. Methods 10-week-old MRL/ lpr mice were orally administered with 100 mg/kg of niclosamide or with 0.5% methylcellulose (MC, vehicle) daily for 7 weeks. TLR7 agonist, resiquimod was topically applied to an ear of 8-week-old C57BL/6 mice 3 times a week for 5 weeks. And they were orally administered with 100 mg/kg of niclosamide or with 0.5% MC daily for 5 weeks. Every mouse was analyzed for lupus nephritis, proteinuria, autoantibodies, immune complex, immune cell subsets at the time of the euthanization. Results Niclosamide treatment greatly improved proteinuria, anti-dsDNA antibody levels, immunoglobulin subclass titers, histology of lupus nephritis, and C3 deposition in MRL/ lpr and R848-induced mice. In addition, niclosamide inhibited the proportion of T FH cells and PCs in the spleens of these animals, and effectively suppressed differentiation of T FH -like cells and expression of associated genes in vitro. Conclusions Niclosamide exerted therapeutic effects on murine lupus models by suppressing T FH cells and plasma cells through STAT3 inhibition.
... Autoreactive antibody production is one of the hallmarks of SLE and plays a critical pathogenic role in LN [28]. T FH cells promote autoantibody-producing B cell differentiation in autoimmune diseases. ...
Preprint
Full-text available
Background. Autoantibody production against endogenous cellular components is pathogenic feature of systemic lupus erythematosus (SLE). Follicular helper T (TFH) cells aid in B cell differentiation into autoantibody-producing plasma cells (PCs). The IL-6 and IL-21 cytokine-mediated STAT3 signaling are crucial for the differentiation to TFH cells. Niclosamide is an anti-helminthic drug used to treat parasitic infections but also exhibits a therapeutic effect on autoimmune diseases due to its potential immune regulatory effects. In this study, we examined whether Niclosamide treatment could relieve lupus-like autoimmunity by modulating the differentiation of TFH cells in two murine models of lupus. Methods. 10-week-old MRL/lpr mice were orally administered with 100 mg/kg of Niclosamide or with 0.5% methylcellulose (MC, vehicle) daily for 7 weeks. TLR7 agonist, resiquimod was topically applied to an ear of 8-week-old C57BL/6 mice 3 times a week for 5 weeks. And they were orally administered with 100 mg/kg of Niclosamide or with 0.5% MC daily for 5 weeks. Every mouse was analyzed for lupus nephritis, proteinuria, autoantibodies, immune complex, immune cell subsets at the time of the euthanization. Results. Niclosamide treatment greatly improved proteinuria, anti-dsDNA antibody levels, immunoglobulin subclass titers, histology of lupus nephritis, and C3 deposition in MRL/lpr and R848-induced mice. In addition, Niclosamide inhibited the proportion of TFH cells and PCs in the spleens of these animals, and effectively suppressed differentiation of TFH-like cells and expression of associated genes in vitro. Conclusions. Niclosamide exerted therapeutic effects on murine lupus models by suppressing TFH cells and plasma cells through STAT3 inhibition.
... These markers are used in the diagnosis of angioimmunoblastic T-cell lymphoma (AITL), a lymphoma originating from TFH cells. However, pathology slides of such lymphomas also include plasmocytes, immature B lymphocytes, chemokines, immunoglobulins, IL-21, and IL-6, thus originating in the largest part by TFH tumors [26][27][28]. Likewise, peripheral T-cell lymphomas, not otherwise specified (PCTL-NOS) and the follicular PCTL variant are lymphomas with TFH phenotypic T cells [29]. ...
Article
Full-text available
Malignant lymphomas are a heterogeneous group of malignancies that develop both in nodal and extranodal sites. The different tissues involved and the highly variable clinicopathological characteristics are linked to the association between the lymphoid neoplastic cells and the tissues they infiltrate. The immune system has developed mechanisms to protect the normal tissue from malignant growth. In this review, we aim to explain how T lymphocyte-driven control is linked to tumor development and describe the tumor-suppressive components of the resistant framework. This manuscript brings forward a new insight with regard to intercellular and intracellular signaling, the immune microenvironment, the impact of therapy, and its predictive implications. A better understanding of the key components of the lymphoma environment is important to properly assess the role of both B and T lymphocytes, as well as their interplay, just as two legendary boxers face each other in a heavyweight title final, as was the case of Ali versus Foreman.
... Despite the prominent role of Tfh cells in humoral immunity for protection against infectious pathogens, accumulating evidence has demonstrated that deregulated frequency and activity of Tfh may lead to impaired immune tolerance, generation of high-a nity autoantibodies and hence development of antibody-mediated autoimmune diseases [12][13][14][15]. However, anatomical localization of Tfh cells in the secondary lymphoid tissues limits their routine studies in human patients [6,8]. ...
Preprint
Full-text available
Objectives: Follicular helper T (Tfh) cells have critical roles in the development and promotion of humoral immune responses. Accumulating evidence points to alterations in the levels and activity of Tfh which may lead to impaired immune tolerance and development of antibody-mediated autoimmune diseases, such as rheumatoid arthritis (RA). To elucidate the status of Tfh cells in patients with RA, we conducted a systematic review and meta-analysis to assess the proportion and types of Tfh cells in the PB of RA patients. Method: We systematically searched databases of Embase, Pubmed, Web of sciences (WOS), and Scopus to identify the eligible articles. We performed meta-analysis to assess the proportion of Tfh cells in PB of RA patients compared to healthy controls (HCs). We also assessed the proportion of Tfh cells based on different parameters including Tfh definition markers, disease activity status, disease stage, treatment status, serum markers, and the geographical location of the included articles. Results: Regardless of definition markers, the proportion of Tfh cells in RA patients was significantly higher than that in HCs (SMD 0.75, [0.56, 0.93], p<0.0001). Based on subgroup analyses, the proportion of Tfh cells with the individual definitions were also significantly higher in PB of RA patients compared to HCs. Tfh cells proportion in untreated-RA and early-RA patients was markedly greater than HCs, when comparisons were made without considering the definition markers, and also when Tfh cells was defined by the specified definition markers. While the proportion of Tfh cells by all definitions was significantly higher in active- and remission-RA compared to HCs, individual analysis of two definitions, CD4⁺CXCR5⁺ and CD4⁺CXCR5⁺ICOS⁺, didn’t show significant differences. In addition, higher proportion of Tfh cells defined by all definitions as well as a specified definition (CD4⁺CXCR5⁺PD-1high) was observed when S⁺RA and S⁻RA patients were compared to the HCs. Higher Tfh cells proportion in RA patients compared with HCs was observed in the Asian populations (SMD 0.89, [0.68, 1.09], p<0.0001), while there was a trend toward significant in the proportion of Tfh between the two groups in non-Asians (SMD 0.29, [-0.003, 0.59], p=0.052). Conclusion: Altogether, our results demonstrate that circulating Tfh are highly elevated in RA patients and are also significantly increased in untreated-, early- and S⁺-RA patients, underscoring the potential pathogenic role of this cell population in RA and its potential use as a biomarker and a target for RA therapy.
... 32 High NaCl treatment also promotes autoimmunity by TET2-induced DNA demethylation and differentiation of Tfh (follicular T helper) cells, thus accelerating the development of lupus syndromes in an experimental MRL/lpr mouse model 33 and demonstrating the critical role of Tfh cells in the production of autoantibodies and pathogenesis of lupus. [34][35][36][37][38] Dendritic cells are professional antigen-presenting cells that present antigen to T cells and initiate adaptive immunity. [39][40][41] CD11c is an ideal surface marker of murine dendritic cells. ...
Article
Full-text available
The increased incidence of systemic lupus erythematosus (SLE) in recent decades might be related to changes in modern dietary habits. Since sodium chloride (NaCl) promotes pathogenic T cell responses, we hypothesize that excessive salt intake contributes to the increased incidence of autoimmune diseases, including SLE. Given the importance of dendritic cells (DCs) in the pathogenesis of SLE, we explored the influence of an excessive sodium chloride diet on DCs in a murine SLE model. We used an induced lupus model in which bone marrow-derived dendritic cells (BMDCs) were incubated with activated lymphocyte-derived DNA (ALD-DNA) and transferred into C57BL/6 recipient mice. We observed that a high-salt diet (HSD) markedly exacerbated lupus progression, which was accompanied by increased DC activation. NaCl treatment also stimulated the maturation, activation and antigen-presenting ability of DCs in vitro. Pretreatment of BMDCs with NaCl also exacerbated BMDC-ALD-DNA-induced lupus. These mice had increased production of autoantibodies and proinflammatory cytokines, more pronounced splenomegaly and lymphadenopathy, and enhanced pathological renal lesions. The p38 MAPK–STAT1 pathway played an important role in NaCl-induced DC immune activities. Taken together, our results demonstrate that HSD intake promotes immune activation of DCs through the p38 MAPK–STAT1 signaling pathway and exacerbates the features of SLE. Thus, changes in diet may provide a novel strategy for the prevention or amelioration of lupus or other autoimmune diseases.
Article
Introduction:Alterations in the levels and activity of Tfh may lead to impaired immune tolerance and autoimmune diseases. The aim of this study was to investigate the proportion and types of Tfh cells in the peripheral blood (PB) of RA patients. Areas covered:Comprehensive databases were searched for studies evaluating the proportion of Tfh cells in the PB of patients with RA compared to healthy control (HCs). The proportion of Tfh cells in RA patients was significantly higher than in HCs (SMD 0.699, [0.513, 0.884], p < 0.0001). Furthermore, Tfh cells proportion in untreated-RA and early-RA patients was markedly greater than HCs, when comparisons done without considering the definition markers, and also when Tfh cells were defined by the specified definition markers. While the proportion of Tfh cells by all definitions was higher in active-RA compared to HCs, analysis of two definitions, CD4⁺CXCR5⁺ and CD4⁺CXCR5⁺ICOS⁺, didn’t show significant differences. Furthermore, higher proportion of Tfh cells defined by all definitions and a specified definition (CD4⁺CXCR5⁺PD-1high) was observed when S⁺RA compared to S⁻RA patients. Expert opinion:The results demonstrate that circulating Tfh are highly elevated in RA patients highlights its potential use as a biomarker and a target for RA therapy.
Article
Full-text available
The BCL6 proto-oncogene encodes a transcriptional repressor that is required for germinal center (GC) formation and whose deregulation by genomic lesions is implicated in the pathogenesis of GC-derived diffuse large B cell lymphoma (DLBCL) and, less frequently, follicular lymphoma (FL). The biological function of BCL6 is only partially understood because no more than a few genes have been functionally characterized as direct targets of BCL6 transrepression activity. Here we report that the anti-apoptotic proto-oncogene BCL2 is a direct target of BCL6 in GC B cells. BCL6 binds to the BCL2 promoter region by interacting with the transcriptional activator Miz1 and suppresses Miz1-induced activation of BCL2 expression. BCL6-mediated suppression of BCL2 is lost in FL and DLBCL, where the 2 proteins are pathologically coexpressed, because of BCL2 chromosomal translocations and other mechanisms, including Miz1 deregulation and somatic mutations in the BCL2 promoter region. These results identify an important function for BCL6 in facilitating apoptosis of GC B cells via suppression of BCL2, and suggest that blocking this pathway is critical for lymphomagenesis.
Article
Full-text available
Despite the sequencing of the human and mouse genomes, few genetic mechanisms for protecting against autoimmune disease are currently known. Here we systematically screen the mouse genome for autoimmune regulators to isolate a mouse strain, sanroque, with severe autoimmune disease resulting from a single recessive defect in a previously unknown mechanism for repressing antibody responses to self. The sanroque mutation acts within mature T cells to cause formation of excessive numbers of follicular helper T cells and germinal centres. The mutation disrupts a repressor of ICOS, an essential co-stimulatory receptor for follicular T cells, and results in excessive production of the cytokine interleukin-21. sanroque mice fail to repress diabetes-causing T cells, and develop high titres of autoantibodies and a pattern of pathology consistent with lupus. The causative mutation is in a gene of previously unknown function, roquin (Rc3h1), which encodes a highly conserved member of the RING-type ubiquitin ligase protein family. The Roquin protein is distinguished by the presence of a CCCH zinc-finger found in RNA-binding proteins, and localization to cytosolic RNA granules implicated in regulating messenger RNA translation and stability.
Article
How Tregs migrate to GCs, and whether they regulate the helper activity of the T cells in GCs (GC-Th cells) remains poorly understood. We found a T cell subset in human tonsils that displays potent suppressive activities toward GC-Th cell-dependent B cell responses. These Tregs with the surface phenotype of CD4 +CD25 +CD69 - migrate well to CCL19, a chemokine expressed in the T cell zone, but poorly to CXCL13, a chemokine expressed in the B cell zone. This migration toward the T cell-rich zone rapidly changes to trafficking toward B cell follicles upon T cell activation. This change in chemotactic behavior upon activation of T cells is consistent with their switch in the expression of the 2 chemokine receptors CXCR5 and CCR7. CD4 +CD25 +CD69 - Tregs suppress GC-Th cells and GC-Th cell-induced B cell responses such as Ig production, survival, and expression of activation-induced cytosine deaminase. Our results have identified a subset of Tregs that is physiologically relevant to GC-Th cell-dependent B cell responses and a potential regulation mechanism for the trafficking of these Tregs to GCs.
Article
The present distribution and ecology of New Zealand plants is discussed from a historical viewpoint.It is suggested that during the Miocene a southern extension of the New Zealand archipelago supported a cool temperste flora, which gave rise to the present mountain flora after the onset of orogeny and climatic cooling in the Pliocene.As there was scarcely any simultaneous development of a distinctive flora adapted to the dry conditions which prevail to the east of the mountain axis, Cockayne§ opinion that extremely arid Pleistocene climates evoked certain characteristic life forms—notably the divaricating juvenile form of some trees—is considered to be substantially incorrect. That these life forms are adapted to still–existing conditions seems more probable.The broader features of present distribution are explllined in terms of Pleistocene glaciation and subsequent climatic amelioration.From the evidence of endemism and discontinuous distribution, it is concluded that Otago and Southland,Nelson and Marlborough, Auckland,the subantarctic regions and the Chatham Islands are areas where much of the present flora survived during the glaciation,whereas the middle portion of the South Island and the south of the North Island were characterised by extinction.Adjustment of the vegetation to post Pleistocene conditions is still incomplete, and complicated by the effect of continuing climatic fluctuations
Article
The mammalian immune system has an extraordinary potential for making receptors that sense and neutralize any chemical entity entering the body. Inevitably, some of these receptors recognize components of our own body, and so cellular mechanisms have evolved to control the activity of these 'forbidden' receptors and achieve immunological self tolerance. Many of the genes and proteins involved are conserved between humans and other mammals. This provides the bridge between clinical studies and mechanisms defined in experimental animals to understand how sets of gene products coordinate self-tolerance mechanisms and how defects in these controls lead to autoimmune disease.
Article
Mitochondria, semi-autonomous organelles possessing their own genetic system, are commonly accepted to descend from free-living eubacteria, namely hydrogen-producing alpha-proteobacteria. The progressive loss of genes from the primitive eubacterium to the nucleus of the eukaryotic cell is strongly justified by the Muller rachet principle, which postulates that asexual genomes, like mitochondrial ones, accumulate deleterious and sublethal mutations faster than sexual genomes, like the nucleus. According to this principle, the mitochondrial genome would be doomed to death; instead, we observe that the mitochondrial genome has a variable size and structure in the different organisms, though it contains more or less the same set of genes. This is an example of genetic conservation versus structural diversity. From an evolutionary point of view the genetic system of organelles is clearly under strong selective pressure and for its survival it needs to utilize strategies to slow down or halt the ratchet. Anyway, the mitochondrial genome changes with time, and the rate of evolution is different for both diverse regions of the mtDNA and between lineages, as demonstrated in the case of mammalian mt genomes. We report here our data on the evolution of the mitochondrial DNA in mammals which demonstrate the suitability of mtDNA as a molecular tool for evolutionary analyses.
Article
MicroRNA (miRNA), a group of short non-coding RNA of approximately 20-22 nucleotides modulating the stability and translational efficiency of target messenger RNA, present an important new layer controlling gene expression. Hundreds to a thousand miRNA have been identified and are predicted to regulate at least one-third of protein-coding transcripts in the mammalian genome. This study reviews the recent advances reinforcing the awareness that miRNA are key players in rheumatic diseases by regulating major pathogenic molecules, such as tumour necrosis factor, central signal pathways, such as type I interferon pathway and critical immunoregulatory cells, such as regulatory T cells. In animals, blockade of miRNA maturation by the deletion of Dicer or Drasha, interference with miRNA function by the mutation of Roquin and the altered expression of individual miRNA (miR-146a) or miRNA cluster (miR-17-92) all lead to the development of autoimmune diseases. Growing evidence also reveals the differential expression of certain immunity-regulating miRNA in rheumatoid patients. The features of miRNA-mediated regulation, the direction of future miRNA study in rheumatic diseases and the application of miRNA in diagnosis, therapy and prognosis will also be briefly discussed.