ArticlePDF Available

Constitutive CD8 expression drives innate CD8 + T-cell differentiation via induction of iNKT2 cells

Authors:

Abstract and Figures

Temporal down-regulation of the CD8 co-receptor after receiving positive-selection signals has been proposed to serve as an important determinant to segregate helper versus cytotoxic lineages by generating differences in the duration of TCR signaling between MHC-I and MHC-II selected thymocytes. By contrast, little is known about whether CD8 also modulates TCR signaling engaged by the non-classical MHC-I–like molecule, CD1d, during development of invariant natural killer T (iNKT) cells. Here, we show that constitutive transgenic CD8 expression resulted in enhanced differentiation of innate memory-like CD8 ⁺ thymocytes in both a cell-intrinsic and cell-extrinsic manner, the latter being accomplished by an increase in the IL-4–producing iNKT2 subset. Skewed iNKT2 differentiation requires cysteine residues in the intracellular domain of CD8α that are essential for transmitting cellular signaling. Collectively, these findings shed a new light on the relevance of CD8 down-regulation in shaping the balance of iNKT-cell subsets by modulating TCR signaling.
Content may be subject to copyright.
Research Article
Constitutive CD8 expression drives innate CD8
+
T-cell
differentiation via induction of iNKT2 cells
Satoshi Kojo* , Michiko Ohno-Oishi*, Hisashi Wada, Sebastian Nieke, Wooseok Seo , Sawako Muroi, Ichiro Taniuchi
Temporal down-regulation of the CD8 co-receptor after receiving
positive-selection signals has been proposed to serve as an
important determinant to segregate helper versus cytotoxic
lineages by generating differences in the duration of TCR sig-
naling between MHC-I and MHC-II selected thymocytes. By con-
trast, little is known about whether CD8 also modulates TCR
signaling engaged by the non-classical MHC-Ilike molecule,
CD1d, during development of invariant natural killer T (iNKT) cells.
Here, we show that constitutive transgenic CD8 expression
resulted in enhanced differentiation of innate memory-like CD8
+
thymocytes in both a cell-intrinsic and cell-extrinsic manner, the
latter being accomplished by an increase in the IL-4producing
iNKT2 subset. Skewed iNKT2 differentiation requires cysteine
residues in the intracellular domain of CD8αthat are essential for
transmitting cellular signaling. Collectively, these ndings shed a
new light on the relevance of CD8 down-regulation in shaping the
balance of iNKT-cell subsets by modulating TCR signaling.
DOI 10.26508/lsa.202000642 | Received 8 January 2020 | Revised 16 January
2020 | Accepted 17 January 2020 | Published online 24 January 2020
Introduction
The thymus provides a specic microenvironment that supports
development of several types of T cells, including innate-like T cells,
such as invariant natural killer T (iNKT) cells and mucosal-associated
invariant T (MAIT) cells, and conventional T cells. Signaling via the TCR
plays a central role in driving differentiation of both innate-like and
conventional T cells (Hogquist & Jameson, 2014), although the TCR
diversity and the selecting MHCpresenting antigens are quite dif-
ferent between these two types of T cells; innate-like T cells such as
iNKT cells and MAIT cells express invariant TCRs that recognize non-
peptide antigens presented on non-classical MHC, CD1d and MR1,
respectively (Godfrey et al, 2015), whereas conventional T cells ex-
press diverse TCRs recognizing peptide antigens presented on
classical MHC (Hogquist & Jameson, 2014).
Essential roles of CD4/CD8 co-receptors in TCR/MHC interaction
during differentiation of the two major conventional T-cell subsets,
CD4
+
helper and CD8
+
cytotoxic cells, from common precursors,
CD4
+
CD8
+
double-positive (DP) thymocytes, are well characterized.
Thymocytes positively selected by class II MHC molecules (MHC-II
selected thymocytes) develop into CD4
+
CD8
single-positive (SP)
thymocytes that are committed to the helper lineage, whereas
MHC-Iselected thymocytes are directed to become CD4
CD8
+
SP
thymocytes committed to the cytotoxic lineage (Ellmeier et al, 1999).
It has been proposed that differences in the duration of the
positive-selection signal instruct distinct fates in post-selection
thymocytes (Singer et al, 2008). Thus, briefer TCR signals in MHC-
Iselected thymocytes caused by temporal down-regulation of the
CD8 co-receptor guide post-selection thymocytes to differentiate
into CD4
CD8
+
SP thymocytes. On the other hand, persistent TCR
signals in MHC-IIselected thymocytes supported by constitutive
CD4 expression activate a developmental program toward the
helper-lineage T cells via induction of the zing-nger transcription
factor ThPOK (He et al, 2005;Sun et al, 2005) through antagonizing
a transcriptional silencer in the Zbtb7b gene encoding ThPOK (He
et al, 2008;Setoguchi et al, 2008). Therefore, in what is called the
kinetic signaling model, distinct expression kinetics between CD4
and CD8 co-receptors have been proposed to play a key role in
segregating helper and cytotoxic lineages (Singer et al, 2008). In line
with this model, perturbation of positive-selection signaling du-
ration in MHC-IIselected thymocytes re-directs them to become
CD8
+
cytotoxic-lineage cells (Sarafova et al, 2005;Singer et al, 2008;
Adoro et al, 2012). On the other hand, constitutive transgenic CD8
expression guides about 30% of MHC-Iselected thymocytes to
differentiate into CD4
+
cells (Bosselut et al, 2001). One proposed
explanation for the low efciency of such redirected differentiation
was down-regulation of the transgenic CD8αchain that hetero-
dimerized with endogenous CD8βchain.
In addition to TCR signals, cytokines play important roles in
controlling T-cell differentiation in the thymus. Signals by IL-7 are
crucial for the differentiation of CD8 SP thymocytes (McCaughtry
et al, 2012). Recently, IL-4 has been shown to support differentiation
of another type of CD8 SP thymocyte with the characteristics of
both the memory and innate cells, which is referred to as innate
memory-like CD8 T cells (Weinreich et al, 2010). The iNKT2 subset of
iNKT cells produces IL-4 and has been shown to be a major source
Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
Correspondence: ichiro.taniuchi@riken.jp
*Satoshi Kojo and Michiko Ohno-Oishi contributed equally to this work
©2020Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 1of13
on 24 January, 2020life-science-alliance.org Downloaded from http://doi.org/10.26508/lsa.202000642Published Online: 24 January, 2020 | Supp Info:
of IL-4 in the thymic environment. Accordingly, an increase in the
numbers of iNKT2 cells, although they represent only a tiny sub-
population of total thymocytes, has a signicant impact on the
generation of innate memory-like CD8 T cells (Lee et al, 2013). In
addition to iNKT2 cells, iNKT1 cells expressing IFN-γand iNKT17 cells
expressing IL-17 are also differentiated from iNKT precursors
(Constantinides & Bendelac, 2013). However, little is known about
how balanced differentiation of such iNKT-cell subsets is regulated.
In this study, we generated a novel transgenic mouse model
expressing the CD8αβ heterodimer or the CD8αα homodimer in the
absence of endogenous CD8α/CD8βchains and MHC-II molecules
and observed that two-thirds of MHC-Iselected thymocytes dif-
ferentiated into CD4
CD8
+
SP thymocytes, most of which acquired
signatures of innate memory-like CD8 T cells in both cell-intrinsic
and cell-extrinsic manner. The cell-extrinsic mechanism was linked
to results from enhanced differentiation of the iNKT2-cell subset.
Thus, our study sheds new light on the physiological relevance of
down-regulation of the Cd8 gene to ne-tune the balance of iNKT-
cell subsets.
Results
Developmental pathway to CD4
+
T cells from the CD8 SP stage in
Zbtb7b
ΔTE/ΔTE
mice
The initial activation of the Zbtb7b gene upon receiving positive-
selection signals is achieved mainly by a thymic enhancer (TE) (He
et al, 2008;Muroi et al, 2013). Therefore, removal of the TE from the
Zbtb7b locus results in delayed and low-level expression of ThPOK
in newly selected thymocytes (Muroi et al, 2013). However, se-
quential activation of a proximal enhancer restores ThPOK ex-
pression in a later developmental stage (Muroi et al, 2008). The
impaired ThPOK expression kinetics due to loss of the TE in
Zbtb7b
ΔTE/ΔTE
mice results in redirected differentiation of a small
proportion of MHC-IIselected thymocytes into CD4
CD8
+
SP thy-
mocytes (Muroi et al, 2013). On the other hand, enforced ThPOK
expression in CD8
+
T cells was shown to activate some helper-
lineage signature genes (Jenkinson et al, 2007). Given the restored
ThPOK expression in the later developmental stage of MHC-II
selected thymocytes in Zbtb7b
ΔTE/ΔTE
mice, we examined whether
there are MHC-IIselected thymocytes from some developmental
stages toward the CD8-lineage that acquire a CD4
+
CD8
phenotype.
We accomplished this by a fate-mapping approach using an E8I-Cre
transgene, whose expression is driven by the E8I enhancer known
to be activated specically in mature CD4
CD8
+
SP thymocytes
(Ellmeier et al, 1997) and can be monitored by GFP expression from
ires-gfp sequences present in the transgene (Seo et al, 2017). By
crossing the E8I-Cre transgenic mice to a Rosa26
YFP
reporter strain,
about 70% of CD8
+
splenic T cells were marked by YFP expression,
which could be distinguished from the weaker GFP signal derived
from the E8I-Cre transgene (Fig 1A), whereas only 0.02% of CD4
+
splenic T cells were marked by YFP. On the other hand, the per-
centage of YFP-positive CD4
+
T cells was signicantly increased, up
to 0.07% on average, in Zbtb7b
ΔTE/ΔTE
mice (Fig 1A and B). This result
indicates that there exists a developmental pathway toward CD4
+
T
cells in the Zbtb7b
ΔTE/ΔTE
mice from a developmental stage where
the E8I-Cre transgene is activated, which occurs after CD4 down-
regulation in control mice (Seo et al, 2017). Such reversible dif-
ferentiation to CD4
+
T cells after possible loss of CD4 expression was
not compatible with the kinetic signaling model, prompting us to
revisit the relevance of distinct co-receptor expression kinetics in
the CD4
+
helper versus CD8
+
cytotoxic lineage choice.
Differentiation of innate memory-like CD8 T cells under
constitutive CD8αβ expression conditions
The effect of constitutive CD8αβ co-receptor expression in vivo was
examined previously in the presence of the endogenous CD8β
chain. Possible down-regulation of the transgenic CD8αchain,
which could dimerize with the endogenous CD8βchain, was dis-
cussed as a possible reason why most of the MHC-Irestricted cells
were differentiated into CD4-negative cytotoxic T cells in this
transgenic model (Bosselut et al, 2001). To analyze the effect of
constitutive CD8αβ expression in the absence of both endogenous
CD8αand CD8βchains, we generated a mutant Cd8 locus, by se-
quential targeting in ES cells, referred to as Cd8
Δab
, that lacks coding
regions for both CD8αand CD8βchains in the Cd8a and Cd8b1
genes, respectively (Figs 2ACand S1). For transgenic expression of
CD8αand CD8βchains, a cDNA encoding either chain was inserted
into the Rosa26 locus, generating a Rosa26
8a
and Rosa26
8b
locus,
respectively (Fig 2A). Induction of CD8αand CD8βchains from the
Rosa26
8a
and Rosa26
8b
loci upon Cre-mediated excision of the stop
cassette was conrmed by ow cytometry analyses (Fig 2D).
However, the expression level of transgenic CD8α, which is rep-
resented in CD4
+
population at the lower left panel in Fig 2D was
lower than that of endogenous CD8α(CD8 SP population at the
upper left panel in Fig 2D), presumably because of the weak pro-
moter activity in the Rosa26 locus.
We next examined expression kinetics of transgenic CD8αex-
pression in Rosa26
8a/8a
:Cd8
Δab/Δab
:Cd4-Cre mice. As expected,
expression level of transgenic CD8αfrom the Rosa26 locus was
stable and did not shown down-regulation after positive selection
(Fig S2), conrming constitutive expression of CD8 co-receptor. To
examine differentiation of MHC-I selectedthymocytes, we then
generated I-Ab
/
:Rosa26
8a/8b
:Cd8
Δab/Δab
:Cd4-Cre mice, hereafter
referred to as MHC-II
o
:CD8ab-Tg mice. Despite lower transgenic
CD8αβ expression levels, the percentage of mature thymocytes,
dened as the CD24
lo
TCRβ
hi
population, was equivalent between Wt
and MHC-II
o
:CD8ab-Tg mice (Fig 3A). In the mature thymocytes
population of MHC-II
o
:CD8ab-Tg mice, although around 30% cells
acquired CD4 expression, about 70% cells emerged as CD4-negative
cells (Fig 3A). The balance of CD4
+
and CD4
cells was maintained
after their egress from the thymus. In the peripheral lymphoid
tissues, such as spleen, of MHC-II
o
:CD8ab-Tg mice, the αβT-cell
population consisted of about 65% CD4
cells and 35% CD4
+
cells
(Fig S3A). This proportion of CD4
+
to CD4
cells in MHC-II
o
:CD8ab-Tg
mice was similar to that reported in the previous transgenic model
expressing CD8αand CD8βfrom mini-transgenes at equivalent
levels to endogenous CD8αβ (Bosselut et al, 2001), indicating that, in
spite of the lower level of transgenic CD8αβ expression, our CD8αβ
transgenic model is likely to mimic developmental processes in the
previous model.
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 2of13
We next examined expression of Runx3, a central transcription
factor for cytotoxic T-cell development, by using a Runx3
tdTomato
reporter allele (Kojo et al, 2017). CD4
mature thymocytes emerged in
MHC-II
o
:CD8ab-Tg mice expressed Runx3-tdTomato, albeit at a lower
level than that in control CD8
+
T cells in control mice (Fig 3A), in-
dicating that these cells are related to the cytotoxic lineage. To
further characterize the CD4
mature thymocytes in MHC-II
o
:CD8ab-
Tg mice, we performed transcriptome analyses by RNA-seq. Principal
component analyses indicated that CD4
mature thymocytes that
had differentiated in MHC-II
o
:CD8ab-Tg mice were different from
control wild-type CD8 SP thymocytes (Fig S3B). Analyses of differ-
entially expressed genes revealed that Gzmk,Zbtb16,andTbx21
genes were up-regulated in the CD4
mature thymocytes (Fig 3B). We
also conrmed higher expression of Eomes, one of the signature
genes for innate memory-like CD8 T cells (Weinreich et al, 2010), in
CD8 SP thymocyte population of MHC-II
o
:CD8ab-Tg mice in subse-
quent qRT-PCR and ow cytometry analyses (Figs 3B and S3C). CD44
and CD122 are known surface markers for innate memory-like CD8 T
cells. Higher CD44 expression was detected in about half of the CD4
mature thymocytes in MHC-II
o
:CD8ab-Tg mice and CD122 expression
was detected in a half of the CD44
hi
cells (Figs 3C and S3D). An in-
crease in CD44
hi
CD122
hi
cells was also observed in spleen CD4
αβT-
cell population (Fig S3E). These observations demonstrate that half of
the CD4
mature thymocytes that emerge in MHC-II
o
:CD8ab-Tg mice
are innate memory-like CD8 T cells. An increase in the CD44
hi
CD122
hi
subset was also induced by transgenic CD8αβ expression in an MHC-
sufcient background (Fig 3C). We also found that expression of just
the CD8αchain that could function as a CD8αα homodimer resulted
in enhanced differentiation of CD44
hi
CD122
hi
innate memory-like CD8
T cells (Fig 3C). Thus, under lower but constitutive CD8αβ or CD8αα
expression, more than half of the MHC-Iselected cells remain to be
differentiated into CD4
cytotoxic lineagerelated cells with a skewed
differentiation toward innate memory-like CD8
+
cells.
Increase in the iNKT2 subset under constitutive CD8αβ expression
During the analysis of promyelocytic leukaemia zinc nger (PLZF)
expression, we noticed that around 40% of CD4
+
mature thymocytes
expressed a higher level of PLZF than that in innate memory-like CD8 T
cells (Fig 4A). Innate-like CD4
+
T cells, including CD1d-restricted
iNKT cells (Kovalovsky et al, 2008) and MR1-restricted MAIT cells
(Rahimpour et al, 2015), have been shown to express PLZF at high
levels. Interestingly, differentiation of innate memory-like CD8 T cells
in the thymus has been shown to be enhanced by IL-4 secreted by
iNKT2 cells (Verykokakis et al, 2010;Weinreich et al, 2010;Lee et al,
2013). We, therefore, examined CD4
+
PLZF
hi
cells in MHC-II
o
:CD8ab-Tg
mice in terms of iNKT characteristics, such as expression of the in-
variant TCR, which can be specically detected by CD1d dimers loaded
with αGalCer (αGC) (Matsuda et al, 2000). About 30% of CD4
+
mature
thymocyte of MHC-II
o
:CD8ab-Tg micewerestainedwithCD1d-αGC and
were positive for transgenic CD8 expression, whereas only 5% of those
cells were stained in control mice (Figs 4B and S4A). Such iNKT cells
werepresentatasimilarratioinCD4
+
T-cell population in MHC-II
o
mice expressing only endogenous CD8αβ (Fig 4B). On the contrary, in
MHC-IIsufcient background, both frequency and numbers of iNKT
cells were reduced in our CD8ab-Tg mice as was previously reported
(Lantz & Bendelac, 1994), whereas these are restored in MHC-II
decient background (Fig S4A). These observations indicate that iNKT
cells are one of the major CD4
+
subsets arising after elimination of
MHC-IIselected thymocytes. Whereas PLZF expression was not de-
tected in the CD4
+
CD1d-αGC
population in wild-type control mice,
there remained a small proportion of CD1d-αGC
PLZF
+
CD4
+
cells in the
thymus of MHC-II
o
:CD8ab-Tg mice as well as MHC-II
o
mice (Fig 4B).
Although the frequency of iNKT cells was similar between MHC-
II
o
and MHC-II
o
:CD8ab-Tg mice, an increase in innate memory-like
CD8
+
T cells occurred only in MHC-II
o
:CD8ab-Tg mice (Fig 3C). Given
the important role of IL-4 to support innate memory-like CD8
+
T-cell
Figure 1. Differentiation pathway to CD4
+
T cells after E8I activation in Thpok
DTE/DTE
mice.
(A) Histograms showing GFP expression from the E8I-Cre transgene and YFP expression from the Rosa26-STOP-YFP allele of splenic CD4
+
CD8
and CD4
CD8
+
T cells of
mice with indicated genotypes. One representative of at least three experiments. (B) Graph showing summary of the percentage of YFP
+
CD4
+
CD8
splenic T cells of Zbtb7
+/+
,
Zbtb7b
+/ΔTE
,andZbtb7b
ΔTE/ΔTE
mice that are hemizygous for E8I-Cre and Rosa26-STOP-YFP transgene. Mean ± SD. ***P< 0.001 (KruskalWallis test with Dunnsmultiple
comparisons test).
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 3of13
differentiation (Verykokakis et al, 2010;Lee et al, 2013), we asked
whether differentiation of iNKT-cell subsets is different between
MHC-II
o
and MHC-II
o
:CD8ab-Tg mice. After CD24 down-regulation, the
developmental stages of iNKT cells are divided into the CD44
NK1.1
stage 1, the CD44
+
NK1.1
stage2,andtheCD44
+
NK1.1
+
stage 3 (Benlagha
et al, 2002). In the thymus of MHC-II
o
:CD8ab-Tg mice, proportions of
CD44
NK1.1
stage 1 and CD44
+
NK1.1
stage 2 cells were signicantly
increasedcomparedwithcontrolandMHC-II
o
mice and, this was
accompanied by a relative decrease in CD44
+
NK1.1
+
stage 3 cells (Fig
4C). The T-bet
lo
PLZF
hi
iNKT2-cell subset is included in the stage 1 and
stage 2 iNKT-cell population (Constantinides & Bendelac, 2013;Lee
et al, 2013). As expected, the proportion of the T-bet
lo
PLZF
hi
iNKT2-cell
subset was signicantly higher in MHC-II
o
:CD8ab-Tg mice than in wild-
type and MHC-II
o
mice (Fig 4C), which was consistent with an increase
in the number of iNKT2 cells in MHC-II
o
:CD8ab-Tg mice (Fig S4B). We
further conrmed an increase in the iNKT2 subset by using other
Figure 2. Transgenic mouse line expressing CD8αand CD8βchain in the absence of endogenous CD8αand CD8βchains.
(A) A scheme showing structures of Rosa26
8a
, Rosa26
8b
,Cd8, and Cd8
Δab
loci on the mouse chromosome 6. Orange triangles and green ovals represent loxP and FRT
sequences, respectively. hCD2 was inserted to replace the Cd8a locus. Arrows indicate direction of transcription at the Cd8 and Rosa26 loci. PA, polyA adenylation signals;
Neo,neomycin-resistance gene; ires, internal ribosomal entry sites. (B) Gel images of genotyping PCR showing deletion of the second exon at the Cd8b1 locus and insertion
of the hCD2 cDNA at the Cd8a locus. (C, D) Flow cytometry analyses for CD4 and CD8a expression of total thymocytes of Cd8
+/+
and Cd8
Δab/Δab
mice (C) and TCRβ
+
spleen T
cells of Rosa26
8a/+
and Rosa26
8a/+
: CD4-Cre mice (D). (C, D) Histogram showing intracellular staining of CD8βchain in total thymocytes of Cd8
+/+
and Cd8
Δab/Δab
mice (C)
and surface CD8βexpression of spleen CD4
+
T cells of Rosa26
8b/+
:CD4-Cre and Rosa26
8a/8b
: CD4-Cre mice (D). CD8βexpression by wild-type CD4
+
T cells is shown as an open
histogram as a negative control. One representative of two independent experiments.
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 4of13
markers, Plzf and RORγt, that dene iNKT2 subset as Plzf
hi
RORγt
population (Fig S4C). In addition, after ex vivo PMA/ionomycin stim-
ulation, thymic iNKT cells of MHC-II
o
:CD8ab-Tg mice produced more
IL-4 than wild-type control mice (Fig 4D), conrming the increase in
the iNKT-cell subset that functionally produces IL-4. These observa-
tions indicate that constitutive expression of the CD8αβ co-receptor
results in a skewed differentiation toward the iNKT2-cell subset.
Cell-intrinsic and cell-extrinsic mechanisms for innate-like CD8
T-cell development
Our results showed that differentiation of both innate memory-like
CD8 thymocytes and the iNKT2-cell subset is enhanced in MHC-II
o
:
CD8ab-Tg mice. Given the known role of IL-4 secreted by iNKT2 cells
in supporting innate memory-like CD8 thymocyte differentiation
(Verykokakis et al, 2010;Lee et al, 2013), we next tested to what
extents the increase in the iNKT2-cell subset has impacts on innate
memory-like CD8 T-cell differentiation in MHC-II
o
:CD8ab-Tg mice.
To accomplish this aim, we set up mixed bone marrow chimera
experiments in which equal numbers of bone marrow progeni-
tors from CD45.1 wild-type mice and CD45.2 wild-type or MHC-II
o
:
CD8ab-Tg mice were co-injected together into sublethal irradiated
MHC-II
o
host mice. In the host mice that received CD45.1 wild-
type and CD45.2 wild-type bone marrow cells, the percentages
of CD44
+
CD122
+
innate memory-like cells in mature CD4
CD8
+
SP
thymocytes were similar between CD45.1
+
and CD45.2
+
populations
Figure 3. Differentiation of innate-like CD8
+
Tcell
with under constitutive CD8αβ expression.
(A) Flow cytometry analyses for CD4, CD8, TCRβ, CD24,
and Runx3-tdTomato expression by various thymocyte
subsets of mice with the indicated genotypes.
Representative results of more than three
independent analyses. Numbers in the plot indicate the
percentage of cells in each quadrant. Graphs
showing summary of cell numbers of total and mature
thymocytes and the percentage of CD4
+
and CD4
subset
in mature thymocytes. Mean ± SD. ***P< 0.001
(unpaired ttest, two-sided). (B) RNA-seq analyses of
CD8 SP cells from indicated genotypes (left). The top 40
differentially expressed genes observed between Wt
and Tg are shown. Histograms showing protein
expression level of selected genes in indicated
genotypes (right). (C) Dot plots showing CD44 and
CD122 expression in mature CD8 SP thymocyte of the
indicated genotypes (left). Representative results of
more than three independent analyses. Graph
showing summary of percentage of CD44
hi
CD122
+
population in mature thymocytes of indicated
genotypes (right). Mean ± SD. **P< 0.01, ***P< 0.001,
****P< 0.0001 (one-way ANOVA with Tukeys multiple
comparison).
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 5of13
Figure 4. Skewed differentiation of the iNKT2 subset by constitutive CD8αβ expression.
(A) Dot plots and histograms showing CD4, CD8, and PLZF expression in mature thymocytes of the indicated genotypes (left two panels). Representative results of at
least three experiments. Graph showing a summary of ve independent experiments (right). Mean ± SD. ***P< 0.001 (unpaired ttest with Welchs correction, two-sided).
(B) Histograms showing CD1d-αGC anti-PLZF staining of the indicated thymocyte subsets and genotypes (left). Graph showing summary of four independent experiments
(right). Mean ± SD. ***P< 0.001 (one-way ANOVA with Tukeys multiple comparison). (C) Contour and dot plots showing frequencies in various stages and subsets of
thymic iNKT cells from the indicated genotypes (upper). CD44
NK1.1
CD24
stage 1 cells, CD44
+
NK1.1
stage 2 cells, and CD44
+
NK1.1
+
stage 3 cells. T-bet
lo
PLZF
hi
is the iNKT2
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 6of13
andwerelessthan1%inbothpopulation(Fig 5A). In striking contrast,
the percentage of CD45.1
+
CD44
+
CD122
+
CD8α
+
cells were increased to
around 5% when progenitors from CD45.2 MHC-II
o
:CD8ab-Tg mice
were included (Fig 5A). The degree of CD45.1
+
CD44
+
CD122
+
CD8 SP
thymocyte differentiation tended to be correlated with the degree
of chimerism of CD45.2
+
cells to CD45.1
+
cells. These results are
consistent with previous studies showing that iNKT2-mediated
cell-extrinsic mechanisms, such as an IL-4enriched thymic envi-
ronment, support innate memory-like CD8 T-cell differentiation
(Verykokakis et al, 2010;Lee et al, 2013). Although actual involve-
ment of IL-4producing iNKT2-cell subsets in enhanced differen-
tiation of innate memory-like CD8 T cells in MHC-II
o
:CD8ab-Tg needs
to get conrmed by genetics using mice decient for IL-4 or CD1d,
it is likely that skewed differentiation of iNKT2 cells could serve
as a cell-extrinsic mechanism driving innate memory-like CD8 T
differentiation.
Interestingly, in this setting, the percentage of CD44
+
CD122
+
cells
differentiated from CD45.2 MHC-II
o
:CD8ab-Tg progenitors was always
higher (threefold) than that from CD45.1 wild-type cells (Fig 5A). This
higher percentage of CD44
+
CD122
+
cells in the CD45.2
+
population
suggests that besides cell-extrinsic mechanisms such as higher level
of IL-4 produced by iNKT2 cells, constitutive CD8αβ expression ac-
tivates uncharacterized cell-intrinsic mechanisms that makes MHC-
Irestricted cells more prone to activate a developmental program
toward the innate memory-like CD8 thymocyte.
We next examined development of the iNKT-cell subset in the
bone marrow chimera host mice by assessing the ratio of CD45.2
+
cells to CD45.1
+
cells at the three stages of iNKT-cell development.
This was performed individually for each host mouse because iNKT-
cell reconstitution efciency varied in each host. In the mixed
chimera setting of CD45.1 wild-type with CD45.2 wild-type progen-
itors, the ratio of CD45.2
+
cells to CD45.1
+
cells in each of the three
iNKT-cell stages was around one in all host mice (Fig 5B). On the
contrary, the proportion of CD45.2
+
cells relative to CD45.1
+
cells at
the CD44
NK1.1
stage 1 and the CD44
+
NK1.1
stage 2 was signicantly
increased in host mice injected with CD45.1 wild-type and CD45.2
MHC-II
o
:CD8ab-Tg progenitors (Fig 5B). We also conrmed an in-
creased proportion of the T-bet
lo
PLZF
hi
iNKT2-cell subset in this
setting (Fig 5B). These results clearly demonstrate that cell-intrinsic
mechanisms are involved in the skewed differentiation toward
iNKT1 and iNKT2-cell subsets in MHC-II
o
:CD8ab-Tg mice, although
involvement of cell-extrinsic mechanisms is not formally excluded.
Intracellular signaling from the CD8αchain is required for iNKT2
skewing
Our results revealed that constitutive CD8αβ expression enhanced
iNKT2-cell differentiation in at least a cell-intrinsic manner. Recently,
TCR signal strength was shown to be involved in regulating the dif-
ferentiation of iNKT-cell subsets. Thus,strongTCRsignalingpromotes
iNKT2 and iNKT17 development (Tuttle et al, 2018;Zhao et al, 2018),
suggesting that constitutive CD8αβ expression might enhance TCR
signaling during iNKT-cell development, either by aiding in antigen
recognition and/or modulating intracellular TCR signals. Consistent
with this notion, expression level of CD5, an indicator of TCR signal
strength, was higher on thymic iNKT cells that were developed under
constitutive CD8 expression (Fig S4D). iNKT cells are selected by CD1d
molecules on DP thymocytes (Constantinides & Bendelac, 2013).
Contrary to the previous study showing CD1d down-regulation by
transgenic CD8 expression (Engel et al, 2010), there was no signicant
change in CD1d expression levels on DP thymocytes upon transgenic
CD8αα or CD8αβ expression (Fig S4E), suggesting that CD1d expression
level is unlikely to be involved in enhancement of TCR signaling by
transgenic CD8 expression. Two cysteine residues in the CD8αcyto-
plasmic tail are essential for co-receptor function in TCR stimulation
by classical MHC-I via interacting with Lck kinase and recruitment of
LAT (Turner et al, 1990;Hoeveler & Malissen, 1993;Bosselutetal,1999).
We, therefore, tested whether these cysteine residues are necessary
to enhance iNKT2-cell differentiation by generating a Rosa26
8aCA
allele that expresses a mutant CD8αchaininwhichtwocysteine
residues are replaced with alanine (Fig 6A). Unlike in the MHC-II
o
:
CD8ab-Tg mice, the frequency and numbers of mature CD8 SP thy-
mocytes were signicantly decreased in the MHC-II
o
mice expressing
CD8α
CA
mutant protein (MHC-II
o
:CD8a
CA
bTg mice) (Fig 6B), indicating
that co-receptor function supporting conventional MHC-Irestricted
cell development was abrogated by these amino acid replacements.
When we examined the iNKT-cell population, dened as
CD24
lo
TCRβ
+
CD1d-αGC
+
,inMHC-II
o
:CD8a
CA
bTg mice, there was no
increase in the percentage of the iNKT2-cell subset (Fig 6C). These
observations indicate that intracellular signaling through the CD8α
chain is necessary to drive enhanced iNKT2-cell development.
It remains unclear whether CD8αβ or CD8αα interacts directly with
the non-classical MHC-I molecule CD1d. Nevertheless, our results
clearly demonstrated that constitutive CD8αexpression alone is
sufcient to affect iNKT-cell subset differentiation. We then exam-
ined expression kinetics of the endogenous CD8αchain during iNKT-
cell development. At the CD24
hi
CD44
NK1.1
stage 0, most of iNKT-cell
precursors express CD8αand CD4 and more than half of these cells
down-regulate CD8αexpression (Fig S5). During the transition from
CD24
hi
stage 0 to the CD24
lo
CD44
NK1.1
stage 1, CD8αexpression was
efciently down-regulated. The ThPOK transcription factor is es-
sential to terminate Cd8 gene expression in conventional MHC-I
restricted cytotoxic T cells (He et al, 2005;Rui et al, 2012)aswellasin
iNKT cells (Engel et al, 2010). Interestingly, CD8αdown-regulation at
stage 0 seemed to precede ThPOK induction (Fig S5). Thus, similar to
conventional cytotoxic T cells (Muroi et al, 2008), the initial CD8 down-
regulation during iNKT-cell development is independent of ThPOK.
Discussion
Results shown in this study clearly demonstrate that constitutive
expression of CD8αβ or CD8αα inuences the proportion of iNKT-
cell subsets, with skewing to the iNKT2 subset in a cell-intrinsic
subpopulation. One representative result of at least four experiments. Graphs showing summary of at least four experiments (lower). Mean ± SD. *P<0.05,**P< 0.01, ***P<
0.001 (one-way ANOVA with Tukeys multiple comparison). (D) Histogram showing intracellular IL-4 staining in the thymic iNKT population after stimulation with PMA and
ionomycin for 4 h. One representative of two experiments.
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 7of13
Figure 5. Cell-intrinsic and cell-extrinsic mechanisms for differentiation of innate-like CD8
+
T cells.
(A) Gating strategy of the ow cytometry analyses for the frequency of the innate memory-like CD8
+
T-cell population in mixed bone marrow chimera experiments. One
representative result from three independent mice (left). Graphs showing summary of innate CD8
+
T-cell frequencyin the indicated genotypes (right). Mean ± SD. Probability is
calculated using two-tailed unpaired ttest with Welchs correction (right upper panel). The scatter plot indicates the relationships between CD45.2 chimerism and frequency of
innate CD8
+
T cells. r indicates Pearsons correlation coefcient. (B) Dot plots showing iNKT-cell stages and iNKT2 subpopulations in mixed bone marrow chimera experiments
(top). One representative result from three independent mice. Graph showing summary of frequencies of iNKT-cell developmental stages and iNKT2 subpopulation (bottom).
Mean ± SD. Indicated probabilities are calculated using two-tailed unpaired ttest with or without Welchs correction (right upper panel).
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 8of13
manner, generating an IL-4rich thymic microenvironment that
fosters differentiation of innate memory-like CD8
+
T cells. In ad-
dition to the cell-extrinsic mechanism, constitutive CD8αβ ex-
pression somehow conditions MHC-Irestricted cells to become
more prone to acquire innate memory-like signatures in a cell-
intrinsic manner.
Recent studies reported that TCR signal strength inuences
iNKT-cell subset differentiation (Tuttle et al, 2018;Zhao et al, 2018).
Attenuated function of ZAP70, which plays an essential role in early
TCR signaling events via phosphorylation of LAT, impaired differ-
entiation of iNKT2 and iNKT17, but not iNKT1 cells (Tuttle et al, 2018;
Zhao et al, 2018). The expression level of CD5, which parallels
TCR signaling intensity, and induction of Nur77, another marker
reecting TCR strength, is higher in iNKT2 cells (Moran et al, 2011;Lee
et al, 2013;Tuttle et al, 2018). These ndings highlight that strong TCR
signaling promotes iNKT2-cell development, at least in part by
sustaining Egr2 expression, which results in increases of chromatin
accessibility of iNKT2-specic regulatory elements harboring Egr2-
and NFAT-binding motifs (Tuttle et al, 2018). Therefore, it is con-
ceivable that constitutive CD8αβ expression enhances TCR sig-
naling in iNKT precursors. However, a previous study failed to detect
direct binding of CD8αβ to the non-classical MHC-I molecule, CD1d
(Engel et al, 2010), leaving open the question of how CD8αβ en-
hances TCR signaling engaged by CD1d. Our results showed that the
CD8αα homodimer, which has a distinct afnity for conventional
MHC-I and a thymus leukemia antigen (TL), another non-classical
MHC-I molecule, from CD8αβ co-receptor (Leishman et al, 2001;
Gangadharan & Cheroutre, 2004), also functions to enhance iNKT2
development. This suggests that CD8αβ is unlikely to help iNKT
precursors recognize antigen on CD1d as it does for antigens on
classical MHC-I as a co-receptor, although the possibility that
CD8αβ increases binding afnity of invariant TCR with antigen/CD1d
molecules is not formally excluded. Using a retrogenic transgenic
mouse system, Cruz Tleugabulova et al showed that TCR half-life is
more important to modulate iNKT subset differentiation than
avidity (Cruz Tleugabulova et al, 2016). This nding is not only in line
Figure 6. Signals from CD8αchain are essential for iNKT2 skewing.
(A) Schematic structure of the C227/229A mutant CD8a chain inserted in the Rosa2 6 locus. Two cysteine residues at positions 227 and 229 in the CD8αchain cytoplasmic
tail were changed to alanine. (B) Dot plots showing CD24 and TCRβexpression to dene mature thymocytes and CD4 and CD8 expression in mature thym ocytes. Numbers
in the dot plots indicate percentage of cells in the indicated gates (left). Graph showing summary of absolute number of mature CD8 SP thymocytes of mice with indicated
genotype (right). Mean ± SD. ***P< 0.001, ****P< 0.0001 (one-way ANOVA with Tukeys multiple comparison). (C) Dot plot showing T-bet and PLZF expression in thymic
iNKT cells of mice of the indicated genotype (top). Numbers indicate percentage of cells in the indicated gate that denes iNKT2 subset. Graph showing a summary of
iNKT2 frequency among iNKT cells of mice with the indicated genotype (bottom). Mean ± SD. **P< 0.01, ***P< 0.0001 (one-way ANOVA wit h Tukeys multiple comparison). TM,
transmembrane domain.
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 9of13
with the idea that CD8αβ does not increase afnity for antigen/
CD1d complexes but also raise the possibility that CD8αβ stabilizes
the interaction between TCR and antigen/CD1d complexes. On the
other hands, our results show that two cysteine residues within the
intracellular domain of CD8αchain are required for skewed dif-
ferentiation toward the iNKT2 subset. These cysteine residues
function as a docking module for Lck and LAT (Turner et al, 1990;
Hoeveler & Malissen, 1993). Such an intracellular association of
CD8αchain with TCR/CD3 complexes and signaling molecules in the
absence of antigen recognition suggests a possible role of CD8α
chain to strengthen TCR signaling independently of extracellular
binding of CD8αto CD1d. Given the involvement of ZAP70 in shaping
the development of iNKT subsets, the capacity of the CD8αchain to
interact with the molecular machinery in early TCR signaling events
is likely to be essential to strengthen or prolong TCR signaling. At
any rate, given that CD4 is constitutively expressed during iNKT-cell
development and interacts with Lck with higher afnity than CD8α,
it is possible that CD8αmodulates intracellular signaling from
invariant TCR engaged by antigen/CD1d complexes differently from
CD4.
Pioneering studies proposed that CD8αβ expression during
iNKT-cell development inhibits this development through a process
of negative selection (Lantz & Bendelac, 1994). However, emergence
of CD8
+
iNKT cells in ThPOK-decient mice indicated that CD8
expression does not provide full inhibitory signals (Engel et al,
2010). Recently, reduction of iNKT-cell numbers in SLAM family
receptordecient mice was shown to stem from increased apo-
ptosis because of strong TCR signals (Lu et al, 2019). Thus, the SLAM
family receptor functions to attenuate TCR signals to foster iNKT-
cell development, implying that differentiation of iNKT cells is
impaired once TCR signal strength exceeds some thresholds.
Therefore, enhanced TCR signaling by constitutive CD8αβ expres-
sion inhibits differentiation of some iNKT precursors that receive
excessive TCR signals, whereas other iNKT precursors that can
survive under constitutive CD8αβ expression are directed toward
the iNKT2-cell developmental pathway. It is possible that such
iNKT2 cells that emerged under constitutive CD8αβ expression are
exposed to prolonged TCR signals and produce an increased
amount of IL-4, generating the IL-4rich thymic microenvironment
despite a small increase in their numbers.
A previous publication discussed the possibility that down-
regulation of endogenous CD8βchain could be what limits the
redirection of MHC-I selected cells into CD4
+
helper T cells (Bosselut
et al, 2001). However, in the absence of endogenous CD8β, the
proportion of CD4
+
versus CD4
cells was about one-third, which is
similar to that observed in the previous study (Bosselut et al, 2001),
implying that this is not the case. Moreover, one-third of the CD4
+
T
cells are CD1d-restricted iNKT cells, and the rest of the CD4
+
T
population contains a signicant proportion of PLZF
+
innate-like
cells with uncharacterized MHC-restriction. Therefore, at least in
our experimental setting for transgenic CD8αβ expression, most
MHC-Iselected cells differentiate into CD8
+
cytotoxic-lineage cells.
This nding demonstrates that a persistent positive-selection
signal alone is not sufcient to instruct the helper-lineage fate
to MHC-I restricted cells. Thus, our results challenge the model in
which a distinct duration of positive-selection signals between
MHC-Iand MHC-IIselected thymocytes generated by distinct
kinetics of expressions of the CD4 versus CD8αβ co-receptor is
central to mechanisms that govern the helper versus cytotoxic
lineage choice. Our results also revealed that down-regulation of
CD8 plays an essential role in preventing development of innate
memory-like CD8 T cells, rather than in inducing conventional
cytotoxic fate, at least in part by shaping differentiation of iNKT-cell
subsets. By using several experimental systems, disruption of CD4/
MHC-IImediated positive-selection signals had been previously
shown to result in a redirected differentiation of MHC-II restricted
cells into CD8
+
cytotoxic-lineage cells (Sarafova et al, 2005;Adoro et
al, 2012). This genetic evidence supports the concept that persis-
tence of positive-selection signals is necessary to guide MHC-II
selected thymocytes to become the helper-lineage T cells. On the
contrary, whether persistence of MHC-Imediated TCR signaling is
sufcient to instruct the helper-lineage fate has not yet been well
established. Given the lower expression level of transgenic CD8αβ
in this study, it remains possible that the signal strength was not
sufcient to instruct the helper-lineage fate in this setting. How-
ever, the ratio of CD4
+
versus CD4
mature thymocytes in this study
was similar to that observed in mice expressing transgenic CD8αβ
at a level similar to endogenous CD8αβ (Bosselut et al, 2001). In
addition, the lower expression level of transgenic CD8αβ or CD8αα
was sufcient to inuence TCR signals from invariant TCR on iNKT
precursors. Therefore, it is conceivable that persistence of MHC-
Iderived positive-selection signals alone is not sufcient to in-
struct the helper-lineage fate, although it is still important to test
thymocyte fate decision by achieving constitutive CD8αβ expres-
sion at the correct level (Littman, 2016). Our results suggest that
there must be uncharacterized differences in positive-selection
signals between MHC-Iand MHC-IIrestricted thymocytes beyond
duration of signal length. Understanding of such differences in
positive-selection signals engaged by distinct MHCs is extremely
important and awaits experiments in future studies.
The unique kinetics of CD8 expression, known as co-receptor
reversal, was discovered 20 yr ago (Brugnera et al, 2000;Cibotti et al,
2000). CD8 co-receptor reversal is controlled at the transcriptional
level and is accomplished by temporal termination of Cd8 gene
expression in all post-selection thymocytes and sequential Cd8
reactivation specically in the cytotoxic-lineage cells. However, the
molecular mechanisms underlying such CD8 expression with its
unique kinetics remains uncharacterized. Our results revealed a
novel relevance of Cd8 down-regulation to shaping iNKT-cell subset
differentiation, raising the possibility that mechanisms causing Cd8
down-regulation might be evolved to ne-tune innate T-cell dif-
ferentiation in species that lack conventional cytotoxic T cells or in
the extant positive-selection process of cytotoxic T cells by classical
MHC-I molecules. However, expression of the CD8αβ co-receptor on
cytotoxic-lineage cells is benecial to efciently mount adaptive
immune responses by assisting recognition of peptide antigen
presented on MHC-I molecules in the periphery, leading us to
speculate that mechanism(s) that reactivate Cd8 gene might have
been acquired later. In mice, the E8I enhancer, one of six enhancers
in the Cd8 locus, shows CD8
+
cytotoxic lineagespecic activity in a
reporter transgenic assay (Ellmeier et al, 1997). However, CD8αβ co-
receptor expression by MHC-Irestricted cells is maintained in the
absence of the E8I enhancer (Ellmeier et al, 1998), indicating that
compensatory mechanisms operate to maintain CD8αβ co-receptor
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 10 of 13
expression on MHC-Irestricted cytotoxic T cells (Ellmeier et al, 2002).
Such mechanisms for CD8 expression are likely to have been estab-
lished under evolutional pressures to secure MHC-Idependent
pathogen clearance.
Materials and Methods
Mice
Thpok
ΔTE
mice (Muroi et al, 2013), Rosa26-STOP-YFP mice (Srinivas
et al, 2001), and Runx3-tdTomato reporter mice (Kojo et al, 2017)
have been described. I-Aβdecient mice were from Taconic. To
generate a Cd8
Δab
allele, we rst replaced exon 2 in the Cd8b1 gene
with a neomycin resistant gene (neo
r
)anked with FRT sites by
homologous recombination with the targeting vector in MI ES cells,
establishing ES clone 67. We next removed the neo
r
gene from the
ES clone 67 by transient transfection of an FLP recombinase ex-
pression vector and isolated an ES clone harboring the Cd8
Δb
allele,
into which the targeting vector used to generate the Cd8a
h2PA
allele
(Wada et al, 2018) was transfected to replace exon 1 of the Cd8a
gene with an hCD2 cDNA and the neo
r
gene. After isolation of ES
clones that had undergone homologous recombination with the
second targeting vector at the Cd8a gene, we screened them to
determine which allele, Cd8 or Cd8
Δb
, was targeted by analyzing the
type of FLP recombinase-mediated recombination occurring after
transduction of a retrovirus vector encoding FLP, and then isolated
ES clones harboring the Cd8
+/ΔbaCD2N
genotype. After removal of the
neo
r
gene from the Cd8
+/ΔbaCD2N
ES clone by transient transfection
of the FLP recombinase expression vector, ES clones were used to
generate chimera mice by aggregation. To generate the Rosa26
8a
or Rosa26
8b
allele, a cDNA fragment encoding CD8αor CD8βwas
PCR-amplied by RT-PCR using thymus mRNA, sequenced, and
cloned into the AscI site of the CTV vector (#159212; Addgene). A
cDNA encoding the mutant CD8α
CA
chain harboring alanine
substitutions at two cysteine residues in the CD8αcytoplasmic tail
was amplied by overlap PCR. All mice were maintained in the
specic pathogen-free animal facility at the RIKEN IMS, and all
animal procedures were in accordance with institutional guideli nes
for animal care and with the protocol (28-017) approved by the
Institutional Animal Care and Use Committee of RIKEN Yoko-
hama Branch.
Flow cytometry and cell sorting
Single-cell suspensions from the thymus, spleen, and lymph nodes
were prepared by mashing tissues through a 70-μm cell strainer (BD
Bioscience). Single-cell suspensions were stained with the fol-
lowing antibodies purchased from BD Bioscience, eBiosciences, or
BioLegend: CD4 (RM4-5), CD8a (53-6.7), CD24 (M1/69), CD44 (IM7),
CD122 (TM-β1), TCRβ(H57-597), NK1.1 (PK136), CD69 (H1.2F3), and CCR7
(4B12). Murine CD1d-dimers X I (557599; BD Biosciences) obtained
from BD Biosciences were loaded with αGalCer (KRN7000; Funa-
koshi) and were labelled with antimouse IgG1 proximal enhancer
(550083; BD Biosciences) or antimouse IgG1 BV510 (740421; BD
Biosciences). For intracellular staining of transcription factors, the
cells were stained with cell surface molecules, and then xed and
permeabilized using the Transcription Factor Buffer Set (562574; BD
Biosciences). Permeabilized cells were stained with antibodies
from BD Biosciences, eBiosciences, or BioLegend: T-bet (4B10), PLZF
(9E12), Eomes (Dan11mag), and ThPOK (T43-94). To stain intracellular
IL-4, total thymocytes were stimulated with 50 ng/ml in ml of
phorbol 12-myristate 13-acetate (PMA) (Sigma-Aldrich) and 1.5 μMof
ionomycin for 4 h in the presence of BD Golgi Stop (554724; BD
Biosciences). Then, the cells were stained with cell surface mole-
cules and were xed and permeabilized using BD Cytox/Cytoperm
Kit (554714; BD Biosciences). Permeabilized cells were stained
with anti-IL-4 antibody (11B11; BD Biosciences). Multi-color ow
cytometry analysis was performed using a BD FACSCanto II (BD
Bioscience), and data were analyzed using FlowJo (BD Bioscience)
software. Cell subsets were sorted using a BD FACSAria II or III (BD
Biosciences).
Mixed bone marrow chimera
Recipient MHC-II-decient mice were sublethally irradiated at 950
rad and were reconstituted with 5 × 10
6
bone marrow cells from
CD45.2
+
wild-type mice or CD45.2
+
MHC-II
o
:CD8αβ Tg mice with 5 × 10
6
CD45.1
+
bone marrow cells. At 812 wk after transfer, the spleen and
thymus were analyzed by ow cytometry.
Quantitative RT-PCR
Total cellular RNA was extracted from puried cell subsets using
Trizol (Thermo Fisher Scientic) and treated with RNase-free DNase
I (Thermo Fisher Scientic). cDNA was synthesized from total RNA
using the SuperScriptII First Strand Synthesis System (Invitrogen).
Quantitative RT-PCR was performed using the ABI/PRISM 7000
sequence detection system with an internal uorescent TaqMan
probe. Primers for Zbtb7b (He et al, 2005)andEomes (Intlekofer
et al, 2005) were previously described.
RNA-seq
RNA was extracted from sorted CD8
+
and CD4
mature thymocytes of
Wt and MHC-II
o
:CD8αβ Tg mice by TRIzol (Thermo Fisher Scientic),
followed by RNeasy Micro Kit (QIAGEN) according to the manu-
facturers protocols. Sequencing libraries were prepared using a
NEBNext RNA Library Prep Kit for Illumina (E7530; NEB) with Poly(A)
mRNA Magnetic Isolation Module (E7490; NEB) according to the
manufacturers protocol. Single-end 50-bp reads were obtained by
Illumina HiSeq 1500. The reads were mapped by Tophat v2.1.1 onto
the mouse genome mm10 and counts were generated using HTSeq
v.0.6.1. The DESeq2 R package was used to perform principle com-
ponent analysis and to generate the heat map of differentially
expressed gene with normalized counts of wild-type (GSE48138)
and Tg (GSE132059)induplicatesinthisstudy.
Accession codes
GEO: raw sequencing data and processed les, GSE48138 and
GSE132059.
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 11 of 13
Supplementary Information
Supplementary Information is available at https://doi.org/10.26508/lsa.
202000642.
Acknowledgements
We are grateful to T Ishikura for aggregation of ES cells, Y Taniguchi for
mouse genotyping, N Yoza for cell sorting, and Dr Wilfried Ellmeier for critical
reading of the manuscript. This work was supported by the Grants-in-Aid for
Scientic Research (B) (19390118) from JSPS and the Grants-in-Aid for Sci-
entic Research on Innovative Areas (17H05805 and 19H04820) from the
MEXT in Japan (I Taniuchi).
Author Contributions
S Kojo: formal analysis and investigation.
M Ohno-Oishi: formal analysis and investigation.
H Wada: formal analysis and investigation.
S Nieke: methodology.
W Seo: formal analysis.
S Muroi: methodology.
I Taniuchi: formal analysis and writingoriginal draft.
Conict of Interest Statement
The authors declare that they have no conict of interest.
References
Adoro S, McCaughtry T, Erman B, Alag A, Van Laethem F, Park JH, Tai X, Kimura
M, Wang L, Grinberg A, et al (2012) Coreceptor gene imprinting governs
thymocyte lineage fate. EMBO J 31: 366377. doi:10.1038/
emboj.2011.388
Benlagha K, Kyin T, Beavis A, Teyton L, Bendelac A (2002) A thymic precursor
to the NK T cell lineage. Science 296: 553555. doi:10.1126/
science.1069017
Bosselut R, Feigenbaum L, Sharrow SO, Singer A (2001) Strength of signaling
by CD4 and CD8 coreceptor tails determines the number but not the
lineage direction of positively selected thymocytes. Immunity 14:
483494. doi:10.1016/s1074-7613(01)00128-5
Bosselut R, Zhang W, Ashe JM, Kopacz JL, Samelson LE, Singer A (1999)
Association of the adaptor molecule LAT with CD4 and CD8
coreceptors identies a new coreceptor function in T cell receptor
signal transduction. J Exp Med 190: 15171526. doi:10.1084/
jem.190.10.1517
Brugnera E, Bhandoola A, Cibotti R, Yu Q, Guinter TI, Yamashita Y, Sharrow SO,
Singer A (2000) Coreceptor reversal in the thymus: Signaled CD4
+
8
+
thymocytes initially terminate CD8 transcription even when
differentiating into CD8
+
T cells. Immunity 13: 5971. doi:10.1016/s1074-
7613(00)00008-x
Cibotti R, Bhandoola A, Guinter TI, Sharrow SO, Singer A (2000) CD8
coreceptor extinction in signaled CD4
+
CD8
+
thymocytes: Coordinate
roles for both transcriptional and posttranscriptional regulatory
mechanisms in developing thymocytes. Mol Cell Biol 20: 38523859.
doi:10.1128/mcb.20.11.3852-3859.2000
Constantinides MG, Bendelac A (2013) Transcriptional regulation of the NKT
cell lineage. Curr Opin Immunol 25: 161167. doi:10.1016/
j.coi.2013.01.003
Cruz Tleugabulova M, Escalante NK, Deng S, Fieve S, Ereno-Orbea J, Savage PB,
Julien JP, Mallevaey T (2016) Discrete TCR binding kinetics control
invariant NKT cell selection and central priming. J Immunol 197:
39593969. doi:10.4049/jimmunol.1601382
Ellmeier W, Sawada S, Littman DR (1999) The regulation of CD4 and CD8
coreceptor gene expression during T cell development. Annu Rev
Immunol 17: 523554. doi:10.1146/annurev.immunol.17.1.523
Ellmeier W, Sunshine MJ, Losos K, Hatam F, Littman DR (1997) An enhancer
that directs lineage-specic expression of CD8 in positively selected
thymocytes and mature T cells. Immunity 7: 537547. doi:10.1016/
s1074-7613(00)80375-1
Ellmeier W, Sunshine MJ, Losos K, Littman DR (1998) Multiple developmental
stage-specic enhancers regulate CD8 expression in developing
thymocytes and in thymus-independent T cells. Immunity 9: 485496.
doi:10.1016/s1074-7613(00)80632-9
Ellmeier W, Sunshine MJ, Maschek R, Littman DR (2002) Combined deletion of
CD8 locus cis-regulatory elements affects initiation but not
maintenance of CD8 expression. Immunity 16: 623634. doi:10.1016/
s1074-7613(02)00309-6
Engel I, Hammond K, Sullivan BA, He X, Taniuchi I, Kappes D, Kronenberg M
(2010) Co-receptor choice by Vα14iNKT cells is driven by Th-POK
expression rather than avoidance of CD8-mediated negative
selection. J Exp Med 207: 10151029. doi:10.1084/jem.20090557
Gangadharan D, Cheroutre H (2004) The CD8 isoform CD8αα is not a
functional homologue of the TCR co-receptor CD8αβ.Curr Opin
Immunol 16: 264270. doi:10.1016/j.coi.2004.03.015
Godfrey DI, Uldrich AP, McCluskey J, Rossjohn J, Moody DB (2015) The
burgeoning family of unconventional T cells. Nat Immunol 16:
11141123. doi:10.1038/ni.3298
He X, Dave VP, Zhang Y, Hua X, Nicolas E, Xu W, Roe BA, Kappes DJ (2005) The
zinc nger transcription factor Th-POK regulates CD4 versus CD8
T-cell lineage commitment. Nature 433: 826833. doi:10.1038/
nature03338
He X, Park K, Wang H, He X, Zhang Y, Hua X, Li Y, Kappes DJ (2008) CD4-CD8
lineage commitment is regulated by a silencer element at the ThPOK
transcription-factor locus. Immunity 28: 346358. doi:10.1016/
j.immuni.2008.02.006
Hoeveler A, Malissen B (1993) The cysteine residues in the cytoplasmic tail of
CD8 alpha are required for its coreceptor function. Mol Immunol 30:
755764. doi:10.1016/0161-5890(93)90147-4
Hogquist KA, Jameson SC (2014) The self-obsession of T cells: How TCR
signaling thresholds affect fate decisionsand effector function. Nat
Immunol 15: 815823. doi:10.1038/ni.2938
Intlekofer AM, Takemoto N, Wherry EJ, Longworth SA, Northrup JT, Palanivel
VR, Mullen AC, Gasink CR, Kaech SM, Miller JD, et al (2005) Effector and
memory CD8
+
T cell fate coupled by T-bet and eomesodermin. Nat
Immunol 6: 12361244. doi:10.1038/ni1268
Jenkinson SR, Intlekofer AM, Sun G, Feigenbaum L, Reiner SL, Bosselut R
(2007) Expression of the transcription factor cKrox in peripheral CD8 T
cells reveals substantial postthymic plasticity in CD4-CD8 lineage
differentiation. J Exp Med 204: 267272. doi:10.1084/jem.20061982
Kojo S, Tanaka H, Endo TA, Muroi S, Liu Y, Seo W, Tenno M, Kakugawa K, Naoe Y,
Nair K, et al (2017) Priming of lineage-specifying genes by Bcl11b is
required for lineage choice in post-selection thymocytes. Nat
Commun 8: 702. doi:10.1038/s41467-017-00768-1
Kovalovsky D, Uche OU, Eladad S, Hobbs RM, Yi W, Alonzo E, Chua K, Eidson M,
Kim HJ, Im JS, et al (2008) The BTB-zinc nger transcriptional regulator
PLZF controls the development of invariant natural killer T cell
effector functions. Nat Immunol 9: 10551064. doi:10.1038/ni.1641
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 12 of 13
Lantz O, Bendelac A (1994) An invariant T cell receptor alpha chain is used by
a unique subset of major histocompatibility complex class I-specic
CD4
+
and CD4
8
T cells in mice and humans. J Exp Med 180: 10971106.
doi:10.1084/jem.180.3.1097
Lee YJ, Holzapfel KL, Zhu J, Jameson SC, Hogquist KA (2013) Steady-state
production of IL-4 modulates immunity in mouse strains and is
determined by lineage diversity of iNKT cells. Nat Immunol 14:
11461154. doi:10.1038/ni.2731
Leishman AJ, Naidenko OV, Attinger A, Koning F, Lena CJ, Xiong Y, Chang HC,
Reinherz E, Kronenberg M, Cheroutre H (2001) T cell responses
modulated through interaction between CD8αα and the nonclassical
MHC class I molecule, TL. Science 294: 19361939. doi:10.1126/
science.1063564
Littman DR (2016) How thymocytes achieve their fate. J Immunol 196:
19831984. doi:10.4049/jimmunol.1600032
Lu Y, Zhong MC, Qian J, Calderon V, Cruz Tleugabulova M, Mallevaey T, Veillette
A (2019) SLAM receptors foster iNKT cell development by reducing TCR
signal strength after positive selection. Nat Immunol 20: 447457.
doi:10.1038/s41590-019-0334-0
Matsuda JL, Naidenko OV, Gapin L, Nakayama T, Taniguchi M, Wang CR,
Koezuka Y, Kronenberg M (2000) Tracking the response of natural
killer T cells to a glycolipid antigen using CD1d tetramers. J Exp Med
192: 741754. doi:10.1084/jem.192.5.741
McCaughtry TM, Etzensperger R, Alag A, Tai X, Kurtulus S, Park JH, Grinberg A,
Love P, Feigenbaum L, Erman B, et al (2012) Conditional deletion of
cytokine receptor chains reveals that IL-7 and IL-15 specify CD8
cytotoxic lineage fate in the thymus. J Exp Med 209: 22632276.
doi:10.1084/jem.20121505
Moran AE, Holzapfel KL, Xing Y, Cunningham NR, Maltzman JS, Punt J, Hogquist
KA (2011) T cell receptor signal strength in T
reg
and iNKT cell
development demonstrated by a novel uorescent reporter mouse.
J Exp Med 208: 12791289. doi:10.1084/jem.20110308
Muroi S, Naoe Y, Miyamoto C, Akiyama K, Ikawa T, Masuda K, Kawamoto H,
Taniuchi I (2008) Cascading suppression of transcriptional silencers
by ThPOK seals helper T cell fate. Nat Immunol 9: 11131121.
doi:10.1038/ni.1650
Muroi S, Tanaka H, Miyamoto C, Taniuchi I (2013) Cutting edge: Fine-tuning of
Thpok gene activation by an enhancer in close proximity to its own
silencer. J Immunol 190: 13971401. doi:10.4049/jimmunol.1203006
Rahimpour A, Koay HF, Enders A, Clanchy R, Eckle SB, Meehan B, Chen Z,
Whittle B, Liu L, Fairlie DP, et al (2015) Identication of phenotypically
and functionally heterogeneous mouse mucosal-associated
invariant T cells using MR1 tetramers. J Exp Med 212: 10951108.
doi:10.1084/jem.20142110
Rui J, Liu H, Zhu X, Cui Y, Liu X (2012) Epigenetic silencing of Cd8 genes by
ThPOK-mediated deacetylation during CD4 T cell differentiation.
J Immunol 189: 13801390. doi:10.4049/jimmunol.1201077
Sarafova SD, Erman B, Yu Q, Van Laethem F, Guinter T, Sharrow SO,
Feigenbaum L, Wildt KF, Ellmeier W, Singer A (2005) Modulation of
coreceptor transcription during positive selection dictates lineage
fate independently of TCR/coreceptor specicity. Immunity 23: 7587.
doi:10.1016/j.immuni.2005.05.011
Seo W, Muroi S, Akiyama K, Taniuchi I (2017) Distinct requirement of Runx
complexes for TCRβenhancer activation at distinct developmental
stages. Sci Rep 7: 41351. doi:10.1038/srep41351
Setoguchi R, Tachibana M, Naoe Y, Muroi S, Akiyama K, Tezuka C, Okuda T,
Taniuchi I (2008) Repression of the transcription factor Th-POK by
Runx complexes in cytotoxic T cell development. Science 319: 822825.
doi:10.1126/science.1151844
Singer A, Adoro S, Park JH (2008) Lineage fate and intense debate: Myths,
models and mechanisms of CD4- versus CD8-lineage choice. Nat Rev
Immunol 8: 788801. doi:10.1038/nri2416
Srinivas S, Watanabe T, Lin CS, William CM, Tanabe Y, Jessell TM, Costantini F
(2001) Cre reporter strains produced by targeted insertion of EYFP
and ECFP into the ROSA26 locus. BMC Dev Biol 1: 4. doi:10.1186/1471-
213x-1-4
Sun G, Liu X, Mercado P, Jenkinson SR, Kypriotou M, Feigenbaum L, Galera P,
Bosselut R (2005) The zinc nger protein cKrox directs CD4 lineage
differentiation during intrathymic T cell positive selection. Nat
Immunol 6: 373381. doi:10.1038/ni1183
Turner JM, Brodsky MH, Irving BA, Levin SD, Perlmutter RM, Littman DR (1990)
Interaction of the unique N-terminal region of tyrosine kinase p56lck
with cytoplasmic domains of CD4 and CD8 is mediated by cysteine
motifs. Cell 60: 755765. doi:10.1016/0092-8674(90)90090-2
Tuttle KD, Krovi SH, Zhang J, Bedel R, Harmacek L, Peterson LK, Dragone LL,
Lefferts A, Halluszczak C, Riemondy K, et al (2018) TCR signal strength
controls thymic differentiation of iNKT cell subsets. Nat Commun 9:
2650. doi:10.1038/s41467-018-05026-6
Verykokakis M, Boos MD, Bendelac A, Kee BL (2010) SAP protein-dependent
natural killer T-like cells regulate the development of CD8
+
T cells with
innate lymphocyte characteristics. Immunity 33: 203215. doi:10.1016/
j.immuni.2010.07.013
Wada H, Yasmin N, Kakugawa K, Ohno-Oishi M, Nieke S, Miyamoto C, Muroi S,
Taniuchi I (2018) Requirement for intron structures in activating the
Cd8a locus. Proc Natl Acad Sci U S A 115: 34403445. doi:10.1073/
pnas.1718837115
Weinreich MA, Odumade OA, Jameson SC, Hogquist KA (2010) T cells
expressing the transcription factor PLZF regulate the development of
memory-like CD8
+
T cells. Nat Immunol 11: 709716. doi:10.1038/ni.1898
Zhao M, Svensson MND, Venken K, Chawla A, Liang S, Engel I, Mydel P, Day J,
Elewaut D, Bottini N, et al (2018) Altered thymic differentiation and
modulation of arthritis by invariant NKT cells expressing mutant
ZAP70. Nat Commun 9: 2627. doi:10.1038/s41467-018-05095-7
License: This article is available under a Creative
Commons License (Attribution 4.0 International, as
described at https://creativecommons.org/
licenses/by/4.0/).
iNKT2-cell development by CD8 expression Kojo et al. https://doi.org/10.26508/lsa.202000642 vol 3 | no 2 | e202000642 13 of 13
... However, is this the only functional significance of CD8 downregulation? Recent work from our laboratory has shed new light on the novel role of CD8 downregulation in balancing invariant natural killer T (iNKT) cell subset differentiation in mice [20]. In this Opinion, we discuss the significance of kinetic CD8 expression in controlling conventional helper and cytotoxic T cell lineage fates, and in balancing iNKT cell subset differentiation. ...
... From another angle, the effects of constitutive expression of the CD8 co-receptor have been tested with the expectation that persistent TCR signaling might guide MHC-I-restricted cells into CD4 + helper T cells ( Figure 2) [20,29]. Initial approaches using transgenic mouse lines expressing CD8α and CD8β chains under the control of human CD2 promoter/enhancer sequences have resulted in a partial redirection in differentiation of MHC-I-selected cells. ...
... We hypothesized that if the CD8 co-receptor is constitutively expressed in MHC-I-restricted thymocytes, we would expect their differentiation to be redirected towards the CD4 + helper T cell lineage. This result, however, was not observed in all MHC-I-restricted thymocytes constitutively and transgenically expressing the CD8 co-receptor [20]. In fact, persistent TCR signaling via transgenic CD8 promoted the differentiation of a majority of MHC-I-restricted thymocytes into CD4 − T cells, which showed similar transcriptional signatures to those of innate memory-like CD8 + T cells [20]. ...
Article
Full-text available
During mammalian T cell development, CD4⁺CD8⁺ double-positive (DP) thymocytes must make a lineage choice to become either conventional CD4⁺ or CD8⁺ T cells, dependent on their specificity for MHC-II or MHC-I, respectively. Alternatively, DP thymocytes can decide to become innate-like T cells in response to nonclassical MHC-I molecules. A key feature is the downregulation of CD8, which causes transient T cell receptor (TCR) signaling in MHC-I-selected DP thymocytes. Hence, this kinetic signaling model postulates that short or long duration of TCR signals during positive selection can direct the development of cytotoxic or helper T cell lineages. In this opinion article, we discuss the effects of constitutive expression of transgenic CD8 and prolonged TCR signaling on T cell lineage choice in MHC-I selected mouse thymocytes.
... Furthermore, we investigated the effect of CD8 up-regulation on T-cell development. It has been reported that constitutively expressed CD8 can promote most of the MHC-I-restricted thymocytes to develop into innate memory-like CD8 + T cells rather than redirecting them to the CD4 helper T-cell lineage (26). Consistently, we observed increased CD8 + T cells with an innate-memory phenotype expressing CD44 and CXCR3 in KO thymocytes (Fig S4A), and significantly elevated innate-memory scores of the T.CD8 subgroup in scRNA-seq data (Fig S4B). ...
... Instead, an innate-memory phenotype of CD8 + cells was found. This is consistent with a previous study in which CD8 was overexpressed in thymocytes (26). However, more investigation into how Lsd1 regulates CD8 and CD4 expression at the DP to CD4 + CD8 lo stage is needed. ...
Article
Full-text available
The histone demethylase Lsd1 has been shown to play multiple essential roles in mammalian biology. However, its physiological functions in thymocyte development remain elusive. We observed that the specific deletion of Lsd1 in thymocytes caused significant thymic atrophy and reduced peripheral T cell populations with impaired proliferation capacity. Single-cell RNA sequencing combined with strand-specific total RNA-seq and ChIP-seq analysis revealed that ablation of Lsd1 led to the aberrant derepression of endogenous retroelements, which resulted in a viral mimicry state and activated the interferon pathway. Furthermore, the deletion of Lsd1 blocked the programmed sequential down-regulation of CD8 expression at the DP→CD4+CD8lo stage and induced an innate memory phenotype in both thymic and peripheral T cells. Single-cell TCR sequencing revealed the kinetics of TCR recombination in the mouse thymus. However, the preactivation state after Lsd1 deletion neither disturbed the timeline of TCR rearrangement nor reshaped the TCR repertoire of SP cells. Overall, our study provides new insight into the function of Lsd1 as an important maintainer of endogenous retroelement homeostasis in early T-cell development.
... The importance of signaling through the CD8 co-receptor is further demonstrated in a mouse model with constitutive expression of CD8αβ. This results in increased numbers of T IM cells in the thymus, partly caused by extrinsic factors involving other thymic cell subsets and partly through cell-intrinsic mechanism [48]. Furthermore, mice deficient for DOCK2, which contributes to cellular signaling events by activating small G proteins, are more sensitive to TCR stimulation with low-affinity antigens and have increased numbers of T VM cells [10]. ...
Article
Full-text available
Conventional CD8 ⁺ memory T cells develop upon stimulation with foreign antigen and provide increased protection upon re-challenge. Over the past two decades, new subsets of CD8 ⁺ T cells have been identified that acquire memory features independently of antigen exposure. These antigen-inexperienced memory T cells (T AIM ) are described under several names including innate memory, virtual memory, and memory phenotype. T AIM cells exhibit characteristics of conventional or true memory cells, including antigen-specific responses. In addition, they show responsiveness to innate stimuli and have been suggested to provide additional levels of protection toward infections and cancer. Here, we discuss the current understanding of T AIM cells, focusing on extrinsic and intrinsic molecular conditions that favor their development, their molecular definitions and immunological properties, as well as their transcriptional and epigenetic regulation.
Article
Full-text available
Phenotypic markers that denote different developmental stages of thymocytes are important for understanding T cell development in the thymus. Here, we show that CD1b is a critical discriminator of thymocyte maturation stage in cynomolgus monkeys. CD1b was expressed by immature thymocytes prior to β-selection, and its expression decreased as cells became fully mature in the thymus. MHC-I expression was lowest at the CD3loCD1b⁺ immature double-positive (DP) stage, while the ratio of CD1d:MHC-I expression was significantly higher at this stage than at other developmental stages. PLZF was expressed by < 0.2% of thymocytes; most PLZF⁺ thymocytes were CD3-/loCD1b⁺ immature DP thymocytes with the potential to produce IL-4. EOMES⁺ thymocytes, which accounted for > 2% of total thymocytes, were mostly CD3⁺CD1b⁻ mature thymocytes and predominantly of the CD8 single-positive (SP) lineage. An unconventional CD8⁺ T cell subset expressing the NKG2AC⁺CXCR3⁺ innate-like T cell marker was identified within the EOMES⁺ CD8 SP lineage; these cells exhibited a memory phenotype. Taken together, these findings show that CD1b is a valuable discriminatory marker of thymocyte development. The data presented herein can be used to characterize the features of PLZF- and EOMES-associated unconventional T cells in the thymus.
Article
Full-text available
Invariant natural killer T cells (iNKT cells) develop through an incompletely understood process that requires positive selection by CD4 ⁺ CD8 ⁺ double-positive thymocytes and SLAM family receptors (SFRs). Here we found that SFRs promoted the development of iNKT cells by reducing the strength of the T cell antigen receptor (TCR) signal after positive selection. This effect improved the survival of iNKT cells and their responses to antigen. Loss of SFRs upregulated the expression of inhibitory receptors, including PD-1, on iNKT cells to mitigate the deleterious effect of SFR deficiency. The role of SFRs could be mimicked by expression of SLAMF6 alone in SFR-deficient mice, and this involved the adaptor SAP–kinase Fyn complex and the phosphatase SHP-1. Thus, SFRs foster iNKT cell development by attenuating TCR signal strength after positive selection. © 2019, The Author(s), under exclusive licence to Springer Nature America, Inc.
Article
Full-text available
During development in the thymus, invariant natural killer T (iNKT) cells commit to one of three major functionally different subsets, iNKT1, iNKT2, and iNKT17. Here, we show that T cell antigen receptor (TCR) signal strength governs the development of iNKT cell subsets, with strong signaling promoting iNKT2 and iNKT17 development. Altering TCR diversity or signaling diminishes iNKT2 and iNKT17 cell subset development in a cell-intrinsic manner. Decreased TCR signaling affects the persistence of Egr2 expression and the upregulation of PLZF. By genome-wide comparison of chromatin accessibility, we identify a subset of iNKT2-specific regulatory elements containing NFAT and Egr binding motifs that is less accessible in iNKT2 cells that develop from reduced TCR signaling. These data suggest that variable TCR signaling modulates regulatory element activity at NFAT and Egr binding sites exerting a determinative influence on the dynamics of gene enhancer accessibility and the developmental fate of iNKT cells.
Article
Full-text available
Various subsets of invariant natural killer T (iNKT) cells with different cytokine productions develop in the mouse thymus, but the factors driving their differentiation remain unclear. Here we show that hypomorphic alleles of Zap70 or chemical inhibition of Zap70 catalysis leads to an increase of IFN-γ-producing iNKT cells (NKT1 cells), suggesting that NKT1 cells may require a lower TCR signal threshold. Zap70 mutant mice develop IL-17-dependent arthritis. In a mouse experimental arthritis model, NKT17 cells are increased as the disease progresses, while NKT1 numbers negatively correlates with disease severity, with this protective effect of NKT1 linked to their IFN-γ expression. NKT1 cells are also present in the synovial fluid of arthritis patients. Our data therefore suggest that TCR signal strength during thymic differentiation may influence not only IFN-γ production, but also the protective function of iNKT cells in arthritis.
Article
Full-text available
T-lineage committed precursor thymocytes are screened by a fate-determination process mediated via T cell receptor (TCR) signals for differentiation into distinct lineages. However, it remains unclear whether any antecedent event is required to couple TCR signals with the transcriptional program governing lineage decisions. Here we show that Bcl11b, known as a T-lineage commitment factor, is essential for proper expression of ThPOK and Runx3, central regulators for the CD4-helper/CD8-cytotoxic lineage choice. Loss of Bcl11b results in random expression of these factors and, thereby, lineage scrambling that is disconnected from TCR restriction by MHC. Initial Thpok repression by Bcl11b prior to the pre-selection stage is independent of a known silencer for Thpok, and requires the last zinc-finger motif in Bcl11b protein, which by contrast is dispensable for T-lineage commitment. Collectively, our findings shed new light on the function of Bcl11b in priming lineage-specifying genes to integrate TCR signals into subsequent transcriptional regulatory mechanisms.
Article
Full-text available
A TCRβ enhancer, known as the Eβ enhancer, plays a critical role in V(D)J recombination and transcription of the Tcrb gene. However, the coordinated action of trans-acting factors in the activation of Eβ during T cell development remains uncharacterized. Here, we characterized the roles of Runx complexes in the regulation of the Eβ function. A single mutation at one of the two Runx binding motifs within the Eβ severely impaired Tcrb activation at the initiation phase in immature thymocytes. However, TCRβ expression level in mature thymocytes that developed under such a single Runx site mutation was similar to that of the control. In contrast, mutations at two Runx motifs eliminated Eβ activity, demonstrating that Runx complex binding is essential to initiate Eβ activation. In cells expressing Tcrb harboring rearranged V(D)J structure, Runx complexes are dispensable to maintain TCRβ expression, whereas Eβ itself is continuously required for TCRβ expression. These findings imply that Runx complexes are essential for Eβ activation at the initiation phase, but are not necessary for maintaining Eβ activity at later developmental stages. Collectively, our results indicate that the requirements of trans-acting factor for Eβ activity are differentially regulated, depending on the developmental stage and cellular activation status.
Article
Significance Understanding the mechanisms controlling reactivation of the Cd8a gene is important to elucidate molecular mechanisms of lineage commitment toward CD8 ⁺ cytotoxic T cells. By analyzing the expression of hCD2 reporter from manipulated Cd8a alleles with distinct polyadenylation signal positions or intron structures defective in RNA splicing, we found that intron structures are essential for hCD2 expression in CD8 ⁺ T cells. This occurred at least in part through the induction of DNA demethylation at intronic regions. In contrast, Cd8a gene was transcribed in CD8 ⁺ T cells from a mutant Cd8a locus lacking intron structures, albeit at a lower level. Our observations highlight the possible involvement of intron structures in the regulation of Cd8a gene during T cell development.
Article
Invariant NKT (iNKT) cells develop and differentiate in the thymus, segregating into iNKT1/2/17 subsets akin to Th1/2/17 classical CD4(+) T cells; however, iNKT TCRs recognize Ags in a fundamentally different way. How the biophysical parameters of iNKT TCRs influence signal strength in vivo and how such signals affect the development and differentiation of these cells are unknown. In this study, we manipulated TCRs in vivo to generate clonotypic iNKT cells using TCR retrogenic chimeras. We report that the biophysical properties of CD1d-lipid-TCR interactions differentially impacted the development and effector differentiation of iNKT cells. Whereas selection efficiency strongly correlated with TCR avidity, TCR signaling, cell-cell conjugate formation, and iNKT effector differentiation correlated with the half-life of CD1d-lipid-TCR interactions. TCR binding properties, however, did not modulate Ag-induced iNKT cytokine production. Our work establishes that discrete TCR interaction kinetics influence iNKT cell development and central priming.
Article
One of the central principles of adaptive immunity is the subdivision of T lymphocytes into functionally distinct subsets. Although multiple T cell subsets were shown in recent years to emerge from the thymus, most mature T cells consist of MHC class II (MHC II)–restricted CD4+ T helper cells and
Article
While most studies of T lymphocytes have focused on T cells reactive to complexes of peptide and major histocompatibility complex (MHC) proteins, many other types of T cells do not fit this paradigm. These include CD1-restricted T cells, MR1-restricted mucosal associated invariant T cells (MAIT cells), MHC class Ib-reactive T cells, and γδ T cells. Collectively, these T cells are considered 'unconventional', in part because they can recognize lipids, small-molecule metabolites and specially modified peptides. Unlike MHC-reactive T cells, these apparently disparate T cell types generally show simplified patterns of T cell antigen receptor (TCR) expression, rapid effector responses and 'public' antigen specificities. Here we review evidence showing that unconventional T cells are an abundant component of the human immune system and discuss the immunotherapeutic potential of these cells and their antigenic targets.