You are on page 1of 12

RESEARCH ARTICLE | IMMUNOLOGY AND INFLAMMATION

SRC2 controls CD4+ T cell activation via stimulating


c-Myc-mediated upregulation of amino acid transporter Slc7a5
Wencan Zhanga,1 , Xu Caob,1 , Xiancai Zhonga , Hongmin Wua, Yun Shia, Mingye Fengb, Yi-Chang Wangc, David Annc, Yousang Gwackd ,
Yate-Ching Yuane , Weirong Shangf , and Zuoming Suna,2

Edited by Douglas Green, St Jude Children’s Research Hospital, Memphis, TN; received December 16, 2022; accepted March 16, 2023

T cell activation stimulates substantially increased protein synthesis activity to accu-


mulate sufficient biomass for cell proliferation. The protein synthesis is fueled by the Significance
amino acids transported from the environment. Steroid nuclear receptor coactivator
2 (SRC2) is a member of a family of transcription coactivators. Here, we show that Steroid receptor coactivator 2
SRC2 recruited by c-Myc enhances CD4+ T cell activation to stimulate immune belongs to a family of SRC
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

responses via upregulation of amino acid transporter Slc7a5. Mice deficient of SRC2 coactivators, which orchestrate
in T cells (SRC2fl/fl/CD4Cre) are resistant to the induction of experimental autoim- transcription programs critical
mune encephalomyelitis (EAE) and susceptible to Citrobacter rodentium (C. roden- for multiple cellular functions.
tium) infection. Adoptive transfer of naive CD4+ T cells from SRC2fl/fl/CD4Cre mice However, the function of SRCs
fails to elicit EAE and colitis in Rag1/ recipients. Further, CD4+ T cells from SRC2fl/ especially SRC2 in the immune
fl
/CD4Cre mice display defective T cell proliferation, cytokine production, and dif-
system has long been ignored.
ferentiation both in vitro and in vivo. Mechanically, SRC2 functions as a coactivator
Here, we show that SRC2
to work together with c-Myc to stimulate the expression of amino acid transporter
Slc7a5 required for T cell activation. Slc7a5 fails to be up-regulated in CD4+ T cells functions as a coactivator for
from SRC2fl/fl/CD4Cre mice, and forced expression of Slc7a5 rescues proliferation, c-Myc to stimulate the expression
cytokine production, and the ability of SRC2fl/fl/CD4Cre CD4+ T cells to induce EAE. of amino acid transporter Slc7a5
Therefore, SRC2 is essential for CD4+ T cell activation and, thus, a potential drug required for T cell activation.
target for controlling CD4+ T cell-mediated autoimmunity. Uncontrolled activation of T cells
is responsible for various
CD4+ T cell | T cell activation | c-Myc | autoimmunity
autoimmune diseases such
as multiple sclerosis and
T cell activation is the initial step in adaptive immune responses essential for clearing path-
ogen infection as well as for the induction of autoimmunity (1). T cell activation triggers immunological bowel disease
the clonal expansion of a few antigen-specific naive T cells into a large pool of antigen-specific and thus inhibiting the activity
effector T cells required for the efficient clearance of pathogens that often propagate expo- of SRC2 hypothetically can
nentially such as bacteria. Naive resting T cells are inactive and thus incapable of mediating prevent such T cell-mediated
immune responses. However, T cell receptor (TCR) engagement induces T cell proliferation autoimmunity. Our study not
and differentiation of naive T cells into effector T cells with cytokine production capabilities. only reveals an important
Activated T cells must accumulate sufficient biomass before cell division, including protein mechanism for SRC2-regulated
and DNA, and reflected by greatly increased cell size. To meet the demands of substantially T cell activation but also identifies
increased protein synthesis activity, activated T cells require the transport of amino acids, SRC2 as a potential therapeutic
the building blocks of proteins, from the environment (2), and amino acid transporter
target for T cell-mediated
expression increases up to 100-fold in activated T cells (3). Further, deletion of the genes
autoimmune diseases.
encoding amino acid transporters, such as solute carrier family 7 member 5 (Slc7a5) and
solute carrier family 1 member 5 (Slc1a5), markedly impairs T cell activation including
proliferation and differentiation (3–6), demonstrating the critical function of amino acid
transporters in T cell activation process.
Transcription factor c-Myc is responsible for stimulating the expression of the amino
Author contributions: Z.S., W.Z., X.C. and Y.G. designed
acid transporters during T cell activation, as deletion of c-Myc prevents the upregulation research; W.Z., X.C., X.Z., H.W., Y.S., Y-C.W., and W.S.
of amino acid transporters including Slc7a5 (3). TCR stimulation rapidly induces c-Myc performed research; D.A., Y-C.W., M.F., and Y-C.Y.
contributed new reagents/analytic tools; Z.S., W.Z., X.C.,
(7) whose expression is then increased at both transcriptional and translation levels X.Z., H.W., Y.S., Y-C.W., and W.S analyzed data; and W.Z.,
(8–10). Interestingly, c-Myc-deficient T cells are unable to proliferate and differentiate X.C., and Z.S. wrote the paper.
in response to TCR stimulation; these T cells are also deficient in amino acid transporter The authors declare no competing interest.
Slc7a5, suggesting that Slc7a5 is an important downstream target of c-Myc during T cell This article is a PNAS Direct Submission.
activation. While it is known that c-Myc directly binds to the promoter region of Slc7a5 Copyright © 2023 the Author(s). Published by PNAS.
This article is distributed under Creative Commons
to regulate its expression (11), it remains unknown about other factors that regulate Attribution-NonCommercial-NoDerivatives License 4.0
Slc7a5 expression. (CC BY-NC-ND).
Steroid receptor coactivator 2 (SRC2) belongs to a family of SRC coactivators (SRC) 1
W.Z. and X.C. contributed equally to this work.
consisting of three members, SRC1 (or NCOA1), SRC2 (or NCOA2/TIF2/GRIP1), and 2
To whom correspondence may be addressed. Email:
zsun@coh.org.
SRC3 (or NCOA3/pCIP/ACTR/AIB1). SRCs do not directly bind to target DNA; they
This article contains supporting information online at
function as coactivators for steroid nuclear receptors and other transcription factors by https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.​
interacting with them to regulate gene transcription (12). As such, SRCs orchestrate tran- 2221352120/-/DCSupplemental.
scription programs critical for multiple cellular functions (12). However, the function of Published April 24, 2023.

PNAS 2023 Vol. 120 No. 18 e2221352120 https://doi.org/10.1073/pnas.2221352120 1 of 12


SRCs in the immune system has long been ignored. Our previous and mRNA (SI Appendix, Fig. S1B) in SRC2fl/fl/CD4Cre CD4+
study illustrated that SRC1 can reciprocally regulate the differen- T cells. In contrast to mice (SRC2fl/fl/Foxp3YFP-Cre) with Treg-specific
tiation of inflammatory Th17 cells and Tregs by promoting Th17 deletion of SRC2 that lost weight with aging due to autoimmunity
while inhibiting Treg differentiation (13). Similar to SRC1, we and (17), the body weight of SRC2fl/fl/CD4Cre mice was comparable to
others showed that SRC3 promotes Th17 differentiation (14, 15). that of wild-type (WT) control SRC2fl/fl mice in both male and
Therefore, SRC1 and SRC3 have nonredundant functions in stim- female at 8 to 20 wk of age (SI Appendix, Fig. S1C). Initial analysis
ulating Th17 differentiation. Although a recent report using of SRC2fl/fl/CD4Cre mice also did not find obvious defects in thymic
germline SRC3−/− mice and SRC3 inhibitor hinted at a possible cellularity (SI Appendix, Fig. S1D) and thymocyte development
function of SRC3 in Treg differentiation (16), SRC3 was found in terms of distribution of different developmental stages of
dispensable for Treg differentiation using a T cell-specific SRC3 thymocytes: CD4−CD8− double-negative, CD4+CD8+ double-
knockout mice (15). As for SRC2, we recently discovered its critical positive, and CD4+ or CD8+ single-positive cells (SI Appendix,
role in the differentiation of induced regulatory T cells (17). Fig. S1E). Further, SRC2fl/fl and SRC2fl/fl/CD4Cre mice showed
However, the function of SRC2 in conventional T cells remains comparable spleen cellularity (SI Appendix, Fig. S1F), which was
unknown. In this study, we investigated the function of SRC2 in supported by a comparable percentage of CD19+ B cells and
the regulation of CD4+ T cell activation in vitro and in vivo. CD3+ T cells including CD4+ and CD8+ T cells in both spleens
(SI Appendix, Fig. S1G) and iLNs (SI Appendix, Fig. S1H).
EAE was then induced by immunization with MOG35–55 (18),
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

Results
and SRC2fl/fl/CD4Cre mice were resistant to EAE induction, indi-
Mice Deficient of SRC2 in T Cells (SRC2fl/fl/CD4Cre) Display Defects cated by delayed onset and much less severity (Fig. 1A) as well as
in T Cell-Mediated Immune Responses. To determine the function greatly reduced incidence (Fig. 1B) of the disease when compared
of SRC2 in T cells, we crossed SRC2fl/fl mice to CD4Cre mice to SRC2fl/fl mice. A significant number of infiltrating CD45+ lym-
(SRC2fl/fl/CD4Cre) to delete SRC2 in T cell compartments, which phocytes (Fig. 1C and SI Appendix, Fig. S1I for gating strategy),
was confirmed by the lack of SRC2 protein (SI Appendix, Fig. S1A) including CD4+ T cells (Fig. 1D) producing IL-17A and IFNγ

A EAE B C D E C. Rodentium infection

Body weight ( of original)


4
110
# of CD45+ in CNS (x 105)

n=4
11/11
*** ***
# of CD4+ in CNS (x 104)
3 100 2.5 6
Clinical score

100
80 2.0
(%) Incidence

SRC2fl/fl SRC2fl/fl
2 n=11 4 n=4
SRC2fl/fl/CD4Cre 60 SRC2fl/fl/CD4Cre 1.5 90
SRC2fl/fl
1
SRC2fl/fl
SRC2fl/fl/CD4Cre * * *
40 2/6 1.0 SRC2fl/fl/CD4Cre 80
2 SRC2fl/fl
n=6 20 0.5 SRC2fl/fl/CD4Cre
0
0 2 4 6 8 10 12 14 16 18 20 70
0 0 0 0 2 4 6 8 10 12 14 16 18 20 22
Days post immunization (d)
Days post inoculation (d)

F 105
IL-17A
105
IL-22 IFN
105
GM-CSF
60 ****
25.0 15.3 105 40.6 13.9
50 SRC2fl/fl
104 104 104 104 SRC2fl/fl/CD4Cre
40
% of CD4

103 103 103 103 ***


SRC2fl/fl
30 ns
GM-CSF

102 102 102 102 20 ***


IL-17A

IFN
IL-22

101 101 101 101 10


101 102 103 104 105 101 102 103 104 105 101 102 103 104 105 101 102 103 104 105 0
CD4 CD4 CD4 CD4 IL-17A IL-22 IFN GM-CSF

105 12.1 105 5.62 105 105 14.9


57.2 SRC2fl/fl SRC2fl/fl/CD4Cre
104 104 104 104 H
103 103 103
SRC2fl/fl/CD4Cre
103
GM-CSF

102 102 102


IL-17A

102
IL-22

IFN

101 101 101 101


101 102 103 104 105 101 102 103 104 105 101 102 103 104 105 101 102 103 104 105
CD4 CD4 CD4 CD4

G IL-17A IL-22 IFN GM-CSF I Foxp3 J Foxp3


ns SRC2fl/fl SRC2fl/fl/CD4Cre
20 6 * 30
8 ns 10 ***
*
6 * 105
8
# of cells (×104)
# of cells (×104)
# of cells (×104)

# of cells (×104)
# of cells (×104)

15 6
% of CD4

4 20 104
4 8.44 4.03 6
10 4 103
4
2 10 2
5 102
2 2
Foxp3

0 101
0 0 0 0 0
101 102 103 104 105 101 102 103 104 105
SRC2fl/fl SRC2fl/fl SRC2fl/fl
CD4 SRC2fl/fl/CD4Cre
SRC2fl/fl/CD4Cre SRC2fl/fl/CD4Cre

Fig. 1. Mice deficient of SRC2 in T cells (SRC2fl/fl/CD4Cre) display defects in T cell-mediated immune responses. (A) Mean clinical EAE scores of indicatedmice
at different day post-EAE induction with MOG35-55 (n ≥ 6 per genotype). (B) Incidence of paralysis of indicatedmice after EAE induction shown in A (n ≥ 6 per
genotype). (C and D) Absolute number of CD45+ (C) and CD4+ (D) cells recovered from the CNS from indicated mice 18 d post EAE induction shown in A (n = 3 per
genotype). (E) Body weight of indicated mice over time after C. Rodentium infection (n = 4 per genotype). (F) Representative flow cytometric analysis (Left) and
the percentage (Right) of IL-17A+, IL-22+, IFNγ+, and GM-CSF+ cells among CD4+ cells from the colon of indicated mice 3 wk after C. Rodentium infection shown in
E (n = 4 per genotype). Numbers indicate the percentage of the cells in the gated area (throughout). (G) Absolute number of IL-17A+, IL-22+, IFNγ+, and GM-CSF+
cells in the colon from indicated mice 3 wk after C. Rodentium infection shown in F (n ≥ 3 per genotype). (H) Hematoxylin and Eosin (H&E)-stained colon section
of indicated mice 3 wk after C. Rodentium infection shown in E. (I) Absolute number of CD4+Foxp3+ cells in the colon from indicated mice 3 wk after C. Rodentium
infection shown in E (n ≥ 3 per genotype). (J) Representative flow cytometric analysis (Left) and the percentage (Right) of Foxp3+ cells among CD4+ T cells in the
colon from indicated mice 3 wk after C. Rodentium infection shown in E (n = 3 per genotype). Data are from three experiments (A–G and I; F and J, Right panels; data
in C–E, G, and I; F and J, Right panels are presented as mean ± SD; data in A are presented as mean ± SEM) or are from one representative of three independent
experiments (H; F and J, Left panels). *P < 0.01; ***P < 0.001; ****P < 0.0005; ns, not significant (two-tailed Students’ t test).

2 of 12 https://doi.org/10.1073/pnas.2221352120 pnas.org
(SI Appendix, Fig. S1 J and K), were recovered from the central shortened colon (Fig. 2 H, Left and I ), and colon tissue damage such
nervous system (CNS) of SRC2fl/fl mice. However, there were very as hyperplasia (Fig. 2J). Smaller spleens (Fig. 2 H, Right) together
few CD45+ infiltrating lymphocytes (Fig. 1C), including CD4+ with significantly fewer total number of splenocytes (SI Appendix,
T cells (Fig. 1D) recovered from CNS of SRC2fl/fl/CD4Cre mice, Fig. S2C), including CD4+ T cells (SI Appendix, Fig. S2D) that pro-
indicating a lack of inflammation that causes EAE. SRC2 is thus duce IFNγ and IL-17A (SI Appendix, Fig. S2 E and F for gating
required to generate T cell-mediated immune responses involved strategy), were found in Rag1 mice with SRC2fl/fl/CD4Cre CD4+
in the induction of EAE. T cells than mice with SRC2fl/fl CD4+ T cells, indicating diminished
While EAE results from T cell activation by self-antigen in CNS, inflammation in the absence of SRC2. Indeed, fewer inflammatory
we next determined the function of SRC2 in T cell responses to the CD45+ lymphocytes (Fig. 2 K, Left), CD4+ T cells (Fig. 2 K, Right)
pathogen Citrobacter rodentium (C. rodentium). Clearance of C. that produce IFNγ (Fig. 2 L, Left and SI Appendix, Fig. S2G for
rodentium infection depends on T cells, as mice lacking T cells are gating strategy) and IL-17A (Fig. 2 L, Right) were recovered from
highly susceptible to infection by this strain of bacteria (19, 20). C. the colon of Rag1 mice with SRC2fl/fl/CD4Cre CD4+ T cells than
rodentium infection caused more weight loss in SRC2fl/fl/CD4Cre from mice with SRC2fl/fl CD4+ T cells. Moreover, although naive
mice compared to SRC2fl/fl mice (Fig. 1E), suggesting ineffective CD4+ T cells from both SRC2fl/fl and SRC2fl/fl/CD4Cre mice had
immune responses against infection. C. rodentium induces inflam- equally low levels of surface T cell activation marker CD44
mation, indicated by lymphocyte infiltration, including IL-17- and (SI Appendix, Fig. S2H), CD4+ T cells recovered from the colon
IL-22-producing CD4+ T cells, which play an important role in (Fig. 2M) and mesenteric lymph nodes (mLN) (Fig. 2N) of
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

clearing C. rodentium infection (21, 22). Indeed, a greatly reduced Rag1 mice with SRC2fl/fl/CD4Cre CD4+ T cells expressed lower
percentage (Fig. 1F and SI Appendix, Fig. S1L for gating strategy) levels of CD44 than those recovered from mice with SRC2fl/fl CD4+
and number (Fig. 1G) of IL-17A+ and IL-22+ but not IFNγ+ and T cells, indicating the defective T cell activation in the absence of
GM-CSF+ CD4+ T cells were recovered from the colon of infected SRC2. SRC2 is thus essential for CD4+ T cell function, as deletion
SRC2fl/fl/CD4Cre mice compared to that of SRC2fl/fl mice, supporting of SRC2 almost abrogated CD4+ T cell-mediated immune responses
impaired immune responses against C. rodentium infection in involved in both EAE and colitis.
SRC2fl/fl/CD4Cre mice. In addition, uncontrolled C. rodentium infec-
fl/fl Cre
tion also caused more tissue damage in the colon including hyper- CD4+ T Cells from SRC2 /CD4 Mice Are Defective in T Cell
plasia and damaged crypts in SRC2fl/fl/CD4Cre mice (Fig. 1H). Activation. Since our results above indicated that defective T cell
Consistent with our previously reported requirement of SRC2 for activation is responsible for the lack of immune responses by
the generation of induced Treg (17), there were reduced number CD4+ T cells from SRC2fl/fl/CD4Cre mice, we further examined the
(Fig. 1I) and percentage (Fig. 1J and SI Appendix, Fig. S1L for gat- role of SRC2 in T cell activation. Upon activation by anti-CD3/
ing strategy) of Tregs in the colon but not spleen and lymph node CD28 antibodies, SRC2fl/fl/CD4Cre CD4+ T cells displayed defects
(SI Appendix, Fig. S1M) of SRC2fl/fl/CD4Cre mice. However, reduced in proliferation (Fig. 3A) and upregulation of activation markers
Tregs cannot explain impaired T cell-dependent immune responses CD25 (Fig. 3B) and CD69 (SI Appendix, Fig. S3A). However, there
against C. rodentium infection. Our results thus support the critical was no obvious difference in the survival between CD4+ T cells
role of SRC2 in T cell-mediated immune responses. from SRC2fl/fl and SRC2fl/fl/CD4Cre mice (SI Appendix, Fig. S3
B and C). Activation with PMA and ionomycin led to similar
CD4+ T Cells from SRC2fl/fl/CD4Cre Mice Are Defective in the defects in proliferation (Fig. 3C) and upregulation of activation
Induction of EAE and Colitis. As SRC2 is deleted in the entire marker CD69 (SI Appendix, Fig. S3D) in SRC2fl/fl/CD4Cre CD4+
T cell compartments of SRC2fl/fl/CD4Cre mice, we next determined T cells. We further used OT-II transgenic mice that express TCR
the function of SRC2 specifically in CD4+ T cells. For this recognizing Ova to monitor CD4+ T cell activation by antigen-
purpose, CD4+ T cells were purified from SRC2fl/fl or SRC2fl/fl/ presenting cells (APCs). CD4+ cells from OT-II/SRC2fl/fl/CD4Cre
CD4Cre mice and adoptively transferred to Rag1 mice that were showed more dramatic defects than above activated by CD3/
then induced to develop EAE (SI Appendix, Fig. S2A). In contrast CD28 cross-linking in the upregulation of activation marker CD25
to SRC2fl/fl CD4+ T cells, CD4+ T from SRC2fl/fl/CD4Cre mice (Fig. 3D). This result indicates that overwhelming stimulation by
failed to induce any sign (Fig. 2A) and incidence (Fig. 2B) of CD3/28 cross-link could overcome the defective activation of
EAE in Rag1 recipients. Consistently, there were significantly SRC2fl/fl/CD4Cre T cells. We next examined T cell differentiation.
fewer CD45+ infiltrating lymphocytes (Fig. 2C), including CD4+ Naive CD4+ T cells from SRC2fl/fl and SRC2fl/fl/CD4Cre mice did not
T cells (Fig. 2D) recovered from CNS of Rag1 mice reconstituted produce IFNγ or IL-17A (SI Appendix, Fig. S3E). However, SRC2fl/
with SRC2fl/fl/CD4Cre CD4+ T cells than from Rag1 mice fl
/CD4Cre CD4+ T cells showed a defective Th17 differentiation when
reconstituted with SRC2fl/fl CD4+ T cells. In contrast, there were activated under two different Th17 polarization conditions, IL-6 +
a higher number of CD4+ T cells in the spleens (SI Appendix, TGFβ + IL-23 (Fig. 3E) or IL-6 + TGFβ (SI Appendix, Fig. S3F)
Fig. S2B) of the Rag1 mice reconstituted with SRC2fl/fl/CD4Cre for 45 h (Fig. 3E and SI Appendix, Fig. S3 F, Top panels) or 72 h
CD4+ T cells compared to mice reconstituted with SRC2fl/fl CD4+ (Fig. 3E and SI Appendix, Fig. S3 F, Bottom). SRC2fl/fl/CD4Cre
T cells. This is presumably due to the mobilization of substantially CD4+ T cells showed greatly decreased ability to differentiate into
more SRC2fl/fl than SRC2fl/fl/CD4Cre CD4+ T cells from the spleen IL-17A- but not IFNγ-producing cells, which was confirmed by
to CNS for induction of EAE. Histological analysis revealed more qPCR (SI Appendix, Fig. S3G). Defective Th17 differentiation
infiltrating lymphocytes and tissue damage (demyelination) in was also observed under another Th17 polarization conditions,
the brain (Fig. 2E) and spinal cord (Fig. 2F) of Rag1 mice with IL-6 + IL-1β + IL-23 (SI Appendix, Fig. S3H); however, there was
SRC2fl/fl CD4+ T cells compared to Rag1 mice with SRC2fl/fl/ an increased IFNγ production by SRC2fl/fl/CD4Cre CD4+ T cells.
CD4Cre CD4+ T cells. SRC2 is thus required for the development Further analysis under Th1 polarization conditions demonstrated
of CD4+ T cell-mediated EAE. that SRC2fl/fl/CD4Cre and OT-II/SRC2fl/fl/CD4Cre CD4+ T cells both
Next, we determined the function of SRC2 in colitis induced showed increased differentiation into IFNγ-producing Th1 cells
by the adoptive transfer of naive CD4+ T cells (23). Adoptive trans- (Fig. 3 F and G). Our in vitro analysis thus supports the essential
fer of naive SRC2fl/fl but not SRC2fl/fl/CD4Cre CD4+ T cells induced function of SRC2 in CD4+ T cell activation-induced proliferation
severe colitis in Rag1 recipients, indicated by weight loss (Fig. 2G), and Th17 differentiation.

PNAS 2023 Vol. 120 No. 18 e2221352120 https://doi.org/10.1073/pnas.2221352120 3 of 12


A Adoptive EAE
B C D G Adoptive Colitis

# of CD45+ in CNS (x 105)

Body weight ( of original)


8 120

# of CD4+ in CNS (x 104)


4
100 6/7 5 ** *** n=6
80 110
4 6
*n=6
SRC2fl/fl to Rag1-/- n=7

(%) Incidence
3
Clinical score

SRC2fl/fl/CD4Cre to Rag1-/- 60 3 100


4
2 40 2 90
20 2
1 1 80 SRC2fl/fl to Rag1-/-
0/7
0 0 0 SRC2fl/fl/CD4Cre to Rag1-/-
n=7 70
0 SRC2fl/fl toRag1-/- 0 2 4 6 8 10
SRC2fl/fl to Rag1-/- SRC2fl/fl to Rag1-/-
0 2 4 6 8 10 12 14 16 18 20 22 24 SRC2fl/fl/CD4Cre to Rag1-/-
SRC2fl/fl/CD4Cre to Rag1-/- SRC2fl/fl/CD4Cre to Rag1-/- Weeks post adoptive transfer
Days post immunization (d)

E Brain F Spinal Cord H I *

Length of colon (cm)


12
SRC2fl/fl to Rag1-/- SRC2fl/fl/CD4Cre to Rag1-/- Colon Spleen
SRC2fl/fl to Rag1-/- SRC2fl/fl/CD4Cre to Rag1-/-
9
SRC2fl/fl to Rag1-/- 6
3
SRC2fl/fl/CD4Cre to Rag1-/-
0
SRC2fl/fl to Rag1-/-
SRC2fl/fl/CD4Cre to Rag1-/-

J K Colon
SRC2fl/fl to Rag1-/- SRC2fl/fl/CD4Cre to Rag1-/-
15
* 10 *

# of CD45+ (x 106)

# of CD4+ (x 106)
8
10
6
5 4
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

2
0
0
SRC2fl/fl to Rag1-/-
SRC2fl/fl/CD4Cre to Rag1-/-
L Colon M Colon N mLN
50 * 8 * 1000 ** 2500 *
# of IL-17A+ (x 105)

6 800
(x 105)

40 2000

MFI of CD44
MFI of CD44

30 4 600 1500
+

20 400
# of IFN

1000
Cell count

2 Cell count
10 200 500
0 0
101 102 103 104 0 102 103 104 0
SRC2fl/fl to Rag1-/- CD44 CD44
SRC2fl/fl to Rag1-/- SRC2fl/fl to Rag1-/-
SRC2fl/fl/CD4Cre to Rag1-/-
SRC2fl/fl/CD4Cre to Rag1-/- SRC2fl/fl/CD4Cre to Rag1-/-

Fig. 2. Naive CD4+ T cells from SRC2fl/fl/CD4Cre mice are defective in the induction of EAE and colitis. (A) Mean clinical EAE scores of Rag1−/− mice adoptively
transferred with CD4+ cells from indicated mice at different days post-EAE induction with MOG35-55 (n = 7 per genotype). (B) Incidence of paralysis of indicated
Rag1−/− recipients after EAE induction with MOG35-55 shown in A (n = 7 per genotype). (C and D) Absolute number of CD45+ (C) and CD4+ (D) cells in the CNS
from indicated Rag1−/− recipients shown in A post-EAE induction (n ≥ 4 per genotype). (E and F) Section of H&E-stained brain (E) and spinal cord (F) from Rag1−/−
recipients post-EAE induction shown in A. (G) Body weight of Rag1−/− recipients over time post-adoptive transfer of indicated naive CD4+ T cells (n = 6 per
genotype). (H) Images of colons (Left) and spleens (Right) from Rag1 recipients 8 wk after adoptive transfer of indicated naive CD4+ T cells shown in G. (I and J).
Colon length (I, n = 6 per genotype) and H&E-stained colon section (J) from Rag1 recipients 8 wk after adoptive transfer of indicated naive CD4+ T cells shown
in G. (K) Absolute number of CD45+ (Left) and CD4+ (Right) cells in the colon from Rag1 recipients 8 wk after adoptive transfer of indicated naive CD4+ T cells
shown in G (n ≥ 4 per genotype). (L) Absolute number of CD4+IFNγ+ (Left) and CD4+IL-17A+ (Right) cells in the colon from Rag1 recipients 8 wk after adoptive
transfer of indicated naive CD4+ T cells shown in G (n ≥ 4 per genotype). (M and N) Representative flow cytometric analysis (Left) and the MFI (Right) of CD44
among CD4+ cells in the colon (M) and mesenteric lymph nodes (mLN) (N) from Rag1−/− recipients 8 wk after adoptive transfer of indicated naive CD4+ T cells
shown in G (n ≥ 5 per genotype). Data are from three experiments (A–D, G, I, K, and L; M and N, Right panel; data in C, D, I, K, and L; M and N, Right panel are
presented as mean ± SD; data in A and G are presented as mean ± SEM) or are from one representative of three independent experiments (E, F, H, and J;
M and N, Left panels). *P < 0.01; **P < 0.005, ***P < 0.001 (two-tailed Students’ t test).

We next examined T cell activation in vivo. A low number of CD4+ T cells from OT-II/SRC2fl/fl/CD4Cre mice displayed very
naive CD4+ T cells purified from SRC2fl/fl or SRC2fl/fl/CD4Cre mice severe defective proliferation (Fig. 4E) and upregulation of acti-
were adoptively transferred to Rag1−/− mice to observe lymphopenia- vation markers CD69 and CD25 (Fig. 4 F and G). While we could
induced proliferation (SI Appendix, Fig. S4A). Three weeks after not detect IL-17A+ cells, there were consistently higher percentage
adoptive transfer, Rag1 mice with SRC2fl/fl/CD4Cre CD4+ T cells of IFNγ-producing (Fig. 4H) CD4+ T cells from
had obviously much smaller spleen and lymph nodes than OT-II/SRC2fl/fl/CD4Cre mice compared to OT-II WT mice. In addi-
Rag1 mice with SRC2fl/fl CD4+ T cells (Fig. 4A). Indeed, there were tion, when naive CD4+ T cells from OT-II/SRC2fl/fl and
a considerably less total number of cells (SI Appendix, Fig. S4B), OT-II/SRC2fl/fl/CD4Cre mice were transferred to WT mice, con-
including a greatly reduced number of CD4+ T cells in the spleens sistent with previous findings, OT-II/SRC2fl/fl/CD4Cre CD4+ cells
and lymph nodes (Fig. 4B) in Rag1 mice with SRC2fl/fl/CD4Cre also showed severe defective proliferation (SI Appendix, Fig. S4G)
CD4+ T cells, supporting defects in T cell proliferation in the and upregulation of activation markers CD69 (SI Appendix,
absence of SRC2. Further, the percentage (Fig. 4C) and number Fig. S4H). SRC2 is thus required for CD4+ T cell activation-induced
(Fig. 4D) of IL-17A-producing CD4+ T cells in the spleens of the proliferation and Th17 differentiation in vivo.
Rag1 mice with SRC2fl/fl/CD4Cre CD4+ T cells were also much Given the dramatic defects in vivo, we noticed that the defects
lower compared to that of Rag1 mice with SRC2fl/fl CD4+ T cells, in activation and proliferation in vitro are relatively mild between
confirming the role of SRC2 in generating Th17 cells. SRC2fl/fl and SRC2fl/fl/CD4Cre CD4+ T cells. One explanation
To monitor antigen-specific T cell activation in vivo, we used would be that the nutrients are more enriched and ready to be
OT-II transgenic mice (OT-II/SRC2fl/fl and OT-II/SRC2fl/fl/CD4Cre) used in culture medium than that in vivo, which may overcome
(SI Appendix, Fig. S4C). As expected, naive CD4+ T cells from the defects in activation of SRC2fl/fl/CD4Cre CD4+ T cells. We
OT-II/SRC2fl/fl and OT-II/SRC2fl/fl/CD4Cre mice expressed compa- thus tested the proliferation and activation of CD4+ cells in a
rable low levels of activation markers CD69 (SI Appendix, nutrition-restricted medium and found more dramatic defects
Fig. S4D) and CD25 (SI Appendix, Fig. S4E) before adoptive of proliferation (SI Appendix, Fig. S4I) and upregulation of acti-
transfer. CD4+ T cells were then labeled with dye for monitoring vation marker CD25 (SI Appendix, Fig. S4J) in SRC2fl/fl/CD4Cre
proliferation CellTrace Violet (CTV, SI Appendix, Fig. S4F) and CD4+ T cells compared to that stimulated in the regular
adoptively transferred to Rag1 mice. Upon Ova immunization, nutrition-enriched medium (Fig. 3 A–C). This result suggests

4 of 12 https://doi.org/10.1073/pnas.2221352120 pnas.org
A anti-CD3 and anti-CD28
SRC2fl/fl B Naïve CD4 (0 h)
anti-CD3 and anti-CD28
4h 20 h Naïve CD4 (0 h)
anti-CD3 and anti-CD28
4h 20 h
SRC2fl/fl SRC2fl/fl/CD4Cre SRC2fl/fl/CD4Cre
80
54321 54321
* 100 150 2500 ***
***

Percentage (%)

MFI of CD25
60

MFI of CD25

MFI of CD25
80 120 2000
40
****
* 60 90 1500
Cell count

40 60 1000

% of Max
20
ns ns 20 ns 30 500 SRC2fl/fl
SRC2fl/fl/CD4Cre
0 10-1 101 103 105 10-1 101 103 105 10-11 101 103 105 0 0 0
101 102 103 104 105 101 102 103 104 105 1 2 3 4 5
CTV CD25
Peaks

C PMA and Ionomycine


SRC2flffl/fl
l/fl SRC2fl/fl/CD
//CD4
4Cre E F IL-12
IL-6+TGFβ+IL-23
54321 54321 60 ** * SRC2fl/fl SRC2fl/fl SRC2fl/fl/CD4Cre SRC2fl/fl SRC2fl/fl/CD4Cre

Percentage (%)
SRC2fl/fl/CD4Cre 25
105 0.72 0.29 0.57 0.017 **** 105 21.8 45.8
40
ns
20 80 ***
104 70 h 104

%of CD4
45 h 15
20 60 *

% of CD4
Cell count

103 103
ns ns 10
40
0 102 5 ns 102
1 2 3 4 5 20
101 102 103 104 105 101 102 103 104 105 Peaks 101 17.7 2.38 0 101
CTV IL-17A IFN

D
0
OT- /SRC2fl/fl OT- /SRC2fl/fl/CD4Cre 105 0.74 0.20 1.34 0.51 40
105
45.7 66.8 70 90
**** **** 104
Differentiation time (h)
105
60 **** 72 h
104
30 90 h

%of CD4
104 103 SRC2fl/fl
29.3 13.6 103
20
% CD25/CD4

SRC2fl/fl/CD4Cre
103
0.1 μg/ml 40 **** 102 102
***

IFN
10

IFN
102 20 101 26.5 6.81 101
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

101 102 103 104 105 101 102 103 104 105 0 101 102 103 104 105 101 102 103 104 105
101 IL-17A IFN CD4
0 IL-17A
105 SRC2fl/fl
0.1 0.5 1.0
SRC2fl/fl/CD4Cre
Ova concentration (μg/ml)
104 51.7 25.4 0.5 μg/ml OT- /SRC2fl/fl
103 OT- /SRC2fl/fl/CD4Cre

102 G Ova 0.1 μg/ml Ova 0.5 μg/ml


OT-
T /S
//SRC2
RC2flffl/fl
l/fl
Ova 1.0 μg/ml
OT-
T /S
//SRC2
RC2flffl/fl
l/fll/CD4Cre
r
20
**** **** ****
101 OT- /SRC2fl/fl OT- /SRC2fl/fl/CD4Cre OT- /SRC2fl/fl OT- /SRC2fl/fl/CD4Cre
105 6.91 16.9 15

% IFN /CD4
105 8.64 14.6 6.58 16.6
1.0 μg/ml 104 10
104 56.3 30.3
103 5
103
102
102 0
IFN

101 0.1 0.5 1.0


CD25

101 Ova concentration (μg/ml)


101 102 103 104 105 101 102 103 104 105 101 102 103 104 105 101 102 103 104 105 101 102 103 104 105 101 102 103 104 105
101 102 103 104 105 101 102 103 104 105 OT- /SRC2fl/fl
CD4
CD4 OT- /SRC2fl/fl/CD4Cre

Fig. 3. CD4+ T cells from SRC2fl/fl/CD4Cre mice are defective in T cell activation in vitro. (A) Representative flow cytometric analysis (Left) and percentage (Right) of
the indicated proliferating CD4+ T cells labeled with CellTrace Violet (CTV), stimulated with anti-CD3/28 antibodies for 42 h (n = 4 per genotype). (B) Representative
flow cytometric analysis (Left) and MFI (Right) of CD25 on indicated CD4+ T cells stimulated with anti-CD3/28 antibodies for the indicated times (0 h, 4 h and 20 h)
(n ≥ 4 per genotype). (C) Representative flow cytometric analysis (Left) and percentage (Right) of the indicated proliferating CD4+ T cells labeled with CTV and
stimulated with PMA+ Ionomycin for 42 h (n = 4 per genotype). (D) Representative flow cytometric analysis (Left) and the percentage of CD25 (Right) on CD4+ T cells
from indicated mice after stimulation with different concentrations of Ova-loaded APCs for 24 h (n = 4 per genotype). (E) Representative flow cytometric analysis
(Left) and the percentage (Right) of IL-17A+ and IFNγ+ cells among indicated CD4+ T cells differentiated under Th17 polarization conditions (IL-6+ TGFβ+IL-23)
for 45 h and 72 h (n = 4 per genotype). (F) Representative flow cytometric analysis (Left) and the percentage (Right) of IFNγ+ cells among indicated CD4+ T cells
differentiated under Th1 polarization conditions for 70 h and 90 h (n = 4 per genotype). (G) Representative flow cytometric analysis (Left) and the percentage
of IFNγ (Right) in CD4+ T cells from indicated mice after stimulation with different concentrations of Ova-loaded APCs for 72 h (n = 4 per genotype). Boxed area:
cell population of interest. Data are from three experiments (A–G, Right panels; presented as mean ± SD) or are from one representative of three independent
experiments (A–G, Left panels). *P < 0.01; **P < 0.005; ***P < 0.001; ****P < 0.0005; ns, not significant (two-tailed Students’ t test).

that nutrition availability limits the activation of SRC2fl/fl/CD4Cre implying defective metabolic pathways. We next compared the
CD4+ T cells. expression of the important genes known to regulate metabolisms
required for T cell activation and proliferation (25) (SI Appendix,
CD4+ T Cells from SRC2fl/fl/CD4Cre Mice Are Defective in c-Myc- Fig. S5D). We found that Hif1a, Batf, Myc, and Irf4 were down-
Dependent Pathways. Since SRC2 is a transcription coactivator regulated in SRC2fl/fl/CD4Cre CD4+ cells from RNA-seq (Fig. 5D),
that regulates cellular function via controlling gene expression, and qPCR analysis confirmed the downregulation of Hif1a and Baft
we performed RNA-seq analysis to determine SRC2-regulated (Fig. 5E). The top down-regulated gene was Hif1a, which is known
genes after the adoptive transfer of SRC2fl/fl and SRC2fl/fl/CD4Cre to promote T cell activation and Th17 differentiation (26, 27).
CD4+ T cells (SI Appendix, Fig. S5A). The computational principal Indeed, SRC2fl/fl/CD4Cre CD4+ cells also had impaired Th17
component analysis revealed that eight RNA-seq samples were differentiation in addition to defective T cell activation (Figs. 3
divided into SRC2fl/fl or SRC2fl/fl/CD4Cre group (SI Appendix, and 4). Down-regulated Hif1a at mRNA (SI Appendix, Fig. S5E)
Fig. S5B), indicating reproducible gene expression patterns within and protein levels (SI Appendix, Fig. S5F) were also confirmed
each group and thus the high quality of RNA-seq results. We in in vitro activated SRC2fl/fl/CD4Cre CD4+ cells. However,
identified 2743 differentially expressed genes (DEGs), with 1408 forced expression of Hif1a did not rescue defective proliferation
up-regulated and 1335 down-regulated genes, between SRC2fl/fl and (SI Appendix, Fig. S5G) or Th17 differentiation (SI Appendix,
SRC2fl/fl/CD4Cre CD4+ cells (Fig. 5A). Ncoa2, which encodes SRC2, Fig. S5H) of SRC2fl/fl/CD4Cre CD4+ cells, suggesting other defects
was among the most down-regulated genes (Fig. 5 A, Left and in the absence of SRC2.
SI Appendix, Fig. S5C), confirming deletion of this gene. KEGG DEGs were then subjected to upstream regulator analysis by
pathway analysis of DEGs identified many dysregulated pathways Ingenuity Pathway Analysis (IPA) to search for dysregulated
in SRC2fl/fl/CD4Cre cells; the most enriched pathways include DNA upstream regulators and c-Myc was identified as the top transcrip-
replication, cell cycle, and amino acid metabolism critical for cell tion factor responsible for the altered gene expression pattern in
proliferation (Fig. 5B). Cell proliferation required ATP to provide SRC2fl/fl/CD4Cre CD4+ cells (Fig. 5F). Indeed, down-regulated
energy for cellular activities including DNA replication and protein c-Myc activity was directly linked to the altered expression of more
synthesis (24). Indeed, SRC2fl/fl/CD4Cre CD4+ cells generated than 150 known c-Myc target genes in SRC2fl/fl/CD4Cre CD4+ cells
much less ATP compared to SRC2fl/fl cells (Fig. 5C), consistent from RNA-seq results (SI Appendix, Fig. S5I). Deletion of c-Myc
with the observed defective proliferation in the absence of SRC2, prevents T cell activation (5, 28, 29), similar to the deletion of

PNAS 2023 Vol. 120 No. 18 e2221352120 https://doi.org/10.1073/pnas.2221352120 5 of 12


A B Spleen iLN C Spleen
l/fl to Rag1
SRC2flffl/fl g −/−
/ SRC2flffl/fl
l/fl//CD4 r to Ra
CD4Cre Rag1−/− 10
SRC2fl/fl to Rag1−/− SRC2fl/fl/CD4Cre to Rag1−/−
8 *
* 10

# of CD4 cells (x106)


105 8
iLN Spleen mLN iLN Spleen mLN
*

# of CD4 cells (x105)


7.44 1.49

%of CD4
6 8 104 6
6 4
4 103
4 2
2 102

IL-17A
2 0
101
0 0 SRC2fl/fl to Rag1−/−
101 102 103 104 105 101 102 103 104 105
SRC2fl/fl to Rag1−/− SRC2fl/fl/CD4Cre to Rag1−/−
CD4
SRC2fl/fl/CD4Cre to Rag1−/−

D Spleen E Day 2
OT-ɉ/SRC2fl/fl to Rag1−/− OT-ɉ/SRC2fl/fl/CD4Cre to Rag1−/−
F Day 1 Day 2 Day 1 Day 2
**
# of IL-17A cells (x105)

4
* 250 800
80 ** **

MFI of CD69
3

% Proliferation
60.1 28.8 200 600

CD69 MFI
60
2 150
SPL 40 400

% of Max
100
Cell count

1 200
20 50
0 0 0
0
SRC2fl/fl toRag1−/− 101 102 103 104 105 OT-ɉ/SRC2fl/fl to Rag1−/− 101 102 103 104 105 101 102 103 104 105
101 102 103 104 105 OT-ɉ/SRC2fl/fl to Rag1−/−
SRC2fl/fl/CD4Cre to Rag1−/− CTV OT-ɉ/SRC2fl/fl/CD4Cre to Rag1−/− CD69
OT-ɉ/SRC2fl/fl/CD4Cre to Rag1−/−

G Day 2 H Day 7
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

OT-ɉ/SRC2fl/fl to Rag1−/− OT-ɉ/SRC2fl/fl/CD4Cre to Rag1−/− OT-ɉ/SRC2fl/fl to Rag1−/− OT-ɉ/SRC2fl/fl/CD4Cre to Rag1−/−


105 60
*
49.7 39.9
80 ** 105
34.5 43.9
% CD25/CD4

104 60 104

% IFN/CD4
40
103 40 103
102 20 20
102
0
CD25

101
IFN

101 0
OT-ɉ/SRC2fl/fl to Rag1−/−
101 102 103 104 105 101 102 103 104 105 101 102 103 104 105 101 102 103 104 105
OT-ɉ/SRC2fl/fl/CD4Cre to Rag1−/− OT-ɉ/SRC2fl/fl to Rag1−/−
CD4 CD4
OT-ɉ/SRC2fl/fl/CD4Cre to Rag1−/−

Fig. 4. CD4+ T cells from SRC2fl/fl/CD4Cre mice are defective in T cell activation in vivo. (A) Representative image of iLNs, spleens, and mesenteric lymph nodes
(mLNs) from Rag1−/− recipients 3 wk post adoptively transfer of 0.4 × 106 naive CD4+ cells from indicated mice. (B) Absolute number of CD4+ T cells in the spleens
(Left) and iLNs (Right) from Rag1−/− recipients 3 wk post adoptively transfer of indicated naive CD4+ cells shown in A (n ≥ 4 per genotype). (C) Representative
flow cytometric analysis (Left) and the percentage (Right) of IL-17A+ cells among CD4+ T cells from the spleens of Rag1−/− recipients 3 wk post adoptive transfer
of indicated naive CD4+ cells shown in A (n = 5 per genotype). (D) Absolute number of IL-17A+ cells in the spleens from Rag1−/− recipients 3 wk post adoptively
transfer of indicated naive CD4+ cells shown in C (n = 5 per genotype). (E) Representative flow cytometric analysis (Left) and percentage (Right) of the indicated
proliferating CD4+ T cells labeled with CTV 2 d after being adoptively transferred to Rag1−/− recipients and stimulated by Ova immunization (n = 10 per genotype).
(F) Representative flow cytometric analysis (Left) and the MFI (Right) of CD69 among indicated CD4+ T cells in the spleens of Rag1−/− recipients, 1 d (Top two panels)
and 2 d (Bottom two panels) after Ova immunization shown in E (n ≥ 4 per genotype). (G) Representative flow cytometric analysis (Left) and the percentage of
CD25+ cells (Right) among indicated CD4+ T cells in the spleen of Rag1−/− recipients 2 d after Ova immunization shown in E (n = 10 per genotype). (H) Representative
flow cytometric analysis (Left) and the percentage of IFNγ+ cells among CD4+ T cells (Right) in the spleen of Rag1−/− recipients 7 d after Ova immunization shown
in L (n = 4 per genotype). Boxed area: cell population of interest. Data are from three experiments (B and D; C and E–H, Right panels; presented as mean ± SD)
or are from one representative of three independent experiments (A; C and E–H, Left panels). *P < 0.01; **P < 0.005, (two-tailed Students’ t test).

SRC2. c-Myc is known to regulate multiple metabolic genes, knockdown of SRC2 (sgSRC2) but not nontargeting control
including glycolysis and amino acid transportation essential for (sgNTC) significantly impaired c-Myc-dependent activation of the
T cell activation (SI Appendix, Fig. S5J). Indeed, critical genes reporter (Fig. 6F), suggesting that SRC2 functions as a coactivator
involved in these c-Myc-regulated metabolic pathways were to stimulate c-Myc activity. Further, c-Myc and SRC2 form protein
down-regulated in SRC2fl/fl/CD4Cre CD4+ cells (Fig. 5G and complexes and could be defected in the same complexes after, but not
SI Appendix, Fig. S5K), and the mRNA levels of the most before, stimulation of CD4+ T cells by anti-CD3/CD28 antibodies
down-regulated genes were confirmed by individual qPCR (Fig. 6 C and D, Bottom and SI Appendix, Fig. S6B for full image
(Fig. 5H). Computational analysis of RNA-seq data thus indicates of the gel) or Th17 differentiation conditions (SI Appendix, Fig. S6
defective c-Myc-dependent pathways in the absence of SRC2. C and B, Right panel for the full image of the gel). These results
thus suggest that c-Myc recruits SRC2 to stimulate transcription.
c-Myc Recruits SRC2 to Stimulate Transcription. We next deter­ It could explain why the forced expression of c-Myc could not
mined whether the computational analysis predicted downregulation rescue the defective activation of SRC2fl/fl/CD4Cre CD4+ cells, as
of c-Myc activity is responsible for defective activation of SRC2fl/ c-Myc is unlikely able to fully activate some target genes without
fl
/CD4Cre CD4+ cells. Forced expression of c-Myc only slightly coactivator SRC2.
reversed defective proliferation (Fig. 6A) of SRC2fl/fl/CD4Cre CD4+
cells but showed no effects on Th17 differentiation (Fig. 6B) of SRC2 Regulates CD4+ T Cell Activation by Controlling c-Myc-
either SRC2fl/fl or SRC2fl/fl/CD4Cre CD4+ cells. We next monitored Dependent Expression of Amino Acid Transporter Slc7a5.
c-Myc protein levels, which were very low in unstimulated naive c-Myc regulates many pathways including well-known glycolysis
SRC2fl/fl and SRC2fl/fl/CD4Cre CD4+ cells (Fig. 6 C, Top Left and and amino acid transportation (28). However, SRC2fl/fl/CD4Cre
SI Appendix, Fig. S6A for the full image of the gel). Consistent with CD4+ cells displayed rather increased than decreased glycolysis
previous reports (5, 30), c-Myc is greatly up-regulated in SRC2fl/ (SI Appendix, Fig. S7 A and B) and no obvious changes in oxidative
fl
CD4+ T cells in response to TCR stimulation. Activated SRC2fl/ phosphorylation (SI Appendix, Fig. S7 C and D) in response to TCR
fl
/CD4Cre CD4+ cells also up-regulated c-Myc to the levels that stimulation, not supporting the positive role of SRC2 in these two
were only slightly lower than the WT cells (Fig. 6 C and D, Top), processes. In addition, c-Myc is responsible for the upregulation of
indicating that c-Myc is not likely regulated at the protein level. multiple amino acid transporters that import basic building blocks
Since SRC2 is a transcriptional coactivator, we determined whether to fuel the high levels of protein synthesis in proliferating T cells
SRC2 can stimulate c-Myc transcriptional activity. Indeed, c-Myc (2). Indeed, several amino acids metabolism pathways were found
together with SRC2 but not alone maximally stimulated the c-Myc altered from RNA-seq results (Fig. 5B). Pharmacological inhibition
luciferase reporter (Fig. 6E). In addition, CRISPR/Cas9-mediated or deletion of the genes encoding amino acid transporters severely

6 of 12 https://doi.org/10.1073/pnas.2221352120 pnas.org
A B C D
DNA replication 6
Cell cycle 200 * Hif1a
****

ATP (uM/106 cells)


5
10 Nicotinate and nicotinamide metabolism 150
C fl/fl/CD4Cre vs. SRC2fl/fl
SRC2 4

-Log10P
Biosynthesis of amino acids 100
8 Batf
3
p53 signaling pathway 50 **
6 2 Myc
Arginine and proline metabolism
-Log10P

1408 0 Irf4
Ncoa2
Fatty acid biosynthesis 0 12 24 36 48 60 1
4 1335 -Log10P
Activation time (h)
D-Glutamine and D-glutamate metabolism Enrichment score 0
2 -2 0 2
Pyrimidine metabolism SRC2fl/fl
SRC2fl/fl/CD4Cre Log2 FC (SRC2fl/fl/CD4re vs. SRC2fl/fl)
0 0 2 4 6 8 10 12 14 16
-8 -7 -6 -5 -4 -3 -2 -1 0 1 2 3 4 5 6 7 8
Log2 FC (SRC2fl/fl/CD4re vs. SRC2fl/fl)

E F Upstream
p regulators
g G Gene name FC (SRC2 /CD4re vs. SRC2 )
fl/fl fl/fl
-Log10P Function
Asns Glutaminolysis
2.5
Tpi1 Glycolysis
ns MYC Glycolysis
Relative mRNA

2.0 Slc16a3
CEBPB Slc7a5 Glutaminolysis
1.5 ns Gfpt1 Polyamine
** * E2F1 Srm Polyamine
1.0 KLF6 Pgam1 Glycolysis
Got2 Glutaminolysis
0.5 FOXM1 Ppat Glutaminolysis
YAP1 Sdhb TCA cycle
0 Oat Polyamine
TBX2 Glycolysis
Slc16a1
Hif1a Batf Myc Irf4 E2F3 Glycolysis
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

-Log10P Pgk1
Pkm Glycolysis
SRC2fl/fl IRF1 - Z score Glycolysis
Tkt
SRC2fl/fl/CD4Cre BHLHE40 Cad Glutaminolysis
Ldha Glycolysis
0 10 20 30 40 Slc2a3 Glycolysis
Got1 Glutaminolysis

H
G6pdx Glycolysis
Fh1 TCA cycle
ns Suclg2 TCA cycle
Aco1 TCA cycle
2.5 TCA cycle
Sdhd
TCA cycle
Relative mRNA

2.0 Suclg1
ns ns Mdh1 TCA cycle
1.5
*** ** * * p=0.1 Idh3a
Gls
TCA cycle
Glutaminolysis
1.0 Mdh2 TCA cycle
Pfkl Glycolysis
0.5 SRC2fl/fl Eno1b Glycolysis
SRC2fl/fl/CD4Cre Aco2 TCA cycle
0 Slc2a1 Glycolysis
H6pd Glycolysis
Gfpt1 Slc7a5 Tpi1 Ppat Asns Pgam1 Got2 Slc16a3
-2.5 -2.0 -1.5 -1.0 -0.5 0.0 0.5 1.0 1.5
0.0 2.0 4.0

+ fl/fl Cre
Fig. 5. CD4 T cells from SRC2 /CD4 mice are defective in c-Myc-dependent pathways. (A) Volcano plot comparison of gene expression between indicated
CD4+ cells adoptively transferred to Rag1−/− recipients (n = 4 per genotype). Differentially up-regulated genes (red) and down-regulated genes (blue) with a cutoff
at P < 0.05 and FC > 1.2. are shown. (B) List of the top down-regulated pathways (P < 0.1) in SRC2fl/fl/CD4Crecells compared to WT SRC2fl/fl CD4+ cells. (C) Intracellular
ATP production by indicated CD4+ cells after stimulation by anti-CD3/28 antibodies for indicated times (n ≥ 6 per genotype). (D) Volcano plot comparison of the
gene expression of top T cell metabolism regulators between SRC2fl/fl/CD4Cre and SRC2fl/fl CD4+ cells. (E) qPCR analysis of indicated genes in indicated CD4+ cells
4 wk after being adoptively transferred to Rag1 recipients (n ≥ 4 per genotype). (F) A list of predicted upstream transcription factors responsible for the changed
gene expression patterns in SRC2fl/fl/CD4CreCD4+ T cells by IPA analysis. (G) A list of c-Myc-regulated metabolic genes and their function that were down-regulated
in SRC2fl/fl/CD4CreCD4+ T cells. (H) qPCR analysis of indicated genes in indicated CD4+ cells 4 wk after being adoptively transferred to Rag1 recipients (n ≥ 4 per
genotype). Data are from three experiments (C, E and H; presented as mean ± SD). *P < 0.01; **P < 0.005; ***P < 0.001; ****P < 0.0005; ns, not significant (two-
tailed Students’ t test).

impairs T cell activation-induced proliferation and cytokine to OT-II WT CD4+ T cells (Fig. 7B). Next, c-Myc or SRC2 gene
production similar to the deletion of c-Myc or SRC2 gene (3– was deleted using CRISPR/Cas9 in CD4+ T cells obtained from
6, 31). Indeed, we found a significantly down-regulated gene mice expressing Cas9 and resulted in reduced T cell proliferation
encoding amino acid transporter Slc7a5 in SRC2fl/fl/CD4Cre CD4+ (SI Appendix, Fig. S7I) and downregulation of CD98 (Fig. 7C).
cells (Fig. 5G), which was one of the top down-regulated genes Therefore, CD98 and Slc7a5 have the same expression pattern,
among metabolic gene set confirmed by qPCR analysis (Fig. 5H). which is regulated by c-Myc and SRC2. Consistent with the
Interestingly, only Slc7a5 was significantly down-regulated among reduced expression of amino acid transporter Slc7a5, the capacity
the amino acid transporters that were reported to be regulated by of amino acid uptake was significantly lower in SRC2fl/fl/CD4Cre
c-Myc (SI Appendix, Fig. S7E) (3), indicating the selective regulation CD4+ T cells than that in SRC2fl/fl CD4+ T cells (Fig. 7D). Since
of Slc7a5 by SRC2. Slc7a5 transports amino acids including leucine, increased uptake of amino acids stimulates mTOR pathway (5), we
tryptophan, and methionine, the first amino acid for all proteins, monitored the levels of pS6 (phosphorylated ribosomal protein S6),
and thus is important for protein synthesis. Since Slc7a5 forms a an indicator for mTOR activation. Indeed, pS6 levels were lower in
heterodimer with CD98 and CD98 expression is greatly reduced SRC2fl/fl/CD4Cre CD4+ cells compared to WT control (SI Appendix,
without Slc7a5 (5), CD98 is often used as a surrogate for Slc7a5 Fig. S7J), consistent with the reduced amino acid uptake activity
expression (28, 32, 33). Same as the Slc7a5 expression pattern, in SRC2fl/fl/CD4Cre CD4+ cells.
CD98 was greatly up-regulated in activated T cells compared to To determine whether the downregulation of Slc7a5 is respon-
naive T cells (Fig. 7A). However, activated SRC2fl/fl/CD4Cre CD4+ sible for the defective T cell activation in the absence of SRC2,
cells failed to up-regulate CD98 to the levels detected on SRC2fl/ we transduced SRC2fl/fl/CD4Cre CD4+ T cells with the retrovirus
fl
cells and such defective upregulation of CD98 became more expressing Slc7a5. Overexpression of Slc7a5 greatly up-regulated
dramatic when SRC2fl/fl/CD4Cre CD4+ cells were stimulated in a CD98 in both SRC2fl/fl (SI Appendix, Fig. S7K) and SRC2fl/fl/
nutrition-restricted medium (SI Appendix, Fig. S7F). Similarly, CD4Cre CD4+ T cells (SI Appendix, Fig. S7L), confirming the pos-
dramatic defective upregulation of CD98 was also found in OT-II/ itive correlation between Slc7a5 and CD98 expression. Further,
SRC2fl/fl/CD4Cre CD4+ cells stimulated by different concentrations forced expression of Slc7a5 promoted proliferation (Fig. 7E) and
of Ova-loaded APCs (SI Appendix, Fig. S7G). Naive CD4+ T cells Th17 differentiation (Fig. 7F) of SRC2fl/fl/CD4Cre but not obvi-
from OT-II and OT-II/SRC2fl/fl/CD4Cre mice were transferred to ously in SRC2fl/fl CD4+ T cells (SI Appendix, Fig. S7M for prolif-
Rag1 mice that were then immunized with Ova to monitor CD98 eration and S7N for Th17 differentiation) compared to the cells
expression in vivo (SI Appendix, Fig. S7H). CD98 expression was transduced with empty virus (EV), supporting the critical function
consistently lower in activated OT-II/SRC2fl/fl/CD4Cre compared of SRC2-regulated Slc7a5 in T cell activation and Th17

PNAS 2023 Vol. 120 No. 18 e2221352120 https://doi.org/10.1073/pnas.2221352120 7 of 12


EV c-Myc
A anti-CD3 and anti-CD28
42 h B 105
IL-17A
SRC2fl/fl/CD4Cre 24.1 23.8
EV c-My
Myc
c-Myc
60 ns 104

Percentage (%)
5 432 1 5 432 1 103 SRC2fl/fl ***
40 ns
30
* 102

20 101
ns ns 20

%of CD4
Cell count

105
0 ns
1 2 3 4 5 10
104 6.28 5.09 SRC2fl/fl/CD4Cre
Peaks
101 102 103 104 105 101 102 103 104 105
CTV SRC2fl/fl/CD4Cre +EV 103 0
SRC2fl/fl/CD4Cre +c-Myc SRC2fl/fl + + - -
102
SRC2fl/fl/CD4Cre - - + +

IL-17A
101 EV + - + -
c-Myc - + - +
101 102 103 104 105 101 102 103 104 105
CD4

C D E ***
12 **
anti-CD3 and anti-CD28
0h 5h 27 h **
2.0 2.0

Luciferase activity
0h 5h 27 h 2.0

c-Myc relative expression


10

c-Myc relative expression

c-Myc relative expression

(Relative fold)
SRC2fl/fl SRC2fl/fl/CD4Cre SRC2fl/fl SRC2fl/fl/CD4Cre SRC2fl/fl SRC2fl/fl/CD4Cre
8
c-Myc 1.5 ns 1.5
*
1.5
ns ***
6
Actin 1.0 1.0 1.0
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

4
SRC2fl/fl SRC2fl/fl SRC2fl/fl 0.5 0.5 0.5 2
MW(kDa) SRC2 IgG SRC2 IgG SRC2 IgG
0 0 0 0
250 SRC2 Reporter + + + +
IP 150
100 SRC2fl/fl c-Myc - + + -
75
c-Myc-SRC2 relative binding intensity
c-Myc SRC2fl/fl/CD4Cre SRC2 - - + +
50 1.0
72 h
F
37 0.8 48 h
25 0.6
** 12
20 * 30 *
0.4 10 * 25

Luciferase activity

Luciferase activity
15

(Relative fold)
0.2 8

(Relative fold)
20
10 0 6 15
0 h 5h 27h
4 10
2 5
0 0
Reporter + + + Reporter + + +
c-Myc - + + c-Myc - + +
sgNTC - + - sgNTC - + -
sgSRC2 - - + sgSRC2 - - +

Fig. 6. c-Myc recruits SRC2 to stimulate transcription. (A) Representative flow cytometric analysis (Left) and percentage (Right) of the indicated proliferating CD4+
T cells transduced with virus expressing Green Fluorescent Protein (GFP) alone (empty vectors, EV) or together with c-Myc, labeled with CTV, and stimulated with
anti-CD3/28 antibodies for 42 h (n = 4 per genotype). (B) Representative flow cytometric analysis (Left) and the percentage (Right) of IL-17A+ among indicated
CD4+ T cells transduced with retrovirus shown in A and differentiated under Th17 polarization conditions (n = 3 per genotype). (C) Immunoblot analysis of
c-Myc in indicated CD4+ T cells stimulated by anti-CD3/28 antibodies for indicated times (Top). Bottom panels are the immunoblot analysis of c-Myc among
anti-SRC2 antibody or IgG control antibody immunoprecipitated (IP) SRC2 complexes from CD4+ cells stimulated with anti-CD3/28 antibodies for indicated
times. (D) Intensity of c-Myc band relative to the control actin shown in Top panels of C (Top) (n = 3 per genotype). Bottom panel is the intensity of c-Myc band
relative to SRC2 shown in C (Bottom). (E) Luciferase activity from c-Myc reporter in HEK 293T cells transfected with c-Myc luciferase reporter with and without
the expression plasmid for c-Myc and SRC2 for 42 h. (n = 4 per treatment). (F) Luciferase activity from c-Myc reporter in HEK 293T cells transfected with c-Myc
luciferase reporter with and without the expression plasmid for c-Myc, nontargeting control single guide RNA (sgNTC), and single-guide RNA for deleting SRC2
(sgSRC2) for 48 h (Left) and 72 h (Right) (n= 3 per treatment). Boxed area: cell population of interest. Data are from three experiments (D–F; A and B, Right panels;
presented as mean ± SD) or are from one representative of three independent experiments (C; A and B, Left panels). *P < 0.01; **P < 0.005; ***P < 0.001; ns,
not significant (two-tailed Students’ t test).

differentiation. As a control, forced expression of Slc7a5 did not expression of Slc7a5. Altogether, our data demonstrate that SRC2,
affect Th1 differentiation in either SRC2fl/fl (SI Appendix, Fig. S7O) as a coactivator for c-Myc, promotes CD4+ T cell-mediated
or SRC2fl/fl/CD4Cre (SI Appendix, Fig. S7P) cells. immune responses by stimulating T cell activation via upregula-
Lastly, an indirect EAE model was used to determine the func- tion of amino acid transporter Slc7a5.
tion of SRC2 in the regulation of Slc7a5 in vivo. CD4+ T cells
from MOG35-55-immunized SRC2fl/fl and SRC2fl/fl/CD4Cre mice Discussion
were transduced with retrovirus expressing Slc7a5 and expanded
in vitro, followed by adoptive transfer to Rag1−/− for induction of T cell activation is critical to fully mount adaptive immune responses
EAE (Fig. 7G). Whereas expression of Slc7a5 did not further essential for pathogen clearance and autoimmune induction.
enhance EAE induced by SRC2fl/fl CD4+ T cells, forced expression Activated T cells have a demand for substantially increased protein
of Slc7a5 partially restored the ability of SRC2fl/fl/CD4Cre CD4+ synthesis activity that depends on amino acids transported by the
T cells in the induction of EAE both in terms of severity (Fig. 7H) amino acid transporters. In this study, we demonstrated the role of
and incidence (Fig. 7I) of EAE. Forced expression of Slc7a5 SRC2 in promoting the expression of amino acid transporter,
increased CD98 levels on SRC2fl/fl/CD4Cre CD4+ T cells recovered Slc7a5, during T cell activation by interacting with c-Myc. We show
from the CNS of Rag1 mice compared to that transduced with that mice deficient in SRC2 in T cells (SRC2fl/fl/CD4Cre) have greatly
empty virus (EV) (SI Appendix, Fig. S7Q), confirming stimulation impaired CD4+ T cell-mediated immune responses responsible for
of CD98 by Slc7a5 expression. Consistently, the number of total EAE and colitis. SRC2fl/fl/CD4Cre mice have reduced Tregs similar
CD4+ T cells and pathogenic IL-17- and IFNγ-producing CD4+ to SRC2fl/fl/Foxp3YFP-Cre mice, which confirms our previous results
T cells recovered from the CNS were significantly increased by showing that SRC2 is required for the generation of Tregs (17).
expression of Slc7a5 in SRC2fl/fl/CD4Cre CD4+ T cells (Fig. 7J), However, reduced Tregs cannot explain the greatly impaired CD4+
reflecting enhanced inflammation responsible for EAE by forced T cell-dependent immunity in SRC2fl/fl/CD4Cre mice, which

8 of 12 https://doi.org/10.1073/pnas.2221352120 pnas.org
A anti-CD3 and anti-CD28 B C ****
Naïve
ï CD4 2h 4h 20 h 1200 ** 12000 **
1000

MFI of CD98
10000

MFI of CD98
800
600 8000

Cell count
400

% of Max
6000 sgNTC
% of Max

200 sgSRC2
0 4000 sgMyc
101 102 103 104 103 104 105
CD98 OT-ɉ/SRC2fl/fl to Rag1−/ −
102 103 104 102 103 104 102 103 104 102 103 104 CD98
OT-ɉ/SRC2fl/fl/CD4Cre to Rag1−/−
CD98
ns
400 700
600
** 800 *
3000
***
D E SRC2fl/fl/CD4Cre
*
MFI of CD98

300 500 200 *** 60


MFI of CD98

MFI of CD98
600

MFI of CD98
EV

Relative AA uptake
Slc7a5
**

Percentage (%)
400 2000 5 4 32 1 5 4 32 1
200 400
300 150 40
100 200 1000
200
100
0 0 0 100 20 *
0
ns *

% of Max

Cell count
SRC2fl/fl
SRC2fl/fl/CD4Cre 50 0
F IL-6+IL-23+TGFβ
101
BPA uptake
102 103 Blank
SRC2fl/fl 102 103 104 105 102 103 104 105
1 2 3
Peaks
4 5

SRC2fl/fl/CD4Cre IL-17A SRC2fl/fl/CD4Cre +EV


SRC2fl/fl/CD4Cre CTV
EV Slc7a5 40 ** SRC2fl/fl/CD4Cre +Slc7a5
Percentage (% of CD4)

105

104
18.1 32.0 30 G Mice immunization
with MOG35-55 Spin infection
Activation with with virus Injection to
20 Polarization under Rag1-/-
103 MOG35-55 and CD28 Th17 conditions
102 10 SRC2fl/fl/CD4Cre +EV
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

SRC2fl/fl/CD4Cre +Slc7a5 11 days 24 h 3h 3 days


IL17A

101 0
101 102 103 104 105 101 102 103 104 105
CD4
SRC2fl/fl SRC2fl/fl/CD4Cre
EV
H 3
I 4/4 4/4
Retrovirus preparation

J
Slc7a5
100
(%) Incidence

80
Clinical score

2 4/6 ns
ns

# of IL-17A in CNS (x 103)


40

# of CD4+ in CNS (x 104)


60 ns
*

# of IFN in CNS (x 103)


20
10
40 2/6
1 SRC2fl/fl +EV
SRC2fl/fl +Slc7a5
20 8
* 15 30 P=0.065
0 6 20 SRC2fl/fl +EV
SRC2fl/fl/CD4Cre +EV 10
SRC2fl/fl +Slc7a5
SRC2fl/fl/CD4Cre +Slc7a5 4
+ 10 SRC2fl/fl/CD4Cre +EV
0 SRC2fl/fl + - - 5
SRC2fl/fl/CD4Cre - - + + 2 SRC2fl/fl/CD4Cre +Slc7a5
0 2 4 6 8 10 12 14 16 18 20 22 24 EV + - + - 0
0 0
Days cells transfer (d) Slc7a5 - + - +

Fig. 7. SRC2 regulates CD4+ T cell activation via controlling c-Myc-dependent expression of amino acid transporter Slc7a5. (A) Representative flow cytometric
analysis (Top) and the MFI of CD98 (Bottom) on indicated CD4+ T cells stimulated with anti-CD3/28 antibodies for indicated times (0 h, 2 h, 4 h, and 20 h) (n ≥ 4 per
genotype). (B) Representative flow cytometric analysis (Left) and MFI of CD98 (Right) among OT-II/SRC2fl/fl or OT-II/SRC2fl/fl/CD4Cre CD4+ T cells adoptively transferred
to Rag1−/− recipients challenged with Ova for 1 d (n ≥ 4 per genotype). (C) Representative flow cytometric analysis (Left) and MFI of CD98 (Right) among CD4+ T cells
expressing Cas9 and transduced with retrovirus expressing sgNTC (nontargeting control), sgSRC2 (deletion of SRC2), and sgMyc (deletion of c-Myc), stimulated by
anti-CD3/28 antibodies for 45 h (n = 5 per genotype). (D) Amino acid uptake of indicated CD4+ T cells stimulated with anti-CD3/28 antibodies for 24 h (n = 4 per
genotype). (E) Representative flow cytometric analysis (Left) and percentage (Right) of proliferating SRC2fl/fl/CD4Cre CD4+ T cells transduced with retrovirus expressing
GFP alone (empty vector, EV) or together with Slc7a5, labeled with CTV, and stimulated with anti-CD3/28 antibodies (n = 4 per genotype). (F) Representative flow
cytometric analysis (Left) and the percentage (Right) of IL-17A+ cells among SRC2fl/fl/CD4Cre CD4+ T cells transduced with retrovirus expressing GFP alone (empty
vector, EV) or together with Slc7a5 and differentiated under Th17 polarization conditions (n = 5 per genotype). (G) Overview of the experimental procedure used
for passive EAE. (H) Mean clinical EAE scores of Rag1−/− mice different days after adoptive transfer of splenic cells obtained from MOG35-55-primed SRC2fl/fl or
SRC2fl/fl/CD4Cre mice and transduced with retrovirus expressing GFP alone (EV) or together with Slc7a5 (n ≥ 4 per genotype per treatment). (I) Incidence of paralysis
of Rag1−/− recipients shown in H. (J) Absolute number of CD4+ (Left), IL-17A+ cells (Middle), and IFNγ+ cells (Right) in the CNS from EAE-induced Rag1−/− recipients
shown in G (n ≥ 4 per genotype per treatment). Boxed area: cell population of interest. Data are from three experiments (H–J; B–F, Right panels; A, Bottom panels;
data in I and J; B–F, Right panels; A, Bottom panels are presented as mean ± SD; data in H are presented as mean ± SEM) or are from one representative of three
independent experiments (B–F, Left panels; A, Top panels). *P < 0.01; **P < 0.005; ***P < 0.001; ****P < 0.0005; ns, not significant (two-tailed Students’ t test).

motivates us to investigate the function of SRC2 in CD4+ T cells. content above the levels of WT naive and thus cannot proliferate and
Our initial in vitro analysis showed impaired SRC2fl/fl/CD4Cre CD4+ differentiate (3, 28). c-Myc regulates multiple metabolic pathways
T cell proliferation, cytokine production, and Th17 differentiation to fulfill the bioenergetic and biosynthetic demands of proliferating
in response to TCR stimulation, indicating defective T cell activa- T cells. One of the c-Myc-regulated pathways is to promote the
tion. We observed more severe defects in CD4+ T cell activation uptake of amino acids from the environment by stimulating the
in vivo analysis. CD4+ T cells from OT-II/SRC2fl/fl/CD4Cre mice expression of amino acid transporters. T cells deficient in c-Myc fail
could not proliferate in vivo upon antigen (Ova) stimulation. to up-regulate several amino acid transporters, including Slc7a5 and
Similarly, CD4+ T cells from SRC2fl/fl/CD4Cre mice were unable to Slc1a5, which are the most abundantly expressed on the surface of
induce EAE or colitis. We barely recovered SRC2fl/fl/CD4Cre CD4+ activated T cells (3). The functional importance of c-Myc-dependent
T cells from CNS in case of EAE or the gut in case of colitis, sug- upregulation of amino acid transporters is demonstrated by the lack
gesting that CD4+ T cells deficient of SRC2 did not migrate to the of T cell proliferation upon stimulation if Slc7a5 or Slc1a5 gene is
target tissues where they cause damage; additionally, these SRC2fl/ deleted (3–6). In this study, we revealed a previously unknown func-
fl
/CD4Cre CD4+ T cells also failed to up-regulate T cell activation tion of SRC2 in c-Myc-mediated upregulation of Slc7a5 during
markers. Such dramatic defects in CD4+ T cell activation were not CD4+ T cell activation. T cells deficient of SRC2 or knockdown of
observed in SRC1- or SRC3-deficient cells (13), suggesting that SRC2 have lower levels of Slc7a5 and Slc7a5-dimerizing partner
other members of SRC family coactivators, although highly con- CD98, similar to c-Myc deficient T cells (3), suggesting the critical
served, cannot compensate for the function of SRC2. Our results role of SRC2 and c-Myc in up-regulating Slc7a5; SRC2 and c-Myc
demonstrate that SRC2 plays the most important role in CD4+ formed complexes in activated CD4+ T cells. Further, SRC2, together
T cells among all three SRC family members. with c-Myc maximally stimulates c-Myc reporter activity. These
Naive T cells express minimum levels of c-Myc, whereas T cell results support that SRC2 functions as a coactivator for c-Myc in
activation greatly stimulates c-Myc expression. The proliferation of stimulating Slc7a5 expression. Lastly, forced expression of Slc7a5 in
activated T cells critically depends on increased c-Myc, which ampli- CD4+ T cells deficient in SRC2 rescues CD4+ T cell proliferation
fies gene transcription and protein synthesis (8, 34). In response to and IL-17A production as well as the ability of the T cells to induce
TCR stimulation, T cells lacking c-Myc fail to increase protein EAE. Thus, SRC2 and c-Myc-mediated upregulation of Slc7a5 is

PNAS 2023 Vol. 120 No. 18 e2221352120 https://doi.org/10.1073/pnas.2221352120 9 of 12


absolutely required for CD4+ T cell activation and CD4+ T cell- from the Jackson Laboratory. SRC2fl/fl mice were obtained from Jianming Xu Lab
dependent immune responses in vivo. Although c-Myc is necessary (Mol & Cell Biology, Baylor College of Medicine, TX). OT-II mice were obtained from
for upregulation of both Slc7a5 and Slc1a5, T cells deficient in SRC2 Jianhua Yu Lab (Department of Hematology & Hematopoietic Cell Transplantation,
only fail to up-regulate Slc7a5 but not Slc1a5, suggesting a selective City of Hope, CA).
function of SRC2 in c-Myc-regulated gene expression. While previ-
Plasmids. The retroviral vector murine stem cell virus (MSCV)-IRES-GFP was
ous reports established c-Myc as a general amplifier of gene transcrip-
a gift from W.S. Pear (University of Pennsylvania, PA). MSCV-Slc7a5-IRES-GFP
tion (8, 34), our results indicate that c-Myc recruits different
and MSCV-SRC2-IRES-GFP were made by cloning cDNA encoding Slc7a5 and
coactivators to regulate different gene expressions. Ncoa2 into MSCV-IRES-GFP vector. PIG-Myc (Plasmid # 177650), HA-HIF1alpha
One interesting observation is that SRC2fl/fl/CD4Cre CD4+ T cells P402A/P564A-pBabe-puro (Plasmid #19005), pBABE-puro (Plasmid #1764),
display more dramatic defective T cell activation when activated 7×E-Box::Renilla (Plasmid #124532), retro-gRNA-eGFP (Plasmid# 116926), and
in vivo than activated by CD3/28 cross-linking in vitro. Our results lentiCRISPR v2 (Plasmid# 52961) were purchased from Addgene.
support that two factors contribute to the relatively more severe
+
phenotypes in T cell activation in vivo: 1) physiologically relevant Isolation of Naive CD4+ Cells and Differentiation. Naive CD4 T cells were iso-
stimulation in vivo. Activation of SRC2fl/fl/CD4cre CD4+ T cells by lated from mice spleens by negative selection using a Naive CD4+ T Cell Isolation
Ova-loaded APCs displayed more severe defective T cell activation Kit (Miltenyi Biotec). Suspensions of 5 × 105 cells/mL in RPMI-1,640 medium
including reduced activation marker CD25, decreased CD98 lev- (Corning Inc) containing 2 mM L-glutamine, 50 μM β-mercaptoethanol, 100
els, and increased Th1 differentiation. This result suggests that U/mL penicillin, 100 mg/mL streptomycin, and 10% Fetal Bovine Serum (FBS)
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

strong stimulation by CD3/CD28 antibodies cross-linking in vitro (Corning Inc) were cultured in 24-well or 48-well plates precoated with 0.1 mg/
partially overcomes the defective T cell activation of SRC2fl/fl/CD4cre mL rabbit anti-hamster. The medium was supplemented with 0.25 μg/mL hamster
T cells; 2) limited nutrition availability in vivo. We observed more anti-CD3 and 0.5 μg/mL hamster anti-CD28 and followed by incubation with
severe defective activation of SRC2fl/fl/CD4cre T cells including polarizing cytokines.
decreased proliferation, reduced activation marker CD25, and In Vivo Activation and Proliferation of Naive CD4+ Cells. For lymphopenia-in-
decreased CD98 levels when they were stimulated in a nutrition- duced proliferation, a total of 4 × 105 naive CD4+ cells from SRC2fl/fl or SRC2fl/fl/
restricted medium. These results confirm long-known differences CD4-Cre mice were intraperitoneally injected into sex-matched Rag1−/− mice. For
between in vitro and in vivo T cell activation. antigen-specific stimulation and activation, naive CD4+ cells from OT-II/SRC2fl/fl
Uncontrolled activation of T cells is responsible for various auto- or OT-II/SRC2fl/fl/CD4Cre mice were labeled with 2.5 μM CellTrace™ Violet (CTV)
immune diseases such as multiple sclerosis, type 1 diabetes, and (C34557, Invitrogen) and a total of 3 × 106 cells were intraperitoneally injected
immunological bowel disease (35, 36). On the other hand, failed into sex-matched Rag1−/− mice or a total of 2 × 104 cells were intraperitoneally
T cell activation leads to severe immunodeficiency and cancer injected into sex-matched C57BL WT mice. Then, 10 μg OVA peptide (Ovalbumin
growth. Therefore, current drugs such as checkpoint inhibitors or (323 to 339), O1641-1MG, Sigma) mixed with Alum Adjuvant (7,7161, Thermo
cyclosporin A either boost or inhibit T cell activation to treat cancer Scientific™) at a 1:1 ratio was also intraperitoneally injected into the recipient
and prevent transplantation rejection, respectively. Our results mice.
demonstrated the essential function of SRC2 in T cell activation
and the potential clinical impact of targeting SRC2 to control T cell In Vitro Activation and Proliferation of Naive CD4+ Cells. First, 5 × 105
+
activation. Members of the SRC family are considered oncogenes, cells/mL naive CD4 cells labeled with 2.5 μM CellTrace™ Violet (CTV) (C34557,
as they play roles in tumorigenesis in different types of cancers (37, Invitrogen) were suspended in RPMI-1,640 culture medium supplemented
38). Small-molecule SRC inhibitors have already been developed with either 0.25 μg/mL hamster anti-CD3 and 0.5 μg/mL hamster anti-CD28 or
5 ng/mL PMA and 1 μg/mL ionomycin. Then, to monitor T cell activation in the
for the treatment of cancers (39, 40). It would be interesting to test
presence of APCs, splenic cells from OT-II/SRC2fl/fl or OT-II/SRC2fl/fl/CD4Cre were
whether SRC2 inhibitors can prevent the above T cell-mediated
cultured at 1 × 106 cells/mL in RPMI-1,640 culture medium supplemented with
autoimmunity by preventing T cell activation. Thus, our study not different concentrations of OVA peptide (0.1 μg/mL, 0.5 μg/mL, and 1.0 μg/mL).
only reveals an important mechanism for SRC2-regulated T cell To assess the activation and proliferation under nutrition-restricted conditions,
activation but also identifies SRC2 as a potential therapeutic target naive CD4+ T cells were cultured in a Base DMEM medium (102353-100, Agilent
for T cell-mediated autoimmune diseases. Seahorse) supplemented with 8% FBS (Corning Inc.) and 1% nonessential amino
In this study, we demonstrated the critical function of SRC2 acid (NEAA) (9304-100ML, Irvine Scientific) for 3 d.
in CD4+ T cell-mediated immune responses via regulating amino
acid transporter Slc7a5. However, the limitation of the study Induction and Assessment of EAE. For active EAE, mice were immunized with
includes forced expression of Slc7a5 in SRC2fl/fl/CD4Cre CD4+ 200 mg MOG35–55 (Hooke Laboratories) in complete Freund’s adjuvant by sub-
T cells only partially rescued T cell activation and EAE induction, cutaneous injection at two dorsal sites of mice, followed by two intraperitoneal
suggesting that there are other defects in addition to failed upreg- injections of 80 ng pertussis toxin (PTX) at days 0 and 1. For adoptive transferred
ulation of Slc75a. For instance, we noticed a lower ATP produc- EAE, 5 × 106 CD4+ cells were adoptively transferred to sex-matched Rag1−/− mice.
tion whereas higher or normal extracellular acidification rate Three weeks post cell transfer, EAE was induced in Rag1−/− recipient mice by
(ECAR) and oxygen consumption rate (OCR) levels in SRC2fl/fl/ immunizing them with MOG35–55 and PTX. For passive EAE, SRC2fl/fl or SRC2fl/fl/
CD4Cre CD4+ T cells, which implies that SRC2 plays a role in CD4Cre donor mice were immunized with MOG35–55 subcutaneously. After 11 d,
other ATP generation pathways including but not limited to beta cells were isolated from the spleen and cultured with 20 μg/mL MOG35–55 (DS-
0111, Hooke Laboratories) and 0.5 μg/mL hamster anti-CD28 for 1 d prior to retro-
oxidation, ketosis, nucleoside diphosphate kinases, and recycled
virus transduction. After transduction, the cells were cultured for another 3 d with
from ADP. This necessitates future work to investigate other
20 μg/mL MOG35–55 and 0.5 μg/mL hamster anti-CD28 under Th17-polarizing
SRC2-regulated pathways in CD4+ T cell-mediated immune conditions (2 ng/mL TGF-β, 20 ng/mL IL-6, and 20 ng/mL IL-23). Rag1 recipient
responses. mice were then transferred intraperitoneally with 2.0 × 107 MOG35–55-specific
Th17 cells. The severity of EAE was monitored and evaluated on a scale from 0 to
Materials and Methods 5 according to Hooke Laboratories' guidelines.
Detailed Materials and Methods can be found in SI Appendix.
C. rodentium Infection. Mice aged 8 to 12 wk were subjected to gavage with a
Mice. Transgenic CD4
Cre
(TgCd4cre, 022071), Rag1−/− (Rag1tm1Mom, 002216), 2 × 109 colony-forming unit of C. rodentium in 200 μL Phosphate-Buffered Saline
Cas9 (Rosa26LSL-Cas9, 028551), and C57BL (B6, 000664) mice were purchased (PBS). Mice were weighed immediately following C. rodentium gavage and every

10 of 12 https://doi.org/10.1073/pnas.2221352120 pnas.org
other day thereafter. Three weeks after C. rodentium infection, the colon, spleen, CRISPR/Cas9-Mediated Genomic DNA Deletion. Pairs of primers containing
and iLN were removed from mice for analysis. sequences of nontargeting control (mouse) sgRNAs (sgNTC), sgRNAs target-
−/− ing mouse c-Myc (sgMyc), and sgRNAs targeting mouse SRC2 (sgSRC2) were
Adoptive Transfer Colitis. Colitis was induced in sex-matched Rag1 mice by designed and cloned into a retro-gRNA-eGFP vector. Naive CD4+ cells were iso-
intraperitoneally injecting 4 × 105 naive CD4+ T cells purified from the spleen of lated from the spleen of Cas9 mice and transduced with the retrovirus contain-
SRC2fl/fl or SRC2fl/fl/CD4Cre mice (8 to 10 wk old). Mice were weighed immedi- ing sgNTC, sgMyc, and sgSRC2, respectively. The sgRNA sequences are listed in
ately following T cells transfer and weekly thereafter. SI Appendix, Table S1.
Intracellular ATP Determination. Intracellular ATP production was deter- Statistics and Reproducibility. The results were analyzed for statistical signifi-
mined by Molecular Probes’ ATP Determination Kit (A22066, Invitrogen). In brief, cance with unpaired Student’s t test. All data are presented as mean ± SD or mean
CD4+ cells stimulated by 0.25 μg/mL hamster anti-CD3 and 0.5 μg/mL hamster ± SEM. P values are calculated using GraphPad Prism and presented where the
anti-CD28 were harvested, counted, washed with PBS, and lysed. Luciferase activ- statistical significance (P < 0.05) was found.
ity was measured by Synergy HTX multi-mode reader (BioTek).
Data, Materials, and Software Availability. All study data are included in the
Metabolic Assays. The Agilent Seahorse XF Glycolysis Stress Test Kit (103020-
article and/or SI Appendix. The high-throughput sequence has been deposited in
100, Agilent Technology) and XF Cell Mito Stress Test Kit (103015-100, Agilent Gene Expression Omnibus with accession no. GSE229633 (41).
Technology) were used to test the ECAR in glycolysis and the OCR of activated
CD4+ cells. Data were analyzed using Seahorse Wave software. ACKNOWLEDGMENTS. We thank W. S. Pear for sharing the plasmids of Murine
Stem Cell Virus/Internal Ribosome Entry Site/Green Fluorescent Protein (MSCV-
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

6
Luciferase Assay. A total of 2 × 10 HEK 293T cells were seeded to each well IRES-GFP). We also thank J. Xu and J. Yu for sharing the mice strain of SRC2fl/fl and
of a 6-well plate and transfected with the reporter vectors 7 × E-Box::Renilla OT-II. Besides, we appreciate the help from the following City of Hope core facili-
(400 ng), firefly luciferase vector (200 ng), and expression vectors (2 μg) using ties: Animal Resource Center, Integrative Genomics Core, Pathology Solid Tumor
BioT transfection reagent (Bioland Scientific, Paramount, CA). Core, and Bioinformatics Core. We thank Dr. Sung Hee Kil for critically reviewing
+
RNA-Seq and Analysis. Naive CD4 T cells isolated from the spleen of SRC2fl/fl or and editing the manuscript. Graphical figures were created using BioRender.
SRC2 /CD4 mice were adoptively transferred to sex-matched Rag1−/− mice.
fl/fl Cre
com. This work was supported by grants from NIH R01-AI109644, R21-AI163256,
Four weeks post adoptively transfer, spleen, iLN, and mLN were removed from institutional pilot funding, Jackie and Bruce Barrow Cancer Research Scholars’
Rag1−/− recipient mice, and CD4+ cells were purified by negative selection using Program, and Caltech-CoH Biomedical Initiative. Research reported in this publi-
CD4+ T Cell Isolation Kit (Miltenyi Biotec) and subjected to RNA extraction. Each cation included work performed in the animal, genomic, and flow cytometry cores
group has four replicates from different mice. Quality control, library prepara- supported under NIH grant P30CA033572. The content is solely the responsibility
tion, and sequencing were performed at Novogene. The analysis was performed of the authors and does not necessarily represent the official views of the NIH.
through Partek® Flow®.
Amino Acid Uptake Assay. The amino acid uptake capacity of CD4+ cells Author affiliations: aDepartment of Immunology & Theranostics, Arthur Riggs Diabetes &
was assessed using the amino acid uptake assay kit (#UP04–12, DOJINDO Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte,
CA 91010; bDepartment of Immuno-Oncology, Beckman Research Institute of the City
Laboratories). Briefly, CD4+ cells activated for 24 h were subjected for analysis. of Hope, Duarte, CA 91010; cDepartment of Diabetes Complication and Metabolism,
Boronophenylalanine (BPA) was used as amino acid analogs, and the BPA uptake Arthur Rigs Diabetes & Metabolism Research Institute, Beckman Research Institute of
capacity was assessed using BPA-probe. The median fluorescent intensity (MFI) the City of Hope, Duarte, CA 91010; dDepartment of Physiology, David Geffen School
of Medicine, University of California, Los Angeles, CA 90095; eDivision of Translational
of the complex of BPA with BPA-probe in the cells was assessed using a flow Bioinformatic, Bioinformatics Core, City of Hope, Duarte, CA 91010; and fDepartment of
cytometer. Gynecology and Obsterics, School of Medicine, Emory University, Atlanta, GA 30322

1. J. E. Smith-Garvin, G. A. Koretzky, M. S. Jordan, T cell activation. Annu. Rev. Immunol. 27, 591–619 17. W. Zhang et al., Steroid nuclear receptor coactivator 2 controls immune tolerance by promoting
(2009). induced treg differentiation via up-regulating Nr4a2. Sci. Adv. 8, eabn7662 (2022).
2. W. Ren et al., Amino-acid transporters in T-cell activation and differentiation. Cell Death Dis. 8, e2757 18. S. D. Miller, W. J. Karpus, Experimental autoimmune encephalomyelitis in the mouse. Curr. Protoc.
(2017). Immunol. Chapter 15, Unit 15–Unit 11 (2007).
3. J. M. Marchingo, L. V. Sinclair, A. J. Howden, D. A. Cantrell, Quantitative analysis of how Myc controls 19. C. P. Simmons et al., Central role for B lymphocytes and CD4+ T cells in immunity to infection by the
T cell proteomes and metabolic pathways during T cell activation. elife 9, 53725 (2020). attaching and effacing pathogen citrobacter rodentium. Infect. Immun. 71, 5077–5086 (2003).
4. K. Hayashi, P. Jutabha, H. Endou, H. Sagara, N. Anzai, LAT1 is a critical transporter of essential amino 20. B. A. Vallance, W. Deng, L. A. Knodler, B. B. Finlay, Mice lacking T and B lymphocytes develop
acids for immune reactions in activated human T cells. J. Immunol. 191, 4080–4085 (2013). transient colitis and crypt hyperplasia yet suffer impaired bacterial clearance during citrobacter
5. L. V. Sinclair et al., Control of amino-acid transport by antigen receptors coordinates the metabolic rodentium infection. Infect. Immun. 70, 2070–2081 (2002).
reprogramming essential for T cell differentiation. Nat. Immunol. 14, 500–508 (2013). 21. R. Basu et al., Th22 cells are an important source of IL-22 for host protection against
6. M. Nakaya et al., Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of enteropathogenic bacteria. Immunity 37, 1061–1075 (2012).
glutamine uptake and mTORC1 kinase activation. Immunity 40, 692–705 (2014). 22. S. Omenetti et al., The intestine harbors functionally distinct homeostatic tissue-resident and
7. B. B. Au-Yeung et al., IL-2 modulates the TCR signaling threshold for CD8 but not CD4 T cell inflammatory Th17 cells. Immunity 51, 77–89.e76 (2019).
proliferation on a single-cell level. J. Immunol. 198, 2445–2456 (2017). 23. K. Eden, Adoptive transfer colitis. Methods Mol. Biol. 1960, 207–214 (2019).
8. C. Y. Lin et al., Transcriptional amplification in tumor cells with elevated c-Myc. Cell 151, 56–67 24. J. Dunn, M. H. Grider, “Physiology, adenosine triphosphate” in StatPearls (StatPearls Publishing,
(2012). Treasure Island, FL, 2022).
9. K. Singh et al., c-MYC regulates mRNA translation efficiency and start-site selection in lymphoma. J. 25. J. A. Shyer, R. A. Flavell, W. Bailis, Metabolic signaling in T cells. Cell Res. 30, 649–659 (2020).
Exp. Med. 216, 1509–1524 (2019). 26. L. Z. Shi et al., HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the
10. V. H. Cowling, M. D. Cole, The Myc transactivation domain promotes global phosphorylation of the differentiation of TH17 and Treg cells. J. Exp. Med. 208, 1367–1376 (2011).
RNA polymerase II carboxy-terminal domain independently of direct DNA binding. Mol. Cell Biol. 27. E. V. Dang et al., Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell 146, 772–784
27, 2059–2073 (2007). (2011).
11. M. Yue, J. Jiang, P. Gao, H. Liu, G. Qing, Oncogenic MYC activates a feedforward regulatory loop 28. R. Wang et al., The transcription factor Myc controls metabolic reprogramming upon T lymphocyte
promoting essential amino acid metabolism and tumorigenesis. Cell Rep. 21, 3819–3832 (2017). activation. Immunity 35, 871–882 (2011).
12. C. A. Walsh, L. Qin, J. C. Tien, L. S. Young, J. Xu, The function of steroid receptor coactivator-1 in 29. H. Tan et al., Integrative proteomics and phosphoproteomics profiling reveals dynamic signaling
normal tissues and cancer. Int. J. Biol. Sci. 8, 470–485 (2012). networks and bioenergetics pathways underlying T cell activation. Immunity 46, 488–503 (2017).
13. S. Sen et al., SRC1 promotes Th17 differentiation by overriding Foxp3 suppression to stimulate 30. T. Lindsten, C. H. June, C. B. Thompson, Multiple mechanisms regulate c-myc gene expression
RORgammat activity in a PKC-theta-dependent manner. Proc. Natl. Acad. Sci. U.S.A. 115, E458–E467 during normal T cell activation. EMBO J. 7, 2787–2794 (1988).
(2018). 31. J. Cantor, M. Slepak, N. Ege, J. T. Chang, M. H. Ginsberg, Loss of T cell CD98 H chain specifically
14. Z. He et al., SRC3 is a cofactor for RORgammat in Th17 differentiation but not thymocyte ablates T cell clonal expansion and protects from autoimmunity. J. Immunol. 187, 851–860 (2011).
development. J. Immunol. 202, 760–769 (2019). 32. M. C. Poffenberger, R. G. Jones, Amino acids fuel T cell-mediated inflammation. Immunity 40,
15. K. Tanaka et al., Regulation of pathogenic T helper 17 cell differentiation by steroid receptor 635–637 (2014).
coactivator-3. Cell Rep. 23, 2318–2329 (2018). 33. H. Jensen, M. Potempa, D. Gotthardt, L. L. Lanier, Cutting edge: IL-2-induced expression of the
16. B. C. Nikolai et al., Steroid receptor coactivator 3 (SRC-3/AIB1) is enriched and functional in mouse amino acid transporters SLC1A5 and CD98 is a prerequisite for NKG2D-mediated activation of
and human tregs. Sci. Rep. 11, 3441 (2021). human NK cells. J. Immunol. 199, 1967–1972 (2017).

PNAS 2023 Vol. 120 No. 18 e2221352120 https://doi.org/10.1073/pnas.2221352120 11 of 12


34. Z. Nie et al., c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem 38. S. Suresh et al., SRC-2-mediated coactivation of anti-tumorigenic target genes suppresses MYC-
cells. Cell 151, 68–79 (2012). induced liver cancer. PLoS Genet. 13, e1006650 (2017).
35. Z. Huang, H. Xie, R. Wang, Z. Sun, Retinoid-related orphan receptor gamma t is a potential 39. X. Song et al., Development of potent small-molecule inhibitors to drug the undruggable steroid
therapeutic target for controlling inflammatory autoimmunity. Expert Opin. Ther. Targets 11, receptor coactivator-3. Proc. Natl. Acad. Sci. U.S.A. 113, 4970–4975 (2016).
737–743 (2007). 40. Y. Wang et al., Small molecule inhibition of the steroid receptor coactivators, SRC-3 and SRC-1. Mol.
36. J. Zhu, H. Yamane, W. E. Paul, Differentiation of effector CD4 T cell populations (*). Annu. Rev. Endocrinol. 25, 2041–2053 (2011).
Immunol. 28, 445–489 (2010). 41. Z. Sun, W. Zhang, SRC2 controls CD4+ T cell activation via stimulating c-Myc-mediated up-
37. A. D. Rohira et al., Targeting SRC coactivators blocks the tumor-initiating capacity of cancer stem-like regulation of amino acid transporterSlc7a5. Gene Expression Omnibus. https://www.ncbi.nlm.nih.
cells. Cancer Res. 77, 4293–4304 (2017). gov/geo/query/acc.cgi?acc=GSE229633. Deposited 12 April 2023.
Downloaded from https://www.pnas.org by TEXAS MEDICAL CENTER LIBRARY TMC SERIALS DEPT on October 12, 2023 from IP address 128.249.96.51.

12 of 12 https://doi.org/10.1073/pnas.2221352120 pnas.org

You might also like