You are on page 1of 15

Articles

Investigating sex differences in T regulatory cells from


cisgender and transgender healthy individuals and patients
with autoimmune inflammatory disease: a cross-sectional
study
George A Robinson, Junjie Peng, Hannah Peckham, Gary Butler, Ines Pineda-Torra*, Coziana Ciurtin*, Elizabeth C Jury*

Summary
Lancet Rheumatol 2022; Background Sexual dimorphisms, which vary depending on age group and pubertal status, have been described across
4: e710–24 both the innate and adaptive immune system. We explored the influence of sex hormones on immune phenotype in
Published Online the context of adolescent health and autoimmunity.
August 31, 2022
https://doi.org/10.1016/
S2665-9913(22)00198-9 Methods In this cross-sectional study, healthy, post-pubertal cisgender individuals (aged 16–25 years); healthy, pre-
See Comment page e652
pubertal cisgender individuals (aged 6–11 years); transgender individuals (aged 18–19 years) undergoing gender-
*Senior authors
affirming treatment (testosterone in individuals assigned female sex at birth and oestradiol in individuals assigned
male sex at birth); and post-pubertal cisgender individuals (aged 14–25 years) with juvenile-onset systemic lupus
Centre for Rheumatology
Research (G A Robinson PhD, erythematosus (SLE) age-matched to cisgender individuals without juvenile-onset SLE were eligible for inclusion.
C Ciurtin PhD, Prof E C Jury PhD), Frequencies of 28 immune-cell subsets (including different T cell, B cell, and monocyte subsets) from each participant
Centre for Adolescent were measured in peripheral blood mononuclear cells by flow cytometry and analysed by balanced random forest
Rheumatology Versus Arthritis
(G A Robinson, J Peng MSc,
machine learning. RNA-sequencing was used to compare sex and gender differences in regulatory T (Treg) cell
H Peckham MSc, C Ciurtin, phenotype between participants with juvenile-onset SLE, age-matched cis-gender participants without the disease,
Prof E C Jury), and Centre for and age matched transgender individuals on gender-affirming sex hormone treatment. Differentially expressed genes
Cardiometabolic and Vascular were analysed by cluster and pathway analysis. Suppression assays assessed the anti-inflammatory function of Treg
Science
(Prof I Pineda-Torra PhD),
cells in vitro.
Division of Medicine,
University College London, Findings Between Sept 5, 2012, and Nov 6, 2019, peripheral blood was collected from 39 individuals in the post-
London, UK; Department of pubertal group (17 [44%] cisgender men, mean age 18·76 years [SD 2·66]; 22 [56%] cisgender women, mean age
Paediatric and Adolescent
Endocrinology, University
18·59 years [2·81]), 14 children in the cisgender pre-pubertal group (seven [50%] cisgender boys, mean age 8·90
College London Hospital and [1·66]; seven [50%] cisgender girls, mean age 8·40 [1·58]), ten people in the transgender group (five [50%]
Great Ormond Street Institute transgender men, mean age 18·20 years [0·47]; five [50%] transgender women, mean age 18·70 years [0·55]), and
of Child Health, University 35 people in the juvenile-onset SLE group (12 [34%] cisgender men, mean age 18·58 years [2·35]; 23 [66%]
College London, London, UK
(Prof G Butler MD); Gender
cisgender women, mean age 19·48 [3·08]). Statistically significantly elevated frequencies of Treg cells were one of
Identity Development Service, the top immune-cell features differentiating young post-pubertal cisgender men from similarly aged cisgender
Tavistock and Portman NHS women (p=0·0097). Treg cells from young cisgender men had a statistically significantly increased suppressive
Foundation Trust, London, UK capacity in vitro compared with those from cisgender women and a distinct transcriptomic signature significantly
(Prof G Butler)
enriched for genes in the PI3K–AKT signalling pathway. Gender-affirming sex hormones in transgender men and
Correspondence to:
Prof Elizabeth C Jury, Centre for
transgender women induced multiple statistically significant changes in the Treg-cell transcriptome, many of
Rheumatology Research, which enriched functional pathways that overlapped with those altered between cisgender men and cisgender
Division of Medicine, University women, highlighting a hormonal influence on Treg-cell function by gender. Finally, sex differences in Treg-cell
College London, London, frequency were absent and suppressive capacity was reversed in patients with juvenile-onset SLE, but sex
W1CE 6JF, UK
e.jury@ucl.ac.uk
differences in Treg-cell transcriptional signatures were significantly more pronounced in patients with juvenile-
onset SLE compared with individuals without juvenile-onset SLE, suggesting that sex hormone signalling could be
dysregulated in autoimmunity.

Interpretation Sex-chromosomes and hormones might drive changes in Treg-cell frequency and function. Young
post-pubertal men have a more anti-inflammatory Treg-cell profile, which could explain inflammatory disease
susceptibilities, and inform sex-tailored therapeutic strategies.

Funding Versus Arthritis, UK National Institute for Health Research University College London Hospital Biomedical
Research Centre, Lupus UK, and The Rosetrees Trust.

Copyright © 2022 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0
license.

e710 www.thelancet.com/rheumatology Vol 4 October 2022


Articles

Research in context
Evidence before this study Added value of this study
Sex differences in the innate and adaptive immune response To our knowledge, this study is the first to report an in-depth
result in differential responses to infection, vaccination, and analysis of immune-cell phenotype comparing sex and gender
susceptibility to autoimmune diseases. Studies have reported differences between young (aged 14–25 years) post-pubertal
different sex specific observations by age group regarding cisgender men and cisgender women both with and without
inflammatory profiles and disease presentation. This is juvenile-onset SLE, and in transgender individuals undergoing
particularly relevant for juvenile-onset systemic lupus gender-affirming sex hormone treatment who did not have
erythematosus (SLE), which predominantly effects young juvenile-onset SLE. During adolescence, rapid changes in sex
women and has a common onset at puberty. We searched hormones drive changes in inflammatory responses and disease
PubMed, Web of Science, and Google Scholar for research susceptibility. Therefore, this age range is important in the
articles published between Jan 1, 1990, and April 30, 2022, context of autoimmune research. We found unique changes in
using the search terms “sex”, “gender”, “transgender”, “sex the immune profile, specifically in regulatory T cells, between
hormones”, “sex chromosomes”, “inflammation”, “immune cisgender men and cisgender women and sex hormone-
response”, “(juvenile-onset) systemic lupus erythematosus”, associated transcriptomic profiles that overlap by gender, which
and “autoimmunity”. We also searched for research articles could play a role in juvenile-onset SLE pathogenesis.
published in the same time window in the top rated
Implications of all the available evidence
rheumatology, immunology, and endocrine-specific journals by
The evidence from this study could be used to improve our
impact factor. Published abstracts were excluded from the
understanding of sexual dimorphisms in immune responses by
searches. The earliest referenced article was published in 2003.
sex to improve sex-specific and gender-specific therapeutic
We found that investigating the role of sex and gender in
strategies in health and disease and to understand sex
immunological research is aiding the understanding of sexual
differences in inflammatory responses, vaccine efficacy, and
dimorphisms in the immune system in both healthy individuals
autoimmune disease susceptibility. We also highlight specific
and in diseases with a sex bias. However, disparities across
immune features and genes that could be targeted
published results have emerged, probably because of
therapeutically or used for sex-specific diagnosis and
differences in study age groups, pubertal status, gender
investigation of juvenile-onset SLE pathogenic mechanisms.
consideration, and use of different animal models.

Introduction due to significant sex bias (nine female patients to every


Men and women differ in their inflammatory response to one male patient) and frequent disease onset during
non-self and self antigens; consequently, they have childbearing age (15–44 years). Between 15% and 20% of
different responses to vaccination and risks of infection.1 all patients with SLE have juvenile-onset disease (onset
This has been highlighted by the COVID-19 pandemic2 before the patient reaches 18 years old). Juvenile-onset
and historically by the risk developing autoimmune SLE often starts at puberty and has a more severe disease
diseases: men are generally more likely to have a more phenotype than adult onset disease.8 Together, this
severe disease course following SARS-CoV-2 infection, highlights a key role for sex hormones in susceptibility to
whereas women represent about 80% of patients with juvenile-onset SLE.
autoimmunity.1,3 Sex differences across the immune Investigating the relationship between sex hormones
system, including fundamental differences in the and inflammation is important for understanding the
frequency and activity of T-cell subsets, have been cause of autoinflammatory diseases. Many clinical trials
described by gender across ethnicities.1,4 Generally, do not consider sex or gender in recruitment and
previous observations show higher CD4+ T-cell and lower outcome measures:9 historically less than 10% of
CD8+ T-cell frequencies4,5 and higher proinflammatory, immunological studies analysed data by sex.10 The
cytotoxic, and antiviral T-cell responses in women absence of consideration of sex in research is particularly
compared with men. Of note, some sex differences in important for the study of autoimmune diseases, which
adaptive immunity are present throughout life whereas are more prevalent in women.3 We aimed to investigate
others manifest after the onset of puberty or post- the influence of sex chromosomes and hormones in
reproductive senescence, implicating both genetic and driving sexual dimorphisms in inflammatory profiles
sex hormonal influences.7 across different sex, gender, age, and disease status
Systemic lupus erythematosus (SLE) is a complex groups.
autoimmune disorder characterised by loss of immune
cell regulation and the presence of autoantibodies against Methods
nuclear and non-nuclear antigens, resulting in chronic Study design and participants
inflammation and organ damage. Sex hormones have In this cross-sectional study healthy, post-pubertal
been hypothesised to be implicated in the cause of SLE cisgender individuals (aged 16–25 years); healthy,

www.thelancet.com/rheumatology Vol 4 October 2022 e711


Articles

pre-pubertal cisgender individuals (aged 6–11 years); analysis and sequencing was done by UCL Genomics
transgender individuals (aged 18–19 years) undergoing (University College London, London, UK). Libraries were
gender-affirming treatment (testosterone in individuals prepped with the NEB Low input kit (New England
assigned female sex at birth and oestradiol in individuals Biolabs, Ipswich, MA, USA) and were sequenced on the
assigned male sex at birth); and post-pubertal cisgender NovaSeq SP flow-cell (Illumina, San Diego, CA, USA)
individuals (aged 14–25 years) with juvenile-onset SLE age- with a 100 base pair single read. Reads were demultiplexed
matched to individuals without juvenile-onset were and converted to fastq files with Illumina’s bcl2fastq
eligible for inclusion. Transgender individuals were conversion software (version 2.19) and analysis was done
contacted for participation via University College London by UCL Genomics. DEGs that were significantly different
Hospital’s (London, UK) young people’s Gender Identity between cohorts, some of which were expanded using
Development Service liaison endocrine clinic, and first-order protein–protein interaction networks, were
participants with juvenile-onset SLE were contacted for analysed by hierarchical clustering (Pearsons), sparce
inclusion via the Rheumatology Clinic at University partial least squares discriminant analysis, open target
College London Hospital. Patients were eligible to be disease association analysis, and gene ontology pathway
included if they had juvenile-onset SLE according to the analysis to investigate differentially regulated immune
American College of Rheumatology revised classification functions and associations in Treg cells (appendix 1 p 6).
criteria for lupus (1997) or the Systemic Lupus Inter­
national Collaborating Clinics (SLICC) criteria (2012), and Statistical analysis
were diagnosed before the age of 18 years. For cisgender For immunophenotyping, the analysis was exploratory
inviduals without juvenile-onset SLE, volunteers who did based on limited sample availability, as a result of
not have the disease or who had non-inflammatory, non- working with rare cohorts of young individuals and
infective conditions (eg, referred for assessment of non- patients. For RNA sequencing, a sample estimation was
inflammatory musculoskeletal conditions) were recruited. carried out using a 90% power calculation (p<0·05) of
Additional inclusion criteria for post-pubertal cohorts was data from a Treg-cell suppression assay (Treg-cell
a puberty Tanner stage of 4-5. Any patient who withheld functional readout), using the proportion of Tresp cells
consent or whose carer withheld consent for inclusion suppressed in cisgender men versus cisgender women
(as appropriate given patient’s competence) and any at a 1:1 Treg-cell to Tresp-cell ratio, yielding the required
patient who withdrew from the study were excluded from sample size of five participants per group (appendix 1
the analyses. p 5). This indicated the number of samples needed to
Ethics approval for the study was obtained see functional transcriptomic changes in Treg cells by
from the London-Harrow Research Ethics Committee sex. Statistical analyses were done on the basis of the
(REC11/LO/0330). Informed written consent was hypothesis that men and women have altered immune
See Online for appendix 1 acquired from all participants (appendix 1 p 1). cell profiles, including Treg-cell phenotype, resulting in
variation in inflammatory disease risk by sex and
Procedures gender. Statistical analyses were done using GraphPad
Peripheral blood mononuclear cell (PMBC) samples were Prism (version 9). Demographic, clinical, immune
taken from cisgender individuals with and without frequency, DEG normalised count, and Treg-cell
juvenile-onset SLE and transgender individuals without suppressive capacity measures data were tested for
juvenile-onset SLE. The samples were assessed for normal distribution. Normally distributed data were
28 immune-cell subsets (including different T-cell, B-cell, assessed with parametric tests; data that were not
and monocyte subsets) by multiparameter flow cytometry, normally distributed data were assessed with non-
as described previously11 and in appendix 1 (pp 2–4). parametric tests. For demographic, clinical, immune
To assess the suppressive capacity of regulatory T (Treg) phenotype, gene expression, and Treg-cell suppression
cells in post-pubertal cisgender individuals with and continuous data, unpaired t tests were used when
without juvenile-onset SLE and transgender individuals comparing two groups and one-way ANOVA (Turkey’s
without juvenile-onset SLE, fluorescence-activated cell post-hoc test) tests were used when comparing more
sorting (FACS) was used to sort Treg cells, responder than two groups. For continuous data that were not
T (Tresp) cells, and monocytes for in-vitro suppression normally distributed, Mann-Whitney tests were used
assays (appendix 1 p 5) using stored PBMCs from study when comparing two groups. For binary data Fisher’s
participants collected during the research project. Treg- exact tests were used to compare two groups and χ² tests
cell purity was confirmed by FOXP3 intracellular staining were used when comparing more than two groups.
(appendix 1 p 5). p values less than 0·050 were considered significant
RNA sequencing was used to assess differentially unless mentioned otherwise. Multiple testing was
expressed genes (DEGs) and gene ontology pathways accounted for using the false discovery rate adjustment
between different sexes and genders with and without for multiple comparisons Benjamini, Krieger, and
juvenile-onset SLE. Total RNA was isolated from FACS- Yekutieli method to calculate p values for immune
sorted Treg cells (appendix 1 pp 5–6). Quality control phenotype data t tests. The balanced random forest

e712 www.thelancet.com/rheumatology Vol 4 October 2022


Articles

Cisgender and Cisgender and p value Cisgender and Cisgender and p value
transgender transgender transgender transgender
men women men women

Cisgender post-puberty (Continued from previous column)


Number of participants 17 22 ·· Tanner Stage at time of sample
Age (years) 18·76 (2·66) 18·59 (2·81) 0·85† Tanner stage 1 0 0 >1·00*
Ethnicity Tanner stage 2–3 0 0 >1·00*
White 8 (47%) 12 (55%) 0·75* Tanner stage 4–5 12 (100%) 23 (100%) >1·00*
Asian 6 (35%) 4 (18%) 0·14* Clinical feature
Black 1 (6%) 2 (9%) 1·00* Age at disease onset 13 (5·37) 12 (3·00) 0·32†
Other 2 (12%) 4 (18%) 0·68* (years)
Tanner Stage at time of sample SLEDAI 2·67 (2·87) 3·21 (3·18) 0·62†
Tanner stage 1 0 0 >1·00* Neurological 1 (8%) 1 (4%) 1·00*
involvement
Tanner stage 2–3 0 0 >1·00*
Serositis involvement 2 (17%) 6 (26%) 0·31*
Tanner stage 4–5 17 (100%) 22 (100%) >1·00*
Cutaneous involvement 10 (83%) 21 (91%) 0·69*
Taking contraception 0 2 (9%) 0·50*
(combined pill) Haematological 5 (42%) 9 (39%) 1·00*
involvement
Cisgender pre-puberty
Musculoskeletal 9 (75%) 21 (91%) 0·31*
Number of participants 7 7 ·· involvement
Age (years) 8·90 (1·66) 8·40 (1·58) 0·68† Renal involvement 4 (33%) 5 (22%) 0·69*
Ethnicity Erythrocyte 14·71 (14·22) 25·26 (32·93) 0·23†
White 4 (57%) 3 (43%) >1·00* sedimentation rate
Asian 2 (29%) 3 (43%) >1·00* dsDNA titre 53·5 9 (3–350) 0·46†
Black 1 (14%) 1 (14%) >1·00* (4·75–138·8)
Other 0 0 >1·00* C3 0·93 (0·29) 1·053 (0·35) 0·28†
Tanner Stage at time of sample Lymphocyte count 1·74 (0·8) 1·68 (0·6) 0·78†
Tanner stage 1 7 (100) 7 (100%) >1·00* Treatment
Tanner stage 2–3 0 0 >1·00* Hydroxychloroquine 11 (92%) 20 (87%) 1·00*
Tanner stage 4–5 0 0 >1·00* Mycophenolate mofetil 5 (42%) 13 (57%) 0·49*
Transgender on gender affirming hormones Prednisolone 5 (42%) 12 (52%) 0·72*
Number of participants 5 5 ·· Prednisolone dose (mg) 4·00 (6·34) 3·83 (4·57) 0·72*
Age (years) 18·20 (0·47) 18·70 (0·55) 0·16† Vitamin D 2 (17%) 6 (26%) 0·19*
Ethnicity Methotrexate 1 (8%) 2 (9%) 1·00*
White 5 (100%) 5 (100) >1·00* Azathioprine 4 (33%) 3 (13%) 0·20*
Asian 0 0 >1·00* Data are mean (SD), n (%), or median (IQR). For transgender individuals, the
Black 0 0 >1·00* Tanner stage was their most recent Tanner stage before puberty blocking therapy.
Tanner stage 1 is classified as pre-puberty, Tanner stages 2–3 were classified as in
Other 0 0 >1·00* the larche or gonadarche, and Tanner stages 4–5 were classified as post-puberty.
Tanner Stage at time of puberty block For patients with juvenile-onset SLE, common clinical measures of disease are
Tanner stage 1 0 0 >1·00* shown as well as treatments. Rituximab treatment was avoided in the cohort.
C3=complement component 3. dsDNA=anti-double-stranded-DNA antibodies.
Tanner stage 2–3 1 (20%) 2 (40%) >1·00* SLE=systemic lupus erythematosus. SLEDAI=systemic lupus erythematosus
Tanner stage 4–5 4 (80%) 3 (60%) >1·00* disease activity index. *p value obtained with Fisher’s exact test. †p value
Time on gender affirming 11·7 (11·08) 12 (7·16) 0·96† obtained with unpaired t test for normally distributed data and Mann-Whitney
hormone treatment test for not normally distributed data.
(months)
Table: Demographic and clinical characteristics
Cisgender post-puberty with juvenile-onset SLE
Number of participants 12 23 ··
Age (years) 18·58 (2·35) 19·48 (3·08) 0·39†
machine-learning approach was used to assess the
Ethnicity extent of the difference in the global immune phenotype
White 3 (25%) 10 (43%) 0·46* between healthy, post-pubertal cisgender men and
Asian 6 (50%) 7 (30%) 0·29* cisgender women and to highlight the key immune cell
Black 3 (25%) 4 (17%) 0·67* variables that were driving this difference. Receiver
Other 0 2 (9%) 0·54* operator characteristic (ROC) analysis and 10-fold cross-
(Table continues in next column) validation were used to assess the balanced random
forest model performance and classification accuracy,
which was optimised and adjusted for age and ethnicity
(appendix 1 p 6).

www.thelancet.com/rheumatology Vol 4 October 2022 e713


Articles

Role of the funding source mean age 19·48 [3·08]; late puberty Tanner stage 4–5).
The funding sources had no role in the study design, Participant characters are reported in the table.
data collection, analysis, interpretation, or writing of the Following optimisation and adjustment for age and
report. ethnicity, ROC analysis of the balanced random forest
model showed a strong classification accuracy (84·76%)
Results and good model efficiency in discriminating cisgender
Between Sept 5, 2012, and Nov 6, 2019, peripheral blood men from cisgender women by immune phenotype
was collected from 39 individuals in the cisgender post- (figure 1A, B). From this analysis, the predictive
pubertal group (17 [44%] cisgender men, mean age sensitivity was 82·61% and specificity was 82·25% for
18·76 years [SD 2·66]; 22 [56%] cisgender women, mean the BRF model, and the 10-times cross-validation
age 18·59 years [2·81]; late puberty Tanner stage 4–5; classification accuracy held a steady measure of 77·40%.
appendix 1 p 1), 14 children in the pre-pubertal cisgender The top contributing immune-cell features segregating
group (seven [50%] cisgender boys, mean age 8·90 years cisgender men from cisgender women were
[1·66]; seven [50%] cisgender girls, mean age 8·40 years CD4+CD25+CD127– Treg cells and CD4+CD25–CD127+
[1·58]; pre-puberty Tanner stage 1), ten individuals in the Tresp cells (figure 1C). This was confirmed by analysis of
transgender group (five [50%] transgender men, mean the frequency and absolute number of Treg cells—which
age 18·20 years [0·47]; five [50%] transgender women, were significantly increased (frequency p=0·0097;
mean age 18·70 years [0·55]; mid to late puberty Tanner absolute number p=0·041)—and Tresp cells—which
stage 2–5 at the time of puberty arrest), and 35 individuals were significantly reduced (frequency p<0·0001; absolute
in the juvenile-onset SLE group (12 [34%] cisgender men, number p=0·0021)—in cisgender men compared with
mean age 18·58 years [2·35]; 23 [66%] cisgender women, cisgender women (figure 1D–F; appendix 1 p 7). This

A B C D
1·0

CD4+ Tresp*
CD8+ EMRA
CD4+ EMRA
Cisgender women

CD8+ Naive
CD4+ Naive

CD4+ Treg*
0·4 Tresp

CD8+ CM
CD4+ CM

CD8+ EM
CD4+ EM
Cisgender men
Out of bag 0·8 Treg

CD8+
CD4+
Error rate CD4 CM
0·3 Cisgender men=0·2353 Bm1
0·6
Sensitivity

Cisgender women=0·1364 Bm2


Error

Late Bm5

*
*
0·4
0·2 Area under CD19 Unswitched memory
the curve=0·8476 Bm2 Translational
0·2 10-fold cross- CD19 Naive Cisgender men vs Cisgender women
0·1 validation=0·7740 CD14 (T cell subsets)
0
0 2500 5000 7500 10 000 1·0 0·8 0·6 0·4 0·2 0·0 0 0·5 1·0 1·5 2·0 2·5 3·0 3·5 p value
Trees Specificity Mean decrease Gini 0·0 0·05 0·3

E F G H
Cisgender men
g

g
g

g
in

in
in

in
di -

di -
vid

vid

Cisgender women
vid

vid
n

p<0·0001 p=0·0021
No

No
Di

Di

80 0·35 50 p=0·032
0·30 0:1
70
Proportion of Tresp suppressed

Unstim
0·25 40
Proportion of CD4+ T cells

60
0·20 1:1
Cell count (×109)/L

50
Treg:Tresp ratio

p=0·0097 0·15 p=0·041 30


40
1:2 p=0·088
8 20
0·025
6 1:4 p=0·033
0·020
4 0·015 10
2 0·010 0:1
0 0·005 0
Tresp Treg Tresp Treg –103 0 103 104 105 –103 0 103 104 105 1:1 1:2 1:4
Cell trace violet Cell trace violet Treg:Tresp ratio

Figure 1: Comparison of immune-cell subsets in young-post pubertal cisgender men and women
Comparison of 28 immune-cell subsets in 17 young, post-pubertal cisgender men versus 22 young, post-pubertal cisgender women using the balanced random forest model approach. (A) out of bag
error rate of the balanced random forest was 0·1795 (82·05% accuracy). (B) Receiver operator characteristic curve analysis used to validate the model, providing an area under the curve of 0·8476
(84·76% accuracy; 82·61% sensitivity; and 81·25% specificity), with 10-fold cross-validation classification accuracy of 77·40%. (C) The top ten variables contributing to the balanced random forest:
higher mean decrease in Gini score represents a higher importance of the variable. (D) Heatmap of p values comparing T-cell subset immunophenotyping in cisgender men versus cisgender women.
Cumulative cell frequency (E) and absolute counts (F) Tresp-cell (CD4+CD25–CD127+) and Treg-cell (CD4+CD25+CD127–) frequencies; data are mean (SE). (G) Treg-cell mediated suppression of activated
Tresp cells in four cisgender men and four cisgender women detected using cell trace violet and flow cytometry following 72 h activation using soluble anti-CD3 and anti-CD28 in the presence of
monocytes. Treg-cell to Tresp-cell ratios of 1:1 1:2, 1:4, and 0:1 were assessed (appendix 1 p 5); each leftward peak represents a round of proliferation. (H) Suppressive capacity of Treg cells at varying
Treg-cell to Tresp-cell ratios in cisgender men compared with cisgender women, calculated using the fold change of proportion of Tresp-cell proliferation with Treg cells (1:1, 1:2, and 1:4) compared
with Tresp cells without Treg cells (0:1); data are mean (SE). CM=central memory. EM=effector memory. EMRA=effector memory re-expressing CD45RA. Treg=regulatory T. Tresp=responder T.
*Significant p value following 10% false discovery rate adjustment for multiple comparisons.

e714 www.thelancet.com/rheumatology Vol 4 October 2022


Articles

balance was reflected in the Treg-cell to Tresp-cell ratio, compared with Treg cells from young post-pubertal
which was significantly higher in cisgender men cisgender women (figure 1G, H; appendix 1 p 5). Thus,
compared with cisgender women (appendix 1 p 7). Of Treg cells from young cisgender men and women are
note, no significant differences in the frequency of other numerically and functionally distinct.
CD4+ and CD8+ T-cell subsets were seen (figure 1D). To investigate functional differences in Treg cells by
Treg cells from young cisgender men had a significantly sex, RNA sequencing, and DEGs from isolated Treg cells
higher capacity to suppress activated Tresp cells in vitro from young post-pubertal cisgender men and women

A B

LOC101928710
LOC389834

LINC00278
KCNQ1OT1
CCDC144A

MZF1-AS1
PPP1R2P9

SLC25A29

CATSPERG

FAM166A

FAM19A2
C9orf135
CLEC11A
MCOLN2

MAP7D2

SAMD12
FAM43A
ADGRA3

TMSB4Y
NLGN4Y

ZSWIM4
GPANK1
SH2D1B

CARD14
PNLDC1

TXLNGY
BCORP1
CXorf38

ZNF648
TOP1P1

TTTY10

TTTY15
TRIM46
SORCS3

RPS4Y1
CFAP58

KDM5D
KDM6A

SMAD5
HDHD1
ERCC6L

HECW2
EIF1AX

NR4A1
DDX3X

MPZL2
TXLNG

DDX3Y
EIF1AY
TIMD4
EIF2S3

TIAM2

PDGFB

USP9Y
BRCA2
SAR1B

ERAP2
FBLN5

AFAP1
CORIN
ZRSR2

KLRB1

WNK4
TCF19
SYAP1

MC1R
ESAM

DTX1
ASB9
NKRF
PRKX
CCR5

PRKY
GYPE

ESR1
TSIX
XIST

IRS1

UTY
ZFX
10–160

ZFY
JPX
DDX3Y
RPS4Y1 XIST
10–120 KDM5D
Cisgender men

10–80 UTY
TTTY15
TXLNGY
10–40 PRKY USP9Y
Split axis
Log10 p value

EIF1AY
10–30
ZFY
PPP1R2P9
10–20 BCORP1
LINC00278 TSIX
Cisgender women

10–8 NLGN4Y
Split axis
TMSB4Y JPX
10–6 FBLN5
TTTY10 ERCC6L
HECW2
10–4 SMAD5 NKRF
EIF1AX
ESAM CCR5
10–2 TCF19 NR4A1 SLC25A29 p=0·010
10–0
–15 –10 –5 0 5 10 15

Cisgender men Cisgender women 2 1 0 –1


Log2 fold change

C E
NR4A1 ESR1 IRS1 PDGFB
120 p=0·0015 80 p=0·0049 100 p=0·0068 200 p=0·0095
Gene ontology pathway

Histone lysine demethylation


Normalised tcounts

80 150
60
PI3K–AKT signalling pathway 80
60
40 100
Translational initiation 40
40
20 20 50
Renal system process
0 0 0 0
0·0 0·5 1·0 1·5 2·0 2·5 3·0 3·5 4·0 Cisgender Cisgender Cisgender Cisgender Cisgender Cisgender Cisgender Cisgender
men women men women men women men women
–Log10 p value

D F RPS4Y1 EIF2S3 EIF1AX G FOXP3


p<0·0001 p=0·00086 p<0·0001 p=0.33
40 Translation 5000 10000 1000 5000
initiation
Nuclear receptor transcription pathway 4000
8000 800 4000
30 ERa genomic pathway 3000
Enrichment

6000 600 3000


PI3K-Akt
2000
20
Normalised counts

signalling 10
pathway Oestrogen signalling 4000 400 2000
8
pathway
10 6
4 2000 200 1000
Cellular response
2
to hormone stimulus
0 0 0 0 0
25 50 75 100 Cisgender Cisgender Cisgender Cisgender Cisgender Cisgender Cisgender Cisgender
–Log10 p value men women men women men women men women

Figure 2: Comparison of Treg-cell transcriptomic profiles between young post-pubertal cisgender men and women
FACS-sorted Treg cells (CD4+CD25+CD127–) from five young, post-pubertal cisgender men and five young, post-pubertal cisgender women were analysed by RNA-sequencing and whole genome
expression was compared by sex. (A) Volcano plot displaying log2 fold changes and log10 p values of DEGs between cisgender men and women, where coloured points represent statistically significant
DEGs below p value threshold of 0·01). (B) Hierarchical clustering heatmap (Clustvis, Pearson’s) of normalised gene counts of statistically significantly altered DEGs between cisgender men and
women. (C) Pathway analysis plot displaying the –log10 p values of enriched DEG pathway ontology terms of Treg cells between cisgender men and women using the 82 statistically significant DEGs.
(D) Pathway analysis plot displaying the p value and enrichment ratios of enriched DEG pathway ontology terms of Treg cells between cisgender men and women using the extended gene list,
incorporating both original seed genes and the genes from respective protein–protein interaction networks; the size of the points are relative to the number of genes contributing to that pathway. Box
and whisker plots displaying Treg-cell gene expression by normalised counts of genes associated with the PI3K–AKT signalling (E), altered translation initiation pathways (F), and FOXP3 expression (G);
data are mean (SE). DEG=differentially expressed gene. ER=oestrogen receptor alpha. Treg=regulatory T. Tresp=responder T.

www.thelancet.com/rheumatology Vol 4 October 2022 e715


Articles

A B C Cisgender Transgender D Cisgender Transgender


10–9 women men men women
10–7
p=0·033 TLR5
10–8
(proportion of CD4+ T cells)

p=0·017 10–6 MTRNR2L1


8 11 10–7
p=0·032 IRS2
10–5

Log10 p value

Log10 p value
10–6
Treg frequency

CXCR4 SOCS2
7 9 HCP5 IER5 CCR5
10–5 10–4 NR4A2 CCR3
SLC25A29 FOSL2 BEND3P3
7 10–4 ADARB2 CTSL TCF19 10–3 SLC25A29
6 CD161
GTF2H4 DRP2
10–3 TEKT4P2 NR4A1
CFAP58 TSPAN12 10–2
SYT6 NR4A1 p=0·010
5 5 10–2 p=0·010
10–1 10–1

4 3 1 1
sg n –10 –5 0 5 10 –10 –5 0 5 10

sg en

sg n

en
r

Tr womer

wo der
sg ys

rls
de

de

de
Ci me

de
an me
d

m
Log2 fold change Log2 fold change
Ci bo

gi
en

en

en

en

en

en
sg

sg

(vs cisgender women) (vs cisgender men)


Ci

Ci

an

E Tr F G
Cytokine-mediated signalling Transgender men Cisgender women Transgender men Cisgender women
Nitric oxide metabolic process
Gene ontology pathway

OAS2
Cytokine secretion IL17RB
HLA-B
Adaptive immune system LTA
NOD2
Cell recognition NOD2
α-amino acid biosynthetic process CD36
CD36
Response to fibroblast growth factor IRS1
Lymphocyte mediated immunity IL32
TLR5
Extracellular structure organisation SPATA2
HLA-DQA1
Gliogenesis NLRP3
IL17RB
0 1 2 3 4 5
–log10 p value -1 0 1 2

H I J

FOXO signaling pathway Transgender women Cisgender men Cisgender men Transgender women
Corticotropin-releasing hormone response TNFSF14
Gene ontology pathway

BCL6
CDKN1A ZFP36L2
Regulation of cell growth CDKN1B EMILIN1
Regulation of cell activation RGS2 IRS2
H3F3B NR4A3
Positive regulation of catabolic process CXCR4 LGALS9C
SGK1 TNFAIP3
Negative regulation of kinase activity ADNP2 CD83
Cellular senescence PTCH2 BCL6
FBP1 CDKN1A
Interleukin-7 signaling SUPV3L1 DUSP10
CISH ZBTB16
Epithelial cell proliferation FBLN5 SOX13
Regulation of macrophage migration SOCS2 CD9
NEDD4L SIRPB1
0 1 2 3 4 5
-log10 p value

K
TLR5 NLRP3 CD36 NOD2 LTA IL32
p<0·0001 p=0·0085 p=0·00087 p=0·0044
500 40 100 p=0·0013 40 40 000 p=0·0054
250
Normalised counts

400 30 80 200 30 30 000


300 60 150
20 20 20 000
200 40 100
100 10 20 10 10 000
50
0 0 0 0 0 0
Tr wo er

Tr wo er

Tr wo er

Tr wo er
Tr wo er

Tr wo er

sg en

sg en

sg en

sg en
sg en

sg en

r
r

en

en

en
en

en

en
de

de

de

de
de

de

d
d

an m

an m

an m

an m
an m

an m

m
m

m
en

en

en

en
en

en

en

en

en

en
en

en
sg

sg

sg

sg

sg
sg
Ci

Ci

Ci

Ci

Ci

Ci

L CD83 BCL6 NR4A3 CXCR4 ZFP36L2 SOCS2

p=0·0080 p=0·00099 p<0·0001 p<0·0001


800 p=0·0029 400 40 15 000 15 000 300
p=0·0070
Normalised counts

600 300 30
10 000 10 000 200
400 200 20
5000 5000 100
200 100 10

0 0 0 0 0 0
en

en

en

en

en

en
wo der

wo er

wo der

wo er

wo der

wo der
en

en

en

en

en

en
nd

nd
rm

rm

rm

rm

rm
m

m
en

en

en
ge

ge

ge
er
de

de

de

de

de
sg

sg

sg
d

ns

ns

ns
en

en

en

en

en

en
an

an

an
a

a
sg

sg

sg

sg

sg

sg
Tr

Tr

Tr

Tr

Tr

Tr
Ci

Ci

Ci

Ci

Ci

Ci

e716 www.thelancet.com/rheumatology Vol 4 October 2022


Articles

were analysed. 82 genes were differentially regulated in was identified between cisgender men and cisgender
Treg cells between cisgender men and women women, despite its gene location on the X chromosome
(figure 2A, B; appendix 2); there were 50 upregulated (figure 2G). See Online for appendix 2
DEGs in cisgender men and 32 upregulated DEGs in To explore the specific influence of sex hormones on
cisgender women. The most significantly altered genes the sexually dimorphic Treg-cell profiles, we used unique
were located on the X or Y chromosomes: 16 (32%) DEGs cohorts of cisgender, pre-pubertal children and young
in cisgender men and 18 (56%) DEGs in cisgender transgender individuals, who had their physiological
women (appendix 1 p 8). The other DEGs were expressed puberty blocked followed by supplementation of gender-
on other chromosomes typically shared equally by sex affirming sex hormones specific to their target gender
(appendix 1 p 8), suggesting both sex chromosomes and (table; appendix 1 p 11).
hormones could affect Treg-cell transcriptomes. Pre-pubertal cisgender boys had increased Treg-cell
Gene ontology pathway enrichment analysis of the frequency compared with pre-pubertal cisgender girls
original 82 DEGs was done and validated using an (figure 3A; p=0·017). Furthermore, gender-affirming sex
extended gene list based on known protein–protein hormone therapy had an influence on Treg-cell
interactions (figure 2C, D; appendix 1 p 8). This frequencies: Treg cells were significantly increased in
highlighted and validated many significantly enriched both cisgender men and transgender men compared
functional pathways in Treg cells that differ by sex, with cisgender women, with a similar trend in cisgender
including altered PI3K–AKT signalling, nuclear receptor men and transgender men compared with transgender
transcription, translation initiation, oestrogen receptor women (figure 3B), suggesting that dimorphism in
signalling, and cellular response to hormone stimulus. circulating Treg cell numbers might be driven by both
Of note, several key node genes from the protein– sex hormones and underlying sex chromosomes.
protein interaction network (appendix 1 p 8) were In addition, large transcriptomic changes in Treg-cell
exclusive to the PI3K–AKT signalling pathway and were gene expression were observed in both transgender men
increased in cisgender men compared with cisgender (treated with testosterone) compared with cisgender
women, including NR4A1, ESR1, IRS1, and PDGFB women (91 genes; figure 3C) and transgender women
(figure 2E; appendix 1 pp 9–10), supporting increased (treated with oestradiol) compared with cisgender men
Treg-cell functionality in cisgender men. Other node (235 genes; figure 3D; appendix 2), highlighting sex
genes from the extended network were associated with hormone-driven effects over chromosome effects.
altered translation initiation pathways, including Pathway enrichment analysis highlighted key functional
EIF2S3, EIF1AX (X-linked), and RPS4Y1 (Y-linked); pathways in Treg cells that were altered by sex hormones
these were increased in a respective sex chromosome- in transgender individuals (figure 3E–J), including
dependent way (figure 2F; appendix 1 pp 9–10). No upregulated cytokine mediated signalling and cytokine
significant difference in Treg cell FOXP3 expression secretion pathways in transgender men (figure 3E–G)
and downregulated cell growth and activation pathways
in transgender women (figure 3H–J). Many of these
Figure 3: Assessment of Treg-cell functional gene expression and frequency
genes have individually been associated with Treg-cell
in cisgender and transgender individuals function (figure 3K, L; appendix 1 pp 9–10).
Violin plots displaying circulating Treg-cell (CD4+CD25+CD127–) frequencies The influence of sex hormones on Treg-cell gene
between seven pre-pubertal cisgender boys and seven cisgender girls, assessed expression was also compared between cisgender men
by t test (A), and 17 post-pubertal cisgender men, 22 cisgender women,
five transgender men, and five transgender women, assessed by one-way
and women and transgender men and women (figure 4A).
ANOVA (B); measured by flow cytometry; data are mean (SE). FACS-sorted Treg Two genes were altered in all gender-unique comparisons:
cells (CD4+CD25+CD127–) from five young, post-pubertal cisgender men and NR4A1 and SLC25A29 (figure 4B; appendix 1 pp 9–10),
five cisgender women and five transgender men and five transgender women and 24 genes overlapped between at least two gender-
were analysed by RNA-sequencing and whole genome expression was compared
by gender. Volcano plots displaying log2 fold changes and log10 p values of DEGs unique comparisons (figure 4A; appendix 1 p 11). We were
between transgender men and cisgender women (C) and transgender women able to cluster gender-unique groups using hierarchical
and cisgender men (D): the red and blue points represent statistically clustering (appendix 1 p 11) and sparse partial least
significantly DEGs below p value threshold (p<0·01). Cluster significance –log10 squares discriminant analysis (figure 4C). The top ranked
p values of enriched DEG pathway ontology terms of Treg cells between
transgender men and cisgender women (E). Hierarchical clustering heatmap genes driving clustering in component 1 (highest
(Clustvis, Pearson’s) of normalised gene counts of DEGs found in the functional contributing component) were TIAM2 and MZF1-AS1
genetic pathway ontology terms in for transgender men versus cisgender (figure 4D; appendix 1 p 11). Of the 62 DEGs unique to the
women (F, G). Cluster significance –log10 p values of enriched DEG pathway
original cisgender analysis that did not overlap with
ontology terms of Treg cells between transgender women versus cisgender men
(H). Hierarchical clustering heatmap (Clustvis, Pearson’s) of normalised gene transgender comparisons, 32 (52%) were X chromosome
counts of DEGs found in the functional genetic pathway ontology terms in for or Y chromosome linked (figure 4A). Of the remaining
transgender women versus cisgender men (I, J). Box and whisker plots of Treg- DEGs that did overlap with transgender comparisons,
cell gene expression by normalised counts of genes associated with the (K)
five genes (BCORP1, MAP7D2, SLC25A29, NR4A1, and
cytokine signalling, and (L) cell growth and activation pathways; data are mean
(SE). ANOVA=analysis of variance. DEGs=differentially expressed genes. KLRB1) were validated as being significantly altered
FACS=fluorescence-activated cell sorting. Treg=regulatory T. Tresp=responder T. between transgender men and transgender women,

www.thelancet.com/rheumatology Vol 4 October 2022 e717


Articles

A B
MZF1-AS1, CARD14, CCR5 NR4A1 SLC25A29
Cisgender women BCORP1, FAM166A, TIAM2 p=0·0034
vs cisgender men MAP7D2, LOC101928710 p=0·0010
IRS1, CFAP58 FBLN5, HECW2, ESAM p=0·0032
62 120 160 p<0·0001
CD161, DTX1 p=0·0068
TRIM46, TCF19 Transgender p=0·0015
LOC389834 women 120 p<0·0001

Normalised counts
7 11 vs cisgender 80
2 men p=0·0048
Transgender 218 80
4
men 78
40
vs cisgender 40
women

0 0
MTUS1, GNLY
NR4A1,

wo der

wo der
en

sg n

en

sg n

sg n

en
Tr om r

Tr om r
an en

an en
de

de

de

de
SIRPG-AS1, GZMB

w de

w de
an me

an me
m

m
en

en

en

en

en

en
SLC25A29

en

en
sg

sg

sg

sg
sg
Ci

Ci
Ci

Ci
Tr

Tr
C D TIAM2 MZF1-AS1
8
Cisgender women p=0·0024 p=0·0064
6 p=0·0011 p=0·0021
80 200
Component 2 (17·1 %)

4
Normalised counts

60 150
2 Cisgender men
40 100
0
Transgender men
–2 20 50
Transgender women
–4 0 0
–6 –4 –2 0 2 4
r

wo der

wo der
sg n

sg n

en

sg n

sg n

en
Tr om r

Tr om r
an en

an en
de

de

de

de
w de

w de
e

an me

an me
m

m
Component 1 (23·2 %)
en

en

en

en

en

en
en

en
sg

sg

sg

sg
Ci

Ci
Ci

Ci
Tr

Tr
E F
Cisgender man Transgender man Cisgender woman Transgender woman p=0·042

Dividing Non- Dividing Non- Dividing Non- Dividing Non- p=0·0099


dividing dividing dividing dividing Proportion of Tresp suppressed
50 p=0·0074
0:1
Unstim 40
Treg:Tresp ratio

30
1:1
20

0:1 10

0
Cell trace violet Cell trace violet Cell trace violet Cell trace violet
Ci end en

n
de men

en
Tr end me
sg r m

om
sg r wo
er
e

rw
Tr end

e
sg

en
Ci

an

sg
an

Figure 4: Sex and gender specific transcriptomic and functional changes in Treg cells
Cohort details and gender-specific terminology descriptions are reported in appendix 1 (p 1). FACS-sorted Treg cells (CD4+CD25+CD127–) from five young, post pubertal
cisgender men, five cisgender women, five transgender men, and five transgender women were analysed by RNA-sequencing and whole genome expression was
compared by gender. (A) Overlap of DEGs (p<0·01) from Treg cells between different group comparisons: cisgender men versus cisgender women, transgender men
versus cisgender women, and transgender women versus cisgender men. (B) Treg-cell gene expression by normalised counts of genes that overlapped in all gender
comparisons; data were assessed by one-way ANOVA and are mean (SE). (C) Sparse partial least squares discriminant analysis plot clustering each gender group using
normalised gene counts of the overlapping genes (n=24); individual distribution points and confidence ellipses (ovals) are plotted for each gender group. (D) Treg-cell
gene expression by normalised counts of the top ranked loaded genes for component 1 (appendix 1 p 11); data were assessed by one-way ANOVA and are mean (SE).
Treg-cell mediated suppression of activated Tresp cells. Treg cells, Tresp cells, and monocytes were isolated from four young, post-pubertal cisgender men, four
cisgender women, three transgender men, and four transgender women. (E) Proliferation of Tresp cells at varying Treg-cell to Tresp-cell ratios in individuals of different
genders. (F) Suppressive capacity of Treg cells at a Treg-cell to Tresp-cell ratio of 1:1 between different gender groups, calculated using the fold change of the percentage
of Tresp-cell proliferation with Treg cells (1:1) compared with Tresp-cell proliferation without Treg cells (0:1); data were assessed with one-way ANOVA and are mean
(SE; appendix 1 p 5). DEGs=differentially expressed genes. FACS=f luorescence-activated cell sorting. ANOVA=analysis of variance. Treg=regulatory T. Tresp=responder
T. Unstim=Unstimulated.

e718 www.thelancet.com/rheumatology Vol 4 October 2022


Articles

supporting a role of sex hormones in driving their 82 sex specific genes in cisgender individuals who did
expression (appendix 1 p 11; appendix 2). not have juvenile-onset SLE (figure 2A; appendix 2). Of
Analysis of DEG lists using first-order protein–protein these genes, 223 were increased in cisgender women
interaction extended network gene lists from cisgender with juvenile-onset SLE compared with 188 in cisgender
and transgender comparisons revealed significant men with juvenile-onset SLE. Only 27 genes overlapped
overlap of functional pathways associated with cell between the cisgender men and cisgender women in the
signalling by both secondary messengers and juvenile-onset SLE and healthy post-pubertal groups
interleukins (appendix 1 p 12). Despite these functional (appendix 1 p 14); all 27 overlapping genes were X or Y
transcriptomic changes, Treg cells from cisgender men chromosome linked. Gene ontology pathway analysis of
were more suppressive in vitro than were Treg cells from the 384 genes altered by sex that were unique to the
all other individuals (figure 4E, F), suggesting that both juvenile-onset SLE group showed significant enrichment
sex chromosomes and hormones might play a role in the of major histocompatibility complex class II antigen
suppressive functions of Treg cells (appendix 1 p 13). presentation, membrane trafficking, protein-DNA
SLE is an autoimmune disease with a 90% female sex complex assembly, histone 3 lys 27 methylation, and ras
bias that is characterised by defective Treg function.12 Of protein signal transduction pathways (appendix 1 p 14).
note, a significant association was identified by open Furthermore, sex differences in Treg-cell functional
target analysis between the original 82 Treg-cell genes pathways were more pronounced in patients with
differentially expressed between cisgender men and juvenile-onset SLE compared with those who did not
women (figure 2A) and genes previously associated with have the disease (appendix 1 p 14).
SLE in public databases (p=0·020). This association was Of note, sex differences in the expression of CCR5,
driven by ten genes (ESR1, CCR5, TIMD4, DTX1, BRCA2, DTX1, and NR4A1 (genes that were significantly altered
KDM6A, PDGFB, MC1R, NR4A1, and IRS1; between cisgender men and cisgender women who did
appendix 1 pp 9–10), and supports a role for these genes not have juvenile-onset SLE, were affected by gender
in sex specific SLE susceptibility. Of note, KDM6A is affirming treatment hormones in transgender individuals
X chromosome linked, and DTX1, IRS1, CCR5, and and were associated with SLE by open target analysis),
NR4A1 were regulated by testosterone, oestradiol, or both were absent in patients with juvenile-onset SLE (figure 5D).
in transgender individuals on gender-affirming sex By contrast, sex differences in sex-chromosome linked
hormone treatment (figure 4A; appendix 1 p 11). Of the genes (eg, KDM6A), that were associated with SLE by open
gender associated and SLE associated genes, CCR5 and target analysis, were maintained in patients with juvenile-
ESR1 (increased in cisgender women), and PDGFB and onset SLE (figure 5E). In addition, comparing patients
MC1R (increased in cisgender men) are already with juvenile-onset SLE with individuals who did not have
established drug targets for HIV (CCR5); breast cancer, the disease by sex showed very little overlap of DEGs
infertility, and obesity (ESR1); macular degeneration between cisgender men and cisgender women, suggesting
(PDGFB); and erythropoietic protoporphyria and kidney that sex hormones might play a differential role in juvenile-
injury (MC1R; appendix 1 p 11). onset SLE pathogenesis and disease presentation by sex
Sex differences in Treg-cell frequencies were absent in (figure 5F; appendix 1 p 13). This was validated by pathway
juvenile-onset SLE, probably due to the significant increase enrichment analysis, in which genes unique to juvenile-
in Treg-cell frequency in cisgender women with juvenile- onset SLE pathogenesis in cisgender men represented
onset SLE (figure 5A). Of note, there was no significant mRNA splicing and cytokine signalling, whereas those in
difference in clinical measures or treatments between cisgender women represented innate immune responses,
cisgender men or cisgender women with juvenile-onset cellular scenenscence, and toll-like receptor signalling
SLE (table 1), nor was there a significant effect of these (figure 5G). As expected, shared pathways included a
features on Treg-cell frequencies in the combined juvenile- strong upregulation of interferon signalling.
onset SLE cohort (appendix 1 p 13). This was observed
despite multiple patients being treated with glucocorticoids Discussion
(17 [49%] of 35 patients), hydroxychloroquine (31 [89%] Our study identified key sex differences in Treg-cell
patients), or methotrexate (three [9%]; appendix 1 p 13). phenotype and function between healthy individuals,
Additionally, Treg cells from patients with juvenile-onset which might explain differences in autoimmune disease
SLE were more suppressive in cisgender women compared susceptibilities and response to infection. Specifically,
with cisgender men, the opposite of that seen in individuals we identified that the global immune profile was altered
who do not have juvenile-onset SLE, probably due to the by sex, with circulating Treg cells more numerous and
significant loss of Treg-cell suppressive function in suppressive in young post-pubertal cisgender men
cisgender men with juvenile-onset SLE (figure 5B). compared, with cisgender women; Treg cells had an
When Treg-cell gene expression was compared between altered transcriptomic profile between young cisgender
cisgender men with juvenile-onset SLE and cisgender men and cisgender women (beyond sex chromosomes)
women with juvenile-onset SLE, 411 DEGs were associated with increased secondary messenger
identified (figure 5C; appendix 2) compared with signalling in cisgender men; sex hormones altered the

www.thelancet.com/rheumatology Vol 4 October 2022 e719


Articles

A Cisgender men
B Cisgender men with Cisgender women with p=0·0285 p=0·031
Cisgender women juvenile-onset SLE juvenile-onset SLE
p=0·0069 p=0·0242
p=0·0089 Dividing Undivided Dividing Undivided
Figure 5: Sex differences in 0:1 p=0·032 p=0·088
8 p=0·0097 50
(proportion of CD4+ T cells)

Treg cells in patients with Unstim


40

Tresp suppressed
juvenile-onset SLE 1:1

Proportion of
6
Treg frequency

(A) Cumulative cell frequency 30


flow cytometry data of Treg 4 1:2
20
cells (CD4+CD25+CD127–) 15
1:4
comparing 17 young post- 2 10
pubertal healthy cisgender 0:1 5
men, 12 cisgender men with 0 0
Healthy Participants –103 0 103 104 105 –103 0 103 104 105

rti cip y

ju tici cipa y
juvenile-onset SLE, 22 young

se h

se th
ni nt ts

ni nt ts
LE

E
Pa arti alth

h
on it

SL
Pa part ealt
ju cip an

ve pa n
on wi
participants with

tS
Cell trace violet Cell trace violet

le- s w
p He

t
le- s
post-pubertal healthy

H
juvenile-onset

i
ve a
cisgender women, and SLE

r
23 cisgender women with 1:1 1:2
juvenile-onset SLE; data were Treg:Tresp ratio
assessed by t test and are
mean (SE). (B) Treg cell- C D E
mediated suppression of 188 223 CCR5 DTX1 NR4A1 KDM6A
activated Tresp cells in five upregulated upregulated (oestradiol) (testosterone) (oestradiol and (X chromosome)
cisgender men and five 10–160 testosterone) p=0·0022
cisgender women with XIST p=0·0008
juvenile-onset SLE, and DDX3Y 800 200 150 2500 p=0·014
p=0·0019 p=0·0059 p=0·0005
four cisgender men and 10–120
Normalised counts

KDM5D
four cisgender women 600 150 p=0·001 p=0·0015 2000
RPS4Y1 p=0·0008
without juvenile-onset SLE; 100
10 –80
TXLNGY
data were assessed with t test TTTY15 400 100 1500
and are mean (SE). (C) Log2 USP9Y
UTY
fold changes and log10 p values 10–40 Split axis 50
of DEGs between 200 50 500
EIF1AY
five cisgender men and PRKY
five cisgender women with 10–30 0 0 0 0
Log10 p value

juvenile-onset SLE, coloured


Ci r wo n

en er n
r w en
en

Ci r wo en

en er n
r w en
en

Ci r wo n

en er n
r w en
en

Ci r wo n

en er n
r w en
en
e
Ci en me

Ci sgen me

e
Ci sgen me

e
Ci sgen me
de m

de m
om

de m

de m
om

de m

de m
om

de m

de m
om
ZFY
points represent statistically FMN1
en er

en er

en er

en er
10–20
sg d

sg d

sg d

sg d

sg d

sg d

sg d

sg d
Ci sgen

Ci sgen

Ci sgen

Ci sgen
significantly upregulated DEGs BCORP1 PPP1R2P9
s g

NLGN4Y
Ci

Ci

Ci

Ci
below p value threshold LINC00278 TSIX
(p<0·01). Treg-cell gene 10–10 Split axis
expression by normalised GPSM3
PLA2G4C
F G
counts of hormone (gender) 10–8 DDX3X Pathways enriched in cisgender women with juvenile-onset SLE
TMSB4Y
(D) or sex chromosome KDM5C Regulation of innate immune response
10–6 LTA HBB Cellular senescence
specific genes (E) that were RNF5 GTF2H4 Regulation of toll-like receptor signalling pathway
significantly associated with 10 –4
MIR6723 Toll-like receptor cascades
HBA2 Regulation of viral life cycle
SLE by open target analysis 10–2 Regulation of nervous system development
(five per group); data were p=0·010 Regulation of cell adhesion
10
0
15 Deubiquitination
assessed with one-way ANOVA Regulation of activin receptor signalling pathway
–15 –10 –5 0 5 10
and are mean (SE); open bars 192
Negative regulation of cell population proliferation
Log2 fold change
are participants without 0 1 2 3 4 5 6 7
juvenile-onset SLE and closed Cisgender women Pathways enriched in cisgender men and cisgender women with juvenile-onset SLE
bars are participants with Cisgender Cisgender Interferon α and β signalling
men women Neutrophil extracellular trap formation
juvenile-onset SLE. (F) Overlap Type II interferon signalling
of DEGs (p<0·01) from Treg Response to interferon-α
Network map of SARS-CoV-2 signalling pathway
cells comparing patients with 44 Extrafollicular B-cell activation
juvenile-onset SLE with Response to interferon β
Regulation of pattern recognition receptor signalling
healthy participants, stratified Non-genomic actions of 1,25 dihydroxyvitamin D3
by sex (cisgender men and Response to bacterium
Cisgender men
cisgender women). (G) 0 5 10 15 20 25
Pathway analysis bar charts of 214
Pathways enriched in cisgender men with juvenile-onset SLE
enriched pathway ontology mRNA splicing, via spliceosome
terms from sex-unique or Cytokine signalling in immune system
Epstein-Barr virus infection
overlapping DEGs. Systemic lupus erythematosus
ANOVA=analysis of variance. Extrafollicular B cell activation
Glial cell activation
SLE=systemic lupus Cognition
erythematosus. Cell activation
Embryonic cranial skeleton morphogenesis
DEGs=differentially expressed Positive regulation of interferon-α production
genes. Treg=regulatory T. 0 2 4 6 8 10 12
Tresp=responder T. –Log10 p value
Unstim=unstimulated.

e720 www.thelancet.com/rheumatology Vol 4 October 2022


Articles

Treg-cell frequency and transcriptomic functional profile expansion15,16 and suppressive capabilities17,18 of Treg cells.
between cisgender men and women, validated using Treg-cell numbers have been shown to increase during
transgender individuals undergoing gender-affirming the menstrual cycle when oestradiol concentrations are
sex hormone therapy; and differences in Treg frequency highest before ovulation,19 suggesting a Treg-cell response
and function were absent or altered in an age-matched to sex hormones. By contrast, men who received
post-pubertal cisgender patients with juvenile-onset gonadotropin-releasing hormone agonists, a puberty
SLE. The information reported here might help us to blocker which significantly reduces testosterone
understand immunopathological mechanisms of concentrations, have lower circulating Treg cells than do
sexually dimorphic autoimmune disease development men who received placebo.20 As observed in cisgender
and contribute to basic understanding of immunology men, we found that transgender men had increased
by sex and gender. Treg-cell frequencies following puberty blocking and
We showed that use of broad-spectrum immune early-stage gender-affirming testosterone administration
phenotyping with machine learning can be used to (mean 12 months). Observed sex differences in Treg-cell
classify sex differences with confidence. To our frequencies in pre-pubertal children matched those seen
knowledge, no other studies have used machine learning in older participants post-puberty suggesting that sex
to address innate and adaptive immunological differences hormones and chromosomes both play dominant roles
between men and women, particularly surrounding in driving changes in Treg-cell frequency by sex. Women
prediction accuracy as a method to describe how different with Turner syndrome, characterised by the presence of
these sexual dimorphisms are. However, other studies, one normal X chromosome and a missing or structurally
including our own, have addressed this from an abnormal second one, have an increased frequency of
autoimmunity standpoint, comparing individuals with Treg cells compared with women with two intact X
and without specific diseases (eg, juvenile-onset SLE chromosomes, suggesting a more suppressive role of the
and juvenile idiopathic arthritis). These studies of similar X chromosome regarding Treg-cell proliferation.21
immune landscapes found that a balanced random Despite these observations, no differences in Treg-cell
forest model had a strong classification accuracy: frequencies have been reported in infants (who have sex
90·9% (4·8% higher than this study) for classifying chromosomes but very low concentrations of sex
patients with juvenile-onset SLE,11 and 89·6% (6·1% higher hormones) from birth to 1 year of age;22 therefore, the
than our study) for classifying patients with juvenile influence of the X chromosome on the number of
idiopathic arthritis13 from healthy controls. For identifying circulating Treg cells might occur after the initial years of
patients with juvenile-onset SLE, there was a diagnostic development in children.
sensitivity of 89·6% (7·0% higher than this study) and Treg-cell FOXP3 expression was not altered by sex in
specificity of 82·1% (0·2% lower than this study). This this study, despite its location on the X chromosome and
highlights that the extent of immune dimorphisms previous reports of increased mRNA expression in
observed between healthy cisgender men and women cisgender men compared with women in an older cohort
are not too dissimilar to those observed between people with a wider age range.23 Our data suggests that differences
who have and those who do not have juvenile-onset SLE, in Treg-cell function by sex might be maintained via
supporting our hypothesis of altered autoimmune risk different mechanisms in younger individuals. Transcrip­
by sex. Treg cells were one of the top contributing tomic analysis identified that the PI3K signalling pathway
immune-cell features responsible for segregating was increased in young cisgender men compared with
cisgender men from cisgender women using machine young cisgender women in our study. PI3K signalling can
learning. This supports previous observations of human support Treg-cell metabolism and function; however,
sexual dimorphism in Treg-cell frequency across overstimulation can reverse this in vitro.24 Inhibiting
different post-pubertal age groups;1,14 however, mouse PI3K signalling in vivo results in reduced Treg-cell
studies of Treg cells by sex are contradictory, probably frequency and suppressive capacity,25 supporting a role for
due use of disease models and examining organ-specific PI3K signalling in the increased suppressive capacity of
rather than peripheral blood Treg cells.14 We found that Treg cells in young men compared with young women.
young post-pubertal cisgender men had increased Treg- Of note, secondary messenger signalling pathways were
cell frequency and suppressive function compared with associated with hormone changes in transgender
cisgender women, suggesting a more anti-inflammatory individuals by extended pathway analysis. A gene of
circulating immune profile. These characteristics could interest from these pathways with increased expression in
contribute to the better response of women to vaccination cisgender men and transgender men was NR4A1 (and
and infection that has been observed (highlighted in the NR4A3 in transgender men); these receptors are crucial
COVID-19 pandemic, in which men had an increased for Treg-cell function,26 highlighting a therapeutic target to
risk of mortality)1,2 and the increased susceptibility of control autoimmune pathogenesis in women. Despite
women to autoimmune disease compared with men.3 this, Treg cells in young cisgender men were more
Despite these observations, multiple studies have suppressive than those in transgender men and cisgender
reported a key role of oestradiol in supporting the and in transgender women, suggesting that both sex

www.thelancet.com/rheumatology Vol 4 October 2022 e721


Articles

hormones and sex chromosomes might play a role in onset SLE, despite the increased number of cells. This
driving increased suppresive function of Treg cells in could explain why anti-inflammatory feedback mech­
cisgender men. Treg cells from women with Turner anisms are often not substantial enough to control
syndrome have impaired suppressive functions compared disease; thus, therapies that promote Treg-cell function
with women with two intact X chromosomes.21 A previous and number could still hold promise in juvenile-onset
Treg-cell transcript­omics study combined a list of genes SLE for both sexes. We also identified strong sex
into a specific human Treg-cell signature, using previously differences in Treg-cell transcriptomic profiles in juvenile-
published datasets, to produce a subsequent microarray onset SLE and highlighted both sex specific (mRNA
test of 62 genes.27 Of these genes, we only identified splicing in cisgender men and immune regulation in
a single gene, CCR5, that was significantly altered between cisgender women) and common (interferon signalling)
cisgender men and cisgender women. PECAM1 was the pathogenic processes, which together might relate to sex
only altered gene in transgender men compared with differences in clinical outcomes for patients. Of note, we
cisgender women in our study. Our findings suggest that did not find an association between oestrogen-enriched
we might have identified multiple novel genes associated and interferon-enriched pathways in the cohort of patients
with Treg-cell function by sex. Pfoertner and colleagues27 with juvenile-onset SLE in our study, which has been
analysed the combined data from six women and five men described in older patients (mean age 38·5 [SD 15·3]) with
(aged 26–58 years) which might have substantially affected SLE.33 Finally, histone lysine demethylation was a key
the results, as highlighted by our observations of sexual pathway altered between young cisgender men and
dimorphisms in Treg cells. cisgender women, supporting previous evidence
Although SLE is more common in females, sexual surrounding the role of female-biased epigenetic alteration
dimorphisms in SLE are important to consider because of T-cell function associated with SLE.31 Together, this
sex-specific clinical features have been observed in adults supports a key role for Treg-cell targeted therapies in SLE
with SLE. Men are affected by more severe renal and juvenile-onset SLE.12
manifestations and higher end-stage renal disease risk, Our study has several limitations. Difficulty in recruiting
requiring increased monitoring in clinical practice.28 Of and taking larger volumes of blood from rare cohorts of
note, DEGs from Treg cells between young cisgender young individuals, especially transgender individuals and
men and cisgender women were significantly associated children, limited our ability to assess the phenotype and
with SLE, some of which were altered in transgender function of Treg cells. Validation in larger cohorts of
individuals. Treg cells have been implicated in SLE young individuals would be beneficial, but this is beyond
pathogenesis and disproportionate T helper 17-cell to Treg- the scope of this study. As highlighted by our study, it is
cell ratios, resulting in a proinflammatory phenotype.29 also possible that Tresp cells could be important for the
However, these studies did not address sex differences inflammatory balance between cisgender men and
and the populations included less than 7% men. It is also women, which would require additional phenotype and
reported that women with SLE have increased plasma functional analyses. The increased suppressive capacity of
oestradiol and decreased testosterone concentration Treg cells in cisgender men could also be due to increased
compared with women without SLE, which could account proliferation of Tresp cells in cisgender women, or these
for differences in Treg-cell function; however, this study cells not being as amenable to suppression compared with
did not take into consideration the individual’s stage of Tresp cells from cisgender men. We attempted to account
menstrual cycle.23 We showed that Treg cells were more for this by plotting the suppressive capacity of Treg cells as
numerous in patients with juvenile-onset SLE compared the fold change between Tresp-cell proliferation in the
with individuals without juvenile-onset SLE in cisgender presence and absence of Treg cells, thus normalising the
women, but not in men; therefore, Treg-cell frequency data for basal Tresp-cell proliferative capacity upon
was not significantly different by sex in patients with stimulation; however, this remains a study limitation. It
juvenile-onset SLE. This increase in Treg-cell frequency in was beyond the scope of the study to measure sex
women with juvenile-onset SLE could represent an anti- hormone concentrations in the serum of study
inflammatory pathway aimed to counter the over-riding participants; however, clinical monitoring aims to keep
inflammation. We also showed that Treg cells from young concentrations within physiological ranges. There was no
cisgender women with juvenile-onset SLE had a signif­ follow-up of transgender individuals, which prevented us
icantly higher suppressive capacity compared with those from observing the long-term effects of hormone therapy
from cisgender men with juvenile-onset SLE, the opposite on Treg cells. Despite using a diverse healthy cohort for
of that seen in healthy individuals. However, this the immunophenotyping and accounting for ethnicity in
suppressive capacity of Treg cells from cisgender women analysis, the RNA-sequencing was done in White
with juvenile-onset SLE was still much lower than that transgender individuals; thus, our findings will require
observed in cisgender men and similar to that observed in validation in larger, more diverse cohorts. Although we
cisgender women who did not have the disease. These have speculated on the functional implications on Treg
findings suggest that the underlying function of Treg cells cells of specific DEGs and pathways between sexes, the
was not improved in cisgender women with juvenile- physiological relevance of these differences would need to

e722 www.thelancet.com/rheumatology Vol 4 October 2022


Articles

be confirmed by mechanistic studies and analysis of Acknowledgments


proteins. The use of conventional flow cytometry limited This work was supported by a Versus Arthritis Career Development
Fellowship (22856), grants from the National Institute for Health and
the number of markers that we could measure for
Care Research University College London Hospital Biomedical Research
phenotyping. In future studies, it would be useful to Centre (grant numbers BRC772/III/EJ/101350 and BRC773/III/
investigate additional surface markers for Treg cells and CC/101350), and Lupus UK and The Rosetrees Trust (M409). The study
Tresp cells to assess function and partition T cells into was done at the Centre for Adolescent Rheumatology Versus Arthritis at
University College London. University College London Hospital and
sublineages. A wider array of markers were used to this Great Ormand Street Hospital supported by grants from Versus
effect by Lambert and colleagues,32 who revealed important Arthritis (21593 and 20164), Great Ormond Street Hospital Charity, and
features that vary with autoimmune disease states and by the National Institute for Health and Care Research-Biomedical
age. Finally, the patients with juvenile-onset SLE were Research Centres at both Great Ormand Street Hospital and University
College London Hospital. The views expressed are those of the authors
clinically heterogeneous and were on different treatments, and not necessarily those of the UK NHS, the National Institute for
presenting possible confounding effects on the immune Health and Care Research or the UK Department of Health.
system, especially considering that dsDNA has previously References
been shown to correlate negatively with Treg cells in 1 Klein SL, Flanagan KL. Sex differences in immune responses.
adults with SLE33 and that immuno­ suppressive drugs, Nat Rev Immunol 2016; 16: 626–38.
2 Peckham H, de Gruijter NM, Raine C, et al. Male sex identified by
such as glucocorticoids, hydroxy­ chloroquine, and global COVID-19 meta-analysis as a risk factor for death and ITU
methotrexate, could influence Treg-cell frequency and admission. Nat Commun 2020; 11: 6317.
function.34,35 Despite finding no difference in clinical 3 Billi AC, Kahlenberg JM, Gudjonsson JE. Sex bias in autoimmunity.
measures or treatments between cisgender men and Curr Opin Rheumatol 2019; 31: 53–61.
4 Uppal SS, Verma S, Dhot PS. Normal values of CD4 and CD8
cisgender women with juvenile-onset SLE, or any lymphocyte subsets in healthy indian adults and the effects of sex,
association of these juvenile-onset SLE features with Treg age, ethnicity, and smoking. Cytometry B Clin Cytom 2003; 52: 32–36.
frequency in our study, the potentially different treatment 5 Abdullah M, Chai PS, Chong MY, et al. Gender effect on in vitro
lymphocyte subset levels of healthy individuals. Cell Immunol 2012;
strategies are important to consider because treatments 272: 214–19.
for the disease could explain the absence of differences in 6 Hewagama A, Patel D, Yarlagadda S, Strickland FM,
Treg-cell frequency in patients with juvenile-onset SLE. It Richardson BC. Stronger inflammatory/cytotoxic T-cell response in
women identified by microarray analysis. Genes Immun 2009;
is possible that this cohort—which includes patients 10: 509–16.
with predominantly well controlled and inactive juvenile- 7 Hughes GC, Choubey D. Modulation of autoimmune rheumatic
onset SLE, with a high proportion of patients on hydroxy­ diseases by oestrogen and progesterone. Nat Rev Rheumatol 2014;
10: 740–51.
chloroquine and several other treatments—might mask
8 Ambrose N, Morgan TA, Galloway J, Ionnoau Y, Beresford MW,
the potential effect of treatment and disease features on Isenberg DA. Differences in disease phenotype and severity in SLE
Treg-cell frequency and function by sex. Because juvenile- across age groups. Lupus 2016; 25: 1542–50.
onset SLE is a severe disease frequently requiring timely 9 Holdcroft A. Gender bias in research: how does it affect evidence
based medicine? J R Soc Med 2007; 100: 2–3.
commencement of therapy, recruitment of treatment-
10 Beery AK, Zucker I. Sex bias in neuroscience and biomedical
naive patients is not usually feasible. research. Neurosci Biobehav Rev 2011; 35: 565–72.
To conclude, we have highlighted sexual dimorphisms 11 Robinson GA, Peng J, Dönnes P, et al. Disease-associated and
in Treg-cell profiles and function by sex in young healthy patient-specific immune cell signatures in juvenile-onset systemic
lupus erythematosus: patient stratification using a machine-
individuals and patients with autoimmunity, showing learning approach. Lancet Rheumatol 2020; 2: e485–96.
differential influences of sex chromosomes and 12 Mizui M, Tsokos GC. Targeting regulatory T cells to treat patients
hormones using unique transgender cohorts. This with systemic lupus erythematosus. Front Immunol 2018; 9: 786.
13 Van Nieuwenhove E, Lagou V, Van Eyck L, et al. Machine learning
information helps us to understand the mechanistic identifies an immunological pattern associated with multiple
pathogenesis of autoimmune disease and the bias juvenile idiopathic arthritis subtypes. Ann Rheum Dis 2019;
towards cisgender women. Our study will help inform 78: 617–28.
14 Afshan G, Afzal N, Qureshi S. CD4+CD25(hi) regulatory T cells in
the future consideration of sex as a biological variable in healthy males and females mediate gender difference in the
inflammatory research and clinical trials. prevalence of autoimmune diseases. Clin Lab 2012; 58: 567–71.
Contributors 15 Tai P, Wang J, Jin H, et al. Induction of regulatory T cells by
ECJ, CC, IP-T, and GAR designed the study. GAR, HP, and GB acquired physiological level estrogen. J Cell Physiol 2008; 214: 456–64.
the data. CC, HP, and GB recruited the patients. All authors had access 16 Polanczyk MJ, Carson BD, Subramanian S, et al. Cutting edge:
to the data. GAR, ECJ, and JP accessed and verified the data. GAR and estrogen drives expansion of the CD4+CD25+ regulatory T cell
compartment. J Immunol 2004; 173: 2227–30.
JP analysied the data. GAR and ECJ wrote the manuscript. CC, IP-T,
and GB reviewed the manuscript. All authors approved the final 17 Goodman WA, Bedoyan SM, Havran HL, Richardson B,
Cameron MJ, Pizarro TT. Impaired estrogen signaling underlies
version.
regulatory T cell loss-of-function in the chronically inflamed
Declaration of interests intestine. Proc Natl Acad Sci USA 2020; 117: 17166–76.
We declare no competing interests. 18 Polanczyk MJ, Hopke C, Huan J, Vandenbark AA, Offner H.
Enhanced FoxP3 expression and Treg cell function in pregnant and For more on the immune
Data sharing estrogen-treated mice. J Neuroimmunol 2005; 170: 85–92. phenotype see http://dx.doi.
Immune phenotype data can be found at Mendeley Data. RNA 19 Arruvito L, Sanz M, Banham AH, Fainboim L. Expansion of org/10.17632/mcsypbk6nz.2
sequencing data can be found at ArrayExpress (Accession number: CD4+CD25+and FOXP3+ regulatory T cells during the follicular
E-MTAB-11919) repositories. This data will be available from manuscript phase of the menstrual cycle: implications for human reproduction. For more on the RNA
publication date. J Immunol 2007; 178: 2572–78. sequencing data see www.ebi.
ac.uk/arrayexpress

www.thelancet.com/rheumatology Vol 4 October 2022 e723


Articles

20 Page ST, Plymate SR, Bremner WJ, et al. Effect of medical 28 Ramírez Sepúlveda JI, Bolin K, Mofors J, et al. Sex differences in
castration on CD4+ CD25+ T cells, CD8+ T cell IFN-gamma clinical presentation of systemic lupus erythematosus.
expression, and NK cells: a physiological role for testosterone and/ Biol Sex Differ 2019; 10: 60.
or its metabolites. Am J Physiol Endocrinol Metab 2006; 29 Yang J, Chu Y, Yang X, et al. Th17 and natural Treg cell population
290: E856–63. dynamics in systemic lupus erythematosus. Arthritis Rheum 2009;
21 Lee YA, Kim HR, Lee JS, et al. CD4+ FOXP3+ regulatory T cells 60: 1472–83.
exhibit impaired ability to suppress effector T cell proliferation in 30 Singh RP, Hahn BH, Bischoff DS. Interferon Genes Are Influenced
patients with turner syndrome. PLoS One 2015; 10: e0144549. by 17β-Estradiol in SLE. Front Immunol 2021; 12: 725325.
22 Collier FM, Tang ML, Martino D, et al. The ontogeny of naïve and 31 Christou EAA, Banos A, Kosmara D, Bertsias GK, Boumpas DT.
regulatory CD4(+) T-cell subsets during the first postnatal year: Sexual dimorphism in SLE: above and beyond sex hormones. Lupus
a cohort study. Clin Transl Immunology 2015; 4: e34. 2019; 28: 3–10.
23 Singh RP, Bischoff DS. Sex hormones and gender influence the 32 Lambert K, Moo KG, Arnett A, et al. Deep immune phenotyping
expression of markers of regulatory T cells in SLE patients. reveals similarities between aging, Down syndrome, and
Front Immunol 2021; 12: 619268. autoimmunity. Sci Transl Med 2022; 14: eabi4888.
24 Pompura SL, Dominguez-Villar M. The PI3K/AKT signaling 33 Lee HY, Hong YK, Yun HJ, Kim YM, Kim JR, Yoo WH. Altered
pathway in regulatory T-cell development, stability, and function. frequency and migration capacity of CD4+CD25+ regulatory T cells
J Leukoc Biol 2018; 103: 1065–76. in systemic lupus erythematosus. Rheumatology (Oxford) 2008;
25 Soond DR, Slack ECM, Garden OA, Patton DT, Okkenhaug K. 47: 789–94.
Does the PI3K pathway promote or antagonize regulatory T cell 34 Mathian A, Jouenne R, Chader D, et al. Regulatory T cell responses
development and function? Front Immunol 2012; 3: 244. to high-dose methylprednisolone in active systemic lupus
26 Hiwa R, Nielsen HV, Mueller JL, Mandla R, Zikherman J. NR4A erythematosus. PLoS One 2015; 10: e0143689.
family members regulate T cell tolerance to preserve immune 35 Scottà C, Fanelli G, Hoong SJ, et al. Impact of immunosuppressive
homeostasis and suppress autoimmunity. JCI Insight 2021; drugs on the therapeutic efficacy of ex vivo expanded human
6: 151005. regulatory T cells. Haematologica 2016; 101: 91–100.
27 Pfoertner S, Jeron A, Probst-Kepper M, et al. Signatures of human
regulatory T cells: an encounter with old friends and new players.
Genome Biol 2006; 7: R54.

e724 www.thelancet.com/rheumatology Vol 4 October 2022

You might also like