Download as pdf or txt
Download as pdf or txt
You are on page 1of 36

Article

Anatomically distinct fibroblast subsets


determine skin autoimmune patterns

https://doi.org/10.1038/s41586-021-04221-8 Zijian Xu1,9, Daoming Chen1,2,9, Yucheng Hu3, Kaiju Jiang1, Huanwei Huang1, Yingxue Du1,


Wenbo Wu1, Jiawen Wang1, Jianhua Sui1, Wenhui Wang4, Long Zhang4, Shuli Li5, Chunying Li5,
Received: 2 March 2020
Yong Yang6, Jianmin Chang7 ✉ & Ting Chen1,8 ✉
Accepted: 5 November 2021

Published online: 15 December 2021


The skin serves as a physical barrier and an immunological interface that protects the
Check for updates body from the external environment1–3. Aberrant activation of immune cells can
induce common skin autoimmune diseases such as vitiligo, which are often
characterized by bilateral symmetric lesions in certain anatomic regions of the
body4–6. Understanding what orchestrates the activities of cutaneous immune cells at
an organ level is necessary for the treatment of autoimmune diseases. Here we
identify subsets of dermal fibroblasts that are responsible for driving patterned
autoimmune activity, by using a robust mouse model of vitiligo that is based on the
activation of endogenous auto-reactive CD8+ T cells that target epidermal
melanocytes. Using a combination of single-cell analysis of skin samples from
patients with vitiligo, cell-type-specific genetic knockouts and engraftment
experiments, we find that among multiple interferon-γ (IFNγ)-responsive cell types in
vitiligo-affected skin, dermal fibroblasts are uniquely required to recruit and activate
CD8+ cytotoxic T cells through secreted chemokines. Anatomically distinct human
dermal fibroblasts exhibit intrinsic differences in the expression of chemokines in
response to IFNγ. In mouse models of vitiligo, regional IFNγ-resistant fibroblasts
determine the autoimmune pattern of depigmentation in the skin. Our study
identifies anatomically distinct fibroblasts with permissive or repressive IFNγ
responses as the key determinant of body-level patterns of lesions in vitiligo, and
highlights mesenchymal subpopulations as therapeutic targets for treating
autoimmune diseases.

Vitiligo is an acquired polygenetic autoimmune disease9. Its defining fea-


ture is skin depigmentation, in which affected skin is unable to spontane- Cellular IFNγ responses in vitiligo-affected skin
ously recover owing to the activity of auto-reactive CD8+ T cells, which To understand the cellular and molecular mechanisms that lead to pat-
causes a loss of epidermal melanocytes7,8,10–15. Vitiligo affects 0.5% to 2% terned autoimmune activity in vitiligo-affected skin (Fig. 1a, Extended
of the population worldwide16–18. More than 80% of patients with vitiligo Data Fig. 1a), we first analysed skin biopsies from patients with vitiligo.
have bilateral symmetric patterns of depigmentation across the central Immunofluorescent staining showed that melanocytes were absent
body axis, which is classified as non-segmental vitiligo4–6. Previous stud- within the lesion but evenly distributed in the basal epidermis of the
ies using a mouse model of vitiligo have shown that IFNγ is important for perilesion region (Fig. 1b). Notably, the majority of the infiltrated CD8+
T cell-induced depigmentation in skin19–21. Although various studies have T cells were concentrated at the junction area between the lesion and
proposed underlying mechanisms22–27, the identity of IFNγ-responsive the perilesion skin, which implied a local recruitment mechanism for
cells that mediate this function is largely unknown. Hypotheses about the CD8+ T cells.
reason behind the bilateral symmetric patterns that are observed in viti- Next we used single-cell RNA sequencing (scRNA-seq) to analyse
ligo pathology include regional variations in microbiota distribution; the all cell types present in patient skin (Extended Data Fig. 1b, c). Unsu-
pattern of antigen expression in melanocytes; and different distributions pervised clustering of more than 50,000 cells from 10 patients with
of neuropeptides released by nerve endings28–32. Despite these studies, vitiligo and 5 healthy donors yielded 8 main cell types defined by
the cellular and molecular mechanisms that orchestrate the patterned signature genes (Extended Data Fig. 1d–h, Supplementary Table 1).
activities of immune cells in the skin of patients with vitiligo remain elusive. Further analysis of melanocytes revealed two distinct sub-clusters

National Institute of Biological Sciences, Beijing, China. 2Peking University–Tsinghua University–National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences,
1

Peking University, Beijing, China. 3Academy for Multidisciplinary Studies, Beijing National Center for Applied Mathematics, Beijing Advanced Innovation Center for Imaging Theory and
Technology, Capital Normal University, Beijing, China. 4Peking University Third Hospital, Beijing, China. 5Department of Dermatology, Xijing Hospital, Xi’an, China. 6Institute of Dermatology,
Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China. 7Department of Dermatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric
Medicine, Chinese Academy of Medical Sciences, Beijing, China. 8Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China. 9These authors contributed
equally: Zijian Xu, Daoming Chen. ✉e-mail: changjm0417@126.com; chenting@nibs.ac.cn

118 | Nature | Vol 601 | 6 January 2022


a b Bright field DAPI CD8 TYR K14 c d
40 100

80

Melanocytes (%)
20
60

t-SNE 2
0
40

−20 20
g M1
M2 0
−40 1 2 3 4 5 6 7 8 9101 2 3 4 5
Perilesion T cell infiltration region Lesion −50 −25 0 25
t-SNE 1 Patient HD

ll
l
e f g h

el

ce
Tcyt in immune cells (%)

ce a te
K14 PDGFRA

Ke cl e i al c

ng go e
T erh cy
20

En obl yte
40

ll ns
La pha cyt
**

M oth t
d as
us el
br c

. o
Fi ano

M tin
15

DAPI pSTAT1
ra
el
NS

M
10
30
in immune cells (%)

Antigen presentation
5
T cell subtypes

IFNγ signalling
0
20 T cell migration
Per cent of pSTAT1+ cells
Vasculature development 0 20 40 60 80 100
3 **
IFNG normalized

10 mRNA catabolic Melanocyte


NS
expression

2 Oxidative stress Fibroblast


Endothelial cell
1 Lymphocyte immunity Muscle
0
Cell killing Keratinocyte
0
M. phagocyte
t

g
ef
cy

re
re
T

T
T

Langerhans cell
4 8 12 >14 1 10 20
Progressive Quiescent Healthy Enrichment −log10(P value) T cell

Fig. 1 | scRNA-seq analysis reveals distinct IFNγ-responsive cell types in the T cells. f, Percentage of CD8+ cytotoxic T cells and expression of IFNG in CD8+
skin of patients with vitiligo. a, Schematic illustration of bilateral symmetric cytotoxic T cells from the skin of patients with vitiligo and healthy donors.
lesions in non-segmental vitiligo. b, Representative bright-field and g, GO categories of genes enriched in each cell type from the skin of patients
immunofluorescent images of vitiligo lesion skin. c, t-distributed stochastic in the progressive state. M. phagocyte; mononuclear phagocyte.
neighbour embedding (t-SNE) plot of melanocytes from the skin of patients h, Representative immunofluorescent images and quantification of pSTAT1
with vitiligo and healthy donor skin. d, Melanocyte composition in each patient signal in different cell types in the skin of patients with vitiligo. Scale bars,
and healthy donor (HD). e, Analysis of T cell subtype composition. Tcyt, 50 μm (b, h). For exact P values, see Source Data. For statistics, P summary and
cytotoxic T cells; Tres, resident T cells; Teff, effector T cells; Treg, regulatory sample sizes, see Methods.

(termed M1 and M2) (Fig. 1c). Upregulated genes (more than twofold; T cells (Extended Data Fig. 1p), T cells in the skin of patients in the
P < 0.01) in melanocytes of the M1 cluster compared to the M2 cluster progressive state did not show an increased IFNγ response compared
showed strong immune responses, especially in terms of the response to healthy skin. This could be related to the low expression level of
to IFNγ (Extended Data Fig. 1i, Supplementary Table 2). Melanocytes IFNGR1 and IFNGR2 in T cells compared to other cell types (Extended
from patients 1–5 were predominantly composed of M1 cluster cells, Data Fig. 1q).
whereas patients 6–10 had a similar composition of melanocytes to that To further validate the IFNγ-responsive cell types, we analysed phos-
of healthy donors (Fig. 1d). On the basis of the expression profile and phorylated (p)STAT1 in different cell types in skin with vitiligo lesions.
composition of melanocytes, we classified patients 1–5 as in a progres- Spatially, the density of CD8+ T cells positively correlated with the den-
sive state, and patients 6–10 as in a quiescent state. sity of pSTAT1+ cells (Extended Data Fig. 1r). Quantification showed
To validate the disease state classifications, we next analysed T cells. that among eight different cell types examined, dermal fibroblasts
Four major sub-clusters of T cells were identified by their expression accounted for most (around 80%) of the pSTAT1+ cells in patient skin
pattern: CD8+ cytotoxic; CD8+ resident; CD4+ effector; and CD4+ reg- (Fig. 1h, Extended Data Fig. 1s).
ulatory T cells (Extended Data Fig. 1j, k, Supplementary Table 3), all
of which were enriched in skin from patients compared to skin from
healthy donors (Fig. 1e, Extended Data Fig. 1l–m). Skin depigmenta- Skin stromal IFNγ response drives vitiligo
tion in vitiligo is mediated by antigen-specific CD8+ cytotoxic T cells To study the functional relevance of our analysis of patient samples, we
targeting melanocytes13,33,34. In patients in the progressive state, there developed a mouse model in which vitiligo was induced by inoculation
were significantly more CD8+ cytotoxic T cells, which also expressed with B16F10 melanoma coupled with depletion of regulatory T (Treg)
significantly higher levels of IFNG (Fig. 1f). cells35, which recapitulated the pathologies of patients with vitiligo
Previous studies showed that IFNγ signalling is important for the pro- (Extended Data Figs. 2, 3). After the same vitiligo induction procedure,
gression of vitiligo in mouse models19–21. Gene ontology (GO) analysis wild-type mice developed vitiligo—as shown by tail skin depigmenta-
of enriched genes (more than 1.5-fold; P < 0.01) in the skin of patients tion, significant loss of melanocytes and abundant infiltration and
in the progressive state showed that melanocytes and fibroblasts were aggregation of CD8+ T cells—but Ifngr1 knockout (KO) mice did not
overrepresented in categories related to the IFNγ signalling pathway develop vitiligo (Fig. 2a, b, Extended Data Fig. 4a–h). This phenomenon
(Fig. 1g, Extended Data Fig. 1n), which was further confirmed by analy- was not due to delayed cytotoxic activity, because even at four months
sis of signature IFNγ response genes in all cell types (Extended Data after vitiligo induction, melanocytes in Ifngr1 KO tail skin were still
Fig. 1o). Although IFNG was predominantly expressed by cytotoxic intact (Extended Data Fig. 4i). RNA-seq and quantitative PCR (qPCR)

Nature | Vol 601 | 6 January 2022 | 119


Article
a WT Ifngr1 KO
b c 104 d WT Pmel
****

WT CD8+ T cell FPKM


100 Ccl5 50 ***
T cells

per 102 CD45+ cells


No. of CD8+ T cells
per 102 CD45+ cells
Gzmb

No. of CD8+ T cells


80 103 40
Gzma hPMEL-AAV
60 30
102 Prf1
40 20
20 101 10
0 0
100
CD8+ T cell Melanocyte 100 101 102 103 104
5 Gy γ Recipient

T
KO

KO
KO CD8+ T cell FPKM

KO

KO
T
W

T
irradiated

W
Vit. induc.

W
Vit. induc. – – + + – – + +

NS f KO KO KO WT
e 2.4 ***

(×104 per cm2)


Skin graft

Melanocytes
KO skin WT skin PDGFRA CD31
1.8
1.2
KO T cell

pSTAT1
0.6

DAPI
0

NS
12 *** Per cent of pSTAT1+ cells

(×104 per cm2)


CD8+ T cells
8 0 20 40 60 80
WT T cell

Fibroblast
4 Endothelial cell
Smooth muscle
0 Immune cell
WT KO T cell
Skin: Melanocyte

KO

T
KO

T
W

W
Melanocyte DAPI DCT CD8 Keratinocyte
T cell: KO WT Langerhans cell

Fig. 2 | Paracrine IFNγ signalling mediated by skin stromal cells drives CD8+ adoptive-transfer-based vitiligo model in wild-type and Ifngr1 KO tail skin
T cell cytotoxicity. a, b, Representative density plot images (a) and FACS epidermis. e, Schematic diagram, representative whole-mount images and
quantification of CD8+ T cells (b) in tail skin epidermis of wild-type (WT) and quantifications of melanocytes and CD8+ T cells in skin graft experiment.
Ifngr1 KO mice at day 33 after vitiligo induction (vit. induc.). c, Scatter plot of f, Representative immunofluorescent images and quantifications of pSTAT1
differentially expressed genes in skin CD8+ T cells from WT and Ifngr1 KO mice signal in different cell types in grafted skin after vitiligo induction in the host
after vitiligo induction. FPKM, fragments per kilobase of transcript per million mouse. Scale bars, 500 µm (e), 50 µm (f). For exact P values, see Source Data.
mapped reads. d, Scheme and FACS quantification of CD8+ T cells in For statistics, P summary and sample sizes, see Methods.

analysis revealed that the cytotoxic genes Gzma, Gzmb, Prf1 and Ccl5 target melanocytes were activated using the melanoma–Treg-induced
were significantly downregulated in CD8+ T cells isolated from Ifngr1 vitiligo method (Extended Data Fig. 5d–g). When wild-type and Ifngr1
KO skin compared to wild-type skin (Fig. 2c, Extended Data Fig. 4j). KO full-thickness tail skin were grafted onto the same Ifngr1 KO host
These results indicated that IFNγ signalling is essential for the skin mice, the activated Ifngr1 KO CD8+ T cells were only recruited into the
recruitment, local aggregation and cytotoxicity of CD8+ T cells that wild-type skin, leading to melanocyte loss, but not into the Ifngr1 KO
target epidermal melanocytes. skin (Fig. 2e). Thus, given a local environment of wild-type skin, the
To further confirm this finding, we used an intervention-free, sponta- Ifngr1 KO auto-reactive CD8+ T cells can be efficiently recruited and
neous model of vitiligo using Pmel TCR transgenic mice; this model gen- activated, leading to vitiligo development.
erates antigen-specific CD8+ T cells that recognize melanocytes36,37. The When we carried out the reverse graft experiment, the results were
Pmel;wild-type (Pmel;WT) mice spontaneously developed vitiligo-like unexpected. When wild-type and Ifngr1 KO full-thickness tail skin were
epidermal depigmentation, melanocyte loss and CD8+ T cell infiltration, grafted onto the same wild-type mice, after induction the host-derived
whereas the Pmel;Ifngr1 KO mice did not (Extended Data Fig. 4k–p). wild-type CD8+ T cells were robustly recruited into both grafted
To distinguish paracrine versus autocrine IFNγ signalling in orches- wild-type and grafted Ifngr1 KO tail skin equivalently, causing a loss of
trating the local activity of T cells in vitiligo-affected skin, we used a both wild-type and Ifngr1 KO melanocytes (Fig. 2e). This result directly
model of vitiligo based on the adoptive transfer of T cells20,38, which contradicted the findings of the adoptive T cell transfer experiment, in
relies on sub-lethally irradiated host mice receiving wild-type Pmel which the transferred wild-type Pmel T cells did not infiltrate the Ifngr1
CD8+ T cells and then hPMEL-AAV for the activation of Pmel T cells KO tail skin or lead to melanocyte loss. One possible explanation for
(Extended Data Fig. 4q–s). After receiving the same number of trans- this discrepancy is that wild-type cells from the host skin migrated into
ferred wild-type Pmel T cells (Extended Data Fig. 4t), wild-type recipient the grafted Ifngr1 KO tail skin, and functionally rescued the KO skin in
mice developed vitiligo, as shown by skin depigmentation, significant terms of driving vitiligo pathogenesis.
melanocyte loss and CD8+ T cell infiltration, whereas Ifngr1 KO recipi- A lineage-tracing experiment using C57 tail skin grafted onto
ent mice did not develop vitiligo (Fig. 2d, Extended Data Fig. 4u–x). In Rosa-mT-host mice showed that host dermal cells (mT+) did indeed
all three different models of vitiligo induction, only significantly more migrate into the grafted skin dermis, but neither host keratinocytes
CD8+ T cells were detected in wild-type skin compared to Ifngr1 KO skin, nor melanocytes did (Extended Data Fig. 5h). After induction of viti-
whereas the other five main immune cell types showed no difference ligo in the host mice, whereas no pSTAT1 signal was present in the
(Extended Data Fig. 5a–c). grafted skin of the KO-to-KO graft, in the KO-to-wild-type graft there
The adoptive T  cell transfer experiment revealed that parac- were abundant nuclear pSTAT1 signals in the grafted Ifngr1 KO skin;
rine IFNγ-signalling is essential for recruiting and orchestrating quantification showed that most of the pSTAT1+ cells were host-derived
auto-reactive CD8+ T cells in skin. To further narrow down whether fibroblasts (around 70%) (Fig. 2f, Extended Data Fig. 5i). On the basis
IFNγ-responsive skin stromal cells are required for vitiligo progression, of these results we concluded that IFNγ-responsive skin stromal cells
we used a skin graft experiment: full-thickness tail skin was grafted onto are required to drive auto-reactive CD8+ T cell-mediated vitiligo in a
the back of the host mouse, and then host-derived CD8+ T cells that paracrine manner.

120 | Nature | Vol 601 | 6 January 2022


a b c RFP-labelled 8 weeks
PdgfraCreER; Vitiligo model
WT Ifngr1fl/fl fibroblasts
induction

Ifngr1 KO fibroblasts WT fibroblasts

DAPI CD8 DCT


Ifngr1 KO Ifngr1 KO

+ Vitiligo induction
Keratinocyte Melanocyte
Endothelial cell Fibroblast
Myeloid immune cell T cell
DAPI CD8 RFP
No No. of CD8+ T cells per scale
induction + Vitiligo induction No. of CD8+ T cells per mm2
0 100 200 300 400 Fibroblast 0 20 40 60 80 100 120
WT WT TekCre K14Cre 120
cKO cKO WT
Vit.

No. of melanocytes
100 cKO WT
Melanocyte

induc.
80

per scale
+ KO
60 R = –0.85
Csf1rCre TyrCreER Cd4Cre PdgfraCreER P = 2.1 × 10–44
cKO cKO cKO cKO 40 R = –0.14 WT
20 P = 0.06 RFP+ region

KO RFP– region
0

d Activated OT-1 Fibroblast-conditioned e f CXCL9+ CXCL10+


CD8+ T cell medium Cells per mm

C CL 0

G A

P
X 3
C CL1

G CL9

LY BP
M 15
IS 5R
G P2
G P4
IL P5
C L5
C L8
C 74

PB
TA 6E
C M
IFNγ

2
0 100 200 300

1
C
C

B
D

B
B
B2

X
3

X
2
C
C
2 250
Progressive
80
Migration ratio (%)

Quiescent 0 200

T cells per mm
60 1× 5× 10× 25× Healthy donor –2 150
40

x 0

p
Ly bp
100

C cl3

Is 5ra
M 15
G cl9
C 74
C cl1

Il1 p5
G p2
G p4

Ta e
pb
C m

C l5
C l8

6
2
Vit. induc.

g
D
B2

b
b
b
c
2
3

x
x
C
C

20 1
– WT 50
+ WT 0
0
IFNγ – + – + – + – + – + – + – + – + + Ifngr1 KO 0
–1
Fibroblast KO WT R = 0.86 R = 0.94
P = 9.4 × 10–7 P = 9.9 × 10–10
g Per cent of CXCL9+ h No. of dermal CD8+ T cells per scale
or CXCL10+ cells
0 20 40 60 80 100 Vit. 0 100 200 300
shRNA H2BRFP induc.
Melanocyte P1 Scrambled – NS
Fibroblast Scrambled NS
+

Endothelial
shCXCL9_1 *
+

Muscle P56
shCXCL9_2 ****
+

Keratinocyte
Vitiligo
M. phagocyte shCXCL10_1 ***
+

CXCL9 induction
Langerhans CXCL10 shCXCL10_2 ****
+

P89
T cell Whole mount RFP+ region RFP– region

Fig. 3 | IFNγ-responsive dermal fibroblasts are uniquely required to fibroblast-conditioned medium. e, Heat map analysis of commonly
orchestrate autoimmune CD8+ T cells through secreted chemokines. a, upregulated secreted factors from vitiligo fibroblasts in human and mice.
Skin illustration and representative density plot images of melanocytes in tail f, Correlation analysis of densities of CD3+ T cells and CXCL9+ and CXCL10+ cells
skin epidermis of mice with the indicated genotypes at day 33 after vitiligo from the skin of patients with vitiligo. g, Quantification of the CXCL9+ and
induction. b, Representative whole-mount images and correlation analysis of CXCL10+ signal in different cell types in the skin of patients with vitiligo.
melanocytes versus CD8+ T cells in tail skin epidermis of wild-type and h, Experimental workflow and quantification of dermal CD8+ T cells from
Pdgfra CreER;Ifngr1 fl/fl cKO mice at day 33 after vitiligo induction. c, Experimental in vivo fibroblast knockdown experiment. P, postnatal day. See Extended Data
workflow, representative whole-mount images and quantification of CD8+ Fig. 4g for scale definition.  Scale bars, 500 µm (b, c). For exact P values, see
T cells in the fibroblast transplant experiment. d, Schematic diagram and Source Data. For statistics, P summary and sample sizes, see Methods.
quantification of the T cell transwell migration assay using

At day 33 of the melanoma–Treg-induced vitiligo model, similar


Vitiligo requires IFNγ-responsive fibroblasts to wild-type mice, TekCre;Ifngr1fl/fl, K14Cre;Ifngr1fl/fl, Csf1rCre;Ifngr1fl/fl,
Next, we used a cell-type-specific knockout strategy to identify the Cd4Cre;Ifngr1fl/fl and TyrCreER;Ifngr1fl/fl mice all developed vitiligo, as indi-
functionally important cell type in skin that drives vitiligo progres- cated by significant loss of melanocytes and CD8+ T cell infiltration in
sion. We ablated Ifngr1 in six different candidate cell types using the skin (Fig. 3a, Extended Data Fig. 6b–e). Only the PdgfraCreER;Ifngr1fl/fl
the cell-type-specific knockout strategy: TekCre;Ifngr1fl/fl mice to tar- mice did not develop vitiligo: skin pigmentation and melanocyte were
get endothelial cells; K14Cre;Ifngr1fl/fl mice to target keratinocytes; not affected, and the number of CD8+ T cells was significantly lower
Csf1rCre;Ifngr1fl/fl mice to target myeloid immune cells; Cd4Cre;Ifngr1fl/fl than that in wild-type and the other conditional knockout (cKO) mice
mice to target T cells; TyrCreER;Ifngr1fl/fl mice to target melanocytes; (Fig. 3b, Extended Data Fig. 6b–f).
and PdgfraCreER;Ifngr1fl/fl mice to target fibroblasts (Fig. 3a). qPCR con- To further validate this discovery, we used two more methods of
firmed the efficient ablation of Ifngr1 in each intended target cell type vitiligo induction. First, the spontaneous vitiligo development in
(Extended Data Fig. 6a). Pmel;WT mice did not occur in Pmel;PdgfraCreER;Ifngr1fl/fl cKO mice,

Nature | Vol 601 | 6 January 2022 | 121


Article
which exhibited no skin depigmentation, melanocyte loss or CD8+ T cell (Extended Data Fig. 8f, g). Together, these in vitro and in vivo results
infiltration (Extended Data Fig. 7a–c). Second, after adoptive-transfer of show that IFNγ-responsive fibroblasts mediate local aggregation of
an equivalent amount of wild-type Pmel T cells (Extended Data Fig. 7d), CD8+ T cells through the CXCL9/CXCL10–CXCR3 axis.
wild-type recipient mice developed vitiligo but PdgfraCreER;Ifngr1fl/fl cKO
recipient mice did not, as shown by the lack of epidermal depigmenta-
tion, melanocyte loss or CD8+ T cell infiltration in skin (Extended Data Regional fibroblasts differ in IFNγ response
Fig. 7e–g). Thus, regardless of the vitiligo induction method used, At the whole-organ level, dermal fibroblasts are not homogeneous:
PdgfraCreER;Ifngr1fl/fl cKO mice phenocopied the Ifngr1 KO mice and did fibroblasts from different anatomic skin regions are intrinsically dif-
not develop vitiligo. ferent in their expression of HOX genes41,43. However, whether skin
To test whether IFNγ-responsive dermal fibroblasts are not only fibroblasts from different anatomic regions are different in regulating
required, but also sufficient to induce local aggregation of CD8+ T cells, the activity of cutaneous immune cells is unknown.
we used a fibroblast transplantation assay (Extended Data Fig. 7h). To investigate this, we first quantified the frequencies of viti-
Intradermal injection of RFP-labelled wild-type—but not Ifngr1 KO— ligo lesions at different anatomic regions of 2,265 patients with
fibroblasts into the tail skin of Ifngr1 KO mice resulted in significant local non-segmental vitiligo (Supplementary Table 7). We quantified eight
aggregation of CD8+ T cells after the induction of vitiligo (Fig. 3c). This main body regions and found large variations in the frequency of vitiligo
finding shows that in a local environment that is otherwise completely among them, with the back of the hand, the chest and the back being
non-responsive to IFNγ, IFNγ-responsive fibroblasts alone are sufficient the most susceptible to vitiligo, and the palm and arm the least (Fig. 4a).
to mediate the local recruitment of CD8+ T cells. Next, we conducted an RNA-seq analysis of in vitro IFNγ-treated
primary human dermal fibroblasts isolated from these eight anatomic
regions. The patterns of HOX gene expression indicated that fibro-
Fibroblasts recruit T cells through CXCL9/CXCL10 blasts retained their positional information during in vitro culture
To distinguish whether this essential fibroblast function is depend- (Extended Data Fig. 9a–c, Supplementary Table 8). Heat map and
ent on cell–cell contact or secreted factors, we used an in vitro T cell qPCR analysis revealed that the numbers and types of upregulated
transwell migration assay using IFNγ-treated fibroblast-conditioned genes in response to IFNγ varied significantly among region-specific
medium (Fig. 3d, Extended Data Fig. 7i); quantifications showed that fibroblasts (Fig. 4b). Although fibroblasts from all anatomic regions
medium from IFNγ-treated wild-type but not Ifngr1 KO fibroblasts could expressed similar levels of the IFNγ signalling pathway genes IFNGR1,
induce the migration of CD8+ T cells in a concentration-dependent JAK1, JAK2 and STAT1 (Extended Data Fig. 9d), multiple IFNγ-induced
manner. To identify the secreted factors involved in this process, we chemokine genes—including CXCL9, CXCL10 and their human-specific
compared the transcriptome analysis of human scRNA-seq data and variant CXCL11—were significantly upregulated in fibroblasts that were
mouse RNA-seq data of fibroblasts isolated from vitiligo skin (Extended isolated from anatomic regions with a higher incidence of vitiligo, such
Data Fig. 7j, Supplementary Tables 4–6). We identified 28 overlapping as the back of the hand, the back of the foot, the chest and the back
genes that encode fibroblast-specific secreted proteins, 11 of which (Fig. 4c, Extended Data Fig. 9e). Further correlation analysis showed
are conserved major histocompatibility complex genes and 5 encode that the percentage of vitiligo incidence positively correlated with the
chemokines—namely CCL5, CCL8, CXCL3, CXCL9 and CXCL10 (Fig. 3e). intrinsic IFNγ response of dermal fibroblasts at different anatomic
A T cell transwell migration assay using these secreted chemokines regions (Fig. 4d).
showed that CXCL9 and CXCL10 induced the migration of CD8+ T cells
in a dosage-dependent manner (Extended Data Fig. 7k).
CXCL9 and CXCL10 have been reported to be involved in vitiligo Distinct fibroblasts drive vitiligo patterns
and can be expressed by many different cell types24,25,39,40. We found To study the functional relevance of this correlation, we turned to the
spatially that the density of CD3+ T cells positively correlated with the melanoma–Treg-induced mouse model of vitiligo. In different anatomic
density of CXCL9+ and CXCL10+ cells in patient skin (Fig. 3f, Extended regions of mouse skin, melanocytes are ubiquitously present in hair
Data Fig. 7l). Co-localization immunofluorescent analysis revealed that follicles but not in epidermis, so we only quantified melanocytes in
fibroblasts accounted for around 80% of the total number of CXCL9- hair follicles at four anatomic regions with a similar hair follicle density
and CXCL10-expressing cells in the skin of patients with vitiligo (Fig, (Extended Data Fig. 9f): dorsal, ventral, paw dorsal and paw ventral. In
3g, Extended Data Fig. 7m, n). These results showed that fibroblasts human vitiligo, hair depigmentation tends to occur after prolonged
are the main source of both chemokines during vitiligo progression. pathogenesis6,44, which was also observed in the skin of vitiligo-affected
To functionally test whether these fibroblast-derived chemokines are mice (Extended Data Fig. 2b). We therefore quantified the number of mel-
involved in vitiligo pathogenesis, we used in vivo fibroblast knockdown anocytes in hair follicles after long-term vitiligo (at day 300). Melanocytes
experiments41 (Fig. 3h). Intradermal injection of lentivirus expressing in dorsal and ventral skin hair follicles were depleted in vitiligo-induced
short hairpin RNA (shRNA) resulted in efficient and specific knockdown mice compared to control mice, but melanocytes in paw dorsal and paw
of Cxcl9 or Cxcl10 in dermal fibroblasts (Extended Data Fig. 8a–c). ventral skin hair follicles remained intact; correspondingly, increases of
After vitiligo induction, CD8+ T cells were increased in skin expressing CD3+CD8+ T cells were significantly higher in the dorsal and ventral skin
scrambled shRNA, but not in skin expressing Cxcl9 and Cxcl10 shRNA than in paw dorsal and paw ventral skin (Fig. 4e, Extended Data Fig. 9g,
(Fig. 3h). Correspondingly, melanocytes in the overlaying epidermis h). These results revealed that there are differences in vitiligo occurrence
of the Cxcl9- and Cxcl10-shRNA-expressing dermis were also intact among different anatomic regions in mice, as in humans.
(Extended Data Fig. 8d). Notably, we found that Cxcl9 and Cxcl10 showed higher expression
Fluorescence-activated cell sorting (FACS) quantifications of six levels in dorsal and ventral fibroblasts compared to paw dorsal and
types of skin immune cell from wild-type and PdgfraCreER;Ifngr1fl/fl cKO paw ventral fibroblasts either from in-vivo-isolated cells in long-term
mice with all three different methods of vitiligo induction showed that vitiligo mice, or in IFNγ-treated cultured cells in vitro (Fig. 4f, Extended
only significantly fewer CD8+ T cells were detected in the cKO compared Data Fig. 9i). To study the functional relevance of this expression level
to the wild-type skin, whereas all of the other immune cells did not difference, we conducted a T cell transwell migration assay with con-
show a significant difference (Extended Data Fig. 8e). CXCR3 is the ditioned medium. The results showed that medium from IFNγ-treated
confirmed receptor for CXCL9 and for CXCL1042. Quantifications in dorsal skin fibroblasts induced significantly more (more than two times
skin from both patients with vitiligo and vitiligo-affected mice showed as much) CD8+ T cell migration than did medium from paw ventral skin
that CD8+ T cells constitute the majority of the CXCR3+ immune cells fibroblasts (Fig. 4g, Extended Data Fig. 9j). Our in vivo data showed that

122 | Nature | Vol 601 | 6 January 2022


a Vitiligo b
Site No. incidence CXCL10 CXCL11 CXCL9 c 1.5 CXCL9
CXCL10

relative to GAPDH
Hand back 558 24.64% Hand back
Chest 465 20.53%

Expression
Foot back 1.0
Back 460 20.31% Chest
Leg 405 17.88% Back
Foot back 298 13.16% 0.5
Leg
Head 296 13.07%
Arm 201 8.87% Arm
Palm 107 4.72% Head 0
Palm
Total 2,265

ba k
C ck
Ba st
Le k
Ar g
H m
Pa d
lm
ot ac

ea
he
Fo d b
2 0 –2

an
H
T cells (%) per hair follicle
d e NS f

Melanocytes
DAPI DCT P-cad 8 **** 8 Cxcl9 Cxcl10
0.905 Foot back 6

Control
R = 0.62 Chest
Enrichment score

realtive to Ppib
0.900 2 6
Back

Expression
0
0.895 Hand back 4
Arm 15 ** ***

Day 300
Leg 10

CD8+
0.890 Head Vitiligo 2
5
0.885 Palm 0 0
5 10 15 20 25 Paw Dorsal Vit. induc. – + – +

d o aw

ve Paw

al

l
ra
Vitiligo incidence (%)

al

Do l
Dorsal

rs
ra

nt
P
rs

nt

Ve
g h DAPI CD8 RFP i j
60 * Cxcl9 Cxcl10
1× 2× 5× 20 **** T0
Ifngr1 KO

25 6
Enrichment score
Migration ratio

Dorsal

15

realtive to Ppib
40 20 5
T200

Expression
(%)

10 15 4
20 3
5 10 T350
Paw dorsal

2
Fibroblast

5 1
0 0 Normal chemotactic fibroblast
0 0
IFNγ – + – + – + – + – + – + Weak chemotactic fibroblast
Paw dorsal Dorsal Activated fibroblast
Fibrolast Paw ventral Dorsal CD8+ T cell Melanocyte
+ Vitiligo induction

k Scrambled shRNA JAK1 shRNA


300
CD8+ T cells per scale

Vitiligo
induction shRNA
+ Vitiligo induction

No. of dermal

200 – Scrambled
+ Scrambled
100 + Ifngr1
+ Jak1
+ Stat1
0
0 25 50 75 100
DAPI CD8 CD8+ T cell DAPI CD8 CD8+ T cell Infected fibroblasts per scale (%)
RFP RFP+ fibroblast RFP RFP+ fibroblast

Fig. 4 | Anatomically distinct fibroblast subsets with differential IFNγ g, Quantification of migration ratio from T cell transwell migration assay using
responses determine organ-level skin autoimmune patterns. IFNγ-treated fibroblast-conditioned medium. h, Representative whole-mount
a, Quantification of lesion frequencies at anatomically distinct skin regions in images and quantification of enrichment score of CD8+ T cells in fibroblast-
patients with non-segmental vitiligo. b, Heat map of 92 upregulated genes in injected area of Ifngr1 KO mice after vitiligo induction. i, qPCR of Cxcl9 and Cxcl10
IFNγ-treated human dermal fibroblasts from eight anatomical regions. c, qPCR of in re-isolated fibroblasts from h. j, Mathematical model predicts the regional
CXCL9 and CXCL10 from IFNγ-treated human dermal fibroblasts in culture. distribution pattern of T cells with mosaic IFNγ-responsive fibroblasts.
d, Correlation analysis of fibroblasts IFNγ response enrichment score versus k, Representative whole-mount and density plot images and correlation analysis
vitiligo incidence frequency. e, Representative immunofluorescent images and of the number of T cells versus the percentage of infected fibroblasts in the in vivo
quantifications of melanocytes and CD8+ T cells in different skin regions in mosaic fibroblast knockdown experiment. See Extended Data Fig. 4g for scale
control and vitiligo-affected mice at day 300. f, qPCR of Cxcl9 and Cxcl10 in definition. Scale bars, 50 µm (e), 500 µm (h, k). For exact P values, see Source
IFNγ-treated-region-specific mouse dermal fibroblasts in culture. Data. For statistics, P summary and sample sizes, see Methods.

a twofold increase in the number of T cells could result in significant To further test whether regional variation in fibroblasts could result
differences in melanocyte loss (Extended Data Fig. 9k). in patterned T cell activity, we built a mathematical model to study the
Next to directly compare the region-specific fibroblasts in CD8+ T cell effect of mosaic fibroblasts on T cell activity (Extended Data Fig. 10a–c,
recruitment in vivo, we intradermally injected RFP-labelled dorsal and paw Supplementary Videos 1–6). In a field of fibroblasts composed of nor-
dorsal wild-type fibroblasts into Ifngr1 KO mice tail skin, then conducted mal and weak chemotactic regions (Fig. 4j, Extended Data Fig. 10d),
vitiligo model induction in the host mice. Quantification showed that there T cells were competitively recruited into the normal chemotactic
was a significantly stronger enrichment of CD8+ T cells in dorsal fibro- region, leading to preferential expansion of T cell clusters in those
blast area than in paw fibroblast area (Fig. 4h). In addition, the re-isolated regions and corresponding melanocyte loss (Supplementary Video 7).
dorsal fibroblasts showed 18 times higher expression of Cxcl9 and 4 times To functionally test this model, we used the fibroblast knockdown
higher expression of Cxcl10 than did paw dorsal fibroblasts (Fig. 4i). approach to create a mosaic field containing fibroblasts with different

Nature | Vol 601 | 6 January 2022 | 123


Article
IFNγ-responsive abilities. By using shRNA against Ifngr1, Jak1 or Stat1 15. Cheuk, S. et al. CD49a expression defines tissue-resident CD8+ T cells poised for
cytotoxic function in human skin. Immunity 46, 287–300 (2017).
with high knockdown efficiency we could efficiently block IFNγ down- 16. Iannella, G. et al. Vitiligo: pathogenesis, clinical variants and treatment approaches.
stream signal in fibroblasts in vitro (Extended Data Fig. 10e, f). Quan- Autoimmun. Rev. 15, 335–343 (2016).
tification showed that in dermis expressing scrambled shRNA, CD8+ 17. Rodrigues, M. et al. Current and emerging treatments for vitiligo. J. Am. Acad. Dermatol.
77, 17–29 (2017).
T cells infiltrated both the RFP+ and RFP− regions equivalently, but in 18. Kovacs, S. O. Vitiligo. J. Am. Acad. Dermatol. 38, 647–666 (1998).
dermis expressing shRNAs against IFNGR1, JAK1 or STAT1, the number 19. Gregg, R. K., Nichols, L., Chen, Y., Lu, B. & Engelhard, V. H. Mechanisms of spatial and
of CD8+ T cells negatively correlated with the percentage of infected temporal development of autoimmune vitiligo in tyrosinase-specific TCR transgenic
mice. J. Immunol. 184, 1909–1917 (2010).
fibroblasts. Correspondingly, in the overlaying epidermis, the number 20. Harris, J. E. et al. A mouse model of vitiligo with focused epidermal depigmentation
of melanocytes positively correlated with the percentage of infected requires IFN-gamma for autoreactive CD8+ T-cell accumulation in the skin. J. Invest.
fibroblasts (Fig. 4k, Extended Data Fig. 10g–i). The fibroblast mosaic Dermatol. 132, 1869–1876 (2012).
21. Chatterjee, S. et al. A quantitative increase in regulatory T cells controls development
knockdown experiments validated the modelling prediction that in a of vitiligo. J. Invest. Dermatol. 134, 1285–1294, (2014).
field with uneven fibroblast responses to IFNγ, CD8+ T cells preferen- 22. Yang, L. et al. Interferon-gamma inhibits melanogenesis and induces apoptosis in
tially aggregate towards a region with fibroblasts capable of stronger melanocytes: a pivotal role of CD8+ cytotoxic T lymphocytes in vitiligo. Acta Derm.
Venereol. 95, 664–670 (2015).
IFNγ responses, hence generating a patterned loss of melanocytes. 23. Wang, S. Q. et al. Interferon-γ induces senescence in normal human melanocytes.
PLoS One 9, e93232 (2014).
24. Tulic, M. K. et al. Innate lymphocyte-induced CXCR3B-mediated melanocyte apoptosis
is a potential initiator of T-cell autoreactivity in vitiligo. Nat. Commun. 10, 2178 (2019).
Discussion 25. Richmond, J. M. et al. Keratinocyte-derived chemokines orchestrate T-cell positioning in
Here we have identified a feedforward system between mobile CD8+ the epidermis during vitiligo and may serve as biomarkers of disease. J. Invest. Dermatol.
T cells and immobile fibroblasts, which functions to prioritize the allo- 137, 350–358 (2017).
26. Biswas, K. B. et al. GPNMB is expressed in human epidermal keratinocytes but disappears
cation of collective cytotoxicity and efficient elimination of target cells. in the vitiligo lesional skin. Sci. Rep. 10, 4930 (2020).
The cyclical nature of the feedforward loop leads to the progressive 27. Boukhedouni, N. et al. Type-1 cytokines regulate matrix metalloprotease-9 production
aggregation of CD8+ T cells in areas with IFNγ-responsive fibroblasts, and E-cadherin disruption to promote melanocyte loss in vitiligo. JCI Insight 4, e133772
(2020).
as well as sharply declined CD8+ T cells in areas with IFNγ-resistant 28. Ganju, P. et al. Microbial community profiling shows dysbiosis in the lesional skin of
fibroblasts. Many other autoimmune skin diseases in addition to viti- vitiligo subjects. Sci Rep. 6, 18761 (2016).
29. Yuan, X. et al. The impact of NBUVB on microbial community profiling in the lesional skin
ligo exhibit regional preference with symmetric distributions45–50. The
of vitiligo subjects. Microb. Pathog. 140, 103943 (2020).
immune cell activity regulated by regional fibroblasts could provide a 30. Lang, K. S. et al. HLA-A2 restricted, melanocyte-specific CD8+ T lymphocytes detected in
general paradigm through which the progression and regional pattern vitiligo patients are related to disease activity and are predominantly directed against
MelanA/MART1. J. Invest. Dermatol. 116, 891–897 (2001).
of skin autoimmune diseases are regulated. Besides the skin, fibro-
31. Namazi, M. R. Neurogenic dysregulation, oxidative stress, autoimmunity, and
blasts are ubiquitously present in essentially all organs of our body; melanocytorrhagy in vitiligo: can they be interconnected? Pigment Cell Res. 20,
our study also provides avenues for investigation of the potentially 360–363 (2007).
32. Lotti, T., D’Erme, A. M. & Hercogova, J. The role of neuropeptides in the control of regional
broad application of this cellular cross-talk.
immunity. Clin. Dermatol. 32, 633–645 (2014).
33. Palermo, B. et al. Specific cytotoxic T lymphocyte responses against Melan-A/MART1,
tyrosinase and gp100 in vitiligo by the use of major histocompatibility complex/peptide
Online content tetramers: the role of cellular immunity in the etiopathogenesis of vitiligo. J. Invest.
Dermatol. 117, 326–332 (2001).
Any methods, additional references, Nature Research reporting sum- 34. Mantovani, S. et al. Dominant TCR-α requirements for a self antigen recognition in
maries, source data, extended data, supplementary information, humans. J. Immunol. 169, 6253–6260 (2002).
35. Byrne, K. T., Zhang, P., Steinberg, S. M. & Turk, M. Autoimmune vitiligo does not require
acknowledgements, peer review information; details of author con-
the ongoing priming of naive CD8 T cells for disease progression or associated
tributions and competing interests; and statements of data and code protection against melanoma. J. Immunol. 192, 1433–1439 (2014).
availability are available at https://doi.org/10.1038/s41586-021-04221-8. 36. Mosenson, J. A. et al. Mutant HSP70 reverses autoimmune depigmentation in vitiligo.
Sci. Transl. Med. 5, 174ra128 (2013).
37. Miao, X. et al. PD-L1 reverses depigmentation in Pmel-1 vitiligo mice by increasing the
1. Larsen, S. B., Cowley, C. J. & Fuchs, E. Epithelial cells: liaisons of immunity. Curr. Opin. abundance of Tregs in the skin. Sci Rep. 8, 1605 (2018).
Immunol. 62, 45–53 (2019). 38. Riding, R. L., Richmond, J. M. & Harris, J. E. Mouse model for human vitiligo. Curr. Protoc.
2. Rognoni, E. & Watt, F. M. Skin cell heterogeneity in development, wound healing, and Immunol. 124, e63 (2019).
cancer. Trends Cell Biol. 28, 709–722 (2018). 39. Richmond, J. M. et al. Resident memory and recirculating memory T cells cooperate
3. Afshar, M. & Gallo, R. L. Innate immune defense system of the skin. Vet. Dermatol. 24, to maintain disease in a mouse model of vitiligo. J. Invest. Dermatol. 139, 769–778
32–38 (2013). (2019).
4. Taieb, A. & Picardo, M. Vitiligo. N. Engl. J. Med. 360, 160–169 (2009). 40. Rashighi, M. et al. CXCL10 is critical for the progression and maintenance of
5. Alikhan, A., Felsten, L. M., Daly, M. & Petronic-Rosic, V. Vitiligo: a comprehensive overview depigmentation in a mouse model of vitiligo. Sci. Transl. Med. 6, 223ra223 (2014).
Part I. Introduction, epidemiology, quality of life, diagnosis, differential diagnosis, 41. Yu, Z. et al. Hoxc-dependent mesenchymal niche heterogeneity drives regional hair
associations, histopathology, etiology, and work-up. J Am. Acad. Dermatol. 65, 473–491 follicle regeneration. Cell Stem Cell 23, 487–500 (2018).
(2011). 42. Groom, J. R. & Luster, A. D. CXCR3 in T cell function. Exp. Cell. Res. 317, 620–631
6. Picardo, M. et al. Vitiligo. Nat. Rev. Dis. Primers 1, 15011 (2015). (2011).
7. Ezzedine, K., Eleftheriadou, V., Whitton, M. & van Geel, N. Vitiligo. Lancet 386, 74–84 43. Chang, H. Y. et al. Diversity, topographic differentiation, and positional memory in
(2015). human fibroblasts. Proc. Natl Acad. Sci. USA. 99, 12877–12882 (2002).
8. Taieb, A. Intrinsic and extrinsic pathomechanisms in vitiligo. Pigment Cell Res. 13, 41–47 44. Paus, R., Ito, N., Takigawa, M. & Ito, T. The hair follicle and immune privilege. J. Investig.
(2000). Dermatol. Symp. Proc. 8, 188–194 (2003).
9. Spritz, R. A. & Santorico, S. A. The genetic basis of vitiligo. J. Invest. Dermatol. 141, 45. Farber, E. M., Nickoloff, B. J., Recht, B. & Fraki, J. E. Stress, symmetry, and psoriasis:
265–273 (2021). possible role of neuropeptides. J. Am. Acad. Dermatol. 14, 305–311 (1986).
10. Frisoli, M. L., Essien, K. & Harris, J. E. Vitiligo: mechanisms of pathogenesis and treatment. 46. Boehncke, W.-H. & Schön, M. P. Psoriasis. Lancet 386, 983–994 (2015).
Annu. Rev. Immunol. 38, 621–648 (2020). 47. Greb, J. E. et al. Psoriasis. Nat. Rev. Dis. Primers 2, 16082 (2016).
11. Le Poole, I. C., van den Wijngaard, R. M., Westerhof, W. & Das, P. K. Presence of T cells and 48. Gabrielli, A., Avvedimento, E. V. & Krieg, T. Scleroderma. N. Engl. J. Med. 360, 1989–2003,
macrophages in inflammatory vitiligo skin parallels melanocyte disappearance. Am. J. (2009).
Pathol. 148, 1219–1228 (1996). 49. Kaul, A. et al. Systemic lupus erythematosus. Nat. Rev. Dis. Primers 2, 16039 (2016).
12. Ogg, G. S., Rod Dunbar, P., Romero, P., Chen, J. L. & Cerundolo, V. High frequency of 50. Boyd, A. S. & Neldner, K. H. Lichen planus. J. Am. Acad. Dermatol. 25, 593–619 (1991).
skin-homing melanocyte-specific cytotoxic T lymphocytes in autoimmune vitiligo. J. Exp.
Med. 188, 1203–1208 (1998).
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
13. Van den Boorn, J. G. et al. Autoimmune destruction of skin melanocytes by perilesional
published maps and institutional affiliations.
T cells from vitiligo patients. J. Invest. Dermatol 129, 2220–2232 (2009).
14. Maeda, Y. et al. Detection of self-reactive CD8+ T cells with an anergic phenotype in
healthy individuals. Science 346, 1536–1540 (2014). © The Author(s), under exclusive licence to Springer Nature Limited 2021

124 | Nature | Vol 601 | 6 January 2022


Methods (stock no. 007906) and Rosa-stop-mTmG (stock no. 007676) mice were
obtained from The Jackson Laboratory. For CreER activation, Tamoxifen
Ethics statement was dissolved in sunflower oil with 10% ethanol. PdgfraCreER;Ifngr1fl/fl
The scRNA-seq and immunofluorescent analysis of human vitiligo skin and TyrCreER;Ifngr1fl/fl mice received an intraperitoneal injection of
biopsies study were reviewed and approved by the Ethics Committee 100 μl of 20 mg ml−1 tamoxifen solution every other day from P42 to
of Beijing Hospital. All donors signed the written informed consent for P56. Knockout efficiency was validated by cell-type-specific qPCR. It
sample collection and data analysis. For minors, informed consent was needs to be mentioned here that TekCre labels both endothelial cells
obtained from the legally authorized representatives of the minors. and haematopoietic cells51, so Ifngr1 expression was ablated in both
Before signing the consent, necessary information including the goals cell types in the TekCre;Ifngr1fl/fl mice. Although Csf1r mainly expresses
and related experimental procedures for the study were provided to and functions in myeloid cells, the Cre activity of Csf1rCre is detected
the donors. In this study, we collected 3 × 3-mm2 skin biopsies from in multiple immune cell types including macrophages, dendritic cells,
treatment-naive patients with vitiligo and control healthy donors who monocytes, granulocytes and T and B cells52,53. Ifngr1 expression was
had surgical operations. The biopsies were then digested into single therefore ablated in the immune cells including macrophages and
cells for further scRNA-seq analysis. For the representative skin lesion dendritic cells in Csf1rCre;Ifngr1fl/fl mice.
pictures shown in Extended Data Fig. 1a, patients provided written
informed consent and agreed to allow photographs of the selected Cell culture
area to be taken and used. All of the cell lines were cultured at 37 °C in a cell incubator with 5% CO2.
The study of anatomically distinct human dermal fibroblasts was B16F10 cells (ATCC, CRL-6475) were maintained in DMEM medium sup-
reviewed and approved by the Peking University Third Hospital Medi- plemented with 10% (v/v) FBS and 1% (v/v) pen–strep. Primary mouse
cal Science Research Ethics Committee. All donors signed the written fibroblasts were maintained in DMEM supplemented with 10% (v/v)
informed consent for sample collection. In this study, we collected skin FBS, 1% (v/v) pen–strep, and 1× antibiotic–antimycotic. The 293FT
biopsies from human embryos between 22 to 24 weeks of gestation cells (Thermo Fisher Scientific, R70007) used for virus packaging were
(weeks after estimated fertilization time). Skin biopsies from anatomi- maintained in DMEM medium supplemented with 10% (v/v) FBS, 1% (v/v)
cally distinct regions were collected, including the back of the hand, pen–strep, 1% (v/v) l-glutamine, 1% (v/v) 100 mM sodium pyruvate, 1%
back of the foot, palm, head, chest, back, leg and arm. Fibroblasts were (v/v) 7.5% sodium bicarbonate and 500 µg ml−1 G418. All cell lines tested
isolated from skin biopsies. Informed consent was obtained from all negative for mycoplasma contamination.
couples. Before giving consent, necessary information including the
goals and related experimental procedures for the study were provided Single-cell collection from human skin biopsies
to the couples. Tissue processing and the single-cell purification process started within
3 h after surgical skin biopsy collection. After removal of subcutaneous
Samples from patients with vitiligo fat, skin tissue was placed dermis side down in 6 ml of 2.4 U ml−1 disease
Seven male and three female patients clinically diagnosed with vitiligo solution and incubated at 37 °C with shaking at 80 rpm for 70 min. Epi-
were enrolled in this study. Their ages ranged from 6 to 55, with a median dermis with hair follicles was carefully separated from the dermis using
age of 24. Among these patients, four were diagnosed as being in the forceps. The separated epidermis was then placed dermis side down
active stage, and the disease stage was uncertain for the other six. The in 6 ml of 0.25% trypsin at 37 °C for 10 min, and afterwards 6 ml of 5%
clinical information regarding disease state is empirically based on FBS medium was added to neutralize trypsin. The epidermal single-cell
observations of whether the white macules or patches have blurred suspension was obtained by repeatedly aspirating and dispensing
boundaries. In addition to this, it relies on feedback from patients about the solution with a 1-ml pipette 10 times, and then filtered through
whether or not the depigmentation areas were progressively expanding two strainers (70 μm followed by 40 μm). Cells were then stained with
in the last month prior to hospital visits. All of the patients were newly anti-human CD45 (BD, cat: 11-9459-41, clone: 2D1, dilution:1:300) and
diagnosed with vitiligo and none of the patients had received therapy anti-human CD117 (BD, cat 555714, clone: YB5.B8, dilution:1:300) for
or UVB treatment. A 3 × 3-mm2 biopsy was taken from each patient in 15 min to detect immune cells and melanocytes, and then washed. The
the junction region between the depigmented lesion region and pig- dermis was placed in 10 ml of 2 mg ml−1 collagenase (Sigma-Aldrich,
mented perilesional region. A biopsy of the same size was also taken C2674) and incubated at 37 °C with shaking at 80 rpm for 1 h; after-
from five healthy donors with other surgical operations as a control. wards, 10 ml of 5% FBS medium was added. Single-cell suspensions
The clinical characteristics of these patients and healthy donors are were obtained by repeatedly aspirating and dispensing with a pipette
summarized in Extended Data Fig. 1c. for 10 times. The cells were then filtered through two strainers (70 μm
followed by 40 μm), and then stained for 15 min with anti-human CD45
Mice (BD, 11-9459-41, clone: 2D1, dilution:1:300) for immune cell detection
Mice were bred and maintained in the National Institute of Biological and then washed.
Sciences (NIBS) specific-pathogen-free facility in accordance with the On the basis of FACS analysis, single cells of different subtypes,
Guide for the Care and Use of Laboratory Animals of NIBS. Procedures including immune cells (CD45+) from both the epidermis and dermis,
were approved by the Laboratory Animal Management Committee of melanocytes (CD117+) from the epidermis, dermal cells (CD45−) from
NIBS and were in compliance with all relevant ethical regulations. All the dermis and keratinocytes (CD45−, CD117−) from the epidermis were
mice used in experiments were socially housed under a 12-h light–dark sorted into collection buffer, which contains 0.04% BSA in PBS to mini-
cycle with free access to food and water, with 23 °C–25 °C and 50%–56% mize cell losses and aggregation. For each skin biopsy sample, all of
humidity. the immune cells, melanocytes and dermal cells, as well as an equal
C57BL/6 mice were purchased from Charles River Laboratories. K14Cre number of keratinocytes to all of the other three cell types combined,
(MGI: 1926500) and K14-H2BGFP (MGI:5286142) mice were provided were pooled together for the next step of single-cell analysis.
by E. Fuchs. Ifngr1 KO mice ( Jackson stock no. 003288) and TekCre mice
( Jackson stock no. 004128) were provided by F. Shao. OT-1 mice (stock Vitiligo induction in mice
no. 003831) were provided by L. Chen. Pmel TCR transgenic mice (stock Human vitiligo pathogenesis is driven by diverse CD8+ cytotoxic T cells
no. 005023), PdgfraCreER (stock no. 018280), Csf1rCre (stock no. 021024), recognizing different endogenous melanocyte-specific antigens,
TyrCreER (stock no. 012328), Cd4Cre (stock no. 017336), Foxp3Cre (stock no. including MART-126-35 and TYR369-37754. Clinically it was discovered that
008694), Ifngr1 flox (stock no. 025394), Ai14 (stock no. 007914) Ai6 melanoma immunotherapy activates CD8+ T cells recognizing these
Article
antigens shared by melanoma cells and melanocytes55, which results in
vitiligo occurrence in 13.5%–25.7% of patients with melanoma receiving Tail skin graft
immunotherapy56–58. In addition, loss of Treg cells that can induce anergy Full-thickness tail skins from 4–6-week-old female mice were removed,
of autoimmune CD8+ T cells is likely to contribute to vitiligo occurrence flattened and cut into 1 × 1-cm squares. Then two 1 × 1-cm square skin
in humans. In mouse studies, vitiligo-like hair coat depigmentation has pieces were cut off from the back of anaesthetized 8-week-old female
been reported after B16 melanoma inoculation followed by CD4+ T cell recipient mice with the indicated genotype. Donor skin pieces were
depletion, but epidermal melanocyte loss—which is the hallmark of placed onto the backs of anaesthetized 8-week-old female recipient
human vitiligo pathology—has not been studied. On the basis of these mice with the indicated genotypes, with each recipient receiving a
previous reports, we developed a melanoma–Treg-induced protocol by wild-type and KO graft. Grafts were secured by sterile gauze and elastic
using B16F10 melanoma inoculation coupled with Treg cell depletion to bandages, which were removed at 8–10 days later. Note that we only
activate endogenous auto-reactive CD8+ T cells targeting epidermal used female mice for both donor and recipient, because sex-unmatched
melanocytes, and induced vitiligo occurrence in mice. First the dorsal graft would result in skin necrosis within 20 days. For C57 to mTmG or
skin in the right flank of 8–9-week old C57 mice was intradermally inocu- CD4Cre;mTmG grafts, the transgenic host mice were crossed to C57 to
lated with 2 × 105 B16F10 melanoma cells (day 0), then CD4 depletion avoid immune rejection.
antibodies were injected on days 4 and 10. Only mice that developed
primary tumours were used for further analysis. Primary tumours were Immunofluorescence staining
surgically removed on day 12. Spontaneous tumour metastases were For section staining, tissues were embedded in OCT compound, fro-
not observed with this B16F10 cell line, and mice with recurrent primary zen, cryosectioned (20–30 µm), and fixed for 10 min in 4% paraform-
tumours after surgery were not used for further study. aldehyde in PBS. Sections were washed in PBS overnight and then
Melanocytes in mouse dorsal skin are located in hair follicles but permeabilized for 20 min in 0.3% H2O2 in methanol at −20 °C and
not in the epidermis; only in mouse tail skin are melanocytes located blocked for 1 h in a solution of 2% normal donkey serum, 1% BSA and
in both the hair follicle and epidermis similar to human skin. Skin epi- 0.3% Triton in PBS at room temperature. The signal of human pSTAT1,
dermis depigmentation is the defining feature of vitiligo pathology, human CD8, human PDGFRA, human CD11c, human langerin, human
therefore only mouse tail skin was used for vitiligo analysis, unless CD3 and mouse pSTAT1 was amplified by the ABC Kit (Vector) and TSA
otherwise specified. The turnover rate of skin epidermis is about 30 Kit (Thermo Fisher Scientific). The following antibodies were used:
days59. Epidermal melanocyte loss peaked at day 30 after induction, anti-human TYR (made by the T.C. laboratory; 1:500), anti-human DCT
and epidermis depigmentation was visually apparent by day 60 after (made by the T.C. laboratory; 1:500), anti-mouse KRT14 (made by the
induction. Tail skin epidermal depigmentation was initially patchy T.C. laboratory; 1:1,000), anti-human pSTAT1 (CST, cat: 9167, clone:
and then progressed to eventually cover the entire epidermal surface, 58D6, dilution: 1:500), anti-human PDGFRA (Abcam, cat: ab203491,
which did not recover even more than a year later. Similar to patients clone: EPR22059-270, dilution: 1:500), anti-human CD31 (Abcam,
with vitiligo, there were no symptoms of skin inflammation in this mela- cat: ab24590, clone: P2B1, dilution: 1:500), anti-human aSMA (Sigma,
noma–Treg-induced vitiligo mouse model, including redness, swelling cat: C6198, clone: 1A4, dilution: 1:500), anti-human CD11c (Abcam,
or epidermal scaliness. cat: ab 52632, clone: EP1347Y, dilution: 1:500), anti-human langerin
The adoptive-transfer-based vitiligo mouse model was generated (Abcam, cat: ab192027, clone: EPR15863, dilution: 1:500), anti-human
according to the protocol described previously. In brief, naive Pmel CD3 (BD, cat: 563423, clone: UCHT1, dilution: 1:300), anti-human CD8
TCR transgenic CD8+ T cells from spleen were magnetically isolated by (BD, cat: 561952, clone: RPA-T8, dilution: 1:300). anti-human FOXP3
CD8 selection according to the manufacturer’s instructions (Miltenyi). (Abcam, cat: ab20034, dilution: 1:300), anti-human KLRB1 (Biosource,
Purified CD8+ T cells were then transferred intravenously at 106 cells per cat: MBS2013141, dilution: 1:300), anti-human CD40LG (Biosource,
mouse. Recipient mice were sub-lethally irradiated (5 Gy, Gammacell cat: MBS4381431, dilution: 1:300), anti-human GZMA (Abcam, cat:
1000) at 24 h prior to the transfer and received AAV-expressing cog- ab10870, dilution:1:300), anti-mouse aSMA (Sigma, cat: C6198, clone:
nate antigen hPMEL (106 pfu) at 30 min after adoptive T cell transfer. 1A4, dilution: 1:500), anti-mouse pSTAT1 (CST, cat: 9167, clone:58D6,
Whole-mount immunofluorescent staining of tail skin epidermis and dilution: 1:500), anti-mouse CD31 (eBioscience, cat: 25-0311-81, clone:
FACS analysis of both tail skin and spleen CD8+ T cells were performed 390, dilution: 1:500). anti-mouse PDGFRA (R&D, cat: AF-307, clone:
at day 26 after adoptive transfer. Mice that were sub-lethally irradiated APA5, dilution: 1:300) and anti-mouse CD45 (BD, cat: 559864, clone:
but did not receive any treatment or received only AVV-hPMEL or Pmel 10-F11, dilution: 1:300).
CD8+ T cell transfer were used as controls. For whole-mount staining, hair shafts that would interfere with
We used three different vitiligo induction models to validate the immunofluorescent imaging process were first removed using Nair
functional importance of fibroblasts. It should be pointed out here treatment. Then the entire tail skin was collected and flattened. Only
that there are some key differences in these models. In the melanoma– the middle one-third section along the long axis of tail skin was used
Treg-induced vitiligo mouse model, Treg cells were initially depleted, for whole-mount staining for the purpose of consistency. Flattened
whereas in the adoptive-transfer-based vitiligo model, the number of skins were cut into 1 × 1-cm squares and placed in 20 mM EDTA solution
functional CCR5+ Treg cells was reported to be increased during vitiligo and incubated at 37 °C with shaking at 80 rpm for 1 h. The epidermis
development60. Future studies using mouse models about the function was separated from the dermis with a fine-tipped tweezer. As hair fol-
of Treg cells and other cell types in driving vitiligo initiation and progres- licles obscure the view of epidermis during whole-mount analysis,
sion should take these differences into consideration. to leave hair follicles in the dermis, the epidermis was peeled from
the dermis along the posterior–anterior direction in a switch motion.
Administration of CD4 and CD8 depletion antibodies Occasional hair follicles still attached to the epidermis were removed
CD4 antibody for cell depletion was purchased from BioXcell. CD8 using a tweezer. The epidermis was then flattened and fixed for 10 min in
antibody for cell depletion was developed by the laboratory of J.S. 4% paraformaldehyde in PBS. Whole-mount skin samples were washed
Anti-CD4 (BioXcell, clone: GK1.5) and anti-CD8 (clone: 2.43) were admin- in PBS overnight and then permeabilized for 20 min in 0.3% H2O2 in
istered by intraperitoneal injection in doses of 10 μg per g mouse body methanol at −20 °C and then blocked for 1 h in a solution of 2% normal
weight. More than 99% depletion of target populations was confirmed donkey serum, 1% BSA and 0.3% Triton in PBS at room temperature.
by FACS. CD4 antibody was administered on day 4 and day 10 after The following antibodies were used: anti-mouse DCT (made by the
B16F10 inoculation. CD8 antibody was administered every fourth day T.C. laboratory; 1:500), anti-mouse CD8a (eBioscience, cat: 17-0081-
after the tumour was removed. 83, clone: 53-6.7, dilution: 1:300), anti-mouse Ki67 (eBioscience, cat:
14-5698-82, clone: SolA15, dilution: 1:500), anti-mouse CD4 (Biolegend, and 1% antibiotic–antimyotic. Two to three days after seeding, the pri-
cat: 100451, dilution: 1:300) and anti-mouse CD11c (Biolegend, cat: mary newborn mouse fibroblasts were infected with lentivirus containing
117316, clone: N418, dilution: 1:300). H2BRFP. At 48 h after infection, the RFP-labelled fibroblasts were then
Whole-mount and section samples were imaged on a Nikon A1-R injected into the tail skin dermis of 8-week-old adult female wild-type
confocal microscope. Images were acquired using a 20 × 0.75 objective or Ifngr1 KO mice as indicated. A total of 3 × 106 fibroblasts at a concen-
lens for representative pictures and a 10 × 0.5 objective lens for quanti- tration of 105 per μl were intradermally injected into three sites per tail
tative pictures. Z-stacks were acquired at a resolution of 1,024 × 1,024 (10 μl per site, 1 cm between sites). Note that the injection procedure can
or 512 × 512. Microscopy data were analysed using Imaris software with only deliver the fibroblasts into lower dermis as indicated by our section
the 3D visualization module (Bitplane). RBG images were assembled staining analysis. Mice were allowed to recover for 3–7 days before vitiligo
and were labelled panel information with Adobe Illustrator CS6. induction. Whole-mount analysis of CD8+ T cells was performed at day
To describe the distribution and density of CD8+ T cells and melano- 33 after vitiligo induction. The density of T cells was analysed by Imaris.
cytes, fluorescent signals of T cells and melanocytes were converted The enrichment score of CD8+ T cells used in Fig. 4h was calculated by
to digital signals using the ‘spot’ function in Imaris software. Position T cell density in the RFP+ region dividing T cell density in RFP− region.
coordinates of each spot were analysed with the ‘smoothScatter’ pack-
age in R to quantify the cell distribution and density. To quantify the FACS analysis
number of RFP+ fibroblasts, melanocytes and CD8+ T cells in each scale For FACS quantification and collection of epidermal immune cells
of tail skin, fluorescent signals of these cells were converted to digital including CD8+ T cells, CD4+ T cells, Langerhans cells and γδ T cells,
signals using the ‘spot’ function in Imaris software, then cell number skin from the middle section of the tail was collected, flattened, cut into
in each scale was quantified using Python. 1 × 1-cm pieces and then floated dermis-side down in 2 ml of 2.4 U ml−1
dispase solution and incubated at 37 °C with shaking at 80 rpm for
Lentivirus infection 50 min. To obtain most of the epithelial cells, the epidermis was then
High-titre lentivirus production was described before61: calcium phos- carefully removed from dermis along the anterior–posterior direc-
phate transfection of 293FT cells was carried out in a 500-cm2 dish with tion with a fine-tipped tweezer. The removed epidermis was floated
pLKO.1 and helper plasmids pMD2.G and psPAX2. Viral supernatant dermis-side down in 3 ml TrypLE Express Enzyme (Gibco) at 37 °C for
was collected 46–48 h after transfection, filtered through a 0.45-μm 10 min, and afterwards 5% FBS was added to neutralize TrypLE.
filter and then concentrated. The final titre was determined by FACS For FACS quantification of epidermal melanocyte, melanocytes
quantification of virus-infected 3T3 cells. located in the hair follicle must be removed. We found out, before skin
For knockdown of Cxcl9, Cxcl10, Ifngr1, Jak1 and Stat1, shRNA len- collection, that waxing using VEET wax paper facilitates subsequent hair
tivirus constructs were obtained from Sigma RNAi Consortium (TRC) follicle separation from epidermis. Waxed tail skin was flattened and
mouse lentivirus library. shRNA was then subcloned into LV-H2BRFP. To floated dermis-side down in 2 ml of 2.4 U ml−1 dispase solution at 37 °C
test knockdown efficiency in vitro, lentivirus was produced by calcium with shaking at 80 rpm for 50 min. To efficiently obtain hair-follicle-free
phosphate transfection of 293FT cells in a six-well plate, followed by epidermis, the epidermis was swiftly peeled from the dermis along the
lentivirus infection of 3T3 cells by centrifugation at 1,000g for 30 min posterior–anterior direction with a fine-tipped tweezer. The occasional
at 37 °C with 10 mg ml−1 polybrene. The knockdown efficiency was remaining hair follicles attached to epidermis were removed using
determined by qPCR. Target sequences of individual shRNA used in tweezers. The hair-follicle-free epidermis was then floated dermis-side
experiments are listed here: down in 3 ml Trypsin-EDTA (0.25%) at 37 °C for 10 min, and afterwards
Ifngr1-shRNA-1: CCACATAGAATATCAGACTTA; 5% FBS was added to neutralize trypsin.
Ifngr1-shRNA-2: GCCAGAGTTAAAGCTAAGGTT; Single-cell suspensions were obtained by repeatedly aspirating and
Jak1-shRNA-1: GCCCTGAGTTACTTGGAAGAT; dispensing with a pipette 20 times followed by filtering through 40-μm
Jak1-shRNA-2: CGGTCCAATCTGCACAGAATA; strainers. The cells were then stained with anti-mouse CD117 (eBiosci-
Stat1-shRNA-1: GCCGAGAACATACCAGAGAAT; ence, cat: 13-1171-82, clone: 2B8, dilution: 1:200) for melanocyte detec-
Stat1-shRNA-2: GCTCACTCAGAACACTCTGAT; tion, anti-mouse CD45 (BD, cat: 559864, clone: 10-F11, dilution: 1:300)
Cxcl9-shRNA-1: CTACACTGAAGAACGGAGAT; for immune cell detection; anti-mouse CD3 (Biolegend, cat: 100220,
Cxcl9-shRNA-2: TCGTCGTTCAAGGAAGACTA; clone: 17A2, dilution: 1:300) and anti-mouse CD8a (eBioscience, cat:
Cxcl10-shRNA-1: CATTGTATATGGAAGAACTT; 17-0081-83, clone: 53-6.7, dilution: 1:300) for CD8+ T cell detection;
Cxcl10-shRNA-2: ATGTCTGAATCCGGAATCTA; anti-mouse CD3 and anti-mouse CD4 (Biolegend, cat: 100451, dilution:
For in vivo knockdown of target genes in dermis, 10 μl high-titre 1:300) for CD4+ T cell detection; anti-mouse MHCII (Biolegend, cat:
lentivirus (>5 × 108 cfu ml−1) was intradermally injected using insulin 107629, dilution: 1:300) and anti-mouse CD207 (Biolegend, cat: 144206,
syringes into P1 mice tail skin. Vitiligo induction began at 8–9 weeks dilution: 1:300) for Langerhans cell detection; and anti-mouse CD3 and
after birth. Samples were collected to perform whole-mount staining anti-mouse TCRγδ (Biolegend, cat: 118108, dilution: 1:300) antibodies
at days 26–33 after vitiligo induction. for γδ T cell detection.
For FACS quantification and collection of dermal fibroblasts,
Fibroblast transplantation assay endothelial cells and immune cells including macrophage and dendritic
To isolate primary mouse fibroblasts, skin from the indicated region of cells from the tail skin, skin from the middle section of the tail was col-
newborn female mice was collected, flattened and placed dermis-side lected, flattened, cut into 1 × 1-cm pieces, and then floated dermis-side
down in 6 ml of 2.4 U ml−1 dispase solution and incubated at 37 °C with down in 2 ml of 2.4 U ml−1 dispase solution and incubated at 37 °C with
shaking at 80 rpm for 60 min. The epidermis along with most of the hair shaking at 80 rpm for 50 min. To get rid of epidermis and hair follicles,
follicles were removed from dermis. The dermis was placed in 10 ml the epidermis was then carefully removed from dermis along the ante-
of 2 mg ml−1 collagenase (Sigma-Aldrich, C2674) at 37 °C with shaking rior–posterior direction with a fine-tipped tweezer. The dermis was
at 80 rpm for 1 h, and afterwards 10 ml of 5% FBS medium was added. then placed in 10 ml of 2 mg ml−1 collagenase (Sigma-Aldrich, C2674)
Single-cell suspensions were obtained by repeatedly aspirating and and incubated at 37 °C with shaking at 80 rpm for 1 h; afterwards 5%
dispensing with pipette 10 times. The cells were then filtered through FBS solution was used for neutralization. Single-cell suspensions were
two strainers (70 μm followed by 40 μm). obtained by repeatedly aspirating and dispensing with a pipette for 20
The isolated primary fibroblasts were cultured in DMEM (Gibco) times followed by filtering through 70-μm and 40-μm strainers. The
medium supplemented with 10% (v/v) FBS (Gibco), 1% (v/v) pen–strep, cells were stained with anti-mouse CD31 (eBioscience, cat: 25-0311-81,
Article
clone: 390, dilution: 1:300), anti-mouse-CD45, and anti-mouse PDGFRA
(eBioscience, cat: 17-1401-81, clone: APA5, dilution: 1:300) antibodies In vitro transwell T cell migration assay
for endothelial cell, total immune cell, and fibroblast detection, respec- Transwell migration of lymphocytes was performed with mature cyto-
tively; anti-mouse/human CD11b (Biolegend, cat: 101215, dilution: toxic T lymphocytes (CTLs) and concentrated fibroblast-conditioned
1:300) and anti-mouse F4/80 (Biolegend, cat: 123116, dilution: 1:300) for medium. In brief, splenocytes isolated from OT-1 mice were stimulated
macrophage detection; and anti-mouse MHC-II and anti-mouse CD11c with OVA257-264 for 2 days in the presence of 10 ng ml−1 IL-2. Cells were
(Biolegend, cat: 117310, dilution: 1:300) for dendritic cell detection. centrifuged and cultured in fresh medium containing 10 ng ml−1 IL-2 for
For analysis and isolation of immune cells from spleens, the spleen two more days, after which most of the cells in the culture were CTLs.
was mechanically dissociated and the red blood cells were lysed with To acquire fibroblast-conditioned medium, primary fibroblasts with
RBC lysis buffer according to the manufacturer’s instructions (Beyo- the indicated genotypes or regions were cultured with DMEM with or
time). Single-cell suspensions were obtained by filtering with strainers without 1,000 U ml−1 IFNγ for 6 h at 37 °C. The medium was then con-
(70 μm and 40 μm) and then cells were stained with anti-mouse CD3, centrated (1×, 2×, 5×, 10× or 25×) for chemo-attractants. In the transwell
anti-mouse CD45 and anti-mouse CD8 antibodies. Anti-mouse VB13 cell migration assay, 96-well 5-μm inserts from Corning were used.
(Biolegend, cat: 140703, clone: MR12-4, dilution:1:300) antibody was Migration medium (200 μl) containing either fibroblast-conditioned
used in Pmel T cell adoptive transfer experiments. medium with the indicated concentrations or control medium were
For CXCR3 expression analysis in skin immune cell types of viti- loaded into the lower chamber. CTLs (1.5 × 105) suspended in 50 μl
ligo mice, the entire tail skin was collected, flattened, then floated medium were then placed in the upper chamber. At 3 h after incubation
dermis-side down in 5 ml trypsin-EDTA (0.25%) at 37 °C for 30 min. at 37 °C under 5% CO2 conditions, the migrated and unmigrated T cells
The skin was inverted and scraped with a scalpel and the cell slurry was were collected from the lower and upper chambers, respectively. To
filtered with strainers (70 μm and 40 μm), then cells were co-stained normalize samples, beads of known concentration were added to all
with anti-mouse CXCR3 (Biolegend, cat: 126506, dilution: 1:300) against samples before counting by FACS. The migration ratio was determined
the immune cell markers mentioned above for further analysis. by dividing the number of migrated cells by the number of migrated
For FACS analysis of CD8+ T cells and melanocytes from mouse dorsal, plus unmigrated cells.
ventral, paw dorsal and paw ventral skin, skin biopsies were collected, To test the ability of secreted chemokines to promote CD8+ T cell
flattened and floated dermis-side down in 3 ml of TrypLE and incubated migration, CXCL2, CXCL3, B2M, CXCL10 and CXCL9 at concentrations
at 37 °C with shaking at 80 rpm for 30 min. The skin epidermis was gen- of 0, 0.2, 0.4, 1 and 2 μg ml−1 were used.
tly scraped with a scalpel and the cell slurry was filtered with strainers
(40 μm), then cells were stained with anti-mouse CD45, CD3, CD8 and RNA isolation and qPCR
CD117 antibodies for further analysis. Fibroblast total RNA was isolated from FACS-sorted cells with TRIzol
For re-isolation of injected RFP-labelled dorsal or paw fibroblasts (Thermo Fisher Scientific) followed by extraction using the Direct-Zol
from Ifngr1 KO mouse tail, tail skin was separated into epidermis and RNA Miniprep Kit (Zymo Research). CD8+ T cell total RNA was isolated
dermis after dispase treatment. After digestion of dermis with colla- from FACS-sorted cells using the Picopure RNA Isolation Kit (Thermo
genase and neutralization, single-cell suspensions were obtained by Fisher Scientific) and RiboAmp HS PLUS RNA Amplification Kit (Thermo
filtering through 70-μm and 40-μm strainers. Then RFP+ fibroblasts Fisher Scientific). For cDNA synthesis, equal amounts of RNA were
were isolated for further qPCR analysis. reverse-transcribed using Oligo-dT (Vazyme, R222-01). Expression
Antibodies described above were incubated on ice for 15–30 min. levels were normalized to the expression of PPIB or GAPDH. qPCR was
DAPI was used to exclude dead cells. Cell analysis and isolation were conducted using a CFX96TM Real-Time system (Bio-Rad) with Power
performed on a BD FACSAria sorter equipped with FACSDiva software SYBRR Green PCR Master Mix (Life Technologies). All primer pairs
(BD Bioscience). Data were analysed with FlowJo software (FlowJo LLC). were designed for the same cycling conditions: 10 min at 95 °C for ini-
tial denaturation, 40 cycles of 10 s at 95 °C for denaturation, 30 s at
IFNγ treatment of fibroblasts in vitro 62 °C for annealing, and 10 s at 65 °C for extension. The primers were
For isolation of human dermal fibroblasts, skin epidermis and der- designed to produce a product spanning an exon–intron boundary in
mis of biopsy were mechanically separated after 2.4 U ml−1 dispase each of the target genes.
treatment for 1 h at 37 °C with shaking at 80 rpm. Further digestion of
the dermis was performed with 10 mg ml−1 collagenase at 37 °C with Single-cell RNA library construction and sequencing
shaking at 80 rpm for 1 h. The fibroblasts were then expanded and Single-cell cDNA libraries were prepared using the Chromium Single
cultured in DMEM supplemented with 10% FBS, 1% pen–strep and 1% Cell 3′ Library and Gel Bead Kit v2 according to the manufacturer’s
antibiotic–antimycotic. Passage-3 or -4 fibroblasts were treated with instructions. In brief, cell suspensions in a chip were loaded on a Chro-
1 U ml−1 recombinant human IFNγ for 3 h followed by RNA-seq. For mium Controller (10x Genomics) to generate single-cell GEMs (gel
each region, the average FPKM value of two individual samples was beads in emulsion). scRNA-seq libraries were then prepared using the
used to estimate the gene expression level. Genes were included in Chromium Single Cell 3′Gel Bead and Library Kit (P/N 120236, 120237,
the heat map if they were differentially expressed in at least one of the 120262; 10x Genomics). Qualitative analysis of each DNA library
eight anatomic regions after IFNγ treatment (log2-transformed fold was performed using an Agilent 2100 Bioanalyzer. The concentra-
change > 1 and P < 0.01). tion of each DNA library was measured using a Qubit fluorometer
For isolation of mouse fibroblasts, skin from different regions was (Invitrogen). Libraries were sequenced on an Illumina NextSeq 500
taken from C57 newborn mice. After enzymatic digestion, the single-cell (2 × 150 paired-end reads).
suspension was stained with anti-mouse PDGFRA. PDGFRA+ fibroblasts
were isolated by FACS and cultured in DMEM supplemented with 10% Processing of single-cell sequencing data
FBS, 1% pen–strep and 1% antibiotic–antimycotic. At 72 h after seeding, The raw sequence reads were aligned and quantified using Cell Ranger
primary mouse fibroblasts were treated with 5 U ml−1 recombinant (v.1.3.1) software, which was obtained from 10x Genomics (https://
mouse IFNγ for 3 h and collected for RNA isolation and qPCR. support.10xgenomics.com/single-cell-gene-expression/software/pipe
For 3T3 cells, shRNA-containing lentivirus-infected cells were seeded lines/latest/installation). The human hg38 assembly reference was
in a 6-well plate 24 h before IFNγ treatment. After treatment with 5 U ml−1 used for analysis. The raw count matrix data were imported into
recombinant mouse IFNγ for 3 h, cells were collected for RNA isolation R (3.5.1) and R studio (1.1.456) using the Seurat (v.2.3.2) package for
and qPCR. further data analysis. For each of the 15 samples, we initially set up a
first filter of min.cells = 3 and min.genes = 200 per sample. Cells with to obtain FPKM. Differential expression analysis was performed using
more than 5,000 genes or more than 25,000 unique molecular identi- the DESeq2 package. Genes with significantly upregulated expres-
fiers (UMIs) were removed. We kept cells with less than 1% mitochon- sion (P < 0.01, fold change > 2) were chosen for further analysis. Heat
drial gene expression. The raw counts were normalized by a factor of maps showing differentially expressed genes were generated using
10,000 and log-transformed to obtain log(T+ 1) values. Variable genes the ‘pheatmap’ and ‘ggrepel’ packages in R.
were identified by fitting the mean-variance relationship and met the
following criteria: 0.0125 < mean of non-zero values < 3 AND standard Mathematical model
deviation > 0.4. We developed a mathematical model to predict T cell cluster expan-
sion and vitiligo progression. The skin tissue was modelled as a square
Cell-type classification using t-SNE lattice with two layers. The upper epidermal layer contains T cells and
Unsupervised clustering of cells was performed with Seurat. Dimen- melanocytes. The lower dermal layer contains fibroblasts with different
sionality reduction was performed using principal component analysis chemotactic effects. The state of each cell changes over time according
(PCA). The first 20 principal components were selected according to to the state of itself, its neighbour cells and the surrounding signals.
the PCA elbow plot and used for clustering with resolution parameter On the upper layer, each lattice site may contain one melanocyte or
0.1. Cell clusters were visualized using t-SNE plots, with all significant one T cell, or it may be empty. At first the melanocytes are uniformly dis-
principal components as input. We integrated data from all samples tributed, with an average spacing of four lattice sites. The melanocyte
using canonical correlation analysis. The shared nearest neighbour does not move, proliferate or die on its own. Only if the surrounding
graph was constructed on a cell-to-cell distance matrix from the top 30 T cells exceed a threshold number will the melanocyte be killed and the
aligned canonical correlation vectors. The shared nearest neighbour lattice site it occupied become free. The T cell occupation at each lattice
graph with different resolution was used as input for the smart local site is controlled by an independent birth–death stochastic process.
moving algorithm to obtain cell clusters, and visualized with t-SNE The death rate is constant. The birth rate depends on the concentration
or uniform manifold approximation and projection (UMAP). Cluster of the T cell-recruiting chemokine produced by fibroblasts on the lower
marker genes were identified using the FindAllMarkers function by layer. In addition, the total birth rate of T cells in the system is limited.
Wilcoxon rank-sum test with the following parameters:min. pct = 0.25, On the lower layer, every lattice site contains one fibroblast. The
thresh.use = 1. On the basis of previous knowledge and consistency fibroblast can respond to its nearby activated T cells in the upper
within the different resolutions, we selected the final number of clus- layer and release chemokine. If there is no activated T cell nearby, the
ters that included all the major cell types in the skin, resulting in eight fibroblasts stay in a quiescent state in which they do not produce any
different clusters. chemokine. By default, all the T cells are inactivated. Only when a mel-
anocyte is killed will the nearby T cells be activated, which in turn will
Profiling differentially expressed genes within each cell cluster induce the fibroblast to release chemokine. The production rate of
To identify differentially expressed genes in two melanocyte subtypes, chemokine per fibroblast is λ. The chemokine diffuses on the lattice
we applied the FindMarkers function from Seurat to the normalized with a rate-constant D and degrades at rate γ. The model details are
gene expression data, with the following parameters: min.pct > 0.25 stated in Algorithm 1.
and thresh.use = 0.25. The highly expressed genes in the M1 melanocyte At first all fibroblasts are inactive and there is no chemokine in the
cluster were identified as C1 signature genes (fold change > 2, P < 0.01). system. Progressive melanocyte death is a consequence of two events.
GO analysis of M1 signature genes was performed using the GO web The first event is the randomly local accumulation of T cells near one
service (http://geneontology.org). For comparing gene expression melanocyte. If the number of T cells near a melanocyte exceeds the
between four T cell clusters, the differentially expressed genes were threshold, the melanocyte will be killed. This melanocyte-killing event
selected using the threshold fold change > 0.25, P < 0.01. The gene will activate the nearby T cells, which will in turn activate the nearby
expression levels shown in the various charts in the manuscript were fibroblasts beneath them in the lower layer. These fibroblasts will
plotted using packages in R. release chemokine to recruit more T cells to this location. As a result,
the local density of T cells will increase. If the local density of T cells near
RNA sequencing, alignment, analysis and visualization a melanocyte exceeds the threshold, subsequent melanocyte-killing
RNA from FACS-purified cells was submitted to Novogene for quanti- events are to be expected. This process constitutes a positive feedback
fication and RNA-seq library preparation and sequencing. The library mechanism that leads to the extermination of melanocytes in a spa-
was sequenced on an Illumina HiSeq platform using the paired-end tially progressive manner. The melanocyte-free region continues to
150-bp sequencing strategy. expand as more T cells concentrate at the border of the T cell cluster.
For RNA-seq analysis of mouse tail fibroblasts, raw RNA-seq data However, in the centre of the T cell cluster, after the activated T cells
were mapped to the mouse genome (GRCm38/mm10) using TopHat undergo apoptosis, the fibroblast will become quiescent and hence
(v.2.0.13) with default settings to produce a reference-guided transcript the number of T cells will drop.
assembly. Cufflinks (v.2.2.1) was used to normalize expression levels
for each sample to obtain FPKM. Algorithm 1 (detailed model implementation)
Cuffdiff was used to quantify changes in gene expression between Initialization.
the control wild-type (WT) fibroblasts, vitiligo WT fibroblasts, and 1. Set chemokine concentration φ(x, t) = 0.
vitiligo Ifngr1 KO fibroblasts. Genes with significantly upregulated 2. Generate melanocytes:
expression levels (P < 0.01, fold change of vitiligo WT/naive WT > 1.5, (a).  Seed melanocytes uniformly on the lattice with an interval of four
vitiligo WT/vitiligo KO > 1.3) were chosen for further analysis. GO analy- lattice sites.
sis of upregulated genes was performed using a GO web service (http:// (b).  Randomly perturb the location of each melanocyte.
geneontology.org). Differentially expressed gene data were presented Repeat until t > Tend.
using the ‘ggplot2’ package in R software. 1. Compute T cell arriving rate a(x, t):
For in vitro IFNγ-treated human fibroblast RNA-seq analysis, raw (a).  a(x, t) = α(φtiny + φ(x, t)).
transcriptome sequence data were mapped to the human genome (b).  If ∑ x a(x, t) > Amax , then a(x, t) = Amax a(x, t)/ ∑ x a(x, t).
(hg38) using STAR (v.2.6.1a) with default settings to produce a 2. For all lattice sites x on the upper layer:
reference-guided transcript assembly. FeatureCount (v.2.2.1) was used (a).  If x is empty, add a T cell (inactive state) with probability a(x, t)dt.
to count reads of genes and normalize expression levels for each sample (b).  If x is occupied by a T cell, remove it with probability βdt.
Article
(c).  If x is occupied by a melanocyte, compute n(x), the number of T cells is greater than nc, the melanocyte will be killed. When this happens,
within a local patch of size Pc × Pc centred at x. If n(x) > nc, remove this its nearby T cells will be activated.
melanocyte and activate all the n(x) T cells. Remark: The choice of Pc = 5 is based on the estimation that the size
3. For all lattice sites x on the lower layer: of a melanocyte is approximately 100 μm. The choice of nc = 15 is to fit
(a).  If there exists an activated T cell within a local patch of size 3 × 3 the experimental data. A larger nc makes the initial melanocyte-killing
centred at x in the upper layer, activate the fibroblast at x. Otherwise event less likely to occur (a longer waiting time and fewer incidents).
set the fibroblast to be inactive.
(b).  Update φ(x, t) according to φ(x, t) = φ(x, t) + ∆φ(x, t)dt + λI (x) Statistics and reproducibility
dt − γφ(x, t). Statistical analyses were performed with GraphPad Prism (v.8.00),
Here ∆ is the discretized Laplacian operator on the lattice. R (v.3.5.1), and RStudio (v.1.1.456). For the representative images and
FACS profiles presented in the paper, each experiment was repeated
 λ , if the fibroblast at x is active
λ I (x) =  at least three times independently with similar results. No statistical
0, otherwise methods were used to predetermine sample size.
4. Set t = t + dt. Figure 1: e, n = 5 individuals for each disease/healthy state. Median,
box: 25/75 percentile, whisker: 5/95 percentile. f, n = 5 individuals for each
Variables. φ(x, t): chemokine concentration at lattice site x at time t. disease/healthy state. Two-tailed unpaired t-test. **P < 0.01, NS, not sig-
a(x, t): T cell arriving rate at lattice site x at time t. nificant. Data are mean ± s.d. h, n ≥ 3 for each disease/healthy state. Data
are mean ± s.d. Figure 2: b, d, n ≥ 4 independent mice for each condition.
Parameters. System settings: Two-tailed unpaired t-test. ***P < 0.001, ****P < 0.0001. e, n = 9 independ-
M × N: system size. The size of the lattice is M × N. M × N = 100 × 100 ent host mice for each condition. Two-tailed paired t-test. ***P < 0.001,
in Extended Data Fig. 10a, b. M × N = 512 × 512 in Extended Data Fig. 10c. NS, not significant. f, n ≥ 3 independent mice. All data are mean ± s.d.
M × N = 400 × 1,024 in Fig. 4j and Extended Data Fig. 10d. The unit of Figure 3: b, n ≥ 150 scales from 3 independent mice for each condition.
length (length of one lattice site) is 20 μm. Correlation test measured by Pearson coefficient r and two-tailed P-value
Tend: total simulation time. Tend = 2,000. The unit of time is 1 h. analysis. c, n = 3 independent mice for each condition. Data are mean ± s.d.
dt: time step. A discrete time step dt = 0.005 (hour) is used through- d, n = 3 independent experiments with at least 2 replicates per experiment.
out our simulation. Data are mean ± s.d. f, n = 20 samples, R, and P from a linear regression
T cell propensity: analysis. g, n ≥ 5 samples. Data are mean ± s.d. h, n ≥ 15 scales from at least
1. α: birth rate of T cells per lattice site. α controls the base rate of T cell 3 independent mice for each condition. Median, box: 25/75 percentile,
arrival on the lattice. In our simulation we chose α = 1.2 for the mouse whisker: 5/95 percentile, dot: outlier. Two-tailed unpaired t-test. *P < 0.05,
experiment (elevated T cell amount), and α = 1.1 for the symmetry ***P < 0.001, ****P < 0.0001, NS, not significant. Figure 4: c, n = 3 individuals
experiment (normal T cell amount). with technical triplicates per condition. Data are mean ± s.d. d, Correlation
2. β: death rate of T cells per lattice site. The rate of removal of T cells. test measured by Spearman coefficient. e, Top, n ≥ 50 hair follicles from
For inactive T cells the removal rate corresponds to how fast a T cell at least 3 mice. Median, box: 25/75 percentile, whisker: 5/95 percentile,
leaves the tissue and possibly re-enters the circulation system. For dot: outlier. Bottom, n = 6 mice for each condition. Two-tailed unpaired
active T cells, the removal rate corresponds to its death rate. In prin- t-test. **P < 0.01, ***P < 0.001, ****P < 0.0001, NS, not significant. Data are
ciple the removal rate for the two different cases may be different. mean ± s.d. f, n = 3 independent mice with technical triplicates per condi-
In our simulation we make them equal at β = 0.04 throughout our tion. Data are mean ± s.d. g, n = 3 independent experiments with at least
simulation. 2 replicates per experiment. Two-tailed unpaired t-test. *P < 0.05. Data
3. Amax: total supply of T cells. Controls the total amount of T cells avail- are mean ± s.d. h, n = 16 samples from at least 10 independent mice per
able to the system. We chose Amax = 0.015 × α × N × N throughout our condition. Two-tailed unpaired t-test. ****P < 0.0001. Data are mean ± s.d.
simulation. i, n ≥ 2 independent mice with technical triplicates per condition. Data
Remark: α, β and Amax are difficult to measure experimentally. We are mean ± s.d. k, Each dot represent median. Correlation test measured
chose their values to fit the experimental data. Changes in these param- by Spearman coefficient. Extended Data Fig. 1: m, n ≥ 3 for each disease/
eters mainly affect the speed of progression of the melanocyte-killing healthy state. Data are mean ± s.d. r, n = 6 samples from at least 3 patients
region, but have little effect on the characteristic behaviour of the with vitiligo. Left and middle, two-tailed unpaired t-test. ***P < 0.001. Data
model, such as the shape of the melanocyte-killing region and the way are mean ± s.d. Right, R, and P from a linear regression analysis. Extended
it expands. Data Fig. 2: e, n = 10 independent mice for each condition. Two-tailed
Chemokine production and distribution: unpaired t-test. ****P < 0.0001. f, n = 6 independent mice for each con-
1. D: diffusion constant of the chemokine. D = 2. dition. Two-tailed unpaired t-test. ****P < 0.0001. g, n ≥ 6 independent
2. λ: production rate of chemokine per active fibroblast. λ = 0.04. mice for each condition. Two-tailed unpaired t-test. ****P < 0.0001. h, i,
3. γ: decay rate of chemokine. γ = 0.2. n = 3 independent mice for each condition. Two-tailed unpaired t-test.
4. φtiny: background chemokine level. φtiny = 0.01. A numerical trick to k, Left, n = 5 independent mice for each condition. Right, n = 2 inde-
avoid division by zero (see a(x, t) in Algorithm 1). pendent mice for each condition. All data are mean ± s.d. Extended
Remark: Together, D, λ, and γ control chemokine dynamics. γ = 0.2 Data Fig. 3: f, n = 5 individuals for each disease/healthy state. Two-tailed
gives a half-life period of 3.5 h for the chemokine. The characteristic unpaired t-test. *P < 0.05, NS, not significant. h, i, n ≥ 4 for each condi-
spreading length (the distance over which the steady-state signal from tion. Two-tailed unpaired t-test. *P < 0.05, **P < 0.01, NS, not significant.
a point source falls by a factor of 1 − 1/e, or about 63%)62 of the chemokine All data are mean ± s.d. Extended Data Fig. 4: d, n = 6 independent mice
is D /γ  = 3.16, or 63 μm (1 unit = 20 μm), which is a reasonable value for each condition. Two-tailed unpaired t-test. **P < 0.01, ****P < 0.0001.
for morphogens, growth factors, cytokines and chemokines (less than Data are mean ± s.d. f, o, t, w, n ≥ 4 independent mice for each condition.
100 μm)63. The production rate λ was chosen to be large enough to Two-tailed unpaired t-test. **P < 0.01, ***P < 0.001, NS, not significant. Data
activate the melanocyte-killing event. are mean ± s.d. h, p, x, n ≥ 100 scales from at least 3 independent mice for
Melanocyte properties: each condition. Correlation test measured by Pearson coefficient r and
1. Pc: local patch size. Pc = 5. two-tailed P-value analysis. j, n = 2 independent mice with technical trip-
2. nc: critical number of T cells for the killing of a melanocyte. nc = 15. If licates for each condition. Two-tailed unpaired t-test. ***P < 0.001. Data
the number of T cells in the local Pc × Pc patch centred at a melanocyte are mean ± s.d. r, n ≥ 3 independent mice for each condition. Two-tailed
unpaired t-test. ***P < 0.001. Data are mean ± s.d. s, n ≥ 8 independent mice
for each condition. Data are mean ± s.d. Extended Data Fig. 5: a, b, c, n ≥ 4 Data availability
independent mice for each condition. Two-tailed unpaired t-test. *P < 0.05, The scRNA-seq data, mouse fibroblast RNA-seq data and human fibro-
**P < 0.01, NS, not significant. Data are mean ± s.d. f, n ≥ 2 independent blast RNA-seq data have been deposited in the Genome Sequence
mice for each condition. Data are mean ± s.d. g, n ≥ 6 independent host Archive (GSA) with accession number PRJCA006797. The code for the
mice for each condition. Two-tailed paired t-test. NS, not significant. vitiligo progression model is available at https://github.com/hydrays/
Extended Data Fig. 6: a, n = 2 independent mice with technical triplicates
vitiligo. Source data are provided with this paper.
for each condition. Data are mean ± s.d. c, n ≥ 7 independent mice for each
condition. Two-tailed unpaired t-test. ***P < 0.001, ****P < 0.0001, NS, not
significant. Data are mean ± s.d. e, n ≥ 9 independent mice for each condi- 51. Tang, Y., Harrington, A., Yang, X., Friesel, R. E. & Liaw, L. The contribution of the Tie2+
lineage to primitive and definitive hematopoietic cells. Genesis 48, 563–567 (2010).
tion. Two-tailed unpaired t-test. *P < 0.05, **P < 0.01, NS, not significant.
52. Deng, L. et al. A novel mouse model of inflammatory bowel disease links mammalian
Data are mean ± s.d. Extended Data Fig. 7: b, f, n ≥ 110 scales from 3 inde- target of rapamycin-dependent hyperproliferation of colonic epithelium to
pendent mice for each condition. Correlation test measured by Pearson inflammation-associated tumorigenesis. Am. J. Pathol. 176, 952–967 (2010).
coefficient r and two-tailed p-value analysis. c, d, g, n ≥ 4 independent mice 53. McCubbrey, A. L., Allison, K. C., Lee-Sherick, A. B., Jakubzick, C. V. & Janssen, W. J.
for each condition. Two-tailed unpaired t-test. *P < 0.05, **P < 0.01, NS, not Promoter specificity and efficacy in conditional and inducible transgenic targeting of
lung macrophages. Front. Immunol. 8, 1618 (2017).
significant. Data are mean ± s.d. i, k, n = 3 independent experiments with
54. Le Gal, F. A. et al. Direct evidence to support the role of antigen-specific CD8+ T cells in
at least 2 replicates per experiment. Data are mean ± s.d. Extended Data melanoma-associated vitiligo. J. Invest. Dermatol. 117, 1464–1470 (2001).
Fig. 8: a, n = 3 independent mice for each condition. Data are mean ± s.d. b, 55. Lo, J. A. et al. Epitope spreading toward wild-type melanocyte-lineage antigens rescues
n = 3 biological replicates for each condition. Data are mean ± s.d. c, n ≥ 3 suboptimal immune checkpoint blockade responses. Sci. Transl. Med. 13, eabd8636
(2021).
independent mice with technical triplicates for each condition. Data are
56. Hua, C. et al. Association of vitiligo with tumor response in patients with metastatic
mean ± s.e.m. d, n ≥ 15 scales from at least 3 independent mice for each melanoma treated with pembrolizumab. JAMA Dermatol. 152, 45–51 (2016).
condition. Median, box: 25/75 percentile, whisker: 5/95 percentile, dot: 57. Herzberg, B. & Fisher, D. E. Metastatic melanoma and immunotherapy. Clin. Immunol. 172,
outlier. Two-tailed unpaired t-test. *P < 0.05, **P < 0.01, ***P < 0.001, NS, 105–110 (2016).
not significant. e, n ≥ 4 independent mice for each condition. Two-tailed 58. Nardin, C. et al. Vitiligo under anti-PD1 therapy is associated with increased survival in
melanoma patients. J. Am. Acad. Dermatol. 82, 770–772 (2020).
unpaired t-test. **P < 0.01, NS, not significant. Data are mean ± s.d. f, n = 5
59. Halprin, K. M. Epidermal "turnover time"–a re-examination. Br. J. Dermatol. 86, 14–19
individuals for each disease/healthy state. Median, box: 25/75 percentile, (1972).
whisker: 5/95 percentile. g, n ≥ 3 independent mice for each condition. 60. Gellatly, K. J. et al. scRNA-seq of human vitiligo reveals complex networks of subclinical
Data are mean ± s.d. Extended Data Fig. 9: c, n = 3 individuals with tech- immune activation and a role for CCR5 in Treg function. Sci. Transl. Med. 13, eabd8995
nical triplicates for each condition. Data are mean ± s.e.m. d, n = 2 indi- (2021).
61. Chen, T. et al. An RNA interference screen uncovers a new molecule in stem cell
viduals for each condition. Data are mean ± s.d. e, n = 2 individuals with
self-renewal and long-term regeneration. Nature 485, 104–108 (2012).
technical triplicates for each condition. Data are mean ± s.d. f, Left, n ≥ 5 62. Lander, A. D. Morpheus unbound: reimagining the morphogen gradient. Cell 128,
mice for each condition. Right, n ≥ 3 mice. Two-tailed unpaired t-test. 245–256 (2007).
NS, not significant. Data are mean ± s.d. g, n ≥ 50 hair follicles from at 63. Chen, C. C. et al. Organ-level quorum sensing directs regeneration in hair stem cell
least 3 mice. Median, box: 25/75 percentile, whisker: 5/95 percentile, dot: populations. Cell 161, 277–290 (2015).

outlier. Two-tailed unpaired t-test. ****P < 0.0001, NS, not significant. h,


n ≥ 5 mice for each condition. Two-tailed unpaired t-test. ***P < 0.001, Acknowledgements This work was supported by grants from the National Key R&D Program of
****P < 0.0001, NS, not significant. Data are mean ± s.d. i, n = 3 independent China (2017YFA0103500), the National Basic Research Program of China 973 Programs
(2014CB849602), National Natural Science Foundation of China (11671415) and the Beijing
mice with technical triplicates for each condition. Data are mean ± s.d. j,
Municipal Natural Science Foundation (7172192). We thank the NIBS Animal Facility for their
n = 3 independent experiments with at least 2 replicates per experiment. expert handling and care of mice; the NIBS Biological Resource Centre for FACS; the NIBS
Two-tailed unpaired t-test. Data are mean ± s.d. k, n ≥ 7 scales for each imaging facility for assistance with the confocal microscope experiment; the NIBS sequencing
condition. Two-tailed unpaired t-test. **P < 0.01, ***P < 0.001. Data are centre for sample processing of 10x genomics RNA-seq; V. Hearing for providing the DCT and
mean ± s.d. Extended Data Fig. 10: e, f, n = 3 biological replicates for each TYR peptide sequences for in-house-made antibodies; F. Shao for help with importing Ifngr1
KO mice; and all members of the laboratory of T.C. for discussions and technical support.
condition. Data are mean ± s.d. h, n ≥ 100 scales from at least 3 independ-
ent mice for each condition. Median, box: 25/75 percentile, whisker: 5/95 Author contributions T.C. and Z.X. conceived the project, designed the experiments and wrote
percentile, dot: outlier. Correlation test measured by Spearman coef- the manuscript. Z.X. and D.C. performed most of the in vivo experiments. Y.D. participated in
ficient. i, Each dot represents the median. Correlation test measured by the lentivirus injection experiment. K.J. performed some of the immunofluorescent staining
experiment. Z.X., H.H. and J.W. conducted bioinformatics analysis of single-cell sequencing
Spearman coefficient. 
data. W. Wu collected fibroblasts from different body positions and participated in the cell
culture experiment. D.C., H.H., J.W. and Z.X. conducted the bioinformatics analysis of the
Data reporting. Participants, mice and samples were assigned into RNA-seq data. Y.H. developed the mathematical model. L.Z., W. Wang, Y.Y. and J.C. collected
different experimental groups randomly. In the experiments with samples from patients with vitiligo and healthy donors. S.L., C.L. and J.C. collected clinical
specific genotypes, samples were sorted into groups based on their data from patients with vitiligo and carried out statistical analysis. J.S. provided the CD8
depletion antibody in the CD8+ T cell depletion experiment.
genotypes. Mice were littermates when available or age-matched
and co-housed to reduce additional variables beyond their genotype Competing interests All authors declare no competing interests related to this work.
difference. Investigators were blinded to the genotype of mice during
vitiligo induction and data collection. Additional information
Supplementary information The online version contains supplementary material available at
https://doi.org/10.1038/s41586-021-04221-8.
Reporting summary
Correspondence and requests for materials should be addressed to Jianmin Chang or Ting
Further information on research design is available in the Nature Chen.
Research Reporting Summary linked to this paper. Reprints and permissions information is available at http://www.nature.com/reprints.
Article

Extended Data Fig. 1 | See next page for caption.


Extended Data Fig. 1 | Comprehensive analysis of different cell populations immunofluorescent images (l) and quantification (m) of T cell subtype markers
from patients with vitiligo and healthy donors. a, Representative images of GZMA, ZNF683, CD40LG, and FOXP3 in T cells (CD3+) in the skin of patients
the skin of patients with vitiligo showing characteristic bilateral symmetric with vitiligo. n, GO categories of genes enriched in each cell type from the skin
lesion patterns in different body regions. b, Experimental workflow and of patients in the progressive state compared to healthy donor skin. o, Heat
representative FACS profiles to obtain single cells from human skin biopsies map analysis of 20 IFNγ response genes in eight cell types from the skin of
for single cell RNA-seq. c, Clinical information for all donors with the number patients in the progressive state and healthy donor skin. p, Violin plot showing
of cells sequenced in single-cell analysis. d, t-SNE visualization of all collected the expression of IFNG in T cell subtypes. q, Violin plots showing IFNGR1 and
cells showing eight main cell types with distinct clusters. e, Heat map analysis IFNGR2 expression in different skin cell types. r, Representative
of differentially expressed genes in each cell type. f–h, violin plot (f), feature immunofluorescent staining images, quantification (left and middle), and
plot (g), and dot plot (h) analysis of signature genes for each cell type. regression analysis (right) between density of CD8+ T cell and pSTAT1+ cell in
i, Volcano plot and GO analysis of genes enriched in melanocytes from the M1 the skin of patients with vitiligo. s, Representative immunofluorescent staining
cluster compared to M2 cluster. j, T-SNE projection of T cells from patients with images of pSTAT1 signal in each cell type in the skin of patients with vitiligo.
vitiligo and healthy donors. k, Volcano plot showing genes differentially Scale bars, 50 µm (l, r, s). For exact p values, see Source Data. For statistics,
expressed (>2-fold, t test p < 0.01) in each T cell sub-cluster. l, m, Representative p summary and sample sizes, see Methods.
Article

Extended Data Fig. 2 | See next page for caption.


Extended Data Fig. 2 | Characterization of the melanoma–Treg-induced enrichment of CD8+ T cells after vitiligo induction. f, Representative whole-
vitiligo mouse model. a, Schematic diagram of the melanoma/Treg-induced mount immunofluorescent staining images and quantifications of DCT+
vitiligo mouse model. First the dorsal skin in the right flank of 8–9-week old melanocytes and CD8+ T cells in mouse tail skin epidermis with combined or
C57 mice was inoculated with B16F10 melanoma cells (Day 0), then CD4 individual B16F10 inoculation and CD4 depletion antibody treatment, showing
depletion antibodies were injected on Days 4 and 10. The tumour was surgically the melanoma/Treg-induced vitiligo model requires both B16F10 inoculation
removed on Day 12 to prevent interference of subsequent analysis. and CD4 depletion antibody injection. g, Schematic diagram, representative
b, Representative hair coat images of control and vitiligo induced mice at whole-mount immunofluorescent staining images and quantification of DCT+
Day 300 after the vitiligo induction procedure. At 4 weeks after induction, melanocytes and CD8+ T cells in the skin epidermis of melanoma/Treg-induced
dorsal skin hair follicles close to the B16F10 tumour cell injection and surgical vitiligo model with or without CD8 depletion antibody treatment, showing
removal sites started to show depigmentation as a result of wounding-induced CD8+ T cells are responsible for melanocyte loss in the melanoma/Treg-
new hair growth. Then the depigmented hair follicles expanded and induced vitiligo mouse model. h, i, Representative whole-mount images
eventually rendered the whole dorsal hair coat depigmented at ~Day 300. (h) and FACS profiles (i) with corresponding quantification of the percentage
c, Representative tail skin images of mice at Day 0, 45, 60, 90, 120, 240, 360, of CD8+ T cells that express CD3 or CD11c in mouse epidermis, showing
480, 720 post melanoma/Treg-induced vitiligo induction procedure. majority of the CD8+ cells are CD3+ T cells, but not CD11c+ dendritic cells.
Melanocytes in mouse dorsal skin are located in hair follicles but not in the j, Representative whole-mount immunofluorescent staining images and
epidermis; only in mouse tail skin are melanocytes located in both the hair density plot images of CD8+ T cells and DCT+ melanocytes in tail skin epidermis
follicle and epidermis similar to human skin. Skin epidermis depigmentation is at Day 19, 26, 33 after vitiligo induction procedure. Note the continuously
the defining feature of vitiligo pathology. Therefore, we only used mouse tail expanding CD8+ T cell clusters in skin epidermis. k, Representative whole-
skin for vitiligo analysis. In tail skin, the depigmentation was initially patchy mount immunofluorescent staining images and quantifications of Ki67+, CD3+
and then progressed to eventually cover the entire epidermal surface, which T cells, and DCT+ melanocytes in tail skin epidermis at Day 33 after vitiligo
did not recover even more than a year later. d, Representative whole-mount induction procedure. Enlarged image on the left represents border of lesion
immunofluorescent staining and density plot images of DCT+ melanocytes and skin, the right one represents lesion region. Quantification showed the
CD8+ T cells in mouse tail skin epidermis at Day 0, 19, 26, 33 after the vitiligo proliferation rates of CD8+ T cells at the border versus inside the clusters are
induction procedure. Prior to vitiligo induction, very few if any CD8+ T cells equivalent; and percentage of Ki67+ cells in melanocytes, T cells and
could be detected in epidermis. Starting from Day 19 after the vitiligo keratinocytes showed majority of the proliferating cells in skin epidermis are
induction procedure, CD8+ T cells infiltration and small regions of melanocyte keratinocytes. These data indicated that the continuous expansion of CD8+
loss could be observed. Note, melanocytes loss only occurred in regions where T cell clusters mainly results from skin-infiltrated CD8+ T cells being actively
CD8+ T cells locally aggregated into clusters; as the CD8+ T cells clusters recruited into the border regions. Scale bars, 500 µm (c, d, f, g, j), 100 µm
continuously expanded so did the regions of melanocyte loss correspondingly. (h, k). For exact p values, see Source Data. For statistics, p summary and sample
e, FACS quantifications of CD45+CD3+CD8+ T cells and CD117+ melanocyte in sizes, see Methods.
mice with or without vitiligo induction confirmed the loss of melanocytes and
Article

Extended Data Fig. 3 | The melanoma–Treg-induced vitiligo mouse model present in mouse. h, i, Quantification and comparison of immune cell types
recapitulates hallmarks of human vitiligo. a, UMAP projection of skin between vitiligo (at Day 33) and control mice. Data in f-g showed the
immune cells from patients with vitiligo and healthy donors. We identified 5 predominantly enriched immune cell type is CD8+ T cell in both patients with
major clusters in immune cells, including two adaptive immune cell clusters vitiligo and the melanoma/Treg-induced vitiligo mouse model, whereas the
(CD4+ T cell and CD8+ T cell) and three innate immune cell clusters other immune cell types did not show significant difference. j, Representative
(Langerhans cell, macrophage, dendritic cell 1/2). Other immune cells of very whole-mount immunofluorescent staining images of CD4+ T cells in WT skin,
low abundance in human skin, such as mast cells and γδ T cells, could also be and Tregs in Foxp3 Cre;Ai14 skin, indicating at Day 33 of vitiligo analysis, the CD4+
observed in our single-cell RNA-seq data, but owing to their limited number T cell population have recovered from systematical CD4+ T cell depletion.
these cell types were not analysed individually. b–e, Heat map (b), dot plot k, Heat map analysis of cytotoxicity-related genes expressed in skin CD8+
(c), violin plot (d) and feature plot (e) analysis of signature genes in each T cells of patients with vitiligo and vitiligo-induced mice, indicating that in
immune cell subtype. f, Quantification and comparison of five immune cell molecular level, this melanoma/Treg-induced vitiligo mouse model elicits
types between progressive-state patients with vitiligo and healthy donors by endogenous auto-reactive CD8+ cytotoxic T cells similar to human vitiligo. Ctl:
scRNA-seq analysis. g, Representative FACS profiles analysing immune cell control, Vit: vitiligo. Scale bars, 200 µm (k). For exact p values, see Source Data.
types in mice vitiligo skin, including epidermal TCRγδhigh and TCRγδlow γδ T cell For statistics, p summary and sample sizes, see Methods.
Extended Data Fig. 4 | See next page for caption.
Article
Extended Data Fig. 4 | Analysis of wild-type and Ifngr1 KO mice with three Ifngr1 KO mice with adoptive T cell transfer-based vitiligo mouse model.
different vitiligo induction methods. a–j, Analysis of WT and Ifngr1 KO q, Schematic diagram. r, Quantification of CD117+ melanocytes and
mice with melanoma/Treg-induced vitiligo model. a, Schematic diagram. CD45+CD3+CD8+ T cells in tail skin epidermis of WT mice after γ irradiation,
b, Representative tail skin images of WT and Ifngr1 KO mice at Day 60 after with or without WT Pmel T cell transfer or AVV-hPMEL intraperitoneal
vitiligo induction. c, d, Representative whole-mount images (c) and injection. These data indicate sub-lethal irradiation alone, irradiation with
quantification (d) of tail skin epidermal melanocytes and CD8+ T cells at Day 33 hPMEL-AAV alone, or irradiation with Pmel CD8+ T cell transfer alone did not
after vitiligo induction. e, f, Representative FACS profile of CD117+ result in melanocyte loss or CD8+ T cell infiltration in WT tail skin epidermis.
melanocytes and CD3+CD8+ T cells (e), and quantification of CD117+ s, Representative immunofluorescent staining images and FACS quantification
melanocytes (f) in the tail skin epidermis at Day 33 after vitiligo induction. of epidermal melanocyte of mice post γ-irradiation 26 days with controls,
g, Whole-mount views of scales in tail skin epidermis (dotted line) in control indicating after 5 Gy γ irradiation leads to ~2-fold increase of melanocyte
and vitiligo mice. This is the unit area we used to quantify melanocyte or CD8+ number in tail skin epidermis compared to untreated mice. t, Representative
T cell density in wholemount analysis throughout this paper. h. Scatter plots FACS profiles and quantification of CD3+CD8+VB13+ Pmel T cells in spleen or
and correlation analysis of melanocyte number versus CD8+ T cell number in skin of WT and Ifngr1 KO mice at Day 26 with or without adoptive transfer-based
each scale of WT and Ifngr1 KO tail skin at Day 33 after vitiligo induction. vitiligo model induction. These data showed although both WT and Ifngr1 KO
i, Representative whole-mount images of DCT+ melanocytes in tail skin host mice contained the same number of transferred WT TCR VB13+ Pmel
epidermis of WT and Ifngr1 KO mice at 4 months after vitiligo induction. j, QPCR T cells in the spleen, only WT host mice exhibit robust VB13+ Pmel T cell
analysis of GZMA, GZMB, PRF1, and CCL5 expression in skin CD8+ T cells isolated infiltration in skin, whereas the Ifngr1 KO host mice did not. u–v, Representative
from WT and Ifngr1 KO mice at Day 33 after vitiligo induction. k–p, Analysis of tail skin images (u, Day 60) and whole-mount immunofluorescent images
WT and Ifngr1 KO mice with Pmel transgenic spontaneous vitiligo mouse (v, Day 26) of WT and Ifngr1 KO mice post vitiligo model-induction. Scale bars,
model. k, Schematic diagram. l–m, Representative tail skin images (l, P70) and 500 µm. w, Representative FACS profiles and quantification of epidermal
whole-mount immunofluorescent staining images (m, P42) of Pmel;WT and CD117+ melanocytes in WT and Ifngr1 KO mice at Day 26 post vitiligo model-
Pmel; Ifngr1 KO mice. n, o, Representative FACS profiles (n) and quantification induction. x, Scatter plots and correlation analysis of melanocyte number
(o) of CD117+ melanocytes and CD45+CD3+CD8+ T cells in tail skin epidermis of versus CD8+ T cell number in each scale in WT and Ifngr1 KO mice at Day 26 post
WT, Ifngr1 KO, Pmel;WT and Pmel;Ifngr1 KO mice at P42. p, Scatter plots and vitiligo model-induction. Scale bars, 500 µm (b, c, g, i, l, m, s, u, v). For exact p
correlation analysis of melanocyte number versus CD8+ T cell number in each values, see Source Data. For statistics, p summary and sample sizes,
scale in Pmel;WT and Pmel;Ifngr1 KO tail skin at P42. q–x, Analysis of WT and see Methods.
Extended Data Fig. 5 | IFNγ-responsive skin stromal cells are required for mice rather than the grafted skin. These data indicate the graft infiltrated
vitiligo progression. a–c, Analysis of six main immune cell types in tail skin of CD8+ T cells were derived from the host mice rather than the grafted skin.
WT and Ifngr1 KO mice with different vitiligo induction methods. a, Day 33 after g, Quantification of melanocytes and CD8+ T cells number in grafted skin
melanoma/Treg-induced vitiligo model induction. b, P42 of Pmel;WT and without vitiligo model induction in host mice. Donor skin pairs grafted onto
Pmel;Ifngr1 KO mice. c, Day 26 after adoptive transfer-based vitiligo the same host mouse are linked by lines. h, Representative immunofluorescent
model-induction. d, Timeline of the graft and vitiligo induction assay. staining of the junction region between grafted C57 tail skin and host dorsal
Representative whole-mount immunofluorescent staining images of grafted skin of a membrane-Tomato (mT) transgenic mouse. After full-thickness C57 tail
WT tail skin epidermis on WT host at Day 0 and Day 21 with or without vitiligo skin was grafted onto membrane-tdTomato (mT)-expressing host mice
induction were showed. These data indicate tail skin did not spontaneously (Rosa-mT), in which all host cells are genetically labelled as mT+, host derived
develop vitiligo after the graft procedure alone; only after vitiligo induction in dermal cells indeed invaded into the grafted skin dermis. But neither K14+
the host mice did the grafted skin develop vitiligo as indicated by melanocyte keratinocytes nor DCT+ melanocytes migrated from the host to grafted skin.
loss and large amounts of CD8+ T cells infiltration. e, f, Lineage tracing i, Representative immunofluorescent staining image of pSTAT1 signal in
experiment of CD8+ T cells: e, Schematic diagram WT tail skin graft on immune cell (CD45+), smooth muscle cell (a-SMA+), Langerhans cells
CD4 Cre;mTmG host and representative immunofluorescent staining images of (Langerin+) of grafted Ifngr1 KO tail skin on WT host after vitiligo induction on a
CD8+ T cells in the grafted skin epidermis after vitiligo induction; f, FACS host mouse. Enlarged image on the right represents Langerin+ cells in the
analysis and quantification of spleen cells from CD4Cre;Ai6 mouse to determine epidermis. Scale bars, 50 µm (d, e, h, i). For exact p values, see Source Data. For
GFP labelling efficiency in CD8+ T cells. Cells were pre-gated on CD45+ live statistics, p summary and sample sizes, see Methods.
singlets, indicating that the infiltrated CD8+ T cells were derived from the host
Article

Extended Data Fig. 6 | See next page for caption.


Extended Data Fig. 6 | IFNγ-responsive skin fibroblasts are required for images of melanocytes and CD8+ T cells in control and six cell-type-specific
driving vitiligo pathogenesis. a, Schematic diagram and representative FACS conditional knockout lines at Day 33 after vitiligo induction. c, Quantification
profiles of skin cells isolation. Detailed gating strategies are described in of skin CD8+ T cells in WT and six cell-type-specific knockout lines at Day 33
method section. QPCR analyses of cell-type-specific signature genes in FACS- after vitiligo induction procedure based on wholemount staining. d, e,
isolated populations include KRT14 for keratinocytes, DCT for melanocytes, Representative FACS profiles (d) and quantification (e) of CD117+ epidermal
CD45 for immune cells, PDGFRA for fibroblasts, and CD31 for endothelial cells melanocytes and CD3+CD8+ T cells in control and six cell-type-specific
respectively. QPCR validation of IFNGR1 knockout used FACS-purified conditional knockout lines post vitiligo induction. f, Schematic diagram of
endothelial cells, keratinocytes, immune cells, melanocytes, fibroblasts and melanoma/Treg-induced vitiligo procedure and representative tail skin images
CD8+ T cells from TekCre;IFNGR1 fl/fl, K14 Cre;IFNGR1 fl/fl, Csf1rCre;IFNGR1 fl/fl, (Day 60) of WT and PdgfraCreER ;IFNGR 1fl/fl cKO mice after vitiligo induction. Vit.
TyrCreER;IFNGR1 fl/fl, CD4 Cre;IFNGR1 fl/fl, and Pdgfra CreER;IFNGR1 fl/fl mice, all with WT induc.: Vitiligo induction. Scale bars, 500 µm (b, f). For exact p values, see
mice as controls. b, Representative whole-mount immunofluorescent staining Source Data. For statistics, p summary and sample sizes, see Methods.
Article

Extended Data Fig. 7 | See next page for caption.


Extended Data Fig. 7 | IFNγ-responsive fibroblasts are both necessary and h, Section immunofluorescent analysis of intradermally injected RFP-labelled
sufficient to mediate local recruitment of CD8+ T cells through secreted fibroblast. Note injected RFP+ fibroblasts located in lower dermis.
chemokines. a–c, Analysis of WT and Pdgfra CreER;IFNGR1 fl/fl cKO mice with Pmel i, Quantification of T cell transwell migration assay showing T cells migration
transgenic spontaneous vitiligo mouse model. a, Schematic diagram and ratio using control medium without fibroblast, corresponding to Fig. 3d.
representative tail skin images (P70) and representative wholemount images j, Analysis of human and mouse vitiligo fibroblasts up regulated genes. Left,
(P42) of Pmel;WT and Pmel;Pdgfra CreER;IFNGR1 fl/fl cKO mice. b, Scatter plots and heat map showing differential expressed genes in tail skin fibroblasts from WT
correlation analysis of melanocyte number versus CD8+ T cell number in each control, WT vitiligo-induced, and Ifngr1 KO vitiligo-induced mice. Middle,
scale from Pmel;WT and Pmel;Pdgfra CreER;IFNGR1 fl/fl cKO mice at P42. volcano plot of differentially expressed genes in human fibroblasts from
c, Representative FACS profiles and quantification of tail skin epidermal progressive-state patients with vitiligo compared with healthy donors. Red
CD117+ melanocytes and CD3+CD8+ T cells in WT, PdgfraCreER;IFNGR1 fl/fl cKO, dots denote genes >1.5 fold upregulated (t test p < 0.01) in fibroblasts from
Pmel;WT and Pmel;Pdgfra CreER;IFNGR1 fl/fl cKO mice at P42. d–g, Analysis of WT progressive-state patients with vitiligo. Right, heat map analysis of the 28
and Pdgfra CreER;IFNGR1 fl/fl cKO mice with adoptive transfer-based vitiligo model. common upregulated secreted factors in fibroblasts from patients with vitiligo
d, Representative FACS profiles and quantification of CD3+CD8+VB13+ Pmel and mice. k, Quantification of migration ratio from the T cell transwell
T cells in spleen or tail skin of WT and PdgfraCreER;IFNGR1 fl/fl cKO mice at Day 26 migration assay with various chemokines. l, Representative
with or without adoptive transfer-based vitiligo model induction. e, Schematic immunofluorescent staining images of CXCL9 and CXCL10 signal with T cells
diagram and representative tail skin images (Day 60) and representative (CD3+) in the skin of patients with vitiligo. m, n, Representative
wholemount immunofluorescent staining images (Day 26) of WT and immunofluorescent staining images of CXCL9 (m) and CXCL10 (n) signals in
Pdgfra CreER;IFNGR1 fl/fl cKO mice after vitiligo induction. f, Scatter plots and melanocytes (DCT+), fibroblasts (PDGFRA+), endothelial cells (CD31+), smooth
correlation analysis of melanocyte number versus CD8+ T cell number in each muscle cells (a-SMA+), keratinocytes (K14+), mononuclear phagocytes
scale WT and Pdgfra CreER;IFNGR1 fl/fl cKO mice at Day 26 after vitiligo induction. (CD11c+), Langerhans cells (Langerin+), and T cells (CD3+) in the skin of patients
g, Representative FACS profiles and quantification of tail epidermal CD117+ with vitiligo. Scale bars, 500 µm (a, e, l), 50 µm (h), 100 µm (m, n). For exact p
melanocytes, and quantification of tail epidermal CD117+ melanocytes and values, see Source Data. For statistics, p summary and sample sizes,
CD3+CD8+ T cells of WT and Pdgfra CreER; IFNGR1 fl/fl cKO mice at Day 26. see Methods.
Article

Extended Data Fig. 8 | See next page for caption.


Extended Data Fig. 8 | Fibroblasts directly affect CD8+ T cells through the epidermis with injection of indicated shRNA-expressing lentivirus. Box-
CXCL9/CXCL10–CXCR3 axis in vitiligo. a, Schematic diagram of the whisker plots of epidermal melanocyte number in each scale of mice tail skin
experimental procedure of lentivirus-infected cell type analysis. FACS profiles injected with the indicated shRNAs. e, FACS quantification of immune cell
and quantification showing majority of lentivirus-infected cell population in types in tail skin of WT and PdgfraCreER;IFNGR1 fl/fl cKO mice with three different
the skin is fibroblast. b, QPCR analysis validated high knockdown efficiency of vitiligo induction methods. f, Feature plot showing CXCR3 expression pattern
shRNAs targeting CXCL9/CXCL10 in vitro. c, Top, experimental design of and quantification of CXCR3 in all cell clusters from patients with vitiligo and
in vivo knockdown efficiency detection in different dermal cell types. Bottom, healthy donors. g, Representative FACS profiles and quantification of CXCR3+
qPCR analysis in FACS-isolated virus-infected (RFP+) fibroblasts, endothelial cells composition in vitiligo-induced mice (at Day 33). Scale bars, 500 μm
cells, and immune cells, indicating CXCL9 and CXCL10 expression were (d). For exact p values, see Source Data. For statistics, p summary and sample
specifically silenced in virus-infected fibroblasts, not in other cell types. sizes, see Methods.
d, Representative whole-mount images showing dermis and overlying
Article

Extended Data Fig. 9 | Anatomically distinct dermal fibroblasts have in the dorsal and paw dorsal skin of mice at P60. g, Representative
intrinsic differences in the IFNγ response. a, Heat map of upregulated genes immunofluorescent images, and quantification of hair follicle located
in human dermal fibroblasts from different anatomical regions after IFNγ melanocytes in ventral and paw ventral skin of control mice and vitiligo mice
treatment (> 2-fold, t test p < 0.01). b, HOX expression pattern (based on (at Day 300). h, FACS analysis and quantification of skin CD3+CD8+ T cells and
RNA-seq results) in human dermal fibroblasts from eight different anatomic CD117+ melanocytes from paw dorsal, paw ventral, dorsal, and ventral regions
regions with or without IFNγ treatment during in vitro culture. c, QPCR analysis at Day 300 after vitiligo induction. i, Schematic diagram of fibroblast isolation
of HOXB8, HOXC8, HOXB13, and HOXD11 in human dermal fibroblasts from and qPCR analysis of CXCL9 and CXCL10 in fibroblasts from four distinct
eight anatomic regions. d, JAK1, JAK2, IFNGR1, and STAT1 expression pattern regions of long-term vitiligo mice. j, Quantification of migration ratio using
(based on RNA-seq results) in human dermal fibroblasts from eight different medium without fibroblast, corresponding to Fig. 4g. k, Relationship between
anatomic regions with or without IFNγ treatment. e, QPCR analysis of TNFSF10, melanocyte number and infiltrated CD8+ T cell number in each scale after
IL6, and CTSH in human dermal fibroblasts from eight anatomically distinct vitiligo induction based on quantification data in Extended Data Fig. 4h. Scale
regions after IFNγ treatment. f, Left, representative whole-mount images and bars, 100 µm (f), 50 μm (g). For exact p values, see Source Data. For statistics,
quantification of hair follicle density in the dorsal and paw dorsal skin of mice p summary and sample sizes, see Methods.
at P21. Right, representative section image and quantification of skin thickness
Extended Data Fig. 10 | See next page for caption.
Article
Extended Data Fig. 10 | Mathematical modelling reveals that fibroblasts or two directions (third row, fibroblasts have 1/2 normal chemotactic effect,
direct collective CD8+ T cell local activity. a, The mathematic model also shown in Supplementary Videos 5, 6). c, The mathematic model predicts
developed to predict local CD8+ T cell recruitment and clonal expansion large-scale T cell clone expansion over the long term. Four representative time
behaviour. The model is a 3D square lattice with two layers. The upper points from T0 to T450 show the initial state and the subsequent T cell cluster
epidermal layer contains T cells and melanocytes. The lower dermal layer expansion process over 450 time units. CD8+ T cells in the normal chemotaxis
contains fibroblasts with different chemotactic abilities. The CD8+ T cell model efficiently coordinate so as to achieve clonal expansion and melanocyte
population in skin is considered to be a decentralized system. Each CD8+ T cell clearance. CD8+ T cells in the no chemotaxis model fail to undergo clonal
is equipped with the means of sensing a change in density. Over time, the expansion and melanocyte death is detected. CD8+ T cells in the weak
behaviour of each cell changes according to its state and the states of its chemotaxis model (migration ability decreases to 1/2) generate small T cell
neighbouring cells and the surrounding signals. The collective pattern can be clones and expand slowly. d, The mathematic model predicts the T cell clone
globally modulated by changing the parameters governing local cell-cell distribution pattern under regional variant fibroblasts with different levels of
interactions. Once the T cell surrounding a melanocyte exceeds a threshold chemotactic effect to T cell. In this model, the fibroblasts marked in blue have a
number, melanocyte death and IFNγ secretion occur. The IFNγ signal induce normal chemotactic effect, and those marked in green have a weak
neighbouring fibroblast chemotactic effect to recruit nearby CD8+ T cells, chemotactic effect. In this region, T cell chemotaxis (migration ability)
reaching the local CD8+ T cell density threshold for adjacent melanocyte decrease to 1/2 of normal value. Six representative time points from T0 to T600
cytotoxicity and IFNγ secretion. This positive feedback loop between CD8+ show the initial state and subsequent T cell cluster expansion (also shown in
T cells and fibroblasts could ensure T cell clonal expansion and vitiligo Supplementary Video 7). The results show that the T cell clones are more likely
progression. b, The mathematic model predicts the expansion process of a to expand and generate white patches on the normal chemotactic region. Both
single T cell clone. Five representative time points from T0 to T80 show the white patches and T cell clone patterns are highly correlated with the regional
initial T cell cluster state and the subsequent T cell expansion process over 80 fibroblast variants. e, f, QPCR analysis validated high knockdown efficiency
time units. In model 1 with normal chemotactic fibroblasts, full spectrum of (e) and the effect blocking IFNγ downstream signal (f) in vitro for shRNAs
T cell cluster formation and expansion patterns observed in WT vitiligo mouse targeting IFNGR1, JAK1, or STAT1. g, Representative epidermis whole-mount
model were reproduced (first row, fibroblasts with normal chemotactic effect, immunofluorescent staining images and density plot of shRNA-mediated
also shown in Supplementary Videos 1, 2). In model 2, in which the fibroblasts knockdown assay, relative to the corresponding dermis in Fig. 4k. h, Box-
were incapable of chemotaxis, we obtained patterns observed in whisker plots and correlation analysis of T cell number versus percentage of
Pdgfra CreER ;IFNGR1 fl/fl cKO mice. In this model, although T cells could still infected fibroblasts (upper panels), and melanocyte number versus
randomly aggregate in the epidermis, they failed to propagate this effect and percentage of infected fibroblasts (lower panels) in each scale of in vivo mosaic
recruit more T cells to the initial site (second row, fibroblasts with no fibroblast knockdown experiment. i, Scatter plots of median of melanocyte
chemotactic effect, also shown in Supplementary Videos 3, 4). In model 3, in number versus percentage of infected fibroblasts in each scale of in vivo
which the chemotactic effect of fibroblasts was turned down to 1/2 of the mosaic fibroblast knockdown experiment. Scale bars, 500 µm(g). For exact
normal value, random CD8+ T cell aggregates only recruited a limited number p values, see Source Data. For statistics, p summary and sample sizes,
of T cells, resulting in slow T cell cluster expansion and melanocyte loss in one see Methods.
nature portfolio | reporting summary
Corresponding author(s): Ting Chen
Last updated by author(s): Sep 28, 2021

Reporting Summary
Nature Portfolio wishes to improve the reproducibility of the work that we publish. This form provides structure for consistency and transparency
in reporting. For further information on Nature Portfolio policies, see our Editorial Policies and the Editorial Policy Checklist.

Statistics
For all statistical analyses, confirm that the following items are present in the figure legend, table legend, main text, or Methods section.
n/a Confirmed
The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement
A statement on whether measurements were taken from distinct samples or whether the same sample was measured repeatedly
The statistical test(s) used AND whether they are one- or two-sided
Only common tests should be described solely by name; describe more complex techniques in the Methods section.

A description of all covariates tested


A description of any assumptions or corrections, such as tests of normality and adjustment for multiple comparisons
A full description of the statistical parameters including central tendency (e.g. means) or other basic estimates (e.g. regression coefficient)
AND variation (e.g. standard deviation) or associated estimates of uncertainty (e.g. confidence intervals)

For null hypothesis testing, the test statistic (e.g. F, t, r) with confidence intervals, effect sizes, degrees of freedom and P value noted
Give P values as exact values whenever suitable.

For Bayesian analysis, information on the choice of priors and Markov chain Monte Carlo settings
For hierarchical and complex designs, identification of the appropriate level for tests and full reporting of outcomes
Estimates of effect sizes (e.g. Cohen's d, Pearson's r), indicating how they were calculated
Our web collection on statistics for biologists contains articles on many of the points above.

Software and code


Policy information about availability of computer code
Data collection BD FACSDiva (8.0.1), Nikon-A1R (4.13.00)

Data analysis Imaris (7.4.2), GraphPad Prism (6.0), R (3.5.1), RStudio (1.1.456), GO analysis (http://geneontology.org), FlowJo (v10.0.7), Seurat (V2.3.2),
TopHat (v2.0.13), Cufflinks (v2.2.1), STAR (v 2.6.1a), FeatureCount (v2.2.1). The code for the vitiligo progression model is available at: https://
github.com/hydrays/vitiligo.
For manuscripts utilizing custom algorithms or software that are central to the research but not yet described in published literature, software must be made available to editors and
reviewers. We strongly encourage code deposition in a community repository (e.g. GitHub). See the Nature Portfolio guidelines for submitting code & software for further information.

Data
Policy information about availability of data
All manuscripts must include a data availability statement. This statement should provide the following information, where applicable:
- Accession codes, unique identifiers, or web links for publicly available datasets
March 2021

- A description of any restrictions on data availability


- For clinical datasets or third party data, please ensure that the statement adheres to our policy

The single-cell RNA-sequencing data, mouse fibroblast RNA-seq data, and human fibroblast RNA-seq data have been deposited in the GSA with accession number
PRJCA006797 (https://ngdc.cncb.ac.cn/bioproject/browse/PRJCA006797).

Source data for all figures are provided with the paper.

1
nature portfolio | reporting summary
Field-specific reporting
Please select the one below that is the best fit for your research. If you are not sure, read the appropriate sections before making your selection.
Life sciences Behavioural & social sciences Ecological, evolutionary & environmental sciences
For a reference copy of the document with all sections, see nature.com/documents/nr-reporting-summary-flat.pdf

Life sciences study design


All studies must disclose on these points even when the disclosure is negative.
Sample size No statistical method were used to predetermine sample size. Sample size was determined based on other similarly designed experiments
and results on cellular immunology analyses (Malik et al., 2017, Science Immunology), aiming for 3-10 mice or sample per group.

Data exclusions No data were excluded, with the exception of vitiligo mouse model induction experiment. Mice that fail to developed primary tumors after
intradermally inoculated B16F10 cells were excluded for further analysis. Mice with recurrent primary tumors after surgery were excluded for
further analysis.

Replication For in vivo experiment, biological replicates and independent mice from at least 2 independent litters were used. For in vitro experiments,
biological replicates and technical triplicates were used.

Randomization Participants, mice, and samples were assigned into different experimental groups randomly. In the experiments with specific genotypes,
samples were sorted into groups based on their genotypes. Mice were littermate when available or age matched and co-housed so as to
reduce additional variables beyond their genotype difference.

Blinding Investigator were blinded to the genotype of mice during vitiligo induction and data collection.

Reporting for specific materials, systems and methods


We require information from authors about some types of materials, experimental systems and methods used in many studies. Here, indicate whether each material,
system or method listed is relevant to your study. If you are not sure if a list item applies to your research, read the appropriate section before selecting a response.

Materials & experimental systems Methods


n/a Involved in the study n/a Involved in the study
Antibodies ChIP-seq
Eukaryotic cell lines Flow cytometry
Palaeontology and archaeology MRI-based neuroimaging
Animals and other organisms
Human research participants
Clinical data
Dual use research of concern

Antibodies
Antibodies used anti-human Tyr (Ting Chen Lab made, 1:500)
anti-human KRT14 (Ting Chen Lab made, 1:1000)
anti-human DCT (Ting Chen Lab made, 1:500)
anti-human CD45 (BD, Cat: 11-9459-41, Clone: 2D1, Dilution:1:300)
Anti-human CD117 (BD, Cat 555714, Clone: YB5.B8, Dilution: 1:300)
anti-human pSTAT1 (CST, Cat: 9167, Clone: 58D6, Dilution: 1:500)
anti-human pdgfra (abcam, Cat: ab203491, Clone: EPR22059-270, Dilution: 1:500)
anti-human CD31 (abcam, Cat: ab24590, Clone: P2B1, Dilution: 1:500)
anti-human aSMA (Sigma, Cat: C6198, Clone: 1A4, Dilution: 1:500)
anti-human CD11c (abcam, Cat: ab 52632, Clone: EP1347Y, Dilution: 1:500)
March 2021

anti-human Langerin (abcam, Cat: ab192027, Clone: EPR15863, Dilution: 1:500)


anti-human CD3 (BD, Cat: 563423, Clone: UCHT1, Dilution: 1:300)
anti-human CD8 (BD, Cat: 561952, Clone: RPA-T8, Dilution: 1:300)
anti-mouse DCT, (Ting Chen Lab made, Dilution: 1:500)
anti-mouse pdgfra, (R&D, Cat: AF-307, Clone: APA5, Dilution: 1:300)
anti-mouse pdgfra, (ebioscience, Cat: 17-1401-81, Clone: APA5, Dilution: 1:300)
anti-mouse VB13, (BIolegend, Cat: 140703, Clone: MR12-4, Dilution:1:300 )
anti-mouse CD11c (Biolegend, Cat: 117316, Clone: N418, Dilution: 1:300)
anti-mouse CD45, (BD, Cat: 559864, Clone: 10-F11, Dilution: 1:300)

2
anti-mouse CXCR3, (Biolegend, Cat: 126506, Dilution: 1:300)
anti-mouse CXCR3, (Biolegend, Cat: 126511, Dilution: 1:300)

nature portfolio | reporting summary


anti-mouse MHCII, (Biolegend, Cat: 107629, Dilution: 1:300)
anti-mouse CD4, (Biolegend, Cat: 100451, Dilution: 1:300)
anti-mouse F4/80, (Biolegend, Cat: 123116, Dilution: 1:300)
anti-mouse CD11c, (Biolegend, Cat: 117310, Dilution: 1:300)
anti-mouse/human CD207, (Biolegend, Cat: 144206, Dilution: 1:300)
anti-mouse/human CD11b, (Biolegend, Cat: 101215, Dilution: 1:300)
anti-mouse TCRγδ, (Biolegend, Cat: 118108, Dilution: 1:300)
anti-mouse TCRγδ, (eBioscience, Cat:11-5711-82, Dilution: 1:300)
anti-mouse CD31, (ebioscience, Cat: 25-0311-81, Clone: 390, Dilution: 1:300)
anti-mouse aSMA, (Sigma, Cat: C6198, Clone: 1A4, Dilution: 1:500)
anti-mouse pSTAT1, (CST, Cat: 9167, Clone:58D6, Dilution: 1:500)
anti-mouse KRT14 (Ting Chen Lab made, Dilution: 1:1000)
anti-mouse CD117 (ebioscience, Cat: 13-1171-82, Clone: 2B8, Dilution: 1:200)
anti-mouse CD3 (Biolegend, Cat: 100220, clone: 17A2, Dilution: 1:300)
anti-mouse CD8a (ebioscience, Cat: 17-0081-83, Clone: 53-6.7, Dilution: 1:300)
anti-mouse Ki67 rat (eBioscience, Cat: 14-5698-82, Clone: SolA15, Dilution: 1:500)
Anti-human FOXP3 (abcam, Cat: ab20034, Dilution: 1:300)
Anti-human KLRB1 (Biosource, Cat: MBS2013141, Dilution: 1:300)
Anti-human CD40LG (Biosource, Cat: MBS4381431, Dilution: 1:300)
Anti-human GZMA (abcam Cat: ab10870, Dilution:1:300 )

The signal of human pSTAT1, human CD8a, human pdgfra, human CD11c, human Langerin, human CD3e, human Foxp3, human
KLRB1, human mouse pdgfra, mouse pSTAT1 was amplified by ABC Kit and TSA Kit.

Validation All the commercial antibodies were validated by the suppliers as indicated in the quality assurance literature provided by the supplier
either through flow cytometry or immunofluorescence assays.

The home made antibodies were validated by immunofluorescence assays in this study (Tyr, 1:500) and previous papers (Krt14,
1:1000, Xu et al, eLife, 2015; DCT, 1:500, Lu et al., eLife, 2020).

Eukaryotic cell lines


Policy information about cell lines
Cell line source(s) B16F10 cells (ATCC, CRL-6475) were purchased from ATCC. 293FT cells (Thermo Fisher Scientific, Cat #R70007) were
purchased from Thermo Fisher Scientific.

Authentication All cell lines were kept at low passages in order to maintain their identity.

Mycoplasma contamination All cell lines were tested negative for mycoplasma contamination.

Commonly misidentified lines No commonly misidentified cell lines were used in this work.
(See ICLAC register)

Animals and other organisms


Policy information about studies involving animals; ARRIVE guidelines recommended for reporting animal research
Laboratory animals C57BL/6 mice were purchased from Charles River Laboratories. K14Cre mice (MGI: 1926500) and K14H2BGFP mice (MGI: 5286142)
were kindly provided by Elaine Fuchs. IFNGR1 KO mice (Jackson Stock NO: 003288) and TekCre mice (Jackson Stock NO: 004128)
were kindly provided by Dr. Feng Shao. OT-1 mice (Stock NO: 003831) were kindly provided by Dr. Liang Chen. PMEL TCR
transgenetic mice (Stock NO: 005023) and PdgfraCreER (Stock NO: 018280), Csf1rCre (Stock NO: 021024), TyrCreER (Stock NO:
012328), CD4Cre (Stock NO: 017336), Foxp3Cre (Stock NO: 008694), IFNGR1 flox (Stock NO: 025394), Ai14 (Stock NO: 007914), Ai6
(Stock NO: 007906) and Rosa-stop-mTmG (Stock NO: 007676) mice were obtained from The Jackson Laboratory. Mice were group
housed (up to 5 animals per cage) on a 12:12 hour light-dark cycle, with23˚C-25˚C, 50%-56% humidity. Unless otherwise specified, all
experiments used balanced groups of male and female mice, and all experiments are conduct and compared using mice of the same
age at 8-9 weeks.

Wild animals This study did not involve wild animals.

Field-collected samples This study did not involve field-collected samples.

Ethics oversight Mice were bred and maintained in National Institute of Biological Sciences specific pathogen-free facility in accordance with the
March 2021

Guide for the Care and Use of Laboratory Animals of the NIBS. Procedures were approved by the Laboratory Animal Management
Committee of NIBS and were in compliance with all relevant ethical regulations.
Note that full information on the approval of the study protocol must also be provided in the manuscript.

3
Human research participants

nature portfolio | reporting summary


Policy information about studies involving human research participants
Population characteristics Seven male and three female patients who were pathologically diagnosed with vitiligo were enrolled in this study. Their ages
ranged from 6 to 55, with a median age of 24. Among these patients, four were diagnosed to be in active stage, while the
other six were not sure. All the patients were newly diagnosed with vitiligo and none of the patients had received therapy or
UVB treatment. A 3×3 mm2 biopsy was taken from each patient in the junction region between depigmented lesion region
and pigmented perilesional region. Biopsy with the same size was also taken from 5 healthy donors with other surgical
operation as control. The clinical characteristics of these patients and healthy donors were summarized in Extended Data
Figure 1c.

In this study of anatomically distinct human fibroblasts, we collected skin biopsies from human embryos (female, 22 to 24
weeks of gestation).

Recruitment Vitiligo patients who were scheduled to have surgery for pathological analysis in Beijing hospital were recruited if they
consented to donate tissue. All the patients were newly diagnosed with vitiligo and none of the patients had received therapy
or UVB treatment. All vitiligo skin tissue used are based on their availability, which would not affect our conclusion. Healthy
donors who were scheduled to have surgery for plastic operation in Beijing hospital were recruit if they consented to donate
tissue. All healthy donors’ tissue used are based on their availability, which would not affect our conclusion.

Research donors of human embryo were recruited from Peking University Third Hospital. Before giving consent, donors have
a suitable opportunity to receive proper counseling about the implication of the donation and potential risk. Embryos were
collect with written informed consent from the donors in this study. Both human embryo used are based on their availability,
which would not affect our conclusion.

Ethics oversight The study of single cell RNA-seq and immunofluorescent analysis of human vitiligo skin biopsy was reviewed and approved by
Ethics Committee of Beijing Hospital. For minors, informed consent was obtained from the LAR’s of the minors. The study of
anatomically distinct human fibroblast was reviewed and approved by the Peking University Third Hospital Medical Science
Research Ethics Committee.
Note that full information on the approval of the study protocol must also be provided in the manuscript.

Flow Cytometry
Plots
Confirm that:
The axis labels state the marker and fluorochrome used (e.g. CD4-FITC).
The axis scales are clearly visible. Include numbers along axes only for bottom left plot of group (a 'group' is an analysis of identical markers).
All plots are contour plots with outliers or pseudocolor plots.
A numerical value for number of cells or percentage (with statistics) is provided.

Methodology
Sample preparation For FACS quantification and collection of epidermal immune cells including CD8+ T cells, CD4+ T cells, Langerhans cells, and
γδ T cells, skin from middle section of the tail was harvested, flattened, cut into 1x1 cm pieces, and then floated dermis side
down in 2 mL of 2.4 U/mL dispase solution and incubated at 37˚C with shaking at 80 rpm for 50 min. To obtain most of the
epithelial cells, the epidermis was then carefully removed from dermis along the anterior-posterior direction with a fine-
tipped tweezer. The removed epidermis was floated dermis side down in 3 mL TrypLE™ Express Enzyme (Gibco) at 37˚C for
10 min, afterwards 5% FBS was added to neutralize TrypLE.

For FACS quantification of epidermal melanocyte, melanocytes located in the hair follicle must be removed. We found out
prior to skin collection, wax using VEET wax paper facilitates subsequent hair follicle separation from epidermis. Waxed tail
skin was flattened and floated dermis side down in 2 mL of 2.4 U/mL dispase solution at 37˚C with shaking at 80 rpm for 50
min. To efficiently obtain hair follicle-free epidermis, the epidermis was swiftly peeled from the dermis along the posterior-
anterior direction with a fine-tipped tweezer. The occasional remaining hair follicles attached to epidermis were removed
using tweezers. The hair follicle-free epidermis was then floated dermis side down in 3 mL Trypsin-EDTA (0.25%) at 37˚C for
10 min, afterwards 5% FBS was added to neutralize trypsin.

Single cell suspensions were obtained by repeatedly aspirating and dispensing with a pipette for 20 times followed by
March 2021

filtering through 40 μm strainers. The cells were then stained with anti-mouse CD117 (ebioscience, Cat: 13-1171-82, Clone:
2B8, Dilution: 1:200) for melanocyte detection, anti-mouse CD45 (BD, Cat: 559864, Clone: 10-F11, Dilution: 1:300) for
immune cell detection; anti-mouse CD3 (Biolegend, Cat: 100220, clone: 17A2, Dilution: 1:300) and anti-mouse CD8a
(ebioscience, Cat: 17-0081-83, Clone: 53-6.7, Dilution: 1:300) for CD8+ T cell detection; anti-mouse CD3 and anti-mouse CD4,
(Biolegend, Cat: 100451, Dilution: 1:300) for CD4+ T cell detection; anti-mouse MHCII (Biolegend, Cat: 107629, Dilution:
1:300) and anti-mouse CD207, (Biolegend, Cat: 144206, Dilution: 1:300) for Langerhans cell detection; anti-mouse CD3 and
anti-mouse TCRγδ (Biolegend, Cat: 118108, Dilution: 1:300) antibodies for γδ T cell detection.

For FACS quantification and collection of dermal fibroblasts, endothelial cells, immune cells including macrophage and

4
dendritic cells from tail skin, skin from middle section of the tail was harvested, flattened, cut into 1x1 cm pieces, and then
floated dermis side down in 2 mL of 2.4 U/mL dispase solution and incubated at 37˚C with shaking at 80 rpm for 50 min. To

nature portfolio | reporting summary


get rid of epidermis and hair follicles, the epidermis was then carefully removed from dermis along the anterior-posterior
direction with a fine-tipped tweezer. The dermis was then placed in 10 mL of 2 mg/mL collagenase (Sigma-Aldrich, C2674)
and incubated at 37˚C with shaking at 80 rpm for 1 h, afterwards 5% FBS solution was used for neutralization. Single cell
suspensions were obtained by repeatedly aspirating and dispensing with a pipette for 20 times followed by filtering through
70 μm and 40 μm strainers. The cells were stained with anti-mouse CD31, (ebioscience, Cat: 25-0311-81, Clone: 390,
Dilution: 1:300), anti-mouse-CD45, and anti-mouse pdgfra, (ebioscience, Cat: 17-1401-81, Clone: APA5, Dilution: 1:300)
antibodies for endothelial cell, total immune cell, and fibroblast detection, respectively; anti-mouse/human CD11b
(Biolegend, Cat: 101215, Dilution: 1:300) and anti-mouse F4/80 (Biolegend, Cat: 123116, Dilution: 1:300) for macrophage
detection; anti-mouse MHC-II and anti-mouse CD11c, (Biolegend, Cat: 117310, Dilution: 1:300) for dendritic cell detection.

For analysis and isolation of immune cells from spleens, the spleen was mechanically dissociated and the red blood cells were
lysed with RBC lysis buffer according to the manufacturer’s instructions (Beyotime). Single cell suspensions were obtained by
filtering with strainers (40 μm) and then cells were stained with anti-mouse CD3, anti-mouse CD45, and anti-mouse CD8
antibodies. anti-mouse VB13 (BIolegend, Cat: 140703, Clone: MR12-4, Dilution:1:300 ) antibody was used in Pmel T cell
adoptive transfer experiments.

For CXCR3 expression analysis in skin immune cell types of vitiligo mice, the entire tail skin was harvested, flattened, then
floated dermis side down in 5 mL Trypsin-EDTA (0.25%) at 37˚C for 30 min. The skin was inverted and scraped with a scalpel
to get total skin cell suspensions. Single cell suspensions were obtained by filtering with strainers (70 μm and 40 μm), then
cells were co-stained with anti-mouse CXCR3, (Biolegend, Cat: 126506, Dilution: 1:300) against immune cell markers
mentioned above for further analysis.

For FACS analysis of CD8+ T cells and melanocytes from mouse dorsal, ventral, paw dorsal, and paw ventral skin, skin biopsies
were harvested, flattened, and floated dermis side down in 3 mL of TrypLE and incubated at 37˚C with shaking at 80 rpm for
30 min. The skin epidermis was gently scraped with a scalpel to get the epithelial cell suspensions. Single cell suspensions
were obtained by filtering with strainers (40 μm), then cells were stained with anti-mouse CD45, CD3, CD8, and CD117
antibodies for further analysis.

For re-isolation of injected RFP-labeled dorsal or paw fibroblasts from IFNGR1 KO mouse tail, tail skin was separated into
epidermis and dermis post dispase treatment. After digestion of dermis with collagenase and neutralization, single cell
suspensions were obtained by filtering through 70 μm and 40 μm strainers. Then RFP+ fibroblasts were isolated for further
qPCR analysis.

Instrument BD FACSAria III, BD FACSAria II

Software BD FACSDiva (8.0.1), FlowJo (v10.0.7)

Cell population abundance For sorting for human single cell RNA sequencing, more than 10,000 immune cells and 2,000 melanocytes were collected.
The purity was more than 95% and was verified by single cell RT-qPCR. For sorting for mouse RNA-seq, at least 200,000
fibroblast were collected. The purity was verified by expression of Pdgfra and Krt14 with RT-qPCR. For immune analysis in
mice, 1,000 CD45+ singlets were collected. For CD117+ melanocytes analysis, 20,000 singlets were collected.

Gating strategy In the FACS for single cell RNA sequencing, cells were gated on live (DAPI-) and FSC/SSC- area. CD45+ and c-Kit+ were used to
enrich immune cells and melanocytes; CD45-, c-Kit- cells are predominantly keratinocytes. In dermis, CD45 was used to
distinguish immune cells from dermal cells (Extended Data Figure 1b).

In the FACS for qPCR validation and mouse fibroblast RNA sequencing, cells were gated on live (DAPI-) and FSC/SSC- area.
CD45+ and c-Kit+ markers were used to enrich immune cells and melanocytes; CD45-, c-Kit- cells are predominantly
keratinocytes. In dermis, CD45, CD31 were used to distinguish immune cells and endothelial cells from fibroblast, PDGFRA
were used to enrich fibroblasts (Extended Data Figure 6a).

For immune analysis, cells were gated on live (DAPI-) and FSC/SSC- area. CD45+ were used to enrich immune cells; CD3
+(medium), CD8+ cells are predominantly CD8+ T cells; CD3+(medium), CD4+ cells are predominantly CD4+ T cells;
F4/80+, CD11b+ cells are predominantly Macrophage; CD207+, MHC-II+ cells are predominantly Langerhans cells; TCR γδ+,
CD3+ cells are predominantly γδ T cells; CD11c+, MHC-II+ cells are predominantly dendritic cells (Extended Data Figure 3f).
Positive and negative gates were set using fluorescence minus one (FMO) background intensity controls. Fluorophores were
chosen to minimize spectral overlap.

Tick this box to confirm that a figure exemplifying the gating strategy is provided in the Supplementary Information.
March 2021

You might also like