commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · commensal–dendritic-cell...

13
LETTER doi:10.1038/nature14052 Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2 {*, Nicolas Bouladoux 1,2 *, Jonathan L. Linehan 1,2 , Seong-Ji Han 1,2 , Oliver J. Harrison 1,2 , Christoph Wilhelm 1,2 , Sean Conlan 3 , Sarah Himmelfarb 1,2 , Allyson L. Byrd 1,2,3 , Clayton Deming 3 , Mariam Quinones 4 , Jason M. Brenchley 1,5 , Heidi H. Kong 6 , Roxanne Tussiwand 7 , Kenneth M. Murphy 7 , Miriam Merad 8 , Julia A. Segre 3 & Yasmine Belkaid 1,2 The skin represents the primary interface between the host and the environment. This organ is also home to trillions of microorgan- isms that play an important role in tissue homeostasis and local immunity 1–4 . Skin microbial communities are highly diverse and can be remodelled over time or in response to environmental challenges 5–7 . How, in the context of this complexity, individual commensal micro- organisms may differentially modulate skin immunity and the con- sequences of these responses for tissue physiology remains unclear. Here we show that defined commensals dominantly affect skin immu- nity and identify the cellular mediators involved in this specification. In particular, colonization with Staphylococcus epidermidis induces IL-17A 1 CD8 1 T cells that home to the epidermis, enhance innate barrier immunity and limit pathogen invasion. Commensal-specific T-cell responses result from the coordinated action of skin-resident dendritic cell subsets and are not associated with inflammation, reveal- ing that tissue-resident cells are poised to sense and respond to alter- ations in microbial communities. This interaction may represent an evolutionary means by which the skin immune system uses fluctuat- ing commensal signals to calibrate barrier immunity and provide heterologous protection against invasive pathogens. These findings reveal that the skin immune landscape is a highly dynamic environ- ment that can be rapidly and specifically remodelled by encounters with defined commensals, findings that have profound implications for our understanding of tissue-specific immunity and pathologies. We first assessed whether individual commensal species could mod- ulate immunity in the context of pre-existing microbial communities. Despite the presence of a diverse microbiota, the skin of specific patho- gen free (SPF) mice was permissive to long-term colonization with S. epidermidis 5 at all skin sites analysed (Fig. 1a, b). At 2 weeks post topical association with as low as 1.3 3 10 6 colony-forming units (c.f.u.) per cm 2 , levels of IL-17A- and IFN-c-expressing T cells but not Foxp3 1 regu- latory T cells were significantly increased at several skin sites analysed (Fig. 1c, d and Extended Data Fig. 1a–d). Long-term accumulation of IL-17A-expressing T cells was not observed at sites distal to the skin and required colonization with live bacteria (Fig. 1c, d, h and Extended Data Fig. 1b). Furthermore, in contrast to responses to intradermal inocula- tion of S. epidermidis, commensal responses were not associated with inflammation (Fig. 1e–h and Extended Data Fig. 1e–h). Thus, an encoun- ter with a new commensal can lead to a robust but non-inflammatory accumulation of effector T cells in the skin. We next assessed the capacity of other constituents of the human (Corynebacterium pseudodiphtheriticum, Propionibacterium acnes and Staphylococcus aureus) and murine (Staphylococcus xylosus, Staphylococ- cus lentus, Rothia nasimurium and S. epidermidis 42E03) skin microbiota to influence T-cell responses (Extended Data Fig. 2a). Six out of eight bacteria tested increased the number of skin IL-17A 1 T cells and half of the commensals also increased the number of IFN-c-expressing T cells (Fig. 2a and Extended Data Fig. 2a, b). Thus, the induction of cytokines, and in particular IL-17A, is a relatively conserved response of the skin to an encounter with a new commensal. The majority of ab T cells found in murine skin are CD4 1 T cells with few resident CD8 1 T cells 8 (Fig. 2b, c). Notably, S. epidermidis isolates were uniquely able to increase the number and frequencies of CD8 1 T cells in the skin in both SPF and germ-free conditions and in response to an application dose as low as 1.3 3 10 6 c.f.u. per cm 2 (Fig. 2c and Ex- tended Data Fig. 2c–h). Similarly to tissue-resident memory (TREM) cells induced by viral challenges 9 , clusters of CD8 1 T cells preferen- tially localized to the basal epidermis or in close proximity to the epithe- lial layer and expressed CD103 and CD69 (Fig. 2c, d and Extended Data Fig. 3a, b). On the other hand, commensal-evoked CD8 1 T cells have a distinct cytokine profile characterized by the production of either IL-17A or IFN-c and in contrast to virally induced TREM cells that localize to the site of injury, commensal-induced CD8 1 T cells accumulated at all skin sites analysed (Fig. 2c and Extended Data Fig. 1b). Although rarely seen at other body sites, Tc17 cells (a subset of CD8 1 T cells) can be found in healthy non-human primate and human skin (Fig. 2e and Extended Data Fig. 3c). This discrete response provided us with the opportunity to explore the factors controlling a commensal-driven immune specification. In germ-free mice, commensals promote T-cell responses through IL-1 (ref. 2). Consistently, mice deficient in IL-1R1 contained significantly fewer skin IL-17A 1 CD8 1 T cells post S. epidermidis association, and in vitro stimulation of purified S. epidermidis-evoked CD8 1 T cells with IL-1 boosted IL-17A release (Fig. 2f and Extended Data Fig. 3d). CD8 1 T-cell response peaked at 2 weeks post association, at which point the number of cells slowly contracted, although increased frequencies were maintained up to 6 months post application (Fig. 2g, h). Dendritic cells are exquisite sensors of their environment and pre- vious studies uncovered a functional specialization of defined dendritic cell subsets in their capacity to drive unique immune modules 10 . What remains unexplored is how this specialization could account for the capacity of the host to regulate defined aspects of its relationship with the microbiota. In the skin, dendritic cells could potentially be exposed to the microbiota via emission of dendrites through epithelial cells and/ or to commensal products passively diffusing at invaginations such as hair follicles, which have more permissive cell adhesions 11 . Supporting the idea that CD8 1 T-cell accumulation post association depends on migrat- ory dendritic cells 12 , the response was largely abolished in Ccr7 2/2 mice (Fig. 3a). Frequencies of skin dendritic cell subsets 13 were not affected *These authors contributed equally to this work. 1 Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA. 2 Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA. 3 Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA. 4 Bioinformatics and Computational Bioscience Branch, National Institute of Allergy and Infectious Diseases, NIH Bethesda, Maryland 20892, USA. 5 Immunopathogenesis Section, Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, NIH Bethesda, Maryland 20892, USA. 6 Dermatology Branch, National Cancer Institute, NIH Bethesda, Maryland 20892, USA. 7 Howard Hughes Medical Institute, Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA. 8 Department of Oncological Sciences, Tisch Cancer Institute and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA. {Present addresses: Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA. 00 MONTH 2015 | VOL 000 | NATURE | 1 Macmillan Publishers Limited. All rights reserved ©2015

Upload: others

Post on 19-Oct-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

LETTERdoi:10.1038/nature14052

Commensal–dendritic-cell interaction specifies aunique protective skin immune signatureShruti Naik1,2{*, Nicolas Bouladoux1,2*, Jonathan L. Linehan1,2, Seong-Ji Han1,2, Oliver J. Harrison1,2, Christoph Wilhelm1,2,Sean Conlan3, Sarah Himmelfarb1,2, Allyson L. Byrd1,2,3, Clayton Deming3, Mariam Quinones4, Jason M. Brenchley1,5,Heidi H. Kong6, Roxanne Tussiwand7, Kenneth M. Murphy7, Miriam Merad8, Julia A. Segre3 & Yasmine Belkaid1,2

The skin represents the primary interface between the host and theenvironment. This organ is also home to trillions of microorgan-isms that play an important role in tissue homeostasis and localimmunity1–4. Skin microbial communities are highly diverse and canbe remodelled over time or in response to environmental challenges5–7.How, in the context of this complexity, individual commensal micro-organisms may differentially modulate skin immunity and the con-sequences of these responses for tissue physiology remains unclear.Here we show that defined commensals dominantly affect skin immu-nity and identify the cellular mediators involved in this specification.In particular, colonization with Staphylococcus epidermidis inducesIL-17A1 CD81 T cells that home to the epidermis, enhance innatebarrier immunity and limit pathogen invasion. Commensal-specificT-cell responses result from the coordinated action of skin-residentdendritic cell subsets and are not associated with inflammation, reveal-ing that tissue-resident cells are poised to sense and respond to alter-ations in microbial communities. This interaction may represent anevolutionary means by which the skin immune system uses fluctuat-ing commensal signals to calibrate barrier immunity and provideheterologous protection against invasive pathogens. These findingsreveal that the skin immune landscape is a highly dynamic environ-ment that can be rapidly and specifically remodelled by encounterswith defined commensals, findings that have profound implicationsfor our understanding of tissue-specific immunity and pathologies.

We first assessed whether individual commensal species could mod-ulate immunity in the context of pre-existing microbial communities.Despite the presence of a diverse microbiota, the skin of specific patho-gen free (SPF) mice was permissive to long-term colonization with S.epidermidis5 at all skin sites analysed (Fig. 1a, b). At 2 weeks post topicalassociation with as low as 1.3 3 106 colony-forming units (c.f.u.) per cm2,levels of IL-17A- and IFN-c-expressing T cells but not Foxp31 regu-latory T cells were significantly increased at several skin sites analysed(Fig. 1c, d and Extended Data Fig. 1a–d). Long-term accumulation ofIL-17A-expressing T cells was not observed at sites distal to the skin andrequired colonization with live bacteria (Fig. 1c, d, h and Extended DataFig. 1b). Furthermore, in contrast to responses to intradermal inocula-tion of S. epidermidis, commensal responses were not associated withinflammation (Fig. 1e–h and Extended Data Fig. 1e–h). Thus, an encoun-ter with a new commensal can lead to a robust but non-inflammatoryaccumulation of effector T cells in the skin.

We next assessed the capacity of other constituents of the human(Corynebacterium pseudodiphtheriticum, Propionibacterium acnes andStaphylococcus aureus) and murine (Staphylococcus xylosus, Staphylococ-cus lentus, Rothia nasimurium and S. epidermidis 42E03) skin microbiota

to influence T-cell responses (Extended Data Fig. 2a). Six out of eightbacteria tested increased the number of skin IL-17A1 T cells and halfof the commensals also increased the number of IFN-c-expressing T cells(Fig. 2a and Extended Data Fig. 2a, b). Thus, the induction of cytokines,and in particular IL-17A, is a relatively conserved response of the skinto an encounter with a new commensal.

The majority ofabT cells found in murine skin are CD41 T cells withfew resident CD81 T cells8 (Fig. 2b, c). Notably, S. epidermidis isolateswere uniquely able to increase the number and frequencies of CD81 Tcells in the skin in both SPF and germ-free conditions and in responseto an application dose as low as 1.3 3 106 c.f.u. per cm2 (Fig. 2c and Ex-tended Data Fig. 2c–h). Similarly to tissue-resident memory (TREM)cells induced by viral challenges9, clusters of CD81 T cells preferen-tially localized to the basal epidermis or in close proximity to the epithe-lial layer and expressed CD103 and CD69 (Fig. 2c, d and Extended DataFig. 3a, b). On the other hand, commensal-evoked CD81 T cells have adistinct cytokine profile characterized by the production of either IL-17Aor IFN-c and in contrast to virally induced TREM cells that localize tothe site of injury, commensal-induced CD81 T cells accumulated at allskin sites analysed (Fig. 2c and Extended Data Fig. 1b). Although rarelyseen at other body sites, Tc17 cells (a subset of CD81 T cells) can be foundin healthy non-human primate and human skin (Fig. 2e and ExtendedData Fig. 3c). This discrete response provided us with the opportunity toexplore the factors controlling a commensal-driven immune specification.

In germ-free mice, commensals promote T-cell responses throughIL-1 (ref. 2). Consistently, mice deficient in IL-1R1 contained significantlyfewer skin IL-17A1 CD81 T cells post S. epidermidis association, andin vitro stimulation of purified S. epidermidis-evoked CD81 T cells withIL-1 boosted IL-17A release (Fig. 2f and Extended Data Fig. 3d). CD81

T-cell response peaked at 2 weeks post association, at which point thenumber of cells slowly contracted, although increased frequencies weremaintained up to 6 months post application (Fig. 2g, h).

Dendritic cells are exquisite sensors of their environment and pre-vious studies uncovered a functional specialization of defined dendriticcell subsets in their capacity to drive unique immune modules10. Whatremains unexplored is how this specialization could account for thecapacity of the host to regulate defined aspects of its relationship withthe microbiota. In the skin, dendritic cells could potentially be exposedto the microbiota via emission of dendrites through epithelial cells and/or to commensal products passively diffusing at invaginations such ashair follicles, which have more permissive cell adhesions11. Supporting theidea that CD81 T-cell accumulation post association depends on migrat-ory dendritic cells12, the response was largely abolished in Ccr72/2 mice(Fig. 3a). Frequencies of skin dendritic cell subsets13 were not affected

*These authors contributed equally to this work.

1Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, NIH, Bethesda 20892, USA. 2Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute ofAllergy and Infectious Diseases, NIH, Bethesda, Maryland 20892, USA. 3Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA.4Bioinformatics and Computational Bioscience Branch, National Institute of Allergy and Infectious Diseases, NIH Bethesda, Maryland 20892, USA. 5Immunopathogenesis Section, Laboratory of MolecularMicrobiology, National Institute of Allergy and Infectious Diseases, NIH Bethesda, Maryland 20892, USA. 6Dermatology Branch, National Cancer Institute, NIH Bethesda, Maryland 20892, USA. 7HowardHughesMedical Institute, Departmentof Pathologyand Immunology,WashingtonUniversitySchool of Medicine, St Louis, Missouri 63110,USA. 8DepartmentofOncological Sciences, Tisch Cancer Instituteand Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA. {Present addresses: Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology andDevelopment, The Rockefeller University, New York, New York 10065, USA.

0 0 M O N T H 2 0 1 5 | V O L 0 0 0 | N A T U R E | 1

Macmillan Publishers Limited. All rights reserved©2015

Page 2: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

by S. epidermidis application, indicating that the induction of Tc17 cellsdid not result from altered dendritic cell frequencies (Extended DataFig. 4a, b).

Mice constitutively deficient in Langerhans cells14 mounted T-cellresponses to S. epidermidis in a manner comparable to their littermatecontrols (Fig. 3b and Extended Data Fig. 4c, and data not shown). Cross-presenting CD1031 dendritic cells15 depend on the expression of IRF8and BATF3 for their development10,16 while CD11b1 dendritic cellsrequire CSF1 for their development and maintenance17,18 and IRF4 forthe formation of peptide–MHC (major histocompatibility complex) classII complexes19. Making use of this differential requirement for transcrip-tion or survival factors, we assessed the relative contribution of thesetwo dendritic cell subsets to CD81 T-cell responses. The selective defectin skin-resident CD1031 dendritic cells (but not lymph-node-residentCD8a1 dendritic cells; Extended Data Fig. 4d) in our Batf32/2 colony

allowed us to evaluate the direct contribution of these cells. Batf32/2

and Irf82/2 mice failed to develop CD81 T-cell responses following col-onization compared to control mice, highlighting a non-redundant rolefor CD1031 dendritic cells in the induction of CD81 T-cell responsesto S. epidermidis (Fig. 3c and Extended Data Fig. 4c, e).

Treatment of mice with an anti-CSF1R antibody led to a marked reduc-tion in Langerhans cells and skin CD11b1 dendritic cells, as well asin skin IL-17A1 CD81 T cells (Fig. 3d and Extended Data Fig. 4c). Asthe specific deletion of Langerhans cells had no consequence on T-cellresponses to commensals (Fig. 3b), these results suggest that the immuneeffect observed was due to CD11b1 dendritic cells. A large fraction ofS. epidermidis-evoked CD81 T cells were in close contact with CSF1R1

CD11c1 cells in the skin and these cells were the most transcriptionally

14 30 60 1800

255060

110160210260

Days post association

c.f

.u.

per

earActinobacteria ***

*****

** **

* ***

** **

*

Bacteroidetes

Deferribacteres

Firmicutes (other)

Firmicutes (S. epi .)

Other/unclassified

Proteobacteria

TenericutesAb

un

dan

ce

100

75

50

25

0

Day 14 Day 180

Ctrl S. epi. Ctrl S. epi.

ba

IL-1

7A

IFN-γ

Skin Lung Gut

9

1

18

9

1

9

1

10

12 3

34

12 2

24

Control

S. epi.

c

TC

Rβ+

IF

N-γ

+ (×1

03)

TC

Rβ+

IF

N-γ

+ (×1

03)

d

0

2

4

6

TC

Rβ+

IL

-17

A+ (×1

03)

TC

Rβ+

IL

-17

A+ (×1

03)

0

2

4

6

8

10

Ctrl

S. epi.

HK S. e

pi.

Ctrl

S. epi.

HK S. e

pi.

Control Topical Intradermal

S. epidermidis (day 7)e g

Day 60Day 14 Day 180Day 30

9

3

10

2

23

15

9

5

19

10

IFN-γ

Control

S. epi.IL-1

7A

13

3

30

15

21

17

h

16

11

2

44

Topical Intradermal

S. epidermidis (day 14)

IL-1

7A

IFN-γ

f

S. epi.Control

NS

NS4

2

3

1

0

60

20

40

1

0

2

Ctrl

Day 7

Day 1

4Ctrl

Day 7

Day 1

4

S. epi. topical S. epi. intradermal

Figure 1 | Remodelling of skin immunity by commensal colonization.a, Relative abundance of bacterial phyla in mouse skin 14 and 180 days afterS. epidermidis topical application. Each bar represents the percentage ofsequences in operational taxonomic units (OTUs) assigned to each phylum foran individual mouse. Ctrl, control. b, Enumeration of colony-forming units(c.f.u.) from the ears after S. epidermidis application (n 5 5–10 per group).c, IFN-c and IL-17A production by skin, lung or gut effector (CD451 TCRb1

Foxp32) T cells in unassociated (control) and S. epidermidis (S. epi.)-associatedmice at day 14. d, Absolute numbers of skin IFN-c1 or IL-17A1 effectorT cells in unassociated mice (control, n 5 3) and mice topically associated withlive (S. epi., n 5 4) or heat-killed (HK S. epi., n 5 4) S. epidermidis at day 14.e, Representative images and histopathological comparison of the ear pinnaeof unassociated (control), topically associated (topical) or intradermallyinoculated (intradermal) mice at day 7. Scale bars, 250mm. f, g, Frequencies andabsolute numbers of skin IFN-c1 or IL-17A1 effector T cells after topicalapplication (n 5 4) or intradermal inoculation (n 5 4–5) of S. epidermidis.h, IFN-c and IL-17A production by skin effector T cells in unassociated andS. epidermidis-associated mice at different time points. Results arerepresentative of 2–3 independent experiments. *P , 0.05, **P , 0.01,***P , 0.001 as calculated by Student’s t-test.

bCD4+ T cells

12

9

6

3

0CD

4+ IL

-17

A+ (×1

03)

Ctrl

S. epi.

S. xylo

.

C. pse

udo.

P. acn

es

S. aure

us

TC

Rβ+

IF

N-γ

+ (×1

03)

9 ***

**

**

*****

*** ***

*****

* ** * *

6

3

0

IFN-γ+ Teff cells

20

15

10

0

TC

Rβ+

IL

-17

A+ (×1

03)

5

IL-17A+ Teff cells

Ctrl

S. epi.

S. xylo

.

C. pse

udo.

P. acn

es

a

S. aure

usCtrl

S. epi.

S. xylo

.

C. pse

udo.

P. acn

es

S. aure

us

cControl S. epi.

3

63

34

44

CD

CD4

IL-1

7A

IFN-γ

17

37

TC

Rβ+

CD

8β+

(×1

03) CD8β+ T cells

25

20

15

10

5

0

Ctrl

S. epi.

S. xylo

.

C. pse

udo.

P. acn

es

S. aure

us

CD

69

CD103

16

7

73

e Human NHP

5

43

11

20

IL-1

7A

IFN-γ

CD8α CD207 (GFP) MergeDAPI

Epidermis

Dermis

d

7

28

25

26

Il1r1–/–

Il1r1–/

Il1r1–/

WT

IL-1

7A

IFN-γ

27

18

9

0

5

4

2

0

3

1

CD

8β+

IL-1

7A

+ (×1

03)

WT

WT

f CD8β+ T cells

1 3 5 7 14 30 18060Days post association

20

15

10

5

0

2.5

2.0

1.5

0.5

1.0

0

CD

8β+

IL

-17

A+ (×1

03)

1 3 5 7 14 30 18060Days post association

1 3 5 7 14 30 18060Days post association

35302520151050T

CRβ+

CD

8β+

(%

)T

CRβ+

CD

8β+

(×1

03)

TC

Rβ+

CD

8β+

(%

)

4

3

2

1

01 3 5 7 14 30 18060Days post association

CD

8β+

IF

N-γ

+ (×1

03)CD8β+ T cellsg h

CtrlS. epi.

CtrlS. epi.

CtrlS. epi.

CtrlS. epi.

***

*****

***

*

*

*****

**

****

*****

****

**

Figure 2 | Distinct commensal species impose specific immune signaturesin the skin. a, Mice were left unassociated (Ctrl, n 5 8) or topically associatedwith S. epidermidis (n 5 7), S. xylosus (n 5 7), S. aureus (n 5 5), C.pseudodiphtheriticum (n 5 7) or P. acnes (n 5 6). Absolute numbers of skinIFN-c1 or IL-17A1 effector T cells are shown 2 weeks after first association.b, Absolute numbers of skin IL-17A1 CD41 effector T cells from mice in a.c, Absolute numbers of skin CD8b1 effector T cells from mice in a. Flowplots show the frequencies of CD41 and CD8b1 effector T cells in unassociatedand S. epidermidis-associated mice, the IFN-c and IL-17A production, andthe expression of CD69 and CD103 by CD8b1 T cells in associated mice.d, Representative imaging volume projected along the x axis of ears fromLangerin–GFP (green fluorescent protein) reporter mice 14 days postS. epidermidis application. Scale bars, 30mm; DAPI, 49,6-diamidino-2-phenylindole. e, CD31 CD81 IFN-c1 and CD31 CD81 IL-17A1 T cells innormal human (n 5 1) and non-human primate (NHP) skin (n 5 8).f, Frequencies and absolute numbers of total CD8b1 or IL-17A1 CD8b1

effector T cells in the skin of wild-type (WT, n 5 4) and Il1r12/2 (n 5 4) miceafter S. epidermidis application. g, h, Frequencies and absolute numbers(mean 6 s.e.m.) of total CD8b1, IFN-c1 CD8b1 and IL-17A1 CD8b1 effectorT cells in the skin over time following S. epidermidis application (n 5 3–5per time point). Results in a–c are a compilation of 2–3 experiments. Resultsin d–h are representative of two independent experiments. *P , 0.05,**P , 0.01, ***P , 0.001 as calculated by Student’s t-test.

RESEARCH LETTER

2 | N A T U R E | V O L 0 0 0 | 0 0 M O N T H 2 0 1 5

Macmillan Publishers Limited. All rights reserved©2015

Page 3: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

altered (including increases in Il1a and Il1b transcripts and a decreasein Il1rn transcripts) following S. epidermidis application (Fig. 3e, f anddata not shown). Furthermore, anti-CSF1R antibody treatment sig-nificantly reduced skin IL-1 levels post association (Extended DataFig. 4f). In mice conditionally depleted of IRF4 in their dendritic cellcompartment19, CD81 T cells accumulated in the skin but failed to pro-duce IL-17A (Extended Data Fig. 4g). Together, these results supportthe idea that through their capacity to produce IL-1, CD11b1 dendriticcells could promote the induction and/or maintenance of IL-17A expres-sion by CD81 T cells. Thus, cooperation between skin-resident dendriticcells promotes and tunes responses to a defined commensal (Fig. 4g).

Of note, healthy human skin contains approximately 20 billion effec-tor lymphocytes of unknown specificity20. To address the possibilitythat T cells accumulating in murine skin in response to S. epidermidiswere commensal specific, CD81 T cells were purified from the skinor regional lymph nodes and exposed to dendritic cells loaded withS. epidermidis antigens. Exposure of S. epidermidis-loaded dendriticcells to CD81 T cells promoted potent IL-17A and IFN-c production(Fig. 4a, b and Extended Data Fig. 5a). In contrast, stimulation with IL-1a

or other bacterial antigens or ligands failed to induce cytokine produc-tion, and S. epidermidis-loaded dendritic cells did not promote cytokinerelease by activated T cells of irrelevant specificity (Fig. 4b and data notshown). A fraction of IFN-cproduction by CD81 T cells was still detect-able in the absence of b2-microglobulin-dependent MHC class I pre-sentation (Fig. 4a, b and Extended Data Fig. 5b), suggesting that someof the IFN-c-producing CD81 T cells may be responding in a bystandermanner or in a b2-microglobulin-independent manner21. However,IL-17A was not observed following exposure to B2m2/2 dendritic cellsloaded with S. epidermidis, demonstrating that Tc17 cells are commensalspecific (Fig. 4a, b). Commensal-specific IL-17A1 cells were also foundin the CD41 T-cell compartment in both the skin and regional lymphnodes following S. epidermidis or S. xylosus application (Extended DataFig. 5c–e). Our present results support the idea that, as in the gut22–24,the majority of IL-17A-producing T cells in the skin may be commensalspecific.

To assess the consequence of adaptive immune responses to com-mensals for skin immunity we employed a model of epicutaneous infec-tion with the fungal pathogen Candida albicans. Prior association with

20 ***15

10

5

0

TC

Rβ+

CD

8β+

(×1

03)

WT

31

46

10

60

CD4

CD

WT Ccr7–/–

Ccr

7–/

a WT Lan–DTA

23 52

6.6

22 0.6

40

CD

11

b

CD207 CD

8β+

IF

N-γ

+ (×1

03)

CD

8β+

IF

N-γ

+ (×1

03)

CD

8β+

IL

-17

A+ (×1

03)

CD

8β+

IL

-17

A+ (×1

03)

0

1

2

3

4

5 12

8

4

0

WT

Lan–D

TAW

T

Lan–D

TA

TC

Rβ+

CD

8β+

(×1

03)

TC

Rβ+

CD

8β+

(×1

03)

0

9

18

27

36

45

WT

Lan–D

TA

b

WT Batf3–/–

Bat

f3–/

Bat

f3–/

1868

6.7

2762

0.1

CD

11

b

CD207

15

10

5

0

3

2

1

0

WT W

T

2.0

1.5

1.0

0.5

0

2.5

WT

CD4

CD

32

44

6

77

c

Rat IgG Anti-CSF1R

18

31

4

42

IL-1

7A

IFN-γ

Rat Ig

G

Anti-CSF1R

Rat Ig

G

Anti-CSF1R

27

18

0

9

CD

8β+

IL

-17

A+ (%

)

CD

8β+

IF

N-γ

+ (%

)

70

60

50

40

30

20

10

0

d

S. epi.

(day 5) Control

Ccr2Cxcl2

Map3k7Mapk14

Ddit3Ly96

Myd88Il18rapIl1aIl1bIl6Il6raTgfb1

Ccl3

0–5 +5

0

50

100

150

200

250

0

20

40

60

80

100 50

40

30

0

mR

NA

co

unts

mR

NA

co

unts

Ctrl

S. epi.

Ctrl

S. epi.

Ctrl

S. epi.

Il1a Il1a Il1rn

Day 5 Day 12

20

10

e

Epidermis

Epidermis

Dermis

CD8α CSF1R (GFP) MergeDAPI CD11cf

NSNS

NS

NS

**

* * *

** **

**

Bat

f3–/

Figure 3 | Distinct dendritic cell subsets cooperate to mediate host–commensal interaction in the skin. a, Frequencies and absolute numbers ofskin CD8b1 effector T cells in wild-type (WT, n 5 3) and Ccr72/2 (n 5 3) mice2 weeks post first S. epidermidis topical application. b, Phenotypic analysisof skin MHCII1 CD11c1 cells and absolute numbers of skin total CD8b1,IFN-c1 CD8b1 and IL-17A1 CD8b1 effector T cells in wild-type (n 5 4)and Langerin–diphtheria toxin subunit A (Lan–DTA, n 5 7) mice afterS. epidermidis application. NS, not significant. c, Phenotypic analysis ofMHCII1 CD11c1 cells and effector T cells in the skin of wild-type (n 5 4) andBatf32/2 (n 5 5) mice after S. epidermidis application. Graphs illustrate theabsolute numbers of skin total CD8b1, IFN-c1 CD8b1 and IL-17A1 CD8b1

effector T cells. Results shown in a–c are representative of 2–3 experiments.**P , 0.01, ***P , 0.001 as calculated by Student’s t-test. d, Frequencies of

IFN-c1 CD8b1 and IL-17A1 CD8b1 effector T cells in S. epidermidis-associated mice treated with anti-CSF1R (n 5 7) or rat IgG isotype control(n 5 7). Graphs are a compilation of the results of two independentexperiments. **P , 0.01 by Student’s t-test. e, Heat map of genes statisticallydifferentially expressed in skin CD11b1 CD1032 dendritic cells from topicallyassociated versus unassociated mice. Graphs summarize Il1a and Il1rnmRNA counts at day 5 and/or day 12 in CD11b1 CD1032 dendritic cells. Eachcolumn of the heat map and each dot of the graph represent gene expressionfrom a biological replicate comprised of skin cells pooled and purified from5 mice (n 5 4 biological replicates, *P , 0.05). f, Representative imagingvolume projected along the z axis of ears from CSF1R GFP reporter mice14 days post application, showing contact (arrows) between CD8a1 andCD11c1 CSF1R1 cells. Scale bars, 40mm.

LETTER RESEARCH

0 0 M O N T H 2 0 1 5 | V O L 0 0 0 | N A T U R E | 3

Macmillan Publishers Limited. All rights reserved©2015

Page 4: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

S. epidermidis significantly improved innate protection against C. albicans,an effect that was abolished by depletion of CD81 T cells or neutral-ization of IL-17A (Fig. 4c, d). Microarray analysis of gene expressionfollowing S. epidermidis association revealed a significant upregulationof the alarmins S100A8 and S100A9 (data not shown), known to elicitmicrobicidal effects and as potent chemoattractants for neutrophils25.Upregulation of these molecules was still detectable at an applicationdose as low as 1.3 3 106 c.f.u. per cm2, and 2 weeks past the peak of CD81

T-cell response in the skin (Extended Data Fig. 5f and data not shown).In keratinocytes, induction of these molecules can be mediated by IL-17A26. Analysis of gene expression by interfollicular keratinocytes27 fromS. epidermidis-associated mice revealed a CD81 T-cell- and IL-17A-dependent upregulation of S100A8 and S100A9 expression (Fig. 4e, f).Thus, S. epidermidis induces CD81 T-cell responses able to promoteskin innate responses in such a way that promotes heterologous pro-tection against invasive microbes (Fig. 4g).

The skin immune system has evolved in the context of its constitutiveexposure to a diverse microbiota that can be remodelled over time or inresponse to environmental challenges5–7,28. Here we show that the skin

is a highly dynamic immune environment that can be finely calibratedby defined commensals. In light of our present findings, it is intriguingto speculate that microbial diversity may also be required as a means totrigger and educate distinct aspects of the immune system. For instance,the capacity of defined commensals to promote CD81 T-cell responsesthat can home to the epidermis may have evolved as a means to spe-cifically reinforce the barrier function of this compartment. Here, weuncovered that tissue-resident dendritic cells are primary sensors of fluc-tuations in the commensal community and that these cells act in a highlycoordinated manner to orchestrate the formation of commensal-specificT-cell responses. The result of this evolving interaction may represent apowerful protective mechanism of host defence by providing hetero-logous immunity against invasive microbes. In the context of highlyperturbed communities, the cumulative cost of these responses may havesevere consequences on tissue homeostasis. Indeed, commensal-specificresponses are characterized by the production of IL-17A, a cytokineknown to contribute to the aetiology and pathology of various skininflammatory disorders, including psoriasis29. Our work also presentsa possible explanation for how variation in microbial communities atdifferent skin sites5,6 may contribute to the site-specificity of dermatol-ogical disorders.

Online Content Methods, along with any additional Extended Data display itemsandSourceData, are available in the online version of the paper; references uniqueto these sections appear only in the online paper.

Received 7 March; accepted 11 November 2014.

Published online 5 January 2015.

1. Grice, E. A. & Segre, J. A. The skin microbiome. Nature Rev. Microbiol. 9, 244–253(2011).

2. Naik, S. et al. Compartmentalized control of skin immunity by residentcommensals. Science 337, 1115–1119 (2012).

3. Chehoud, C. et al. Complement modulates the cutaneous microbiome andinflammatory milieu. Proc. Natl Acad. Sci. USA 110, 15061–15066 (2013).

4. Sanford, J. A. & Gallo, R. L. Functions of the skin microbiota in health and disease.Semin. Immunol. 25, 370–377 (2013).

5. Grice, E. A. et al. Topographical and temporal diversity of the human skinmicrobiome. Science 324, 1190–1192 (2009).

6. Costello, E. K. et al. Bacterial community variation in human body habitats acrossspace and time. Science 326, 1694–1697 (2009).

7. Kong, H. H. et al. Temporal shifts in the skin microbiome associated with diseaseflares and treatment in children with atopic dermatitis. Genome Res. 22, 850–859(2012).

f

g

100

75

50

0

25

125

C. alb

ican

s

Anti-CD8

Rat Ig

G2b

Mouse

IgG

1

Anti-IL

-17A

S. epi. + C. albicans

400

300

200

0

100

S. epi. + C. albicans

Fo

ld in

cre

ase

over

naiv

e

S100a8 S100a9

1

0

23

46

6

27

5 2

12

00 19

IFN-γ

IL-1

7A

Control PMA S. epi.

B2m–/–WTWTWT

a

b

0

9

18

27

36

45C

D8β+

IL

-17

A+ (%

)

5040

30

05

10

20

60

CD

8β+

IF

N-γ

+ (%

)

Ctrl

S. epi.

PMA

B2m–/– S. e

pi.

S. xylo

.

S. aure

usLPS

P3C

IL-1α/L

PS/P3C

IFN-γ+ cells IL-17A+ cells

12

0

4

8 129

6

03

15

18

Fo

ld in

cre

ase

over

naiv

e

S100a8 S100a9

Interfollicular keratinocytes

Days0 62 4 8 10 12

S. epidermidis

Anti-CD8 or Anti-IL-17A or

isotype

Analysis

C. albicans

c

20

15

10

0

5

25

C.

alb

ican

s c

.f.u

. (×

10

2)

S. epi + C. albicans

d

e

CD103+

DC

Ep

iderm

isD

erm

is

CD11b+

DC

IL-17A

IFN-γCD8+ T cell

Draining lymph node

IL-1

IL-17A

Pathogen

S100A9

S100A8

S. epidermidisMicrobiota

Keratinocytes

****

*****

******* ****

****

Ctrl

S. epi.

PMA

B2m–/– S. e

pi.

S. xylo

.

S. aure

usLPS

P3C

IL-1α/L

PS/P3C

****

****

C. alb

ican

s

Anti-CD8

Rat Ig

G2b

Mouse

IgG

1

Anti-IL

-17A

C. alb

ican

s

Anti-CD8

Rat Ig

G2b

Mouse

IgG

1

Anti-IL

-17A

***

******

**

Anti-CD8

Rat Ig

G2b

Mouse

IgG

1

Anti-IL

-17A

Anti-CD8

Rat Ig

G2b

Mouse

IgG

1

Anti-IL

-17A

****

*

*

Figure 4 | Commensal-driven CD81 T cell response is specific for S.epidermidis antigen. a, CD8b1 effector T cells from the skin of S. epidermidis-associated mice were co-cultured with wild-type (WT) or B2m2/2 splenicdendritic cells (SpDC) untreated (Ctrl) or pre-incubated with heat-killedS. epidermidis, S. xylosus or S. aureus; LPS, Pam3Cys (P3C) or IL-1a. Flow plotsillustrate the frequencies of IFN-c1 CD8b1 and IL-17A1 CD8b1 T cells inovernight co-cultures. PMA, naive SpDC 1 PMA/ionomycin. b, Frequencies ofIFN-c1 CD8b1 and IL-17A1 CD8b1 T cells in overnight co-cultures asdescribed in a (mean 6 standard deviation of triplicate cultures). Data arerepresentative of 2–3 independent experiments. ***P , 0.001, ****P , 0.0001as calculated by Student’s t-test. c, Unassociated or S. epidermidis-topicallyassociated mice were infected with C. albicans and treated with anti-CD8,anti-IL-17A or corresponding isotype control antibodies. d, Enumeration of C.albicans colony-forming units from dorsal skin biopsies from mice in c 2 dayspost C. albicans infection. e, S100a8 and S100a9 gene expression (fold increaseover naive unassociated) by interfollicular keratinocytes purified from the earsof mice 2 weeks after S. epidermidis application. f, S100a8 and S100a9 geneexpression (fold increase over naive unassociated and uninfected) in dorsal skinbiopsies from mice in c 2 days post C. albicans infection. Results in d–f arerepresentative of two independent experiments. *P , 0.05, **P , 0.01,***P , 0.001 as calculated by Student’s t-test. g, Model of the response of theskin immune system to colonization with a new commensal. Skin-residentCD1031 dendritic cells (DC) may acquire commensals or commensal-derivedantigens by reaching into skin appendages or via capture of soluble factors.CD81 T cells primed by CD1031 dendritic cells in the lymph node, migrate tothe skin and are locally tuned by IL-1 produced by CD11b1 dendritic cells.Commensal-specific CD81 T cells can enhance antimicrobial defence ofkeratinocytes in an IL-17 dependent manner. Dotted lines indicate points thatare not addressed in this work.

RESEARCH LETTER

4 | N A T U R E | V O L 0 0 0 | 0 0 M O N T H 2 0 1 5

Macmillan Publishers Limited. All rights reserved©2015

Page 5: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

8. Belkaid, Y. et al. CD81 T cells are required for primary immunity in C57BL/6 micefollowing low-dose, intradermal challenge with Leishmania major. J. Immunol. 168,3992–4000 (2002).

9. Mueller, S. N., Gebhardt, T., Carbone, F. R. & Heath, W. R. Memory T cell subsets,migration patterns, and tissue residence. Annu. Rev. Immunol. 31, 137–161(2013).

10. Murphy, K. M. Transcriptional control of dendritic cell development. Adv. Immunol.120, 239–267 (2013).

11. Mittal, A. et al. Non-invasive delivery of nanoparticles to hair follicles: a perspectivefor transcutaneous immunization. Vaccine 31, 3442–3451 (2013).

12. Caux, C. et al. Regulation of dendritic cell recruitment by chemokines.Transplantation 73, S7–S11 (2002).

13. Tamoutounour, S. et al. Origins and functional specialization of macrophages andof conventional and monocyte-derived dendritic cells in mouse skin. Immunity 39,925–938 (2013).

14. Igyarto, B. Z. et al. Skin-resident murine dendritic cell subsets promote distinct andopposing antigen-specific T helper cell responses. Immunity 35, 260–272 (2011).

15. Bedoui, S. et al. Cross-presentation of viral and self antigens by skin-derivedCD1031 dendritic cells. Nature Immunol. 10, 488–495 (2009).

16. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8a1 dendritic cells incytotoxic T cell immunity. Science 322, 1097–1100 (2008).

17. Merad, M., Sathe, P., Helft, J., Miller, J. & Mortha, A. The dendritic cell lineage:ontogeny and function of dendritic cells and their subsets in the steady state andthe inflamed setting. Annu. Rev. Immunol. 31, 563–604 (2013).

18. Hashimoto, D. et al. Pretransplant CSF-1 therapy expands recipient macrophagesand ameliorates GVHD after allogeneic hematopoietic cell transplantation. J. Exp.Med. 208, 1069–1082 (2011).

19. Vander Lugt, B. et al. Transcriptional programming of dendritic cells for enhancedMHC class II antigen presentation. Nature Immunol. 15, 161–167 (2014).

20. Clark, R. A. et al. The vast majority of CLA1 T cells are resident in normal skin.J. Immunol. 176, 4431–4439 (2006).

21. Lehmann-Grube, F., Dralle, H., Utermohlen, O. & Lohler, J. MHC class I molecule-restricted presentation of viral antigen in beta 2-microglobulin-deficient mice.J. Immunol. 153, 595–603 (1994).

22. Yang, Y. et al. Focused specificity of intestinal TH17 cells towards commensalbacterial antigens. Nature 510, 152–156 (2014).

23. Goto, Y. et al. Segmented filamentous bacteria antigens presented by intestinaldendritic cells drive mucosal Th17 cell differentiation. Immunity 40, 594–607(2014).

24. Lecuyer, E. et al. Segmented filamentous bacterium uses secondary and tertiarylymphoid tissues to induce gut IgA and specific T helper 17 cell responses.Immunity 40, 608–620 (2014).

25. Gebhardt, C., Nemeth, J., Angel, P. & Hess, J. S100A8 and S100A9 in inflammationand cancer. Biochem. Pharmacol. 72, 1622–1631 (2006).

26. Mose, M., Kang, Z., Raaby, L., Iversen, L. & Johansen, C. TNFa- and IL-17A-mediatedS100A8 expression is regulated by p38 MAPK. Exp. Dermatol. 22, 476–481(2013).

27. Nowak, J. A. & Fuchs, E. Isolation and culture of epithelial stem cells. Methods Mol.Biol. 482, 215–232 (2009).

28. Oh, J., Conlan, S., Polley, E. C., Segre, J. A. & Kong, H. H. Shifts in human skin andnares microbiota of healthy children and adults. Genome med. 4, 77 (2012).

29. Martin, D. A. et al. The emerging role of IL-17 in the pathogenesis of psoriasis:preclinical and clinical findings. J. Invest. Dermatol. 133, 17–26 (2013).

AcknowledgementsThisworkwassupportedby theDivisionof IntramuralResearchofthe National Institute of Allergy and Infectious Diseases (NIAID) and by the HumanFrontier Science Program (C.W.). We thank the NIAID animal facility staff, in particularA. Gozalo (isolation of S. xylosus); D. Trageser-Cesler and C. Acevedo (NIAID gnotobioticfacility); K. Holmes, C. Eigsti and E. Stregevsky (NIAID sorting facility); K. Frank andF. Stock (MALDI-TOF analysis); B. Malissen (Langerin–GFP reporter mice); H. C. Morse(Irf82/2 mice); D. Kaplan (Langerin-DTA mice); R. Bosselut (B2m2/2 mice);S. B. Hopping (collection of human skin tissue samples); J. Oh, K. Lore, and theBrenchley laboratory (technical advice and reagents); and K. Beacht and L. Martins dosSantos for technical assistance.We also thank the Belkaid laboratory for critical readingof the manuscript.

Author Contributions S.N.,N.B., andY.B.designed the studies. S.N. andN.B.performedthe experiments and analysed the data. J.L.L. assisted with in vitro co-culturestudies and S.-J.H. with innate cell analysis and imaging. O.J.H. and C.W. providedtechnical assistance. S.C. and C.D. provided technical advice and performed 454pyrosequencing. S.C. and M.Q. analysed 454 pyrosequencing data. S.H. assisted inprocessing of human and non-human primate skin tissue samples. A.L.B. performedNanoString data analysis. J.M.B. and H.H.K. provided technical advice and skin tissuesamples from non-human primates and human patients, respectively. R.T., K.M.M. andM.M. assisted with design of dendritic cell depletion strategies. J.A.S. helped to designsequencing studies and provided guidance on bacterial isolates. S.N., N.B. and Y.B.wrote the manuscript.

Author Information 454 sequencing data are deposited in the Sequence Read Archiveunder accession number SRP039428. Reprints and permissions information isavailable at www.nature.com/reprints. The authors declare no competing financialinterests. Readers are welcome to comment on the online version of the paper.Correspondence and requests for materials should be addressed toY.B. ([email protected]).

LETTER RESEARCH

0 0 M O N T H 2 0 1 5 | V O L 0 0 0 | N A T U R E | 5

Macmillan Publishers Limited. All rights reserved©2015

Page 6: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

METHODSMice. C57BL/6 and BALB/c specific pathogen free (SPF) mice were purchased fromTaconic Farms. Germ-free C57BL/6 mice were bred at Taconic Farms and main-tained in the NIAID gnotobiotic facility. B6.SJL, C57BL/6-[KO]IL1r1 (Il1r12/2)and C57BL/6J-[KO]B2m-[KO]Abb (Abb2/2 B2m2/2) mice were obtained throughthe NIAID-Taconic exchange program. C57BL/6-Tg(Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6)2Bck/J (CSF1R GFP reporter)30, B6.129P2(C)-Ccr7tm1Rfor/J (Ccr72/2),B6.129P2(C)-Batf3tm1Kmm/J (Batf32/2) mice and their C57BL/6 wild-type con-trols were purchased from The Jackson Laboratory. B6(Cg)-Irf8tm1.2Hm/J (Irf82/2),B6.FVB-Tg(CD207-Dta)312Dhka/J (Lan-DTA), B6.129S2-Cd207tm3(DTR/GFP)Mal/J(Lan-GFP) mice were obtained from H. C. Morse (National Institute of Allergy andInfectious Diseases, NIH), D. Kaplan (University of Minnesota) and B. Malissen(Centre d’immunologie de Marseille Luminy, France), respectively. Congenic B2m2/2

(B6.129-B2mtm1Jae N12 x B6-LY5.2/Cr) mice were provided by R. Bosselut (NationalCancer Institute, NIH). Irf4fl/fl 3 Cd11cre1 mice were obtained by breeding B6.129S1-Irf4tm1Rdf/J (Irf4fl/fl) mice with B6.Cg-Tg(Itgax–Cre)1-1Reiz/J (Itgax–Cre) mice (bothstrains from The Jackson Laboratory). All mice were bred and maintained underpathogen-free conditions at an American Association for the Accreditation ofLaboratory Animal Care (AAALAC)-accredited animal facility at the NIAID andhoused in accordance with the procedures outlined in the Guide for the Care andUse of Laboratory Animals. All experiments were performed at the NIAID underan animal study proposal approved by the NIAID Animal Care and Use Com-mittee. Gender- and age-matched mice between 6 and 12 weeks of age were usedfor each experiment. When possible, preliminary experiments were performed todetermine requirements for sample size, taking into account resources availableand ethical, reductionist animal use. In general, each mouse of the different experi-mental groups is reported. Exclusion criteria such as inadequate staining or lowcell yield due to technical problems were pre-determined. Animals were assignedrandomly to experimental groups.Human and non-human primate skin tissue. Healthy human skin samples fromanonymous patients were obtained as discarded material after cosmetic surgeryaccording to a protocol approved by the Institutional Review Board of NIAID,NIH. All subjects gave informed consent. Non-human primate skin tissue wasobtained from the glabella of eight healthy rhesus (Macaca mulatta) or pigtail(Macaca nemestrina) macaques immediately post euthanasia. All monkeys werehoused and cared in accordance with AAALAC standards in AAALAC-accreditedfacilities, and all animal procedures performed according to protocols approved bythe NIAID Animal Care and Use Committee.Topical association and intradermal infection. S. epidermidis strain 42E0331 andR. nasimurium were isolated from mouse colonies. S. epidermidis strain NIHLM08732,S. epidermidis strain 42E03, S. lentus, S. xylosus (isolated from mice as previouslydescribed33), S. aureus strain NCTC 8325, C. pseudodiphtheriticum strain NIHLM0865

and R. nasimurium were cultured for 18 h in tryptic soy broth at 37 uC. P. acnes ATCC11827 was cultured for up to 72 h in tryptic soy broth in an anaerobic chamber.Bacteria were enumerated before topical application by assessing colony-formingunits using traditional bacteriology techniques and by measuring optical density(OD) at 600 nm using a spectrophotometer.

For topical association of bacteria, each mouse was associated with bacteria byapplying bacterial suspension (approximately 109 ml21, 5 ml or for Extended DataFigs 1c, d, 2d and 5f, 108 or 107 c.f.u. per ml as indicated) across the entire skin sur-face (approximately 36 cm2) using a sterile cotton swab. Application of bacterialsuspension was repeated every other day a total of four times. In experiments involv-ing topical application of various bacterial species or strains, 18-h cultures werenormalized using OD600 to achieve similar bacterial density (approximately 109 c.f.u.per ml). In some experiments, mice were infected intradermally in the ear pinnaewith 107 c.f.u. of S. epidermidis strain LM087. Mice were euthanized at differenttime points after the first topical association or intradermal inoculation with bacteria.Tissue processing. Murine tissues. Cells from the lung, the small intestine laminapropria, the skin draining lymph nodes, the ear pinnae or flank tissue were isolatedas previously described2,34. Isolation of cells from epidermal and dermal compart-ments of the ear skin tissue were performed as previously reported35.

Human and non-human primate skin. Subcutaneous fat tissue was scrapped offwith a number 10 scalpel. Skin tissue was then weighted and perfused with 100–500mlof digestion media (RPMI 1640 media supplemented with 2 mM L-glutamine, 1 mMsodium pyruvate and nonessential amino acids, 20 mM HEPES, 100 U ml21 peni-cillin, 100mg ml21 streptomycin and 0.25 mg ml21 Liberase Cl purified enzyme blend(Roche)). Perfused skin tissue was placed in 5 ml of digestion media dermal sidedown and incubated for 1 h at 4 uC. Tissue was then minced with scissors in 5 ml offresh digestion media and incubated at 37 uC with shaking. After 1 h, digestion wasstopped by adding 100ml of 0.5 M EDTA and 1 ml of fetal bovine serum. Digestedtissue was then smashed on a 70mm cell strainer to obtain a single-cell suspension.Immunofluorescence/confocal microscopy. Ear pinnae were split, fixed in 1%paraformaldehyde solution (Electron Microscopy Sciences) overnight at 4 uC and

blocked in 1% BSA 0.25% Triton X blocking buffer for 2 h at room temperature.Tissues were first stained with anti-CD8a (clone 53-6.7, eBioscience), anti-CD11c(clone N418, eBioscience) and/or rabbit anti-GFP (Life Technologies) antibodiesovernight at 4 uC, washed three time with PBS and then stained with 49,6-diamidino-2-phenylindole (DAPI, Sigma-Aldrich) for 5 min at room temperature and beforebeing mounted with ProLong Gold (Life Technologies) anti-fade reagent. Imageswere captured on a Leica TCS SP8 confocal microscope with a 403 oil objective(HC PL APO 403/1.3 oil). Images were analysed using Imaris Bitplane software.In vivo antibody administration. Mice were treated intraperitoneally (i.p.) withanti-CSF1R antibody (clone AFS98, BioXCell) or Rat IgG2a isotype control anti-body (clone 2A3, BioXCell). Treatment schedule was as follows: 2 mg of eitherantibody was injected i.p. in each mouse 5 and 3 days before the first S. epidermidistopical application (days 25 and 23); each mouse then received 0.5 mg of eitherantibodies on days 22, 21 and days 0, 2, 4, 6, 9, 11 and 13 post topical application18.Mice were analysed two weeks after the first S. epidermidis topical application (or19 days after the first antibody injection).

For CD81 T-cell depletion, mice were treated i.p. with 0.5 mg of anti-CD8 anti-body (clone 2.43) or rat IgG2b isotype control antibody (clone LTF-2, BioXCell)every 2–3 days starting at day 6 after the first topical application of S. epidermidis.For IL-17A neutralization, mice were treated i.p. with 0.5 mg of anti-IL-17A anti-body (clone 17F3, BioXCell) or mouse IgG1 isotype control antibody (clone MOPC-21, BioXCell) every 2 days starting at day 6 after the first topical application ofS. epidermidis.In vitro re-stimulation. For detection of basal cytokine potential, single-cell sus-pensions from various tissues were cultured directly ex vivo in a 96-well U-bottomplate in complete medium (RPMI 1640 supplemented with 10% fetal bovine serum(FBS), 2 mM L-glutamine, 1 mM sodium pyruvate and nonessential amino acids,20 mM HEPES, 100 U ml21 penicillin, 100mg ml21 streptomycin, 50 mM b-mer-captoethanol) and stimulated with 50 ng ml21 phorbol myristate acetate (PMA)(Sigma-Aldrich) and 5mg ml21 (mouse) or 1mg ml21 (human and monkey) iono-mycin (Sigma-Aldrich) in the presence of brefeldin A (GolgiPlug, BD Biosciences)for 2.5 h at 37 uC in 5% CO2. After stimulation, cells were assessed for intracellularcytokine production as described below.Phenotypic analysis. Murine single-cell suspensions were incubated with fluoro-chrome-conjugated antibodies against surface markers CD3 (145-2C11), CD4 (cloneRM4-5), CD8a (53-6.7), CD8b (eBioH35-17.2), CD11b (M1/70), CD11c (N418 orHL3), CD19 (6D5), CD45.2 (104), CD45R (RA3-6B2), CD49b (DX5), CD69 (H1.2F3),CD103 (2E7), DEC205 (NLDC-145), KLRG1 (2F1), MHCII (M5/114.15.2) and/or TCRb (H57-597) in Hank’s buffered salt solution (HBSS) for 20 min at 4 uC andthen washed. LIVE/DEAD Fixable Blue Dead Cell Stain Kit (Invitrogen Life Tech-nologies) was used to exclude dead cells. Cells were then fixed for 15 min at 4 uCusing 2% paraformaldehyde solution (Electron Microscopy Sciences) and washedtwice. For simultaneous Foxp3 and intracellular cytokine staining, cells were stainedwith fluorochrome-conjugated antibodies against Foxp3 (FJK-16 s), IFN-c (XMG-1.2) and IL-17A (eBio17B7) in HBSS containing 0.5% saponin (Sigma-Aldrich) for45 min at 4 uC. For detection of Langerin, cells were incubated with a fluorochrome-conjugated antibody against CD207 (929F3.01) in permeabilization buffer suppliedwith the BD Cytofix/Cytoperm kit (BD Biosciences) for 1 h at 4 uC. Each stainingwas performed in the presence of purified anti-mouse CD16/32 (93), 0.2 mg ml21

purified rat IgG and 1 mg ml21 of normal mouse serum (Jackson Immunoresearch).Staining of cells from human or non-human primate skin tissue was performedusing a similar protocol and the following antibodies against human proteins: anti-CD3 (SP34-2), anti-CD4 (L200), anti-CD8a (RPA-T8), anti-IFN-c (4S.B3) andanti-IL-17A (eBio64DEC17). All antibodies were purchased from eBioscience, BDBiosciences, Miltenyi Biotec or Dendritics. Cell acquisition was performed on anLSRII flow cytometer using FACSDiVa software (BD Biosciences) and data wereanalysed using FlowJo software (TreeStar).Analysis of skin microbiota after topical association. DNA extraction from skinand 454 pyrosequencing. Mouse ear skin samples were sterilely obtained and pro-cessed using a protocol adapted from ref. 31. For 16S rRNA amplicon sequencing,the DNA from each sample was amplified using Accuprime High Fidelity Taq poly-merase (Invitrogen Life Technologies) with universal primers flanking variableregions V1 (primer 27 F; 59-AGAGTTTGATCCTGGCTCAG-39) and V3 (pri-mer 534 R; 59-ATTACCGCGGCTGCTGG-39). For each sample, the universalprimers were tagged with unique sequences (‘barcodes’) to allow for multiplexing/demultiplexing36. PCR products were then purified using the Agencourt AmpureXP kit (Beckman Counter Genomics) and quantitated using the QuantIT dsDNAHigh-Sensitivity Assay kit (Invitrogen Life Technologies). Approximately equiva-lent amounts of each PCR product were then pooled and purified on a column fromthe MinElute PCR Purification Kit (Qiagen) into 30ml TE buffer before sequencingat the NIH Intramural Sequencing Center on a 454 GS FLX (Roche) instrumentusing titanium chemistry. Sequencing data were analysed as previously described2.

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2015

Page 7: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

Bacteria quantitation. The ear skin of topically associated or unassociated con-trol mice was swabbed with a sterile cotton swab previously soaked in tryptic soybroth. Swabs were streaked on either tryptic soy agar or blood agar plates. Plateswere then placed at 37 uC under aerobic or anaerobic conditions for 18 h. Colony-forming units on each plate were enumerated and the identity of the isolates wasconfirmed by matrix-assisted laser desorption/ionization time of flight (MALDI-TOF) mass spectrometry.Purification of skin dendritic cell subsets. Dendritic cell subsets were purifiedfrom the epidermis and dermis compartment of the ear skin tissue of unassociatedmice and S. epidermidis-associated mice at days 5 and 12 post topical application.In brief, cell suspensions obtained from the dermis compartment were incubatedwith a mixture of antibodies containing anti-CD16/32 (93), anti-CD11b (M1/70), anti-CD11c (N418), anti-CD45.2 (104), anti-CD103 (2E7), anti-MHCII (M5/114.15.2)in the presence of DAPI (Sigma-Aldrich). Cells from the epidermis compartmentwere incubated with anti-CD16/32, anti-CD45.2, anti-MHCII and DAPI. The fol-lowing two subsets of dendritic cells were sorted from the dermis by flow cytometryon a FACSAria (BD Biosciences): CD45.21 CD11c1 MHCII1 CD11b2 CD1031

cells (CD1031 dendritic cells) and CD45.21 CD11c1 MHCII1 CD11b1 CD1032

cells (CD11b1 dendritic cells). Langerhans cells were sorted from the epidermis asCD45.21 MHCII1 cells.Gene expression analysis by NanoString. The nCounter analysis system (Nano-String Technologies) was used to screen for the expression of signature genes associatedwith inflammation pathway in the different dendritic cell subsets37. Two specificprobes (capture and reporter) for each gene of interest were employed. In brief, RNAfrom each dendritic cell subset was obtained by lysing the sorted cells (103 cells perml) in RLT buffer (Qiagen) and then hybridized with the customized ReporterCodeSet and Capture ProbeSet of the Mouse Inflammation Panel including 150selected genes (NanoString Technologies), according to the manufacturer’s instruc-tions. Messenger RNA molecules were counted on a NanoString nCounter, as previ-ously described2. Data analysis was performed according to NanoString Technologiesrecommendations. mRNA counts were processed to account for hybridization effi-ciency, background noise and sample content using the R package NanoStringNormwith arguments: CodeCount 5 ‘geo.mean’, Background 5 ‘mean.2sd’, SampleContent5 ‘housekeeping.geo.mean’. Each sample profile was normalized to geometric meanof two housekeeping genes, Cltc and Pgk1. Post normalization, genes with meancounts less than 20 were disregarded and differential expression of remaining geneswas determined using a nonparametric Welch t-test with correction for multipletesting using the Benjamini–Hochberg false discovery rate (FDR) controlling pro-cedure in the multtest package in R. Based on genes with a FDR , 0.05, a heat mapwas rendered using the R package gplots.Dendritic cell and T-cell co-culture assay. CD451 CD90.21 CD8b1 or CD451

CD90.21 CD41 effector T cells (.95% purity) were sorted by flow cytometry fromthe ear skin tissue of C57BL/6 mice 2 weeks after topical association with S. epidermidisstrain LM087 or S. xylosus using a FACSAria cell sorter. For CD8b 1 and CD41

T-cell purification from the skin draining lymph nodes, single-cell suspension fromthe ear skin tissue of mice topically associated with S. epidermidis strain LM087 orS. xylosus were first magnetically enriched for CCR61 cells by positive selectionusing PE-conjugated CCR6 antibody (clone 29-2L17), anti-PE MicroBeads andMACS separation columns (Miltenyi Biotec). The enriched fraction was furtherlabelled with fluorochrome-conjugated antibodies against CD45, CD90.2, CD4 and/orCD8b and CD451 CD90.21 CD8b1 or CD451 CD90.21 CD41 were sorted (.95%purity) by flow cytometry on a FACSAria cell sorter. For splenic dendritic cell (SpDC)purification, single-cell suspensions from the spleen of congenic wild-type, B2m2/2

or Abb2/2 B2m2/2 mice were magnetically enriched for CD11c1 cells by positiveselection using CD11c MicroBeads and MACS separation columns (Miltenyi Biotec).Purified SpDCs and CD8b1 or CD41 T cells were co-cultured at a 15:1 ratio (5 3 103

CD8b1 T cells) in a 96-well U-bottom plate in complete medium for 16 h at 37 uCin 5% CO2. Brefeldin A (GolgiPlug) was added for the final 4 h of culture. SpDCwere previously incubated for 3 h with or without heat-killed S. epidermis, heat-killed S. xylosus or heat-killed S. aureus (bacteria:SpDC ratio, 500:1) and washedbefore co-culture with CD8b1 T cells. In some experiments, co-cultures of CD8b1

T cells and naive SpDC were supplemented with 5mg ml21 ultra-pure lipopoly-saccharide (LPS, InvivoGen), 100 nM N-a-palmitoyl-S-[2,3-bis(palmitoyloxy-(2RS)-propyl]-L-cysteine (Pam3Cys, InvivoGen) and/or 10 ng ml21 murine IL-1a(Peprotech). CD8b1 T cell cytokine production following co-culture was assessedby flow cytometry after intracellular cytokine staining using the following antibodies:anti-CD4, anti-CD8b, anti-CD45.1 (clone A20), anti-CD45.2, anti-TCRb, anti-IFN-cand anti-IL-17A.

Cytokine measurement. Leukocytes isolated from the skin of S. epidermidis-associated mice were cultured in 100ml of complete medium for 3 h at 37 uC in 5%CO2. FACS-purified CD8b1 T cells from the skin of S. epidermidis-associated micewere cultured overnight at 37 uC in 5% CO2 in 30ml of complete culture medium ina 96-well U-bottom plate coated with anti-CD3e (1mg ml21, clone 145-2C11, BDBiosciences) in the presence or absence of IL-1a and IL-1b (10 ng ml21 each, Pepro-tech). Supernatants were collected and levels of inflammatory cytokines were assessedusing a bead-based cytokine detection assay (FlowCytomix, eBioscience). Cytokineconcentrations were adjusted to the plated density of 2 3 104 cells in 100 or 30mlculture volume.Purification of keratinocytes. Interfollicular keratinocytes were purified by cellsorting from the ear skin tissue of unassociated mice and S. epidermidis-associatedmice at 14 days post topical application. In brief, cell suspensions obtained fromear skin tissue were incubated with the following antibodies: anti-CD16/32 (93), anti-CD45 (30-F11), anti-CD49f (eBioGoH3), anti-CD117 (2B8), anti-CD140a (APA5)and anti-Sca-1 (D7) in the presence of DAPI. Interfollicular keratinocytes weresorted by flow cytometry on a FACSAria (BD Biosciences) as CD452 CD1172

CD140a2 CD49f1 Sca-11 cells.Candida albicans infection. Eleven days after the first topical application ofS. epidermidis, mice were infected with C. albicans along the dorsal midline as pre-viously described38. In brief, C. albicans was grown in YPAD medium at 30 uC for18 h and re-suspended at 107 C. albicans yeast per 1 ml of sterile PBS. One hundredmicrolitres of this suspension was applied to sandpaper (100 grit)-treated dorsalskin to achieve an infectious dose of 106 C. albicans. At 2 days post infection a 6 mmpunch biopsy of skin was homogenized in sterile PBS containing penicillin andstreptomycin before plating on YPAD plates. Colony-forming units were countedafter culture at 30 uC for 48 h.RNA purification and quantitative PCR. Total RNA was isolated from purifiedkeratinocytes or skin tissue biopsies using the RNeasy Mini Kit (QIAGEN). Com-plementary DNA was prepared using the Omniscript Reverse Transcription Kit(QIAGEN) according to the manufacturer’s instructions. Quantitative real-timePCR was performed on a Bio-Rad iCycler using the iQ SYBR Green Supermix(BioRad) and predesigned Quantitect primers (QIAGEN) specific for the follow-ing genes: Hprt, S100a8 and S100a9.Histology. Mice were euthanized 7 days after topical application or intraperitonealinjection of S. epidermidis LM087 in the ear. Unassociated mice were used as con-trols. The ears from each mouse were removed and fixed in PBS containing 10%formalin. Paraffin-embedded sections were cut at 0.5 mm, stained with haematox-ylin and eosin and examined histologically.Statistics. Data are presented as mean 6 standard error of the mean or mean 6

standard deviation. Group sizes were determined based on the results of prelim-inary experiments. No statistical method was used to predetermine sample size.Mice were assigned at random to groups. Mouse studies were not performed in ablinded fashion. Generally, each mouse of the different experimental groups isreported. Statistical significance was determined with the two-tailed unpaired Student’st-test, under the untested assumption of normality. Within each group there was anestimate of variation, and the variance between groups was similar. All statisticalanalysis was calculated using Prism software (GraphPad). Differences were consid-ered to be statistically significant when P , 0.05.

30. Burnett, S. H. et al. Conditional macrophage ablation in transgenic miceexpressing a Fas-based suicide gene. J. Leukoc. Biol. 75, 612–623 (2004).

31. Grice, E. A. et al. Longitudinal shift in diabetic wound microbiota correlates withprolonged skin defense response. Proc. Natl Acad. Sci. USA 107, 14799–14804(2010).

32. Conlan, S. et al. Staphylococcus epidermidis pan-genome sequence analysisreveals diversity of skin commensal and hospital infection-associated isolates.Genome Biol. 13, R64 (2012).

33. Gozalo, A. S. et al. Spontaneous Staphylococcus xylosus infection in mice deficientin NADPH oxidase and comparison with other laboratory mouse strains. J. Am.Assoc. Lab. Anim. Sci. 49, 480–486 (2010).

34. Spencer, S. P. et al. Adaptation of innate lymphoid cells to a micronutrientdeficiency promotes type 2 barrier immunity. Science 343, 432–437 (2014).

35. Helft, J. & Merad, M. Isolation of cutaneous dendritic cells. Methods Mol. Biol. 595,231–233 (2010).

36. Lennon, N. J. et al. A scalable, fully automated process for construction ofsequence-ready barcoded libraries for 454. Genome Biol. 11, R15 (2010).

37. Geiss, G. K. et al. Direct multiplexed measurement of gene expression withcolor-coded probe pairs. Nature Biotechnol. 26, 317–325 (2008).

38. Rittig,M.G. et al.Coiling phagocytosis of trypanosomatids and fungal cells. Infect.Immun. 66, 4331–4339 (1998).

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2015

Page 8: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

0

20

40

60

80

IL-13

0

10

20

30

40

50

IL-1β

Ctrl 24h48

h0

200

400

600

800

IL-1α

0

1000

2000

3000

4000

IL-6

0

10

20

30

40

50

IL-2

0

20

40

60

80

100

IL-27 IFN-γ

0

50

100

150

200

TNF-α

0

5

10

15

20

25

GM-CSF

(pg/

ml)

0

10

20

30

40

S. epi.Ctrl 24

h48

h

S. epi.Ctrl 24

h48

h

S. epi.Ctrl 24

h48

h

S. epi.Ctrl 24

h48

h

S. epi.Ctrl 24

h48

h

S. epi.Ctrl 24

h48

h

S. epi.Ctrl 24

h48

h

S. epi.Ctrl 24

h48

h

S. epi.

f

14

8

3

34

12

2

Topical Intradermal

S. epidermidis (day 7)

Control

IL-1

7A

IFN-γ

g h

���������

���

Ctrl

Day 7

Day 14

Ctrl

Day 7

Day 14

30

24

12

18

6

0TC

Rβ+

IL-1

7A+

(%)

5060

40

2030

100T

CRβ+

IFN

-γ+

(%)

S. epi. topicalS. epi. intrad.

IFN-γ+ Teff cells IL-17A+ Teff cells

30

2010

1

40

CD

11bhi

Ly6

Ghi (

%)

0

500

321

1000

0

15002000

CD

11bhi

Ly6

Ghi (

x103 )

Ctrl

Topica

l

Intrad.

Ctrl

Topica

l

Intrad.

�9

6

3

0CD

11bhi

Ly6

Chi (

%)

100

20

10

200

0

300

CD

11bhi

Ly6

Chi (

x103 )

Ctrl

Topica

l

Intrad.

Ctrl

Topica

l

Intrad.

Neutrophils Monocytese

���

21

14

7

0CD

4+ F

oxp3

+ (x

103 )

Ctrl

S. epi.

Ctrl

S. epi.

60

40

20

0

80

CD

4+ F

oxp3

+ (%

)

a

��� NS NS6

4

2

0

90

60

30

0

90

60

30

0

TC

Rβ+

IL-1

7A+

(x10

3 )

Skin

Lung Gut

Ctrl

S. epi.

Ctrl

S. epi.

Ctrl

S. epi.

b

���

Ctrl

S. epi.

2

1.5

1

0

0.5

Ear Flank

010202090

160230

300

c.f.u

./ear

0

5

10

15

20

TC

Rβ+

IFN

-γ+

(x10

2 )

TC

Rβ+

IL-1

7A+

(x10

2 )

0

2102030405060

���

����

��

��

��

S. epi. Ctrl 107 108 109

S. epi. Ctrl 107 108 109

S. epi. Ctrl 107 108 109

IFN-γ+ Teff cells IL-17A+ Teff cellsc d

Extended Data Figure 1 | Assessment of Foxp31 regulatory T cells andcytokine production by effector T cells after S. epidermidis topicalapplication and/or intradermal inoculation. a, Frequencies and absolutenumbers of skin regulatory (CD451 TCRb1 CD41 Foxp31) T cells inunassociated mice (Ctrl, n 5 4) and mice associated with S. epidermidis (n 5 4)at day 14 post first topical application. b, Absolute numbers of effector T cellsproducing IL-17A after PMA/ionomycin stimulation in the skin (ear pinnaeand flank), the lung or the small intestine lamina propria (gut) at day 14 posttopical association (Ctrl, n 5 4–5; S. epi., n 5 4–5). c, d, Enumeration ofcolony-forming units and absolute numbers of effector T cells producing IFN-cor IL-17A (PMA/ionomycin) from the skin 2 weeks post application withdifferent doses (107, 108 or 109 c.f.u. per ml) of S. epidermidis (n 5 4 per group).e, Frequencies and absolute numbers of neutrophils and monocytes in the skin

of mice 14 days after the first topical application or intradermal inoculationwith S. epidermidis (n 5 4 per group). f, Assessment of cytokine production(mean 6 s.e.m., n 5 3 per time point) by leukocytes from the ear skin tissue24 and 48 h after topical association with S. epidermidis. Unassociated micewere used as controls. No significant amounts of IL-4, IL-5, IL-17A, IL-18,IL-21 or IL-22 could be detected at the time of analysis. g, IFN-c and IL-17Aproduction by skin effector T cells in mice 7 days after S. epidermidistopical application or intradermal inoculation. h, Frequencies of IFN-c andIL-17A-producing effector T cells in the skin of mice 7 and 14 days after the firsttopical application or intradermal inoculation of S. epidermidis (n 5 4–5mice per group). All results shown are representative of 2–3 experiments withsimilar results. *P , 0.05; **P , 0.01; ***P , 0.001; NS, not statisticallysignificant as calculated by Student’s t-test.

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2015

Page 9: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

0

4060

20

200

400

600

c.f.u

./ear

Ctrl

S. epi.

S. xylo

.

C. pse

udo.

P. acn

es

S. aureus

S. epi 42E03

S. lentus

a

R. nasi.

S. epi. 42E03 S. lentus R. nasi.

IL-1

7A

IFN-γ

Control7

0.9

20

3

12

0.6

14

0.5

Ctrl

S. epi. 4

2E03

S. lentus

IFN-γ+ Teff cells

Ctrl

S. epi. 4

2E03

S. lentus

TC

Rβ+

IFN

-γ+

(x10

2 )

4

3

1

0

5

2

TC

Rβ+

IL-1

7A+

(x10

2 )

20

15

50

25

10

30

IL-17A+ Teff cells

b

R. nasi.

R. nasi.

S. epi. 42E03 S. lentus R. nasi.

CD

CD4

Control

5

53

13

50

5

46

6

50

c

TC

Rβ+

CD

8β+

(x10

2 )

129

30

15

6

1821

CD8β+ T cells

Ctrl

S. epi. 4

2E03

S. lentus

R. nasi.

01234567

CD

8β+

IL-1

7A+

(x10

2 )

0

15

152535

45C

D8β

+IF

N-γ

+(x

102 )

���

��

��

�� ��

��

CD8β+ T cellsd

S. epi. Ctrl 107 108 109

S. epi. Ctrl 107 108 109

0

3

6

9

TC

Rβ+

CD

8β+

(x10

3 )

NS

e

SPF

GF

Control S. epi.

4 24

47 57

1 17

52 75

CD4

CD

��

65

4321

0SPF GFT

CRβ+

CD

8β+

(x10

3 )

ControlS. epi.

f

��

���

3210

15

CD

8β+

IFN

-γ+

(x10

2 )

35

55

43210

10

18

26

4

CD

8β+

IL-1

7A+

(x10

2 )

Ctrl

S. epi.

Ctrl

S. epi.

C57Bl/6 BALB/c

Ctrl

S. epi.

Ctrl

S. epi.

C57Bl/6 BALB/c

gControl S. epi.+

CD4

CD

7 15

59

±1 ±4

±1 65±9

0

50

100

150

n.d.

c.f.u

./ear

0

1

2

3 ��

Ctrl

S. epi.+ Ctrl

S. epi.+

TC

Rβ+

CD

8β+

(x10

2 )

hS. e

pi. 1 m

l

S. epi. 5

ml

Ctrl

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2015

Page 10: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

Extended Data Figure 2 | Assessment of CD81 T-cell responses in the skinof specific pathogen-free and germ-free mice after topical applicationwith skin commensals. a, Mice were left unassociated (Ctrl, n 5 5) or topicallyassociated with S. epidermidis human isolate (n 5 5), S. xylosus (n 5 3),S. epidermidis murine isolate (S.epi 42E03, n 5 2), S. lentus (n 5 2),R. nasimurium (n 5 2), S. aureus (n 5 5), C. pseudodiphtheriticum (n 5 3) orP. acnes (n 5 3). Quantification of colony-forming units from the ears aftertopical application is shown 2 weeks after first association. b, Frequencies andnumbers of effector (CD451 TCRb1 CD41 Foxp32) T cells producing IFN-cor IL-17A after PMA/ionomycin stimulation in the skin of mice from a atday 14 post first topical application. Bar graphs represent the mean value fromtwo mice. c, Frequencies of skin CD41 and CD8b1 effector T cells in mice froma at day 14 post first topical application. d, Absolute numbers of IFN-c- and IL-17A-producing CD8b1 effector T cells in the skin of unassociated (Ctrl) miceor mice associated with different doses (107, 108 or 109 c.f.u. per ml) of S.epidermidis (n 5 4 per group). e, Absolute numbers of skin CD8b1 effector T

cells in unassociated (Ctrl, n 5 3) mice or mice associated with 1 ml (n 5 5) or5 ml (n 5 5) of a suspension (109 c.f.u. per ml) of S. epidermidis. f, Flowcytometric assessment of the frequencies of CD41 and CD8b1 effector T cellsand absolute numbers of CD8b1 effector T cells in SPF (n 5 3 per group) andgerm-free (GF, n 5 4 per group) mice 2 weeks after S. epidermidis topicalapplication. g, Absolute numbers of IFN-c- and IL-17A-producing CD8b1

effector T cells in the skin of unassociated (Ctrl) or S. epidermidis-associatedC57BL/6 and BALB/c mice at 14 days post first topical application (n 5 5 pergroup). For d–g, all results shown are representative of 2–3 independentexperiments with similar results. *P , 0.05; **P , 0.01; ***P , 0.001; NS, notstatistically significant as calculated with Student’s t-test. h, Quantification ofcolony-forming units from the ears of adult mice born from S. epidermidis-associated (S. epi1, n 5 3) or unassociated (Ctrl, n 5 3) breeder pairs. Flowplots and bar graphs (mean 6 s.e.m.) illustrate the frequencies of CD41 andCD8b1 effector T cells and absolute numbers of CD8b1 effector T cells,respectively. n.d., not detected; **P , 0.01 as calculated with Student’s t-test.

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2015

Page 11: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

0

10

2030

40C

D8β

+ (x

104

/ gra

m)

IL-17A+

IFN-γ+

NHPc

��180

120

60

0IL

-17A

(pg

/ml)

Ctrl+ IL

-1

IL-17Ad

5

15

epidermis

dermis

CD8α CD207 (GFP) MergeDAPI

b

Dermis Epidermis

20

60

80

40

100

0

% o

f tot

al T

CRβ+

CD

8β+

a

Ctrl

S. epi

Extended Data Figure 3 | CD81 T cells accumulate preferentially in theepidermidis after topical application of S. epidermidis. a, Proportion ofeffector (CD451 TCRb1 Foxp32) CD8b1 T cells in the epidermal and dermalcompartments of the ear skin tissue 2 weeks after the first S. epidermidis topicalapplication. b, Representative imaging volume projected along the x axis ofears from Langerin–GFP reporter mice at 14 days post first topical applicationwith S. epidermidis. Scale bars, 30mm. c, Numbers of CD31 CD8b1 cells

producing IFN-c or IL-17A (after PMA/ionomycin stimulation) from normalnonhuman primate (NHP) skin (n 5 8). d, Assessment of IL-17A productionin the supernatant of CD8b1 T cells purified from the skin of mice topicallyassociated with S. epidermidis and cultured overnight in presence of anti-CD3ealone (Ctrl) or with IL-1a and IL-1b (1 IL-1). Bars represent the meanvalue 6 s.e.m. (n 5 3, **P , 0.01 as calculated with Student’s t-test). Resultsshown in a, c and d are representative of 2–3 experiments with similar results.

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2015

Page 12: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

SS

C-A

FS

C-W

SS

C-W

SS

C-A

SS

C-A

CD

11c

CD

11b

FSC-A FSC-A SSC-A DAPI CD45 MHC-II CD207

Leukocytes FSC singlet SSC singlet Live cellsHematopoietic

cellsDendritic cells DC subsets

LC

CD103DC

CD11bDC

a

CD

11b

CD207

Control S. epi

2754

8.3

2654

8.3

b

��

80

60

40

20

0

Cel

ls (

x103 )

LC

CD103 DC

CD11b D

C

Lan-DTA

��

NS

NS50

30

10

0246

Cel

ls (

x103 )

Irf8 ─/─

LC

CD103 DC

WTLan-DTA

WTIrf8 ─/─

��

NS

Batf3 ─/─

60

40

2010

0

5Cel

ls (

x103 )

LC

CD103 DC

WTBatf3 ─/─

CD11b D

C

CD11b D

C

c

���

��50

40

30

20

0

10

10

8

6

4

0

2

32

24

16

0

8

LC CD103 DC CD11b DC

Cel

ls (

x103 )

Rat IgG

αCSF1R

Rat IgG

αCSF1R

Rat IgG

αCSF1R

αCSF1R

����32

16

24

8

0

CD

8α+

DE

C20

5+ (x

104 )

Batf3 ─/─

WT

12

6

9

3

0

Batf3 ─/─

WT

Spleen dLN

d

WT Irf8 ─/─ 30 50

7.6

21 77

0.2

CD

11b

CD207

��18

12

6

0

��

8

6

4

2

0

��1.2

0.9

0.6

0.3

0

Irf8 ─/─

WT

TC

Rβ+

CD

8β+

(x10

3 )

CD

8β+

IL-1

7A+

(x10

3 )

CD

8β+

IFN

-γ+

(x10

3 )

Irf8 ─/─

WT

Irf8 ─/─

WT

e�600

500

400

300

200

IL-1

(pg

/ml)

Rat IgG

αCSF1R

IL-1f

����

20

10

5

0

25

15

TC

Rβ+

CD

8β+

(%)

NS

40

20

10

0

50

30

CD

8β+

IFN

-γ+

(%)

8

4

2

0

10

6

CD

8β+

IL-1

7A+

(%)

Irf4 fl/fl x Cd11c cre+

Control

IL-1

7A

IFN-γ

Control7.4

27

0.6

37

Irf4 fl/fl xCd11c cre+

g

Extended Data Figure 4 | Depletion strategies for the different subsets ofskin dendritic cells. a, Gating strategy for various dendritic cell subsets in theskin. Cells are first gated on live CD451 CD11c1 MHCII1. Subsets of dendriticcells are then defined as follows: Langerhans cells (LC) are gated on CD11b1

CD207(Langerin)1 cells, CD1031 dendritic cells (CD103 DC) on CD11b2

CD2071 cells and CD11b1 dermal dendritic cells (CD11b DC) on CD11b1

CD2072 cells. b, Comparative assessment by flow cytometry of Langerhanscell, CD103 DC and CD11b DC in the ear skin of unassociated mice (control)and mice first topically associated with S. epidermidis 2 weeks earlier.c, Absolute numbers of Langerhans cell, CD103 DC and CD11b DC 2 weeksafter the first topical application of S. epidermidis in wild-type (WT, n 5 3),Langerin–DTA (Lan–DTA, n 5 3), Batf32/2 (n 5 3) or Irf82/2 (n 5 3) miceand in mice treated with anti-CSF1R (n 5 3) or isotype control (rat IgG, n 5 3)antibodies. d, Absolute numbers of CD11chi MHCII1 CD81 DEC2051

dendritic cells in the spleen and the skin draining lymph node (dLN) ofwild-type (n 5 5) and Batf32/2 (n 5 6) mice. e, Phenotypic analysis of CD451

MHCII1 CD11c1 cells by flow cytometry and absolute numbers of effector(CD451 TCRb1 Foxp32) CD8b1 T cells and IL-17A- or IFN-c-producingCD8b1 T cells in wild-type (n 5 3) and Irf82/2 (n 5 3) mice 2 weeks after thefirst topical application of S. epidermidis. f, Assessment of IL-1 production byleukocytes from the ear skin tissue of S. epidermidis-associated mice treatedwith anti-CSF1R (n 5 4) or isotype control (rat IgG, n 5 5) antibodies.g, Frequencies of total and IFN-c- or IL-17A- producing CD8b1 effector T cellsin S. epidermidis-associated Irf4 fl/fl 3 CD11ccre1 (n 5 3) and littermate control(n 5 3) mice. All data shown in this figure are representative of 2–3experiments with similar results. *P , 0.05; **P , 0.01; ***P , 0.001; NS, notstatistically significant as calculated with Student’s t-test.

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2015

Page 13: Commensal-dendritic-cell interaction specifies a unique ..._2015.pdf · Commensal–dendritic-cell interaction specifies a unique protective skin immune signature Shruti Naik 1,2

Control S. epi

Skin

dLNIL-1

7A

IFN-γ

1

1

0

1

1

0

7

2

8

2

1

16

����

���� ����18

15

12

0

3

6

9

30

25

20

0

5

10

15

CD

4+ IF

N-γ

+ (%

)

CD

4+ IL

-17A

+ (%

)

IFN-γ+ cells IL-17A+ cells

Ctrl

S. epi

Ctrl

S. epi

Skin CD4+

0

1

2

3

CD

4+ IF

N-γ

+ (%

)

12

0

3

6

9

CD

4+ IL

-17A

+ (%

)

IFN-γ+ cells IL-17A+ cells

Ctrl

S. epi

Ctrl

S. epi

dLN CD4+

��������

c d

b

�����

Ctrl

S. epi

Ctrl

S. epi

0

2

4

6C

D8β

+ IF

N-γ

+ (%

)

0

9

18

27

45

36

CD

8β+

IL-1

7A+

(%)

IFN-γ+ cells IL-17A+ cells

dLN CD8β+

4

0

0 23

25

4 3

14

123

IL-1

7A

IFN-γ

Control S. epi

Abb ─/─

x B2m ─/─ WTWT

�� ���

36

27

18

0

9

45

20

10

0

30

CD

8β+

IFN

-γ+

(%)

CD

8β+

IL-1

7A+

(%)

IFN-γ+ cells IL-17A+ cells

Ctrl

S. epi

Ctrl

S. epi

�����

Abb / B2m

S. epi

a

Abb / B2m

S. epi

Abb / B2m

S. epi

Abb / B2m

S. epi

����

����

����

����

����

����16

0

4

8

12

CD

4+ IF

N-γ

+ (%

) 50

40

30

0

10

20

CD

4+ IL

-17A

+ (%

)

0

5

10

15

CD

4+ IL

-17A

+ (%

)

0

0.5

1

CD

4+ IF

N-γ

+ (%

)

Ctrl

S. xylo

Abb / B2m

S. xylo

IFN-γ+ cells IL-17A+ cells

Skin CD4+

IFN-γ+ cells IL-17A+ cells

dLN CD4+e

Ctrl

S. xylo

Abb / B2m

S. xylo Ctrl

S. xylo

Abb / B2m

S. xylo Ctrl

S. xylo

Abb / B2m

S. xylo

01234567

Fol

d in

crea

se

over

con

trol

01234567

S100a8 S100a9

S. epi 107 108 109

S. epi 107 108 109

f

Extended Data Figure 5 | Commensal-driven CD41 and CD81 T-cellresponses in the skin tissue and the skin draining lymph nodes are specificfor commensal antigens. a, Frequencies of IFN-c- or IL-17A-producingCD8b1 T cells in overnight co-cultures of splenic dendritic cells (SpDC)and CD8b1 T cells purified from the skin draining lymph node (dLN) of micefirst topically associated with S. epidermidis 2 weeks earlier. b, Frequenciesof IFN-c- and IL-17A-producing CD8b1 T cells in overnight co-cultures ofSpDC and CD8b1 T cells purified from the skin of mice 14 days after the firstS. epidermidis application. Dendritic cells were purified from either wild-type(WT) or Abb2/2 B2m2/2 mice. c, d, Frequencies of IFN-c- and IL-17A-producing CD41 T cells in overnight co-cultures of SpDC and CD8b1 T cellspurified from the skin ear tissue or the skin dLN of mice 14 days after thefirst S. epidermidis application. For a, b and d, Ctrl, naive SpDC; S. epi,

SpDC 1 heat-killed S. epidermidis; Abb/B2m S. epi, Abb2/2 B2m2/2

SpDC 1 heat-killed S. epidermidis. e, Frequencies of IFN-c- and IL-17A-producing CD41 T cells in overnight co-cultures of SpDC and CD8b1 T cellspurified from the skin ear tissue or the skin dLN of mice 14 days after thefirst S. xylosus application. Ctrl, naive SpDC; S. xylo, SpDC 1 heat-killedS. xylosus; Abb/B2m S. xylo, Abb2/2 B2m2/2 SpDC 1 heat-killed S. xylosus.All data shown in a–d are representative of three independent experiments.Graph bars represent the mean 6 standard deviation of triplicate cultures.**P , 0.01, ***P , 0.0001, ****P , 0.0001 as calculated with Student’s t-test.f, S100a8 and S100a9 gene expression in dorsal skin biopsies of mice associatedwith different doses (107, 108 or 109 c.f.u. per ml) of S. epidermidis 2 weeksafter the first topical application (n 5 4 per group). Data are expressed as foldincrease over gene expression in unassociated control mice.

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2015