International Immunology International Immunology Advance Access published May 12, 2014

Regulation of intestinal health and disease by Innate lymphoid cells

International Immunology

r Fo

Manuscript ID: Manuscript Type: Date Submitted by the Author:

Keywords:

Invited Review

02-May-2014

Sonnenberg, Gregory; University of Pennsylvania,

Pe

Complete List of Authors:

INTIMM-14-0065.R1

mucosal immunology, inflammatory bowel disease, intestinal homeostasis, innate lymphoid cells

er ew

vi

Re

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

Journal:

Page 1 of 22

1

International Immunology

Regulation of intestinal health and disease by Innate lymphoid cells

2 3

Gregory F. Sonnenberg

4 5

Division of Gastroenterology, Department of Medicine, and Institute for Immunology, Perelman School of

6

Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA

7 CORRESPONDENCE Gregory F. Sonnenberg, [email protected]

r Fo

9 10

Running title: Intestinal innate lymphoid cells

11 Word count: 3,241

13 Figures count: 2

15

er

14

Pe

12

Re

16

Keywords: Intestinal homeostasis, mucosal immunology, inflammatory bowel disease, innate lymphoid

17

cell.

vi

18 19

ew

20 21 22 23 24 25 26 27 28 29 1

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

8

International Immunology

Page 2 of 22

Abstract

2

Innate lymphoid cells (ILCs) are a recently appreciated immune cell population that is constitutively found

3

in the healthy mammalian gastrointestinal (GI) tract and associated lymphoid tissues.

4

studies have revealed that alterations in ILC populations are associated with GI disease in patients, such

5

as inflammatory bowel disease (IBD), HIV infection and colon cancer, suggesting a potential role for ILCs

6

in either maintaining intestinal health or promoting intestinal disease. Mouse models identified that ILCs

7

have context-dependent protective and pathologic functions either during the steady state, or following

8

infection, inflammation or tissue damage. This review will discuss the associations of altered intestinal

9

ILCs with human GI diseases, and the functional consequences of targeting ILCs in mouse models.

10

Collectively, our current understanding of ILCs suggests that the development of novel therapeutic

11

strategies to modulate ILC responses will be of significant clinical value to prevent or treat human GI

12

diseases.

17

vi

18

Re

16

19

ew

20 21 22 23 24 25 26 27 28 29 2

Translational

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

15

er

14

Pe

13

r Fo

1

Page 3 of 22

International Immunology

2

Innate lymphoid cells (ILCs) are a family of innate immune cells that critically contribute to the state of

3

health or disease in the mammalian gastrointestinal (GI) tract. The development, heterogeneity, plasticity

4

and regulation of ILCs have been extensively reviewed elsewhere (1-7). In brief, ILCs develop from

5

lymphoid precursors in a process regulated by the common γ-chain cytokine receptor and the transcription

6

factors Id2, TCF1 and GATA3. Mature ILCs express the IL-7 receptor (CD127), but lack surface markers

7

associated with other immune cell lineages, including T cells, B cells, granulocytes and myeloid cells.

8

Further, ILCs can be divided into three groups based on transcriptional regulation. Group 1 ILCs (ILC1)

9

constitutively express T-bet but not RORγt, group 2 ILCs (ILC2) constitutively express GATA3 but not

10

RORγt, and group 3 ILCs (ILC3) constitutively express RORγt. There are numerous shared regulatory and effector pathways that are associated with each group

12

of ILCs. For example, ILC1 respond to IL-12 and IL-18 to produce IFN-γ and TNF-α, ILC2 respond to IL-

13

25, IL-33 and TSLP to produce IL-5, IL-9, IL-13 and amphiregulin, and ILC3 respond to IL-1β and IL-23 to

14

produce IL-17 and IL-22.

15

populations. Currently, ILC1 encompass classical NK cells (which will not be discussed in this review) and

16

recently identified ILC1 that lacks expression of eomesodermin and cytolytic potential, and can be

17

heterogeneous in expression of natural cytotoxicity receptors (NCRs), such as NKp46 and NKp44. ILC3

18

encompass CCR6 cells that most closely resemble fetal lymphoid tissue inducer (LTi) cells and CCR6

19

cells that can co-express T-bet and NCRs.

20

heterogeneity in groups of ILCs remain unclear and are active areas of investigation.

er

Pe

11

However, significant heterogeneity has been found in ILC1 and ILC3

vi

Re

+

-

ew

The lineage relationships, plasticity and significance of

21

Analyses of ILCs in humans and mice have revealed that the GI tract and associated lymphoid

22

tissues constitutively contain populations of ILCs under homeostatic conditions. Further, studies in patient

23

populations with diseases that affect the GI tract, including inflammatory bowel disease (IBD), HIV and

24

cancer, have revealed significant alterations in either the composition or the functional potential of ILC

25

populations. Mouse models have identified several critical pathways by which ILCs can directly modulate

26

intestinal homeostasis, immunity, inflammation and tissue repair, suggesting that further interrogation of

27

ILCs could provoke the development of novel preventative and therapeutic strategies for GI diseases. The

28

focus of this review will be to discuss data supporting the idea that that alterations in ILC populations occur

3

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

Introduction

r Fo

1

International Immunology

Page 4 of 22

1

in the intestine of healthy versus diseased humans, and to highlight functional studies in mice identifying

2

context-dependent protective and pathologic roles for intestinal ILCs.

3 4

Human innate lymphoid cells in gastrointestinal health and disease

5

Subsets of ILCs were first discovered in the intestinal tract and associated lymphoid tissues of fetal and

6

adult donors.

7

identified—in the tonsils, Peyer’s patches and lamina propria of adult donors—that constitutively

8

expressed NKp44, RORC (which encodes RORγ isoforms) and IL-22 (8). Similarly, a report by Cupedo

9

and Spits identified a population of NCR CD127 innate immune cells present in the fetal intestine and

10

mesenteric lymph nodes that expressed IL-17 and IL-22, and further characterized a post-natal

11

development of NKp44 CD127 cells that express IL-22 and RORC in human tonsils (9). Subsequent

12

studies suggested that IL-22-expressing NKp44 cells might be a subset of immature NK cells (10), and

13

highlighted many questions regarding the developmental and lineage relationships of innate IL-22- and IL-

14

17-expressing cell types in the GI tract and associated lymphoid tissues of humans.

In a seminal study by Colonna and colleagues, a subset of innate immune cells was

+

+

er

Pe

+

Since those initial reports, more recent studies have demonstrated that these cells are indeed a

16

novel innate immune cell population, now termed ILC3, that are distinct from classical NK cells (11) and

17

uniquely regulated by environmental and inflammatory stimuli (12-15).

18

produce IL-22 and IL-17, two cytokines that induce expression of anti-microbial peptides and inflammatory

19

mediators (16), provoked the hypothesis that they were critical regulators of intestinal immunity and

20

inflammation.

The ability of ILC3 to rapidly

ew

vi

Re

15

21

Consistent with this, ILC3 were found to be the dominant source of IL-22 in the intestine and

22

associated lymphoid tissue of healthy human donors (17) and, as discussed below, alterations in ILC3

23

populations were identified in multiple infectious, inflammatory and metabolic diseases affecting the GI

24

tract. For example, it was reported by multiple groups that IL-22- and IL-17-expressing ILC3 are depleted

25

from the GI tract of non-human primates following infection with a highly pathogenic strain of simian

26

immunodeficiency virus (SIV) and induction of tissue damage in the intestine (18-20), a pathway critically

27

linked to the progression of AIDS. Further, the presence of IL-22-producing ILC3 populations has been

28

correlated with preserved integrity of the GI tract of HIV-infected individuals (21,22), suggesting a

29

beneficial role for ILC3 and IL-22 in maintaining intestinal health during chronic viral infection.

4

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo +

-

Page 5 of 22

International Immunology

1

IBD patients have also been reported to exhibit significant alterations in intestinal ILCs. IL-22-

2

producing NKp44 ILC3 were found to be reduced in patients with Crohn’s disease (23,24); however,

3

these ILC3 were preserved in ankylosing spondylitis patients exhibiting subclinical intestinal inflammation

4

(25), suggesting that this population of cells may play a role in limiting the development of clinical GI

5

disease. This loss of a potentially ‘protective’ ILC3 population in IBD was also associated with an increase

6

in potentially ‘pathologic’ ILCs (23,24,26). Powrie and colleagues identified an expansion of CD127 IL-

7

23-responsive, IL-17-producing ILC3 in the intestinal lamina propria of Crohn’s disease patients (26). In a

8

seminal study by Spits and colleagues, a subset of IFN-γ-producing ILC1 were found to be increased in

9

Crohn’s disease patients relative to non-inflamed controls, and further expansion of human ILC1 could be

10

driven by experimental intestinal damage in a humanized mouse model (24). Colonna and colleagues

11

also identified a population of T-bet IFN-γ-producing ILC1 residing in the intra-epithelial cell compartment,

12

which accumulated in Crohn’s disease patients (27). Collectively, these reports suggest that there are

13

significant alterations in the composition or functional potential of intestine-resident ILCs during chronic

14

infection or tissue inflammation in humans.

+

+

er

Pe

The mechanisms that result in dysregulated human ILC populations in these chronic diseases

Re

16

currently remain unclear.

17

pathologic ILCs may occur through altered lineage plasticity or dysregulated exposure to regulatory

18

cytokines or microbial stimuli (12-14,24,26,28,29). Further, a recent report identified that IL-22 ILC3 are

19

enriched in tumors of patients with colorectal cancer (30), suggesting that alterations in ILCs may be

20

present in a broad spectrum of human GI diseases. Finally, although the presence of ILC2 in the intestine

21

and associated lymphoid tissues has been identified in non-diseased fetal and adult donors (31), it

22

remains poorly defined whether they are dysregulated in the context of intestinal diseases. However, one

23

recent report identified an accumulation of IL-13-producing cells that resemble ILC2 in fibrotic Crohn’s

24

disease patients (32), suggesting an involvement in pathologic collagen deposition. In future analyses of

25

ILCs in the human intestine, it will be important to undertake extensive phenotypic and functional

26

characterization to fully elucidate groups of ILCs in the healthy intestine and alterations that may occur in

27

infectious, inflammatory or metabolic diseases.

Several studies suggest the loss of protective ILCs and accumulation of

ew

vi

28 29

+

The role of ILCs in maintaining intestinal health

5

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

15

+

International Immunology

Page 6 of 22

1

Functional studies in mice have revealed that ILCs can play a significant role in maintaining intestinal

2

health by promoting immunity to pathogens, limiting inappropriate inflammatory responses to commensal

3

bacteria or dietary antigens, or mediating repair following tissue damage. This is accomplished through

4

expression of a number of factors that can influence stromal, epithelial, myeloid, granulocyte and adaptive

5

immune cell responses (Fig. 1).

6

8

ILCs can be a rapid source of protective cytokines following initial exposure to a variety of pathogens. For

9

example, a recently described ILC1 population was found to be an important innate source of IFN-γ and

10

TNF-α following oral infection with Toxoplasma gondii, which promoted the recruitment of inflammatory

11

monocytes and control of parasite replication (33). ILC2 in the small intestine are critical for promoting

12

tissue immunity via rapid production of IL-5 and IL-13, which induces intestinal goblet cell responses and

13

expulsion of intestinal nematodes, such as Nippostrongylus brasiliensis (34,35). Finally, ILC3 are a rapid

14

source of IL-22 and IL-17, which induce intestinal epithelial cell expression of anti-microbial peptides and

15

innate immunity to extracellular bacteria, such as Citrobacter rodentium (8,16,36,37). Therefore, ILCs

16

provide rapid cytokine production that is critical to control pathogen replication and clearance early

17

following infection.

er

Pe

Re

As previously reviewed (38), ILCs also play a significant role in modulating adaptive immunity in

19

the intestine through either indirect interactions with stromal cells, or direct interactions with adaptive

20

immune cells. Studies by Mebius and colleagues identified a subset of ILC3, termed lymphoid tissue

21

inducer (LTi) cells, that interacts with stromal cells to induce secondary lymphoid organogenesis prior to

22

birth (39,40). Following birth, a complex interplay between mammalian hosts and commensal bacteria

23

facilitate the maturation of intestinal cryptopatches to isolate lymphoid follicles by RORγt ILC3 (41,42).

ew

vi

18

+

24

ILC3 promote the development of GALT through expression of lymphotoxin (LT) and interactions

25

with stromal cells, allowing the recruitment of immune cells to discrete organized locations (40). The

26

formation of secondary lymphoid tissues in the intestine by ILC3 is critical for orchestrating optimal

27

adaptive immunity. For example, regulation of these functions of ILC3 by dietary retinoids significantly

28

impacts the magnitude of protective adaptive immune cell responses to mucosal viruses (43). Expression

29

of surface-bound LTα1β2 or soluble LTα3 by ILC3 is also essential for generating host-protective IgA in the

6

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

ILCs promote intestinal immunity to pathogens

r Fo

7

Page 7 of 22

International Immunology

1

GI tract in a T cell-independent and T cell-dependent manner, respectively (44,45). ILC2 have recently

2

been described to regulate eosinophil homeostasis in the intestine through constitutive production of IL-5

3

(46). While the homeostatic functions of intestinal eosinophils are poorly understood, emerging evidence

4

suggests they can contribute to optimal induction of IgA and regulatory T cells (47)

5

ILCs can also directly interact with adaptive immune cells to promote tissue immunity.

ILC3 can

promote B cell responses through production of GM-CSF, BAFF and co-stimulatory receptors, and ILC2

7

have been shown to be critical regulators of B1 cell proliferation (14,34,48).

8

promote maintenance of memory CD4 T cell responses to pathogens through expression of Ox40L and

9

CD30L (49). Collectively, ILCs can orchestrate intestinal immunity to pathogens through rapid cytokine-

11

+

r Fo

10

ILC3 can also directly

dependent responses, or by facilitating optimal adaptive immune cell responses

ILCs limit inflammatory responses to commensal bacteria and dietary antigens

13

A growing body of evidence suggests that ILCs play a critical role in limiting inappropriate immune

14

responses to non-harmful environmental stimuli, such as commensal bacteria and dietary antigens. In the

15

absence of pathogen infection, loss of ILC3 or IL-22 results in peripheral dissemination of commensal

16

bacteria and systemic inflammation (17). Interestingly, disseminating bacteria were found to be of the

17

genus Alcaligenes or Achromobacter (17), which were characterized to constitutively colonize the GALT of

18

healthy humans, non-human primates and mice (50), suggesting that the ILC3–IL-22 pathway selectively

19

regulates the anatomical containment of commensal bacteria in order to limit chronic inflammation.

er

Pe

12

ew

vi

Re

20

Consistent with this, it has been demonstrated that regulation of ILC3 and IL-22 by the aryl

21

hydrocarbon receptor (Ahr) is critical to limit colonization of intestinal epithelial cells with segmented

22

filamentous bacteria (SFB). Ahr-deficient mice exhibit an overgrowth of SFB and a subsequent increase

23

in pro-inflammatory Th17 cells (51). Collectively, these studies suggest that the ILC3–IL-22 pathway is

24

critical to maintain intestinal homeostasis by limiting the dissemination and replication of commensal

25

bacteria that are intimately associated with mammalian hosts, such as Alcaligenes and Achromobacter

26

that are resident in lymphoid tissue, or SFB that are associated with intestinal epithelial cells.

27

Emerging evidence suggests that ILC3 may also limit inappropriate immune responses to

28

commensal bacteria through other effector pathways. For example, it was recently shown that ILC3 can

29

directly limit pro-inflammatory CD4 T cell responses to commensal bacteria through expression of MHC

+

7

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

6

International Immunology

Page 8 of 22

+

1

class II (MHCII) (52).

2

responses to commensal bacteria and spontaneous intestinal inflammation (52). A recent report by Merad

3

and colleagues also identified that ILC3 are a dominant source of GM-CSF in the intestine and critically

4

regulate myeloid cell homeostasis (53).

5

required to maintain optimal Treg cell responses and tolerance to dietary antigens (53). Collectively, these

6

results suggest that ILC3 play a central role in regulating intestinal homeostasis by limiting inappropriate

7

immune responses to normally beneficial or harmless environmental stimuli.

Loss of ILC-intrinsic MHCII resulted in dysregulated effector CD4 Th17 cell

Intriguingly, ILC3-mediated regulation of myeloid cells was

r Fo

9

ILCs mediate intestinal tissue repair

Production of IL-22 is critical in mediating repair of the intestinal epithelium in the context of tissue damage

11

(16). Studies by Eberl and colleagues have highlighted that ILC3 rapidly respond to experimental tissue

12

damage in the intestine and mediate IL-22-dependent tissue repair (54). Interestingly, in a mouse model

13

of graft versus host disease, ILC3-derived IL-22 maintained intestinal epithelial barrier integrity following

14

hematopoietic stem cell transplantation by directly protecting intestinal stem cells from immune-mediated

15

damage (55). Acting directly on intestinal stem cells may be one mechanism by which a relatively rare

16

population, such as ILC3, can substantially influence homeostasis in the intestine. Collectively, ILCs

17

promote multiple beneficial processes in order to maintain a state of health in the mammalian intestine.

er

Pe

10

vi

Re

18 The role of ILCs in promoting intestinal disease

20

In contrast to the tissue protective roles of intestinal ILCs, dysregulated ILC responses or changes in the

21

composition of intestinal ILC populations can promote intestinal inflammation and the development or

22

progression of intestinal cancers (Fig. 2).

ew

19

23 24

Dysregulated ILC responses promote intestinal inflammation

25

Pioneering research by Powrie and colleagues (56) identified that IL-23 production in response to

26

Helicobacter hepaticus infection, or following dysregulated activation of dendritic cells via anti-CD40

27

antibody administration, results in the expansion of a T-bet ILC3 population that promotes colitis in an IL-

28

17- and IFN-γ-dependent manner. Later research identified that T-bet co-expression in ILC3 is associated

29

with IFN-γ production and enterocolitis following Salmonella infection (57). IL-22 production by ILC3 can

+

8

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

8

Page 9 of 22

International Immunology

1

also promote colitis in some contexts (58), which may be dependent on co-expression of other pro-

2

inflammatory cytokines (16).

4

promote IFN-γ-dependent intestinal inflammation following anti-CD40 antibody administration (27).

5

Emerging evidence suggest that a shift from protective to pathologic ILC responses in the intestine may

6

involve lineage plasticity, altered expression of transcriptional regulators, or engagement of alternative

7

activation pathways (24,28,29). For example, recent work suggests that loss of RORγt expression in ILC3

8

is accompanied with an upregulation of T-bet and IFNγ expression (24,28). As RORγt is a defining

9

transcription factor of ILC3, these cells have been termed ‘ex-ILC3’, but are distinct from ILC1, which have

10

no history RORγt expression (33). Collectively, these data suggest that dysregulation or expansion of pro-

11

inflammatory ILC populations may directly promote colitis through the production of cytokines IL-17, IL-22,

12

TNF-α and IFN-γ.

13

promote dysregulation of the composition or functional potential of intestinal ILCs, and further, a role for

14

ILC2 in intestinal inflammation.

Pe

er

15

Additional studies are required to carefully define the mechanistic pathways that

Re

Dysregulated ILC responses support intestinal cancers

17

In addition to chronic inflammation, dysregulated ILC responses have been linked to the development of

18

intestinal cancers. Administration of a carcinogen following Helicobacter hepaticus infection results in the

19

formation of aberrant crypt lesions, which were enriched in expression of IL-23, IL-22 and IL-17 (30). ILC3

20

and IL-22 were required for cancer progression in this model (30), suggesting that sustained activation of

21

ILC3 can promote the formation of intestinal tumors.

ew

vi

16

22

In support of this, it was recently highlighted that uncontrolled IL-22-dependent signals, occurring

23

through a loss of the regulatory IL-22 binding protein (IL-22BP), results in increased tumorigenesis in the

24

intestine following tissue damage (59). Further, it has also been shown that loss of intestinal barrier

25

function can contribute to sustained activation of the IL-23–IL-17 pathway and promote growth of intestinal

26

tumors (60). It is possible to speculate that the ILC3–IL-22 axis may be particularly poised to promote

27

tumorigenesis by its ability to act directly on intestinal stem cells, as previously observed following

28

hematopoietic stem cell transplantation (55).

9

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

In addition to ILC3, recent research suggests that dysregulated intra-epithelial ILC1 responses can

r Fo

3

International Immunology

Page 10 of 22

1

Therefore, sustained activation of ILCs, likely the result of chronic infection or impaired intestinal

2

barrier function, can promote the development and progression of inflammation and cancer within the GI

3

tract. Despite these advances, little is known about whether other factors expressed by ILC3, such as LT,

4

MHCII or GM-CSF, can in some contexts promote the development of intestinal inflammation or tumors,

5

and this will be an important area of future investigation.

6

8

As discussed above, translational studies in healthy human donors and patient populations have revealed

9

a significant association of altered intestinal ILC responses with human disease, and basic mouse models

10

have demonstrated that ILCs have the functional potential to promote both tissue protective and

11

pathologic responses. Therefore, it is likely that developing novel targets to manipulate ILCs will likely be

12

of significant value to prevent or limit human intestinal diseases. Additional analyses are required to

13

identify the regulatory pathways that control the composition and functional potential of ILC responses in

14

the intestine. Recent reports have highlighted that microbial and dietary signals can significantly influence

15

intestinal ILCs (43,54,61), suggesting that not only will this be important to consider in our interpretation of

16

basic and translational studies, but also that this may be a useful strategy to identify pathways to boost

17

protective ILC responses while limiting potentially limiting pathologic ILC responses.

er

Pe

vi

Re

18 Acknowledgements

20

Members of the Sonnenberg laboratory are thanked for discussions and critical reading of the manuscript.

21

Research in the Sonnenberg laboratory is supported by the National Institutes of Health (DP5OD012116),

22

the NIAID Mucosal Immunology Studies Team (MIST) Scholar Award in Mucosal Immunity, a pilot grant

23

from the NIDDK P30 center for Molecular Studies in Digestive and Liver Diseases (P30DK50306) and the

24

Institute for Translational Medicine and Therapeutics Transdisciplinary Program in Translational Medicine

25

and Therapeutics (UL1-RR024134 from the US National Center for Research Resources).

ew

19

26 27

Abbreviations

28

BAFF, B-cell activating factor; GATA3, GATA-binding protein 3; GI, gastrointestinal; IBD, inflammatory

29

bowel disease; Id2, inhibitor of DNA binding 2; ILCs, innate lymphoid cells; NCR, natural cytotoxicity

10

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

Summary and outstanding questions

r Fo

7

Page 11 of 22

International Immunology

1

receptor,

2

immunodeficiency virus; T-bet, T-box expressed in T cells; TCF1, T-cell factor 1; TSLP, thymic stromal

3

lymphopoietin.

Ox40L, Ox40 ligand; RORγt, retinoic acid-related orphan receptor γt; SIV, simian

4 5

Figure legends

6

Figure 1.

7

promote optimal innate and adaptive immunity, maintain immune cell homeostasis and mediate tissue

8

repair. This occurs through regulation of multiple epithelial, stromal, granulocyte, myeloid and adaptive

9

immune cell lineages.

Intestinal ILCs can

11

Figure 2.

12

Dysregulated ILC responses, or changes in the composition of ILC populations, can promote chronic

13

intestinal inflammation or initiate the development and progression of intestinal tumors. This can occur

14

through loss of regulation or sustained activation.

Dysregulated innate lymphoid cell responses can promote intestinal disease.

er

Pe

15

Re

16

References

17

1

18

relationships, and function. Annu Rev Immunol 30:647.

19

2

20

intestinal innate lymphoid cells. Curr Opin Immunol 24:277.

21

3

22

them? Nat Rev Immunol 13:75.

23

4

24

of innate lymphoid cells. Int Immunol 26:119.

Spits, H. and Cupedo, T. 2012. Innate lymphoid cells: emerging insights in development, lineage

ew

vi

Cherrier, M., Ohnmacht, C., Cording, S., and Eberl, G. 2012. Development and function of

Walker, J. A., Barlow, J. L., and McKenzie, A. N. 2013. Innate lymphoid cells--how did we miss

Tanriver, Y. and Diefenbach, A. 2014. Transcription factors controlling development and function

11

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

10

Innate lymphoid cells are critical regulators of intestinal health.

International Immunology

Page 12 of 22

1

5

2

Immunity 39:622.

3

6

4

implications for intestinal health and disease. Immunity 37:601.

5

7

6

R. M., McKenzie, A. N., Mebius, R. E., Powrie, F., and Vivier, E. 2013. Innate lymphoid cells--a proposal

7

for uniform nomenclature. Nat Rev Immunol 13:145.

8

8

9

and Colonna, M. 2009. A human natural killer cell subset provides an innate source of IL-22 for mucosal

Sonnenberg, G. F., Mjosberg, J., Spits, H., and Artis, D. 2013. SnapShot: Innate Lymphoid Cells.

Sonnenberg, G. F. and Artis, D. 2012. Innate lymphoid cell interactions with microbiota:

r Fo

Cella, M., Fuchs, A., Vermi, W., Facchetti, F., Otero, K., Lennerz, J. K., Doherty, J. M., Mills, J. C.,

Pe

immunity. Nature 457:722.

11

9

12

Cornelissen, J. J., and Spits, H. 2009. Human fetal lymphoid tissue-inducer cells are interleukin 17-

13

producing precursors to RORC+ CD127+ natural killer-like cells. Nat Immunol 10:66.

14

10

15

Yu, J., and Caligiuri, M. A. 2009. Stage 3 immature human natural killer cells found in secondary lymphoid

16

tissue constitutively and selectively express the TH 17 cytokine interleukin-22. Blood 113:4008.

17

11

18

cells and LTi-like cells constitute a stable RORC+ lineage distinct from conventional natural killer cells. J

19

Exp Med 207:281.

20

12

21

C., Nuovo, G., Wei, L., Zhang, X., Gavrilin, M. A., Wewers, M. D., and Caligiuri, M. A. 2010. Interleukin-

er

10

Cupedo, T., Crellin, N. K., Papazian, N., Rombouts, E. J., Weijer, K., Grogan, J. L., Fibbe, W. E.,

vi

Re

ew

Hughes, T., Becknell, B., McClory, S., Briercheck, E., Freud, A. G., Zhang, X., Mao, H., Nuovo, G.,

Crellin, N. K., Trifari, S., Kaplan, C. D., Cupedo, T., and Spits, H. 2010. Human NKp44+IL-22+

Hughes, T., Becknell, B., Freud, A. G., McClory, S., Briercheck, E., Yu, J., Mao, C., Giovenzana,

12

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

Spits, H., Artis, D., Colonna, M., Diefenbach, A., Di Santo, J. P., Eberl, G., Koyasu, S., Locksley,

Page 13 of 22

International Immunology

1

1beta selectively expands and sustains interleukin-22+ immature human natural killer cells in secondary

2

lymphoid tissue. Immunity 32:803.

3

13

4

Regulation of cytokine secretion in human CD127(+) LTi-like innate lymphoid cells by Toll-like receptor 2.

5

Immunity 33:752.

6

14

7

IL-1beta reveals intrinsic functional plasticity. Proc Natl Acad Sci U S A 107:10961.

8

15

9

Mjosberg, J. M., Spits, H., and Cupedo, T. 2012. Functional Differences between Human NKp44(-) and

Crellin, N. K., Trifari, S., Kaplan, C. D., Satoh-Takayama, N., Di Santo, J. P., and Spits, H. 2010.

r Fo

Hoorweg, K., Peters, C. P., Cornelissen, F., Aparicio-Domingo, P., Papazian, N., Kazemier, G.,

Pe

NKp44(+) RORC(+) Innate Lymphoid Cells. Front Immunol 3:72.

11

16

12

inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat Immunol 12:383.

13

17

14

Shibata, N., Grunberg, S., Sinha, R., Zahm, A. M., Tardif, M. R., Sathaliyawala, T., Kubota, M., Farber, D.

15

L., Collman, R. G., Shaked, A., Fouser, L. A., Weiner, D. B., Tessier, P. A., Friedman, J. R., Kiyono, H.,

16

Bushman, F. D., Chang, K. M., and Artis, D. 2012. Innate lymphoid cells promote anatomical containment

17

of lymphoid-resident commensal bacteria. Science 336:1321.

18

18

19

L. K., Pan, L., Vinton, C. L., Tabb, B., Canary, L. A., Dang, Q., Hirsch, V. M., Alter, G., Belkaid, Y., Lifson,

20

J. D., Silvestri, G., Milner, J. D., Paiardini, M., Haddad, E. K., and Brenchley, J. M. 2012. Loss of mucosal

21

CD103+ DCs and IL-17+ and IL-22+ lymphocytes is associated with mucosal damage in SIV infection.

22

Mucosal Immunol 5:646.

er

10

Sonnenberg, G. F., Fouser, L. A., and Artis, D. 2011. Border patrol: regulation of immunity,

vi

Re

Sonnenberg, G. F., Monticelli, L. A., Alenghat, T., Fung, T. C., Hutnick, N. A., Kunisawa, J.,

ew

Klatt, N. R., Estes, J. D., Sun, X., Ortiz, A. M., Barber, J. S., Harris, L. D., Cervasi, B., Yokomizo,

13

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

Cella, M., Otero, K., and Colonna, M. 2010. Expansion of human NK-22 cells with IL-7, IL-2, and

International Immunology

Page 14 of 22

1

19

2

17-producing innate lymphoid cells are restricted to mucosal tissues and are depleted in SIV-infected

3

macaques. Mucosal Immunol 5:658.

4

20

5

V., Carville, A., and Johnson, R. P. 2011. Gut inflammation and indoleamine deoxygenase inhibit IL-17

6

production and promote cytotoxic potential in NKp44+ mucosal NK cells during SIV infection. Blood

7

118:3321.

8

21

9

C., Shin, L. Y., Grin, A., Kandel, G., Loutfy, M., Ostrowski, M., Gommerman, J. L., Kaushic, C., and Kaul,

10

R. 2012. A role for mucosal IL-22 production and Th22 cells in HIV-associated mucosal

11

immunopathogenesis. Mucosal Immunol 5:670.

12

22

13

J., Veiga-Fernandes, H., and Sousa, A. E. 2014. Enteric mucosa integrity in the presence of a preserved

14

innate IL-22 compartment in HIV-1 treated individuals. J Infect Dis.

15

23

16

Suzuki, S., Sugita, A., Koganei, K., Hisamatsu, T., Kanai, T., and Hibi, T. 2010. Imbalance of

17

NKp44(+)NKp46(-) and NKp44(-)NKp46(+) natural killer cells in the intestinal mucosa of patients with

18

Crohn's disease. Gastroenterology 139:882.

19

24

20

Hreggvidsdottir, H. S., Heinsbroek, S. E., Legrand, N., Buskens, C. J., Bemelman, W. A., Mjosberg, J. M.,

21

and Spits, H. 2013. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat

22

Immunol 14:221.

Xu, H., Wang, X., Liu, D. X., Moroney-Rasmussen, T., Lackner, A. A., and Veazey, R. S. 2012. IL-

Reeves, R. K., Rajakumar, P. A., Evans, T. I., Connole, M., Gillis, J., Wong, F. E., Kuzmichev, Y.

er

Pe

Fernandes, S. M., Pires, A. R., Ferreira, C., Foxall, R. B., Rino, J., Santos, C., Correia, L., Pocas,

vi

Re

ew

Takayama, T., Kamada, N., Chinen, H., Okamoto, S., Kitazume, M. T., Chang, J., Matuzaki, Y.,

Bernink, J. H., Peters, C. P., Munneke, M., Te Velde, A. A., Meijer, S. L., Weijer, K.,

14

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Kim, C. J., Nazli, A., Rojas, O. L., Chege, D., Alidina, Z., Huibner, S., Mujib, S., Benko, E., Kovacs,

Page 15 of 22

International Immunology

1

25

2

Giardina, A., De Leo, G., and Triolo, G. 2011. Interleukin-22 and IL-22-producing NKp44(+) NK cells in the

3

subclinical gut inflammation of patients with ankylosing spondylitis. Arthritis Rheum.

4

26

5

Travis, S. P., and Powrie, F. 2011. IL-23-responsive innate lymphoid cells are increased in inflammatory

6

bowel disease. J Exp Med 208:1127.

7

27

8

M. 2013. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive

9

IFN-gamma-producing cells. Immunity 38:769.

Ciccia, F., Accardo-Palumbo, A., Alessandro, R., Rizzo, A., Principe, S., Peralta, S., Raiata, F.,

Geremia, A., Arancibia-Carcamo, C. V., Fleming, M. P., Rust, N., Singh, B., Mortensen, N. J.,

Pe

10

28

11

Bengsch, B., Thimme, R., Holscher, C., Honig, M., Pannicke, U., Schwarz, K., Ware, C. F., Finke, D., and

12

Diefenbach, A. 2010. Regulated expression of nuclear receptor RORgammat confers distinct functional

13

fates to NK cell receptor-expressing RORgammat(+) innate lymphocytes. Immunity 33:736.

14

29

15

N., Seidl, R., Seifarth, C., Grone, J., Lenarz, M., Stolzel, K., Fugmann, D., Porgador, A., Hauser, A.,

16

Karlas, A., and Romagnani, C. 2013. RORgammat(+) innate lymphoid cells acquire a proinflammatory

17

program upon engagement of the activating receptor NKp44. Immunity 38:1223.

18

30

19

O., and Powrie, F. 2013. Innate lymphoid cells sustain colon cancer through production of interleukin-22 in

20

a mouse model. J Exp Med 210:917.

21

31

22

J., Cupedo, T., and Spits, H. 2011. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are

23

defined by expression of CRTH2 and CD161. Nat Immunol 12:1055.

Vonarbourg, C., Mortha, A., Bui, V. L., Hernandez, P. P., Kiss, E. A., Hoyler, T., Flach, M.,

er

vi

Re

Glatzer, T., Killig, M., Meisig, J., Ommert, I., Luetke-Eversloh, M., Babic, M., Paclik, D., Bluthgen,

ew

Kirchberger, S., Royston, D. J., Boulard, O., Thornton, E., Franchini, F., Szabady, R. L., Harrison,

Mjosberg, J. M., Trifari, S., Crellin, N. K., Peters, C. P., van Drunen, C. M., Piet, B., Fokkens, W.

15

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Fuchs, A., Vermi, W., Lee, J. S., Lonardi, S., Gilfillan, S., Newberry, R. D., Cella, M., and Colonna,

International Immunology

Page 16 of 22

1

32

2

and Whiting, C. V. 2012. IL-13 promotes collagen accumulation in Crohn's disease fibrosis by down-

3

regulation of fibroblast MMP synthesis: a role for innate lymphoid cells? PLoS One 7:e52332.

4

33

5

V., Fonseca-Pereira, D., Domingues, R. G., Veiga-Fernandes, H., Arnold, S. J., Busslinger, M., Dunay, I.

6

R., Tanriver, Y., and Diefenbach, A. 2014. Differentiation of Type 1 ILCs from a Common Progenitor to All

7

Helper-like Innate Lymphoid Cell Lineages. Cell 157:340.

8

34

9

M., Fujii, H., and Koyasu, S. 2010. Innate production of T(H)2 cytokines by adipose tissue-associated c-

Bailey, J. R., Bland, P. W., Tarlton, J. F., Peters, I., Moorghen, M., Sylvester, P. A., Probert, C. S.,

Klose, C. S., Flach, M., Mohle, L., Rogell, L., Hoyler, T., Ebert, K., Fabiunke, C., Pfeifer, D., Sexl,

Pe

Kit(+)Sca-1(+) lymphoid cells. Nature 463:540.

11

35

12

M., Fallon, P. G., Pannell, R., Jolin, H. E., and McKenzie, A. N. 2010. Nuocytes represent a new innate

13

effector leukocyte that mediates type-2 immunity. Nature 464:1367.

14

36

15

lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 34:122.

16

37

17

Mention, J. J., Thiam, K., Cerf-Bensussan, N., Mandelboim, O., Eberl, G., and Di Santo, J. P. 2008.

18

Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal

19

immune defense. Immunity 29:958.

20

38

21

innate lymphoid cells. Curr Opin Immunol in press.

er

10

Neill, D. R., Wong, S. H., Bellosi, A., Flynn, R. J., Daly, M., Langford, T. K., Bucks, C., Kane, C.

vi

Re

Sonnenberg, G. F., Monticelli, L. A., Elloso, M. M., Fouser, L. A., and Artis, D. 2011. CD4(+)

ew

Satoh-Takayama, N., Vosshenrich, C. A., Lesjean-Pottier, S., Sawa, S., Lochner, M., Rattis, F.,

Hepworth, M. R. and Sonnenberg, G. F. 2014. Regulation of the adaptive immune system by

16

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Moro, K., Yamada, T., Tanabe, M., Takeuchi, T., Ikawa, T., Kawamoto, H., Furusawa, J., Ohtani,

Page 17 of 22

International Immunology

1

39

2

LTbeta+ cells that can differentiate to APC, NK cells, and follicular cells but not T or B cells. Immunity

3

7:493.

4

40

Mebius, R. E. 2003. Organogenesis of lymphoid tissues. Nat Rev Immunol 3:292.

5

41

Eberl, G., Marmon, S., Sunshine, M. J., Rennert, P. D., Choi, Y., and Littman, D. R. 2004. An

6

essential function for the nuclear receptor RORgamma(t) in the generation of fetal lymphoid tissue inducer

7

cells. Nat Immunol 5:64.

8

42

9

Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature

Mebius, R. E., Rennert, P., and Weissman, I. L. 1997. Developing lymph nodes collect CD4+CD3-

Pe

456:507.

11

43

12

L., Almeida, F. F., Ibiza, S., Barbosa, I., Goverse, G., Labao-Almeida, C., Godinho-Silva, C., Konijn, T.,

13

Schooneman, D., O'Toole, T., Mizee, M. R., Habani, Y., Haak, E., Santori, F. R., Littman, D. R., Schulte-

14

Merker, S., Dzierzak, E., Simas, J. P., Mebius, R. E., and Veiga-Fernandes, H. 2014. Maternal retinoids

15

control type 3 innate lymphoid cells and set the offspring immunity. Nature 508:123.

16

44

17

Eberl, G., Littman, D. R., Heikenwalder, M., Tumanov, A. V., and Nedospasov, S. A. 2013. Nonredundant

18

function of soluble LTalpha3 produced by innate lymphoid cells in intestinal homeostasis. Science

19

342:1243.

20

45

21

and Fagarasan, S. 2008. Requirement for lymphoid tissue-inducer cells in isolated follicle formation and T

22

cell-independent immunoglobulin A generation in the gut. Immunity 29:261.

er

10

van de Pavert, S. A., Ferreira, M., Domingues, R. G., Ribeiro, H., Molenaar, R., Moreira-Santos,

ew

vi

Re

Kruglov, A. A., Grivennikov, S. I., Kuprash, D. V., Winsauer, C., Prepens, S., Seleznik, G. M.,

Tsuji, M., Suzuki, K., Kitamura, H., Maruya, M., Kinoshita, K., Ivanov, II, Itoh, K., Littman, D. R.,

17

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Bouskra, D., Brezillon, C., Berard, M., Werts, C., Varona, R., Boneca, I. G., and Eberl, G. 2008.

International Immunology

Page 18 of 22

1

46

2

Thornton, E. E., Krummel, M. F., Chawla, A., Liang, H. E., and Locksley, R. M. 2013. Type 2 innate

3

lymphoid cells control eosinophil homeostasis. Nature 502:245.

4

47

5

C. 2014. Eosinophils promote generation and maintenance of immunoglobulin-a-expressing plasma cells

6

and contribute to gut immune homeostasis. Immunity 40:582.

7

48

8

Chudnovskiy, A., Gentile, M., Llige, D., Cols, M., Serrano, S., Arostegui, J. I., Juan, M., Yague, J., Merad,

9

M., Fagarasan, S., and Cerutti, A. 2014. Innate lymphoid cells integrate stromal and immunological signals

Nussbaum, J. C., Van Dyken, S. J., von Moltke, J., Cheng, L. E., Mohapatra, A., Molofsky, A. B.,

Chu, V. T., Beller, A., Rausch, S., Strandmark, J., Zanker, M., Arbach, O., Kruglov, A., and Berek,

Pe

to enhance antibody production by splenic marginal zone B cells. Nat Immunol 15:354.

11

49

12

N. A., White, A. J., Flores-Langarica, A., McConnell, F. M., Anderson, G., and Lane, P. J. 2012. Cutting

13

edge: lymphoid tissue inducer cells maintain memory CD4 T cells within secondary lymphoid tissue. J

14

Immunol 189:2094.

15

50

16

Shibata, N., Gohda, M., Kagiyama, Y., Nochi, T., Yuki, Y., Fukuyama, Y., Mukai, A., Shinzaki, S.,

17

Fujihashi, K., Sasakawa, C., Iijima, H., Goto, M., Umesaki, Y., Benno, Y., and Kiyono, H. 2010. Indigenous

18

opportunistic bacteria inhabit mammalian gut-associated lymphoid tissues and share a mucosal antibody-

19

mediated symbiosis. Proc Natl Acad Sci U S A 107:7419.

20

51

21

Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon

22

receptor signaling and regulation of microflora. Immunity 39:386.

er

10

Withers, D. R., Gaspal, F. M., Mackley, E. C., Marriott, C. L., Ross, E. A., Desanti, G. E., Roberts,

vi

Re

ew

Obata, T., Goto, Y., Kunisawa, J., Sato, S., Sakamoto, M., Setoyama, H., Matsuki, T., Nonaka, K.,

Qiu, J., Guo, X., Chen, Z. M., He, L., Sonnenberg, G. F., Artis, D., Fu, Y. X., and Zhou, L. 2013.

18

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Magri, G., Miyajima, M., Bascones, S., Mortha, A., Puga, I., Cassis, L., Barra, C. M., Comerma, L.,

Page 19 of 22

International Immunology

1

52

2

Mantegazza, A. R., Ma, H. L., Crawford, A., Angelosanto, J. M., Wherry, E. J., Koni, P. A., Bushman, F.

3

D., Elson, C. O., Eberl, G., Artis, D., and Sonnenberg, G. F. 2013. Innate lymphoid cells regulate CD4+ T-

4

cell responses to intestinal commensal bacteria. Nature 498:113.

5

53

6

Merad, M. 2014. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal

7

homeostasis. Science 343:1249288.

8

54

9

Santo, J. P., and Eberl, G. 2011. RORgammat(+) innate lymphoid cells regulate intestinal homeostasis by

Hepworth, M. R., Monticelli, L. A., Fung, T. C., Ziegler, C. G., Grunberg, S., Sinha, R.,

Mortha, A., Chudnovskiy, A., Hashimoto, D., Bogunovic, M., Spencer, S. P., Belkaid, Y., and

Pe

integrating negative signals from the symbiotic microbiota. Nat Immunol 12:320.

11

55

12

Jenq, R. R., Holland, A. M., Kappel, L. W., Ghosh, A., Tsai, J. J., Rao, U. K., Yim, N. L., Smith, O. M.,

13

Velardi, E., Hawryluk, E. B., Murphy, G. F., Liu, C., Fouser, L. A., Kolesnick, R., Blazar, B. R., and van den

14

Brink, M. R. 2012. Interleukin-22 protects intestinal stem cells from immune-mediated tissue damage and

15

regulates sensitivity to graft versus host disease. Immunity 37:339.

16

56

17

2010. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 464:1371.

18

57

19

Croxford, A. L., Waisman, A., Tanriver, Y., and Diefenbach, A. 2013. A T-bet gradient controls the fate and

20

function of CCR6-RORgammat+ innate lymphoid cells. Nature 494:261.

21

58

22

colitis via interleukin-22-dependent mechanism. Mucosal Immunol 7:143.

er

10

Hanash, A. M., Dudakov, J. A., Hua, G., O'Connor, M. H., Young, L. F., Singer, N. V., West, M. L.,

ew

vi

Re

Buonocore, S., Ahern, P. P., Uhlig, H. H., Ivanov, II, Littman, D. R., Maloy, K. J., and Powrie, F.

Klose, C. S., Kiss, E. A., Schwierzeck, V., Ebert, K., Hoyler, T., d'Hargues, Y., Goppert, N.,

Eken, A., Singh, A. K., Treuting, P. M., and Oukka, M. 2014. IL-23R+ innate lymphoid cells induce

19

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Sawa, S., Lochner, M., Satoh-Takayama, N., Dulauroy, S., Berard, M., Kleinschek, M., Cua, D., Di

International Immunology

Page 20 of 22

1

59

2

O'Connor, W., Jr., Murphy, A. J., Valenzuela, D. M., Yancopoulos, G. D., Booth, C. J., Cho, J. H., Ouyang,

3

W., Abraham, C., and Flavell, R. A. 2012. IL-22BP is regulated by the inflammasome and modulates

4

tumorigenesis in the intestine. Nature 491:259.

5

60

6

G. Y., Osterreicher, C. H., Hung, K. E., Datz, C., Feng, Y., Fearon, E. R., Oukka, M., Tessarollo, L.,

7

Coppola, V., Yarovinsky, F., Cheroutre, H., Eckmann, L., Trinchieri, G., and Karin, M. 2012. Adenoma-

8

linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature 491:254.

9

61

Huber, S., Gagliani, N., Zenewicz, L. A., Huber, F. J., Bosurgi, L., Hu, B., Hedl, M., Zhang, W.,

Grivennikov, S. I., Wang, K., Mucida, D., Stewart, C. A., Schnabl, B., Jauch, D., Taniguchi, K., Yu,

Pe

10

J. F., Jr., Wang, J., Ramalingam, T. R., Bhandoola, A., Wynn, T. A., and Belkaid, Y. 2014. Adaptation of

11

innate lymphoid cells to a micronutrient deficiency promotes type 2 barrier immunity. Science 343:432.

er ew

vi

Re 20

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Spencer, S. P., Wilhelm, C., Yang, Q., Hall, J. A., Bouladoux, N., Boyd, A., Nutman, T. B., Urban,

International Immunology Page 21 of 22

er Pe

ew vi Re

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Page 22 of 22 International Immunology

er Pe

ew vi Re

Downloaded from http://intimm.oxfordjournals.org/ at University of New Hampshire Library on May 29, 2014

r Fo

Regulation of intestinal health and disease by innate lymphoid cells.

Innate lymphoid cells (ILCs) are a recently appreciated immune cell population that is constitutively found in the healthy mammalian gastrointestinal ...
541KB Sizes 0 Downloads 0 Views