Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 May 17.
Published in final edited form as: Nat Med. 2015 Jun 29;21(7):698–708. doi: 10.1038/nm.3892

Innate lymphoid cells in the initiation, regulation and resolution of inflammation

Gregory F Sonnenberg 1, David Artis 1
PMCID: PMC4869856  NIHMSID: NIHMS784852  PMID: 26121198

Abstract

A previously unappreciated cell type of the innate immune system, termed innate lymphoid cells (ILCs), has been characterized in mice and humans, and found to profoundly influence the induction, regulation and resolution of inflammation. ILCs play an important role in these processes in murine models of infection, inflammatory disease and tissue repair. Further, disease association studies in defined patient populations have identified significant alterations in ILC responses, suggesting a potential role for these cell populations in human health and disease. In this review, we discuss the emerging family of ILCs, the role of ILCs in inflammation, and how current or novel therapeutic strategies could be employed to selectively modulate ILC responses and limit chronic inflammatory diseases in patients.


Inflammation is defined as heat, redness, pain, swelling and loss of function. While acute inflammation is a necessary process to protect against infection and promote tissue repair, chronic inflammation directly contributes to the pathogenesis and progression of multiple infectious, inflammatory and metabolic disorders, including HIV/AIDS, inflammatory bowel disease, arthritis, psoriasis, allergy, asthma, diabetes, obesity and cancer1,2. While there are many well characterized cellular and molecular components of the innate and adaptive immune system that influence inflammatory processes, recent characterization of an emerging family of innate immune cells, termed ILCs, has revealed an essential role for these populations in the initiation, regulation and resolution of inflammation. ILCs are a population of innate lymphocytes that are relatively rare in comparison to adaptive lymphocytes in lymphoid tissues, but are enriched at barrier surfaces of the mammalian body, such as the skin, lung and intestine, as well as adipose and some mucosal-associated lymphoid tissues36. ILCs rapidly respond to cytokine and microbial signals and are potent innate cellular sources of multiple pro-inflammatory and immuno-regulatory cytokines, and recent research has also identified a critical role for ILCs in modulating adaptive immunity. Mature ILC subsets can be identified by a lack of known lineage markers associated with T cells, B cells, myeloid cells, or granulocytes, but share expression of the common gamma chain (γc, CD132), IL-7Rα (CD127), IL-2Rα (CD25), and Thy1 (CD90), with some exceptions noted below36.

A combination of advances in multi-parameter flow cytometry and the identification of novel cytokine pathways regulating immunity and inflammation, including the interleukin (IL-)23-IL-22 pathway712 and epithelial-derived cytokines IL-25, IL-33 and thymic stromal lymphopoietin (TSLP)1317, contributed to our emerging knowledge of ILCs. Prototypical members of the ILC family were discovered many years prior, including the natural killer (NK) cells in 197518,19, and subsequently lymphoid tissue-inducer (LTi) cells20. However, it was not until more recently that other members of the ILC family were characterized. These included simultaneous reports of innate lymphocytes that are predominant cellular sources of the cytokines IL-17 and IL-222128, or IL-5 and IL-132933, in the steady state or early following infection. These rapid and fundamental advances also generated redundant nomenclature based upon functional potential of the identified cells, including NK-22 cells, LTi-like cells, natural helper cells, nuocytes and innate helper cells. To limit confusion, leaders in the field later unified a common terminology to classify these emerging cell populations as a new family of ILCs which encompasses three subsets, termed group 1,2 or 3 ILCs, based on common expression or dependence of surface markers, transcription factors and cytokines3.

Recent investigations of ILCs has caused a shift in our understanding of innate and adaptive immunity, and has fuelled additional extensive investigation into these cells due to the potential influence of ILCs in human health and disease. Mouse models indicate that ILCs play a fundamental role in the immune system by initiating, regulating and resolving inflammation. Further, studies in humans have revealed that ILC responses are significantly altered in several disease states. Below we discuss the development and heterogeneity of ILCs, the role of human and mouse ILCs in inflammatory processes, and how current or novel therapeutic strategies could be employed to modulate ILC responses and benefit human health.

Development and heterogeneity of the innate lymphoid cell family

ILCs initially develop in the fetal liver and later in the adult bone marrow from common lymphoid progenitors (CLPs)3436. CLPs also differentiate into cells of the adaptive immune system, such as T cells and B cells, but development of ILCs from CLPs occurs independent of somatic recombination, a defining feature of the adaptive immune system that permits the generation antigen-specific receptors or secreted proteins such as the T cell receptor, B cell receptor and immunoglobulin. ILC develop is regulated at the transcriptional level, with several precursor populations and transcription factors regulating each lineage (Fig. 1)4,37,38. Differentiation of all ILCs from a CLP requires the transcription factors inhibitor of DNA binding 2 (Id2), nuclear factor interleukin-3 regulated (NFIL3)3,4,36,3943, and thymocyte selection-associated high mobility group box (Tox)44,45 and involves additional precursor populations (Fig. 1). These include NK cell precursors (NKp) that give rise to NK cells, and a common helper innate lymphoid precursor (CHILP) that gives rise to all other defined ILCs in a process that requires T cell factor 1 (TCF1)46,47 and GATA binding protein 3 (GATA3)48,49. From CHILPs, several distinct progenitors expressing α4β7 integrin give rise to LTi cells34, while a PLZF-dependent ILC progenitor (ILCp) can give rise to other defined ILC populations35. While the specific interactions and functions of transcription factors in ILC development are not well defined, one recent study elegantly demonstrated that IL-7 signaling can induce NFIL3 and subsequently Id2 to support the generation of ILC progenitors39. However additional studies are required to further define these functional interactions as NFIL3-deficient mice do not completely replicate the phenotype of Id2-deficeint mice, which lack nearly all ILCs. Further, while most of these developmental studies have necessarily employed mice, recent research by Romagnani and colleagues defined that CD34+ hematopoietic progenitor cells (HSCs) in the bone marrow or peripheral blood can give rise to α4β7 integrin expressing CD34+ progenitors in the lymphoid tissues and intestinal lamina propria with the potential to differentiate into either NK cells or LTi-like cells in a process that influenced by the presence of aryl hydrocarbon receptor (Ahr) ligands, stem cell factor (SCF) and IL-1550.

Figure 1. Development and heterogeneity of the ILC family.

Figure 1

ILCs develop from distinct progenitors in the fetal liver or bone marrow. All ILCs develop from common lymphoid progenitors (CLPs), which can differentiate into NK cell progenitors (NKp) or common helper innate lymphoid precursors (CHILPs). CHIPs can further differentiation to lymphoid tissue inducer (LTi) cells through α4β7 integrin-expressing intermediate populations, or to other ILC populations through differentiation to a PLZF-dependent ILC progenitor (ILCp). Further sequential engagement of transcription factors, cytokines and microbial signals is critical for the development of three distinct groups of mature ILCs. ILC1 express T-bet, are responsive to IL-12, and produce IFNγ. ILC2 highly express GATA3, are responsive to IL-25, IL-33 and TSLP, and produce IL-4, IL-5, IL-9, IL-13 and Areg. ILC3 express RORγt, are responsive to IL-1β and IL-23, and produce IL-17 and/or IL-22.

Mature ILCs provide a potent and early source of cytokines in response to various stimuli, including direct cytokine stimulation, or as a consequence of colonization with commensal microbes or pathogen infection. There is considerable phenotypic and functional heterogeneity in the mature ILC family, and broadly three groups of ILCs have been defined based upon shared expression of surface markers, transcription factors and effector cytokines (Fig. 1)36.

Group 1 ILCs (ILC1) respond to IL-12, constitutively express T-bet, and produce the effector cytokines including IFNγ and TNF36,51,52. ILC1 are considerably heterogeneous and can be distinguished into at least three subsets based up differential expression and requirements for eomesodermin (eomes), T-bet, IL-15 and IL-7. Conventional NK cells are a subset of ILC1 requiring eomes and IL-15 for development from NKp, but not T-bet or IL-736. ILC1 also include CD103+ intraepithelial ILC1 that develop independent of IL-15 from an unknown precursor, express eomes, and require T-bet for differentiation52. Further, a CD127+ ILC1 was also identified in humans and mice, which does not express eomes, develops independently of IL-7 from a CHILP and ILCp, but requires T-bet and IL-1536,51.

Group 2 ILCs (ILC2) respond to cytokines IL-25, IL-33 and TSLP, constitutively express high levels of GATA3, and produce the effector cytokines IL-4, IL-5, IL-9, IL-13 and amphiregulin2933. While GATA3 is required for the development of most ILC subsets48,49, it is also required for the maintenance and function of mature ILC2 in humans and mice5356. RORα is also required for ILC2 development in mice57,58, but is also highly expressed by other ILC subsets and its function is poorly defined59. Gfi1 and Bcl11b also contributes to the differentiation or lineage stability of mouse ILC26062. Although mature ILC2 can be found in most anatomical locations, they appear to be enriched in the healthy lung, skin and adipose tissue of mice and humans29,31,32,6366.

Group 3 ILCs (ILC3) respond to IL-1β, IL-6 and IL-23, constitutively express RORγt, and produce the effector cytokines IL-17 and/or IL-222128. Although all ILC3 share a developmental requirement for RORγt24,67, significant heterogeneity exists within this group. Fetal and adult LTi cells are members of ILC3, which develop independently of PLZF-dependent ILCp, are CCR6+, heterogeneous in expression of CD4, express IL-22 and IL-17, and are a source of lymphotoxin (LT). A subset of adult ILC3 can develop from PLZF-dependent ILCp after birth are CCR6, co-express T-bet, are heterogeneous in expression of natural cytoxicity receptors (NCR, such as NKp46 and NKp44), and can co-express IL-22 and IFNγ. This T-bet+ ILC3 population requires T-bet, the presence of commensal bacteria and the aryl hydrocarbon receptor (AhR) for development6872. T-bet+ ILC3 are almost exclusively found in the skin and intestinal lamina propria, while LTi-like ILC3 are enriched in the intestine and lymphoid tissues28,68,7375. Further, as discussed below, ILC3 with a unique phenotype can develop in some inflammatory contexts in the intestine and liver24,76.

Although still an emerging field, plasticity between ILC groups has been observed. ILC3 subsets can downregulate RORγt expression in mice and humans, resulting in a dominant expression of T-bet and sustained expression of IFNγ51,68,72. However, it is currently unclear whether to classify these populations as ILC1 or ILC3, and it is difficult to distinguish them from a stable ILC1 lineage without fate mapping approaches that genetically mark cells that express or previously expressed RORγt. Therefore many groups have termed these cells ex-ILC336,51,68,72. Additional evidence suggests that a transient IL-25-responsive ILC2 population can differentiate into an IL-17-producing ILC3-like cell77, however additional research is required to fully characterize the extent of ILC plasticity, as well as define the potential causes and consequences of this plasticity.

ILCs orchestrate acute inflammation to promote immunity to infection

Acute inflammation is necessary to mount an effective immune response to various infectious organisms. ILCs were first identified based on their ability to promote rapid and essential innate immune responses to different classes of pathogens in part by modulating local epithelial cell, myeloid cell or granulocyte responses (Fig. 2).

Figure 2. ILCs promote acute inflammation to mediate innate immunity to pathogens.

Figure 2

ILCs promote innate immune responses to a number of pathogens in the intestine. (a) ILC1 promote innate immunity to intracellular pathogens, such as Toxoplasma gondii, by producing TNF and IFNγ in response to DC-derived IL-12 and subsequently promoting recruitment of inflammatory myeloid cells. (b) Following infection with the helminth parasites Nippostrongylus brasiliensis or Trichuris muris, ILC2 produce IL-13 in response to epithelial-cell derived-IL-25 and IL-33, which increases smooth muscle contractility and mucus production from goblet cells. (c) ILC3 produce IL-17 and IL-22 in response to DC-derived IL-23 and IL-1β, which promotes innate immunity to fungi and extracellular bacteria, such as Citrobacter rodentium and Candida albicans. IL-17 and IL-22 promote neutrophil recruitment to the intestine and the production of antimicrobial peptides from IECs.

ILC1 and intracellular pathogens

ILC1 populations play a critical role in promoting immunity to intracellular pathogens (Fig. 2a). Rapid NK cell responses have been well characterized following exposure to multiple intracellular pathogens in both humans and mice78. However, the role of other ILC1 populations has been less clear. Recent evidence suggests that ILC1 are the dominant innate source of IFNγ and TNF in mice following infection with the oral pathogen Toxoplasma gondii, and play a role in recruiting inflammatory myeloid cells that control infection36. Consistent with this, genomic T-bet-deficiency rendered mice highly susceptible to Toxoplasma gondii infection, and adoptive transfer of ILC1 to lymphocyte-deficient mice (Rag2/Il2rg/) was sufficient to boost immunity36.

ILC2 and extracellular parasites

ILC2 rapidly respond following exposure to multicellular parasites that are typically extracellular (Fig. 2b). For example, ILC2 are an important innate cellular source of IL-13 in the intestine following infection of mice with the parasite Nippostrongylus brasiliensis29,30,33. IL-13 can act on goblet cells to induce mucus production and on smooth muscle to enhance contractility, both of which are thought to contribute to expulsion of the parasites from the gastrointestinal tract79. IL-4 and IL-13 can also induce goblet cell expression of Relmβ, which limits parasitic infection80. Epithelial cell-derived IL-25 and IL-33 primarily promote the population expansion and cytokine production of ILC2s following parasite infection of mice29,30,33. ILC2 can similarly promote IL-13-mediated immunity to other parasites in mice, including Trichuris muris81. Notably, this response could be enhanced by Vitamin A-deficiency81, suggesting that ILC2 can respond to dietary stress and that the type 2 immune response may have adapted to support anti-parasite immunity in human populations with malnutrition.

ILC3 and extracellular bacteria and fungi

ILC3 rapidly respond to infections of mice with either extracellular bacteria or fungi (Fig. 2c). NCR+ ILC3 rapidly respond to infection of mice with the gram-negative enteric pathogen Citrobacter rodentium by producing IL-2222, which is essential for host protection7. These ILC3 responses in humans and mice can be promoted by dendritic cell (DC)-derived IL-2322, and in mice ILC3 are a dominant and essential source of IL-22 for innate immunity to Citrobacter rodentium in the intestine21,23. IL-22 acts almost exclusively on non-hematopoietic cells, such as intestinal epithelial cells (IECs), and stimulates production of antimicrobial peptides (RegIIIγ and RegIIIβ), element-sequestering proteins (lipocalin-2, S100A8 and S100A9), mucus production (Muc1, 3, 10 and 13) and epithelial fucosylation (Fut2)79,8285. These responses collectively limit the replication, dissemination and tissue damage induced by pathogenic and opportunistic bacteria. Similarly, ILC3 located in the oral mucosa promote IL-17-dependent innate immunity to infection with the fungal pathogen Candida albicans in mice86. IL-17 can act alone or synergistically with IL-22 to also promote antimicrobial peptide production and induces the expression of chemokines (Cxcl1 and Cxcl9) to recruit neutrophils to the site of infection79,87. ILC3-derived IL-17 also critically regulates neutrophils in neonatal mice, which is important for resistance to sepsis with gram negative opportunistic bacteria, and is critically dependent upon the presence of a commensal bacteria and microbial sensing pathways88. Collectively, this suggests that the ILC family plays a significant role in mediating acute inflammation in response to infection, which is important for the control and clearance of various classes of pathogens.

ILCs promote the resolution of inflammation and tissue repair

In addition to their role in initiating acute inflammatory responses and immunity to pathogens, ILCs directly contribute to the resolution of inflammation by repairing damaged tissues including the lung, various lymphoid tissues, and gastrointestinal tract (Fig. 3). These repair processes are essential to limit sustained inflammation, prevent against re-infection, and restore tissues to a state of homeostasis.

Figure 3. ILC2 and ILC3 promote the resolution of inflammation and tissue repair.

Figure 3

(a) Following viral infection in the lung, airway epithelial cells are damaged and produce IL-33 in conjunction with resident myeloid cell populations. ILC2 respond to IL-33 and produce amphiregulin, which promotes repair of the airway epithelium. (b) In lymphoid tissues, such as the spleen and thymus, stromal cell damage induced by viral infection or irradiation results in increased numbers of ILC3 and cytokine production, in part through production of DC-derived IL-23. ILC3 directly promote restoration of stromal cell compartments through production of LTα1β2 and IL-22, which increase the proliferation and survival of tissue resident stromal cells. (c) In the intestine, ILC3 responses can be limited by a regulatory loop whereby commensal bacteria induce IEC expression of IL-25, which acts on DCs to limit ILC3 cytokine responses in a contact-dependent manner. In contrast, upon chemical-, infection- or irradiation-induced damage of the intestine, ILC3 are activated by DC-derived IL-1β, IL-23, TL1A and retinoic acid (RA). Activation of ILC3 induces IL-22 production that directly promotes mucus production and epithelial cell repair, in part by acting directly on intestinal stem cells or progenitors.

ILC2 and resolution of inflammation in the lung

In the lung of mice, ILC2 expand in response to IL-33 following influenza virus infection and subsequent immune-mediated tissue damage (Fig. 3a)31. Experimental depletion of ILC2 in mice did not impair innate immunity to influenza, but rather limited repair of the airway epithelium and reduced lung function. This repair function was not mediated by ILC2 cytokine responses, such as IL-13, but rather was promoted by production of amphiregulin, a member of the epidermal growth factor family that was highly expressed by ILC231. Further, following infection and subsequent tissue damage induced by the migration of Nippostrongylus brasiliensis in the lungs of mice, autocrine production of IL-9 contributes to the survival of ILC2 and sustained amphiregulin production that mediates repair of the lung tissues89. ILC2 therefore represent a major ILC population in the lung that promotes tissue repair following infection. Murine ILC2s also promote cutaneous wound healing in an excisional wound model (Artis and Volk, pers. comm.). Although it has been demonstrated that human ILC2 express amphiregulin at the transcript level90, additional work is needed to define whether human ILC2 are potent sources of amphiregulin and mediate tissue repair. Also, it currently remains unclear whether ILC2 can promote tissue repair at different anatomical locations.

ILC3 and resolution of inflammation in lymphoid tissues and the intestine

ILC3 promote tissue repair through several distinct mechanisms (Fig. 3b, c). Systemic viral infections can impair the architecture of secondary lymphoid organs following CD8+ T cell-mediated killing of infected stromal cells91. If restoration of tissue homeostasis does not occur it can render a host susceptible to secondary infections. Disruption of lymphoid tissue architecture in mice promotes a local accumulation of LTi-like ILC3s that express LTα1β2 and act on LTβR-expressing stromal cells to enhance their proliferation and survival (Fig. 3b)91. Further, ILC3 can also promote tissue repair in the thymus of mice following total body irradiation92. In this context, CCR6+ ILC3 are partially radio-resistant and respond to irradiation-induced IL-23, promoting IL-22-dependent restoration of tissue integrity in the thymus92. IL-22 acts on thymic epithelial cells to promote cell survival and proliferation92. Further, IL-22-dependent tissue repair in mice has also been reported in the liver following chemical-induced hepatitis79 and lung following influenza infection or bleomycin-induced damage87,93,94, which are two other organs where the tissue resident non-hematopoietic cells highly express the IL-22 receptor8.

In the intestine, ILC3 play an important role in regulating tissue repair (Fig. 3c). This may be particularly important in the context of human inflammatory bowel disease (IBD) because several reports have identified reduced numbers of ILC3 in intestinal tissues from disease patients relative to non-IBD controls51,95,96. In mouse models, ILC3 production of IL-22 mediates tissue repair following experimental tissue damage induced by hematopoietic stem cell transplantation (HSCT) and subsequent graft versus host disease (GVHD), or administration of dextran sodium sulfate (DSS)9799. Following whole body irradiation and HSCT, radio-resistant ILC3 respond to DC-derived IL-23 and produce IL-22. IL-22 acts on the intestinal stem cell or progenitor compartments to limit apoptosis and preserve intestinal barrier function98. If alloreactive T cells with the capacity to attack donor tissues are co-transferred into mice following irradiation, this causes GVHD and is associated with a decrease in intestinal ILC3 and enhanced intestinal tissue damage due to a loss of IL-2298. Critically, in humans, ILC3 are not normally observed in the circulation, but are observed following chemotherapy for HSCT100. Furthermore, the levels of circulating ILC3s positively correlate with a reduced incidence of developing GVHD100, suggesting a critical role for human ILC3 in limiting GVHD.

In the DSS model of intestinal damage and inflammation, ILC3 responses and production of IL-22 also promote tissue repair and maintain intestinal barrier function82,97. This process is regulated by commensal bacteria, which induce expression of IL-25 by IECs following colonization in the postnatal period97,101. IL-25 acts on IL-25R+ DCs to subsequently limit ILC3 responses in a contact-dependent manner97. However, upon induction of experimental tissue damage with DSS, IL-25 expression was reduced and ILC3 responses were enhanced to mediated IL-22-dependent tissue repair97. ILC3 responses and IL-22-dependent tissue repair can also be enhanced in mice and humans following recognition of pathogenic or commensal microbes by CX3CR1+ myeloid cells and subsequent production of IL-1β, IL-23 and TL1A102105. One potential reason for the differential roles of commensal bacteria in promoting or suppressing ILC3 responses could be explained by the differential regulation of IL-1β secretion by pathogenic bacteria and selective subsets of commensal bacteria106,107. The vitamin A metabolite retinoic acid (RA) can also enhance ILC3 responses in mice through multiple mechanisms including direct binding to the Rorc or Il22 loci, promoting maturation of LTi-like ILC3, and regulating ILC3 proliferation81,99,108. In addition to promoting maintenance of the intestinal epithelium, during fetal development RA and Vitamin A promotes maturation of and larger secondary lymphoid tissues via LTi cells in mice, which could enhance protection from viral infections later in life108. Thus, ILC3 play a significant role in repairing damaged lymphoid tissues, airway epithelia, liver, and intestinal epithelia, thus preserving organ function.

ILCs promote chronic inflammation

ILCs can also promote chronic inflammation in several mouse models, and dysregulated ILC responses have been characterized in patient populations with chronic inflammatory disease of the lung, skin and intestine (Fig. 4).

Figure 4. ILCs can promote chronic inflammation.

Figure 4

(a) In response to infection or allergens ILC2 responses are elicited in the lung (and skin) by epithelial cell- and myeloid cell-derived IL-25, IL-33 and TSLP. Further, ILC2 responses can be enhanced by basophil-derived IL-4 or mast cell-derived prostoglandin D2 (PGD2). Activated ILC2 can subsequently promote chronic inflammation via IL-5-dependent eosinophil recruitment, IL-13-mediated contraction of smooth muscle cells, collagen deposition, and alternatively activated macrophage (AAMac) differentiation, or MHCII-mediated enhancement of Th2 cell responses, resulting in allergy and fibrosis. (b) In patients with psoriasis and mouse models of skin inflammation, ILC3 responses are increased, which can occur in response to DC-derived IL-23. ILC3 largely promote skin inflammation through production of IL-22 and IL-17. Further, ILC3 are increased in the BAL of patients with asthma and in mouse models of obesity-induced asthma. In mice this occurs through activation of the NLRP3 inflammasome and macrophage production of IL-1β. IL-1β activates ILC3 to produce IL-17 and directly mediate airway inflammation. (c) In the intestine ILC3 can promote IL-22-dependent progression of tumors, which is in part dependent upon DC-derived IL-23. Further, ILC3 can mediate tissue inflammation in the intestine in response to DC-derived IL-23 and IL-12. This may occur through production of IL-17 by ILC3, production of IFNγ following loss of RORγt in ILC3 and differentiation to ex-ILC3, or direct activation of tissue resident ILC1.

ILC2 and chronic inflammation in the lung and skin

Increased ILC2 responses have been observed in multiple allergic diseases, including the skin of individuals with atopic dermatitis63,90, in the nasal polyps of individuals with chronic rhinosinusitis32, circulating in the blood of individuals with asthma109, and in the bronchalveolar lavage of individuals with idiopathic pulmonary fibrosis110. In mouse models, dysregulated ILC2 responses contribute to chronic inflammation in the lung and skin (Fig. 4a). For example, ILC2 responses are elicited by epithelial cell- or myeloid cell-derived TSLP, IL-25 and IL-33 following exposure to allergens, chemicals, helminth parasites, or following influenza virus infection2932,63,90,111115. Basophils can also promote pro-inflammatory ILC2 responses in the skin and lung through production of IL-4 in mouse models of chemical-induced atopic dermatitis and protease allergen-induced airway inflammation116,117. Further, human mast cells co-localize near ILC2 in the human lung and could directly promote ILC2 responses in vitro through production of prostaglandin D2 (PGD2)118. The population expansion of ILC2 is thought to contribute to chronic inflammation through multiple mechanisms. Production of ILC2-dervied IL-5 can promote the recruitment of eosinophils into the lung and skin of mice, which contributes to tissue inflammation114. Further, ILC2-derived IL-13 can impair lung function of mice by enhancing airway smooth muscle cell contractility, increasing epithelial cell mucus production, polarizing macrophages to an alternatively activated phenotype (AAMacs) and increasing collagen deposition110,111. ILC2 in mice can also promote chronic inflammation by enhancing Th2 cell responses either indirectly by IL-13-elicited migration of activated DCs to the lung draining lymph node and subsequent Th2 cell priming119, or through direct MHCII-dependent interactions with CD4 T cells120,121.

ILC3, chronic inflammation and cancer in the skin, lung and intestine

Increases in ILC3 frequencies, cell numbers and cytokine production have also been observed in individuals with chronic inflammatory diseases, including in the skin of patients with psoriasis75,122, in tumors of patients with colitis-associated colon cancer123, or in the BAL of patients with asthma124. In a mouse model of psoriasis, ILC3 are the dominant source of IL-17 and IL-22 in the skin and are reduced by an p40-specific monoclonal antibody125, which likely blocks the IL-23-mediated population expansion or activation of ILC3. ILC3 are necessary and sufficient to induce psoriatic plaque formation in mice via production of IL-17 and IL-22 (Fig. 4b)125. Further, although normally absent in the lungs of healthy mice31, Umestu and colleagues identified in a mouse model of obesity-induced asthma that ILC3 expand in response to NLRP3-dependent production of IL-1β by macrophages124. Further ILC3 were sufficient to promote IL-17-dependent airway hyper-responsiveness in this mouse model124.

The role of ILC3 in promoting intestinal inflammation is more complex given the significant heterogeneity observed in this cell lineage, potential lineage plasticity, and differential gating strategies employed by different groups conducting human studies. Two reports observed an increased frequency in pro-inflammatory ILC3 in intestinal tissues from individuals with IBD126,127, while another identified increased ILC3 production of IL-22102. However, as described in the next section, there have been many reports of tissue protective functions of ILC3 in mouse models of intestinal inflammation, and additional reports of decreased ILC3 frequencies in IBD patients. A potential explanation for the different reports may be due to the selected mouse models, heterogeneity and potential plasticity of ILC3, as well as the clinical phenotypes and tissue sources of the patient populations studied. Notwithstanding this, one study elegantly demonstrated a role for a unique ILC3 population in two innate mouse models of intestinal inflammation (Fig. 4c). In that study, administration of anti-CD40 monoclonal antibody or colonization with Helicobacter hepaticus in Rag1/ mice induces colitis that is dependent upon ILCs, RORγt, IL-17 and IFNγ24. The pro-inflammatory ILC3 observed in these models are largely absent in the steady state, lack expression of c-kit, and a portion co-expressed T-bet, IFNγ and IL-17A24. ILCs with a similar phenotype are also observed in mice that lack both T-bet and Rag2 (TRUC mice), a spontaneous model of innate cell-driven intestinal inflammation76,128. TRUC mice have increased IL-17-producing ILCs relative to littermate controls, which act in synergy with DC-derived TNF to promote intestinal inflammation76,128. IL-6 was also found to be a critical cytokine that promotes IL-17 production from human and mouse pro-inflammatory ILC3127. ILC3 can also promote intestinal inflammation indirectly, as it has recently been demonstrated that following Toxoplasma gondii infection, ILC3-derived IL-22 acts on IECs to promote expression of the Th1 cell promoting cytokine IL-18, and subsequent tissue inflammation129. Further, IEC production of IL-18 was also important for ILC3 production of IL-22, indicating a previously unappreciated cross-regulatory circuit modulating inflammation and ILC3 responses129.

Some of the pro-inflammatory ILCs may arise from a loss of RORγt expression in ILC3 and an upregulation of T-bet68,72. In fate-mapping mouse models, infection with Salmonella enterica or anti-CD40 monoclonal antibody administration to Rag2−/− mice resulted in the development of ILCs that lost expression of RORγt, resemble ILC1 in expression of T-bet and IFNγ, and contribute to driving intestinal inflammation76,128. In human samples and humanized mice this transition of RORγt+ ILC3 to RORγt ex-ILC3 is promoted by IL-12 or can be induced following experimental induction of intestinal tissue damage with DSS51. Consistent with these findings, a novel gating strategy from above revealed reduced frequencies of ILC3 and increased frequencies of ILC1, in intestinal tissue from individuals with IBD patients relative to non-IBD controls51. Further, a unique intraepithelial ILC1 that develops independently of ILC3 were also enriched in intestinal tissues of individuals with IBD relative to non-IBD controls, and may contribute to the development of intestinal inflammation through production of IFNγ following administration of anti-CD40 monoclonal antibody to Rag2-deficient mice52.

In the colon, ILC3 also promote colitis-associated tumor progression via production of IL-22123. In a novel model of colitis-associated cancer, Rag1/ mice, which exhibit chronic intestinal inflammation when colonized with Helicobater hepaticus, were administered the carcinogen azoxymethane (AOM) and found to develop colitis-associated colorectal cancer in an ILC- and IL-22-dependent manner123. The timing and regulation of IL-22 production is important to consider in the context of intestinal tumors, as Flavell and colleagues recently demonstrated that IL-22 production during the peak of intestinal inflammation is protective against tumor formation, while uncontrolled IL-22 production during the resolution of inflammation promotes intestinal tumorigenesis130. In contrast, in an implantable model of mouse melanoma, ILCs that were previously marked with RORγt expression could promote IL-12-mediated tumor rejection131. In this mouse model, ILCs induced expression of adhesion molecules in the tumor vasculature, which subsequently enhanced recruitment of immune cells and anti-tumor immunity131, however this occurred in the context of a genetically modified tumor that overexpressed IL-12. Further, given that the characterized ILCs were described to previously express RORγt, the possibility remains that these anti-tumor ILC responses were mediated by ex-ILC3 that lost RORγt expression and now produced IFNγ and TNF. Further studies are required to define the role of ILCs in influencing pro- versus anti-tumor immune responses, and in particular a role for these cells in the presence of adaptive immunity is lacking. Collectively, these studies defined a complex role for ILC1, ILC2 and pro-inflammatory ILC3 or ex-ILC3 in promoting chronic inflammation in mice and humans.

ILCs limit chronic inflammation

ILCs can also play a significant role in limiting chronic inflammation either by influencing metabolic homeostasis, or by directly regulating innate and adaptive immune cell responses to non-harmful environmental stimuli in the intestine, such as commensal bacteria or dietary antigens (Fig. 5).

Figure 5. ILCs can prevent or limit chronic inflammation.

Figure 5

(a) In the intestine, ILC2 respond to epithelial cell-derived IL-33, IL-25 and vasoactive intestinal peptide (VIP) to promote IL-5 and IL-13-dependent recruitment of eosinophils and differentiation of alternatively activated macrophages (AAMacs). This process also occurs in the adipose tissues, however the sources of IL-25 or IL-33 are less well defined. Differentiation of AAMacs or direct stimulation of adipocytes with IL-13 or methionine-enkephalin peptides (met-enk) can promote metabolic homeostasis through a process known as beiging in the adipocytes. (b) ILC3 can limit chronic inflammation by regulating innate and adaptive immune responses in the intestine. ILC3 responses are induced in response to myeloid cell- and DC-derived IL-1β and IL-23 following recognition of pathogenic or commensal microbes. Production of ILC3-derived LTα1β2 or LTα3 can promote IgA production by B cells indirectly by modulating stromal cell or DC responses. Production of ILC3-derived GM-CSF can influence myeloid cell homeostasis to subsequently promote regulatory T cell (Treg) responses to food antigens. ILC3-intrisic MHCII can directly kill commensal bacteria-specific CD4 T cells with the potential to cause intestinal inflammation. Production of IL-22 by ILC3 can promote antimicrobial peptides by IECs to limit colonization with commensal bacteria, such as segmented filamentus bacteria (SFB), or regulate the anatomical localization of lymphoid tissue resident commensal bacteria. Further, ILC3-derived IL-22 can induce fucosylation of IECs to promote colonization with beneficial bacteria.

ILC2 and metabolic homeostasis

ILC2 have been characterized in the intestine, lung and adipose tissues of healthy humans and mice2933. In the intestine of mice, ILC2 controls the homeostasis of circulating eosinophils through constitutive production of IL-5, as well as tissue-resident eosinophils in the intestine through induced production of IL-13 and subsequent eotaxin expression132. Tissue recruitment of eosinophils in the intestine is regulated by nutrient intake and central circadian rhythms that induce vasoactive intestinal peptide (VIP) expression by IECs and stimulates ILC2 production of IL-13 through the VPAC2 receptor132. However, the functional significance of ILC2-mediated regulation of eosinophil homeostasis in the intestine is poorly understood. In adipose tissue of mice, ILC2 can regulate eosinophil homeostasis and alternatively activated macrophage polarization64,65. In obese mice fed a high-fat diet, or in obese humans, frequencies of ILC2 are reduced in the adipose tissues as compared to non-obese controls, and this reduction in ILC2 is further associated with a reciprocal increase in chronic low grade systemic inflammation6466, suggesting a critical role for ILC2 in regulating metabolic homeostasis. Consistent with this, experimental depletion of ILCs exacerbates weight gain in mice and causes insulin resistance, whereas treatment of mice on a high-fat diet with exogenous IL-25 or IL-33 increases ILC2 numbers and limits metabolic disease6466. Mechanistically, this occurred either indirectly through recruitment of eosinophils and differentiation of alternatively activated macrophage in adipose tissues, or by directly acting on adipocytes via ILC2-derived IL-13 or methionine-enkephalin peptides66,133. Activation of adipocytes through these pathways induces expression of the transcription factor Ucp1 and beiging of white adipocytes66,133, a process which induces thermogenesis, protects against insulin resistance and regulates metabolic homeostasis134. Additional research is required to further define how ILC2 orchestrate metabolic homeostasis, and whether this may also involve crosstalk with other IL-33R+ cells, such as recently described subsets of Tregs135137.

ILC3s in chronic systemic and intestinal inflammation

ILC3 also limit chronic inflammation through several distinct mechanisms (Fig. 5b), which may particularly important in the intestine, as there are several reports of reduced frequencies of ILC3 in individuals with IBD or following human immunodeficiency virus (HIV) infection relative to controls51,95,96,138141. In the intestine and associated lymphoid tissues of healthy humans, non-human primates and mice, ILC3 are a dominant source of IL-22 that is in part influenced by the colonization of the intestine with commensal bacteria6,142. Production of IL-22 restricts the anatomical localization or replication of specific species of commensal bacteria in mice. Impairment of this pathway promotes systemic dissemination of lymphoid tissue-resident commensal bacteria or increased colonization in the intestinal epithelium with segmented filamentous bacteria, resulting in low-grade systemic and intestinal inflammation142,143. This may be particularly important in the context of HIV infection and progression to acquired immune deficiency syndrome (AIDS), where there are defects in intestinal barrier function and systemic dissemination of commensal bacteria promote chronic immune activation and contribute to viral replication, loss of CD4 T cells and disease progression144. Strikingly, in humans and non-human primates, numbers of ILC3 and production of IL-22 are reduced in the intestine following pathogenic infection with HIV or SIV138141, and thus may be a novel therapeutic target to limit disease progression. Production of IL-22 in mice can also promote colonization of the intestine with beneficial and diverse commensal bacteria that provide protection from intestinal inflammation and infection8385. This occurs via ILC3- and IL-22-dependent fucosylation or glycosylation of IECs via Fut2, thus providing a sugar food source for beneficial microbiota, which in turn limit the growth of pathogens or opportunistic pathogens and protect from intestinal tissue damage8385. IL-22 production by ILC3 also requires maintained expression of Id2 and subsequent regulation of the IL-23R and Ahr pathway, which is necessary to limit infection-induced damage in mice by regulating colonization resistance and the composition of intestinal commensal bacteria145.

ILC3 can also limit chronic inflammation through indirect and direct regulation of the adaptive immune cell response (Fig. 5b)146. ILCs regulate homeostasis of myeloid cells in the intestine of mice through production of GM-CSF147. This process is regulated by macrophage sensing of intestinal commensal bacteria and production of IL-1β, which can act on ILC3 to promote GM-CSF expression147. Conversely, production of GM-CSF by ILC3 was essential to modulate myeloid cells and subsequently generate regulatory T cell responses to food antigens in the intestine and maintain oral tolerance147. Through production of LTα1β2 or a soluble LTα3, ILC3 can influence the production of T cell-independent and T cell-dependent IgA production in the intestine, respectively, which modulates the composition of the intestinal commensal bacteria148,149. Human and mouse ILC3 were found to also express MHCII, process and present antigens, and directly interact with CD4 T cells150. Genetic deletion of ILC3-intrisic MHCII in mice results in the development of spontaneous CD4 T cell-mediated intestinal inflammation, which was dependent upon commensal bacteria150,151. Mechanistically, MHCII+ ILC3 induced cell death of activated commensal bacteria-specific CD4 T cells in the intestine of mice, revealing a previously unappreciated selection pathway in the intestine whereby T cells with the potential to cause local inflammation are deleted, akin to the process that occurs for self-reactive T cells during thymic selection152. While no differences were observed in the frequency of ILC3 in intestinal biopsies of pediatric IBD patients, a significant reduction of MHCII on ILC3 was observed relative to non-IBD controls, and this reduction inversely correlated with increased intestinal Th17 cells152. These studies identify an essential role for ILC3 in maintaining tissue homeostasis and limiting chronic inflammation in the intestine of humans and mice. Further studies are necessary to interrogate what causes dysregulated ILC3 numbers or responses in the context of chronic human diseases, such as HIV infection or IBD.

Potential therapeutic modulation of ILCs

Given the role of ILCs in the initiation, regulation and resolution of inflammation in mice, and characterized alterations of ILCs in defined patient populations, there is an urgent need to investigate whether therapeutic strategies can be employed to modulate ILC responses and provide clinical benefit. As a proof of principle, ILC responses were recently shown to be modulated in patients with multiple sclerosis (MS) following anti-CD25 monoclonal antibody (daclizumab) therapy153. Although the role of ILCs in MS is poorly defined, this study identified increased circulating numbers of ILC3-like cells in individuals with MS that were reduced following anti-CD25 monoclonal antibody treatment and associated with reduced inflammatory markers in the cerebrospinal fluid153. Despite this advance, further high resolution profiling of ILC responses in defined patient populations, and during defined treatment strategies, is required to fully elucidate how we can modulate ILCs to limit human disease. There are many other therapeutics in the clinic or currently under development that could likely influence ILC differentiation, homeostasis or function. These include targeting the cytokine-cytokine receptor pathways that are critical for the differentiation, function and maintenance of ILCs such as IL-2-IL-2R, IL-12-IL-12R, IL-23-IL-23R, IL-1-IL-1R, TSLP-TSLPR and IL-6-IL-6R, targeting molecules critical for migration of ILCs such as α4β7 and MAdCAM-1, or targeting effector molecules of ILCs such as TNF-TNFR, IL-17-IL-17R, IFNγ and IL-13-IL-13R. It will be important to consider how these strategies may influence the pathologic versus protective functions of ILCs. For example, targeting the IL-23-IL-17 pathway has demonstrated efficacy in psoriasis and rheumatoid arthrisits154157. However in IBD patients, blockade of IL-17 had limited efficacy and in some cases resulted in enhanced disease and susceptibility to fungal infections158161. Given the role of ILC3 and IL-17 in promoting anti-fungal immunity86, one possibility is that targeting IL-17 may have limited efficacy in certain conditions as it also targeted protective ILC3 responses. Therefore, the design of novel therapeutics may be necessary in order to find strategies that selectively modulate protective versus pathologic ILC responses. These could include novel small molecule inhibitors of transcription factors, other recently identified ILC modulators in mice and humans, such as the vitamin A metabolite retinoic acid, Lipoxin A4, or exogenous cytokines that may promote protective functions of ILCs or limit their pathologic potential81,99,108,118.

Outstanding questions and future directions

Research on the biology of ILCs has already advanced our understanding of their development and the role they play in regulating acute and chronic inflammation as well as tissue repair. One future challenge is to better define this family of innate immune cells, and delineate how they specifically interact with other innate, adaptive and non-hematopoietic cells to promote, limit or resolve inflammation. A universal consensus of gating strategies on human and murine ILCs should be considered, as well as critical evaluation of translational studies, such as number of patients, current medication usage, longitudinal samples, source of healthy or non-diseased control tissues, tissue digestion protocols, and genetic and environmental factors like commensal bacteria. Further, additional mouse studies are needed to carefully define the potential plasticity of ILC populations, and identify novel functions and regulatory pathways influencing ILC responses. Defining these outstanding questions could prompt the development of novel therapeutic strategies, or promote new approaches that will permit selective regulation of protective versus pathologic ILC responses. These advances will be critical to advance our understanding of the cellular and molecular basis of inflammation and determine whether and how we can manipulate ILC responses to maintain healthy tissues and limit chronic inflammation.

Acknowledgments

Research in the Sonnenberg laboratory is supported by the National Institutes of Health (DP5OD012116), the NIAID Mucosal Immunology Studies Team (MIST) Scholar Award in Mucosal Immunity and the Institute for Translational Medicine and Therapeutics Transdisciplinary Program in Translational Medicine and Therapeutics (UL1-RR024134 from the US National Center for Research Resources). Research in the Artis lab is supported by the National Institutes of Health (AI061570, AI074878, AI095466, AI095608, AI102942, AI097333 and AI106697), the Burroughs Wellcome Fund Investigator in Pathogenesis of Infectious Disease Award and Crohn’s and Colitis Foundation of America.

References

  • 1.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008;454:428–435. doi: 10.1038/nature07201. [DOI] [PubMed] [Google Scholar]
  • 3.Spits H, et al. Innate lymphoid cells--a proposal for uniform nomenclature. Nat Rev Immunol. 2013;13:145–149. doi: 10.1038/nri3365. [DOI] [PubMed] [Google Scholar]
  • 4.Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells--how did we miss them? Nat Rev Immunol. 2013;13:75–87. doi: 10.1038/nri3349. [DOI] [PubMed] [Google Scholar]
  • 5.Sonnenberg GF, Mjosberg J, Spits H, Artis D. SnapShot: innate lymphoid cells. Immunity. 2013;39:622–622. e621. doi: 10.1016/j.immuni.2013.08.021. [DOI] [PubMed] [Google Scholar]
  • 6.Sonnenberg GF, Artis D. Innate lymphoid cell interactions with microbiota: implications for intestinal health and disease. Immunity. 2012;37:601–610. doi: 10.1016/j.immuni.2012.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zheng Y, et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat Med. 2008;14:282–289. doi: 10.1038/nm1720. [DOI] [PubMed] [Google Scholar]
  • 8.Sonnenberg GF, Fouser LA, Artis D. Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat Immunol. 2011;12:383–390. doi: 10.1038/ni.2025. [DOI] [PubMed] [Google Scholar]
  • 9.Aujla SJ, et al. IL-22 mediates mucosal host defense against Gram-negative bacterial pneumonia. Nat Med. 2008;14:275–281. doi: 10.1038/nm1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ouyang W, Kolls JK, Zheng Y. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity. 2008;28:454–467. doi: 10.1016/j.immuni.2008.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Harrington LE, Mangan PR, Weaver CT. Expanding the effector CD4 T-cell repertoire: the Th17 lineage. Curr Opin Immunol. 2006;18:349–356. doi: 10.1016/j.coi.2006.03.017. [DOI] [PubMed] [Google Scholar]
  • 12.Ahern PP, Izcue A, Maloy KJ, Powrie F. The interleukin-23 axis in intestinal inflammation. Immunol Rev. 2008;226:147–159. doi: 10.1111/j.1600-065X.2008.00705.x. [DOI] [PubMed] [Google Scholar]
  • 13.Fallon PG, et al. Identification of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, and IL-13 at the onset of helminth expulsion. J Exp Med. 2006;203:1105–1116. doi: 10.1084/jem.20051615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Saenz SA, Taylor BC, Artis D. Welcome to the neighborhood: epithelial cell-derived cytokines license innate and adaptive immune responses at mucosal sites. Immunol Rev. 2008;226:172–190. doi: 10.1111/j.1600-065X.2008.00713.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Liew FY, Pitman NI, McInnes IB. Disease-associated functions of IL-33: the new kid in the IL-1 family. Nat Rev Immunol. 2010;10:103–110. doi: 10.1038/nri2692. [DOI] [PubMed] [Google Scholar]
  • 16.Liu YJ, et al. TSLP: an epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation. Annu Rev Immunol. 2007;25:193–219. doi: 10.1146/annurev.immunol.25.022106.141718. [DOI] [PubMed] [Google Scholar]
  • 17.Fort MM, et al. IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity. 2001;15:985–995. doi: 10.1016/s1074-7613(01)00243-6. [DOI] [PubMed] [Google Scholar]
  • 18.Kiessling R, Klein E, Pross H, Wigzell H. “Natural” killer cells in the mouse. II. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Characteristics of the killer cell. Eur J Immunol. 1975;5:117–121. doi: 10.1002/eji.1830050209. [DOI] [PubMed] [Google Scholar]
  • 19.Pross HF, Jondal M. Cytotoxic lymphocytes from normal donors. A functional marker of human non-T lymphocytes. Clin Exp Immunol. 1975;21:226–235. [PMC free article] [PubMed] [Google Scholar]
  • 20.Mebius RE, Rennert P, Weissman IL. Developing lymph nodes collect CD4+CD3- LTbeta+ cells that can differentiate to APC, NK cells, and follicular cells but not T or B cells. Immunity. 1997;7:493–504. doi: 10.1016/s1074-7613(00)80371-4. [DOI] [PubMed] [Google Scholar]
  • 21.Sonnenberg GF, Monticelli LA, Elloso MM, Fouser LA, Artis D. CD4(+) lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity. 2011;34:122–134. doi: 10.1016/j.immuni.2010.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Cella M, et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature. 2009;457:722–725. doi: 10.1038/nature07537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Satoh-Takayama N, et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity. 2008;29:958–970. doi: 10.1016/j.immuni.2008.11.001. [DOI] [PubMed] [Google Scholar]
  • 24.Buonocore S, et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature. 2010;464:1371–1375. doi: 10.1038/nature08949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sawa S, et al. Lineage relationship analysis of RORgammat+ innate lymphoid cells. Science. 2010;330:665–669. doi: 10.1126/science.1194597. [DOI] [PubMed] [Google Scholar]
  • 26.Cupedo T, et al. Human fetal lymphoid tissue-inducer cells are interleukin 17-producing precursors to RORC+ CD127+ natural killer-like cells. Nat Immunol. 2009;10:66–74. doi: 10.1038/ni.1668. [DOI] [PubMed] [Google Scholar]
  • 27.Crellin NK, et al. Regulation of cytokine secretion in human CD127(+) LTi-like innate lymphoid cells by Toll-like receptor 2. Immunity. 2010;33:752–764. doi: 10.1016/j.immuni.2010.10.012. [DOI] [PubMed] [Google Scholar]
  • 28.Sanos SL, et al. RORgammat and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat Immunol. 2009;10:83–91. doi: 10.1038/ni.1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Moro K, et al. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. Nature. 2010;463:540–544. doi: 10.1038/nature08636. [DOI] [PubMed] [Google Scholar]
  • 30.Price AE, et al. Systemically dispersed innate IL-13-expressing cells in type 2 immunity. Proc Natl Acad Sci U S A. 2010;107:11489–11494. doi: 10.1073/pnas.1003988107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Monticelli LA, et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat Immunol. 2011;12:1045–1054. doi: 10.1031/ni.2131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mjosberg JM, et al. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat Immunol. 2011;12:1055–1062. doi: 10.1038/ni.2104. [DOI] [PubMed] [Google Scholar]
  • 33.Neill DR, et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature. 2010;464:1367–1370. doi: 10.1038/nature08900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cherrier M, Sawa S, Eberl G. Notch, Id2, and RORgammat sequentially orchestrate the fetal development of lymphoid tissue inducer cells. J Exp Med. 2012;209:729–740. doi: 10.1084/jem.20111594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Constantinides MG, McDonald BD, Verhoef PA, Bendelac A. A committed precursor to innate lymphoid cells. Nature. 2014;508:397–401. doi: 10.1038/nature13047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Klose CS, et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell. 2014;157:340–356. doi: 10.1016/j.cell.2014.03.030. [DOI] [PubMed] [Google Scholar]
  • 37.Diefenbach A, Colonna M, Koyasu S. Development, differentiation, and diversity of innate lymphoid cells. Immunity. 2014;41:354–365. doi: 10.1016/j.immuni.2014.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Artis D, Spits H. The biology of innate lymphoid cells. Nature. 2015;517:293–301. doi: 10.1038/nature14189. [DOI] [PubMed] [Google Scholar]
  • 39.Xu W, et al. NFIL3 Orchestrates the Emergence of Common Helper Innate Lymphoid Cell Precursors. Cell Rep. 2015 doi: 10.1016/j.celrep.2015.02.057. [DOI] [PubMed] [Google Scholar]
  • 40.Geiger TL, et al. Nfil3 is crucial for development of innate lymphoid cells and host protection against intestinal pathogens. J Exp Med. 2014;211:1723–1731. doi: 10.1084/jem.20140212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Yu X, et al. The basic leucine zipper transcription factor NFIL3 directs the development of a common innate lymphoid cell precursor. Elife. 2014;3 doi: 10.7554/eLife.04406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Seillet C, et al. Nfil3 is required for the development of all innate lymphoid cell subsets. J Exp Med. 2014;211:1733–1740. doi: 10.1084/jem.20140145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kobayashi T, et al. NFIL3-deficient mice develop microbiota-dependent, IL-12/23-driven spontaneous colitis. J Immunol. 2014;192:1918–1927. doi: 10.4049/jimmunol.1301819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Seehus CR, et al. The development of innate lymphoid cells requires TOX-dependent generation of a common innate lymphoid cell progenitor. Nat Immunol. 2015 doi: 10.1038/ni.3168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Aliahmad P, de la Torre B, Kaye J. Shared dependence on the DNA-binding factor TOX for the development of lymphoid tissue-inducer cell and NK cell lineages. Nat Immunol. 2010;11:945–952. doi: 10.1038/ni.1930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yang Q, et al. T cell factor 1 is required for group 2 innate lymphoid cell generation. Immunity. 2013;38:694–704. doi: 10.1016/j.immuni.2012.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mielke LA, et al. TCF-1 Controls ILC2 and NKp46+RORgammat+ Innate Lymphocyte Differentiation and Protection in Intestinal Inflammation. J Immunol. 2013;191:4383–4391. doi: 10.4049/jimmunol.1301228. [DOI] [PubMed] [Google Scholar]
  • 48.Yagi R, et al. The transcription factor GATA3 is critical for the development of all IL-7Ralpha-expressing innate lymphoid cells. Immunity. 2014;40:378–388. doi: 10.1016/j.immuni.2014.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Serafini N, et al. Gata3 drives development of RORgammat+ group 3 innate lymphoid cells. J Exp Med. 2014;211:199–208. doi: 10.1084/jem.20131038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Montaldo E, et al. Human RORgammat(+)CD34(+) cells are lineage-specified progenitors of group 3 RORgammat(+) innate lymphoid cells. Immunity. 2014;41:988–1000. doi: 10.1016/j.immuni.2014.11.010. [DOI] [PubMed] [Google Scholar]
  • 51.Bernink JH, et al. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat Immunol. 2013;14:221–229. doi: 10.1038/ni.2534. [DOI] [PubMed] [Google Scholar]
  • 52.Fuchs A, et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-gamma-producing cells. Immunity. 2013;38:769–781. doi: 10.1016/j.immuni.2013.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hoyler T, et al. The transcription factor GATA-3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity. 2012;37:634–648. doi: 10.1016/j.immuni.2012.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mjosberg J, et al. The transcription factor GATA3 is essential for the function of human type 2 innate lymphoid cells. Immunity. 2012;37:649–659. doi: 10.1016/j.immuni.2012.08.015. [DOI] [PubMed] [Google Scholar]
  • 55.Klein Wolterink RG, et al. Essential, dose-dependent role for the transcription factor Gata3 in the development of IL-5+ and IL-13+ type 2 innate lymphoid cells. Proc Natl Acad Sci U S A. 2013;110:10240–10245. doi: 10.1073/pnas.1217158110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Furusawa J, et al. Critical role of p38 and GATA3 in natural helper cell function. J Immunol. 2013;191:1818–1826. doi: 10.4049/jimmunol.1300379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wong SH, et al. Transcription factor RORalpha is critical for nuocyte development. Nat Immunol. 2012;13:229–236. doi: 10.1038/ni.2208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Halim TY, et al. Retinoic-acid-receptor-related orphan nuclear receptor alpha is required for natural helper cell development and allergic inflammation. Immunity. 2012;37:463–474. doi: 10.1016/j.immuni.2012.06.012. [DOI] [PubMed] [Google Scholar]
  • 59.Robinette ML, et al. Transcriptional programs define molecular characteristics of innate lymphoid cell classes and subsets. Nat Immunol. 2015;16:306–317. doi: 10.1038/ni.3094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Spooner CJ, et al. Specification of type 2 innate lymphocytes by the transcriptional determinant Gfi1. Nat Immunol. 2013;14:1229–1236. doi: 10.1038/ni.2743. [DOI] [PubMed] [Google Scholar]
  • 61.Walker JA, et al. Bcl11b is essential for group 2 innate lymphoid cell development. J Exp Med. 2015 doi: 10.1084/jem.20142224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yu Y, et al. The transcription factor Bcl11b is specifically expressed in group 2 innate lymphoid cells and is essential for their development. J Exp Med. 2015 doi: 10.1084/jem.20142318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kim BS, et al. TSLP elicits IL-33-independent innate lymphoid cell responses to promote skin inflammation. Sci Transl Med. 2013;5:170ra116. doi: 10.1126/scitranslmed.3005374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Molofsky AB, et al. Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages. J Exp Med. 2013;210:535–549. doi: 10.1084/jem.20121964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hams E, Locksley RM, McKenzie AN, Fallon PG. Cutting edge: IL-25 elicits innate lymphoid type 2 and type II NKT cells that regulate obesity in mice. J Immunol. 2013;191:5349–5353. doi: 10.4049/jimmunol.1301176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Brestoff JR, et al. Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature. 2015;519:242–246. doi: 10.1038/nature14115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Eberl G, et al. An essential function for the nuclear receptor RORgamma(t) in the generation of fetal lymphoid tissue inducer cells. Nat Immunol. 2004;5:64–73. doi: 10.1038/ni1022. [DOI] [PubMed] [Google Scholar]
  • 68.Klose CS, et al. A T-bet gradient controls the fate and function of CCR6-RORgammat+ innate lymphoid cells. Nature. 2013;494:261–265. doi: 10.1038/nature11813. [DOI] [PubMed] [Google Scholar]
  • 69.Kiss EA, et al. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science. 2011;334:1561–1565. doi: 10.1126/science.1214914. [DOI] [PubMed] [Google Scholar]
  • 70.Lee JS, et al. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat Immunol. 2012;13:144–151. doi: 10.1038/ni.2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Qiu J, et al. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity. 2012;36:92–104. doi: 10.1016/j.immuni.2011.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Vonarbourg C, et al. Regulated expression of nuclear receptor RORgammat confers distinct functional fates to NK cell receptor-expressing RORgammat(+) innate lymphocytes. Immunity. 2010;33:736–751. doi: 10.1016/j.immuni.2010.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Bouskra D, et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature. 2008;456:507–510. doi: 10.1038/nature07450. [DOI] [PubMed] [Google Scholar]
  • 74.Mackley EC, et al. CCR7-dependent trafficking of RORgamma(+) ILCs creates a unique microenvironment within mucosal draining lymph nodes. Nat Commun. 2015;6:5862. doi: 10.1038/ncomms6862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Teunissen MB, et al. Composition of innate lymphoid cell subsets in the human skin: enrichment of NCR(+) ILC3 in lesional skin and blood of psoriasis patients. J Invest Dermatol. 2014;134:2351–2360. doi: 10.1038/jid.2014.146. [DOI] [PubMed] [Google Scholar]
  • 76.Powell N, et al. The transcription factor T-bet regulates intestinal inflammation mediated by interleukin-7 receptor+ innate lymphoid cells. Immunity. 2012;37:674–684. doi: 10.1016/j.immuni.2012.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Huang Y, et al. IL-25-responsive, lineage-negative KLRG1(hi) cells are multipotential ‘inflammatory’ type 2 innate lymphoid cells. Nat Immunol. 2015;16:161–169. doi: 10.1038/ni.3078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sun JC, Lanier LL. NK cell development, homeostasis and function: parallels with CD8(+) T cells. Nat Rev Immunol. 2011;11:645–657. doi: 10.1038/nri3044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Maizels RM, Hewitson JP, Smith KA. Susceptibility and immunity to helminth parasites. Curr Opin Immunol. 2012;24:459–466. doi: 10.1016/j.coi.2012.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Artis D, et al. RELMbeta/FIZZ2 is a goblet cell-specific immune-effector molecule in the gastrointestinal tract. Proc Natl Acad Sci U S A. 2004;101:13596–13600. doi: 10.1073/pnas.0404034101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Spencer SP, et al. Adaptation of innate lymphoid cells to a micronutrient deficiency promotes type 2 barrier immunity. Science. 2014;343:432–437. doi: 10.1126/science.1247606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sugimoto K, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest. 2008;118:534–544. doi: 10.1172/JCI33194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Pham TA, et al. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization resistance to an opportunistic pathogen. Cell Host Microbe. 2014;16:504–516. doi: 10.1016/j.chom.2014.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Goto Y, et al. Innate lymphoid cells regulate intestinal epithelial cell glycosylation. Science. 2014;345:1254009. doi: 10.1126/science.1254009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Pickard JM, et al. Rapid fucosylation of intestinal epithelium sustains host-commensal symbiosis in sickness. Nature. 2014;514:638–641. doi: 10.1038/nature13823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Gladiator A, Wangler N, Trautwein-Weidner K, LeibundGut-Landmann S. Cutting edge: IL-17-secreting innate lymphoid cells are essential for host defense against fungal infection. J Immunol. 2013;190:521–525. doi: 10.4049/jimmunol.1202924. [DOI] [PubMed] [Google Scholar]
  • 87.Sonnenberg GF, et al. Pathological versus protective functions of IL-22 in airway inflammation are regulated by IL-17A. J Exp Med. 2010;207:1293–1305. doi: 10.1084/jem.20092054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Deshmukh HS, et al. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat Med. 2014;20:524–530. doi: 10.1038/nm.3542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Turner JE, et al. IL-9-mediated survival of type 2 innate lymphoid cells promotes damage control in helminth-induced lung inflammation. J Exp Med. 2013;210:2951–2965. doi: 10.1084/jem.20130071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Salimi M, et al. A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J Exp Med. 2013;210:2939–2950. doi: 10.1084/jem.20130351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Scandella E, et al. Restoration of lymphoid organ integrity through the interaction of lymphoid tissue-inducer cells with stroma of the T cell zone. Nat Immunol. 2008;9:667–675. doi: 10.1038/ni.1605. [DOI] [PubMed] [Google Scholar]
  • 92.Dudakov JA, et al. Interleukin-22 drives endogenous thymic regeneration in mice. Science. 2012;336:91–95. doi: 10.1126/science.1218004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Matsumoto A, et al. IL-22-producing RORgammat-dependent innate lymphoid cells play a novel protective role in murine acute hepatitis. PLoS One. 2013;8:e62853. doi: 10.1371/journal.pone.0062853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kumar P, Thakar MS, Ouyang W, Malarkannan S. IL-22 from conventional NK cells is epithelial regenerative and inflammation protective during influenza infection. Mucosal Immunol. 2013;6:69–82. doi: 10.1038/mi.2012.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Takayama T, et al. Imbalance of NKp44(+)NKp46(-) and NKp44(-)NKp46(+) natural killer cells in the intestinal mucosa of patients with Crohn’s disease. Gastroenterology. 2010;139:882–892. 892 e881–883. doi: 10.1053/j.gastro.2010.05.040. [DOI] [PubMed] [Google Scholar]
  • 96.Ciccia F, et al. Interleukin-22 and interleukin-22-producing NKp44+ natural killer cells in subclinical gut inflammation in ankylosing spondylitis. Arthritis Rheum. 2012;64:1869–1878. doi: 10.1002/art.34355. [DOI] [PubMed] [Google Scholar]
  • 97.Sawa S, et al. RORgammat+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat Immunol. 2011;12:320–326. doi: 10.1038/ni.2002. [DOI] [PubMed] [Google Scholar]
  • 98.Hanash AM, et al. Interleukin-22 protects intestinal stem cells from immune-mediated tissue damage and regulates sensitivity to graft versus host disease. Immunity. 2012;37:339–350. doi: 10.1016/j.immuni.2012.05.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mielke LA, et al. Retinoic acid expression associates with enhanced IL-22 production by gammadelta T cells and innate lymphoid cells and attenuation of intestinal inflammation. J Exp Med. 2013;210:1117–1124. doi: 10.1084/jem.20121588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Munneke JM, et al. Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease. Blood. 2014;124:812–821. doi: 10.1182/blood-2013-11-536888. [DOI] [PubMed] [Google Scholar]
  • 101.Zaph C, et al. Commensal-dependent expression of IL-25 regulates the IL-23-IL-17 axis in the intestine. J Exp Med. 2008;205:2191–2198. doi: 10.1084/jem.20080720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Longman RS, et al. CX(3)CR1(+) mononuclear phagocytes support colitis-associated innate lymphoid cell production of IL-22. J Exp Med. 2014;211:1571–1583. doi: 10.1084/jem.20140678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Manta C, et al. CX(3)CR1(+) macrophages support IL-22 production by innate lymphoid cells during infection with Citrobacter rodentium. Mucosal Immunol. 2013;6:177–188. doi: 10.1038/mi.2012.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Aychek T, et al. IL-23-mediated mononuclear phagocyte crosstalk protects mice from Citrobacter rodentium-induced colon immunopathology. Nat Commun. 2015;6:6525. doi: 10.1038/ncomms7525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Satoh-Takayama N, et al. The chemokine receptor CXCR6 controls the functional topography of interleukin-22 producing intestinal innate lymphoid cells. Immunity. 2014;41:776–788. doi: 10.1016/j.immuni.2014.10.007. [DOI] [PubMed] [Google Scholar]
  • 106.Franchi L, et al. NLRC4-driven production of IL-1beta discriminates between pathogenic and commensal bacteria and promotes host intestinal defense. Nat Immunol. 2012;13:449–456. doi: 10.1038/ni.2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Seo SU, et al. Distinct Commensals Induce Interleukin-1beta via NLRP3 Inflammasome in Inflammatory Monocytes to Promote Intestinal Inflammation in Response to Injury. Immunity. 2015;42:744–755. doi: 10.1016/j.immuni.2015.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.van de Pavert SA, et al. Maternal retinoids control type 3 innate lymphoid cells and set the offspring immunity. Nature. 2014;508:123–127. doi: 10.1038/nature13158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bartemes KR, Kephart GM, Fox SJ, Kita H. Enhanced innate type 2 immune response in peripheral blood from patients with asthma. J Allergy Clin Immunol. 2014;134:671–678. e674. doi: 10.1016/j.jaci.2014.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hams E, et al. IL-25 and type 2 innate lymphoid cells induce pulmonary fibrosis. Proc Natl Acad Sci U S A. 2014;111:367–372. doi: 10.1073/pnas.1315854111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Chang YJ, et al. Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat Immunol. 2011;12:631–638. doi: 10.1038/ni.2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Halim TY, Krauss RH, Sun AC, Takei F. Lung natural helper cells are a critical source of Th2 cell-type cytokines in protease allergen-induced airway inflammation. Immunity. 2012;36:451–463. doi: 10.1016/j.immuni.2011.12.020. [DOI] [PubMed] [Google Scholar]
  • 113.Bartemes KR, et al. IL-33-responsive lineage- CD25+ CD44(hi) lymphoid cells mediate innate type 2 immunity and allergic inflammation in the lungs. J Immunol. 2012;188:1503–1513. doi: 10.4049/jimmunol.1102832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Imai Y, et al. Skin-specific expression of IL-33 activates group 2 innate lymphoid cells and elicits atopic dermatitis-like inflammation in mice. Proc Natl Acad Sci U S A. 2013;110:13921–13926. doi: 10.1073/pnas.1307321110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kabata H, et al. Thymic stromal lymphopoietin induces corticosteroid resistance in natural helper cells during airway inflammation. Nat Commun. 2013;4:2675. doi: 10.1038/ncomms3675. [DOI] [PubMed] [Google Scholar]
  • 116.Kim BS, et al. Basophils promote innate lymphoid cell responses in inflamed skin. J Immunol. 2014;193:3717–3725. doi: 10.4049/jimmunol.1401307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Motomura Y, et al. Basophil-derived interleukin-4 controls the function of natural helper cells, a member of ILC2s, in lung inflammation. Immunity. 2014;40:758–771. doi: 10.1016/j.immuni.2014.04.013. [DOI] [PubMed] [Google Scholar]
  • 118.Barnig C, et al. Lipoxin A4 regulates natural killer cell and type 2 innate lymphoid cell activation in asthma. Sci Transl Med. 2013;5:174ra126. doi: 10.1126/scitranslmed.3004812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Halim TY, et al. Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation. Immunity. 2014;40:425–435. doi: 10.1016/j.immuni.2014.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Oliphant CJ, et al. MHCII-mediated dialog between group 2 innate lymphoid cells and CD4(+) T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Immunity. 2014;41:283–295. doi: 10.1016/j.immuni.2014.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Mirchandani AS, et al. Type 2 innate lymphoid cells drive CD4+ Th2 cell responses. J Immunol. 2014;192:2442–2448. doi: 10.4049/jimmunol.1300974. [DOI] [PubMed] [Google Scholar]
  • 122.Villanova F, et al. Characterization of innate lymphoid cells in human skin and blood demonstrates increase of NKp44+ ILC3 in psoriasis. J Invest Dermatol. 2014;134:984–991. doi: 10.1038/jid.2013.477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kirchberger S, et al. Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J Exp Med. 2013;210:917–931. doi: 10.1084/jem.20122308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kim HY, et al. Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat Med. 2014;20:54–61. doi: 10.1038/nm.3423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Pantelyushin S, et al. Rorgammat+ innate lymphocytes and gammadelta T cells initiate psoriasiform plaque formation in mice. J Clin Invest. 2012;122:2252–2256. doi: 10.1172/JCI61862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Geremia A, et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J Exp Med. 2011;208:1127–1133. doi: 10.1084/jem.20101712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Powell N, et al. Interleukin-6 Increases Production of Cytokines by Colonic Innate Lymphoid Cells in Mice and Patients with Chronic Intestinal Inflammation. Gastroenterology. 2015 doi: 10.1053/j.gastro.2015.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Ermann J, Staton T, Glickman JN, de Waal Malefyt R, Glimcher LH. Nod/Ripk2 signaling in dendritic cells activates IL-17A-secreting innate lymphoid cells and drives colitis in T-bet−/−.Rag2−/− (TRUC) mice. Proc Natl Acad Sci U S A. 2014;111:E2559–2566. doi: 10.1073/pnas.1408540111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Munoz M, et al. Interleukin-22 induces interleukin-18 expression from epithelial cells during intestinal infection. Immunity. 2015;42:321–331. doi: 10.1016/j.immuni.2015.01.011. [DOI] [PubMed] [Google Scholar]
  • 130.Huber S, et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. 2012;491:259–263. doi: 10.1038/nature11535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Eisenring M, vom Berg J, Kristiansen G, Saller E, Becher B. IL-12 initiates tumor rejection via lymphoid tissue-inducer cells bearing the natural cytotoxicity receptor NKp46. Nat Immunol. 2010;11:1030–1038. doi: 10.1038/ni.1947. [DOI] [PubMed] [Google Scholar]
  • 132.Nussbaum JC, et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature. 2013;502:245–248. doi: 10.1038/nature12526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Lee MW, et al. Activated type 2 innate lymphoid cells regulate beige fat biogenesis. Cell. 2015;160:74–87. doi: 10.1016/j.cell.2014.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Brestoff JR, Artis D. Immune Regulation of Metabolic Homeostasis in Health and Disease. Cell. 2015;161:146–160. doi: 10.1016/j.cell.2015.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Vasanthakumar A, et al. The transcriptional regulators IRF4, BATF and IL-33 orchestrate development and maintenance of adipose tissue-resident regulatory T cells. Nat Immunol. 2015;16:276–285. doi: 10.1038/ni.3085. [DOI] [PubMed] [Google Scholar]
  • 136.Burzyn D, et al. A special population of regulatory T cells potentiates muscle repair. Cell. 2013;155:1282–1295. doi: 10.1016/j.cell.2013.10.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Schiering C, et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature. 2014;513:564–568. doi: 10.1038/nature13577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Klatt NR, et al. Loss of mucosal CD103+ DCs and IL-17+ and IL-22+ lymphocytes is associated with mucosal damage in SIV infection. Mucosal Immunol. 2012;5:646–657. doi: 10.1038/mi.2012.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Gray EE, Friend S, Suzuki K, Phan TG, Cyster JG. Subcapsular sinus macrophage fragmentation and CD169+ bleb acquisition by closely associated IL-17-committed innate-like lymphocytes. PLoS One. 2012;7:e38258. doi: 10.1371/journal.pone.0038258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kim CJ, et al. A role for mucosal IL-22 production and Th22 cells in HIV-associated mucosal immunopathogenesis. Mucosal Immunol. 2012;5:670–680. doi: 10.1038/mi.2012.72. [DOI] [PubMed] [Google Scholar]
  • 141.Li H, et al. Hypercytotoxicity and rapid loss of NKp44+ innate lymphoid cells during acute SIV infection. PLoS Pathog. 2014;10:e1004551. doi: 10.1371/journal.ppat.1004551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Sonnenberg GF, et al. Innate lymphoid cells promote anatomical containment of lymphoid-resident commensal bacteria. Science. 2012;336:1321–1325. doi: 10.1126/science.1222551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Qiu J, et al. Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon receptor signaling and regulation of microflora. Immunity. 2013;39:386–399. doi: 10.1016/j.immuni.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Brenchley JM, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
  • 145.Guo X, et al. Innate Lymphoid Cells Control Early Colonization Resistance against Intestinal Pathogens through ID2-Dependent Regulation of the Microbiota. Immunity. 2015;42:731–743. doi: 10.1016/j.immuni.2015.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hepworth MR, Sonnenberg GF. Regulation of the adaptive immune system by innate lymphoid cells. Curr Opin Immunol. 2014;27:75–82. doi: 10.1016/j.coi.2014.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Mortha A, et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science. 2014;343:1249288. doi: 10.1126/science.1249288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Kruglov AA, et al. Nonredundant function of soluble LTalpha3 produced by innate lymphoid cells in intestinal homeostasis. Science. 2013;342:1243–1246. doi: 10.1126/science.1243364. [DOI] [PubMed] [Google Scholar]
  • 149.Tsuji M, et al. Requirement for lymphoid tissue-inducer cells in isolated follicle formation and T cell-independent immunoglobulin A generation in the gut. Immunity. 2008;29:261–271. doi: 10.1016/j.immuni.2008.05.014. [DOI] [PubMed] [Google Scholar]
  • 150.Hepworth MR, et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature. 2013;498:113–117. doi: 10.1038/nature12240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Goto Y, et al. Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Th17 cell differentiation. Immunity. 2014;40:594–607. doi: 10.1016/j.immuni.2014.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Hepworth MR, et al. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4+ T cells. Science. 2015 doi: 10.1126/science.aaa4812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Perry JS, et al. Inhibition of LTi cell development by CD25 blockade is associated with decreased intrathecal inflammation in multiple sclerosis. Sci Transl Med. 2012;4:145ra106. doi: 10.1126/scitranslmed.3004140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Papp KA, et al. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N Engl J Med. 2012;366:1181–1189. doi: 10.1056/NEJMoa1109017. [DOI] [PubMed] [Google Scholar]
  • 155.Leonardi C, et al. Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N Engl J Med. 2012;366:1190–1199. doi: 10.1056/NEJMoa1109997. [DOI] [PubMed] [Google Scholar]
  • 156.Genovese MC, et al. LY2439821, a humanized anti-interleukin-17 monoclonal antibody, in the treatment of patients with rheumatoid arthritis: A phase I randomized, double-blind, placebo-controlled, proof-of-concept study. Arthritis Rheum. 2010;62:929–939. doi: 10.1002/art.27334. [DOI] [PubMed] [Google Scholar]
  • 157.Bowman EP, Chackerian AA, Cua DJ. Rationale and safety of anti-interleukin-23 and anti-interleukin-17A therapy. Curr Opin Infect Dis. 2006;19:245–252. doi: 10.1097/01.qco.0000224818.42729.67. [DOI] [PubMed] [Google Scholar]
  • 158.Hueber W, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut. 2012;61:1693–1700. doi: 10.1136/gutjnl-2011-301668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Targan SR, et al. Mo2083 A Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety, Tolerability, and Efficacy of AMG 827 in Subjects With Moderate to Severe Crohn’s Disease. Gastroenterology. 143:e26. [Google Scholar]
  • 160.Kaser A. Not all monoclonals are created equal - lessons from failed drug trials in Crohn’s disease. Best Pract Res Clin Gastroenterol. 2014;28:437–449. doi: 10.1016/j.bpg.2014.04.005. [DOI] [PubMed] [Google Scholar]
  • 161.Colombel JF, Sendid B, Jouault T, Poulain D. Secukinumab failure in Crohn’s disease: the yeast connection? Gut. 2013;62:800–801. doi: 10.1136/gutjnl-2012-304154. [DOI] [PubMed] [Google Scholar]

RESOURCES