Open Access

Role of innate lymphoid cells in obesity and metabolic disease (Review)

  • Authors:
    • Jirakrit Saetang
    • Surasak Sangkhathat
  • View Affiliations

  • Published online on: November 13, 2017     https://doi.org/10.3892/mmr.2017.8038
  • Pages: 1403-1412
  • Copyright: © Saetang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The immune system has previously been demonstrated to be associated with the pathophysiological development of metabolic abnormalities. However, the mechanisms linking immunity to metabolic disease remain to be fully elucidated. It has previously been suggested that innate lymphoid cells (ILCs) may be involved in the progression of numerous types of metabolic diseases as these cells act as suppressors and promoters for obesity and associated conditions, and are particularly involved in adipose tissue inflammation, which is a major feature of metabolic imbalance. Group 2 ILCs (ILC2s) have been revealed as anti‑obese immune regulators by secreting anti‑inflammatory cytokines and promoting the polarization of M2 macrophages, whereas group 1 ILCs (ILC1s), including natural killer cells, may promote adipose tissue inflammation via production of interferon‑γ, which in turn polarizes macrophages toward the M1 type. The majority of studies to date have demonstrated the pathological association between ILCs and obesity in the context of adipose tissue inflammation, whereas the roles of ILCs in other organs which participate in obesity development have not been fully characterized. Therefore, identifying the roles of all types of ILCs as central components mediating obesity‑associated inflammation, is of primary concern, and may lead to the discovery of novel preventative and therapeutic interventions.

Introduction

Obesity, as defined by an excess of adipose tissue, has increased globally in most of the westernized world over recent decades. This condition is associated with several chronic complications, such as hypertension, coronary heart disease, hyperglycemia, hypertriglyceridemia, dyslipidemia and insulin resistance (1). Although over 30 gene loci combinations have been identified as being associated with the development of obesity and metabolic disease (2), these loci are responsible for only 2–3% of the incidence of these conditions. Other causes of obesity are known to include energy-dense diets high in saturated fat and sugar, and the sedentary lifestyles common in the modern world (35). In recent years, animal models have been developed to study such things as diet-induced obesity to study the underlying mechanisms of obesity and related diseases.

It is further generally accepted that obesity and metabolic disorders are associated with chronic, low-grade systemic inflammation (6). This kind of inflammation originates predominantly in adipose tissue, which leads to greater numbers of immune cells, resulting in the activation of other cells, such as adipocytes, which cause adipose tissue remodeling and further incitement of the inflammatory process (7). Interestingly, malnutrition, including excessive energy intake, increase fat accumulation, and lipotoxicity, can activate the expression of pro-inflammatory effector molecules in metabolic tissues and cells involved in innate immunity (8,9). Although many recent studies have focused on the role of adipose tissue macrophages (ATMs), which are known to be the main factor initiating systemic inflammatory promotion in obesity, the recently identified innate lymphoid cells (ILCs) have also been reported to play a role in the inflammatory process in obese adipose tissue (10).

In this article, we review the current understanding of the complex interplay between the types of ILCs that have been found to link inflammation to obesity. We also discuss the cellular and molecular basis of obesity-induced inflammation and the functional role of each type of ILC. Finally, we open a new discussion by noting that even if adipose tissue is accepted as a major cause of systemic inflammation, it is still unclear whether any metabolic-related organs can influence obesity-related pathologies.

Interplay between diets, inflammation and gut microbiota in obesity

In the last decade, various mechanisms linking immunity, metabolic abnormalities, and intestinal microbiota have been proposed (11). Although the cause of metabolic inflammation in obesity has not been fully clarified, some studies have shown that diet-induced dysbiosis may be the origin of this inflammation (9,12), and this scenario is related to increased intestinal permeability caused by changes of normal flora and their metabolites (13,14). The studies found that diet can be associated with structural variations in gut microbiota, especially the ‘Western’ diet, which has various effects on host gastrointestinal tract (GI) metabolism, microbiota and immune homeostasis (15,16).

Changes in microbiota can affect gut metabolic activity in various way, such as increasing the production of short-chain fatty acids (SCFAs), which leads to alterations of intestinal homeostasis. Decreased bacterial diversity and alteration of representative bacterial genes and metabolic pathways can be found in obese individuals (17). In particular, high-fat diets or diets low in fiber have been associated with a higher abundance of Firmicutes (18), while studies comparing obese individuals and their lean twins have also shown a higher predominance of Firmicutes and lower abundance of Bacteroidetes (17,19) in the obese subjects. It must be noted, however, that other studies have not found similar differences (20,21).

Further investigations have been shown that the complex interplay between diet and the intestinal microbiota in the context of obesity can lead to the release of gut-derived inflammatory factors into the circulation, resulting in the development of obesity (22). Lipopolysaccharide (LPS), a potent inflammatory mediator of Gram-negative bacteria, has been recently shown to trigger inflammation in obese and metabolic syndrome individuals by signaling through the CD14/TLR4 pathway (23). Such LPS-induced systemic inflammation may result from intestinal permeability mediated by a high-fat diet since increases in the translocation of intestinal Gram-negative bacteria (which produce LPS) to the mesenteric lymph nodes (mLNs) and mesenteric fat can be found in high-fat diet-fed mice (24). One recent study found that antibiotic treatment or CD14 suppression appeared to reduce inflammatory cytokine expression and improve weight gain in high-fat diet mice, indicating a role of the microbiota in the inflammatory process (25). Therefore, it is possible that intestinal inflammation may lead to GI permeability, resulting in an increase in circulating LPS and bacterial DNA, which promote systemic inflammation and insulin resistance in both mice and humans (26).

This metabolic inflammation, which does not necessarily involve pathogens, is associated with inflammatory adipose tissue and higher immune cell accumulation in fatty tissue regions (Fig. 1) (6). ATMs appear to play a major function in the regulation of obesity-induced inflammation, and different types of macrophage can cause the different effects in adipose tissue. Currently, macrophages are divided into 2 groups, the M1 and M2 types. M1 macrophages characterized by the expression of F4/80+ CD11b+ CD11c+ iNOS+ (inducible nitric oxide synthase) and the production of pro-inflammatory cytokines [IL-1β, IL-6, IL-12, tumor necrosis factor (TNF)-α, MCP-1] is considered to be involved in adipose tissue inflammation, while the M2 type macrophages, which express F4/80+ CD11c CD301+ Arg1+ CD206+ and produce anti-inflammatory cytokines [IL-1 receptor antagonist, IL-4, IL-10, transforming growth factor (TGF)-β1], have been known to suppress inflammation in adipose tissue (27,28). Other studies have also suggested that high levels of inflammatory cytokines in adipose tissue during obesity are consistent with increasing macrophage numbers, which can be described as a metabolic activation instead of the classical activation related to infections (29,30). In addition to macrophages, lymphocytes are strongly associated with inflammatory processes in obesity. Although there are several types of lymphocytes that are related to obesity and metabolic syndrome, pro-inflammatory Th1, Th17 and CD8+ T cells predominate over anti-inflammatory regulatory T (Treg) cells and Th2 cells, which are found in higher proportions in lean adipose tissue (7,31). One study found that mice fed a high-fat diet displayed more Th1 polarization and IFN-γ production, which occurred several months after macrophage accumulation and insulin resistance (32), while the number of Treg cells was decreased in the adipose tissue of obese mice; insulin sensitivity was also improved when these cells were increased (Fig. 1) (31).

Innate lymphoid cells (ILCs)

ILCs are a recently discover type of innate immune cells, which have been identified as an important player in lymphoid organogenesis, tissue defense, epithelial tissue homeostasis and the amplification of immune responses (33,34). Although it has been agreed that ILCs participate in the defense against pathogens (35), some studies have suggested they could also be involved in some systemic conditions, such as chronic inflammation and autoimmune disorders (36,37). ILCs have been defined as lymphoid-derived, immune lineage negative (Lin), Th cytokine expressing cells (33). Currently, three distinct groups of ILCs which exhibit a functional diversity mirroring three types of effector CD4+ T-cells have been identified: Group 1 ILCs, equivalent to Th1 T cells, group 2 ILCs to Th2 cells, and group 3 ILCs to Th17 and Th22 cells (Fig. 2) (35).

Group 1 ILCs

Group 1 ILCs are characterized based on their ability to produce IFN-γ, and have been sub-grouped into two main types, conventional NK (cNK) cells and ILC1 cells (38). cNK cells have cytotoxic ability and can be found in numerous organs as they recirculate between the blood and tissues while ILC1s have only limited cytotoxicity, but have the ability to produce several types of inflammatory cytokines mirroring Th1 cells (39,40). Both types of group 1 ILCs can produce TNF and IFN-γ as a uniquely inflammatory profile when these cells are stimulated with IL-12, IL-15, or IL-18, and rely on T-bet transcription factor (T-bet) as a key transcription factor (41,42). Generally, mice resting mature cNK cells have been identified as Lin CD3ε+ NK1.1+ NKp46+ CD49b(DX5)+ Eomes+ whereas Lin CD3ε CD56+ NKp46+ NKp44 has been used in humans (Fig. 2) (43). Two main groups of cNK cells have been identified, including the majority of blood cNK cells, which are CD56low and highly cytotoxic, and CD56high low cytotoxicity cNK cells, which produce a high number of inflammatory cytokines (44).

Unconventional NK cells or, also known as ILC1s, are tissue-resident NK-like cells that are not derived from NK cell precursors and are normally found in non-lymphoid organs (45). A defining distinction between NK cells and ILC1s is the expression of the transcription factor, Eomes (39). NK cells are Eomes+ and require this transcription factor to develop while ILC1s are Eomes and do not need Eomes for their development (39). Another noticeable characteristic that differentiates ILC1s from NK cells is their tissue resident markers, CD49a and CD69 (39). These tissue resident ILC1s have been found in a variety of non-lymphoid tissues, including the small intestine mucosa, liver, salivary glands, and female reproductive tract (45). Interestingly, recirculation of these tissue-resident ILC1s does not seem to occur, and, in mice, the existence of these ILC1s is likely maintained predominantly through local self-renewal instead of replenishment from blood-derived ILC1s or their precursors (46,47).

Emerging evidence in recent years has demonstrated that ILC1s and NK cells in different organs show coincident but different phenotypes and functions. Intestinal and hepatic ILC1s share the CD49a+ CD49b phenotype, produce high IFN-γ levels and are Eomes and less cytotoxic than NK cells (48). The cytotoxicity of hepatic resident ILC1s is regularly mediated by TRAIL rather than perforin (48,49), which is found in NK cell-mediated cytotoxicity, although mice and human NK cells can express TRAIL (50). Moreover, various studies have found that several types of group 1 ILCs that differ from cNK cells are resident in the gastrointestinal mucosa, e.g., intraepithelial ILC1s (ieILC1s), which express surface markers typical of intraepithelial T cells, such as CD49a, CD69, CD160, and in humans, the integrin CD103 that makes them distinct from cNK cells although ieILC1s also display canonical NK cell receptors and need the transcription factors T-bet and Eomes in their development, similar to cNK cells (42). Another type is lamina propria-resident ILC1s (LP ILC1s), which express NKp46 and NK1.1, which are found in both cNK cells and ieILC1s (45). However, mouse LP ILC1s express high levels of the IL-7 receptor α chain (CD127) and are negative for Eomes but positive for T-bet, which is not common in either cNK or ieILC1 cells (Fig. 2) (45,48).

The major function of group 1 ILCs is the potent expression of IFN-γ, which plays a crucial role in promoting immunity against intracellular pathogens (51). cNK cells are known for their rapid response after exposure to a variety of viral and bacterial pathogens (52). It is now suggested that ILC1s are the major source of IFN-γ and TNF in orally Toxoplasma gondii-infected mice while cNK cells contribute to a lesser extent (48). Another study found that T-bet deficient mice were highly susceptible to T. gondii infection, and adoptive transfer of ILC1s to Rag2−/− Il2rg−/− mice boosted anti-bacterial immunity (48). Importantly, it seems likely that some results from the previous studies regarded all NK-like phenotypes with an ability to produce IFN-γ as cNK cells, which make the un-recognition of ILC1s as a separate lineage (45). Studies that specifically investigated the role of ILC1s, as compared to cNK cells, in host defenses to pathogens are only now being conducted; using specific markers for ILC1s, such as CD49a, CD127 and Eomes, may make it possible to more accurately study the specific roles of ILC1s compared to cNK cells (45).

Group 2 ILCs

ILC2s derive from common lymphoid progenitors like most lymphoid cells but lack the common lineage marker expression associated with T cells (CD3, CD4, and αβ/γδ TCR), B cells (CD19 and CD20), and other leukocytes including CD11c, CD14, CD16, CD56, and FcεR1 (53). Moreover, they are positive for CD90 (Thy1), CD25 (IL-2 receptor α), IL-25R (IL-17RB), IL-33 receptor (IL-33R; ST2), and CD127 (IL-7 receptor α) (Fig. 2) (54). Similar to Th2 cells, GATA3 acts as a key transcription factor for development and function of ILC2s (55), and transcription factors Id2 and retinoic acid-related orphan receptor α (ROR)α have also be recently include as important regulators in their development (54,56,57).

ILC2s can respond to IL-25, IL-33 (57,58) and thymic stromal lymphopoietin (TSLP) (59), and produce type 2 cytokines, predominantly IL-5 and IL-13 (60). Several studies have shown that ILC2s can produce high levels of IL-5 and IL-13, which contribute to the immune response against helminth infection in the GI, lungs and skin (61,62). ILC2s also produce IL-9, which one study found supported the accumulation of mast cells and mucus production (63). However, IL-9 expression by ILC2s was transient and dependent on IL-2 and intact adaptive immunity, suggesting that IL-9 could amplify ILC2 function (64). ILC2s can also activate CD4+ T cells for the efficient induction of Th2 cell development by presenting an antigen to non-activated CD4+ T cells (65). Another study also showed that OX40/OX40 L interactions and the production of IL-4 by ILC2s contributed to CD4+ T-cell responses in vitro supporting the role of ILC2s-CD4+ T cell relation (66). Interestingly, lipid mediators, such as CysLTs and PGD2, and the TNF-like ligand 1A (TL1A), which have all been associated with Th2-driven diseases, have also been found to be activators of ILC2s (67,68). Furthermore, basophilic- and Th2-derived IL-4 also promote ILC2 activation (69). Cell-cell interactions through molecular signaling of ICOS (binds ICOS-L) and KLRG1 (binds cadherins) in ILC2s also promote ILC2 activation and survival (70,71). A recent report also demonstrated that ILC2 activation can be influenced by Treg cells, and activated ILC2 also conversely promote Treg cell maintenance (72).

It has been proposed that ILC2-derived Th2 cytokines contribute to several types of Th2-related diseases, such as chronic rhinosinusitis, asthma, atopic dermatitis, and gastrointestinal allergic disease, which have all been considered as resident sites of ILC2s (Fig. 2) (64). Clinical experiments with asthmatics have shown that IL-4, IL-5, and IL-13 inhibition give beneficial effects to asthma patients, reflecting the importance of Th2 cytokines, which are partly derived from ILC2s in asthma (73). Notably, ILC2s seem to play an important role in lung inflammation in responding to protease-containing allergens. One study showed that papain, an allergen used in the experiment, promoted allergic lung inflammation even in RAG-deficient mice, suggesting a role of ILC2 in lung inflammation (74). Moreover, airway hyper-reactivity mediated by ILC2s is found not only in non-infectious inflammation but also after influenza viral infections (75).

Another role of ILC2s in the metabolic pathway has also been found, based on the knowledge that this kind of ILC can regulate beige adipose tissue development and may promote the lean phenotype (76). Moreover, ILC2s can be detected in visceral adipose tissue (VAT) where they were thought to be responsible for eosinophil accumulation (77). Another study found that ILC2-depleted Rag1−/− mice became obese and showed impaired glucose tolerance, but these problems improved when ILC2 cells were transferred into these obese mice (78). Moreover, nutritional status also likely influences ILC2 biology, as vitamins A and D are known to skew the ILC3/ILC2 balance in the intestines (79).

Group 3 ILCs

There are currently two recognized sub-populations of ILC3s, the CCR6+ lymphoid-tissue inducer (LTi) ILC3s and the CCR6 ILC3s, of which the latter can be divided into two groups based on the expression of NKp46 in mice (80) and NKp44 in humans (81) (Fig. 2). All groups of ILC3s need the nuclear hormone retinoic acid receptor-related orphan receptor γt (RORγt) as a key regulator for their development and function. These ILCs can be activated by IL-23 or IL-1β stimulation to produce IL-17 or IL-22 mirroring Th17 and Th22 cells (82).

LTi cells appearing during embryonic development were initially regarded as strictly required for prenatal lymph node development and Peyer's patches (PPs), and also thought to be important in the development of the adaptive immune system (82,83). However, CCR6-expressing LTi-ILC3s (CCR6+ ILC3s), which share several characteristics with embryonic LTi cells, such as co-expression of CD4 and the production of IL-17 (Fig. 2) (84), can be found in mLNs and the colon lamina propria (cLPL) of healthy adult mice (85).

CCR6 ILC3s, which produce only IL-22 and account for a small proportion of ILC3s perinatally, increased significantly within 4 weeks after birth (84,86), and PLZF+ common helper-like ILC progenitor (CHILP) cells seem likely to be a specific CCR6 ILC3 progenitor, which make CCR6 ILC3s differ from other ILC3s (84). Moreover, some studies have demonstrated that CCR6 ILC3s use distinct transcriptional regulation pathways for their development (84,87), and there are some differences within CCR6 ILC3s in terms of their activation since CCR6NKp46 ILC3s do not require T-bet for cell differentiation and maintenance, but CCR6NKp46+ ILC3s do require T-bet (48).

Although the proportion of ILC3s accounts for less than 5% of lamina propria lymphocytes, it has been shown that this cell type is one of the major sources of IL-22, which plays a crucial role in mucosal immune defense. CCR6+ ILC3s regulate immunity against infection and disease through the secretion of IL-17 and IL-22, in addition to their key role in organogenesis (82,88). For example, one study found that intestinal tissue homeostasis could be supported by CCR6+ ILC3-derived IL-22 in a graft-vs.-host disease model (89). Another study showed that this CCR6+ ILC3-derived IL-22 also promotes the induction of intestinal fucosylation of the intestinal epithelium with the cooperation of lymphotoxin, which is also derived from CCR6+ ILC3s (90). CCR6NKp46+ ILC3s in the small intestine also contribute to mucosal immunity against Citrobacter rodentium (C. rodentium) through production of IL-22 in Rag2−/− Il2rg−/− mice (80). In addition, ILC3s have been thought to play a role in the intestinal defenses in Salmonella typhimurium (91), Candida albicans (92) and Streptococcus pneumoniae (93) models. It has also been proposed that IL-22-producing CCR6NKp46+ ILC3s have an impact on the resistance to bacterial invasion in the colitis model (94,95), and modulate eosinophil infiltration and lymphocyte invasion in allergic airway hyperreactivity (AHR) in the lung (96). These all suggest a role of ILC3s in homeostasis in multiple tissues following inflammation or damage.

Relationship between ILCs and obesity

Although it is accepted that genetic and environmental factors were originally thought to be the major influences on the development of obesity, many researches have now shown that immunological factors can also contribute to the pathogenesis of obesity. Nowadays, several types of immune cells have been recognized as critical regulators of metabolic homeostasis (97,98), and this crosstalk likely involves immune cells and low-grade inflammation, particularly in many organs besides the adipose tissue, including the pancreas, liver and intestines, which all showed as an emerging characteristics and made a potentially regulatory force behind the development of obesity (97,99).

ILCs are now recognized as a new regulator involved in adipose tissue and in metabolic homeostasis (10,100). Most studies of ILCs and their role in metabolism have focused on ILC2s, which have been reported to play a role in maintaining metabolic homeostasis, since ILC2s and eosinophils are predominantly resident in lean adipose tissue, and are considered as ‘upstream’ regulators of M2 macrophages in adipose tissue (77). Various studies have shown that, with the cooperation of eosinophils and M2 macrophages, ILC2s regulate obesity, beige conversion of white adipose tissue and beige fat biogenesis (Fig. 3) (76,77,101). The production of IL-5 and IL-13 from ILC2s in VAT has been shown to be essential for eosinophil and M2 macrophage differentiation and activation, both of which act as important regulators of obesity (77). In addition, ILC2 deficiency in Rag1−/− mice resulted in significantly reduced numbers of eosinophils and M2 macrophages, suggesting that ILC2s alone can promote the development of eosinophils and M2 macrophages in adipose tissue leading to obesity regulation (77,78). IL-33 is another important inducer of ILC2s and can influence the development of obesity. Recent studies have shown the effect of ILC2s on the biogenesis of VAT under IL-33 stimulation, which might regulate obesity (Fig. 3) (76,102). In addition, the numbers of ILC2s were decreased in obese murine epididyma, and this scenario is also found in human abdominal subcutaneous white adipose tissue (76). Notably, IL-33 knock-out mice were shown to have weight gain and reduced frequency and absolute numbers of ILC2s, even when the mice were fed a normal diet (76). However, this situation could be reversed by administration of IL-33, which led to increased numbers of ILC2s, consequently promoting the recovery of M2 macrophage numbers (76).

Roles of cNK cells and ILC1s in obesity have also been demonstrated. One study found that large quantities of IFN-γ, which may trigger M1 macrophages in VAT, may be derived from cNK cells in HFD-induced obesity (103). Moreover, systemic depletion of NK1.1+ or NKp46+ cells decreased diet-induced insulin resistance by restricting the polarization of M1 macrophages, but did not decrease obesity, suggesting that cNK cells affect inflammation-related insulin resistance rather than metabolism directly (103,104). In addition, adoptive transfer of splenic NK cells into the VAT of IFN-γ knock-out mice could restore insulin resistance following HFD induction (103), implying that this cell type may be a major regulator promoting insulin resistance. Tissue resident group 1 ILCs have also been recently reported to contribute to obesity-associated insulin resistance in the absence of the influence of T and/or NKT cells (105). The same study also reported that IL-12, which were undefined for upstream signals and cellular sources, could activate adipose ILC1s, leading to the production of IFN-γ, and the polarization of M1 macrophages in adipose tissue at the early stage of HFD consumption (Fig. 3) (105). Mirroring the Th1-Th2 balance, IFN-γ, which is partly derived from ILC1s, can counteract the IL-33 function and interfere with the activation of ILC2s in infected tissues, as well as in healthy adipose tissues (72). Therefore, ILC1s may indirectly affect ILC2-mediated regulation of obesity (Fig. 3).

Although there are currently no specific experiments describing either the effects of ILC3s in adipose tissues or its role or roles in HFD-induced obesity, it has been hypothesized that IL-17 and/or IL-22 secreted from ILC3s might affect obesity or metabolic homeostasis (10). ILC3-derived IL-17 has been shown to be associated with obesity-related diseases such as AHR (106). This same study also demonstrated that AHR lesions were abundant with CCR6+NKp46 ILC3s, which have the ability to secrete excessive IL-17 (106). Moreover, the same study found that Rag1−/− IL-17A−/− mice did not develop AHR under HFD conditions, but transferring CCR6+NKp46 ILC3s into the Rag−/−IL-2Rγ−/− mice resulted in AHR under HFD feeding, suggesting a role of IL-17 and ILC3s on AHR pathogenesis (106). However, it has also been reported that IL-22, for which ILC3s were the major source, has been found to alleviate metabolic abnormalities, including insulin resistance and hyperglycemia via changes in liver metabolism (107). This same study showed that IL-22R−/− mice were highly susceptible to HFD-induced obesity and insulin resistance, but these events improved when IL-22 was administrated to the obese mice. Another study also demonstrated a protective role of IL-22 against diabetes, partly through modulating oxidative stress and inflammation pathways related to islet β cells, promoting the secretion of insulin and fully restored glucose insulin sensitivity in obese mice (108).

Determining whether intestinal ILCs influence the development of obesity

As mentioned previously, energy balance and gut homeostasis of the host may be influenced by diet and gut microbiota composition, and, in metabolically abnormal conditions, increased intestinal permeability and inflammation may play a role in the adipose tissue inflammatory response (9,24). The gut is resided by extensive immune system, and recent studies have investigated changes in intestinal inflammatory and immune cell populations with regard to their roles in obesity and insulin resistance (109,110).

Early reports have shown increasing levels of gut inflammatory cytokines, such as TNF-α, IL-1β and IL-12, which are mostly produced by innate immune cells during HFD feeding, and these increases are related to weight gain, adiposity, plasma insulin and glucose levels (99,111). Interestingly, these expressions were detected in bowel immune cells, especially in PPs and lymphoid aggregates, in which ILCs are resident (112). However, this intestinal inflammatory response was not found in HFD-fed germ-free mice, suggesting the microbiota are a driving force behind intestinal inflammation (111). Recent evidence has also demonstrated high expression levels of several pro-inflammatory cytokines, including IFN-γ and IL-1β, in the duodenum of insulin-resistant obese patients (113), and IL-23, TNF-α, TGF-β, CCL5, and IFN-γ in the combined lamina propria and epithelial fraction of obese subjects (114). Specifically, there are many studies which have investigated changes of particular types of intestinal immune cells in obese subjects. One study found that the intestinal frequency of γδT cells was changed in mice with HFD feeding (109). This type of innate immune cell is predominantly found in the intraepithelial lymphocyte fraction, and the same study found that the numbers of IL-17-producing γδT cells in the small intestine increased after 3 weeks of HFD consumption, and after 12 weeks in both the colon and small bowel (109). Another study found that intestinal eosinophils were also decreased in both number and proportion after 1 week of HFD feeding (115). This reduction was correlated with intestinal permeability of HFD mice predominantly in the ileum. Another study also demonstrated that total macrophage density and the numbers of mature DCs and NK cells were increased in obese diabetic and obese non-diabetic humans (114).

HFD has been shown not only to be active in innate immune cells, but it can also alter the proportions of adaptive immune cells in the intestinal community. One study found that the percentages of Treg cells, which play an important role in inflammatory suppression, were decreased after 3 weeks of HFD consumption, while the quantity of CD4+ and CD8+ cells did not show any significant differences in this period, but were higher after 12 weeks of HFD feeding (109). Obese human subjects also showed increased T-bet expression and CD8+ T cells and reduced Foxp3 (Treg) cells in both the small bowel and colon compared to lean subjects (109,114). Reduced numbers and frequency of Th17 cells were also found in HFD-fed mice, apparently related to alterations of the commensal bacteria populations, such as segmented filamentous bacteria (SFB) and Porphyromonas gingivalis (110). Another study found that decreased numbers of Th17 and SFB were correlated with an abnormal gut barrier and increased adiposity, linking the role of microbiota and adaptive immunity to obesity (116).

Although the roles of ILCs on metabolic homeostasis have been intensively studied, to date such studies have focused only on adipose tissue ILCs, and these findings may not represent the overall contributions of this cell type on obesity, since other organs such as the intestine also displayed low-grade chronic inflammatory changes in obese persons. There are few studies to date which have examined the role of gut ILCs on intestinal inflammation and homeostasis after HFD-feeding, and until now only ILC3s, which are very abundant within the intestine, have been reported to be associated with mucosal homeostasis during obesity (107). This information is consistent with the reduction of CCR6+NKp46+ ILC3s in the lamina propria of HFD-fed mice, and this seems to be related to lower mucosal barrier integrity and increased serum LPS (109). However, no current reports have demonstrated a role for HFD in all types of intestinal ILCs, which directly contact with diets and gut microbiota, and may contribute to colon homeostasis in obesity.

Conclusion

Like macrophages, ILCs were first described as a player in the innate immune system against several types of pathogens, but new studies on these immune cells have proposed that ILCs may also be a player in metabolic homeostasis. ILC2s appear to be regulators of anti-inflammatory conditions in the lean state while ILC1s seem to be involved in promoting the development of obesity and other metabolic diseases by secreting inflammatory factors in the obese state. Although these roles and the characterizations of ILCs have been well documented in many studies, most investigations to date have focused on the function of this cell in adipose tissue, but since the primary functions of this type of cell are carried out in barrier tissues such as the liver, intestine and other abdominal viscera, which all affect the progression of obesity and metabolic diseases, new research should examine the influence of these cells in other organs on the development of obesity and metabolic abnormalities.

References

1 

Boulangé CL, Neves AL, Chilloux J, Nicholson JK and Dumas ME: Impact of the gut microbiota on inflammation, obesity and metabolic disease. Genome Med. 8:422016. View Article : Google Scholar : PubMed/NCBI

2 

Lu Y and Loos RJ: Obesity genomics: Assessing the transferability of susceptibility loci across diverse populations. Genome Med. 5:552013. View Article : Google Scholar : PubMed/NCBI

3 

Westerterp KR and Plasqui G: Physically active lifestyle does not decrease the risk of fattening. PLoS One. 4:e47452009. View Article : Google Scholar : PubMed/NCBI

4 

DiNicolantonio JJ, O'Keefe JH and Lucan SC: Added fructose: A principal driver of type 2 diabetes mellitus and its consequences. Mayo Clin Proc. 90:372–381. 2015. View Article : Google Scholar : PubMed/NCBI

5 

DiNicolantonio JJ, Lucan SC and O'Keefe JH: The evidence for saturated fat and for sugar related to coronary heart disease. Prog Cardiovasc Dis. 58:464–472. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Gregor MF and Hotamisligil GS: Inflammatory mechanisms in obesity. Annu Rev Immunol. 29:415–445. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Sell H, Habich C and Eckel J: Adaptive immunity in obesity and insulin resistance. Nat Rev Endocrinol. 8:709–716. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Purkayastha S and Cai D: Neuroinflammatory basis of metabolic syndrome. Mol Metab. 2:356–363. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, et al: Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 56:1761–1772. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Yang D, Yang W, Tian Z, van Velkinburgh JC, Song J, Wu Y and Ni B: Innate lymphoid cells as novel regulators of obesity and its-associated metabolic dysfunction. Obes Rev. 17:485–498. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Hotamisligil GS: Inflammation and metabolic disorders. Nature. 444:860–867. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H and Flier JS: TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 116:3015–3025. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD and Gordon JI: Obesity alters gut microbial ecology. Proc Natl Acad Sci USA. 102:11070–11075. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, Qiu Z, Maher L, Redinbo MR, Phillips RS, et al: Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity. 41:296–310. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Zhang C, Zhang M, Wang S, Han R, Cao Y, Hua W, Mao Y, Zhang X, Pang X, Wei C, et al: Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J. 4:232–241. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Sekirov I, Russell SL, Antunes LC and Finlay BB: Gut microbiota in health and disease. Physiol Rev. 90:859–904. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, et al: A core gut microbiome in obese and lean twins. Nature. 457:480–484. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, Blanchard C, Junt T, Nicod LP, Harris NL and Marsland BJ: Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 20:159–166. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Ley RE, Turnbaugh PJ, Klein S and Gordon JI: Microbial ecology: Human gut microbes associated with obesity. Nature. 444:1022–1023. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C and Hardt PD: Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring). 18:190–195. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Duncan SH, Lobley GE, Holtrop G, Ince J, Johnstone AM, Louis P and Flint HJ: Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes (Lond). 32:1720–1724. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Montiel-Castro AJ, González-Cervantes RM, Bravo-Ruiseco G and Pacheco-López G: The microbiota-gut-brain axis: Neurobehavioral correlates, health and sociality. Front Integr Neurosci. 7:702013. View Article : Google Scholar : PubMed/NCBI

23 

Harris K, Kassis A, Major G and Chou CJ: Is the gut microbiota a new factor contributing to obesity and its metabolic disorders? J Obes. 2012:8791512012. View Article : Google Scholar : PubMed/NCBI

24 

Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermúdez-Humarán LG, Smirnova N, Bergé M, Sulpice T, Lahtinen S, et al: Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: Molecular mechanisms and probiotic treatment. EMBO Mol Med. 3:559–572. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Cox LM and Blaser MJ: Pathways in microbe-induced obesity. Cell Metab. 17:883–894. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Burcelin R, Garidou L and Pomié C: Immuno-microbiota cross and talk: The new paradigm of metabolic diseases. Semin Immunol. 24:67–74. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Mosser DM and Edwards JP: Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 8:958–969. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Eagle Red A and Chawla A: In obesity and weight loss, all roads lead to the mighty macrophage. J Clin Invest. 120:3437–3440. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Kratz M, Coats BR, Hisert KB, Hagman D, Mutskov V, Peris E, Schoenfelt KQ, Kuzma JN, Larson I, Billing PS, et al: Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metab. 20:614–625. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Xu X, Grijalva A, Skowronski A, van Eijk M, Serlie MJ and Ferrante AW Jr: Obesity activates a program of lysosomal-dependent lipid metabolism in adipose tissue macrophages independently of classic activation. Cell Metab. 18:816–830. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, Lee J, Goldfine AB, Benoist C, Shoelson S and Mathis D: Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 15:930–939. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Strissel KJ, DeFuria J, Shaul ME, Bennett G, Greenberg AS and Obin MS: T-cell recruitment and Th1 polarization in adipose tissue during diet-induced obesity in C57BL/6 mice. Obesity (Silver Spring). 18:1918–1925. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie AN, Mebius RE, et al: Innate lymphoid cells – a proposal for uniform nomenclature. Nat Rev Immunol. 13:145–149. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Spits H and Cupedo T: Innate lymphoid cells: Emerging insights in development, lineage relationships and function. Annu Rev Immunol. 30:647–675. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Artis D and Spits H: The biology of innate lymphoid cells. Nature. 517:293–301. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Villanova F, Flutter B, Tosi I, Grys K, Sreeneebus H, Perera GK, Chapman A, Smith CH, Di Meglio P and Nestle FO: Characterization of innate lymphoid cells in human skin and blood demonstrates increase of NKp44+ ILC3 in psoriasis. J Invest Dermatol. 134:984–991. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Fuchs A and Colonna M: Innate lymphoid cells in homeostasis, infection, chronic inflammation and tumors of the gastrointestinal tract. Curr Opin Gastroenterol. 29:581–587. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Cortez VS, Robinette ML and Colonna M: Innate lymphoid cells: New insights into function and development. Curr Opin Immunol. 32:71–77. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Cortez VS and Colonna M: Diversity and function of group 1 innate lymphoid cells. Immunol Lett. 179:19–24. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Sun JC and Lanier LL: NK cell development, homeostasis and function: Parallels with CD8+ T cells. Nat Rev Immunol. 11:645–657. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Robinette ML, Fuchs A, Cortez VS, Lee JS, Wang Y, Durum SK, Gilfillan S and Colonna M: Immunological Genome Consortium: Transcriptional programs define molecular characteristics of innate lymphoid cell classes and subsets. Nat Immunol. 16:306–317. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Fuchs A, Vermi W, Lee JS, Lonardi S, Gilfillan S, Newberry RD, Cella M and Colonna M: Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-γ-producing cells. Immunity. 38:769–781. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM and Ugolini S: Innate or adaptive immunity? The example of natural killer cells. Science. 331:44–49. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Cichicki F, Schlums H, Theorell J, Tesi B, Miller JS, Ljunggren HG and Bryceson YT: Diversification and functional specialization of human NK cell subsets. Curr Top Microbiol Immunol. 395:63–94. 2016.PubMed/NCBI

45 

Fuchs A: ILC1s in tissue inflammation and infection. Front Immunol. 7:1042016. View Article : Google Scholar : PubMed/NCBI

46 

Gasteiger G, Fan X, Dikiy S, Lee SY and Rudensky AY: Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Science. 350:981–985. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Peng H, Jiang X, Chen Y, Sojka DK, Wei H, Gao X, Sun R, Yokoyama WM and Tian Z: Liver-resident NK cells confer adaptive immunity in skin-contact inflammation. J Clin Invest. 123:1444–1456. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Klose CSN, Flach M, Möhle L, Rogell L, Hoyler T, Ebert K, Fabiunke C, Pfeifer D, Sexl V, Fonseca-Pereira D, et al: Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell. 157:340–356. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Daussy C, Faure F, Mayol K, Viel S, Gasteiger G, Charrier E, Bienvenu J, Henry T, Debien E, Hasan UA, et al: T-bet and Eomes instruct the development of two distinct natural killer cell lineages in the liver and in the bone marrow. J Exp Med. 211:563–577. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Spits H, Bernink JH and Lanier L: NK cells and type 1 innate lymphoid cells: Partners in host defense. Nat Immunol. 17:758–764. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Sonnenberg GF and Artis D: Innate lymphoid cells in the initiation, regulation and resolution of inflammation. Nat Med. 21:698–708. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Vivier E, Tomasello E, Baratin M, Walzer T and Ugolini S: Functions of natural killer cells. Nat Immunol. 9:503–510. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Karta MR, Broide DH and Doherty TA: Insights into group 2 innate lymphoid cells in human airway disease. Curr Allergy Asthma Rep. 16:82016. View Article : Google Scholar : PubMed/NCBI

54 

Walker JA, Barlow JL and McKenzie AN: Innate lymphoid cells – how did we miss them? Nat Rev Immunol. 13:75–87. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Hoyler T, Klose CS, Souabni A, Turqueti-Neves A, Pfeifer D, Rawlins EL, Voehringer D, Busslinger M and Diefenbach A: The transcription factor GATA-3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity. 37:634–648. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Wong SH, Walker JA, Jolin HE, Drynan LF, Hams E, Camelo A, Barlow JL, Neill DR, Panova V, Koch U, et al: Transcription factor RORα is critical for nuocyte development. Nat Immunol. 13:229–236. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, Furusawa J, Ohtani M, Fujii H and Koyasu S: Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. Nature. 463:540–544. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Saenz SA, Siracusa MC, Perrigoue JG, Spencer SP, Urban JF Jr, Tocker JE, Budelsky AL, Kleinschek MA, Kastelein RA, Kambayashi T, et al: IL25 elicits a multipotent progenitor cell population that promotes T(H)2 cytokine responses. Nature. 464:1362–1366. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Gentek R, Munneke JM, Helbig C, Blom B, Hazenberg MD, Spits H and Amsen D: Modulation of signal strength switches notch from an inducer of T cells to an inducer of ILC2. Front Immunol. 4:3342013. View Article : Google Scholar : PubMed/NCBI

60 

Price AE, Liang HE, Sullivan BM, Reinhardt RL, Eisley CJ, Erle DJ and Locksley RM: Systemically dispersed innate IL-13-expressing cells in type 2 immunity. Proc Natl Acad Sci USA. 107:11489–11494. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Tait Wojno ED and Artis D: Innate lymphoid cells: Balancing immunity, inflammation and tissue repair in the intestine. Cell Host Microbe. 12:445–457. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Neill DR, Wong SH, Bellosi A, Flynn RJ, Daly M, Langford TK, Bucks C, Kane CM, Fallon PG, Pannell R, et al: Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature. 464:1367–1370. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Wilhelm C, Turner JE, Van Snick J and Stockinger B: The many lives of IL-9: A question of survival? Nat Immunol. 13:637–641. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Doherty TA and Broide DH: Group 2 innate lymphoid cells: New players in human allergic diseases. J Investig Allergol Clin Immunol. 25:1–11; quiz 2p following 11. 2015.PubMed/NCBI

65 

Oliphant CJ, Hwang YY, Walker JA, Salimi M, Wong SH, Brewer JM, Englezakis A, Barlow JL, Hams E, Scanlon ST, 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. 41:283–295. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Drake LY, Iijima K and Kita H: Group 2 innate lymphoid cells and CD4+ T cells cooperate to mediate type 2 immune response in mice. Allergy. 69:1300–1307. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Fajt ML, Gelhaus SL, Freeman B, Uvalle CE, Trudeau JB, Holguin F and Wenzel SE: Prostaglandin D2 pathway upregulation: Relation to asthma severity, control and TH2 inflammation. J Allergy Clin Immunol. 131:1504–1512. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Yu X, Pappu R, Ramirez-Carrozzi V, Ota N, Caplazi P, Zhang J, Yan D, Xu M, Lee WP and Grogan JL: TNF superfamily member TL1A elicits type 2 innate lymphoid cells at mucosal barriers. Mucosal Immunol. 7:730–740. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Motomura Y, Morita H, Moro K, Nakae S, Artis D, Endo TA, Kuroki Y, Ohara O, Koyasu S and Kubo M: Basophil-derived interleukin-4 controls the function of natural helper cells, a member of ILC2s, in lung inflammation. Immunity. 40:758–771. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Maazi H, Patel N, Sankaranarayanan I, Suzuki Y, Rigas D, Soroosh P, Freeman GJ, Sharpe AH and Akbari O: ICOS: ICOS-ligand interaction is required for type 2 innate lymphoid cell function, homeostasis and induction of airway hyperreactivity. Immunity. 42:538–551. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Salimi M, Barlow JL, Saunders SP, Xue L, Gutowska-Owsiak D, Wang X, Huang LC, Johnson D, Scanlon ST, McKenzie AN, et al: A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J Exp Med. 210:2939–2950. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Molofsky AB, Van Gool F, Liang HE, Van Dyken SJ, Nussbaum JC, Lee J, Bluestone JA and Locksley RM: Interleukin-33 and interferon-γ counter-regulate group 2 innate lymphoid cell activation during immune perturbation. Immunity. 43:161–174. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Walford HH and Doherty TA: Diagnosis and management of eosinophilic asthma: A US perspective. J Asthma Allergy. 7:53–65. 2014.PubMed/NCBI

74 

Oboki K, Ohno T, Kajiwara N, Arae K, Morita H, Ishii A, Nambu A, Abe T, Kiyonari H, Matsumoto K, et al: IL-33 is a crucial amplifier of innate rather than acquired immunity. Proc Natl Acad Sci USA. 107:18581–18586. 2010. View Article : Google Scholar : PubMed/NCBI

75 

Chang YJ, Kim HY, Albacker LA, Baumgarth N, McKenzie AN, Smith DE, Dekruyff RH and Umetsu DT: Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat Immunol. 12:631–638. 2011. View Article : Google Scholar : PubMed/NCBI

76 

Brestoff JR, Kim BS, Saenz SA, Stine RR, Monticelli LA, Sonnenberg GF, Thome JJ, Farber DL, Lutfy K, Seale P and Artis D: Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature. 519:242–246. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Molofsky AB, Nussbaum JC, Liang HE, Van Dyken SJ, Cheng LE, Mohapatra A, Chawla A and Locksley RM: Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages. J Exp Med. 210:535–549. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Hams E, Locksley RM, McKenzie AN and Fallon PG: Cutting edge: IL-25 elicits innate lymphoid type 2 and type II NKT cells that regulate obesity in mice. J Immunol. 191:5349–5353. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Spencer SP, Wilhelm C, Yang Q, Hall JA, Bouladoux N, Boyd A, Nutman TB, Urban JF Jr, Wang J, Ramalingam TR, et al: Adaptation of innate lymphoid cells to a micronutrient deficiency promotes type 2 barrier immunity. Science. 343:432–437. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Satoh-Takayama N, Vosshenrich CA, Lesjean-Pottier S, Sawa S, Lochner M, Rattis F, Mention JJ, Thiam K, Cerf-Bensussan N, Mandelboim O, et al: Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity. 29:958–970. 2008. View Article : Google Scholar : PubMed/NCBI

81 

Cella M, Fuchs A, Vermi W, Facchetti F, Otero K, Lennerz JK, Doherty JM, Mills JC and Colonna M: A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature. 457:722–725. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Satoh-Takayama N: Heterogeneity and diversity of group 3 innate lymphoid cells: New cells on the block. Int Immunol. 28:29–34. 2016.PubMed/NCBI

83 

van de Pavert SA and Vivier E: Differentiation and function of group 3 innate lymphoid cells, from embryo to adult. Int Immunol. 28:35–42. 2016.PubMed/NCBI

84 

Klose CS, Kiss EA, Schwierzeck V, Ebert K, Hoyler T, d'Hargues Y, Göppert N, Croxford AL, Waisman A, Tanriver Y and Diefenbach A: A T-bet gradient controls the fate and function of CCR6-RORγt+ innate lymphoid cells. Nature. 494:261–265. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Hepworth MR, Fung TC, Masur SH, Kelsen JR, McConnell FM, Dubrot J, Withers DR, Hugues S, Farrar MA, Reith W, et al: Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4+ T cells. Science. 348:1031–1035. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Sawa S, Cherrier M, Lochner M, Satoh-Takayama N, Fehling HJ, Langa F, Di Santo JP and Eberl G: Lineage relationship analysis of RORγt+ innate lymphoid cells. Science. 330:665–669. 2010. View Article : Google Scholar : PubMed/NCBI

87 

Rankin LC, Groom JR, Chopin M, Herold MJ, Walker JA, Mielke LA, McKenzie AN, Carotta S, Nutt SL and Belz GT: The transcription factor T-bet is essential for the development of NKp46+ innate lymphocytes via the Notch pathway. Nat Immunol. 14:389–395. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, Littman DR and O'Shea JJ: Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med. 206:35–41. 2009. View Article : Google Scholar : PubMed/NCBI

89 

Hanash AM, Dudakov JA, Hua G, O'Connor MH, Young LF, Singer NV, West ML, Jenq RR, Holland AM, Kappel LW, et al: Interleukin-22 protects intestinal stem cells from immune-mediated tissue damage and regulates sensitivity to graft versus host disease. Immunity. 37:339–350. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Pickard JM, Maurice CF, Kinnebrew MA, Abt MC, Schenten D, Golovkina TV, Bogatyrev SR, Ismagilov RF, Pamer EG, Turnbaugh PJ and Chervonsky AV: Rapid fucosylation of intestinal epithelium sustains host-commensal symbiosis in sickness. Nature. 514:638–641. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Goto Y, Obata T, Kunisawa J, Sato S, Ivanov II, Lamichhane A, Takeyama N, Kamioka M, Sakamoto M, Matsuki T, et al: Innate lymphoid cells regulate intestinal epithelial cell glycosylation. Science. 345:12540092014. View Article : Google Scholar : PubMed/NCBI

92 

Gladiator A, Wangler N, Trautwein-Weidner K and LeibundGut-Landmann S: Cutting edge: IL-17-secreting innate lymphoid cells are essential for host defense against fungal infection. J Immunol. 190:521–525. 2013. View Article : Google Scholar : PubMed/NCBI

93 

Van Maele L, Carnoy C, Cayet D, Ivanov S, Porte R, Deruy E, Chabalgoity JA, Renauld JC, Eberl G, Benecke AG, et al: Activation of type 3 innate lymphoid cells and interleukin 22 secretion in the lungs during Streptococcus pneumoniae infection. J Infect Dis. 210:493–503. 2014. View Article : Google Scholar : PubMed/NCBI

94 

Kirchberger S, Royston DJ, Boulard O, Thornton E, Franchini F, Szabady RL, Harrison O and Powrie F: Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J Exp Med. 210:917–931. 2013. View Article : Google Scholar : PubMed/NCBI

95 

Sawa S, Lochner M, Satoh-Takayama N, Dulauroy S, Bérard M, Kleinschek M, Cua D, Di Santo JP and Eberl G: RORγt+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat Immunol. 12:320–326. 2011. View Article : Google Scholar : PubMed/NCBI

96 

Taube C, Tertilt C, Gyülveszi G, Dehzad N, Kreymborg K, Schneeweiss K, Michel E, Reuter S, Renauld JC, Arnold-Schild D, et al: IL-22 is produced by innate lymphoid cells and limits inflammation in allergic airway disease. PLoS One. 6:e217992011. View Article : Google Scholar : PubMed/NCBI

97 

Osborn O and Olefsky JM: The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med. 18:363–374. 2012. View Article : Google Scholar : PubMed/NCBI

98 

Jin C, Henao-Mejia J and Flavell RA: Innate immune receptors: Key regulators of metabolic disease progression. Cell Metab. 17:873–882. 2013. View Article : Google Scholar : PubMed/NCBI

99 

Winer DA, Luck H, Tsai S and Winer S: The intestinal immune system in obesity and insulin resistance. Cell Metab. 23:413–426. 2016. View Article : Google Scholar : PubMed/NCBI

100 

Bostick JW and Zhou L: Innate lymphoid cells in intestinal immunity and inflammation. Cell Mol Life Sci. 73:237–252. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Hashiguchi M, Kashiwakura Y, Kojima H, Kobayashi A, Kanno Y and Kobata T: IL-33 activates eosinophils of visceral adipose tissue both directly and via innate lymphoid cells. Eur J Immunol. 45:876–885. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Lee MW, Odegaard JI, Mukundan L, Qiu Y, Molofsky AB, Nussbaum JC, Yun K, Locksley RM and Chawla A: Activated type 2 innate lymphoid cells regulate beige fat biogenesis. Cell. 160:74–87. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Wensveen FM, Jelenčić V, Valentić S, Šestan M, Wensveen TT, Theurich S, Glasner A, Mendrila D, Štimac D, Wunderlich FT, et al: NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol. 16:376–385. 2015. View Article : Google Scholar : PubMed/NCBI

104 

Lee BC, Kim MS, Pae M, Yamamoto Y, Eberlé D, Shimada T, Kamei N, Park HS, Sasorith S, Woo JR, et al: Adipose natural killer cells regulate adipose tissue macrophages to promote insulin resistance in obesity. Cell Metab. 23:685–698. 2016. View Article : Google Scholar : PubMed/NCBI

105 

O'Sullivan TE, Rapp M, Fan X, Weizman OE, Bhardwaj P, Adams NM, Walzer T, Dannenberg AJ and Sun JC: Adipose-resident group 1 innate lymphoid cells promote obesity-associated insulin resistance. Immunity. 45:428–441. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Kim HY, Lee HJ, Chang YJ, Pichavant M, Shore SA, Fitzgerald KA, Iwakura Y, Israel E, Bolger K, Faul J, et al: IL-17 producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat Med. 20:54–61. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Wang X, Ota N, Manzanillo P, Kates L, Zavala-Solorio J, Eidenschenk C, Zhang J, Lesch J, Lee WP, Ross J, et al: Interleukin-22 alleviates metabolic disorders and restores mucosal immunity in diabetes. Nature. 514:237–241. 2014.PubMed/NCBI

108 

Hasnain SZ, Borg DJ, Harcourt BE, Tong H, Sheng YH, Ng CP, Das I, Wang R, Chen AC, Loudovaris T, et al: Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress. Nat Med. 20:1417–1426. 2014. View Article : Google Scholar : PubMed/NCBI

109 

Luck H, Tsai S, Chung J, Clemente-Casares X, Ghazarian M, Revelo XS, Lei H, Luk CT, Shi SY, Surendra A, et al: Regulation of obesity-related insulin resistance with gut anti-inflammatory agents. Cell Metab. 21:527–542. 2015. View Article : Google Scholar : PubMed/NCBI

110 

Garidou L, Pomié C, Klopp P, Waget A, Charpentier J, Aloulou M, Giry A, Serino M, Stenman L, Lahtinen S, et al: The gut microbiota regulates intestinal CD4 T cells expressing RORγt and controls metabolic disease. Cell Metab. 22:100–112. 2015. View Article : Google Scholar : PubMed/NCBI

111 

Ding S, Chi MM, Scull BP, Rigby R, Schwerbrock NM, Magness S, Jobin C and Lund PK: High-fat diet: Bacteria interactions promote intestinal inflammation which precedes and correlates with obesity and insulin resistance in mouse. PLoS One. 5:e121912010. View Article : Google Scholar : PubMed/NCBI

112 

Hashiguchi M, Kashiwakura Y, Kojima H, Kobayashi A, Kanno Y and Kobata T: Peyer's patch innate lymphoid cells regulate commensal bacteria expansion. Immunol Lett. 165:1–9. 2015. View Article : Google Scholar : PubMed/NCBI

113 

Veilleux A, Mayeur S, Bérubé JC, Beaulieu JF, Tremblay E, Hould FS, Bossé Y, Richard D and Levy E: Altered intestinal functions and increased local inflammation in insulin-resistant obese subjects: A gene-expression profile analysis. BMC Gastroenterol. 15:1192015. View Article : Google Scholar : PubMed/NCBI

114 

Monteiro-Sepulveda M, Touch S, Mendes-Sá C, André S, Poitou C, Allatif O, Cotillard A, Fohrer-Ting H, Hubert EL, Remark R, et al: Jejunal T cell inflammation in human obesity correlates with decreased enterocyte insulin signaling. Cell Metab. 22:113–124. 2015. View Article : Google Scholar : PubMed/NCBI

115 

Johnson AM, Costanzo A, Gareau MG, Armando AM, Quehenberger O, Jameson JM and Olefsky JM: High fat diet causes depletion of intestinal eosinophils associated with intestinal permeability. PLoS One. 10:e01221952015. View Article : Google Scholar : PubMed/NCBI

116 

Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, Kim SG, Li H, Gao Z, Mahana D, et al: Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 158:705–721. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 17 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Saetang J and Saetang J: Role of innate lymphoid cells in obesity and metabolic disease (Review). Mol Med Rep 17: 1403-1412, 2018
APA
Saetang, J., & Saetang, J. (2018). Role of innate lymphoid cells in obesity and metabolic disease (Review). Molecular Medicine Reports, 17, 1403-1412. https://doi.org/10.3892/mmr.2017.8038
MLA
Saetang, J., Sangkhathat, S."Role of innate lymphoid cells in obesity and metabolic disease (Review)". Molecular Medicine Reports 17.1 (2018): 1403-1412.
Chicago
Saetang, J., Sangkhathat, S."Role of innate lymphoid cells in obesity and metabolic disease (Review)". Molecular Medicine Reports 17, no. 1 (2018): 1403-1412. https://doi.org/10.3892/mmr.2017.8038