Skip to main content
Advertisement
  • Loading metrics

Innate lymphoid cells—Underexplored guardians of immunity

  • Irina Tsymala,

    Roles Conceptualization, Writing – original draft, Writing – review & editing

    Affiliation Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria

  • Karl Kuchler

    Roles Conceptualization, Writing – review & editing

    karl.kuchler@meduniwien.ac.at

    Affiliation Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria

Introduction

Innate lymphoid cells (ILCs) show remarkable plasticity and influence immunity as early sensors of pathogenic cues [14]. The general term “ILC” describes innate immune cells that display most common features of lymphocytes. ILCs comprise 5 different subsets: cytotoxic natural killer cells (NK cells), lymphoid tissue inducer cells (LTi), and 3 helper-like subsets, group 1 ILCs (ILC1), group 2 ILCs (ILC2), and group 3 ILCs (ILC3) [3,57]. ILCs are defined by their specific cytokine and transcription factor profiles, largely mirroring functions of T helper (Th) subsets [3,4,8]. However, the fate of ILCs is determined by Id2, which blocks T cell differentiation and drives ILC development [2,9,10]. As a consequence, ILCs typically lack antigen-specific T or B cell receptors [3], sandwiching their roles in between adaptive and innate immunity. In steady-state, ILC1s depend on T-bet and express interferon gamma (IFN-γ), as well as tumor necrosis factor alpha (TNF-α). ILC2s engage GATA3, RORα, Bcl11b along with GFI1 upon differentiation [2,3] and produce IL-13, IL-5, IL-4, IL-9, and amphiregulin [2,11,12]. Finally, ILC3s require RORγt, as well as AhR while secreting IL-22 and IL-17 [4,1315].

Most ILCs are long-lived, self-renewing cells residing at tissue barriers, were they readily respond to physiological or pathological triggers by cytokine release [2,4,16,17] (Table 1). Thereby, ILCs may acquire long-lasting immunological memories relevant for effective immune surveillance [1821]. ILCs can also fuel dysregulated immune responses causing chronic inflammation and thus worsen disease outcomes [4,22,23] (Table 1). However, biological mechanisms modulating polarization of ILCs in response to pathogens remain ill-posed, and even less is known about the plasticity between NK cells and ILCs [4,24]. Further, considerable differences between human and murine ILCs have been posing road blocks to gain a better understanding of their immunological functions [25,26]. In this work, we provide an overview about the properties of main ILC subsets, emphasizing possible roles in infectious diseases. We will discuss plasticity in response to different pathogens. Finally, we shall inspect the immunomodulatory potential of ILCs in patients, as well as future challenges in ILC biology.

thumbnail
Table 1. Relevance, functions, and regulation of ILCs in infectious diseases.

https://doi.org/10.1371/journal.ppat.1011678.t001

NK cells and ILC1s—Two sides of the same coin?

Although NK cells and ILC1s arise from a common lymphoid progenitor [2], both express IFN-γ and both are implicated in the early clearance of intracellular pathogens [27]. Especially human NK cells are phenotypically difficult to distinguish from ILC1s due to their overlapping surface receptor expression, such as NKp46, NK1.1 (mouse), CD56 (human) [6,27] (Fig 1), and owing to a lack of unique, subset-defining hallmarks.

thumbnail
Fig 1. ILC1s and conventional NK cells (cNKs) express common and distinct cell surface receptors.

Both subsets share interspecific conserved features, including cell surface markers (Ly49-family receptors in mouse/Killer Cell Immunoglobulin like Receptors, KIRs, in human, DNAM-1, NK1.1 in mouse/CD161 in human, Nkp46, CD44, TRAIL, TIGIT), transcription factors (T-bet, Hobit), cytotoxic features (granzyme A, B and granzyme C in mouse, perforin expression), and cytokine expression profile (IFN-γ, TNF-α, GM-CSF). Mouse ILC1s can be distinguished by the lack of lineage-defining surface molecules (T or B cell receptor, CD19, B220) and the expression of CD200r1, Syndecan-4, CD49a, and IL-7Rα. Lineage-negative human ILC1s may express Eomes at low levels as well as surface receptors associated with cNK cells; (*)-marked features are only present in certain environments. Created with BioRender.com. cNK, conventional NK; ILC, innate lymphoid cell.

https://doi.org/10.1371/journal.ppat.1011678.g001

The NK lineage commitment is initiated by Eomesodermin (Eomes), whereas ILC1 up-regulates T-bet instead, along with surface receptors such as IL-7Rα, CD90, CD49a, and CD200r1 [6,21,28,29] (Fig 1). Expression of these subset markers may differ depending on their microenvironment and activation state, often resulting in a rather blurred distinction between tissue-resident NK cells and ILC1s [3,3032]. Although NK cells and ILC1s diverge during development, some degree of plasticity may be maintained. Of note, TGF-β induces a metabolic transition in NK cells, promoting oxidative phosphorylation and adoption of an ILC1-like phenotype, again highlighting the close relation of these subsets [6,20].

ILC1s can be primed by infections that favor the acquisition of memory-like phenotypes [33]. Similar to memory T cells, pathogen-trained ILC1s show distinct transcriptional patterns [19,20]. Trained ILC1s can increase expression of Ly6c, IL-2Rα, IL-7Rα, and IL-18R and contribute to pathogen resistance after re-challenge by increasing IFN-γ secretion [1820,34,35]. In a mouse model of pulmonary Mycobacterium tuberculosis (Mtb) infection, adoptive transfer of IL-18R+ IFN-γ-expressing ILCs improves pathogen clearance in lymphocyte-deficient mice [20]. A pathogen-experienced ILC1-like subset also appears after Toxoplasma gondii infections [18], since a Stat4-dependent ILC subset persists in the circulation resembling mature NK-cells by surface Ly6c [18,35].

Although ILC1s can produce cytolytic granules, their response mainly entails cytokine secretion and signaling [24,29]. Transition to a mature cytotoxic effector state in ILC1s results in the down-regulation of IL-7Rα regulated by Hobit (Homolog of Blimp-1 in T cells), similar to invariant natural killer T cells (iNKT) and NK cells [36]. Notably, ILC1s share key-effector properties with iNKTs and NK cells, while their physiological functions as cytotoxic effector cells remain unresolved. ILC1s may be dispensable for the control of intracellular pathogens if they are compensated for by other cytotoxic subsets, including conventional NK cells or cytotoxic T cells [37,38]. Nonetheless, IFN-γ secretion by ILC1s can impede viral replication in various mouse models [20,28,34,39,40]. ILC1s are mainly located at epithelial barrier sites and respond within the first hours of infection, acting even before cytotoxic NK cells. This fast-acting surveillance or danger-sensing may offer a crucial advantage to host defense [38,39].

ILC2s—Indispensable for mucosal type-2-immunity?

ILC2s respond to mucosal alarmins such as IL-25, thymic stromal lymphopoietin (TSLP) as well as IL-33 [41]. Especially engagement of ST2 (IL-33R) triggers fast expansion of ILC2s [4143]. Consequently, ILC2s amplify allergic response, eosinophilia, and hyperreactivities in the skin, lung, and respiratory tract.

ILC2s are also associated with excessive inflammation in mucosal infections [22,43]. For example, ILC2s exacerbate Aspergillus fumigatus infections in a mouse model of cystic fibrosis, where mast cells activate ILCs via IL-2 signaling, in turn resulting in IL-9 release by ILC2s and Th9 cells [14]. Furthermore, depletion of ILC2s leads to prolonged survival of mice infected by Cryptococcus neoformans, as type-1-immunity and pro-inflammatory activation of pulmonary macrophages is enhanced [22]. ILC2 expansion during inflammation is tightly linked to their distinct metabolic profile as defined by Arginase-1 (Arg1) expression [4,44]. Inhibition of Arg1 dampens ILC2s but not ILC3s, and affects several metabolic pathways, including aerobic glycolysis [4,44]. Further, Mtb-associated type-1-inflammation and reprograming towards glycolysis induces a conversion of ILC2s into IFN-γ+ ILC1-like cells [4,20]. In contrast, in vitro challenge with Pseudomonas aeroginosa, Staphylococcus aureus, or a combination of IL-23, IL-1β, and TGF-β induces transition towards an ILC3-like phenotype [45].

Of note, a targeted depletion of ILC subsets has been difficult to achieve, since they share common surface receptors and lineage-defining transcription factors with conventional NK or T cells. Interestingly, the recently identified neuromedin U receptor 1 (Nmur1) might form an exception, since ILC2s constitutively express Nmur1, which distinguishes them from other immune cells. Thus, ILC2s may also play a role in the cross-talk with sensory neurons [2,46,47]. Indeed, ectopic Nmur1 expression allows for the generation of mouse models suitable for in vivo tracking and targeted gene-deletion in ILC2s [46,47]. For instance, depletion of ILC2s, using Nmur1iCre-eGFP Id2fl/fl or Nmur1iCre-eGFP Gata3fl/fl mice, reveals that eosinophil infiltration during pulmonary helminth infections is directly linked to early IL-5 secretion by ILC2s [47]. In addition, ILC2-deficiency in Klrg1creGata3fl/fl mice significantly delays complete clearance of intestinal Nippostrongylus brasiliensis infections [48]. Consequently, the presence of ILC2s is vital for clearance of helminth infections, as well as for inducing early type-2-inflammation [47,48]. Furthermore, ILC2s are a main source of amphiregulin in intestinal tissues, exerting a protective regulatory impact on mucosal epithelia in homeostasis but also during parasite infection [46]. ILC2s can also regulate inflammation via IL-10 secretion [3,49,50]. In contrast to Treg cells, ILC2s do not require FoxP3 [51]. Instead, IL-10 induction in ILC2s is associated with the up-regulation of Id3, but also Atf3, Blimp-1, cMaf, Klf2, and Foxf1 may be involved [4951]. Further, retinoic acid, IL-2, IL-10, IL-27, IL-4, and NMU can induce regulatory ILC2s in vitro while the TL1A functions as a negative regulator [50,51]. Although regulatory ILC2s are pivotal in allergy and tissue integrity, their relevance and potential roles during infections remain open [49,50].

ILC3s—Threat or treat for intestinal immunity

RORγt guides the maturation of ILC3s and LTis, the latter being essential for the development of secondary lymphoid tissues [2,9]. LTis differ from ILC3s by their cell surface expression of CCR6, CD4, and c-Kit [2,52]. ILC3s inhabit the lamina propria of the small intestine and colon, as well as other mucosal tissues, where they respond to microbial and dietary compounds engaging the AhR-signaling pathway [2,4,52,53]. In contrast to Th cells, ILC3s develop also in germ-free mice [9].

Mucosal ILC3s secrete IL-17 and GM-CSF in the early phase of infections to recruit neutrophils and to promote their activation [2,5,54]. In addition, ILC3s-mediated cross-talk with epithelial cells via IL-22 sustains barrier integrity and supports the release of defensins and antimicrobial peptides to restrain excessive damage after infection [55,56]. Resident ILC3s, as well as recruited IL-17-producing RORγt+ ILC2s, are early sources of IL-17A, thus contributing to the control and perhaps clearance of mucosal candidiasis in absence of adaptive immunity [43,57]. Of note, dysregulated colonization of the gut with Candida spp. is instead associated with activation of ILC3s that display tumorgenic potential [58]. Further, IL-17A+ILC3s can also contribute to Salmonella enterica Typhimurium serovar (S. Typhimurium)-induced gut fibrosis [12]. IL-17A production at later stages of chronic disease is regulated by RORα and becomes a driving factor of gut fibrosis.

ILC3s can release IFN-γ after gradually up-regulating T-bet. Interestingly, these beneficial T-bet+ILC3s (ex-ILC3s) restrain S. Typhimurium as well as Listeria monocytogenes infections [40,59]. Contrary to IL-22+ ILC3s, ex-ILC3s are redundant in intestinal Citrobacter rodentium infections, and they promote inflammation via GM-CSF signaling [54,60,61].

ILC3s also cooperate with other lymphocytes to orchestrate intestinal immunity and to regulate the response to microbiota and pathogens during aging [17,6264]. For instance, certain E3 ubiquitin ligases, such as cIAP1/2, modulate proliferation and function of γδ-T cells and intestinal ILC3s [64]. Defects in cIAP1/2 affect immunity only gradually over time, comprising ILC3-related IL-22 production and increasing susceptibility to C.rodentium infection.

A subset of ILC3s can also execute MHCII antigen presentation, and they express co-stimulatory surface molecules [63,65]. Although a few studies indicate that ILC3s can process and present antigens to T cells, this matter remains controversial [63,66,67]. A regulatory effect on T cell homeostasis may arise from competition for IL-2 recognition, albeit without providing co-stimulatory signals [63]. Of note, a novel extrathymic autoimmune regulator (AIRE) and MHCII-expressing cell lineage could mediate microbial-tolerance in early development. This subset appears related but transcriptionally distinct from dendritic cells and ILCs subsets [66,67].

Notably, MHCII-related functions are not restricted to ILC3-like polarization, as they are also observed in ILC2s and NK cells [6870]. Moreover, a variety of other innate immune cells including NK cells and myeloid cells may have similar barrier-protective properties. Accordingly, ILC3s are important for barrier integrity, but their role in mucosal immune surveillance needs to be further elucidated.

Can helper-ILCs exhibit immunomodulatory functions in patients?

Current knowledge about the identification and function of helper ILCs is largely based on mouse models. However, surface markers that separate ILCs clearly from other lineages, especially human ILC1s from NK cells, are still missing. Further, more clinical studies and disease implications will be critical to determine whether ILCs can offer targetable cell types in infectious disease settings. Engraftment of human ILC precursors into humanized or immunodeficient (NSG) mice may be a first step to explore the immunomodulatory potential of ILCs. This approach may also provide appropriate models to investigate the development, expansion, and plasticity of transferred ILC-precursors in the human setting [26,71,72]. The low abundance of peripheral blood ILCs pose extreme technical challenges for isolation and identification. Hence, robust methods for ex vivo expansion of ILC-progenitors from peripheral blood, bone marrow, or umbilical cord blood are urgently needed [42,71,73]. Of note, proof-of-concept studies show that human ILCs can be obtained from various sources, remain viable and functional under in vitro expansion conditions, offering interesting options for cell-based therapies similar to T or NK cells [73].

Importantly, haplo-identical and allogeneic NK cells are tested in several clinical studies [7476]. NK cells can be engineered in vitro to enhance their CD16-mediated antibody-dependent cytotoxicity (ADCC) rendering them effective against hematopoietic malignancies [74]. Several strategies are currently used to enhance NK cytotoxicity in the context of refractory viral infections. A combination of NK cell activation by the IL-15 super-agonist ALT-803, anti-CD16-fusion antibodies or vaccines with antiretroviral therapy (ART) are in clinical studies to treat HIV infections [75,76]. NK cell therapies offer several advantages when compared to T cell-based approaches. First, NK cells do not rely on prior antigen sensitization. Second, they are less likely to trigger a cytokine-release syndrome. Third, they may have less toxic effects and do not drive excessive graft-versus-host disease [74,76]. Finally, NK cells can even come from non-autologous sources and may be beneficial for treating lymphopenic patients. Interestingly enough, links for ILC functions in systemic infections by HIV, Mtb, SARS-CoV-2 or bacterial sepsis are just emerging [7782]. Hence, it is tempting to speculate that modulating ILC functions in certain infectious disease settings could be effective and support treatments of otherwise refractory diseases.

Current and future challenges

In general, ILCs are primarily tissue-resident and organ-specific immune cells of very low abundance. Low cell numbers pose serious challenges for in vivo studies in either mice or humans, but such studies are pivotal for determining relevant in vivo functions. Correspondingly, an age-dependent decline in different ILC subsets may be relevant for health, but studies in aging mice or individuals are still scarce [30,64,80,83,84]. Moreover, human ILC1s and NK cells are very difficult to distinguish (Fig 1) [6,27], and unique, subset defining features remain elusive. One possibility is a high degree of plasticity as well as redundancy between these 2 subsets. This scenario mirrors Th subsets were transcriptional reprogramming or adaptive rewiring can derepress lineage-specific genes that determine T cell cytotoxicity [85]. In this case by down-regulating ThPOK and subsequent up-regulation of Runx3 along with the CBFβ-complexes [15,40,86]. Accordingly, Runx3 is also essential for development and maintenance of ILC1s and Nkp46+ILC3s, while ThPOK negatively regulates RORγt and IL23R expression [15,40,59]. In contrast, neither Runx3 nor the loss of ThPOK correlates with the induction of a cytotoxic effector program in ILCs, highlighting essential differences in regulation of these cell types. In mice, acquisition of an ILC1-associated transcriptional profile is rather linked to phenotypes associated with exhaustion [24,85,87]. In view of NK-cell-based therapies, it will be mandatory to understand the dynamics in human ILCs [85]. Other transcriptional regulators, in particular, histone deacetylase 1 (HDAC1) and HDAC2 are also potent regulators of cytotoxicity in T cells [88]. It will be exciting to further explore whether these chromatin modifiers can also contribute to the adaptive potential and plasticity of ILCs [89] similar to the situation in T cell lineages, where HDACs have been further associated with T helper subset defects in health and autoimmunity [88,90,91].

Further, transcriptional and phenotypical heterogeneities within ILC subsets are dynamically influenced by their immunological microenvironments. Thus, ILCs may reflect functional continua and appear as transition cell types [30,31]. Current strategies rely on negative selection of ILCs, sorted by the lack of lineage-defining surface markers and by the expression of IL-7Rα, CD90, and CD49a [2,9295]. However, canonical ILC markers such as CD90 and IL-7R are subject to regulation and thus not constantly expressed on the surface of intestinal or pulmonary ILCs, respectively [31,32,96]. Moreover, the integrin CD49a may also show tissue-related variation [3,31]. Lineage-defining negative markers such as Ly6c may be in addition up-regulated following inflammatory challenges or in distinct subsets during steady state [18,19,31]. Categorization into canonical subsets can therefore create a bias in the interpretation of skyrocketing next-generation single-cell RNA-seq datasets. Hence, the analysis and clustering of single-cell data should be done in great detail and with caution, since interpretations should require at least some validation. Nonetheless, single-cell–based technologies still constitute an essential tool in studying small cell populations in general and ILCs particularly in an organ- or disease-specific context. Such technologies will without any doubt propel our understanding of the complex cellular interplay in vivo to further unravel relevant features of ILCs in disease.

Acknowledgments

Finally, we apologize to contributors to ILC biology for not referencing some relevant papers about ILC functions unrelated to infections, owing to space limitations of this format.

References

  1. 1. Klose CSN, Artis D. Innate lymphoid cells control signaling circuits to regulate tissue-specific immunity. Cell Res. 2020 306 [Internet]. 2020 May 6 [cited 2021 Oct 28];30(6):475–91. Available from: https://www.nature.com/articles/s41422-020-0323-8. pmid:32376911
  2. 2. Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. Innate Lymphoid Cells: 10 Years On. Cell. 2018 Aug 23;174(5):1054–66. pmid:30142344
  3. 3. Bal SM, Golebski K, Spits H. Plasticity of innate lymphoid cell subsets. Nat Rev Immunol [Internet]. 2020;20(9):552–65. pmid:32107466
  4. 4. Pelletier A, Stockmann C. The Metabolic Basis of ILC Plasticity. Front Immunol [Internet]. 2022 Apr 29 [cited 2022 Nov 25];13. Available from: https://pubmed.ncbi.nlm.nih.gov/35572512/. pmid:35572512
  5. 5. Croft CA, Thaller A, Marie S, Doisne JM, Surace L, Yang R, et al. Notch, RORC and IL-23 signals cooperate to promote multi-lineage human innate lymphoid cell differentiation. Nat Commun [Internet]. 2022 Dec 1 [cited 2022 Nov 25];13(1). Available from: https://pubmed.ncbi.nlm.nih.gov/35896601/. pmid:35896601
  6. 6. Krabbendam L, Bernink JH, Spits H. Innate lymphoid cells: from helper to killer. Curr Opin Immunol [Internet]. 2021 Feb 1 [cited 2022 Nov 25];68:28–33. Available from: https://pubmed.ncbi.nlm.nih.gov/32971468/. pmid:32971468
  7. 7. Olguín-Martínez E, Ruiz-Medina BE, Licona-Limón P. Tissue-Specific Molecular Markers and Heterogeneity in Type 2 Innate Lymphoid Cells. Front Immunol. 2021 Oct 25;12:4197. pmid:34759931
  8. 8. Bernink JH, Peters CP, Munneke M, Te Velde AA, Meijer SL, Weijer K, et al. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat Immunol [Internet]. 2013 [cited 2022 Dec 7];14(3):221–9. Available from: https://pubmed.ncbi.nlm.nih.gov/23334791/. pmid:23334791
  9. 9. Colonna M. Innate Lymphoid Cells: Diversity, Plasticity, and Unique Functions in Immunity. Immunity. 2018 Jun 19;48(6):1104–17. pmid:29924976
  10. 10. Shin SB, McNagny KM. ILC-You in the Thymus: A Fresh Look at Innate Lymphoid Cell Development. Front Immunol. 2021 May 6;12:681110. pmid:34025680
  11. 11. Lo BC, Canals Hernaez D, Scott RW, Hughes MR, Shin SB, Underhill TM, et al. The Transcription Factor RORα Preserves ILC3 Lineage Identity and Function during Chronic Intestinal Infection. J Immunol [Internet]. 2019 Dec 15 [cited 2023 Aug 29];203(12):3209–15. pmid:31676672
  12. 12. Lo BC, Gold MJ, Hughes MR, Antignano F, Valdez Y, Zaph C, et al. The orphan nuclear receptor RORa and group 3 innate lymphoid cells drive fibrosis in a mouse model of Crohn’s disease. Sci Immunol [Internet]. 2016 [cited 2023 Aug 17];1(3). Available from: https://pubmed.ncbi.nlm.nih.gov/28783681/.
  13. 13. Taggenbrock RLRE, Gisbergen KPJM van. ILC1: Development, maturation, and transcriptional regulation. Eur J Immunol [Internet]. 2022 Nov 27 [cited 2022 Nov 28];0:1–12. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/eji.202149435. pmid:36408791
  14. 14. Moretti S, Renga G, Oikonomou V, Galosi C, Pariano M, Iannitti RG, et al. A mast cell-ILC2-Th9 pathway promotes lung inflammation in cystic fibrosis. Nat Commun [Internet]. 2017 Jan 16 [cited 2022 Oct 2];8. Available from: /pmc/articles/PMC5241810/. pmid:28090087
  15. 15. Ebihara T, Song C, Ryu SH, Plougastel-Douglas B, Yang L, Levanon D, et al. Runx3 specifies lineage commitment of innate lymphoid cells. Nat Immunol. 2015;16(11):1124–33. pmid:26414766
  16. 16. Korchagina AA, Koroleva E, Tumanov A V. Innate Lymphoid Cells in Response to Intracellular Pathogens: Protection Versus Immunopathology. Front Cell Infect Microbiol. 2021 Dec 6;11:1168. pmid:34938670
  17. 17. Klose CSN, Artis D. Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis. Nat Immunol. 2016 Jun 21;17(7):765–74. pmid:27328006
  18. 18. Park E, Patel S, Wang Q, Andhey P, Zaitsev K, Porter S, et al. Toxoplasma gondii infection drives conversion of NK cells into ILC1-like cells. Elife. 2019 Aug 1;8. pmid:31393266
  19. 19. Weizman O El, Song E, Adams NM, Hildreth AD, Riggan L, Krishna C, et al. Mouse cytomegalovirus-experienced ILC1s acquire a memory response dependent on the viral glycoprotein m12. Nat Immunol. 2019 208 [Internet]. 2019 Jul 1 [cited 2022 Dec 7];20(8):1004–11. Available from: https://www.nature.com/articles/s41590-019-0430-1. pmid:31263280
  20. 20. Corral D, Charton A, Krauss MZ, Hepworth MR, Neyrolles O, Hudrisier D, et al. ILC precursors differentiate into metabolically distinct ILC1-like cells during Mycobacterium tuberculosis infection. Cell Rep [Internet]. 2022 Apr 19 [cited 2022 Dec 14];39(3):110715. pmid:35443177
  21. 21. Favaro RR, Phillips K, Delaunay-Danguy R, Ujčič K, Markert UR. Emerging Concepts in Innate Lymphoid Cells, Memory, and Reproduction. Front Immunol [Internet]. 2022 Jun 14 [cited 2022 Nov 25];13. Available from: https://pubmed.ncbi.nlm.nih.gov/35774779/. pmid:35774779
  22. 22. Kindermann M, Knipfer L, Obermeyer S, Müller U, Alber G, Bogdan C, et al. Group 2 Innate Lymphoid Cells (ILC2) Suppress Beneficial Type 1 Immune Responses During Pulmonary Cryptococcosis. Front Immunol [Internet]. 2020 Feb 14 [cited 2022 Dec 5];11:209. Available from: /pmc/articles/PMC7034304/. pmid:32117319
  23. 23. Muraoka WT, Korchagina AA, Xia Q, Shein SA, Jing X, Lai Z, et al. Campylobacter infection promotes IFNγ-dependent intestinal pathology via ILC3 to ILC1 conversion. Mucosal Immunol 2020 143 [Internet]. 2020 Nov 19 [cited 2022 Dec 13];14(3):703–16. Available from: https://www.nature.com/articles/s41385-020-00353-8.
  24. 24. Nixon BG, Chou C, Krishna C, Dadi S, Michel AO, Cornish AE, et al. Cytotoxic granzyme C-expressing ILC1s contribute to antitumor immunity and neonatal autoimmunity. Sci Immunol [Internet]. 2022 Apr 1 [cited 2022 Nov 25];7(70). Available from: https://pubmed.ncbi.nlm.nih.gov/35394814/. pmid:35394814
  25. 25. Crinier A, Milpied P, Escalière B, Piperoglou C, Galluso J, Balsamo A, et al. High-Dimensional Single-Cell Analysis Identifies Organ-Specific Signatures and Conserved NK Cell Subsets in Humans and Mice. Immunity. 2018 Nov 20;49(5):971–986.e5. pmid:30413361
  26. 26. Horowitz NB, Mohammad I, Moreno-Nieves UY, Koliesnik I, Tran Q, Sunwoo JB. Humanized Mouse Models for the Advancement of Innate Lymphoid Cell-Based Cancer Immunotherapies. Front Immunol. 2021 Apr 22;0:977. pmid:33968039
  27. 27. Seillet C, Brossay L, Vivier E. Natural killers or ILC1s? That is the question. Curr Opin Immunol. 2021 Feb 1;68:48–53. pmid:33069142
  28. 28. Trittel S, Vashist N, Ebensen T, Chambers BJ, Guzmán CA, Riese P. Invariant NKT cell-mediated modulation of ILC1s as a tool for mucosal immune intervention. Front Immunol. 2019 Aug 7;10(AUG):1849. pmid:31440243
  29. 29. Di Censo C, Marotel M, Mattiola I, Müller L, Scarno G, Pietropaolo G, et al. Granzyme A and CD160 expression delineates ILC1 with graded functions in the mouse liver. Eur J Immunol [Internet]. 2021 Nov 1 [cited 2022 Dec 7];51(11):2568–75. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/eji.202149209. pmid:34347289
  30. 30. Yudanin NA, Schmitz F, Flamar AL, Thome JJC, Tait Wojno E, Moeller JB, et al. Spatial and Temporal Mapping of Human Innate Lymphoid Cells Reveals Elements of Tissue Specificity. Immunity [Internet]. 2019;50(2):505–519.e4. pmid:30770247
  31. 31. McFarland AP, Yalin A, Wang SY, Cortez VS, Landsberger T, Sudan R, et al. Multi-tissue single-cell analysis deconstructs the complex programs of mouse natural killer and type 1 innate lymphoid cells in tissues and circulation. Immunity [Internet]. 2021;54(6):1320–1337.e4. pmid:33945787
  32. 32. Schroeder JH, Beattie G, Lo JW, Zabinski T, Powell N, Neves JF, et al. CD90 is not constitutively expressed in functional innate lymphoid cells. Front Immunol. 2023;14(April):1–13.
  33. 33. Wang X, Peng H, Tian Z. Innate lymphoid cell memory. Cell Mol Immunol. 2019 165 [Internet]. 2019 Feb 22 [cited 2022 Dec 5];16(5):423–9. Available from: https://www.nature.com/articles/s41423-019-0212-6. pmid:30796350
  34. 34. Steigler P, Daniels NJ, McCulloch TR, Ryder BM, Sandford SK, Kirman JR. BCG vaccination drives accumulation and effector function of innate lymphoid cells in murine lungs. Immunol Cell Biol. 2018;96(4):379–89. pmid:29363172
  35. 35. Pionnier N, Furlong-Silva J, Colombo SAP, Marriott AE, Chunda VC, Ndzeshang BL, et al. NKp46+ natural killer cells develop an activated/memory-like phenotype and contribute to innate immunity against experimental filarial infection. Front Immunol [Internet]. 2022 Sep 27 [cited 2022 Nov 25];13. Available from: https://pubmed.ncbi.nlm.nih.gov/36238293/. pmid:36238293
  36. 36. Friedrich C, Taggenbrock RLRE, Doucet-Ladevèze R, Golda G, Moenius R, Arampatzi P, et al. Effector differentiation downstream of lineage commitment in ILC1s is driven by Hobit across tissues. Nat Immunol [Internet]. 2021 Oct 1 [cited 2022 Nov 25];22(10):1256–67. Available from: https://pubmed.ncbi.nlm.nih.gov/34462601/. pmid:34462601
  37. 37. Vély F, Barlogis V, Vallentin B, Neven B, Piperoglou C, Ebbo M, et al. Evidence of innate lymphoid cell redundancy in humans. Nat Immunol. 2016 1711 [Internet]. 2016 Sep 12 [cited 2022 Dec 5];17(11):1291–9. Available from: https://www.nature.com/articles/ni.3553. pmid:27618553
  38. 38. Weizman O El, Adams NM, Schuster IS, Krishna C, Pritykin Y, Lau C, et al. ILC1 Confer Early Host Protection at Initial Sites of Viral Infection. Cell. 2017 Nov 2;171(4):795–808.e12. pmid:29056343
  39. 39. Shannon JP, Vrba SM, Reynoso G V., Wynne-Jones E, Kamenyeva O, Malo CS, et al. Group 1 innate lymphoid-cell-derived interferon-γ maintains anti-viral vigilance in the mucosal epithelium. Immunity. 2021 Feb 9;54(2):276–290.e5.
  40. 40. Yin S, Yu J, Hu B, Lu C, Liu X, Gao X, et al. Runx3 mediates resistance to intracellular bacterial infection by promoting IL12 signaling in group 1 ILC and NCR+ILC3. Front Immunol. 2018;9(SEP):1–15. pmid:30258450
  41. 41. Van Dyken SJ, Nussbaum JC, Lee J, Molofsky AB, Liang HE, Pollack JL, et al. A tissue checkpoint regulates type 2 immunity. Nat Immunol [Internet]. 2016 Dec 1 [cited 2022 Nov 25];17(12):1381–7. Available from: https://pubmed.ncbi.nlm.nih.gov/27749840/. pmid:27749840
  42. 42. Trabanelli S, Ercolano G, Wyss T, Gomez-Cadena A, Falquet M, Cropp D, et al. c-Maf enforces cytokine production and promotes memory-like responses in mouse and human type 2 innate lymphoid cells. EMBO J [Internet]. 2022 Jun 14 [cited 2022 Nov 25];41(12). Available from: https://pubmed.ncbi.nlm.nih.gov/35467036/.
  43. 43. Huang Y, Guo L, Qiu J, Chen X, Hu-Li J, Siebenlist U, et al. IL-25-responsive, lineage-negative KLRG1 hi cells are multipotential “inflammatory” type 2 innate lymphoid cells HHS Public Access. Nat Immunol. 2015;16(2):161–9.
  44. 44. Monticelli LA, Buck MD, Flamar AL, Saenz SA, Wojno EDT, Yudanin NA, et al. Arginase 1 is an innate lymphoid-cell-intrinsic metabolic checkpoint controlling type 2 inflammation. Nat Immunol. 2016;17(6):656–65. pmid:27043409
  45. 45. Golebski K, Ros XR, Nagasawa M, van Tol S, Heesters BA, Aglmous H, et al. IL-1β, IL-23, and TGF-β drive plasticity of human ILC2s towards IL-17-producing ILCs in nasal inflammation. Nat Commun [Internet]. 2019 Dec 1 [cited 2023 Aug 29];10(1). Available from: https://pubmed.ncbi.nlm.nih.gov/31089134/.
  46. 46. Tsou AM, Yano H, Parkhurst CN, Mahlakõiv T, Chu C, Zhang W, et al. Neuropeptide regulation of non-redundant ILC2 responses at barrier surfaces. Nature [Internet]. 2022 [cited 2022 Nov 25];611(7937). Available from: https://pubmed.ncbi.nlm.nih.gov/36323781/. pmid:36323781
  47. 47. Jarick KJ, Topczewska PM, Jakob MO, Yano H, Arifuzzaman M, Gao X, et al. Non-redundant functions of group 2 innate lymphoid cells. Nature. 2022 6117937 [Internet]. 2022 Nov 2 [cited 2022 Dec 13];611(7937):794–800. Available from: https://www.nature.com/articles/s41586-022-05395-5. pmid:36323785
  48. 48. Gurram RK, Wei D, Yu Q, Zhao K, Leonard WJ, Zhu J. Crosstalk between ILC2s and Th2 cells varies among mouse models. Cell Rep [Internet]. 2023 [cited 2023 Aug 17];42:112073. pmid:36735533
  49. 49. Golebski K, Layhadi JA, Sahiner U, Steveling-Klein EH, Lenormand MM, Li RCY, et al. Induction of IL-10-producing type 2 innate lymphoid cells by allergen immunotherapy is associated with clinical response. Immunity [Internet]. 2021 Feb 9 [cited 2023 Aug 17];54(2):291–307.e7. Available from: https://pubmed.ncbi.nlm.nih.gov/33450188/. pmid:33450188
  50. 50. Bando JK, Gilfillan S, Di Luccia B, Fachi JL, Fachi JL, Sécca C, et al. ILC2s are the predominant source of intestinal ILC-derived IL-10. J Exp Med. 2020;217(2). pmid:31699824
  51. 51. Jegatheeswaran S, Mathews JA, Crome SQ. Searching for the Elusive Regulatory Innate Lymphoid Cell. J Immunol [Internet]. 2021 Oct 15 [cited 2022 Nov 25];207(8):1949–57. Available from: https://pubmed.ncbi.nlm.nih.gov/34607908/. pmid:34607908
  52. 52. Sonnenberg GF, Monticelli LA, Elloso MM, Fouser LA, Artis D. CD4+ Lymphoid Tissue-Inducer Cells Promote Innate Immunity in the Gut. Immunity. 2011 Jan 28;34(1):122–34. pmid:21194981
  53. 53. de Araújo EF, Loures F V., Preite NW, Feriotti C, Galdino NAL, Costa TA, et al. AhR Ligands Modulate the Differentiation of Innate Lymphoid Cells and T Helper Cell Subsets That Control the Severity of a Pulmonary Fungal Infection. Front Immunol. 2021 Apr 16;12:1277. pmid:33936043
  54. 54. Song C, Lee JS, Gilfillan S, Robinette ML, Newberry RD, Stappenbeck TS, et al. Unique and redundant functions of NKp46+ ILC3s in models of intestinal inflammation. J Exp Med [Internet]. 2015 Oct 10 [cited 2022 Dec 5];212(11):1869. Available from: /pmc/articles/PMC4612098/. pmid:26458769
  55. 55. Zindl CL, Witte SJ, Laufer VA, Gao M, Yue Z, Janowski KM, et al. A nonredundant role for T cell-derived interleukin 22 in antibacterial defense of colonic crypts. Immunity. 2022;55(3):494–511.e11.
  56. 56. Van Maele L, Carnoy C, Cayet D, Ivanov S, Porte R, Deruy E, et al. Activation of type 3 innate lymphoid cells and interleukin 22 secretion in the lungs during streptococcus pneumoniae infection. J Infect Dis. 2014;210(3):493–503. pmid:24577508
  57. 57. 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 [Internet]. 2013 Jan 15 [cited 2022 Dec 5];190(2):521–5. Available from: https://journals.aai.org/jimmunol/article/190/2/521/39486/Cutting-Edge-IL-17-Secreting-Innate-Lymphoid-Cells. pmid:23255360
  58. 58. Zhu Y, Shi T, Lu X, Xu Z, Qu J, Zhang Z, et al. Fungal-induced glycolysis in macrophages promotes colon cancer by enhancing innate lymphoid cell secretion of IL-22. EMBO J [Internet]. 2021 Jun 1 [cited 2022 Dec 5];40(11):e105320. Available from: https://onlinelibrary.wiley.com/doi/full/10.15252/embj.2020105320. pmid:33591591
  59. 59. Gao X, Shen X, Liu K, Lu C, Fan Y, Xu Q, et al. The Transcription Factor ThPOK Regulates ILC3 Lineage Homeostasis and Function During Intestinal Infection. Front Immunol. 2022;13(July):1–17. pmid:35844574
  60. 60. Elemam NM, Ramakrishnan RK, Hundt JE, Halwani R, Maghazachi AA, Hamid Q. Innate Lymphoid Cells and Natural Killer Cells in Bacterial Infections: Function, Dysregulation, and Therapeutic Targets. Front Cell Infect Microbiol. 2021 Nov 5;11:1093. pmid:34804991
  61. 61. Reynders A, Yessaad N, Vu Manh TP, Dalod M, Fenis A, Aubry C, et al. Identity, regulation and in vivo function of gut NKp46+RORγt+ and NKp46+RORγt- lymphoid cells. EMBO J [Internet]. 2011 Jul 20 [cited 2022 Dec 5];30(14):2934–47. Available from: https://pubmed.ncbi.nlm.nih.gov/21685873/.
  62. 62. Valle-Noguera A, Ochoa-Ramos A, Gomez-Sánchez MJ, Cruz-Adalia A. Type 3 Innate Lymphoid Cells as Regulators of the Host-Pathogen Interaction. Front Immunol [Internet]. 2021 Sep 29 [cited 2022 Dec 13];12. Available from: /pmc/articles/PMC8511434/. pmid:34659248
  63. 63. Hepworth MR, Fung TC, Masur SH, Kelsen JR, McConnell FM, Dubrot J, et al. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4+ T cells. Science (80-) [Internet]. 2015 May 29 [cited 2022 Nov 30];348(6238):1031–5. Available from: https://www.science.org/doi/10.1126/science.aaa4812.
  64. 64. Rizk J, Mörbe UM, Agerholm R, Virginia Baglioni M, Catafal Tardos E, Fares da Silva MGF, et al. The cIAP ubiquitin ligases sustain type 3 γδ T cells and ILC during aging to promote barrier immunity. 2023 [cited 2023 Aug 17]; pmid:37440178
  65. 65. Fei Teng A, Tachó -Piñ ot R, Sung B, Lambrecht BN, Hepworth MR, Sonnenberg Correspondence GF, et al. ILC3s control airway inflammation by limiting T cell responses to allergens and microbes. Cell Rep [Internet]. 2021 [cited 2022 Dec 5];37. pmid:34818549
  66. 66. Akagbosu B, Tayyebi Z, Shibu G, Paucar Iza YA, Deep D, Parisotto YF, et al. Novel antigen-presenting cell imparts Treg-dependent tolerance to gut microbiota. Nature. 2022 6107933 [Internet]. 2022 Sep 7 [cited 2022 Dec 2];610(7933):752–60. Available from: https://www.nature.com/articles/s41586-022-05309-5. pmid:36070798
  67. 67. Kedmi R, Najar TA, Mesa KR, Grayson A, Kroehling L, Hao Y, et al. A RORγt+ cell instructs gut microbiota-specific Treg cell differentiation. Nature. 2022 6107933 [Internet]. 2022 Sep 7 [cited 2022 Dec 13];610(7933):737–43. Available from: https://www.nature.com/articles/s41586-022-05089-y.
  68. 68. Oliphant CJ, Hwang YY, Walker JA, Salimi M, Wong SH, Brewer JM, 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 [Internet]. 2014 Aug 8 [cited 2022 Dec 14];41(2):283. Available from: /pmc/articles/PMC4148706/. pmid:25088770
  69. 69. van der Ploeg EK, Carreras Mascaro A, Huylebroeck D, Hendriks RW, Stadhouders R. Group 2 Innate Lymphoid Cells in Human Respiratory Disorders. J Innate Immun [Internet]. 2020 Jan 1 [cited 2021 Oct 28];12(1):47–62. Available from: https://www.karger.com/Article/FullText/496212. pmid:30726833
  70. 70. Niehrs A, Altfeld M. Regulation of NK-Cell Function by HLA Class II. Front Cell Infect Microbiol. 2020 Feb 18;10:55. pmid:32133304
  71. 71. Lim AI, Li Y, Lopez-Lastra S, Stadhouders R, Paul F, Casrouge A, et al. Systemic Human ILC Precursors Provide a Substrate for Tissue ILC Differentiation. Cell. 2017 Mar 9;168(6):1086–1100.e10. pmid:28283063
  72. 72. Alisjahbana A, Gao Y, Sleiers N, Evren E, Brownlie D, von Kries A, et al. CD5 Surface Expression Marks Intravascular Human Innate Lymphoid Cells That Have a Distinct Ontogeny and Migrate to the Lung. Front Immunol. 2021 Nov 18;12:4881. pmid:34867984
  73. 73. Magnusson FC, Bahhar I. Helper Innate Lymphoid Cells as Cell Therapy for Cancer. Immunology [Internet]. 2022 Oct 26 [cited 2022 Oct 28]. Available from: https://onlinelibrary.wiley.com/doi/10.1111/imm.13599. pmid:36288454
  74. 74. Laskowski TJ, Biederstädt A, Rezvani K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat Rev Cancer. 2022 2210 [Internet]. 2022 Jul 25 [cited 2022 Dec 9];22(10):557–75. Available from: https://www.nature.com/articles/s41568-022-00491-0. pmid:35879429
  75. 75. Duan S, Liu S. Targeting NK Cells for HIV-1 Treatment and Reservoir Clearance. Front Immunol. 2022 Mar 16;13:692. pmid:35371060
  76. 76. Liu S, Galat V, Galat Y4, Lee YKA, Wainwright D, Wu J. NK cell-based cancer immunotherapy: from basic biology to clinical development. J Hematol Oncol. 2021 141 [Internet]. 2021 Jan 6 [cited 2022 Dec 14];14(1):1–17. Available from: https://jhoonline.biomedcentral.com/articles/10.1186/s13045-020-01014-w.
  77. 77. Kløverpris HN, Kazer SW, Mjösberg J, Mabuka JM, Wellmann A, Ndhlovu Z, et al. Innate Lymphoid Cells Are Depleted Irreversibly during Acute HIV-Infection in the Absence of Viral Suppression. Immunity. 2016 Feb 16;44(2):391–405.
  78. 78. Singh A, Kazer SW, Roider J, Krista KC, Millar J, Asowata OE, et al. Innate Lymphoid Cell Activation and Sustained Depletion in Blood and Tissue of Children Infected with HIV from Birth Despite Antiretroviral Therapy. Cell Rep. 2020 Sep 15;32(11):108153. pmid:32937142
  79. 79. García M, Kokkinou E, Carrasco García A, Parrot T, Palma Medina LM, Maleki KT, et al. Innate lymphoid cell composition associates with COVID-19 disease severity. Clin Transl Immunol. 2020;9(12). pmid:33343897
  80. 80. Silverstein NJ, Wang Y, Manickas-Hill Z, Carbone C, Dauphin A, Boribong BP, et al. Innate lymphoid cells and COVID-19 severity in SARS-CoV-2 infection. Elife. 2022 Mar 1;11. pmid:35275061
  81. 81. Pan L, Chen X, Liu X, Qiu W, Liu Y, Jiang W, et al. Innate lymphoid cells exhibited IL-17-expressing phenotype in active tuberculosis disease. BMC Pulm Med [Internet]. 2021 Dec 1 [cited 2022 Nov 30];21(1):1–11. Available from: https://bmcpulmmed.biomedcentral.com/articles/10.1186/s12890-021-01678-1.
  82. 82. Carvelli J, Piperoglou C, Bourenne J, Farnarier C, Banzet N, Demerlé C, et al. Imbalance of Circulating Innate Lymphoid Cell Subpopulations in Patients With Septic Shock. Front Immunol [Internet]. 2019 Sep 20 [cited 2022 Dec 9];10:2179. Available from: /pmc/articles/PMC6763762/. pmid:31616411
  83. 83. Darboe A, Nielsen CM, Wolf AS, Wildfire J, Danso E, Sonko B, et al. Age-Related Dynamics of Circulating Innate Lymphoid Cells in an African Population. Front Immunol. 2020 Dec 2;11.
  84. 84. D’Souza SS, Shen X, Fung ITH, Ye L, Kuentzel M, Chittur SV, et al. Compartmentalized effects of aging on group 2 innate lymphoid cell development and function. Aging Cell. 2019 Dec 1;18(6). pmid:31429526
  85. 85. Corvino D, Kumar A, Bald T. Plasticity of NK cells in Cancer Available from: www.frontiersin.org.
  86. 86. Mucida D, Husain MM, Muroi S, Van Wijk F, Shinnakasu R, Naoe Y, et al. Transcriptional reprogramming of mature CD4 + helper T cells generates distinct MHC class II-restricted cytotoxic T lymphocytes. Nat Immunol. 2013;14(3):281–9.
  87. 87. Krabbendam L, Heesters BA, Kradolfer CMA, Haverkate NJE, Becker MAJ, Buskens CJ, et al. CD127+ CD94+ innate lymphoid cells expressing granulysin and perforin are expanded in patients with Crohn’s disease. Nat Commun [Internet]. 2021 Dec 1 [cited 2022 Nov 25];12(1). Available from: https://pubmed.ncbi.nlm.nih.gov/34615883/.
  88. 88. Ellmeier W, Seiser C. Histone deacetylase function in CD4+ T cells. Nat Rev Immunol [Internet]. 2018;18(10):617–34. pmid:30022149
  89. 89. Raghu D, Xue HH, Mielke LA. Control of Lymphocyte Fate, Infection, and Tumor Immunity by TCF-1. Trends Immunol [Internet]. 2019 Dec 1 [cited 2023 Jun 20];40(12):1149–62. Available from: http://www.cell.com/article/S1471490619302157/fulltext. pmid:31734149
  90. 90. Göschl L, Preglej T, Hamminger P, Bonelli M, Andersen L, Boucheron N, et al. A T cell-specific deletion of HDAC1 protects against experimental autoimmune encephalomyelitis. J Autoimmun. 2018 Jan 1;86:51–61. pmid:28964722
  91. 91. Göschl L, Preglej T, Boucheron N, Saferding V, Müller L, Platzer A, et al. Histone deacetylase 1 (HDAC1): A key player of T cell-mediated arthritis. J Autoimmun. 2020 Mar 1;108:102379. pmid:31883829
  92. 92. Chen Y, Wang X, Hao X, Li B, Tao W, Zhu S, et al. Ly49E separates liver ILC1s into embryo-derived and postnatal subsets with different functions. J Exp Med. 2022 May 2;219(5). pmid:35348580
  93. 93. Wu X, Kasmani MY, Zheng S, Khatun A, Chen Y, Winkler W, et al. BATF promotes group 2 innate lymphoid cell–mediated lung tissue protection during acute respiratory virus infection. Sci Immunol. 2022;7(67). pmid:35030033
  94. 94. Suffiotti M, Carmona SJ, Jandus C, Gfeller D. Identification of innate lymphoid cells in single-cell RNA-Seq data. Immunogenetics. 2017 Jul;69(7):439–50. pmid:28534222
  95. 95. Ardain A, Domingo-Gonzalez R, Das S, Kazer SW, Howard NC, Singh A, et al. Group 3 innate lymphoid cells mediate early protective immunity against tuberculosis. Nature. 2019 Jun 27;570(7762):528–32. pmid:31168092
  96. 96. Entwistle LJ, Gregory LG, Oliver RA, Branchett WJ, Puttur F, Lloyd CM. Pulmonary Group 2 Innate Lymphoid Cell Phenotype Is Context Specific: Determining the Effect of Strain, Location, and Stimuli. Front Immunol. 2020 Jan 22;10. pmid:32038635
  97. 97. Abt MC, Lewis BB, Caballero S, Xiong H, Carter RA, Susac B, et al. Innate immune defenses mediated by two ilc subsets are critical for protection against acute clostridium difficile infection. Cell Host Microbe. 2015;18(1):27–37. pmid:26159718
  98. 98. Klose CSN, Flach M, Möhle L, Rogell L, Hoyler T, Ebert K, et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell. 2014;157(2):340–56. pmid:24725403
  99. 99. Serafini N, Jarade A, Surace L, Goncalves P, Sismeiro O, Varet H, et al. Trained ILC3 responses promote intestinal defense. Science (80-). 2022;375(6583):859–63. pmid:35201883
  100. 100. Schmalzl A, Leupold T, Kreiss L, Waldner M, Schürmann S, Neurath MF, et al. Interferon regulatory factor 1 (IRF-1) promotes intestinal group 3 innate lymphoid responses during Citrobacter rodentium infection. Nat Commun. 2022;13(1):1–15.
  101. 101. Lin YD, Arora J, Diehl K, Bora SA, Cantorna MT. Vitamin D is required for ILC3 derived IL-22 and protection from Citrobacter rodentium infection. Front Immunol. 2019;10(JAN):1–10. pmid:30723466
  102. 102. Ibiza S, García-Cassani B, Ribeiro H, Carvalho T, Almeida L, Marques R, et al. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature. 2016;535(7612):440–3. pmid:27409807
  103. 103. Rankin LC, Girard-Madoux MJH, Seillet C, Mielke LA, Kerdiles Y, Fenis A, et al. Complementarity and redundancy of IL-22-producing innate lymphoid cells. Nat Immunol. 2016;17(2):179–86. pmid:26595889
  104. 104. Ahlfors H, Morrison PJ, Duarte JH, Li Y, Biro J, Tolaini M, et al. IL-22 Fate Reporter Reveals Origin and Control of IL-22 Production in Homeostasis and Infection. J Immunol. 2014;193(9):4602–13. pmid:25261485
  105. 105. Chu C, Parkhurst CN, Zhang W, Zhou L, Yano H, Arifuzzaman M, et al. The ChAT-acetylcholine pathway promotes group 2 innate lymphoid cell responses and anti-helminth immunity. Sci Immunol. 2021;6(57). pmid:33674322
  106. 106. Oherle K, Acker E, Bonfield M, Wang T, Gray J, Lang I, et al. Insulin-like Growth Factor 1 Supports a Pulmonary Niche that Promotes Type 3 Innate Lymphoid Cell Development in Newborn Lungs. Immunity. 2020;52(2):275–294.e9. pmid:32075728
  107. 107. Xiong H, Keith JW, Samilo DW, Carter RA, Leiner IM, Pamer EG. Innate Lymphocyte/Ly6Chi Monocyte Crosstalk Promotes Klebsiella Pneumoniae Clearance. Cell. 2016;165(3):679–89. pmid:27040495
  108. 108. Mear JB, Gosset P, Kipnis E, Faure E, Dessein R, Jawhara S, et al. Candida albicans airway exposure primes the lung innate immune response against Pseudomonas aeruginosa infection through innate lymphoid cell recruitment and interleukin-22-associated mucosal response. Infect Immun. 2014;82(1):306–15. pmid:24166952
  109. 109. Monticelli LA, Sonnenberg GF, Abt MC, Alenghat T, Ziegler CGK, Doering TA, et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat Immunol. 2011;12(11):1045–54. pmid:21946417
  110. 110. Vashist N, Trittel S, Ebensen T, Chambers BJ, Guzmán CA, Riese P. Influenza-activated ILC1s contribute to antiviral immunity partially influenced by differential GITR expression. Front Immunol. 2018;8(MAR):1–20. pmid:29623077
  111. 111. Chang YJ, Kim HY, Albacker LA, Baumgarth N, McKenzie ANJ, Smith DE, et al. Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat Immunol. 2011;12(7):631–8. pmid:21623379