Fact-checked by Grok 2 weeks ago

Innate lymphoid cell

Innate lymphoid cells (ILCs) are a diverse of innate immune lymphocytes that lack rearranged antigen-specific receptors, distinguishing them from adaptive T and B cells, and instead respond rapidly to environmental signals such as cytokines and alarmins to orchestrate early immune responses. Derived from common lymphoid progenitors in the or fetal liver, ILCs are predominantly tissue-resident, populating mucosal barriers, lymphoid organs, and non-lymphoid tissues where they mirror the effector functions of subsets through the production of signature cytokines. First identified in the early 2000s as rare populations with potent cytokine-secreting capabilities, ILCs have since been recognized as essential sentinels for maintaining host-microbe and mounting defenses against pathogens at barrier sites. ILCs are classified into three main groups based on their developmental pathways, transcription factors, and profiles, with natural killer (NK) cells often included as a distinct cytotoxic subset within group 1. Group 1 ILCs (ILC1s and NK cells) express the T-bet and produce interferon-γ (IFN-γ) and (TNF), enabling them to combat intracellular pathogens and tumors, particularly in the liver, intestine, and salivary glands. Group 2 ILCs (ILC2s), driven by GATA3 and RORα, secrete type 2 s such as interleukin-5 (IL-5), IL-13, and IL-4 in response to helminths, allergens, or tissue damage, promoting anti-parasitic immunity, allergic inflammation, and epithelial repair in the , gut, and . Group 3 ILCs (ILC3s), characterized by RORγt expression, produce IL-17, , and IL-23 to support antifungal and antibacterial defenses while fostering microbiota tolerance and lymphoid tissue organization, with subsets including lymphoid tissue inducer (LTi) cells active during embryogenesis. Beyond immunity, ILCs play multifaceted roles in tissue homeostasis, , and repair by integrating signals from the , neurons, and stromal cells to regulate barrier integrity and physiological processes. For instance, ILC3-derived promotes intestinal epithelial regeneration and containment of commensal bacteria, while ILC2s contribute to and through amphiregulin production. Their plasticity allows interconversion between subsets in response to environmental cues, such as the transdifferentiation of ILC3s into ILC1-like cells during , enhancing adaptive responses but also contributing to chronic diseases. Dysregulated ILC activity is implicated in a spectrum of pathologies, underscoring their therapeutic potential. In inflammatory bowel disease (IBD), aberrant ILC3 production of IL-17 and IFN-γ exacerbates gut inflammation, whereas regulatory ILC3 subsets mitigate it via IL-10. ILC2s drive type 2 inflammation in asthma and atopic dermatitis, often resisting glucocorticoid therapy, while group 1 ILCs promote insulin resistance in obesity and can contribute to tumor immunosurveillance or progression in cancer, depending on the context. Recent advances, including single-cell transcriptomics and spatial mapping, reveal tissue-specific adaptations, with ongoing research as of 2025 highlighting ILC plasticity in tumor microenvironments and spatial diversity in mucosal antiviral responses, paving the way for targeted interventions like IL-33 blockade in allergic diseases.

Discovery and History

Initial Identification

The concept of innate lymphoid cells (ILCs) built upon earlier discoveries of natural killer (NK) cells in 1975 and lymphoid tissue inducer (LTi) cells in 1997, which were later recognized as part of the ILC family. The identification of innate lymphoid cells (ILCs) occurred in the early through studies revealing cytokine-producing lymphocytes in mucosal tissues that lacked rearranged receptors, distinguishing them from adaptive T and B cells. A pivotal example came from investigations in the mouse spleen, where Thy1+ lineage-negative cells were found to produce IL-17 in response to microbial signals, representing an innate source of this cytokine independent of engagement. These cells were characterized as resident in the and capable of rapid activation, highlighting their role in early mucosal defense. Key experiments further delineated ILC populations in the late 2000s. In , studies showed that RORγt+ cells from lymphoid tissues were essential for organizing secondary lymphoid structures during development, as these cells expressed lymphoid tissue inducer-like properties without adaptive immune features. Concurrently, studies in helminth infection models identified IL-25-responsive lineage-negative cells in the intestinal mucosa that rapidly produced type 2 cytokines such as IL-13, driving expulsion of parasites through an innate pathway. These findings, using models like Nippostrongylus brasiliensis infection, showed that such cells proliferated in response to epithelial-derived IL-25, underscoring their position at barrier sites. Early descriptions of these cells faced challenges, with terms like "natural helper cells," "nuocytes," and "innate helper cells" emerging for overlapping populations based on context-specific functions.30911-5) This fragmentation reflected their diverse localization and profiles, complicating unified . Resolution came in 2013 through an international that proposed the term "innate lymphoid cells" to encompass these non-B, non-T lymphocytes, grouped by their effector functions mirroring T helper subsets but activated via innate signals. In historical context, ILCs were recognized as the "innate counterparts" to T helper cells due to their shared ability to produce signature cytokines—such as IFN-γ, IL-5/IL-13, or IL-17/—without requiring antigen-specific receptors, enabling swift responses to infections and tissue damage.30911-5) This emphasized their evolutionary conservation and foundational role in innate immunity at mucosal barriers.

Evolution of Classification

The classification of innate lymphoid cells (ILCs) underwent significant refinement starting with the 2013 consensus, which established a framework dividing them into three groups analogous to subsets based on effector production and key transcription factors. Group 1 ILCs were defined by their production of IFN-γ and dependence on T-bet, Group 2 ILCs by secretion of IL-4, IL-5, and IL-13 under the control of GATA3, and Group 3 ILCs by expression of IL-17 and regulated by RORγt. In this initial proposal, natural killer (NK) cells were positioned separately from other members due to their distinct cytotoxic capabilities, while lymphoid tissue inducer (LTi) cells were treated as a unique entity outside the core groups despite shared profiles with Group 3. Post-2013 developments, particularly around 2015, expanded the taxonomy by integrating cells into ILCs, recognizing their shared developmental origins from common lymphoid progenitors and T-bet dependency, alongside non-cytotoxic ILC1s. Similarly, LTi cells were reclassified as a specialized subset of ILCs, given their RORγt expression and role in production during early lymphoid organ formation. These updates solidified the use of dependence and signatures as primary classification criteria, enabling clearer distinctions from adaptive lymphocytes while accommodating functional parallels to Th1, Th2, and Th17 cells. By 2018, recognition of intra-group heterogeneity prompted a pivotal revision toward a five-subgroup system: cells, ILC1s, ILC2s, ILC3s, and LTi cells, emphasizing developmental trajectories over strict groupings. This framework separated cells from ILC1s to account for differences in Eomes expression and perforin/granzyme-mediated , while distinguishing LTi cells from other ILC3s due to their unique ontogenetic timing and lack of certain receptors. Reviews from that period also integrated emerging of diversity, such as ILC2s (responsive to IL-33 and TSLP for baseline ) versus inflammatory ILC2s (induced by IL-25 to produce IL-17 alongside type 2 cytokines during acute responses). Ongoing scientific debates centered on cell inclusion, with arguments for separation highlighting their circulating nature and distinct lineage commitment compared to tissue-resident ILC1s. Recent advances in single-cell sequencing have further evolved the , with 2024 studies proposing an additional ILC0 category for precursor populations that exhibit immature transcriptional profiles lacking commitment to the three main effector groups. These precursors, identified through trajectory analysis in fetal and adult tissues, bridge early lymphoid progenitors and mature ILCs, potentially resolving ambiguities in developmental classification.

Definition and Characteristics

Core Features

Innate lymphoid cells (ILCs) are a distinct family of innate immune s derived from common lymphoid progenitors (CLPs) that lack rearranged antigen-specific receptors, setting them apart from adaptive lymphocytes like T and B s. These cells originate within the lymphoid lineage but function in innate immunity, contributing to early host defense, tissue homeostasis, and repair at environmental interfaces. Unlike adaptive immune s, ILCs respond rapidly to environmental cues such as cytokines and alarmins without requiring prior sensitization or clonal expansion. Key phenotypic markers define ILCs as lineage-negative (Lin-) cells, meaning they do not express surface markers associated with T cells (e.g., CD3), B cells (e.g., ), or myeloid lineages (e.g., CD11b). Most ILC subsets express the alpha chain (IL-7Rα, or CD127), which is essential for their survival and maintenance, particularly in peripheral tissues, though natural killer () cells typically do not. Developmentally, ILCs depend on the inhibitor of DNA binding 2 (ID2), which is critical for their from progenitors and to tissue environments. Morphologically, ILCs display typical lymphoid features, including small, round nuclei with densely packed and minimal , resembling small lymphocytes under light microscopy. Some ILCs, particularly those with cytotoxic potential, exhibit granular containing effector molecules. Functionally, they are equipped for immediate effector responses, secreting cytokines and other mediators upon without the need for , which allows them to bridge innate and adaptive immunity in real time. Non-NK ILCs are predominantly tissue-resident, with high abundance in mucosal barrier sites such as the , lungs, and , where they integrate into local stromal networks to sense and respond to threats. Their frequency is notably low in circulation and secondary lymphoid organs like lymph nodes, comprising only 0.01%–0.1% of peripheral blood mononuclear s in healthy individuals, in stark contrast to the more abundant and recirculating adaptive lymphocytes, whereas cells are more abundant (5–15%) and primarily circulating. This residency underscores their as sentinels at entry points for pathogens and stressors. The following descriptions primarily pertain to human ILCs, with broadly similar features observed in mice.

Distinctions from Other Lymphocytes

Innate lymphoid cells (ILCs) are distinguished from adaptive lymphocytes, such as T and B cells, primarily by their lack of somatic recombination for antigen receptor generation. Unlike T and B cells, which undergo V(D)J recombination to produce diverse, antigen-specific T cell receptors (TCRs) and B cell receptors (BCRs), ILCs do not express these rearranged receptors and instead rely on germline-encoded pattern recognition receptors for activation. This fundamental difference results in pre-programmed effector functions for ILCs, enabling rapid cytokine production in response to environmental cues or alarmins, whereas adaptive lymphocytes require antigen-driven clonal expansion and differentiation for specificity. Consequently, ILCs contribute to early innate immunity without the capacity for long-term antigen-specific memory typical of T and B cells, although recent studies have debated the existence of memory-like responses in ILCs through epigenetic modifications or enhanced recall responses to stimuli. Compared to natural killer (NK) cells, which are now classified within the Group 1 ILC subset, conventional ILCs exhibit distinct phenotypic and functional traits. Classical NK cells are predominantly circulating lymphocytes that mediate through perforin- and granzyme-dependent mechanisms, targeting virus-infected or malignant cells via activating and inhibitory receptors. In contrast, most ILCs, including non-NK ILCs (ILC1s), are non-, tissue-resident populations that prioritize cytokine secretion, such as interferon-γ (IFN-γ), over direct killing, and they maintain long-term residence in barrier tissues like the gut and lungs. While both share a common lymphoid origin, NK cells display higher expression of transcription factors like Eomesodermin, enabling their migratory and cytotoxic profile, whereas ILCs emphasize localized and inflammation control. ILCs also differ markedly from myeloid cells in developmental lineage and primary functions. ILCs arise from common lymphoid progenitors (CLPs) in the , sharing a lymphoid and origin with T, B, and cells, whereas myeloid cells derive from common myeloid progenitors and include phagocytic populations like macrophages and dendritic cells. Functionally, ILCs focus on orchestrating immune responses through soluble cytokines rather than direct pathogen engulfment or , which are hallmarks of myeloid cells via and (MHC) pathways. This lymphoid commitment underscores ILCs' role as innate counterparts to helper T cells, bridging innate and adaptive immunity by promoting T cell priming through cytokine-mediated amplification of , without themselves forming immunological memory.

Classification

Group 1 ILCs

Group 1 innate lymphoid cells (ILCs) constitute a subset of ILCs that mirror the functions of T helper 1 (Th1) cells, primarily producing interferon-gamma (IFN-γ) to combat intracellular pathogens. These cells are characterized by their dependence on the T-bet for development and effector function, with distinct subgroups including natural killer () cells and ILC1s. Unlike other ILC groups, Group 1 ILCs are equipped for rapid responses at barrier sites and in circulation, emphasizing their role in early innate immunity. NK cells, a cytotoxic subgroup of Group 1 ILCs, are typically circulating and express both Eomesodermin (Eomes) and T-bet transcription factors. They are identified by markers such as CD56bright or CD56dim and CD3- in humans, enabling their distinction from adaptive lymphocytes. Upon activation, NK cells produce IFN-γ along with cytotoxic molecules like perforin and granzymes, facilitating direct of infected or aberrant cells. In contrast, ILC1s are non-cytotoxic, tissue-resident cells that lack Eomes expression but rely on T-bet, producing IFN-γ without significant perforin or granzyme activity. ILC1s are marked by CD49a expression, particularly in tissues like the liver and , underscoring their localized adaptation. The core effector profile of Group 1 ILCs centers on IFN-γ production, triggered by cytokines such as interleukin-12 (IL-12), IL-15, and IL-18 from infected tissues or antigen-presenting cells. This response supports basic antiviral defense by enhancing activation and inhibiting , while also aiding in anti-intracellular bacterial immunity through promotion of Th1-like environments. For instance, cells rapidly control viruses like murine via IFN-γ and , whereas ILC1s provide tissue-specific protection in mucosal sites.

Group 2 ILCs

Group 2 innate lymphoid cells (ILC2s) are defined by their expression of key transcription factors such as GATA3 and RORα, which drive their development and function, alongside surface markers including CD127 (IL-7Rα) for both mice and humans, and CRTH2 (chemoattractant receptor-homologous molecule expressed on T helper type 2 cells) predominantly in human ILC2s. In mice, additional markers like ST2 (IL-33 receptor) and IL-17RB (IL-25 receptor) are commonly used for identification, while human ILC2s often co-express CD161 and KLRG1, reflecting their lymphoid origin and activation state. These markers distinguish ILC2s from other innate lymphoid cell groups and adaptive lymphocytes, enabling their isolation via in lin⁻ CD45⁺ populations. ILC2s encompass distinct subsets, primarily natural ILC2s (nILC2s) and inflammatory ILC2s (iILC2s), which differ in their responsiveness to environmental cues. Natural ILC2s are predominantly tissue-resident, expressing high levels of ST2 and responding robustly to IL-33, as first demonstrated in early studies of and gut populations. In contrast, inflammatory ILC2s, often characterized by KLRG1ʰⁱ expression and lower ST2 levels, are recruited during inflammation and preferentially activated by IL-25 via IL-17RB, allowing rapid expansion in response to parasitic or allergic challenges. This subset distinction highlights the adaptive heterogeneity within ILC2s, with iILC2s showing migratory potential from or blood to inflamed tissues. Upon activation, ILC2s secrete a signature profile of type 2 cytokines, including IL-4, IL-5, IL-13, and IL-9, which orchestrate recruitment, production, and Th2 . Additionally, they produce , an epidermal growth factor-like molecule that promotes epithelial repair without excessive , underscoring their role in . These cytokines are rapidly induced following alarmin stimulation, with IL-5 and IL-13 being the most abundant effectors in both and models. ILC2s are primarily localized in barrier tissues such as the , gut, and , where they reside in close proximity to epithelial layers and stromal cells. Their activation is triggered by epithelial-derived alarmins, including IL-25, IL-33, and (TSLP), which bind specific receptors to initiate signaling cascades independent of recognition. For instance, IL-33 acts on ST2⁺ nILC2s in the to drive immediate release, while IL-25 targets iILC2s in the gut during helminth infection. TSLP further enhances ILC2 survival and function, particularly in and airway epithelia. ILC2s display significant heterogeneity influenced by tissue microenvironments, with variations in marker expression (e.g., up to 35% CRTH2⁻ in ILC2s) and functional states across sites like or . Notably, ILC2s exhibit higher proliferative potential than or ILCs, driven by IL-2, IL-7, and alarmin co-stimulation, enabling rapid population expansion during type 2 immune responses. This proliferative capacity, coupled with metabolic adaptations like oxidation, supports their sustained activity in chronic settings. In some contexts, ILC2s demonstrate plasticity toward phenotypes via IL-12 and IL-1β signaling.

Group 3 ILCs

Group 3 innate lymphoid cells (ILC3s) are characterized by the expression of the RORγt and the common γ-chain receptor CD127. These cells lack rearranged antigen receptors and are distinguished from adaptive lymphocytes by their innate-like responses to environmental cues. ILC3s comprise heterogeneous subsets primarily defined by the expression of natural cytotoxicity receptors (NCRs). The NCR+ subset, marked by NKp46 expression, predominantly produces high levels of with lower IL-17 output, representing about 70% of intestinal ILC3s. In contrast, the NCR- subset favors IL-17 production and is often LTi-like in phenotype. Some NCR- ILC3s can transition to an NCR+ state upon stimulation. The cytokine profile of ILC3s includes IL-17A and IL-17F, , and (GM-CSF), with certain subsets also secreting lymphotoxin to support mucosal immunity. These s enable rapid responses to microbial challenges without prior exposure. ILC3s are enriched in mucosal sites such as the intestinal and cryptopatches, as well as secondary lymphoid tissues like tonsils. They are activated by proinflammatory signals including IL-23 and IL-1β from tissue-resident myeloid cells, which drive secretion. In barrier tissues, ILC3s contribute to integrity by producing , which promotes epithelial proliferation and antimicrobial expression to prevent invasion.

Lymphoid Tissue Inducer Cells

Lymphoid tissue inducer (LTi) cells represent a specialized subset of group 3 innate lymphoid cells (ILC3s), distinguished by their pivotal role in orchestrating the development of secondary lymphoid organs. These cells are phenotypically defined by expression of the RORγt, surface markers including and IL-7Rα (CD127), absence of CD3, and high levels of lymphotoxin α1β2 (LTα1β2) on their surface. Development of LTi cells critically depends on RORγt, which drives their differentiation from common lymphoid progenitors during embryogenesis. Unlike conventional lymphocytes, LTi cells lack rearranged antigen receptors and instead rely on innate signaling for activation and function. LTi cells secrete key cytokines including IL-17, , and lymphotoxin, with their production of TNF superfamily ligands such as LTα1β2 setting them apart from other ILC3 subsets that primarily emphasize antimicrobial responses. These molecules enable LTi cells to coordinate remodeling and immune during lymphoid formation. In the embryonic context, fetal LTi cells emerge around embryonic day 9.5 in mice from hemogenic endothelium-derived progenitors and migrate to nascent lymphoid anlagen, where they initiate clustering between embryonic days 12.5 and 15.5 to drive the development of lymph nodes and Peyer's patches. This process is absent in RORγt- or LT-deficient models, underscoring the indispensability of LTi cells for these structures. In adulthood, bone marrow-derived LTi cells persist at low frequencies and play a supportive role in maintaining tertiary lymphoid structures (TLS) under conditions of chronic inflammation, , or tumorigenesis, where they promote ectopic lymphoid aggregate formation similar to their embryonic functions but on a smaller scale. Central to both phases is the interaction between LTi-derived LTα1β2 and the lymphotoxin β receptor (LTβR) on stromal organizer cells, which triggers activation and upregulation of chemokines (e.g., ) and adhesion molecules (e.g., ), thereby recruiting hematopoietic cells and facilitating vascularization and compartmentalization essential for . This bidirectional signaling loop ensures the structural integrity and functionality of lymphoid tissues across developmental stages.

Development and Ontogeny

Progenitor Cells

Innate lymphoid cells (ILCs) originate from hematopoietic stem cells (HSCs) within the , progressing through a series of committed progenitors that restrict their developmental potential to the innate lymphoid lineage, though an earlier HSC-independent wave emerges from yolk sac-derived innate lymphoid-biased multipotent . The foundational step involves HSCs differentiating into common lymphoid progenitors (CLPs), which are multipotent cells capable of giving rise to all lymphoid lineages, including B cells, T cells, and ILCs. In mice, CLPs are characterized as ⁻ IL-7Rα⁺ Flt3⁺ and serve as the immediate upstream precursor to ILC-specific progenitors. From CLPs, development proceeds to the common innate lymphoid progenitor (CILP), also known as the α-lymphoid progenitor (αLP), marked by expression of ID2 and integrin α4β7 (ID2⁺ α4β7⁺). This progenitor represents a committed stage dedicated exclusively to ILC fates, lacking potential for B or T cell differentiation. The CILP was identified in 2014 through single-cell sorting of Lin⁻ IL-7Rα⁺ c-Kit⁺ α4β7⁺ cells from mouse fetal liver and adult bone marrow, revealing high expression of PLZF (Zbtb16) as a hallmark of early commitment. These cells efficiently generate all ILC subsets in vitro and in vivo but not conventional NK cells or other adaptive lymphocytes. Downstream of the CILP, specified progenitors emerge with further lineage restriction. For helper-like ILCs (groups 1-3 excluding conventional cells), the common helper innate lymphoid progenitor (CHILP) arises, defined by markers such as Lin⁻ ID2⁺ IL-7Rα⁺ α4β7⁺ CD25⁻ Flt3⁻. Identified in 2014, the CHILP differentiates into ILC1, , and ILC3 lineages but retains separation from the pre-NK progenitor pathway, which branches earlier from CLPs to support conventional NK cell development. ILC progenitors differ in their primary sites of origin during development. In early , yolk sac-derived progenitors contribute the primitive wave, followed by fetal liver as a predominant source, where committed progenitors like CILPs generate the initial waves of tissue-resident ILCs seeding peripheral organs; the also contributes, particularly to neonatal ILC1 production and potentially other subsets from early T-cell precursors. In adults, remains a main niche, but tissue-resident progenitors, such as those in the intestinal , sustain local differentiation and replenishment of ILCs from CLPs and downstream stages. Interleukin-7 (IL-7) signaling is indispensable for the survival, proliferation, and of these progenitors across the hierarchy. IL-7Rα expression begins at the CLP stage and persists through CILP and CHILP, promoting progression to mature ILCs; its absence severely impairs ILC development in both fetal and adult contexts.

Transcriptional Regulation

The transcriptional regulation of innate lymphoid cells (ILCs) relies on a core set of transcription factors that orchestrate lineage commitment from common lymphoid progenitors. ID2 acts as an obligatory factor for all ILC subsets by inhibiting E protein transcription factors, thereby enabling the specification and development of ILCs; its absence results in a complete loss of ILCs across all groups. NFIL3 promotes early ILC commitment in a TOX-dependent manner, where it induces TOX expression to drive the of progenitors into lineage-restricted ILC precursors, with NFIL3 deficiency leading to severe reductions in all ILC populations. Additionally, TCF-1, activated via Wnt signaling, is essential for the progression of early ILC progenitors, enforcing their commitment to the ILC fate while suppressing alternative . Group-specific transcription factors further refine ILC identity and function following initial commitment. In group 1 ILCs, including natural killer cells and ILC1s, T-bet is critical for lineage specification and interferon-γ production, while Eomes supports maturation particularly in natural killer cells, with sequential actions of Eomes followed by T-bet ensuring stepwise development. For group 2 ILCs, GATA3 is indispensable for development, maintenance, and type 2 expression, cooperating with RORα to stabilize the phenotype and prevent deviation to other lineages. Group 3 ILCs and lymphoid tissue inducer cells depend on RORγt for their generation and interleukin-17/22 production, with AHR enhancing their functionality and survival in mucosal environments. The dynamics of involve sequential expression patterns and epigenetic modifications that fine-tune ILC specification. PLZF is expressed early in progenitors committed to non-natural killer ILC lineages, priming them for subsequent group-specific factors like T-bet, GATA3, or RORγt, which then dominate to enforce effector programs. Epigenetic mechanisms, such as the repression of Runx3 in through Runx/Cbfβ complex activity, prevent interference with GATA3-driven type 2 responses, allowing full activation of the ILC2 genetic program under homeostatic conditions. Recent studies highlight the role of Tox2 in maintaining group 3 ILCs, where it supports metabolic adaptations necessary for their residency and function in the gut.

Tissue Seeding and Maturation

Innate lymphoid cell (ILC) progenitors primarily seed peripheral tissues during fetal development, with the earliest contributions from yolk sac-derived cells migrating to the fetal liver, the main hematopoietic site at that stage. These progenitors, including common innate lymphoid progenitors (ILCPs), emerge around embryonic day 13.5 (E13.5) in mice and migrate via the bloodstream to colonize barrier sites such as the intestine and lymphoid organs before birth. In humans, circulating ILCPs with a type 3 signature are detectable in fetal blood and tissues from early , facilitating early establishment of tissue-resident populations. This fetal seeding ensures rapid adaptation to environmental challenges postnatally, with liver-derived ILCPs using like α4β7 and receptors such as CCR9 to home to the gut mucosa. For lymphoid tissues, homing involves CCR6 and CCR7, which guide progenitors to secondary lymphoid organs during embryogenesis. Upon arrival, ILC progenitors mature into functional, tissue-resident cells under the influence of local environmental signals, particularly tissue-derived cytokines. In the liver, IL-15 provides essential survival and maturation cues for ILC1s, promoting T-bet expression and enhancing their residency and effector functions. Similarly, in the gut, transforming growth factor-β (TGF-β) supports ILC3 differentiation by suppressing alternative pathways like Eomes and facilitating adaptation to the mucosal niche, though it is not strictly required for overall ILC3 numbers in the . These signals integrate with transcriptional programs to finalize lineage commitment, enabling ILCs to respond to tissue-specific demands. Tissue-specific heterogeneity among ILCs emerges during maturation, shaped by local and dietary factors. In the gut, the influences ILC3 diversity through metabolites that activate pathways like (AhR), enhancing production and functional specialization. Diet-derived further drives this process by inducing gut-homing receptors (e.g., CCR9 and α4β7) on ILC3s, promoting their retention and to the intestinal environment while suppressing lymphoid-homing markers like CCR7. In adulthood, mature ILCs are maintained primarily through local self-renewal within s and from tissue-resident s, with minimal contribution from circulating s under steady-state conditions. studies in mice demonstrate that over 95% of ILCs in organs like the intestine, , and liver remain host-derived even after prolonged mixing of circulatory systems, indicating robust tissue residency. This self-renewal and local activity sustain ILC pools under homeostatic conditions and during mild perturbations, though limited replenishment from blood (~5%) can occur in .

Functions in Immunity and Homeostasis

Barrier Defense Against Pathogens

Innate lymphoid cells (ILCs) play a critical role in the early innate at epithelial barriers, where they detect and orchestrate rapid defense mechanisms to prevent systemic . Positioned at mucosal surfaces such as the gut, , and , ILCs respond swiftly to microbial threats by producing signature cytokines that enhance barrier integrity and activate other immune cells. This function is particularly vital in the absence of adaptive immunity, as demonstrated in models of acute where ILC depletion leads to increased pathogen burden and mortality. Group 1 ILCs, including natural killer () cells and tissue-resident ILC1s, contribute to barrier defense primarily through the production of interferon-gamma (IFN-γ), which activates and promotes antiviral and antibacterial states in infected tissues. In murine cytomegalovirus (MCMV) infection models, cells rapidly produce IFN-γ upon recognition of infected cells via activating receptors like Ly49H, limiting viral replication in the spleen and liver during the first few days post-infection. Similarly, ILC1s in the liver and intestine produce IFN-γ in response to intracellular bacteria such as , enhancing killing capacity and restricting bacterial dissemination from the gut mucosa. These IFN-γ-mediated responses are essential for controlling early spread at barrier sites, with deficiencies in IFN-γ signaling resulting in heightened susceptibility. Group 2 ILCs (ILC2s) defend against helminth parasites at mucosal barriers by secreting type 2 cytokines, including IL-13 and IL-5, which drive hyperplasia, , and recruitment to facilitate worm expulsion. In infections with the gastrointestinal Nippostrongylus brasiliensis, ILC2-derived IL-13 is indispensable for rapid parasite clearance, as ILC2-deficient mice exhibit delayed expulsion and increased worm burden compared to wild-type controls. IL-5 from ILC2s promotes accumulation in the lung and gut, where eosinophils contribute to tissue eosinophilia and direct against larval stages, underscoring ILC2s' role in type 2 immunity at epithelial interfaces. Group 3 ILCs (ILC3s) bolster barrier defense against extracellular bacteria and fungi through and IL-17 production, which strengthen epithelial tight junctions, induce antimicrobial peptide secretion, and limit microbial translocation. During rodentium infection, a model for attaching-effacing bacterial pathogens, ILC3-derived is crucial for host survival, as it promotes crypt proliferation and mucus production to restrict bacterial colonization in the colon; IL-22-deficient mice succumb to infection with severe barrier breach. ILC3s also produce IL-17 to recruit neutrophils and enhance fungal clearance, such as against at oral and gut barriers, highlighting their non-redundant role in maintaining epithelial homeostasis during extracellular pathogen challenge. ILCs are activated at barrier sites by epithelial-derived alarmins, such as IL-25, IL-33, and , released upon sensing or tissue damage, which bind receptors on ILCs to trigger cytokine production and proliferation. This activation amplifies downstream innate responses by coordinating with s and macrophages; for instance, ILC3-derived indirectly enhances recruitment via induction from epithelial cells, while ILC1 IFN-γ primes macrophages for enhanced and production against intracellular invaders. Such interplay ensures a layered defense, where ILCs bridge epithelial sensing to broader innate immunity without requiring antigen-specific recognition.

Tissue Repair and Remodeling

Innate lymphoid cells (ILCs) contribute to tissue repair and remodeling through the production of effector cytokines that promote epithelial integrity, modulation, and interactions with stromal components, particularly in barrier tissues such as the , gut, , and liver. Group 2 ILCs (ILC2s) play a prominent role in this process by secreting (AREG) and interleukin-13 (IL-13), which drive epithelial proliferation and repair following sterile injury. In models of bleomycin-induced fibrosis, ILC2-derived AREG activates (EGFR) signaling in fibroblasts, enhancing alveolar epithelial regeneration while mitigating excessive fibrotic deposition. Similarly, in the gut, ILC2s support mucosal repair by inducing differentiation and crypt expansion via IL-13, thereby restoring barrier function after damage. Group 3 ILCs (ILC3s) facilitate intestinal homeostasis and repair primarily through , which upregulates like regenerating islet-derived 3 gamma (Reg3γ) in epithelial cells to maintain barrier integrity under steady-state conditions. This IL-22 signaling also promotes protection and epithelial regeneration during injury, independent of inflammatory contexts. ILC3s interact with stromal fibroblasts via lymphotoxin and IL-22 to organize lymphoid structures and support tissue remodeling, as evidenced by fibroblastic reticular cell niches that sustain ILC3 maintenance and effector functions in the . Group 1 ILCs (ILC1s) modulate remodeling through interferon-gamma (IFN-γ) production, which influences deposition in tissues like the liver and . In the liver, ILC1-derived IFN-γ protects against acute chemical by promoting survival and limiting fibrotic progression via STAT1-dependent pathways in non-parenchymal cells. In the intestine and , IFN-γ from ILC1s drives epithelial turnover and reorganization, contributing to controlled remodeling during resolution. Recent studies highlight ILC1 involvement in repair, where type 1 immunity, including IFN-γ, balances to favor regenerative deposition over scarring. Mechanisms underlying ILC contributions to repair involve bidirectional with stromal cells, including fibroblasts and endothelial components, which provide survival signals like IL-7 and respond to ILC cytokines to orchestrate matrix dynamics. In , ILC2s participate in obesity-associated remodeling by producing IL-13 to regulate recruitment and limit inflammatory , as shown in high-fat diet models where ILC2 activation preserves tissue architecture. These interactions ensure adaptive responses to damage, with ILC allowing subset conversion to meet remodeling demands in injured tissues.

Metabolic and Microbiota Regulation

Innate lymphoid cells (ILCs) play a pivotal role in regulating host metabolism and maintaining microbiota homeostasis, particularly through subset-specific mechanisms that respond to microbial cues and influence energy balance. Group 3 ILCs (ILC3s) are essential in the intestine, where they produce interleukin-22 (IL-22) to reinforce epithelial barrier integrity and limit bacterial translocation from the gut lumen. This IL-22 secretion is indirectly modulated by commensal and pathogenic bacteria, which stimulate ILC3 function via intermediary signals, thereby preserving microbial containment without direct pathogen engagement. Furthermore, the aryl hydrocarbon receptor (AHR) in ILC3s senses microbiota-derived metabolites, such as indole-3-aldehyde and other tryptophan catabolites, to enhance IL-22 production and support gut homeostasis. These indole derivatives, generated by commensal bacteria like Lactobacillus reuteri, act as AHR ligands to promote ILC3 activation and maintain microbial balance. Group 2 ILCs (ILC2s) contribute to metabolic regulation in by orchestrating -mediated processes that influence and energy expenditure. In , ILC2-derived IL-5 recruits and sustains , which in turn promote beiging of adipocytes—the conversion of energy-storing white fat into heat-generating beige fat—thereby counteracting fat accumulation. This axis is disrupted in models, such as high-fat diet (HFD)-fed mice, where ILC2 numbers decline, leading to reduced infiltration and impaired beiging, which exacerbates metabolic dysfunction. In the liver, group 1 ILCs (ILC1s) modulate through interferon-γ (IFN-γ) production, which influences hepatic handling and prevents excessive accumulation. Liver-resident ILC1s respond to local cues to secrete IFN-γ, thereby regulating and mitigating in metabolic contexts. Additionally, ILC3s exhibit circadian oscillations in output, driven by clock genes like Bmal1, which align microbial regulation with daily metabolic rhythms in the gut-liver axis. Recent studies (as of 2025) highlight emerging links between ILC3 dysfunction and as precursors to metabolic imbalances. For instance, short-term HFD exposure impairs ILC3 production within days, rapidly altering composition and increasing permeability, which precedes broader metabolic perturbations. Similarly, microbiota-dependent regulation of ILC3 via pathways like signaling underscores their role in sensing microbial shifts to sustain .

Plasticity and Heterogeneity

Phenotypic Switching Mechanisms

Innate lymphoid cells (ILCs) exhibit phenotypic switching, a form of where cells transition between subsets in response to environmental cues, allowing to changing demands without altering core developmental commitments. This switching is primarily driven by cytokines and epigenetic modifications that reprogram expression and states. Such mechanisms enable ILCs to shift effector functions, such as from type 2 immunity dominated by ILC2s to type 1 responses via ILC1-like phenotypes, particularly in mucosal tissues like the and gut. Cytokine signaling plays a central role in inducing to ILC1-like conversion, especially during inflammatory conditions in the . Exposure to IL-12 and IL-18 promotes downregulation of the ILC2-defining GATA3 and upregulation of T-bet, the master regulator of ILC1 identity, resulting in IFN-γ production by formerly ILC2 cells. This process requires IL-12 receptor signaling and is observed in viral infections or type 1 inflammation, where converted cells localize to inflamed areas. Similarly, ILC3 to ILC1 switching occurs in the gut, influenced by cytokine withdrawal and alternative signals. Withdrawal of IL-23, which sustains RORγt expression and ILC3 maintenance, combined with IL-15 exposure, drives downregulation of RORγt and upregulation of T-bet, yielding ex-ILC3 cells with ILC1-like features including IFN-γ secretion. This shift is prominent in human ILC3 cultured with IL-15 or IL-2, highlighting cytokine-mediated reprogramming in intestinal and inflammation.00263-0) Epigenetic modifications underpin these transitions by altering accessibility and stability. Histone modifications, such as repressive marks at plasticity-associated loci, facilitate reversible switching between subset identities, as demonstrated where ILC conversions can be bidirectionally induced by modulation. The Runx3 contributes to this process by enforcing lineage-specific epigenetic landscapes in ILC1 and ILC3, promoting stable T-bet expression while restricting alternative fates during environmental challenges. Plasticity varies by ILC subset and origin, with inflammatory ILC2 (iILC2) showing greater adaptability than natural ILC2 (nILC2). iILC2, recruited during acute via IL-25 signaling, readily undergo switching to ILC1- or ILC3-like states under type 1 or type 3 exposure, whereas nILC2 maintain tissue-resident stability with limited conversion potential. Single-cell sequencing analyses from 2022 reveal intermediate transcriptional states during these transitions, capturing cells co-expressing markers like GATA3 and T-bet, which underscore the continuum of ILC heterogeneity in mucosal sites.

Functional and Tissue-Specific Adaptations

Innate lymphoid cells (ILCs) exhibit remarkable plasticity that allows them to adapt their functions to diverse tissue microenvironments, enabling rapid responses to environmental perturbations such as dietary changes, , or . This adaptability is particularly evident in barrier tissues like the gut, lungs, , and tumors, where ILCs shift phenotypes to balance , defense, and repair. For instance, cytokines like IL-12 can briefly induce such shifts, but the resulting functional changes are context-dependent and tissue-specific. In the gut, ILC3s demonstrate plasticity by sensing dietary metabolites and microbiota-derived signals through receptors such as the (AhR), which helps maintain microbial balance during diet shifts. For example, high-salt or metabolite-altered diets can prompt ILC3s to adjust and IL-17 production, supporting epithelial integrity and preventing without full subset conversion. This adaptation enhances survival by promoting tolerance to fluctuating nutrient environments, as seen in models where AhR signaling integrates dietary cues to modulate ILC3 effector functions. Similarly, in the lungs, ILC2s can switch to a pro-inflammatory state during exacerbations, acquiring ILC3-like features such as IL-17A production in response to IL-1β and IL-18. In severe mixed granulocytic , sputum-derived intermediate ILC2s co-express CRTH2, IL-5, IL-13, c-Kit, and IL-17A, driving neutrophilic and contributing to treatment resistance. This provides adaptive benefits by amplifying type 3 responses in chronic allergic contexts, aiding clearance but exacerbating airway damage. In hypoxic tumor environments, ILC3s convert to ILC1-like cells via HIF-1α stabilization, upregulating T-bet and IFN-γ while downregulating RORγt, which improves their persistence in low-oxygen niches. This shift, observed in intestinal and tumor models, confers survival advantages by enhancing proinflammatory signaling for antitumor immunity, though it may disrupt local if prolonged. Recent studies highlight similar dynamics in , where ILC2 plasticity during integrates IL-33 signals to promote remodeling, with partial shifts toward regulatory phenotypes accelerating repair in inflammatory lesions. Despite these benefits, has limitations, including incomplete phenotypic conversion that results in cells retaining original traits, such as persistent IL-13 in ex-ILC2s. Converted ILC1s from ILC3s, for instance, produce IFN-γ but fail to acquire full cytotoxic capabilities like granzyme expression, restricting their effector potential compared to natural killer cells. These constraints underscore the role of plasticity in generating heterogeneity, which fine-tunes responses but may limit in extreme pathological settings like persistent tumors or chronic wounds.

Roles in Disease

Inflammatory and Allergic Conditions

Innate lymphoid cells (ILCs) play a pivotal role in driving inflammatory and allergic responses through production that amplifies type 2 immunity and tissue-specific . Group 2 ILCs (ILC2s) are particularly implicated in allergic airway diseases such as and , where they respond to environmental allergens by secreting IL-5 and IL-13. These s promote eosinophil recruitment and activation, leading to , while IL-13 induces hyperplasia and mucus hypersecretion, exacerbating airway obstruction. In preclinical models of (HDM)-induced allergic sensitization, ILC2 activation by epithelial-derived alarmins like IL-33 drives these IL-5- and IL-13-dependent features, linking innate responses to adaptive Th2 immunity and persistent inflammation. Group 3 ILCs (ILC3s) contribute to inflammatory conditions like by producing IL-17 in lesional tissue, promoting hyperproliferation and infiltration. In psoriatic lesions, IL-23 from dendritic cells stimulates RORγt+ ILC3s to secrete IL-17A and IL-17F, which sustain the inflammatory cascade and epidermal thickening characteristic of the disease. This IL-23/IL-17 axis in ILC3s is central to plaque formation, as evidenced by elevated ILC3 numbers and output in human psoriatic compared to healthy . Group 1 ILCs (ILC1s) exacerbate (COPD) by producing IFN-γ, particularly following viral infections that trigger acute flares. In COPD lungs, ILC2s can undergo plasticity to adopt an ILC1-like phenotype, releasing IFN-γ that activates macrophages and amplifies post-viral inflammation, contributing to tissue damage and progression. This IFN-γ-driven response heightens susceptibility to exacerbations, as seen in influenza-challenged models where ILC1-derived cytokines correlate with worsened airway . In human studies, ILC2s are elevated in the skin of patients with , correlating with disease severity and type 2 profiles that drive barrier dysfunction and pruritus. Recent data from 2023 indicate that numbers increase during pollen seasons in individuals with , associating with heightened IL-13 production and symptom exacerbation upon aeroallergen exposure.

Autoimmune and Chronic Inflammation

Innate lymphoid cells (ILCs) play a pivotal role in the of systemic autoimmune diseases and chronic conditions by dysregulating production and . In (IBD), ILC1s accumulate in the inflamed gut and secrete IFN-γ, which disrupts epithelial and vascular barriers, exacerbating and correlating with disease severity. Similarly, ILC3s contribute to barrier dysfunction through IL-17 production, promoting intestinal in models of dextran sulfate sodium-induced . These mechanisms highlight how ILC-derived perpetuate chronic mucosal damage in IBD, distinct from their homeostatic roles in regulation. In (), CNS-resident ILC1s drive in experimental autoimmune (EAE) models by producing IFN-γ and regulating Th17 responses, with T-bet-dependent ILC1s enriching in the , , and parenchyma during disease progression. This positioning enables ILC1s to amplify T cell-mediated , contributing to demyelination and underscoring their underappreciated involvement in central . ILC2s have been implicated in joint inflammation during (RA), where (AREG) secretion from synovial s activates fibroblast-like synoviocytes, enhancing their migration and promoting in arthritic models. This pro-inflammatory effect contrasts with potential regulatory functions in other contexts, illustrating ILC2 plasticity in chronic joint disease. Natural killer (NK) cells, a subset of ILC1s, exhibit functional defects in RA, including reduced cytotoxicity and altered receptor expression (e.g., lower and ), which impair immune regulation and correlate with persistent synovial inflammation. Dysregulation of ILC3s manifests in systemic (SLE), with studies reporting varying circulating ILC3 frequencies in active disease. Plasma IL-22 levels are significantly lower in new-onset SLE patients compared to healthy controls, correlating with disease activity. However, IL-22 from ILC3 has been linked to promoting inflammation in . Recent studies have linked dysregulation to chronic inflammation in post-acute sequelae of infection (), particularly persistent , where decreased frequencies alongside expanded ILC precursors sustain immune abnormalities up to 12 months post-infection in symptomatic patients. This persistence in altered states may drive ongoing type 2 imbalances, contributing to and other chronic symptoms in cohorts.

Cancer and Tumor Microenvironment

Innate lymphoid cells (ILCs) exhibit dual roles within the (TME), acting as both sentinels for anti-tumor immunity and contributors to cancer progression depending on their subset and contextual signals. ILC1s and natural (NK) cells, as part of ILCs, primarily exert protective effects through and production that curb tumor growth and . In contrast, ILC2s and ILC3s can foster pro-tumor environments by promoting , , and tissue remodeling that favor . This dichotomy is modulated by TME factors such as and inflammatory , which drive ILC toward tumor-supportive phenotypes. Understanding these dynamics is crucial for harnessing ILCs in . Group 1 ILCs, including ILC1s and cells, contribute to tumor surveillance by deploying perforin- and granzyme-mediated against malignant cells and secreting interferon-gamma (IFN-γ) to activate downstream anti-tumor responses. In models, cell-derived IFN-γ inhibits by enhancing macrophage polarization toward an anti-tumor state and suppressing (VEGF) expression in tumor cells. Similarly, ILC1s in models produce IFN-γ to regulate myeloid cell function and limit tumor progression, with depletion studies showing accelerated in their absence. These protective mechanisms highlight the potential of ILCs as early responders in solid tumors. Conversely, ILC3s promote tumor advancement in colorectal cancer through IL-22 secretion, which activates STAT3 signaling in epithelial and stromal cells to drive angiogenesis via upregulated VEGF production. In these models, ILC3-derived IL-22 sustains tumor growth by enhancing epithelial proliferation and barrier dysfunction, with blockade reducing metastasis in experimental settings. ILC2s further aid immune suppression by releasing IL-13, which recruits myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) to dampen cytotoxic responses; in lung and colorectal cancers, the IL-25/IL-33 axis amplifies this pro-tumor ILC2 activity, correlating with increased tumor burden. The TME profoundly influences ILC , shifting subsets toward pro-tumor states under hypoxic or inflammatory conditions. Hypoxia-inducible factor 1α (HIF-1α) in low-oxygen niches enhances ILC3 survival and IL-17 production, fostering an immunosuppressive milieu that supports tumor evasion. IL-23, often elevated in inflamed TMEs, drives NCR-negative ILC3s to adopt pro-tumor phenotypes, as seen in (HCC) where it promotes IL-17-mediated and recruitment. These adaptations underscore how TME cues reprogram ILC functions, potentially overriding innate anti-tumor potentials. Clinically, elevated ILC3 infiltration correlates with poor in HCC, where tumor-associated ILC3s express high and IL-17 levels, linking to advanced and reduced in patient cohorts. Recent studies indicate synergies between ILC modulation and checkpoint inhibitors; for instance, IL-33-activated ILC2s enhance PD-1 blockade efficacy in by boosting + T cell infiltration and function. In HCC, microbial interventions to rebalance ILC3 responses amplify anti-PD-1 effects, suggesting combinatorial strategies as promising avenues for 2025-era immunotherapies. As of 2025, advances in single-cell multi-omics and CAR-ILC engineering highlight potential for targeted interventions in ILC-driven cancers.

Therapeutic Implications

Targeting ILCs in Disease

Therapeutic strategies targeting innate lymphoid cells (ILCs) focus on modulating their activation, proliferation, and production to address dysregulated responses in various diseases. Inhibitors and enhancers of specific ILC subsets have entered clinical development, leveraging the cells' roles in and antitumor immunity. These approaches often target s produced by or acting on ILCs, such as IL-5 for ILC2s in allergic conditions and IL-15 for ILC1s and natural killer (NK) cells in cancer. Monoclonal antibodies (mAbs) inhibiting IL-5 have demonstrated efficacy in reducing activity in severe eosinophilic , where s drive through IL-5 and IL-13 secretion. For instance, , an anti-IL-5 mAb, attenuates proliferation and release in asthmatic patients, leading to decreased exacerbations and improved . Clinical studies show that treatment reduces expression of homing receptors like and GPR183, thereby limiting their recruitment to inflamed tissues. Similarly, in (IBD), IL-22 blockers targeting ILC3-derived are under investigation in preclinical models, as excessive signaling can exacerbate despite its protective epithelial roles; small protein antagonists of the IL-22 receptor have shown promise in suppressing in animal studies. Enhancers of ILC1 and NK cell function, particularly IL-2/IL-15 agonists, are being explored to bolster antitumor responses in cancer. NKTR-255, a pegylated IL-15 receptor agonist, expands and activates cells and CD8+ T cells, enhancing in solid tumors and lymphomas. In phase II trials for relapsed/refractory large B-cell lymphoma following CD19-directed CAR-T therapy, NKTR-255 increased complete response rates at six months post-infusion by sustaining IL-15 signaling and promoting memory T-cell formation. This approach indirectly supports ILC1 functions, as cells represent a subset of ILC1s critical for early tumor surveillance. Exploiting ILC plasticity offers additional therapeutic avenues, such as redirecting ILC3s toward antitumor phenotypes in the . Cytokine combinations involving IL-12 can induce of ILC3s into ILC1-like cells, enhancing IFN-γ production and tumor suppression independent of adaptive immunity. In preclinical tumor models, IL-12 stimulation of NKp46+ ILC3s promotes their conversion and cytotoxic activity against cancer cells. Key challenges in ILC-targeted therapies include achieving tissue specificity, as ILCs exhibit heterogeneous responses across organs due to their adaptation to local microenvironments. Non-specific modulation risks off-target effects, such as disrupting barrier immunity in the gut or lungs. Recent advancements include FDA approvals for biologics targeting in allergic diseases, such as expansions in indications for existing anti-IL-4/IL-13 agents like , which indirectly influence ILC2-driven responses, though no novel ILC-specific approvals occurred in 2023.

Emerging Research Directions

Recent investigations into microbiome interactions with innate lymphoid cells (ILCs) have elucidated how group 3 ILCs (ILC3s) sense microbial-derived short-chain fatty acids (SCFAs), such as butyrate, acetate, and propionate, which modulate their function and cytokine production to maintain gut barrier integrity. For instance, SCFAs promote ILC3-derived interleukin-22 (IL-22) secretion, enhancing epithelial protection against pathogens, as demonstrated in studies linking dietary fiber metabolism to ILC homeostasis. Emerging 2024 research further connects gut dysbiosis—characterized by reduced SCFA-producing bacteria—to neurodevelopmental disorders like autism spectrum disorder. Similarly, high-fat diets have been shown to rapidly impair ILC3 functions within 48 hours, reducing IL-22 output and highlighting dietary influences on microbiome-ILC crosstalk in metabolic contexts. In , ILC2s and ILC3s are increasingly recognized for their roles in the brain-gut axis, particularly through production that influences inflammation. A in dark rats demonstrated that heightened regulatory activity of intestinal ILC3s during disease induction correlates with reduced severity of experimental autoimmune , suggesting therapeutic potential in targeting ILC3 plasticity to mitigate relapse. These findings build on broader evidence of driving in via ILC-dependent pathways. Post-viral syndromes represent another frontier, with persistent ILC alterations observed in patients. Studies from 2024 report significant shifts in circulating ILC subsets, including increased innate lymphoid cell progenitors (ILCPs) and decreased mature s, alongside pro-inflammatory signatures that sustain symptoms like and . In severe acute , an subpopulation expressing has been identified in the lungs, contributing to type 2 immune responses and recruitment during viral persistence. Regarding emerging infections, 2025 research highlights the protective role of "trained" ILCs—epigenetically modified for enhanced responses—in conferring immunity against strains, independent of adaptive mechanisms. Advanced single-cell techniques are unveiling previously unrecognized ILC heterogeneity. Single-cell sequencing (scRNA-seq) analyses from 2024 have revealed transcriptomic diversity among ILC subsets in lymph nodes, identifying transitional states and tissue-specific imprints that challenge traditional classifications. These approaches have delineated "ILC0"-like progenitors or uncommitted subsets in peripheral blood and tissues, characterized by intermediate transcriptional profiles between classical ILC1, , and ILC3 groups. Complementing this, /Cas9-mediated editing has enabled precise modulation of transcription factors in primary ILCs; for example, 2023 protocols achieved efficient knockout in ILC2s to dissect regulatory mechanisms of production, paving the way for functional studies post-2023. Such innovations, including non-viral ribonucleoprotein delivery for mature ILC editing, are accelerating insights into ILC and plasticity in disease contexts.

References

  1. [1]
    Versatile roles of innate lymphoid cells at the mucosal barrier - Nature
    Sep 11, 2023 · Innate lymphoid cells (ILCs) are innate lymphocytes that do not express antigen-specific receptors and largely reside and self-renew in mucosal tissues.
  2. [2]
    Innate lymphoid cells: More than just immune cells - Frontiers
    Since their discovery, innate lymphoid cells (ILCs) have been described as the innate counterpart of the T cells. Indeed, ILCs and T cells share many ...Introduction · The innate lymphoid cell family · Exchange with environment...
  3. [3]
    Approaches to investigate tissue-resident innate lymphocytes ...
    Nov 30, 2024 · This review explores the current understanding of the metabolism of tissue-resident innate lymphocytes, comprehensively examines current methodologies,
  4. [4]
    The discovery of innate lymphoid cells | Nature Reviews Immunology
    130 years after the identification of lymphoid and myeloid cells, and more than 40 years after the identification of T and B ...
  5. [5]
    Lymphoid tissue inducer–like cells are an innate source of IL-17 and ...
    Dec 29, 2008 · It is well known that Stat3 is activated by IL-23 and other inducers of Th17 cell differentiation, and the absence of Stat3 abrogates IL-17 ...Missing: Thy1+ | Show results with:Thy1+
  6. [6]
    Development and regulation of RORγt+ innate lymphoid cells
    Nov 17, 2014 · RORγt + innate lymphoid cells (ILCs), or ILC3, play a fundamental role in the development of lymphoid tissues, as well as in homeostasis and defence of mucosal ...
  7. [7]
    The evolution of innate lymphoid cells - PMC - NIH
    Jun 21, 2016 · Innate lymphoid cells (ILCs) are the most recently discovered group of immune cells. Understanding their biology poses many challenges.
  8. [8]
    Innate lymphoid cells — a proposal for uniform nomenclature - Nature
    Jan 7, 2013 · Innate lymphoid cells (ILCs) are a family of developmentally related cells that are involved in immunity and in tissue development and remodelling.
  9. [9]
    Page not found | Nature
    Insufficient relevant content.
  10. [10]
  11. [11]
    Innate Lymphoid Cells: Diversity, Plasticity, and Unique Functions in ...
    Jun 19, 2018 · Type 1, 2, and 3 innate lymphoid cells (ILCs) have emerged as tissue-resident innate correlates of T helper 1 (Th1), Th2, and Th17 cells.Main Text · Acknowledgments · Declaration Of Interests
  12. [12]
    Dynamic regulation of innate lymphoid cell development during ...
    To elucidate the underlying mechanisms, we utilized single-cell RNA sequencing (scRNA-seq) to reconstruct the ILC developmental pathways, identifying a ...Missing: ILC0 | Show results with:ILC0
  13. [13]
    Human innate lymphoid cells | Blood - ASH Publications
    Jul 31, 2014 · Innate lymphoid cells (ILCs) are lymphoid cells that do not express rearranged receptors and have important effector and regulatory functions in innate ...
  14. [14]
    Innate Lymphoid Cells (ILCs): Cytokine Hubs Regulating Immunity ...
    In response, ILCs rapidly secrete effector cytokines, which allow them to survey and maintain the mucosal integrity. Uncontrolled action of ILCs might ...
  15. [15]
    Innate Lymphoid Cells: a new paradigm in immunology - PMC
    Innate lymphoid cells (ILCs) are a growing family of immune cells that mirror the phenotypes and functions of T cells.
  16. [16]
    Lymphoid Cell - an overview | ScienceDirect Topics
    The size divides lymphoid cells into small (T and B) and large granular (NK cells). ... Small, round, with usually undetectable cytoplasm and nucleoli;.
  17. [17]
    Tissue-specific features of innate lymphoid cells in antiviral defense
    Apr 29, 2024 · This review focuses on the specialized functions of each ILC subtype and their roles in antiviral immune responses across tissues.
  18. [18]
  19. [19]
  20. [20]
  21. [21]
  22. [22]
  23. [23]
  24. [24]
    Functional and phenotypic heterogeneity of group 3 innate lymphoid ...
    ILC3 consist largely of two major subsets, NCR + ILC3 and LTi‐like ILC3, but also demonstrate significant plasticity and heterogeneity.Missing: NCR+ | Show results with:NCR+
  25. [25]
    ILC3 function as a double-edged sword in inflammatory bowel ...
    Apr 8, 2019 · Group 3 innate lymphoid cells (ILC3) are responsible for gastrointestinal mucosal homeostasis through moderate generation of IL-22, IL-17 and GM ...
  26. [26]
    Subsets of ILC3-ILC1-like cells generate a diversity ... - PubMed - NIH
    In the present study we found that ILC3s and ILC1s in human tonsils represented the ends of a spectrum that included additional discrete subsets. RNA ...
  27. [27]
    Developing lymph nodes collect CD4+CD3- LTbeta+ cells ... - PubMed
    Immunity. 1997 Oct;7(4):493-504. doi: 10.1016/s1074-7613(00)80371-4. Authors. R E Mebius , P Rennert, I L Weissman. Affiliation. 1 Department of Cell Biology ...
  28. [28]
    Lymphoid Tissue inducer (LTi) cell ontogeny and functioning ... - NIH
    Lymphoid Tissue inducer (LTi) cells are an ILC member and essential for embryonic lymph node (LN) formation. LNs are formed at pre-defined and strategic ...
  29. [29]
    Lymphoid tissue inducer–like cells are an innate source of IL-17 and ...
    We report that splenic lymphoid tissue inducer–like cells (LTi-like cells) constitutively express RORγt, IL-23R, CCR6, and aryl hydrocarbon receptor (AHR) (14), ...Missing: organogenesis | Show results with:organogenesis
  30. [30]
  31. [31]
    Cellular pathways in the development of human and murine innate ...
    When purified and cultured the EILP gives rise to a more restricted progenitor known as the common helper ILC progenitor (CHILP), that is functionally distinct ...
  32. [32]
    A committed hemopoietic precursor to innate lymphoid cells - PMC
    We identified and characterized a novel subset of lymphoid precursors in fetal liver and adult bone marrow that transiently expressed high amounts of PLZF.
  33. [33]
    Polarizing influence of TCRs - Nature Immunology
    **Summary of Common Innate Lymphoid Progenitor (CILP) Discovery:**
  34. [34]
  35. [35]
  36. [36]
  37. [37]
    Delineating spatiotemporal and hierarchical development of human ...
    Jul 8, 2021 · Our study illuminates the precise cellular and molecular features underlying the stepwise formation of human fetal ILC hierarchy with remarkable spatiotemporal ...Missing: traits paper<|control11|><|separator|>
  38. [38]
    Interleukin-7 Receptor Alpha in Innate Lymphoid Cells
    In addition to the BM, IL-7 is produced in the fetal liver where earliest progenitors of ILCs can be found (72). Moreover, IL-7 is known to orchestrate the ...
  39. [39]
    The basic leucine zipper transcription factor NFIL3 directs the ... - eLife
    Oct 13, 2014 · Here we show that the transcription factor NFIL3 directs the development of a committed bone marrow precursor that differentiates into all known ILC lineages.
  40. [40]
  41. [41]
  42. [42]
    Embryonic ILC-poiesis across tissues - PMC - NIH
    Dec 20, 2022 · Tissue seeding of ILCs is initiated already during fetal development, which on the one hand facilitates balanced homeostatic conditions early in ...
  43. [43]
    Metabolic Control of Innate Lymphoid Cell Migration - Frontiers
    ILC homing to the small intestine is mediated by α4β7 integrin and the chemokine receptor CCR9 and their respective ligands MAdCAM-1 and CCL25. ILC2s in the ...<|separator|>
  44. [44]
    Gut migration of innate lymphoid cells - Nature
    Jul 21, 2015 · The researchers found that chemokine receptor (CCR)7, CCR9 and α4β7 integrin were expressed in ILCs residing in gastrointestinal tissues and the ...
  45. [45]
    ILC1: Development, maturation, and transcriptional regulation - PMC
    The bone marrow is an important site harboring developmental precursor stages of ILCs with increasingly restricted lymphoid potential [Fig. 1] [6, 31, 32].
  46. [46]
    Transforming Growth Factor-β Signaling Guides the Differentiation of ...
    May 17, 2016 · TGF-β induces SG ILC differentiation by suppressing Eomes. TGF-β acted through a JNK-dependent, Smad4-independent pathway.
  47. [47]
    The interplay between innate lymphoid cells and microbiota | mBio
    Jun 15, 2023 · ILC3s are characterized by the expression of transcription factor retinoic acid-related orphan receptor gamma subtype t (RORγt) and the ...
  48. [48]
    Retinoic acid differentially regulates the migration of innate lymphoid ...
    We report that prior to migration to the intestine ILCs first undergo a `switch' in their expression of homing receptors from lymphoid to gut homing receptors.
  49. [49]
    Tissue residency of innate lymphoid cells in lymphoid and ... - Science
    Nov 20, 2015 · Innate lymphoid cells (ILCs) are a subset of immune cells that promote barrier immunity in tissues such as the gut and lungs and help to ...Missing: classification | Show results with:classification
  50. [50]
    Innate Lymphoid Cells in Response to Intracellular Pathogens
    Here, we review recent advances in understanding the mechanisms controlling protective and pathogenic ILC responses to intracellular pathogens.
  51. [51]
    Requirement for natural killer cell-produced interferon gamma in ...
    The presence of natural killer (NK) cells contributes to early defense against murine cytomegalovirus (MCMV) infection. Although NK cells can mediate in ...Missing: seminal | Show results with:seminal
  52. [52]
    Type 1 Innate Lymphoid Cell Biology: Lessons Learnt from Natural ...
    Oct 12, 2016 · The ILC1 family, such as Th1 cells, is composed of the T-bet-expressing cells that secrete interferon (IFN)-γ. Group 2 ILCs (ILC2s), initially ...Conventional Nk Cells · Ilc1s · Ilc1 Plasticity
  53. [53]
    Systemically dispersed innate IL-13–expressing cells in type ... - PNAS
    Jun 7, 2010 · These cells expand robustly in response to exogenous IL-25 or IL-33 and after infection with the helminth Nippostrongylus brasiliensis, and they ...Missing: ILC2 seminal
  54. [54]
    ILC2s and T cells cooperate to ensure maintenance of M2 ... - Nature
    Apr 27, 2015 · Here we report that lung CD4 + T cells and Group 2 innate lymphoid cells (ILC2s) work in concert to block Nippostrongylus brasiliensis (Nb) development in the ...
  55. [55]
    Group 3 innate lymphoid cells mediate host defense against ...
    Jul 15, 2021 · ILC3 are tissue-resident lymphoid cells that help orchestrate mucosal immunity under healthy and disease conditions. •. ILC3 are activated and ...Group 3 Innate Lymphoid... · Ilc3 As Gatekeepers Of The... · Impact Of The Gut...
  56. [56]
    Targeting the Epithelium-Derived Innate Cytokines
    Feb 22, 2022 · Epithelium-derived cytokines, or alarmins, are IL-33, IL-25, and TSLP. They are produced by epithelial cells in response to threats and alert ...
  57. [57]
  58. [58]
    Unconventional immune cells in the gut mucosal barrier - Nature
    Sep 11, 2023 · Retinoic acids induce gut-homing receptors in ILC1s and ILC3s but not in ILC2s. While the regulation of ILCs by dietary retinoic acids has been ...
  59. [59]
    Innate lymphoid cells control signaling circuits to regulate tissue ...
    May 6, 2020 · IL-25 is released following helminth infections and ... cell-derived IL-25 regulates an intestinal ILC2-epithelial response circuit.
  60. [60]
    Intestinal fibroblastic reticular cell niches control innate lymphoid cell ...
    Apr 19, 2022 · Our study indicates critical fibroblastic niches within the intestinal lamina propria that control ILC homeostasis and functionality and thereby secure ...Missing: repair | Show results with:repair
  61. [61]
    Regulatory T cells in skin injury: At the crossroads of tolerance and ...
    Type 1 immunity contributes most prominently to the inflammatory stage of tissue repair by promoting antimicrobial defense at the expense of tissue regeneration ...<|separator|>
  62. [62]
    Commensal and Pathogenic Bacteria Indirectly Induce IL-22 but Not ...
    Mar 28, 2019 · These data demonstrate that human gut microbiota, including commensal bacteria, indirectly modulate colonic ILC3 function to induce IL-22.
  63. [63]
    Colonic interleukin‐22 protects intestinal mucosal barrier and ...
    Feb 9, 2022 · Colonic interleukin-22 protects intestinal mucosal barrier and microbiota abundance in severe acute pancreatitis2.3 5% Taurocholic... · 3 Results · 4 Discussion
  64. [64]
    Tryptophan Catabolites from Microbiota Engage Aryl Hydrocarbon ...
    Aug 22, 2013 · Metabolic profiling revealed that, of the different putative metabolites, indole-3-aldehyde (IAld), a molecule with AhR ligand activity (Figure ...
  65. [65]
    Interaction between Group 3 Innate Lymphoid Cells, Microbiota, and ...
    Jun 18, 2025 · In addition, intestinal microbiota metabolite indole-3-methanol acts as an AhR ligand to directly activate the AhR signaling pathway of ILC3 ...
  66. [66]
    Group 2 innate lymphoid cells promote beiging of adipose and limit ...
    To test whether ILC2s in WAT are also dysregulated in murine obesity, mice were fed a control diet (CD) or high fat diet (HFD). HFD-induced obese mice exhibited ...
  67. [67]
    Elevating adipose eosinophils in obese mice to physiologically ...
    Adipose tissue eosinophils declined with high fat diet induced weight gain. •. Recombinant interleukin 5 treatment restored adipose eosinophils during obesity.
  68. [68]
    Natural Killer Cells and Type 1 Innate Lymphoid Cells Are New ...
    Lipid accumulation in the liver contributes to hepatocyte cell death and promotes liver ... Murine liver ILC1 have the potent ability to produce cytokines (IFN-γ, ...
  69. [69]
    A circadian clock is essential for homeostasis of group 3 innate ... - NIH
    Oct 4, 2020 · In particular, group 3 ILCs (ILC3) play numerous roles in regulating intestinal health through production of IL-22, as well as their ability to ...
  70. [70]
    Transcriptional control of ILC identity - Frontiers
    In this review, we discuss recent advances in understanding transcriptional regulation of ILCs in homeostatic and inflammatory conditions. 1 Introduction.
  71. [71]
    Innate Lymphoid Cell Plasticity in Mucosal Infections - MDPI
    Type 1 inflammatory cytokines (IL-12+IL-1β) drive ILC2→ILC1 plasticity by downregulating GATA3 and upregulating T-bet. T-bet induces production of IFNγ by ILC1s ...
  72. [72]
    ILC1: Development, maturation, and transcriptional regulation
    Nov 21, 2022 · In vivo, lung infection has been shown to induce the conversion of ILC2s into ILC1s in an IL-12 and IL-18-dependent manner to upregulate anti- ...
  73. [73]
    Innate Lymphoid Cells: Diversity, Plasticity and Unique Functions in ...
    Type 1, 2 and 3 innate lymphoid cells (ILCs) have emerged as tissue resident innate correlates of T helper-1 (Th1), Th2 and Th17 cells.
  74. [74]
    Epigenetic regulation of innate lymphoid cells - Wiley Online Library
    Jun 2, 2024 · In this review, we summarize recent discoveries on epigenetic mechanisms regulating ILC development and function, and how these regulations affect disease ...Introduction · Ilc2s · Ilc3s
  75. [75]
    Runx3 specifies lineage commitment of innate lymphoid cells - PMC
    May 1, 2016 · Here we report that the transcription factor Runx3 was essential for normal development of ILC1 and ILC3, but not ILC2 cells.Missing: repression | Show results with:repression
  76. [76]
    Inflammatory group 2 innate lymphoid cells - PMC - NIH
    Distinct from homeostatic or natural ILC2 cells (nILC2 cells) that ... The plasticity of inflammatory ILC2 cells. In the CD4 lineage, naive cells ...
  77. [77]
    Heterogeneity of ILC2s in the Intestine; Homeostasis and Pathology
    ILC2s are classified into different phenotypes depending on tissue and phase of inflammation, mainly inflammatory and natural ILC2 cells.
  78. [78]
    Identification and characterization of innate lymphoid cells ...
    Nov 1, 2022 · Our study provides insights into the generation of ILCs in animals transplanted with PSC-derived iHPCs as a cell source.Missing: CILP | Show results with:CILP
  79. [79]
    The transcription factor HIF-1α mediates plasticity of NKp46+ innate ...
    Jan 13, 2022 · Loss of HIF-1α in NKp46+ cells prevents ILC3-to-ILC1 conversion, increases the expression of IL-22–inducible genes, and confers protection ...
  80. [80]
    ILC3, a Central Innate Immune Component of the Gut-Brain Axis in ...
    ILC3 can sense diet-based compounds and changes in the gut microbiota through AhR (115). AhR is highly expressed in ILC3 and is essential for the maintenance of ...
  81. [81]
  82. [82]
    A population of c-kit+ IL-17A+ ILC2s in sputum from individuals with ...
    Jan 15, 2025 · We have identified an intermediate-ILC2 phenotype in the airways of individuals with severe mixed granulocytic asthma, representing a candidate therapeutic ...Results · Airway Cytokines In Severe... · Acquisition Of Ilc3 Features...
  83. [83]
    The role of type 2 innate lymphoid cells in eosinophilic asthma - Salter
    May 7, 2019 · With respect to ILC2 plasticity, multiple studies have demonstrated that ILC2s have the capacity to undergo trans-differentiation into ILC1 or ...
  84. [84]
    Hypoxia enhances ILC3 responses through HIF-1α-dependent ...
    Jan 14, 2021 · We found increased proliferation and activation of intestinal ILC3 at low oxygen levels, a response that was phenocopied when HIF-1α was ...
  85. [85]
    Innate lymphoid cells in the spotlight: from biomarkers to blueprint ...
    Sep 2, 2025 · For example, IL-1β, IL-12 and IL-18 promote the conversion of ILC2s and ILC3s to ILC1-like cells, driven by upregulation of T-bet and Aiolos ...
  86. [86]
    Controversies and caveats in innate lymphoid cell plasticity - NIH
    Aug 16, 2022 · ILC3s are activated in response to IL-1β or IL-23 and produce IL-17 and IL-22, which are important for defence against extracellular pathogens, ...Ilc2 Plasticity In The Lungs... · Figure 2 · Ilc3 Plasticity In The Gut...
  87. [87]
  88. [88]
  89. [89]
    Epidermal Notch1 recruits RORγ+ group 3 innate lymphoid cells to ...
    Apr 21, 2016 · Group 3 or ILC3s are characterized by the expression of RORγ transcription factor and have key roles in the maintenance and repair of epithelial ...
  90. [90]
    Skin-resident innate lymphoid cells converge on a ... - PubMed - NIH
    Feb 3, 2021 · Here we show that the induction of psoriasis in mice by IL-23 or imiquimod reconfigures a spectrum of skin ILCs, which converge on a pathogenic ILC3-like state.Missing: lesions | Show results with:lesions
  91. [91]
    Increased frequencies of basophils, type 2 innate lymphoid cells and ...
    Jul 15, 2017 · Frequencies of basophils, ILC2 and Th2 but not mast cells were significantly elevated in skin, and not blood, of AD relative to psoriasis.
  92. [92]
    Immunotherapy for asthma | Cellular & Molecular Immunology - Nature
    Oct 27, 2025 · As such, grass pollen AIT was shown to suppress the increase in ILC2 numbers normally observed during the pollen season and to regulate both ...
  93. [93]
    Innate Lymphoid Cells in Inflammatory Bowel Disease - MDPI
    Innate lymphoid cells (ILCs) have recently emerged as key regulators of mucosal immunity and tissue homeostasis and are increasingly implicated in IBD.Innate Lymphoid Cells In... · 3.2. Ilc2 · 3.3. Ilc3
  94. [94]
    Innate Lymphoid Cells - Neglected Players in Multiple Sclerosis
    To assess the role of this ILC3 subtype, the authors induced EAE with MOG35-55 in the C57BL/6 mice and found an increased number of IFN-γ, GM-CSF, and IL-17 ...
  95. [95]
    Group 2 Innate Lymphoid Cells Aggravates Development of ...
    This study uncovers a new pathogenic function of ILC2s in arthritis, demonstrating that modulating ILC2s can significantly influence RA progression.
  96. [96]
    Natural Killer Cells and Their Role in Rheumatoid Arthritis: Friend or ...
    NK cells have the capacity to damage normal cells or through interaction with other cells such as dendritic cells, macrophages, and T cells cause autoimmune ...NK Cell Receptors · NK Cell Signal Transduction · Regulation of NK Cell Effector...
  97. [97]
    Innate Lymphoid Cells in Autoimmune Diseases - Frontiers
    Innate lymphoid cells (ILC) are a heterogeneous group of immune cells characterized by lymphoid morphology and cytokine profile similar to T cells.Missing: terminology | Show results with:terminology
  98. [98]
    Decreased Plasma IL‐22 Levels and Correlations with IL‐22 ...
    Dec 9, 2013 · Moreover, plasma IL-22 levels as well as peripheral Th17 and Th22 cells correlated with SLE disease activity index (SLEDAI) scores and ...
  99. [99]
    Persistent immune abnormalities discriminate post-COVID ...
    Feb 7, 2024 · Several studies detected a reduction in total circulating ILCs in severe COVID-19 patients, while relative group 2 innate lymphoid cells (ILC2) ...
  100. [100]
    Mepolizumab attenuates ILC2s proliferation and activity in adults ...
    Mepolizumab attenuates ILC2s proliferation and activity in adults with severe eosinophilic asthma.
  101. [101]
    NKTR-255, a novel polymer-conjugated rhIL-15 with potent ...
    May 17, 2021 · NKTR-255 has more sustained IL-15R engagement and prolonged NK cell increase compared with precomplexed rhIL-15 N72D/IL-15Rα Fc NK and CD8+ ...Missing: ILC1 | Show results with:ILC1
  102. [102]
    Severe asthma ILC2s demonstrate enhanced proliferation that is ...
    Apr 28, 2023 · Mepolizumab may have a more direct effect on ILC2s as it has been demonstrated that a single dose can markedly reduce serum TSLP and IL-25 in ...Abstract · INTRODUCTION · RESULTS · DISCUSSION
  103. [103]
    Severe asthma ILC2s demonstrate enhanced proliferation ... - PubMed
    Apr 28, 2023 · Both mepolizumab and omalizumab were associated with reduced ILC2s release of IL-5 and IL-13, only mepolizumab reduced IL-6. Conclusion: ILC2s ...
  104. [104]
    Human IL-22 receptor-targeted small protein antagonist suppress ...
    Oct 1, 2024 · To study a role of IL-22-mediated signaling axis during intestinal inflammation, we generated a set of small protein blockers of IL-22R1 and ...Missing: trials ILC
  105. [105]
    Nektar Therapeutics Announces NKTR-255 Following CD19 ...
    Dec 7, 2024 · The results of this study demonstrated that NKTR-255 enhanced CAR T-cell counts and increased the number of patients who achieve a CR at 6 months post infusion.
  106. [106]
    Nektar Therapeutics Announces its First Publication of Preclinical ...
    May 18, 2021 · NKTR-255 is a novel recombinant human Interleukin-15 (rhIL-15) receptor agonist designed to activate the IL-15 pathway to expand both natural ...Missing: ILC1 | Show results with:ILC1
  107. [107]
    Innate lymphoid cells and cancer: Role in tumor progression and ...
    Jun 29, 2021 · ILCs are classified into NK cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue inducer (LTi) cells [1, 7, 8]. ILC1s respond to tumors and viruses, ...Role Of Ilcs During Tumor... · Plasticity Of Ilcs And Tumor... · Ilcs And Cancer...<|control11|><|separator|>
  108. [108]
    Tissue-Specific Features of Innate Lymphoid Cells - ScienceDirect
    Innate lymphoid cells (ILCs) are predominantly tissue-resident, and accumulating data indicate that they have significant tissue-specific functions.Missing: core | Show results with:core
  109. [109]
    Emerging concepts and future challenges in innate lymphoid cell ...
    Despite the parallels between ILC and Th subsets, ILCs lack expression of antigen-specific receptors and do not directly mediate antigen-specific responses ( ...
  110. [110]
    2023 Biological License Application Approvals | FDA
    This list reflects information regarding the applications as of the approval date. It is not updated with regard to applicant or application status changes.
  111. [111]
    Immunoregulatory Sensory Circuits in Group 3 Innate Lymphoid Cell ...
    Vitamin D is required for ILC3 derived IL-22 and protection from Citrobacter rodentium infection. Front Immunol. (2019) 10:1. doi: 10.3389/fimmu.2019.00001.
  112. [112]
    ILC3, a Central Innate Immune Component of the Gut-Brain Axis in ...
    ILC3 is of special interest in MS research, as they represent the innate cell counterpart of the major pathogenic cell population in MS, ie T helper (Th)17 ...
  113. [113]
    Neurodevelopmental Disorders Associated with Gut Microbiome ...
    We reviewed recent evidence regarding the impact of the gut microbiome on early brain development, alongside its correlation with significant NDDs.
  114. [114]
    Article Acute exposure to high-fat diet impairs ILC3 functions and gut ...
    May 13, 2025 · We show that within 48 h, HFD impairs intestinal group 3 innate lymphoid cells (ILC3s) and their capacity to produce interleukin-22 (IL-22), critical for ...
  115. [115]
    Increased regulatory activity of intestinal innate lymphoid cells type 3 ...
    Jan 18, 2024 · Our results suggest that different clinical outcomes in DA rats are under determinative influence of intestinal ILC3 activity during the inductive phase of EAE.
  116. [116]
    Beyond the gut: decoding the gut–immune–brain axis in health and ...
    Aug 14, 2025 · Recent studies suggest that gut microbiota dysbiosis may drive neuroinflammation in multiple neurological disorders, highlighting the ...
  117. [117]
    Persistent immune abnormalities discriminate post-COVID ...
    We found significant changes in circulating ILC subsets, including increased ILCPs and concurrent decreased ILC2 levels. In addition, pro-inflammatory ...
  118. [118]
    Severe COVID-19 patients exhibit an ILC2 NKG2D + population in ...
    Dec 14, 2020 · The ILC2 subpopulation was shown to mediate Type 2 responses and to recruit eosinophils during viral lung infections upon the release of alarmins.
  119. [119]
    Trained ILCs confer adaptive immunity-independent protection ...
    Aug 4, 2025 · These trained cells provide broad protection against various flu strains, including dangerous bird flu variants that could cause future ...
  120. [120]
    Transcriptomic diversity of innate lymphoid cells in human lymph ...
    Jun 25, 2024 · In this study, we determined the heterogeneity of ILCs in human LNs, including abdominal and thoracic LNs, using single-cell RNA sequencing ( ...Missing: ILC0 | Show results with:ILC0
  121. [121]
    Dynamic single-cell regulomes characterize human peripheral ... - NIH
    Aug 24, 2023 · Here, we show high transcriptional and epigenetic heterogeneity among human circulating ILCs in healthy individuals.
  122. [122]
    Efficient and stable CRISPR/Cas9-mediated genome-editing ... - NIH
    Oct 5, 2023 · We detail a CRISPR/Cas9 protocol for efficient genetic knockout to aid in studying molecular mechanism regulating human ILC2s.
  123. [123]
    CRISPR-Cas9 Ribonucleoprotein-Mediated Genomic Editing in ...
    May 19, 2020 · We describe an optimized strategy for non-viral CRISPR-Cas9 ribonucleoprotein (cRNP) genomic editing of mature primary mouse innate lymphocyte cells (ILCs) and ...