Next Article in Journal
Combined Toxicological Effects of Di (2-Ethylhexyl) Phthalate and UV-B Irradiation through Endoplasmic Reticulum Stress—Tight Junction Disruption in Human HaCaT Keratinocytes
Previous Article in Journal
Role of Signaling Pathways in the Viral Life Cycle 2.0
Previous Article in Special Issue
Cell Type-Specific Induction of Inflammation-Associated Genes in Crohn’s Disease and Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

NK Cells and Other Cytotoxic Innate Lymphocytes in Colorectal Cancer Progression and Metastasis

1
Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy
2
Istituto Pasteur Italia—Fondazione Cenci-Bolognetti, Viale Regina Elena, 291, 00161 Rome, Italy
3
IRCCS Neuromed, Pozzilli, 86077 Isernia, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(14), 7859; https://doi.org/10.3390/ijms23147859
Submission received: 30 June 2022 / Revised: 12 July 2022 / Accepted: 14 July 2022 / Published: 16 July 2022

Abstract

:
Colorectal cancer (CRC) is one of the most common malignancies and leading causes of cancer-related deaths worldwide. Despite its complex pathogenesis and progression, CRC represents a well-fitting example of how the immune contexture can dictate the disease outcome. The presence of cytotoxic lymphocytes, both CD8+ T cells and natural killer (NK) cells, represents a relevant prognostic factor in CRC and is associated with a better overall survival. Together with NK cells, other innate lymphocytes, namely, innate lymphoid cells (ILCs), have been found both in biopsies of CRC patients and in murine models of intestinal cancer, playing both pro- and anti-tumor activities. In particular, several type 1 innate lymphoid cells (ILC1) with cytotoxic functions have been recently described, and evidence in mice shows a role for both NK cells and ILC1 in controlling CRC metastasis. In this review, we provide an overview of the features of NK cells and the expanding spectrum of innate lymphocytes with cytotoxic functions. We also comment on both the described and the potential roles these innate lymphocytes can play during the progression of intestinal cancer leading to metastasis. Finally, we discuss recent advances in the molecular mechanisms underlying the functional regulation of cytotoxic innate lymphocytes in CRC.

1. Introduction

Colorectal cancer (CRC) is the third most common malignancy and one of the leading causes of cancer-related deaths, responsible for almost 1 million annual deaths world-wide, according to the International Association of Cancer Registries [1]. The majority of CRC cases are associated with sporadic mutations linked to risk factors or lifestyle; 10–30% of the cases, instead, present family history, while less than 5% of patients show hereditary forms of the disease [2,3,4]. Diet, smoke, alcohol consumption, as well as chronic inflammation in patients with inflammatory bowel disease (IBD), all represent critical independent risk factors for CRC development [5,6,7,8,9]. CRC pathogenesis and progression are driven by distinct genetic features and events of genomic instability which lead to different CRC phenotypes characterized by chromosomal instability (CIN), hypermethylation of promoter CpG island sites (CpG island methylator phenotype, CIMP), and a high level of microsatellite instability (MSI-High) [3,10,11]. Despite these genetic features, CRC represents a well-fitting example of how the immune contexture can also dictate the outcome of tumor progression. Indeed, along with CRC classifications based on the features of cancer cells, including microsatellite instability and TNM staging, the WHO has introduced the “immunoscore” as a prognostic value for predicting disease-specific recurrence and survival [12,13,14,15,16]. Among the parameters, this immunoscore includes the density of tumor-infiltrating cytotoxic and memory T cells, which are associated with favorable prognosis [17]. Moreover, based on the Cancer Genome Atlas Consortium (TCGA) transcriptomic datasets, six immune subtypes of the tumor microenvironment (TME) have been identified, named wound healing, IFN-γ dominant, inflammatory, lymphocyte-depleted, immunologically quiet, and TGF-β dominant [18]. Besides TME and immune contexture, this classification also considers genetics.
By employing the same effector machinery of CD8+ cytotoxic T lymphocytes, Natural Killer (NK) cells represent the prototype of cytotoxic innate lymphocytes [19], and their frequency has been associated with a favorable outcome in CRC patients [20,21]. Together with NK cells, innate lymphoid cells (ILCs) have been also found both in tumor biopsies of CRC patients and in mouse models of CRC, playing both pro- and anti-tumor activities [22,23,24,25]. Added complexity is provided by the identification of ILCs expressing perforin and granzymes able to kill target cells, as well as by evidence in mice showing that NK cells and cytotoxic ILC can limit cancer growth [26,27,28,29,30,31,32,33,34,35]. In this review, we provide an overview of the features of NK cells and the expanding spectrum of innate lymphocytes with cytotoxic functions. We also discuss the potential roles of these cells in the progression and metastasis of intestinal cancer, as well as recent advances in the molecular mechanisms underlying NK cell functions in CRC.

2. NK Cells and the Expanding Family of Cytotoxic Innate Lymphocytes

NK cells remained the only known cell type belonging to the innate lymphoid branch until 2008, when several ILC populations able to elicit polarized effector phenotypes started to be discovered [33,36,37]. Paralleling the functions of CD8+ cytotoxic T cells and CD4+ helper T cells, innate lymphocytes are currently classified in five prototypical subsets: cytotoxic NK cells; three “helper-like” subsets, namely, ILC1, ILC2 and ILC3; and lymphoid tissue-inducer (LTi) cells, which represent a separate lineage involved in the generation of secondary lymphoid organs [38]. This nomenclature contemplates not only the functional properties, but also the distinct developmental trajectories of NK cells and ILC subsets and their different transcriptional requirements. NK cells and ILC1 were originally grouped together as type 1 ILCs because of their ability to produce Interferon (IFN)-γ as well as for their shared expression of the lineage-defining transcription factor (TF) T-bet and many surface makers, including natural cytotoxicity receptor 1 (NCR1, NKp46) and NK1.1 in C57Bl/6 mice [39,40,41,42]. This transcriptional similarity has made it difficult to specifically target ILC1 by using genetic tools in mice without affecting NK cells, except for the TF Hobit which is selectively required by liver ILC1 and not by NK cells or other known tissue-resident ILC1 [30,43,44,45]. However, unlike NK cells, ILC1 show limited ability to circulate in the peripheral blood (PB) at steady state and have been considered poor cytotoxic cells [4,41].
Advances in single-cell transcriptomic approaches have helped to refine the phenotypes and the identity of NK cells and other innate lymphocytes in humans and mice [46,47,48]. Human PB NK cells include several subsets corresponding to distinct differentiation stages and are divided, by convention, in two major subsets based on the expression of CD56, namely, CD56bright and CD56dim NK cells [49,50]. CD56dim NK cells represent around 90% of total PB NK cells and are characterized by the high expression of CD16 and a higher potential to kill tumor cells, while CD56bright NK cells express higher levels of CD94-NKG2A heterodimers and primarily produce large amounts of cytokines including IFN-γ and tumor necrosis factor (TNF)-α [51,52]. These NK cell subsets also differ for the expression of chemokine receptors and homing properties as well as for transcriptomic and epigenetic regulatory programs [47,53,54,55,56,57]. In mice, three main NK cell subsets have been identified according to the expression of CD27, a member of the TNF receptor superfamily, and the integrin chain CD11b; these cells also differ for functions and homing receptors [58,59,60,61].
NK cell activation is regulated by a dynamic balance between positive and negative signals from cell surface activating and inhibitory receptors which recognize ligands on potential target cells; their functions have been intensively reviewed and are only discussed briefly below (interested readers are referred to other outstanding reviews [62,63,64,65]). NK cells mainly receive inhibitory signals by the leukocyte antigen class I (HLA-I)-binding receptors, including killer cell Ig-like receptors (KIRs) in humans and Ly49 receptors in mice, and by CD94/NKG2A heterodimers [66,67]. KIR and Ly49 receptors bind HLA-I(A-C) and H-2K and H-2D, respectively, while CD94/NKG2A heterodimers recognize human HLA-E and mouse Qa1. These interactions transmit negative signals that limit NK cell function. This inhibition is required both to prevent NK cell responses against healthy cells as well as to ensure functional competence during development, via a process named education or licensing [68,69,70,71]. The frequent downregulation or absence of HLA-I molecules on virus-infected and neoplastic cells limit the delivery of inhibitory signals to NK cells. Moreover, during NK cell activation, other immune checkpoint receptors can be upregulated, as well as their ligands, on target cells. These include T-cell immunoglobulin and ITIM domain (TIGIT) and Programmed Cell Death Protein 1 (PD-1) and their ligands, CD155 and PDL-1/2 in virus-infected and tumor cells [66,72]. Activating signals, instead, are determined by a wide array of receptors, such as Natural Killer receptor group 2, member D (NKG2D), DNAX accessory molecule-1 (DNAM-1), natural cytotoxicity receptors (NCRs: NKp30, NKp44 and NKp46), CD94/NKG2C, and KIRs with activating intracellular domains [66,73]. A peculiarity of these receptors consists in their ability to interact with two or more ligands. For instance, NKG2D ligands include proteins major histocompatibility complex (MHC) class I-related chain A (MICA) and B (MICB) and UL16-binding proteins (ULBP1-6), while DNAM-1 ligands consist of CD155 and CD112. Tumor transformation or viral infection leads to the upregulation or neo-induction of ligands of different NK cell activating receptors. Engagement of these activating receptors by their cognate ligands promotes NK cell-mediated recognition and killing of target cells. Additionally, the low-affinity FcγRIIIA receptor (CD16) allows NK cells to exert antibody-dependent cellular cytotoxicity (ADCC) [74]. Notably, different levels of activation associated with distinct functional outcomes can be obtained by the concomitant triggering of two or more NK cell activating receptors [75]. Since virus-infected and tumor cells have evolved evasion mechanisms to reduce the expression of activating ligands (e.g., proteolytic shedding), the combined activity of distinct activating receptors could guarantee NK cell immunosurveillance.
Transcriptional and genetic approaches in mice as well as the identification of humans carrying germline mutations of selected TFs have helped to deconvolute the transcriptional requirements of NK cells [76,77,78]. Murine NK cells differ from other ILCs for the selective expression of the TF Eomesodermin (Eomes) [79,80,81], while other TFs originally considered NK cell-specific, among innate lymphocytes, are also required for other ILCs, such as T-bet, which controls ILC1 and NCR+ ILC3 [82,83,84,85,86], or Nfil3 [87,88,89,90,91,92,93,94,95,96] and Id2, which are required for the generation of the whole innate lymphoid compartment [85,97,98,99,100]. In humans, mutation of the GATA2 gene leads to altered differentiation of NK cells and higher susceptibility of opportunistic infections and cancer [101]. As well, T-BET and IRF-8 deficiency leads to defects in the differentiation and functions of human NK cells and innate-like adaptive lymphocytes [102,103,104].
Selective expression of Eomes and perforin-dependent cytotoxic functions are considered, at least in mice, two key factors to discriminate NK cells from ILC1. On the other hand, both definition of human ILC1 and identification of specific TFs have been, and still are, more problematic [105]. Several observations blur the line separating NK cells from ILC1 identities, and we will consider this issue below. Recent findings have provided evidence for the existence of ILC1 subsets expressing Eomes and/or perforin and granzymes and able to efficiently kill target cells [26]. In the intraepithelial compartment of the human and mouse intestine, a subset of ILC1 (ieILC1) expresses Eomes and can exert cytotoxic activity [28]. These cells develop in the absence of Il15ra in mice and accumulate during colitis in both humans and mice [28]. In the TME of human head and neck cancer, PB NK cells can differentiate towards cells that resemble ieILC1 endowed with potent in vivo antitumor activity [27]. In contrast, conversion of NK cells into ILC1-like cells has been reported as a mechanism of tumor immune evasion in the methylcholanthrene (MCA)-induced fibrosarcoma cell line MCA1956 mouse model [106]. Thus, further fate-tracing and functional studies are needed to discriminate between ILC1-like NK cells with anti- and pro- tumor potentials. Recently, our group [29] and others [34,44] have found murine Eomes−/lo ILC1 able to express perforin and granzyme A, B, and C and to kill target cells. Liver granzyme A+ ILC1 are sensitive to Hobit deletion [30,44] and can originate from fetal ILC1 [107] or as a product of ILC3–ILC1 plasticity [29], but not from NK cells. In humans, a population of cytotoxic ILC1 was also found in the gut in conditions of IBD; this population is characterized by the expression of CD127, CD94, granulysin, and perforin and the presence of features of both CD127+ ILC1 and CD94+ NK cells [31]. In light of these phenotypic and functional similarities as well as of NK–ILC1 plasticity occurring in both physiological and pathological conditions, further work is needed to better classify the spectrum of cytotoxic and non-cytotoxic ILC1, their mechanisms of recognition, and their functions.

3. NK Cells and Cytotoxic Innate Lymphocytes in Intestinal Cancer

The role of NK cells in CRC was initially assessed by retrospective analysis of clinical data and by employing mouse models recapitulating distinct features of human CRC [108]. A general understanding is that both a low degree of NK cell infiltration and/or impaired NK cell functions are associated with poor overall patient survival and CRC relapse after treatment [20,109,110]. Findings obtained in mice parallel these observations, since the frequency of tumor-infiltrating NK cells progressively decreases throughout tumor progression both in the hereditary and in the sporadic mouse models of CRC [111]. In these models, in vivo administration of an antibody targeting NK1.1-expressing cells vastly exacerbates tumor formation, suggesting a role for NK cells in limiting CRC growth [111]. Since anti-NK1.1 administration also depletes ILC1, a subset of ILC3 and NKT cells, genetic approaches need to be employed to discriminate the distinct roles of murine innate and innate-like populations in CRC development. In this context, a protective role of NK cells in mouse models of colitis has been shown by using Ncr1-CreEomesfl/fl mice, which selectively lack NK cells [112]; however, the molecular mechanisms underlying the increased inflammation in absence of NK cells are still poorly understood.
Comprehensive transcriptomic and high-dimensional flow cytometry studies have revealed a high degree of phenotypic complexity among innate lymphocytes in CRC [22,23,24,25,113,114,115,116]. Thus, together with NK cells, ieILC1-like cells represent the most abundant tumor-infiltrating innate lymphoid subset in CRC patients [114]. These cells express high levels of perforin and granzymes and are particularly enriched in mismatch repair-deficient CRC tumors [115], suggesting a potential anti-tumor role for these cells in CRC patients. Recently, a CRC tissue-specific ILC1-like population has been identified by single-cell RNA-seq analysis, characterized by the expression of TIGIT, CTLA4, and TNFRSF4 [113]. SLAMF1 was also found on both CRC-specific ILC1 and ILC,2 and its expression on circulating ILCs has been associated with a significantly higher survival rate in CRC patients [113]. Interestingly, circulating SLAMF1+ ILC2 are also increased in patients with Crohn’s Disease showing less active disease [117]. Thus, SLAMF1+ cells might play a protective function both during intestinal inflammation and in CRC. In the colitis-induced CRC mouse model, tumor-infiltrating ILC1 decrease over time together with their functional abilities [24,116]; however, whether these cells play a role in limiting cancer growth or progression remains to be established.
ILC1 with cytotoxic functions have been brought to the attention only recently, and many functional aspects remain to be elucidated, while more findings are available about the features of NK cells in CRC. Circulating and tumor-infiltrating NK cells in CRC patients display profound differences in terms of both receptor repertoire and effector functions, showing a drastic reduction in the expression of activating receptors, including NKG2D, NKp30, NKp46, and DNAM-1, as well as in perforin-containing lytic granules [118,119,120,121,122]. These alterations result in NK cell functional impairment in terms of IFN-γ secretion, degranulation, and cytotoxicity and are paralleled by an increased expression of the inhibitory receptors, including CD85j and NKG2A [120,122] (Figure 1A and Table 1). Moreover, NK cells from CRC patients are also characterized by an altered ADCC, showing a poor response to cetuximab treatment [123]. Different studies have shown the association between the reduced expression of NKp44 and NKp46 on circulating CD56dim NK cells in CRC patients [118,124] and the presence of NCR ligands in the tumor microenvironment [124]. These findings indicate that CD56dim and CD56bright NK cells might differently contribute to CRC progression and suggest the involvement of additional NK cell subsets in these mechanisms [125]. To this regard, several human NK cell populations characterized by intermediate phenotypes and multiple functions have been recently identified and deserve further attention [126,127,128,129,130,131,132].
To elude adaptive immune surveillance, solid tumors, including CRC, downregulate MHC-I molecules; however, specific KIR/HLA associations and CRC susceptibility to NK cell anti-tumor response remain to be elucidated [126,136,137,138]. Regarding the action of inhibitory receptors linked with NK cell dysfunction other than KIR, in several patients affected by gastrointestinal cancers, including CRC, the expression of the immune checkpoints NKG2A and TIGIT is increased on tumor-infiltrating NK cells and is correlated with impaired NK cell cytotoxicity, advanced disease stage, and poor survival [133,134,139]. Consistently, blockade of the NKG2A or TIGIT axes promotes both NK and CD8+ T cell effector functions and potentiates the anti-tumor immune response [133,134]. Similarly, NK cell-mediated antitumor activity is synergistically enhanced by blocking NKG2A along with PD-1 in mice [134]. Thus, NKG2A, TIGIT, and CD96 along with the PD-1 axis represent potent candidates for CRC immunotherapy. Finally, changes in tumor cells of the ligands triggering NK cell activating receptors could compromise NK cell functionality during tumor progression. Indeed, a significant reduction of tumor NKG2D and NCR-ligands together with a low expression of MHC class I molecules correlates not only with disease stages but also with a decrease of NK cell killing susceptibility [139,140]. Altogether, these observations support the use of therapeutic strategies to target novel check-point inhibitory mechanisms able to ensure the full activity of NK cells and preserving activating ligand expression on tumor cells.

4. NK Cells and ILC1 in CRC Liver Metastasis

CRC patients frequently develop liver metastases that often cause death [141]. The liver is enriched with immune cells and contains a high percentage of NK cells, which represent 25–30% of the total lymphocytes in humans and 15–20% in mice [142,143,144]. Although it has long been recognized that human liver NK cells are different from their circulating counterpart [145,146,147,148], detailed phenotypic analysis of these cells have been performed only recently [149,150,151,152,153,154,155,156]. Studies based on HLA-mismatched human liver transplants have allowed us to unambiguously define the features of conventional circulating and tissue-resident NK cells [154,157]. Liver-resident NK cells are characterized by unique transcription factors and surface markers expression profiles, being defined as CD56bright EomeshiT-betlo and characterized for the expression of the chemokine receptor CXCR6 and the tissue residence/activation marker CD69; in contrast, circulating NK cells can be distinguished for their EomesloT-bethi profile and for the absence of CXCR6 expression [157]. RNAseq data showed a lower expression of cytotoxic molecules, such as perforin and granzyme B, by resident NK cells compared to their circulating counterparts, associated with a lower ability to kill target cells in in vitro functional assays [154]. Liver-resident NK cells represent a long-lived population which is thought to accumulate upon the recruitment to the liver of a small subset of circulating EomesloT-bethi CXCR6+ NK cells, via the chemokine CXCL16 expressed on the surface of sinusoidal endothelial cells [154,155]. The liver microenvironment is enriched with IL-15 and TGF-β, two cytokines able to upregulate, in vitro, Eomes expression on PB NK cells along with the expression of selected adhesion molecules involved in tissue retention, including the integrins CD103 and CD49a [158]. However, it cannot be ruled out that the liver microenvironment can also promote the in situ differentiation of circulating CD34+ or NK cell precursors into liver-resident NK cells. These observations support the crucial role played by the hepatic microenvironment in shaping NK cell phenotype and functions by providing a specific cytokine and chemokine milieu [159].
Since the recent identification and characterization of liver-resident NK cells, only few data have become available in regard to their role both in homeostatic and in pathological conditions [143]. A general positive correlation between the frequency of liver metastasis-infiltrating NK cells and CRC patient’s overall survival has been reported [160]. Indeed, a retrospective study on a large cohort of CRC patients showed a 5-year overall survival for those patients with a higher frequency of metastasis-infiltrating NK cells [160]. In particular, the association of neo-adjuvant chemotherapy with an EGFR inhibitor or neutralizing antibody correlates with an increase of NK cell infiltrate [160]. Accordingly, blocking the EGFR pathway has been reported to promote the tumoral production of chemokines and cytokines able to enhance the recruitment and the activation of NK cells within the tumor [161]. These findings underline the importance of the liver microenvironment in shaping the immune response toward both anti- or pro-tumor activity by promoting alterations in NK cell phenotype, functions, and frequency. A reduced density of liver-resident CD56bright EomeshiT-betlo NK cells in colorectal liver metastasis (CLM) has also been associated with metastasis recurrence after resection [135]. Metastasis development significantly alters the liver microenvironment, as cancer cells preferentially use glycolysis rather than oxidative phosphorylation to produce energy [162]. In particular, an increased concentration of lactate was observed in CLM as a consequence of the Warburg metabolism, and this metabolite induced a strong reduction of the intracellular pH, causing mitochondrial dysfunction that led to ROS-mediated apoptosis of liver-resident NK cells [135]. The acid tumor microenvironment strongly impacted CD56brightEomeshiT-betlo NK cells with respect to circulating CD56dimEomesloT-bethi, whose frequency remained unaffected [135]. The ability of a cell to control the intracellular pH is strictly dependent on mitochondrial mass and CO2 production as a consequence of cellular respiration; in line with this observation, liver-resident-NK cells show a lower mitochondrial mass and higher ROS production with respect to CD56dim NK cells [163]. These findings highlight the importance of liver-resident NK cells in the control of tumor growth and metastasis spreading. In line with these observations, a phase I clinical study investigated the effect of in situ delivery of allogeneic NK cells combined with cetuximab and high doses of IL-2 as a possible treatment for CLM [164]. The results of this study reported the safety of allogeneic NK cell adoptive transfer and showed that better patient outcome was achieved when infused NK cells displayed higher KIR–ligand mismatches.
Evidence supporting a role for innate lymphocytes in the control of CLM has been obtained also in mouse models mainly based on intrasplenic injection of tumor cells derived from a murine primary colon carcinoma (MC38) [165]. The mouse liver contains at steady state two main NKp46+ populations: NK cells and ILC1 [90,166,167]. Liver NK cells are defined as CD49aCD49b+ cells [79,168], while liver ILC1 express CD49a, TRAIL, CD69, CXCR6, and the inhibitory receptor CD200R1, but lack CD49b [43,80,169]. Moreover, liver ILC1 are very heterogeneous and include populations that differ for phenotypic and functional properties. Our group recently identified functionally distinct ILC1 populations defined by the expression of granzyme A (GzmA) and CD160 [29]. These populations display a differential ability to mediate granule-dependent killing and produce cytokines. Compared with other ILC1 populations, GzmA+CD160 ILC1 are more cytotoxic and produce less IFN-γ when stimulated with cytokines [29].
Both NK cells and ILC1 play a role in controlling hepatic metastasis; indeed, conditional deletion of NK cells (by using Ncr1iCreEomesfl/fl mice) or ILC1 (granzyme A (GzmA) Hobit−/−, Ncr1Cre/+Rorα−/− mice) led to an increased MC38-derived liver metastatic load, thus indicating that both populations are needed to restrict liver CRC metastasization [33,170]. However, it has been proposed that NK cells and ILC1 control different steps of this process: while ILC1 interfere with the seeding of cancer cells, NK cells prevent metastatic outgrowth and progression. A differential distribution of these populations may account for these distinct functions. Indeed, ILC1s are mainly localized in the hepatic sinusoids and outside metastatic nodules, whereas NK cells can infiltrate MC38 metastases [33]. However, in advanced metastatic disease, ILC1 are still functional and able to kill tumor cells, while NK cells lose their cytotoxic ability. The metastatic microenvironment deeply affects the liver NK cell phenotype, leading to the generation of novel NK cell subsets. Notably, NK cells appear differently re-modelled in metastasis derived from colon or lung cancer cells, suggesting that tumor-specific features play a key role in these mechanisms. In particular, MC38-induced metastases concur to generate a unique NK cell subset expressing ILC1 markers, such as Thy1, CD69, CD27, and CD49a. This subset, defined as Eomes+CD49a+CD49b+, is supposed to arise from NK cells in the presence of TGF-β and IL-15. Consistently, this NK cell population is enriched in genes typically associated with TGF-β and IL-15 signaling [33] and appears in metastatic livers in Hobit−/− but not in Ncr1iCreEomesfl/fl or Ncr1iCreTgfbr2fl/fl mice. These findings confirmed the relevance of the tumor microenvironment in shaping the phenotype and function of immune effector cells.
The anti-metastatic efficacy of liver NK cells is regulated by the inflammasome pathway. Mice lacking key inflammasome components, such as caspase 1 or caspase 11 (Ice−/− or Nlrp3 (Nlrp3−/− ), have exacerbated CLM after MC38 intrasplenic injection, which supports the requirement of this pathway to suppress CRC growth in this organ [171]. In particular, IL-18 plays a critical role in inflammasome-mediated immune surveillance against metastasis. Indeed, ablation of IL-18 signaling in IL-18−/− and Il18r1−/− mice also resulted in higher tumor burden in the liver, and consistently recombinant IL-18 was able to reduce metastasis growth in Ice/– mice. The cytokine operates independently of the adaptive immune system, by promoting the anti-tumor activity of NK cells. Indeed, depletion of NK cells via anti-asialo GM1 antibodies blocks the anti-metastatic effect of IL-18, while specific loss of IL18r on NK cells recapitulates the impairment of immune surveillance found in inflammasome-deficient mice. IL-18 promotes NK cell maturation because the frequencies and numbers of fully mature NK cells (CD11bhi CD27lo) significantly decreases in IL-18−/− mice, but it also regulates FasL surface expression on NK cells, thus impacting on their cytotoxic activity [171].
The anti-metastatic role of the IL-18 pathway and NK cells in the liver is also controlled by IL-1R8, a member of the IL-1R family serving as a checkpoint for NK cell maturation and effector functions [172]. Il1r8−/− mice exhibit protection against MC38 liver metastasis, but IL-18 genetic deficiency abrogates this effect, thus indicating a role for IL-1R8 in promoting the metastasization process via blocking inflammasome-mediated immune surveillance. In particular, adoptive transfer of Il1r8−/− NK cells significantly reduced liver metastatic load, demonstrating a direct contribution for liver IL-1R8+ NK cells in these mechanisms.
The antimetastatic function of NK cells is not restricted to the liver but was also demonstrated for pulmonary CRC metastases. T-bet-deficient mice lack fully functional peripheral CD27loKLRG1+ NK cells and are more highly susceptible to lung CRC metastases, suggesting that terminally developed NK cells are required for controlling the metastatic disease [173]. Accordingly, the adoptive transfer of wild-type CD27loKLRG1+ NK cells succeeded in partially protecting Tbx21−/− mice from metastasis growth. Moreover, Prf1−/− but not Ifng−/− mice developed pulmonary CRC metastases in a way comparable to T-bet-deficient mice, indicating that cytotoxic activity is key for the protective function of NK cells.
Although very few and limited human and murine studies indicate a role for NK/ILC1 in the control of CRC metastasis (Figure 1B and Table 1); however, it remains poorly understood how their activity is impaired thus promoting cancer metastasization.

5. Concluding Remarks

It is now increasingly clear that the immune signatures associated with cancer have profound implications in the clinical development and outcome of this disease. In this context, the immune infiltrate represents a relevant prognostic factor in CRC, in which the presence of cytotoxic lymphocytes, both CD8+ T cell and NK cells, is associated with a better overall survival. Although NK cells and other cytotoxic innate lymphocytes are found at lower numbers than T cells in CRC and CLM specimens, their effector phenotypes should not be overlooked and rather have the potential to be employed to design novel targeted therapies and to define new biomarkers. Indeed, taking advantage of the fact that specific NK populations or ILC subsets might be found at a low level in the PB of healthy donors, these cells can represent a highly sensitive biomarker for tissue alterations/disruption if are released into the circulation, as discussed above for the expression of SLAMF1 in ILCs.
Another important aspect is that, among innate lymphocytes, ieILC1 and NK cells are the main CRC-infiltrating cells. Several findings suggest that NK cells and ieILC1 regulate different steps of CRC tumorigenesis. With the rapid advances of spatial transcriptomic approaches, it will be possible to decipher the relative positioning of these two populations in the tumor environment, as well as their activation signatures, phenotypes, and functional abilities, which will affect future choices for therapeutic strategies. This approach will also lead to the identification of novel ILC subsets with cytotoxic potential which could play a role in the immune response against CRC. Thus, understanding the complexity of cytotoxic innate lymphocytes may provide not only further prognostic tools but also wider options for the treatment of CRC and CLM and the design of specific therapies. Indeed, being localized in the tissues, these cells have the potential not only to eliminate altered cells at an early stage of transformation but also to limit tissue invasion by cancer cells. In addition, the comprehension of the complex mechanisms impacting on the anti-metastatic potential of these lymphocytes will be key to delineate strategies to contrast metastasis growth and spreading.

Author Contributions

C.F. wrote the manuscript and prepared the figure and table. H.S., G.S. (Gianluca Scarno), C.D.C., R.M. and R.P. participated in the investigation, writing, and editing of the manuscript. A.G., S.S. and A.S. critically revised the manuscript. G.S. (Giuseppe Sciumè) designed, wrote, and edited the manuscript. The final manuscript was a result of the joint efforts of all the authors. All authors have read and agreed to the published version of the manuscript.

Funding

A.G. was supported by PRIN [20174T7NXL_003]. G.S. (Giuseppe Sciumè), R.P., S.S., and A.S. were supported by the Italian Association for Cancer Research (AIRC), [MFAG-21311], [AIRC IG-24955], [AIRC IG-20776] and [5x1000-21147], respectively, and by Institut Pasteur (France), Transversal Research Program, [PTR-113-17] program. G.S. (Giuseppe Sciumè) and R.P. were supported by Istituto Pasteur Italia-Fondazione Cenci Bolognetti [Under 45-213], [2020-366], respectively. The APC was funded by AIRC [MFAG-21311].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any conflict of interest.

Abbreviations

Azoxymethane (AOM); colorectal cancer (CRC); dextran sodium sulfate (DSS); inflammatory bowel disease (IBD); innate lymphoid cells (ILCs); interferon (IFN); interleukin (IL); intraepithelial (ie); lymphoid tissue-inducer (LTi); natural killer (NK); natural cytotoxicity receptors (NCRs); T helper (Th); transcription factor (TF); tumor necrosis factor (TNF); Tumor microenvironment (TME); Colorectal liver metastasis (CLM); T-cell immunoglobulin and ITIM domain (TIGIT); Programmed Cell Death Protein 1 (PD-1); T-cell immunoglobulin mucin-3 (Tim3); Natural-Killer receptor group 2, member D (NKG2D); DNAX accessory molecule-1 (DNAM-1); antibody-dependent cellular cytotoxicity (ADCC); major histocompatibility complex (MHC) class I-related chain A (MICA) and B (MICB); UL16-binding proteins (ULBP1-6); killer cell Ig-like receptors (KIRs); granzyme A (GzmA); leukocyte antigen class I (HLA-I).

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Kanth, P.; Grimmett, J.; Champine, M.; Burt, R.; Samadder, J.N. Hereditary Colorectal Polyposis and Cancer Syndromes: A Primer on Diagnosis and Management. Am. J. Gastroenterol. 2017, 112, 1509–1525. [Google Scholar] [CrossRef]
  3. Fearon, E.R.; Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 1990, 61, 759–767. [Google Scholar] [CrossRef]
  4. Fidelle, M.; Yonekura, S.; Picard, M.; Cogdill, A.; Hollebecque, A.; Roberti, M.P.; Zitvogel, L. Resolving the Paradox of Colon Cancer Through the Integration of Genetics, Immunology, and the Microbiota. Front. Immunol. 2020, 11, 600886. [Google Scholar] [CrossRef] [PubMed]
  5. Dashti, S.G.; Buchanan, D.D.; Jayasekara, H.; Ait Ouakrim, D.; Clendenning, M.; Rosty, C.; Winship, I.M.; Macrae, F.A.; Giles, G.G.; Parry, S.; et al. Alcohol Consumption and the Risk of Colorectal Cancer for Mismatch Repair Gene Mutation Carriers. Cancer Epidemiol. Biomark. Prev. 2017, 26, 366–375. [Google Scholar] [CrossRef] [Green Version]
  6. Murphy, N.; Ward, H.A.; Jenab, M.; Rothwell, J.A.; Boutron-Ruault, M.-C.; Carbonnel, F.; Kvaskoff, M.; Kaaks, R.; Kühn, T.; Boeing, H.; et al. Heterogeneity of Colorectal Cancer Risk Factors by Anatomical Subsite in 10 European Countries: A Multinational Cohort Study. Clin. Gastroenterol. Hepatol. 2018, 17, 1323–1331.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Chan, A.T.; Giovannucci, E.L. Primary Prevention of Colorectal Cancer. Gastroenterology 2010, 138, 2029–2043.e10. [Google Scholar] [CrossRef] [Green Version]
  8. Dahm, C.C.; Keogh, R.H.; Spencer, E.A.; Greenwood, D.C.; Key, T.J.; Fentiman, I.S.; Shipley, M.J.; Brunner, E.J.; Cade, J.E.; Burley, V.J.; et al. Dietary Fiber and Colorectal Cancer Risk: A Nested Case-Control Study Using Food Diaries. J. Natl. Cancer Inst. 2010, 102, 614–626. [Google Scholar] [CrossRef] [Green Version]
  9. Lin, Y.-C.; Lin, Y.-C.; Chen, C.-J. Cancers Complicating Inflammatory Bowel Disease. N. Engl. J. Med. 2015, 373, 194–195. [Google Scholar] [CrossRef]
  10. Boland, C.R.; Goel, A. Microsatellite instability in colorectal cancer. Gastroenterology 2010, 138, 2073–2087.e3. [Google Scholar] [CrossRef]
  11. Advani, S.M.; Advani, P.S.; Brown, D.W.; DeSantis, S.M.; Korphaisarn, K.; VonVille, H.; Bressler, J.; Lopez, D.S.; Davis, J.S.; Daniel, C.R.; et al. Global differences in the prevalence of the CpG island methylator phenotype of colorectal cancer. BMC Cancer 2019, 19, 964. [Google Scholar] [CrossRef] [PubMed]
  12. Mlecnik, B.; Bindea, G.; Angell, H.K.; Maby, P.; Angelova, M.; Tougeron, D.; Church, S.E.; Lafontaine, L.; Fischer, M.; Fredriksen, T.; et al. Integrative Analyses of Colorectal Cancer Show Immunoscore Is a Stronger Predictor of Patient Survival Than Microsatellite Instability. Immunity 2016, 44, 698–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ascierto, P.A.; Marincola, F.M.; Fox, B.A.; Galon, J. No time to die: The consensus immunoscore for predicting survival and response to chemotherapy of locally advanced colon cancer patients in a multicenter international study. OncoImmunology 2020, 9, 1826132. [Google Scholar] [CrossRef] [PubMed]
  14. Lanzi, A.; Pagès, F.; Lagorce-Pagès, C.; Galon, J. The consensus immunoscore: Toward a new classification of colorectal cancer. OncoImmunology 2020, 9, 1789032. [Google Scholar] [CrossRef]
  15. Nagtegaal, I.D.; Odze, R.D.; Klimstra, D.; Paradis, V.; Rugge, M.; Schirmacher, P.; Washington, K.M.; Carneiro, F.; Cree, I.A.; the WHO Classification of Tumours Editorial Board. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020, 76, 182–188. [Google Scholar] [CrossRef] [Green Version]
  16. Quezada-Marín, J.I.; Lam, A.K.; Ochiai, A.; Odze, R.D.; Washington, K.M.; Fukayama, M.; Rugge, M.; Klimstra, D.S.; Nagtegaal, I.D.; Tan, P.-H.; et al. Gastrointestinal tissue-based molecular biomarkers: A practical categorisation based on the 2019 World Health Organization classification of epithelial digestive tumours. Histopathology 2020, 77, 340–350. [Google Scholar] [CrossRef]
  17. Galon, J.; Costes, A.; Sanchez-Cabo, F.; Kirilovsky, A.; Mlecnik, B.; Lagorce-Pagès, C.; Tosolini, M.; Camus, M.; Berger, A.; Wind, P.; et al. Type, Density, and Location of Immune Cells Within Human Colorectal Tumors Predict Clinical Outcome. Science 2006, 313, 1960–1964. [Google Scholar] [CrossRef] [Green Version]
  18. Thorsson, V.; Gibbs, D.L.; Brown, S.D.; Wolf, D.; Bortone, D.S.; Ou Yang, T.-H.; Porta-Pardo, E.; Gao, G.F.; Plaisier, C.L.; Eddy, J.A.; et al. The Immune Landscape of Cancer. Immunity 2018, 48, 812–830.e14. [Google Scholar] [CrossRef] [Green Version]
  19. Sun, J.C.; Lanier, L.L. NK cell development, homeostasis and function: Parallels with CD8+ T cells. Nat. Rev. Immunol. 2011, 11, 645–657. [Google Scholar] [CrossRef]
  20. Coca, S.; Perez-Piqueras, J.; Martinez, D.; Colmenarejo, A.; Saez, M.A.; Vallejo, C.; Martos, J.A.; Moreno, M. The prognostic significance of intratumoral natural killer cells in patients with colerectal carcinoma. Cancer 1997, 79, 2320–2328. [Google Scholar] [CrossRef]
  21. Zhong, F.; Lin, Y.; Jing, X.; Ye, Y.; Wang, S.; Shen, Z. Innate tumor killers in colorectal cancer. Cancer Lett. 2021, 527, 115–126. [Google Scholar] [CrossRef] [PubMed]
  22. Atreya, I.; Kindermann, M.; Wirtz, S. Innate lymphoid cells in intestinal cancer development. Semin. Immunol. 2019, 41, 101267. [Google Scholar] [CrossRef] [PubMed]
  23. Pietropaolo, G.; Scarno, G.; Stabile, H.; Grimaldi, A.; Gismondi, A.; Santoni, A.; Sciumè, G. NK cell and ILC heterogeneity in colorectal cancer. New perspectives from high dimensional data. Mol. Asp. Med. 2021, 80, 100967. [Google Scholar] [CrossRef] [PubMed]
  24. Marchalot, A.; Mjösberg, J. Innate lymphoid cells in colorectal cancer. Scand. J. Immunol. 2022, 95, e13156. [Google Scholar] [CrossRef]
  25. Huang, Q.; Cao, W.; Mielke, L.; Seillet, C.; Belz, G.T.; Jacquelot, N. Innate Lymphoid Cells in Colorectal Cancers: A Double-Edged Sword. Front. Immunol. 2020, 10, 3080. [Google Scholar] [CrossRef] [Green Version]
  26. Cella, M.; Robinette, M.L. Intraepithelial ILC1-like cells: Front-line fighters in human head and neck squamous cell carcinoma. Proc. Natl. Acad. Sci. USA 2021, 118, e21108751. [Google Scholar] [CrossRef]
  27. Moreno-Nieves, U.Y.; Tay, J.K.; Saumyaa, S.; Horowitz, N.B.; Shin, J.H.; Mohammad, I.A.; Luca, B.; Mundy, D.C.; Gulati, G.S.; Bedi, N.; et al. Landscape of innate lymphoid cells in human head and neck cancer reveals divergent NK cell states in the tumor microenvironment. Proc. Natl. Acad. Sci. USA 2021, 118, e2101169118. [Google Scholar] [CrossRef]
  28. Fuchs, A.; Vermi, W.; Lee, J.S.; Lonardi, S.; Gilfillan, S.; Newberry, R.D.; Cella, M.; Colonna, M. Intraepithelial Type 1 Innate Lymphoid Cells Are a Unique Subset of IL-12- and IL-15-Responsive IFN-γ-Producing Cells. Immunity 2013, 38, 769–781. [Google Scholar] [CrossRef] [Green Version]
  29. Di Censo, C.; Marotel, M.; Mattiola, I.; Müller, L.; Scarno, G.; Pietropaolo, G.; Peruzzi, G.; Laffranchi, M.; Mazej, J.; Hasim, M.S.; et al. Granzyme A and CD160 expression delineates ILC1 with graded functions in the mouse liver. Eur. J. Immunol. 2021, 51, 2568–2575. [Google Scholar] [CrossRef]
  30. Yomogida, K.; Bigley, T.M.; Trsan, T.; Gilfillan, S.; Cella, M.; Yokoyama, W.M.; Egawa, T.; Colonna, M. Hobit confers tissue-dependent programs to type 1 innate lymphoid cells. Proc. Natl. Acad. Sci. USA 2021, 118, e21179651. [Google Scholar] [CrossRef]
  31. Krabbendam, L.; Heesters, B.A.; Kradolfer, C.M.A.; Haverkate, N.J.E.; Becker, M.A.J.; Buskens, C.J.; Bemelman, W.A.; Bernink, J.H.; Spits, H. CD127+ CD94+ innate lymphoid cells expressing granulysin and perforin are expanded in patients with Crohn’s disease. Nat. Commun. 2021, 12, 5841. [Google Scholar] [CrossRef] [PubMed]
  32. Krabbendam, L.; Heesters, B.A.; Kradolfer, C.M.; Spits, H.; Bernink, J.H. Identification of human cytotoxic ILC3s. Eur. J. Immunol. 2020, 51, 811–823. [Google Scholar] [CrossRef] [PubMed]
  33. Ducimetière, L.; Lucchiari, G.; Litscher, G.; Nater, M.; Heeb, L.; Nuñez, N.G.; Wyss, L.; Burri, D.; Vermeer, M.; Gschwend, J.; et al. Conventional NK cells and tissue-resident ILC1s join forces to control liver metastasis. Proc. Natl. Acad. Sci. USA 2021, 118, e2026271118. [Google Scholar] [CrossRef] [PubMed]
  34. Nixon, B.G.; Chou, C.; Krishna, C.; Dadi, S.; Michel, A.O.; Cornish, A.E.; Kansler, E.R.; Do, M.H.; Wang, X.; Capistrano, K.J.; et al. Cytotoxic granzyme C–expressing ILC1s contribute to antitumor immunity and neonatal autoimmunity. Sci. Immunol. 2022, 7, eabi8642. [Google Scholar] [CrossRef]
  35. Kansler, E.R.; Dadi, S.; Krishna, C.; Nixon, B.G.; Stamatiades, E.G.; Liu, M.; Kuo, F.; Zhang, J.; Zhang, X.; Capistrano, K.; et al. Cytotoxic innate lymphoid cells sense cancer cell-expressed interleukin-15 to suppress human and murine malignancies. Nat. Immunol. 2022, 23, 904–915. [Google Scholar] [CrossRef]
  36. Spits, H.; Di Santo, J. The expanding family of innate lymphoid cells: Regulators and effectors of immunity and tissue remodeling. Nat. Immunol. 2010, 12, 21–27. [Google Scholar] [CrossRef]
  37. Walker, J.A.; Barlow, J.L.; McKenzie, A.N.J. Innate lymphoid cells—How did we miss them? Nat. Rev. Immunol. 2013, 13, 75–87. [Google Scholar] [CrossRef]
  38. Vivier, E.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate Lymphoid Cells: 10 Years On. Cell 2018, 174, 1054–1066. [Google Scholar] [CrossRef] [Green Version]
  39. Spits, H.; Bernink, H.S.J.H.; Lanier, L. NK cells and type 1 innate lymphoid cells: Partners in host defense. Nat. Immunol. 2016, 17, 758–764. [Google Scholar] [CrossRef]
  40. Cortez, V.S.; Colonna, M. Diversity and function of group 1 innate lymphoid cells. Immunol. Lett. 2016, 179, 19–24. [Google Scholar] [CrossRef] [Green Version]
  41. Seillet, C.; Brossay, L.; Vivier, E. Natural killers or ILC1s? That is the question. Curr. Opin. Immunol. 2020, 68, 48–53. [Google Scholar] [CrossRef] [PubMed]
  42. Spits, H.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate lymphoid cells—A proposal for uniform nomenclature. Nat. Rev. Immunol. 2013, 13, 145–149. [Google Scholar] [CrossRef] [PubMed]
  43. Mackay, L.K.; Minnich, M.; Kragten, N.A.M.; Liao, Y.; Nota, B.; Seillet, C.; Zaid, A.; Man, K.; Preston, S.; Freestone, D.; et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 2016, 352, 459–463. [Google Scholar] [CrossRef] [Green Version]
  44. Friedrich, C.; Taggenbrock, R.L.R.E.; Doucet-Ladevèze, R.; Golda, G.; Moenius, R.; Arampatzi, P.; Kragten, N.A.M.; Kreymborg, K.; de Agüero, M.G.; Kastenmüller, W.; et al. Effector differentiation downstream of lineage commitment in ILC1s is driven by Hobit across tissues. Nat. Immunol. 2021, 22, 1256–1267. [Google Scholar] [CrossRef]
  45. Weizman, O.-E.; Adams, N.M.; Schuster, I.S.; Krishna, C.; Pritykin, Y.; Lau, C.; Degli-Esposti, M.A.; Leslie, C.S.; Sun, J.C.; O’Sullivan, T.E. ILC1 Confer Early Host Protection at Initial Sites of Viral Infection. Cell 2017, 171, 795–808.e12. [Google Scholar] [CrossRef]
  46. McFarland, A.P.; Yalin, A.; Wang, S.-Y.; Cortez, V.S.; Landsberger, T.; Sudan, R.; Peng, V.; Miller, H.L.; Ricci, B.; David, E.; 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 2021, 54, 1320–1337.e4. [Google Scholar] [CrossRef] [PubMed]
  47. Collins, P.L.; Cella, M.; Porter, S.I.; Li, S.; Gurewitz, G.L.; Hong, H.S.; Johnson, R.P.; Oltz, E.M.; Colonna, M. Gene Regulatory Programs Conferring Phenotypic Identities to Human NK Cells. Cell 2018, 176, 348–360.e12. [Google Scholar] [CrossRef] [Green Version]
  48. Crinier, A.; Milpied, P.; Escalière, B.; Piperoglou, C.; Galluso, J.; Balsamo, A.; Spinelli, L.; Cervera-Marzal, I.; Ebbo, M.; Girard-Madoux, M.; et al. High-Dimensional Single-Cell Analysis Identifies Organ-Specific Signatures and Conserved NK Cell Subsets in Humans and Mice. Immunity 2018, 49, 971–986.e5. [Google Scholar] [CrossRef] [Green Version]
  49. Freud, A.G.; Mundy-Bosse, B.L.; Yu, J.; Caligiuri, M.A. The Broad Spectrum of Human Natural Killer Cell Diversity. Immunity 2017, 47, 820–833. [Google Scholar] [CrossRef] [Green Version]
  50. Huntington, N.; Vosshenrich, C.A.J.; Di Santo, J.P. Developmental pathways that generate natural-killer-cell diversity in mice and humans. Nat. Rev. Immunol. 2007, 7, 703–714. [Google Scholar] [CrossRef]
  51. Caligiuri, M.A. Human natural killer cells. Blood 2008, 112, 461–469. [Google Scholar] [CrossRef] [PubMed]
  52. Morvan, M.; Lanier, L.L. NK cells and cancer: You can teach innate cells new tricks. Nat. Rev. Cancer 2015, 16, 7–19. [Google Scholar] [CrossRef] [PubMed]
  53. Grégoire, C.; Chasson, L.; Luci, C.; Tomasello, E.; Geissmann, F.; Vivier, E.; Walzer, T. The trafficking of natural killer cells. Immunol. Rev. 2007, 220, 169–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bernardini, G.; Gismondi, A.; Santoni, A. Chemokines and NK cells: Regulators of development, trafficking and functions. Immunol. Lett. 2012, 145, 39–46. [Google Scholar] [CrossRef]
  55. Soriani, A.; Stabile, H.; Gismondi, A.; Santoni, A.; Bernardini, G. Chemokine regulation of innate lymphoid cell tissue distribution and function. Cytokine Growth Factor Rev. 2018, 42, 47–55. [Google Scholar] [CrossRef]
  56. Koues, O.I.; Collins, P.L.; Cella, M.; Robinette, M.L.; Porter, S.I.; Pyfrom, S.C.; Payton, J.E.; Colonna, M.; Oltz, E.M. Distinct Gene Regulatory Pathways for Human Innate versus Adaptive Lymphoid Cells. Cell 2016, 165, 1134–1146. [Google Scholar] [CrossRef] [Green Version]
  57. Fernando, N.; Sciumè, G.; O’Shea, J.J.; Shih, H.-Y. Multi-Dimensional Gene Regulation in Innate and Adaptive Lymphocytes: A View from Regulomes. Front. Immunol. 2021, 12, 655590. [Google Scholar] [CrossRef]
  58. Chiossone, L.; Chaix, J.; Fuseri, N.; Roth, C.; Vivier, E.; Walzer, T. Maturation of mouse NK cells is a 4-stage developmental program. Blood 2009, 113, 5488–5496. [Google Scholar] [CrossRef] [Green Version]
  59. Vosshenrich, C.A.; Di Santo, J. Developmental programming of natural killer and innate lymphoid cells. Curr. Opin. Immunol. 2013, 25, 130–138. [Google Scholar] [CrossRef] [Green Version]
  60. Lim, A.I.; Di Santo, J.P. ILC-poiesis: Ensuring tissue ILC differentiation at the right place and time. Eur. J. Immunol. 2018, 49, 11–18. [Google Scholar] [CrossRef] [Green Version]
  61. Hayakawa, Y.; Huntington, N.D.; Nutt, S.; Smyth, M.J. Functional subsets of mouse natural killer cells. Immunol. Rev. 2006, 214, 47–55. [Google Scholar] [CrossRef] [PubMed]
  62. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef] [PubMed]
  63. Lanier, L.L. Up on the tightrope: Natural killer cell activation and inhibition. Nat. Immunol. 2008, 9, 495–502. [Google Scholar] [CrossRef]
  64. Martinet, L.; Smyth, M. Balancing natural killer cell activation through paired receptors. Nat. Rev. Immunol. 2015, 15, 243–254. [Google Scholar] [CrossRef]
  65. Moffett, A.; Colucci, F. Co-evolution of NK receptors and HLA ligands in humans is driven by reproduction. Immunol. Rev. 2015, 267, 283–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Guia, S.; Fenis, A.; Vivier, E.; Narni-Mancinelli, E. Activating and inhibitory receptors expressed on innate lymphoid cells. Semin. Immunopathol. 2018, 40, 331–341. [Google Scholar] [CrossRef]
  67. Long, E.O. Negative signaling by inhibitory receptors: The NK cell paradigm. Immunol. Rev. 2008, 224, 70–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Anfossi, N.; André, P.; Guia, S.; Falk, C.S.; Roetynck, S.; Stewart, C.A.; Breso, V.; Frassati, C.; Reviron, D.; Middleton, D.; et al. Human NK Cell Education by Inhibitory Receptors for MHC Class I. Immunity 2006, 25, 331–342. [Google Scholar] [CrossRef]
  69. Kadri, N.; Wagner, A.; Ganesan, S.; Kärre, K.; Wickström, S.; Johansson, M.H.; Höglund, P. Dynamic Regulation of NK Cell Responsiveness. Curr. Top. Microbiol. Immunol. 2016, 395, 95–114. [Google Scholar] [CrossRef]
  70. Kim, S.; Poursine-Laurent, J.; Truscott, S.M.; Lybarger, L.; Song, Y.-J.; Yang, L.; French, A.R.; Sunwoo, J.B.; Lemieux, S.; Hansen, T.H.; et al. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature 2005, 436, 709–713. [Google Scholar] [CrossRef]
  71. Shifrin, N.; Raulet, D.H.; Ardolino, M. NK cell self tolerance, responsiveness and missing self recognition. Semin. Immunol. 2014, 26, 138–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Sciumè, G.; Fionda, C.; Stabile, H.; Gismondi, A.; Santoni, A. Negative regulation of innate lymphoid cell responses in inflammation and cancer. Immunol. Lett. 2019, 215, 28–34. [Google Scholar] [CrossRef] [PubMed]
  73. Raulet, D.H.; Gasser, S.; Gowen, B.G.; Deng, W.; Jung, H. Regulation of Ligands for the NKG2D Activating Receptor. Annu. Rev. Immunol. 2013, 31, 413–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kanakaraj, P.; Duckworth, B.; Azzoni, L.; Kamoun, M.; Cantley, L.; Perussia, B. Phosphatidylinositol-3 kinase activation induced upon FcγRIIIA-ligand interaction. J. Exp. Med. 1994, 179, 551–558. [Google Scholar] [CrossRef] [PubMed]
  75. Bryceson, Y.; March, M.; Ljunggren, H.-G.; Long, E.O. Activation, coactivation, and costimulation of resting human natural killer cells. Immunol. Rev. 2006, 214, 73–91. [Google Scholar] [CrossRef] [Green Version]
  76. Mace, E.M.; Orange, J.S. Emerging insights into human health and NK cell biology from the study of NK cell deficiencies. Immunol. Rev. 2018, 287, 202–225. [Google Scholar] [CrossRef]
  77. Cherrier, D.E.; Serafini, N.; Di Santo, J.P. Innate Lymphoid Cell Development: A T Cell Perspective. Immunity 2018, 48, 1091–1103. [Google Scholar] [CrossRef] [Green Version]
  78. Stokic-Trtica, V.; Diefenbach, A.; Klose, C.S.N. NK Cell Development in Times of Innate Lymphoid Cell Diversity. Front. Immunol. 2020, 11, 813. [Google Scholar] [CrossRef]
  79. Gordon, S.M.; Chaix, J.; Rupp, L.J.; Wu, J.; Madera, S.; Sun, J.C.; Lindsten, T.; Reiner, S.L. The Transcription Factors T-bet and Eomes Control Key Checkpoints of Natural Killer Cell Maturation. Immunity 2012, 36, 55–67. [Google Scholar] [CrossRef] [Green Version]
  80. Daussy, C.; Faure, F.; Mayol, K.; Viel, S.; Gasteiger, G.; Charrier, E.; Bienvenu, J.; Henry, T.; Debien, E.; Hasan, U.; et al. T-bet and Eomes instruct the development of two distinct natural killer cell lineages in the liver and in the bone marrow. J. Exp. Med. 2014, 211, 563–577. [Google Scholar] [CrossRef]
  81. Zhang, J.; Le Gras, S.; Pouxvielh, K.; Faure, F.; Fallone, L.; Kern, N.; Moreews, M.; Mathieu, A.-L.; Schneider, R.; Marliac, Q.; et al. Sequential actions of EOMES and T-BET promote stepwise maturation of natural killer cells. Nat. Commun. 2021, 12, 5446. [Google Scholar] [CrossRef] [PubMed]
  82. Townsend, M.J.; Weinmann, A.S.; Matsuda, J.L.; Salomon, R.; Farnham, P.J.; Biron, C.A.; Gapin, L.; Glimcher, L.H. T-bet Regulates the Terminal Maturation and Homeostasis of NK and Vα14i NKT Cells. Immunity 2004, 20, 477–494. [Google Scholar] [CrossRef] [Green Version]
  83. Sciume, G.; Hirahara, K.; Takahashi, H.; Laurence, A.; Villarino, A.; Singleton, K.L.; Spencer, S.; Wilhelm, C.; Poholek, A.C.; Vahedi, G.; et al. Distinct requirements for T-bet in gut innate lymphoid cells. J. Exp. Med. 2012, 209, 2331–2338. [Google Scholar] [CrossRef] [Green Version]
  84. Klose, C.S.N.; Kiss, E.A.; Schwierzeck, V.; Ebert, K.; Hoyler, T.; D’Hargues, Y.; Göppert, N.; Croxford, A.L.; Waisman, A.; Tanriver, Y.; et al. A T-bet gradient controls the fate and function of CCR6−RORγt+ innate lymphoid cells. Nature 2013, 494, 261–265. [Google Scholar] [CrossRef] [PubMed]
  85. Klose, C.S.N.; Flach, M.; Möhle, L.; Rogell, L.; Hoyler, T.; Ebert, K.; Fabiunke, C.; Pfeifer, D.; Sexl, V.; Fonseca-Pereira, D.; et al. Differentiation of Type 1 ILCs from a Common Progenitor to All Helper-like Innate Lymphoid Cell Lineages. Cell 2014, 157, 340–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Rankin, L.C.; Groom, J.; Chopin, M.; Herold, M.; Walker, J.A.; Mielke, L.; McKenzie, A.N.J.; Carotta, S.; Nutt, S.; Belz, G.T. The transcription factor T-bet is essential for the development of NKp46+ innate lymphocytes via the Notch pathway. Nat. Immunol. 2013, 14, 389–395. [Google Scholar] [CrossRef] [Green Version]
  87. Gascoyne, D.M.; Long, E.; Veiga-Fernandes, H.; de Boer, J.; Williams, O.; Seddon, B.; Coles, M.; Kioussis, D.; Brady, H. The basic leucine zipper transcription factor E4BP4 is essential for natural killer cell development. Nat. Immunol. 2009, 10, 1118–1124, Erratum in Nat. Immunol. 2011, 11, 543. [Google Scholar] [CrossRef] [PubMed]
  88. Cortez, V.S.; Fuchs, A.; Cella, M.; Gilfillan, S.; Colonna, M. Cutting Edge: Salivary Gland NK Cells Develop Independently of Nfil3 in Steady-State. J. Immunol. 2014, 192, 4487–4491. [Google Scholar] [CrossRef]
  89. Crotta, S.; Gkioka, A.; Male, V.; Duarte, J.H.; Davidson, S.; Nisoli, I.; Brady, H.; Wack, A. The Transcription Factor E4BP4 Is Not Required for Extramedullary Pathways of NK Cell Development. J. Immunol. 2014, 192, 2677–2688. [Google Scholar] [CrossRef] [Green Version]
  90. Sojka, D.K.; Plougastel-Douglas, B.; Yang, L.; A Pak-Wittel, M.; Artyomov, M.N.; Ivanova, Y.; Zhong, C.; Chase, J.; Rothman, P.B.; Yu, J.; et al. Tissue-resident natural killer (NK) cells are cell lineages distinct from thymic and conventional splenic NK cells. eLife 2014, 3, e01659. [Google Scholar] [CrossRef]
  91. Doisne, J.-M.; Balmas, E.; Boulenouar, S.; Gaynor, L.M.; Kieckbusch, J.; Gardner, L.; Hawkes, D.A.; Barbara, C.F.; Sharkey, A.M.; Brady, H.J.M.; et al. Composition, Development, and Function of Uterine Innate Lymphoid Cells. J. Immunol. 2015, 195, 3937–3945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. O’Sullivan, T.E.; Rapp, M.; Fan, X.; Weizman, O.-E.; Bhardwaj, P.; Adams, N.M.; Walzer, T.; Dannenberg, A.J.; Sun, J.C. Adipose-Resident Group 1 Innate Lymphoid Cells Promote Obesity-Associated Insulin Resistance. Immunity 2016, 45, 428–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Seillet, C.; Rankin, L.C.; Groom, J.; Mielke, L.; Tellier, J.; Chopin, M.; Huntington, N.; Belz, G.T.; Carotta, S. Nfil3 is required for the development of all innate lymphoid cell subsets. J. Exp. Med. 2014, 211, 1733–1740. [Google Scholar] [CrossRef]
  94. Geiger, T.L.; Abt, M.C.; Gasteiger, G.; Firth, M.A.; O’Connor, M.H.; Geary, C.D.; O’Sullivan, T.; Brink, M.R.V.D.; Pamer, E.G.; Hanash, A.M.; et al. Nfil3 is crucial for development of innate lymphoid cells and host protection against intestinal pathogens. J. Exp. Med. 2014, 211, 1723–1731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Yu, X.; Wang, Y.; Deng, M.; Li, Y.; Ruhn, K.A.; Zhang, C.C.; Hooper, L.V. The basic leucine zipper transcription factor NFIL3 directs the development of a common innate lymphoid cell precursor. eLife 2014, 3, e04406. [Google Scholar] [CrossRef]
  96. Xu, W.; Domingues, R.G.; Fonseca-Pereira, D.; Ferreira, M.; Ribeiro, H.; Lopez-Lastra, S.; Motomura, Y.; Moreira-Santos, L.; Bihl, F.; Braud, V.; et al. NFIL3 Orchestrates the Emergence of Common Helper Innate Lymphoid Cell Precursors. Cell Rep. 2015, 10, 2043–2054. [Google Scholar] [CrossRef]
  97. Yokota, Y.; Mansouri, A.; Mori, S.; Sugawara, S.; Adachi, S.; Nishikawa, S.-I.; Gruss, P. Development of peripheral lymphoid organs and natural killer cells depends on the helix–loop–helix inhibitor Id2. Nature 1999, 397, 702–706. [Google Scholar] [CrossRef]
  98. Ikawa, T.; Fujimoto, S.; Kawamoto, H.; Katsura, Y.; Yokota, Y. Commitment to natural killer cells requires the helix–loop–helix inhibitor Id2. Proc. Natl. Acad. Sci. USA 2001, 98, 5164–5169. [Google Scholar] [CrossRef] [Green Version]
  99. Boos, M.D.; Yokota, Y.; Eberl, G.; Kee, B.L. Mature natural killer cell and lymphoid tissue–inducing cell development requires Id2-mediated suppression of E protein activity. J. Exp. Med. 2007, 204, 1119–1130. [Google Scholar] [CrossRef]
  100. Moro, K.; Yamada, T.; Tanabe, M.; Takeuchi, T.; Ikawa, T.; Kawamoto, H.; Furusawa, J.-I.; Ohtani, M.; Fujii, H.; Koyasu, S. Innate production of TH2 cytokines by adipose tissue-associated c-Kit+Sca-1+ lymphoid cells. Nature 2010, 463, 540–544. [Google Scholar] [CrossRef]
  101. Mace, E.M.; Hsu, A.P.; Monaco-Shawver, L.; Makedonas, G.; Rosen, J.B.; Dropulic, L.; Cohen, J.I.; Frenkel, E.P.; Bagwell, J.C.; Sullivan, J.L.; et al. Mutations in GATA2 cause human NK cell deficiency with specific loss of the CD56bright subset. Blood 2013, 121, 2669–2677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Yang, R.; Mele, F.; Worley, L.; Langlais, D.; Rosain, J.; Benhsaien, I.; Elarabi, H.; Croft, C.A.; Doisne, J.-M.; Zhang, P.; et al. Human T-bet Governs Innate and Innate-like Adaptive IFN-γ Immunity against Mycobacteria. Cell 2020, 183, 1826–1847.e31. [Google Scholar] [CrossRef] [PubMed]
  103. Mace, E.; Bigley, V.; Gunesch, J.T.; Chinn, I.K.; Angelo, L.S.; Care, M.A.; Maisuria, S.; Keller, M.D.; Togi, S.; Watkin, L.; et al. Biallelic mutations in IRF8 impair human NK cell maturation and function. J. Clin. Investig. 2016, 127, 306–320. [Google Scholar] [CrossRef] [PubMed]
  104. Orange, J.S. How I Manage Natural Killer Cell Deficiency. J. Clin. Immunol. 2019, 40, 13–23. [Google Scholar] [CrossRef] [PubMed]
  105. Bernink, J.H.; Mjösberg, J.; Spits, H. Human ILC1: To Be or Not to Be. Immunity 2017, 46, 756–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Gao, Y.; Souza-Fonseca-Guimaraes, F.; Bald, T.; Ng, S.; Young, A.; Ngiow, S.F.; Rautela, J.; Straube, J.; Waddell, N.; Blake, S.J.; et al. Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat. Immunol. 2017, 18, 1004–1015. [Google Scholar] [CrossRef]
  107. Chen, Y.; Wang, X.; Hao, X.; Li, B.; Tao, W.; Zhu, S.; Qu, K.; Wei, H.; Sun, R.; Peng, H.; et al. Ly49E separates liver ILC1s into embryo-derived and postnatal subsets with different functions. J. Exp. Med. 2022, 219, e20211805. [Google Scholar] [CrossRef]
  108. Clark, C.R.; Starr, T.K. Mouse models for the discovery of colorectal cancer driver genes. World J. Gastroenterol. 2016, 22, 815–822. [Google Scholar] [CrossRef]
  109. Halama, N.; Braun, M.; Kahlert, C.; Spille, A.; Quack, C.; Rahbari, N.; Koch, M.; Weitz, J.; Kloor, M.; Zoernig, I.; et al. Natural Killer Cells are Scarce in Colorectal Carcinoma Tissue Despite High Levels of Chemokines and Cytokines. Clin. Cancer Res. 2011, 17, 678–689. [Google Scholar] [CrossRef] [Green Version]
  110. Sconocchia, G.; Eppenberger, S.; Spagnoli, G.C.; Tornillo, L.; Droeser, R.; Caratelli, S.; Ferrelli, F.; Coppola, A.; Arriga, R.; Lauro, D.; et al. NK cells and T cells cooperate during the clinical course of colorectal cancer. OncoImmunology 2014, 3, e952197. [Google Scholar] [CrossRef] [Green Version]
  111. Pan, P.; Kang, S.; Wang, Y.; Liu, K.; Oshima, K.; Huang, Y.-W.; Zhang, J.; Yearsley, M.; Yu, J.; Wang, L.-S. Black Raspberries Enhance Natural Killer Cell Infiltration into the Colon and Suppress the Progression of Colorectal Cancer. Front. Immunol. 2017, 8, 997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Bank, U.; Deiser, K.; Plaza-Sirvent, C.; Osbelt, L.; Witte, A.; Knop, L.; Labrenz, R.; Jänsch, R.; Richter, F.; Biswas, A.; et al. c-FLIP is crucial for IL-7/IL-15-dependent NKp46+ ILC development and protection from intestinal inflammation in mice. Nat. Commun. 2020, 11, 1056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Qi, J.; Crinier, A.; Escalière, B.; Ye, Y.; Wang, Z.; Zhang, T.; Batista, L.; Liu, H.; Hong, L.; Wu, N.; et al. Single-cell transcriptomic landscape reveals tumor specific innate lymphoid cells associated with colorectal cancer progression. Cell Rep. Med. 2021, 2, 100353. [Google Scholar] [CrossRef] [PubMed]
  114. Simoni, Y.; Fehlings, M.; Kløverpris, H.N.; McGovern, N.; Koo, S.-L.; Loh, C.Y.; Lim, S.; Kurioka, A.; Fergusson, J.R.; Tang, C.-L.; et al. Human Innate Lymphoid Cell Subsets Possess Tissue-Type Based Heterogeneity in Phenotype and Frequency. Immunity 2017, 46, 148–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. De Vries, N.L.; Van Unen, V.; Ijsselsteijn, M.E.; Abdelaal, T.; Van Der Breggen, R.; Sarasqueta, A.F.; Mahfouz, A.; Peeters, K.C.M.J.; Höllt, T.; Lelieveldt, B.P.F.; et al. High-dimensional cytometric analysis of colorectal cancer reveals novel mediators of antitumour immunity. Gut 2020, 69, 691–703. [Google Scholar] [CrossRef] [Green Version]
  116. Wang, S.; Qu, Y.; Xia, P.; Chen, Y.; Zhu, X.; Zhang, J.; Wang, G.; Tian, Y.; Ying, J.; Fan, Z. Transdifferentiation of tumor infiltrating innate lymphoid cells during progression of colorectal cancer. Cell Res. 2020, 30, 610–622. [Google Scholar] [CrossRef]
  117. Mazzurana, L.; Bonfiglio, F.; Forkel, M.; D’Amato, M.; Halfvarson, J.; Mjösberg, J. Crohn’s Disease Is Associated with Activation of Circulating Innate Lymphoid Cells. Inflamm. Bowel Dis. 2020, 27, 1128–1138. [Google Scholar] [CrossRef]
  118. Krijgsman, D.; de Vries, N.L.; Skovbo, A.; Andersen, M.N.; Swets, M.; Bastiaannet, E.; Vahrmeijer, A.L.; van de Velde, C.J.H.; Heemskerk, M.H.M.; Hokland, M.; et al. Characterization of circulating T-, NK-, and NKT cell subsets in patients with colorectal cancer: The peripheral blood immune cell profile. Cancer Immunol. Immunother. 2019, 68, 1011–1024. [Google Scholar] [CrossRef] [Green Version]
  119. Gharagozloo, M.; Rezaei, A.; Kalantari, H.; Bahador, A.; Hassannejad, N.; Maracy, M.R.; Nouri, N.; Sedghi, M.; Ghazanfari, H.; Bayat, B. Decline in peripheral blood NKG2D+CD3+CD56+ NKT cells in metastatic colorectal cancer patients. Bratisl. Med. J. 2018, 119, 6–11. [Google Scholar] [CrossRef] [Green Version]
  120. Gharagozloo, M.; Kalantari, H.; Rezaei, A.; Maracy, M.R.; Salehi, M.; Bahador, A.; Hassannejad, N.; Narimani, M.; Sanei, M.H.; Bayat, B.; et al. The decrease in NKG2D+ Natural Killer cells in peripheral blood of patients with metastatic colorectal cancer. Bratisl. Med. J. 2015, 116, 296–301. [Google Scholar] [CrossRef]
  121. Peng, Y.-P.; Zhu, Y.; Zhang, J.-J.; Xu, Z.-K.; Qian, Z.-Y.; Dai, C.-C.; Jiang, K.-R.; Wu, J.-L.; Gao, W.-T.; Li, Q.; et al. Comprehensive analysis of the percentage of surface receptors and cytotoxic granules positive natural killer cells in patients with pancreatic cancer, gastric cancer, and colorectal cancer. J. Transl. Med. 2013, 11, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Rocca, Y.S.; Roberti, M.P.; Arriaga, J.M.; Amat, M.; Bruno, L.; Pampena, M.B.; Huertas, E.; Loria, F.S.; Pairola, A.; Bianchini, M.; et al. Altered phenotype in peripheral blood and tumor-associated NK cells from colorectal cancer patients. Innate Immun. 2012, 19, 76–85. [Google Scholar] [CrossRef] [PubMed]
  123. Rocca, Y.S.; Roberti, M.P.; Juliá, E.P.; Pampena, M.B.; Bruno, L.; Rivero, S.; Huertas, E.; Loria, F.S.; Pairola, A.; Caignard, A.; et al. Phenotypic and Functional Dysregulated Blood NK Cells in Colorectal Cancer Patients Can Be Activated by Cetuximab Plus IL-2 or IL-15. Front. Immunol. 2016, 7, 413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Krijgsman, D.; Roelands, J.; Andersen, M.N.; Wieringa, C.H.; Tollenaar, R.A.; Hendrickx, W.; Bedognetti, D.; Hokland, M.; Kuppen, P.J. Expression of NK cell receptor ligands in primary colorectal cancer tissue in relation to the phenotype of circulating NK- and NKT cells, and clinical outcome. Mol. Immunol. 2020, 128, 205–218. [Google Scholar] [CrossRef]
  125. Stabile, H.; Fionda, C.; Gismondi, A.; Santoni, A. Role of Distinct Natural Killer Cell Subsets in Anticancer Response. Front. Immunol. 2017, 8, 293. [Google Scholar] [CrossRef] [Green Version]
  126. Mahapatra, S.; Mace, E.M.; Minard, C.G.; Forbes, L.R.; Vargas-Hernandez, A.; Duryea, T.K.; Makedonas, G.; Banerjee, P.P.; Shearer, W.; Orange, J.S. High-resolution phenotyping identifies NK cell subsets that distinguish healthy children from adults. PLoS ONE 2017, 12, e0181134. [Google Scholar] [CrossRef]
  127. Stannard, K.A.; Lemoine, S.; Waterhouse, N.J.; Vari, F.; Chatenoud, L.; Gandhi, M.K.; Martinet, L.; Smyth, M.J.; Guillerey, C. Human peripheral blood DNAM-1neg NK cells are a terminally differentiated subset with limited effector functions. Blood Adv. 2019, 3, 1681–1694. [Google Scholar] [CrossRef]
  128. Pesce, S.; Tabellini, G.; Cantoni, C.; Patrizi, O.; Coltrini, D.; Rampinelli, F.; Matta, J.; Vivier, E.; Moretta, A.; Parolini, S.; et al. B7-H6-mediated downregulation of NKp30 in NK cells contributes to ovarian carcinoma immune escape. OncoImmunology 2015, 4, e1001224. [Google Scholar] [CrossRef] [Green Version]
  129. Salomé, B.; Gomez-Cadena, A.; Loyon, R.; Suffiotti, M.; Salvestrini, V.; Wyss, T.; Vanoni, G.; Ruan, D.F.; Rossi, M.; Tozzo, A.; et al. CD56 as a marker of an ILC1-like population with NK cell properties that is functionally impaired in AML. Blood Adv. 2019, 3, 3674–3687. [Google Scholar] [CrossRef]
  130. Lopez-Vergès, S.; Milush, J.M.; Pandey, S.; York, V.A.; Arakawa-Hoyt, J.; Pircher, H.; Norris, P.J.; Nixon, D.; Lanier, L.L. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood 2010, 116, 3865–3874. [Google Scholar] [CrossRef] [Green Version]
  131. Jonges, L.E.; Albertsson, P.; Van Vlierberghe, R.L.P.; Ensink, N.G.; Johansson, B.R.; Van De Velde, C.J.H.; Fleuren, G.J.; Nannmark, U.; Kuppen, P. The Phenotypic Heterogeneity of Human Natural Killer Cells: Presence of at least 48 Different Subsets in the Peripheral Blood. Scand. J. Immunol. 2001, 53, 103–110. [Google Scholar] [CrossRef] [PubMed]
  132. Stabile, H.; Nisti, P.; Morrone, S.; Pagliara, D.; Bertaina, A.; Locatelli, F.; Santoni, A.; Gismondi, A. Multifunctional human CD56low CD16low natural killer cells are the prominent subset in bone marrow of both healthy pediatric donors and leukemic patients. Haematologica 2015, 100, 489–498. [Google Scholar] [CrossRef] [Green Version]
  133. Zhang, Q.; Bi, J.; Zheng, X.; Chen, Y.; Wang, H.; Wu, W.; Wang, Z.; Wu, Q.; Peng, H.; Wei, H.; et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 2018, 19, 723–732. [Google Scholar] [CrossRef]
  134. André, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Bléry, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells. Cell 2018, 175, 1731–1743.e13. [Google Scholar] [CrossRef] [Green Version]
  135. Harmon, C.; Robinson, M.W.; Hand, F.; Almuaili, D.; Mentor, K.; Houlihan, D.D.; Hoti, E.; Lynch, L.; Geoghegan, J.; O’Farrelly, C. Lactate-Mediated Acidification of Tumor Microenvironment Induces Apoptosis of Liver-Resident NK Cells in Colorectal Liver Metastasis. Cancer Immunol. Res. 2019, 7, 335–346. [Google Scholar] [CrossRef] [Green Version]
  136. Ghanadi, K.; Shayanrad, B.; Ahmadi, S.A.Y.; Shahsavar, F.; Eliasy, H. Colorectal cancer and the KIR genes in the human genome: A meta-analysis. Genom. Data 2016, 10, 118–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Beksac, K.; Beksaç, M.; Dalva, K.; Karaagaoglu, E.; Tirnaksiz, M.B. Impact of “Killer Immunoglobulin-Like Receptor /Ligand” Genotypes on Outcome following Surgery among Patients with Colorectal Cancer: Activating KIRs Are Associated with Long-Term Disease Free Survival. PLoS ONE 2015, 10, e0132526. [Google Scholar] [CrossRef] [PubMed]
  138. Menon, A.G.; Morreau, H.; Tollenaar, R.A.E.M.; Alphenaar, E.; Van Puijenbroek, M.; Putter, H.; Rhijn, C.M.J.-V.; Van De Velde, C.J.H.; Fleuren, G.J.; Kuppen, P.J.K. Down-Regulation of HLA-A Expression Correlates with a Better Prognosis in Colorectal Cancer Patients. Lab. Investig. 2002, 82, 1725–1733. [Google Scholar] [CrossRef] [Green Version]
  139. Zhou, X.; Li, W.-Q.; Wu, Y.-H.; Han, L.; Cao, X.-G.; Yang, X.; Wang, H.-F.; Zhao, W.-S.; Zhai, W.-J.; Qi, Y.-M.; et al. Intrinsic Expression of Immune Checkpoint Molecule TIGIT Could Help Tumor Growth in vivo by Suppressing the Function of NK and CD8+ T Cells. Front. Immunol. 2018, 9, 2821. [Google Scholar] [CrossRef] [Green Version]
  140. Tallerico, R.; Todaro, M.; Di Franco, S.; Maccalli, C.; Garofalo, C.; Sottile, R.; Palmieri, C.; Tirinato, L.; Pangigadde, P.N.; La Rocca, R.; et al. Human NK Cells Selective Targeting of Colon Cancer–Initiating Cells: A Role for Natural Cytotoxicity Receptors and MHC Class I Molecules. J. Immunol. 2013, 190, 2381–2390. [Google Scholar] [CrossRef] [Green Version]
  141. Cardoso, R.; Guo, F.; Heisser, T.; Hackl, M.; Ihle, P.; De Schutter, H.; Van Damme, N.; Valerianova, Z.; Atanasov, T.; Májek, O.; et al. Colorectal cancer incidence, mortality, and stage distribution in European countries in the colorectal cancer screening era: An international population-based study. Lancet Oncol. 2021, 22, 1002–1013. [Google Scholar] [CrossRef]
  142. Freitas-Lopes, M.A.; Mafra, K.; David, B.A.; Carvalho-Gontijo, R.; Menezes, G.B. Differential Location and Distribution of Hepatic Immune Cells. Cells 2017, 6, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Mikulak, J.; Bruni, E.; Oriolo, F.; Di Vito, C.; Mavilio, D. Hepatic Natural Killer Cells: Organ-Specific Sentinels of Liver Immune Homeostasis and Physiopathology. Front. Immunol. 2019, 10, 946. [Google Scholar] [CrossRef] [Green Version]
  144. Highton, A.J.; Schuster, I.S.; Degli-Esposti, M.A.; Altfeld, M. The role of natural killer cells in liver inflammation. Semin. Immunopathol. 2021, 43, 519–533. [Google Scholar] [CrossRef]
  145. Doherty, D.G.; Norris, S.; Madrigal-Estebas, L.; McEntee, G.; Traynor, O.; Hegarty, J.E.; O’Farrelly, C. The human liver contains multiple populations of NK cells, T cells, and CD3+CD56+ natural T cells with distinct cytotoxic activities and Th1, Th2, and Th0 cytokine secretion patterns. J. Immunol. 1999, 163, 2314–2321. [Google Scholar]
  146. Luo, D.Z.; Vermijlen, D.; Ahishali, B.; Triantis, V.; Plakoutsi, G.; Braet, F.; Vanderkerken, K.; Wisse, E. On the cell biology of pit cells, the liver-specific NK cells. World J. Gastroenterol. 2000, 6, 1–11. [Google Scholar] [CrossRef] [PubMed]
  147. Norris, S.; Collins, C.; Doherty, D.G.; Smith, F.; McEntee, G.; Traynor, O.; Nolan, N.; Hegarty, J.; O’Farrelly, C. Resident human hepatitis lymphocytes are phenotypically different from circulating lymphocytes. J. Hepatol. 1998, 28, 84–90. [Google Scholar] [CrossRef]
  148. Hata, K.; van Thiel, D.H.; Herberman, R.B.; Whiteside, T.L. Phenotypic and functional characteristics of lymphocytes isolated from liver biopsy specimens from patients with active liver disease. Hepatology 1992, 15, 816–823. [Google Scholar] [CrossRef]
  149. Harmon, C.; Robinson, M.W.; Fahey, R.; Whelan, S.; Houlihan, D.D.; Geoghegan, J.; O’Farrelly, C. Tissue-resident Eomes(hi) T-bet(lo) CD56(bright) NK cells with reduced proinflammatory potential are enriched in the adult human liver. Eur. J. Immunol. 2016, 46, 2111–2120. [Google Scholar] [CrossRef] [Green Version]
  150. Stegmann, K.A.; Robertson, F.; Hansi, N.; Gill, U.; Pallant, C.; Christophides, T.; Pallett, L.J.; Peppa, D.; Dunn, C.; Fusai, G.; et al. CXCR6 marks a novel subset of T-betloEomeshi natural killer cells residing in human liver. Sci. Rep. 2016, 6, 26157. [Google Scholar] [CrossRef]
  151. Marquardt, N.; Béziat, V.; Nyström, S.; Hengst, J.; Ivarsson, M.A.; Kekäläinen, E.; Johansson, H.; Mjösberg, J.; Westgren, M.; Lankisch, T.O.; et al. Cutting Edge: Identification and Characterization of Human Intrahepatic CD49a+ NK Cells. J. Immunol. 2015, 194, 2467–2471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Yeang, H.X.A.; Piersma, S.J.; Lin, Y.; Yang, L.; Malkova, O.N.; Miner, C.; Krupnick, A.S.; Chapman, W.C.; Yokoyama, W.M. Cutting Edge: Human CD49e NK Cells Are Tissue Resident in the Liver. J. Immunol. 2017, 198, 1417–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Hudspeth, K.; Donadon, M.; Cimino, M.; Pontarini, E.; Tentorio, P.; Preti, M.; Hong, M.; Bertoletti, A.; Bicciato, S.; Invernizzi, P.; et al. Human liver-resident CD56bright/CD16neg NK cells are retained within hepatic sinusoids via the engagement of CCR5 and CXCR6 pathways. J. Autoimmun. 2015, 66, 40–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Cuff, A.O.; Robertson, F.P.; Stegmann, K.A.; Pallett, L.J.; Maini, M.K.; Davidson, B.R.; Male, V. Eomeshi NK Cells in Human Liver Are Long-Lived and Do Not Recirculate but Can Be Replenished from the Circulation. J. Immunol. 2016, 197, 4283–4291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Moroso, V.; Famili, F.; Papazian, N.; Cupedo, T.; van der Laan, L.J.W.; Kazemier, G.; Metselaar, H.J.; Kwekkeboom, J. NK cells can generate from precursors in the adult human liver. Eur. J. Immunol. 2011, 41, 3340–3350. [Google Scholar] [CrossRef]
  156. Hydes, T.; Abuhilal, M.; Armstrong, T.; Primrose, J.; Takhar, A.; Khakoo, S. Natural killer cell maturation markers in the human liver and expansion of an NKG2C+KIR+ population. Lancet 2015, 385, S45. [Google Scholar] [CrossRef]
  157. Male, V. Liver-Resident NK Cells: The Human Factor. Trends Immunol. 2017, 38, 307–309. [Google Scholar] [CrossRef]
  158. Harmon, C.; Jameson, G.; AlMuaili, D.; Houlihan, D.D.; Hoti, E.; Geoghegan, J.; Robinson, M.W.; O’Farrelly, C. Liver-Derived TGF-β Maintains the EomeshiTbetlo Phenotype of Liver Resident Natural Killer Cells. Front. Immunol. 2019, 10, 1502. [Google Scholar] [CrossRef]
  159. Fernández, J.P.; Luddy, K.A.; Harmon, C.; O’Farrelly, C. Hepatic Tumor Microenvironments and Effects on NK Cell Phenotype and Function. Int. J. Mol. Sci. 2019, 20, 4131. [Google Scholar] [CrossRef] [Green Version]
  160. Donadon, M.; Hudspeth, K.; Cimino, M.; Di Tommaso, L.; Preti, M.; Tentorio, P.; Roncalli, M.; Mavilio, D.; Torzilli, G. Increased Infiltration of Natural Killer and T Cells in Colorectal Liver Metastases Improves Patient Overall Survival. J. Gastrointest. Surg. 2017, 21, 1226–1236. [Google Scholar] [CrossRef]
  161. Hoffmann, T.K.; Schirlau, K.; Sonkoly, E.; Brandau, S.; Lang, S.; Pivarcsi, A.; Balz, V.; Müller, A.; Homey, B.; Boelke, E.; et al. A novel mechanism for anti-EGFR antibody action involves chemokine-mediated leukocyte infiltration. Int. J. Cancer 2009, 124, 2589–2596. [Google Scholar] [CrossRef] [PubMed]
  162. Pavlova, N.N.; Thompson, C.B. The Emerging Hallmarks of Cancer Metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Casey, J.R.; Grinstein, S.; Orlowski, J. Sensors and regulators of intracellular pH. Nat. Rev. Mol. Cell Biol. 2009, 11, 50–61. [Google Scholar] [CrossRef] [PubMed]
  164. Adotevi, O.; Godet, Y.; Galaine, J.; Lakkis, Z.; Idirene, I.; Certoux, J.M.; Jary, M.; Loyon, R.; Laheurte, C.; Kim, S.; et al. In situ delivery of allogeneic natural killer cell (NK) combined with Cetuximab in liver metastases of gastrointestinal carcinoma: A phase I clinical trial. OncoImmunology 2018, 7, e1424673. [Google Scholar] [CrossRef] [Green Version]
  165. Rosenberg, S.A.; Spiess, P.; LaFreniere, R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 1986, 233, 1318–1321. [Google Scholar] [CrossRef]
  166. Meininger, I.; Carrasco, A.; Rao, A.; Soini, T.; Kokkinou, E.; Mjösberg, J. Tissue-Specific Features of Innate Lymphoid Cells. Trends Immunol. 2020, 41, 902–917. [Google Scholar] [CrossRef]
  167. Takeda, K.; Cretney, E.; Hayakawa, Y.; Ota, T.; Akiba, H.; Ogasawara, K.; Yagita, H.; Kinoshita, K.; Okumura, K.; Smyth, M. TRAIL identifies immature natural killer cells in newborn mice and adult mouse liver. Blood 2005, 105, 2082–2089. [Google Scholar] [CrossRef] [Green Version]
  168. Kamizono, S.; Duncan, G.S.; Seidel, M.G.; Morimoto, A.; Hamada, K.; Grosveld, G.; Akashi, K.; Lind, E.F.; Haight, J.P.; Ohashi, P.S.; et al. Nfil3/E4bp4 is required for the development and maturation of NK cells in vivo. J. Exp. Med. 2009, 206, 2977–2986. [Google Scholar] [CrossRef] [Green Version]
  169. Zhang, L.H.; Shin, J.H.; Haggadone, M.D.; Sunwoo, J.B. The aryl hydrocarbon receptor is required for the maintenance of liver-resident natural killer cells. J. Exp. Med. 2016, 213, 2249–2257. [Google Scholar] [CrossRef]
  170. Song, J.; Song, H.; Wei, H.; Sun, R.; Tian, Z.; Peng, H. Requirement of RORα for maintenance and antitumor immunity of liver-resident natural killer cells/ILC1s. Hepatology 2021, 75, 1181–1193. [Google Scholar] [CrossRef]
  171. Lee, H.H.; Kim, D.; Jung, J.; Kang, H.; Cho, H. NLRP3 Deficiency in Hepatocellular Carcinoma Enhances Surveillance of NK-92 through a Modulation of MICA/B. Int. J. Mol. Sci. 2021, 22, 9285. [Google Scholar] [CrossRef] [PubMed]
  172. Molgora, M.; Bonavita, E.; Ponzetta, A.; Riva, F.; Barbagallo, M.; Jaillon, S.; Popovic, B.; Bernardini, G.; Magrini, E.; Gianni, F.; et al. IL-1R8 is a checkpoint in NK cells regulating anti-tumour and anti-viral activity. Nature 2017, 551, 110–114. [Google Scholar] [CrossRef] [PubMed]
  173. Malaisé, M.; Rovira, J.; Renner, P.; Eggenhofer, E.; Sabet-Baktach, M.; Lantow, M.; Lang, S.A.; Koehl, G.E.; Farkas, S.A.; Loss, M.; et al. KLRG1+ NK Cells Protect T-bet–Deficient Mice from Pulmonary Metastatic Colorectal Carcinoma. J. Immunol. 2014, 192, 1954–1961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Role of NK cells and ILC1 in CRC progression and metastasis. (A) Both NK cells and ILC1 infiltrate CRC at an early stage to block tumor growth. However, tumor-infiltrating NK cells and ILC1 acquire a different phenotype and decrease over time together with their functional abilities. (B) CRC liver metastasization occurs along with the reduction of NK cells in humans and significant alterations of phenotypic and functional properties of NK cells in mice. Arrow indicates a decrease or increase.
Figure 1. Role of NK cells and ILC1 in CRC progression and metastasis. (A) Both NK cells and ILC1 infiltrate CRC at an early stage to block tumor growth. However, tumor-infiltrating NK cells and ILC1 acquire a different phenotype and decrease over time together with their functional abilities. (B) CRC liver metastasization occurs along with the reduction of NK cells in humans and significant alterations of phenotypic and functional properties of NK cells in mice. Arrow indicates a decrease or increase.
Ijms 23 07859 g001
Table 1. Contribution of NK/ILC1 in CRC progression and metastasis. Grey: Decrease; Bold: Increase; -: Not determined.
Table 1. Contribution of NK/ILC1 in CRC progression and metastasis. Grey: Decrease; Bold: Increase; -: Not determined.
PRIMARY CRCLIVER CRC METASTASIS
PhenotypeFunctionRef.PhenotypeFunctionRef.
Ijms 23 07859 i001NKCD16, NKG2D, NKp30, NKp46, CD161, DNAM-1, CD158b, CD158a/h
NKp44
TIGIT+ NKG2A+
-Frequency at late stage
-Degranulation
-IFN-γ, TNF production



-Low infiltrate and functional impairment associated with poor overall survival and relapse
[122]


[133]
[134]

[20]
[109]
[110]
CD56bright
CXCR6+TRAIL+CD69+
NKG2D, NKG2C,
NKp46, NKp44,
PERFORIN+
GRANZYME+
-Apoptosis-induced depletion
-A reduced density associated with metastasis recurrence.
[135]
ILC1TIGIT+ CTLA4+ TNFRSF4+
SLAMF1+
PERFORIN+GRANZYME+
KIR2DL1, KIR2DL2/3
-Frequency at late stage[113]


[116]
Ijms 23 07859 i002NKTIGIT, Ly49a, CD96,
LAG3, TIM3
NKG2D, DNAM-1
-Frequency at late stage
-Degranulation
-IFN-γ, TNF production
[111]

[133]
Eomes+CD49a+CD49b+
Thy1.2+CD69+CD27 +
-Infiltrate the metastatic nodules
-Restrain metastasis growth
-Cytotoxicity at late stage
[33]
ILC1NKp46, CD94, NK1.1,
IL-12RB2
Ly49G2, NKG2I
-Frequency at late stage
-IFN-γ production
[116]--Outside the metastatic nodules
-Control metastasis seeding
-Cytotoxicity at late stage
[33]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fionda, C.; Scarno, G.; Stabile, H.; Molfetta, R.; Di Censo, C.; Gismondi, A.; Paolini, R.; Sozzani, S.; Santoni, A.; Sciumè, G. NK Cells and Other Cytotoxic Innate Lymphocytes in Colorectal Cancer Progression and Metastasis. Int. J. Mol. Sci. 2022, 23, 7859. https://doi.org/10.3390/ijms23147859

AMA Style

Fionda C, Scarno G, Stabile H, Molfetta R, Di Censo C, Gismondi A, Paolini R, Sozzani S, Santoni A, Sciumè G. NK Cells and Other Cytotoxic Innate Lymphocytes in Colorectal Cancer Progression and Metastasis. International Journal of Molecular Sciences. 2022; 23(14):7859. https://doi.org/10.3390/ijms23147859

Chicago/Turabian Style

Fionda, Cinzia, Gianluca Scarno, Helena Stabile, Rosa Molfetta, Chiara Di Censo, Angela Gismondi, Rossella Paolini, Silvano Sozzani, Angela Santoni, and Giuseppe Sciumè. 2022. "NK Cells and Other Cytotoxic Innate Lymphocytes in Colorectal Cancer Progression and Metastasis" International Journal of Molecular Sciences 23, no. 14: 7859. https://doi.org/10.3390/ijms23147859

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop