Abstract
IL-15 belongs to the common gamma chain cytokine family and has pleiotropic immunological functions. IL-15 is a homeostatic cytokine essential for the development and maintenance of NK cells and memory CD8+ T cells. In addition, IL-15 plays a critical role in the activation, effector functions, tissue residency, and senescence of CD8+ T cells. IL-15 also activates virtual memory T cells, mucosal-associated invariant T cells and γδ T cells. Recently, IL-15 has been highlighted as a major trigger of TCR-independent activation of T cells. This mechanism is involved in T cell-mediated immunopathogenesis in diverse diseases, including viral infections and chronic inflammatory diseases. Deeper understanding of IL-15-mediated T-cell responses and their underlying mechanisms could optimize therapeutic strategies to ameliorate host injury by T cell-mediated immunopathogenesis. This review highlights recent advancements in comprehending the role of IL-15 in relation to T cell responses and immunopathogenesis under various host conditions.
Keywords: Interleukin-15; T-lymphocytes, immunopathogenesis
INTRODUCTION
IL-15 is a cytokine composed of four alpha-helical bundles (1) and is well-known as a common gamma chain (γc, CD132)-dependent cytokine (2), a class that also includes IL-2, IL-4, IL-7, IL-9, IL-21, and TSLP (3). IL-15 is frequently referred to as a pleiotropic cytokine due to its mediation of multiple functions in various types of immune cells. Since its discovery nearly 30 years ago (1,4), IL-15 has emerged as a pivotal cytokine with well-established functions in mediating lymphoid homeostasis, most notably in compartments of NK cells and memory CD8+ T cells (5). However, the functions of IL-15 are not only limited to homeostasis, but are also closely associated with a range of contexts, including the activation, effector functions, tissue residency, and senescence of CD8+ T cells. IL-15 has been recognized as a potent stimulatory cytokine that enhances T-cell immune responses with potential for cancer treatment (6). Dysregulated expression of IL-15 has been also demonstrated in various types of diseases (7). Moreover, IL-15 activates diverse types of unconventional T cells and triggers NK-like cytotoxicity, which could contribute to immunopathogenesis in diverse diseases (8,9). This emphasizes the need for a better understanding of the IL-15-mediated T-cell response in various contexts. Although IL-15 is also indispensable in shaping NK cell biology, as comprehensively reviewed elsewhere (10), this review focuses on recent advancements in understanding the role of IL-15 in relation to T-cell responses under varying host conditions.
EXPRESSION OF IL-15 AND ITS SIGNAL TRANSDUCTION
Despite the abundant expression of IL-15 transcripts in a wide variety of tissues and both hematopoietic and non-hematopoietic cells (11,12,13,14,15), IL-15 protein is mainly produced by monocytes, dendritic cells, and epithelial cells (12,14,16). IL-15 primarily operates in a cell contact-dependent manner through the trans-presentation of membrane-bound complexes comprised of IL-15 and IL-15 receptor alpha (IL-15Rα) on the producing cells to IL-2/IL-15 receptor-β chain (CD122) and γc on the responding cells (17). This process begins with the preassembly of IL-15 with IL-15Rα in a complex within the endoplasmic reticulum/Golgi, followed by its subsequent transportation to the cell surface. IL-15 has also been reported to signal as soluble IL-15-IL-15Rα complexes or IL-15 alone, but trans-presentation has been demonstrated to be the dominant mechanism eliciting potent IL-15 signals (18,19,20).
IL-15 production is triggered by inflammation, infection, or prolonged cellular stress (7). Various types of inflammatory stimuli have been shown to trigger IL-15 expression. Treatment with polyinosinic:polycytidylic acid or LPS, either in vivo or in vitro, enhances the expression of IL-15 and IL-15Rα on dendritic cells (14,21). Type I IFNs, including IFNα and IFNβ, have also been shown to upregulate IL-15 mRNA and protein expression in dendritic cells, monocytes, macrophages, and epithelial cells (14,22,23,24,25). In addition, IFNγ, a type II IFN, can elevate the expression of IL-15 and IL-15Rα in monocytes, endothelial cells, and epithelial cells (12,25,26,27). More recently, in epithelial cells, IFNγ was shown to upregulate the expression of IL-15 and IL-15Rα more potently, promoting the effector functions of NK cells via IL-15 trans-presentation compared to type I IFNs (25). This previous study showed that IFN regulatory factor 1 (IRF1) plays a critical role in IL-15 expression induced by IFNγ and type I IFNs (Fig. 1). This result suggests that the IFNs-IRF1-IL-15 axis may serve as a regulatory target for the treatment of diseases in which the expression of IL-15 is dysregulated.
Figure 1. IFNγ- and type I IFN-mediated IL-15 trans-presentation.
IFNγ and type I IFNs activate distinct signaling pathways leading to the upregulation of IL-15. However, these pathways eventually converge at IRF1. IFNγ signaling primarily depends on STAT1 homodimers for the activation of IRF1. In type I IFN signaling, the ISGF3 complex is formed by a phosphorylated STAT1 and STAT2 heterodimer, which subsequently activates IRF1.
ISGF3, IFN-stimulated gene factor 3.
IL-15 activates three main downstream signaling pathways, including the JAK/STAT, PI3K/AKT/mTOR, and Ras/Raf/MAPK pathways (10). Upon binding to IL-15, the IL-2/15Rβγ receptors recruit and activate JAK1, leading to the subsequent phosphorylation of STAT3 via the β chain (28,29). Simultaneously, JAK3 is recruited to the γ chain, where it phosphorylates STAT5. This cascade of reactions results in the formation of phosphorylated STAT3/STAT5 heterodimers, which subsequently translocate to the nucleus, leading to the activation of anti-apoptotic (Bcl-2 and Mcl-1) and oncogenic (c-Myc) transcription factors (30,31,32). In the second signaling pathway, activated adaptor protein, Shc is recruited to a phosphorylated site on the β chain and triggers phosphorylation of GAB2 through the adaptor Grb2, which in turn activates the PI3K/AKT/mTOR signaling pathway crucial for cell proliferation and survival (33). This pathway leads to accumulation of mTORC1 in the nucleus, which induces E4 promoter-binding protein 4 (E4BP4)-mediated upregulation of Eomes (34,35). Subsequently, Eomes binds to the il2rb promoter, increasing CD122 expression and establishing a positive feedback loop of IL-15 signaling. A recent study demonstrated that IL-15-mediated phosphorylation of AKT accumulates X-box binding protein 1 (XBP1) in the nucleus, where it recruits T-bet to induce the transcription of genes encoding effector molecules, such as IFNγ and granzyme B (36). In addition to the PI3K/AKT/mTOR signaling pathway, IL-15-induced activation of Grb2 also triggers the Ras/Raf/MAPK signaling pathway, which subsequently activates c-Fos, c-Jun, and c-Myc, which are responsible for cell proliferation (37,38).
The IL-15 signaling pathways can be negatively regulated by intracellular checkpoints, namely, suppressor of cytokine signaling (SOCS) family members. STAT5 has been shown to upregulate genes that encode SOCS proteins (39,40). Cytokine-inducible SH2 protein (CIS) is a member of the SOCS family that inhibits the enzymatic activity of JAK1, thereby suppressing JAK-STAT signaling (41). Similarly, SOCS3 suppresses IL-15-induced STAT5 phosphorylation, thereby inhibiting the IL-15 responsiveness of cells (42). Furthermore, zinc fingers and homeoboxes 2 (ZHX2), OTU domain-containing ubiquitin aldehyde-binding protein 1 (OTUB1), tumor necrosis factor-α-induced protein 8-like 2 (TIPE2), and forkhead box protein O1 (FOXO1) have been reported to be transcription repressors that negatively regulate the IL-15-induced transcriptional activity of STAT5 (43), AKT (44), mTOR (45), and T-bet (46), respectively.
IL-15 AND T-CELL RESPONSES
Memory CD8+ T cells
IL-15 has been demonstrated to play a significant role in homeostatic proliferation of memory CD8+ T cells. Previous studies have demonstrated that the absence of IL-15 results in a reduction in the memory CD8+ T-cell population (5,47). Moreover, IL-15 induces the proliferation of stem cell-like memory T cells (TSCM) (48) and prevents attrition of the pre-existing memory CD8+ T-cell population (49). CD8+ T cells are activated upon TCR-mediated recognition of cognate peptides presented by MHC-I (50). Notably, IL-15 promotes TCR-mediated activation of CD8+ T cells by reducing the TCR activation threshold. This was demonstrated by upregulation of TCR-mediated ERK phosphorylation and Nur77 expression in CD8+ T cells in the presence of concurrent IL-15 stimulation (51,52). IL-15 also contributes to TCR-mediated proliferation and IFNγ production by CD8+ T cells (53,54). Correspondingly, IL-15 significantly augments Ag-specific memory CD8+ T-cell responses and provides protective immunity to viral infection (47,55,56,57) and bacterial and parasitic infections (58,59,60).
Senescent CD8+ T cells
Memory CD8+ T cells in an advanced stage of differentiation present senescent-like features, including the expression of senescence-associated cell surface (CD57 and KLRG1) and intracellular (p38 and γH2AX) molecules (61), low telomerase activity (62), and shortened telomeres (63). In addition, senescent CD8+ T cells are hypo-responsive to TCR stimulation and exhibit impaired TCR-mediated proliferation (64,65). On the other hand, compared to their non-senescent counterparts, senescent CD8+ T cells have a greater responsiveness to IL-15. IL-15 induces more robust proliferation in highly differentiated and senescent CD8+ T cells than their less differentiated counterparts (66). Therefore, IL-15 contributes to the generation and expansion of the senescent CD8+ T-cell population (64,67).
Recently, CD5 was shown to function as a negative regulator of the IL-15 response in memory CD8+ T cells (66). The expression of CD5 on human CD8+ T cells progressively decreases during cellular differentiation and senescence (68,69). Consequently, highly differentiated and senescent memory CD8+ T cells with low expression of CD5 exhibit a heightened responsiveness to IL-15, revealing an inverse correlation between the level of CD5 expression and IL-15 responsiveness. Further analysis showed that CD5 directly suppresses the IL-15-induced proliferation of human memory CD8+ T cells by inhibiting mTOR pathways (66).
Upon exposure to IL-15, memory CD8+ T cells typically acquire NK cell-like phenotypes and functions, including the upregulation of various NK receptors (NKRs) and cytotoxic molecules (70,71,72). IL-15 was shown to activate promyelocytic leukemia zinc finger (PLZF) transcription factor, which upregulates NKRs on memory CD8+ T cells (72). Senescent CD8+ T cells are characterized by enhanced NK cell-like functions mediated by NK-activating receptors (73). Sestrins are stress-sensing proteins produced in response to glucose deprivation, oxidative stress, or cellular senescence (74). Sestrins have been shown to promote senescence-associated features in CD8+ T cells, such as down-regulation of TCR-induced intracellular signaling molecules, including LAT, Zap70, and Lck (75). Moreover, sestrins upregulate the expression of NKG2D and its adaptor molecule DAP12, which trigger cytokine secretion and NKG2D-mediated cytotoxicity without TCR stimulation. This indicates that senescent CD8+ T cells are reprogrammed by sestrins as they differentiate and exert NK-like cytotoxic activity. Thus, the poor sensitivity to TCR signals and enhanced responsiveness to IL-15 render senescent CD8+ T cells sensitive to TCR-independent, IL-15-induced activation. This may explain the loss of immunity to previously encountered pathogens, decreased vaccine efficacy, and enhanced immunopathological tissue injury often demonstrated in aged individuals (73). Further work is required to elucidate the molecular mechanisms underlying IL-15 hyper-responsiveness and the resulting NK-like functions of senescent CD8+ T cells.
Virtual memory T cells (TVM)
Memory CD8+ T cells develop from naïve CD8+ T cells after encountering their cognate Ags (50). CD8+ T cells exhibiting memory-like characteristics can also develop during routine T-cell homeostasis, referred to as TVM cells (76). TVM cells originate from naïve CD8+ T cells and acquire memory-like characteristics without prior exposure to Ags (77). The development and maintenance of TVM cells significantly depend on IL-15-mediated stimulation through CD122 (78). The transcription factor Eomes upregulates expression of CD122, which is essential for the TVM cell response to IL-15. Notably, a significant reduction of TVM cells was found in mice with a T cell-specific conditional deletion of Eomes (78). Moreover, IL-15 induces TCR-independent protective immunity mediated by NKG2D and granzyme B expression in TVM cells, as demonstrated in a mouse model of Listeria monocytogenes infection (79).
In humans, the expression of inhibitory NKRs, killer cell immunoglobulin-like receptors (KIRs) and/or NKG2A, on CD8+ T cells defines TVM cells (79,80,81). CD8+ T cells expressing KIR/NKG2A exhibit NK-like effector functions following stimulation with IL-12/IL-18 and/or IL-15, without TCR stimulation (80). The mutually exclusive expression of KIRs or NKG2A on human TVM cells has been identified as an indicator of different functionalities (82). Specifically, KIR+CD8+ T cells express high levels of NKRs, such as 2B4, CD16, CD56, and NKG2C, compared to NKG2A+CD8+ T cells, together with perforin and granzyme B. In addition, KIR+CD8+ T cells exhibit enhanced responsiveness to IL-15 with higher levels of STAT5 phosphorylation compared to NKG2A+CD8+ T cells (81,82). IL-15 stimulation leads to the upregulation of CD107a, perforin, granzyme B, and CD16 and promotes the antibody-dependent cellular cytotoxicity of KIR+CD8+ T cells. These findings demonstrate that the IL-15 responsiveness differs among heterogenous human TVM cells recognized by KIRs or NKG2A.
Tissue-resident memory CD8+ T cells (TRM)
TRM cells are a long-lasting, non-circulating population that establishes residence in the tissue (83). TRM cells have the capacity to rapidly provide on-site immune responses against invading pathogens by not only degranulating perforin and granzyme B, but also producing cytokines, such as IFNγ and TNF, that effectively coordinate both local innate and adaptive immune cells (84,85). TRM cell formation begins with the initial induction of TRM precursor cells into epithelial tissues. The local cytokine environment consists of TGFβ and IL-15, promoting the residence and development of these cells into long-lived TRM cells (86). IL-15 plays a crucial role in preserving TRM cells across diverse tissues, including the skin, liver, salivary glands, lungs, and kidneys (87,88,89,90). However, certain TRM subsets located in non-lymphoid tissues, such as the female reproductive tract, pancreas, and small intestines, can persist without IL-15 (89). Nevertheless, though some TRM populations may not depend on IL-15 for their maintenance, these cells still undergo proliferation in response to IL-15 (91).
NKR-expressing CD8+ T cells
Subpopulations of CD8+ T cells express NKRs such as NKG2C (92). The expansion of NKG2C-expressing CD8+ T cells have been observed in response to various pathological conditions, including cytomegalovirus (CMV) infection, Stevens-Johnson syndrome, toxic epidermal necrolysis, and celiac disease (92). NKG2C+CD8+ T cells expressing high levels of cytotoxic molecules have been shown to effectively lyse target cells upon co-stimulation with anti-CD94 and anti-CD3 Abs (71,93). However, another study reported that, even in the absence of TCR stimulation, the ligation of NKG2C alone can trigger T cells to proliferate and eliminate HLA-E-transfected target cells lacking expression of other MHC-I molecules (94). This suggests that NKG2C signaling may serve as a potential alternative to TCR-mediated activation of CD8+ T cell cytotoxicity.
NKR-expressing CD8+ T cells exist at a higher frequency in the liver than the peripheral blood (95). Recent advancements in multi-omics analysis have investigated the diverse subgroups of liver sinusoidal CD8+ T cells that express NKRs. A specific NK-like CD8+TCRαβ+ liver sinusoidal T-cell population characterized by high expression of CD56 without CD161 expression (CD56hiCD161-CD8+ T cells) was found to express various NKRs, including CD94, KIRs, and NKG2C (96). In addition, this population exhibits hyper-responsiveness to IL-15, IL-12, and IL-18 but weak responsiveness to TCR stimulation. Upon stimulation with IL-15, in addition to both IL-12 and IL-18, these CD56hiCD161-CD8+ T cells expand and exert NK-like effector functions through NKG2D and NKG2C in a TCR-independent manner (96). Further investigation is required to elucidate the precise role and regulatory mechanisms of CD56hiCD161-CD8+ T cells in various types of disease, particularly in relation to IL-15.
Innate-like, unconventional T cells
Beyond its role in conventional CD8+ T cells, IL-15 also plays a critical role in homeostasis and effector functions of innate-like unconventional T cells including mucosal-associated invariant T cells (MAIT) and γδ T cells (97). MAIT cells recognize intermediates of riboflavin (microbial vitamin B2 metabolites) biosynthesis, presented on class I-related molecule 1 (MR1) on Ag-presenting cells, through semi-variant TCRs composed of Vα 7.2-Ja33, 12, or 20 with limited TCR Vβ diversity (98). IL-15 activates mTORC1 and upregulates T-bet, inducing proliferation and maintenance of MAIT cells (99). In addition, IL-15, in combination with IL-12 or IL-18, can activate MAIT cells to produce IFNγ and granzyme B in a STAT5-dependent manner (100,101).
The γδ T cells express lineage-specific γδ TCR and share numerous characteristics with αβ T cells in terms of cytotoxic effector functions and pro-inflammatory cytokine production (102). However, γδ T cells do not rely on MHC molecules. This MHC-independent property involves the recognition of both exogenous and endogenous Ags, encompassing both foreign and self-Ags (103,104). IL-15 plays a pivotal role in proliferation and homeostasis of γδ T cells (105). γδ T cells sensitized by IL-15 were shown to maintain the expression of Mcl-1 following the activation of STAT5 and ERK for promoting cell survival against apoptosis (106). Moreover, IL-15 has been shown to activate γδ T cells and enhance their anti-tumor immune response (102,106,107).
IL-15 PROMOTES CD8+ T CELL-MEDIATED ANTI-TUMOR IMMUNE RESPONSES
IL-15 can enhance the anti-tumor immune response of CD8+ T cells (108). Previous studies in mouse tumor models demonstrated that IL-15 treatment increases the number of tumor-infiltrating CD8+ T cells and their IFNγ production (109,110). In addition, IL-15 has been shown to improve the CD8+ T-cell response to immune checkpoint blockade. IL-15 reinvigorated tumor-infiltrating CD28-PD-1+CD8+ T cells that are unresponsive to PD-1 blockade in non-small cell lung cancer (NSCLC) (111). IL-15 also plays a crucial role in the self-renewal of progenitor exhausted TCF-1+PD-1+CD8+ T cells, which are characterized by stem-like properties and associated with a more favorable response to PD-1 blockade (112).
Various forms of IL-15 have been developed as immunotherapeutic agents and are presently undergoing clinical trials in combination with anti-PD-1 Abs for the treatment of cancer patients (113,114,115). N-803 (formerly known as ALT-803) is a superagonist complex consists of an IL-15 mutant bound to a bivalent IL-15Rα sushi domain and IgG1-Fc fusion protein (116). N-803 has been recognized as a potent inducer of the activation, proliferation and cytotoxicity of CD8+ T cells and NK cells, thereby conferring anti-tumor efficacy in preclinical models including breast cancer (117), colon cancer (116) and glioblastoma (110). N-803 is currently being evaluated in several ongoing clinical trials (118). Results from the recent clinical trials demonstrated that N-803 yielded promising responses with a tolerable safety profile when used in patients with NSCLC (113) and bladder cancer (119). IL-15 has also been integrated in chimeric Ag receptor T cell (CAR-T) engineering for the treatment of cancer through adoptive cell therapy. CAR-T cells expanded ex vivo with IL-15 exhibit a less differentiated phenotype, reduced expression of exhaustion and pro-apoptotic molecules, and enhanced mitochondrial metabolism, with administration resulting in a stronger anti-tumor response (120,121,122,123).
IL-15 has been shown to activate tumor-infiltrating bystander memory CD8+ T cells. A considerable portion of memory CD8+ T cells present within tumor infiltrates is specific for tumor-unrelated viruses, such as CMV and Epstein-Barr virus (EBV) (124). Though tumor-specific CD8+ T cells typically express CD39, bystander CD8+ T cells within tumor tissues lack CD39 expression. IL-15-induced activation of memory CD8+ T cells has been shown to contribute to the anti-tumor immune response via an NKG2D-dependent mechanism, enhancing tumor control even in the absence of cognate Ag recognition (125).
IL-15-INDUCED T CELL-MEDIATED IMMUNOPATHOGENESIS
Viral infection
IL-15 has been shown to activate memory CD8+ T cells to proliferate and exert effector functions in the absence of TCR stimulation (126). During viral infection, the upregulation of IL-15 triggers bystander activation of memory CD8+ T cells (8,127). IL-15 induces polyclonal expansion of bystander memory CD8+ T cells with a highly diverse TCR repertoire (128). Though several studies have reported a protective effect (129,130,131), a significant number of studies have demonstrated a detrimental effect of IL-15-induced bystander activation of memory CD8+ T cells, leading to host tissue damage during microbial infections (95,132,133,134,135,136,137).
The pathological contribution of IL-15-drvien bystander-activated memory CD8+ T cells is well described in acute hepatitis A virus (HAV) infection (Fig. 2). In adults, acute HAV infection often causes severe liver injury (138). During acute HAV infection, independent of TCR, elevated IL-15 activates pre-existing memory CD8+ T cells specific for HAV-unrelated viruses, such as human CMV, EBV, influenza A virus (IAV), respiratory syncytial virus, and vaccinia virus (133). These bystander-activated memory CD8+ T cells express increased levels of NK cell-activating receptors (NKG2D and NKp30) and cytotoxic molecules (perforin and granzyme B), as well as activation markers (CD38 and HLA-DR) and proliferation marker Ki-67. In addition, IL-15 upregulates chemokine receptor CCR5 via ERK signaling pathway, facilitating the migration of bystander-activated memory CD8+ T cells to the infected liver during acute HAV infection (134). This finding aligns with previous studies indicating that IL-15 promotes the migration of memory CD8+ T cells to the infection site without antigenic stimulation (139,140,141). In the HAV-infected liver, bystander-activated memory CD8+ T cells exert NKG2D-dependent NK-like cytotoxicity and kill hepatocytes expressing NKG2D ligands (133). This NK-like cytotoxicity significantly correlates with liver damage in patients with acute HAV infection, indicating the presence of IL-15-induced immunopathological mechanisms performed by bystander-activated memory CD8+ T cells. Similarly, this NK-like cytotoxic attribute driven by IL-15 has been also observed in MAIT cells (142), the most prevalent innate-like T cells in human liver (100). In the absence of a TCR-MR1 interaction, the combination of IL-15 with IL-12 and IL-18 upregulates of NKG2D, granzyme B, and CD2 in a PI3K-mTOR signaling-dependent manner (142). Subsequently, MAIT cells kill hepatocytes through NK-like cytotoxicity which is associated with severe liver damage in patients with acute HAV infection.
Figure 2. Bystander-activated memory CD8+ T cells induced by IL-15 contribute to liver damage during acute HAV infection.
During acute HAV infection, elevated IL-15 activates pre-existing memory CD8+ T cells that are specific for HAV-unrelated viruses, independent of TCR engagement. This IL-15-induced bystander activation of memory CD8+ T cells occurs both outside the liver and in the areas surrounding infected hepatocytes (left). These bystander-activated memory CD8+ T cells express upregulated NKG2D and CCR5. Bystander-activated memory CD8+ T cells migrate into the HAV-infected liver via a CCR5-dependent mechanism (right). Within the HAV-infected liver, NKG2D triggers NK-like cytotoxicity against hepatocytes expressing NKG2D ligands. Notably, in patients with acute HAV infection, the NK-like cytotoxicity of bystander-activated memory CD8+ T cells significantly correlates with liver injury, as reflected by the level of ALT in serum.
ALT, alanine transaminase.
Importantly, though IL-15 upregulates surface expression of NKG2D and CCR5 on memory CD8+ T cells, TCR stimulation has no impact on these markers (133,134). Interestingly, concurrent TCR stimulation abrogates the IL-15-induced upregulation of NKG2D and CCR5. Therefore, the upregulation of NKG2D and CCR5 serves as a marker of IL-15-induced bystander activation of memory CD8+ T cells (143). Consistent with this, IFN-induced transmembrane protein 3 (IFITM3) has been recognized as an indicator of IL-15-induced bystander activation of memory CD8+ T cells in a murine model of IAV infection (144). This suggests that IL-15 promotes innate-like features, whereas TCR stimulation is responsible for preserving the intrinsic adaptive nature of memory CD8+ T cells. However, the regulatory mechanisms underlying IL-15-induced bystander activation in memory CD8+ T cells have not been fully elucidated yet.
Chronic inflammatory diseases
Overexpression of IL-15 is associated with pathology in various chronic inflammatory diseases (7). The pathological contribution of TCR-independent, IL-15-induced upregulation of NKG2D on CD8+TCRαβ+ intraepithelial lymphocytes (IELs) has been demonstrated in celiac disease (145). In celiac disease, intestinal epithelial cells overexpress IL-15 and the NKG2D ligand MHC class I polypeptide-related protein (MICA) (146). Dysregulated IL-15 production upregulates NKG2D on CD8+TCRαβ+ IELs in the absence of TCR stimulation, and they contribute to intestinal-tissue damage through NK-like killing activity (145).
The intestinal mucosa serves as a primary site for extrathymic lymphopoiesis of γδ T cells, often referred to as γδ intestinal IELs (147). The homeostatic proliferation of γδ IELs is mainly induced by the IL-15 produced by adjacent epithelial cells (148). γδ IELs exhibit NK-like features, responding rapidly and consistently to tissue alarmins and stress-induced ligands of NKRs without requiring Ag recognition (149). Although γδ IELs play a critical role in mucosal protection, dysregulated activation of γδ IELs can exacerbate inflammation and contribute to the progression of intestinal diseases (150,151). Interestingly, the Ikaros zinc finger (IKZF) transcription factor Aiolos has been identified as a regulator that suppresses expression of NKRs, cytotoxic molecules, and chemokines in γδ IELs (9). This Aiolos-induced regulatory mechanism involves a partial attenuation of IL-15 signaling in γδ IELs and has been shown to ameliorate colitis in a mouse model. As unrestricted IL-15 production can lead to T cell-mediated tissue damage, the Aiolos-mediated regulation of IL-15 signaling is crucial for maintaining intestinal homeostasis. Further investigation into other regulatory factors suppressing IL-15 signaling, as well as the expression of NKRs and cytotoxic mediators (43,44,45,46,66) that may contribute to tissue damage and disease pathogenesis, is warranted. These could serve as promising therapeutic targets for alleviating IL-15-induced T cell-mediated immunopathological tissue damage.
The TCR-independent, IL-15-induced NK-like cytotoxicity of effector memory CD8+ T cells was also shown to contribute to the pathogenesis of alopecia areata (152). IL-15 was shown to expand TVM cells expressing remarkably high levels of CD44 and lacking CD49d (CD44s-hiCD49dlo), which cause alopecia areata in a mouse model (153). IL-15 upregulated NKG2D and cytotoxic molecules in CD44s-hiCD49dlo TVM cells and facilitated their migration to the cutaneous tissues. Here, the local expression of IL-15, IL-12, and IL-18, as well as NKG2D ligands, leads to the damage of hair follicles by CD44s-hiCD49dlo TVM cells through their NKG2D-dependent killing activity. This finding was further supported by effectively controlling disease progression through the administration of blocking Abs targeting CD122, IL-12, and IL-18 or NKG2D in the mouse model (153).
Interestingly, a study reported that germline STAT3 gain-of-function mutations in mice induce CD122-dependent expansion of CD8+ T cells, which contribute to the autoimmune-like pathology (154). These expanded CD8+ T cells were characterized by the upregulation of NKG2D and effector molecules (IFNγ, granzymes, and perforin), and their accumulation significantly correlated with the development of autoimmune-like pathology.
In non-alcoholic steatohepatitis (NASH), IL-15 drives hepatic accumulation of CXCR6+CD8+ T cells, which cause liver damage (155). During NASH, uncontrolled production of IL-15 in the liver down-regulates FOXO1, rendering liver-resident CXCR6+CD8+ T cells susceptible to metabolic stimuli, including acetate and extracellular ATP. Upon exposure to acetate, liver-resident CXCR6+CD8+ T cells upregulate granzyme B expression and TNF production to become auto-aggressive CD8+ T cells. With subsequent exposure to extracellular ATP, which signals through P2X7 purinergic receptor, auto-aggressive CD8+ T cells kill hepatocytes in an MHC-I-independent, Fas ligand-dependent manner, contributing to the liver damage in NASH (155). Further research to elucidate the mechanisms underlying uncontrolled IL-15 production and how IL-15 reprograms T cells to acquire pathological functions could support the identification of therapeutic targets for ameliorating T cell-mediated tissue injury.
CONCLUSION
In summary, IL-15 is essential for the homeostasis and effector functions of T cells. However, dysregulated IL-15 production can trigger TCR-independent, NK-like activation of T cells that contributes to immunopathological tissue damage (Fig. 3). Although significant progress has been made in demonstrating the pathological contribution of IL-15, further investigations are required to explore T cell-mediated immunopathogenesis in various diseases, encompassing those not yet thoroughly examined. Moreover, it is necessary to determine the specific types of T cells that exhibit the most pronounced responsiveness to IL-15 and play a significant role in inducing immunopathology in each distinct disease. Furthermore, the molecular mechanisms underlying IL-15-induced T cell-mediated immunopathogenesis remain poorly understood. Therefore, it is crucial to gain further insights into the regulatory mechanisms involved in IL-15-induced T-cell activation. These insights will shed light on therapeutic targets to alleviate immunopathology observed in various infectious diseases and chronic inflammatory diseases.
Figure 3. IL-15-induced T cell-mediated immunopathogenesis in various diseases.
Dysregulated production of IL-15 triggers TCR-independent and NK-like cytotoxicity of T cells that contributes to immunopathological tissuse damage in various diseases. IL-15 activates bystander memory CD8+ T cells to kill hepatocytes in an NKG2D-dependent manner, leading to severe liver injury during acute HAV infection. This IL-15-induced immunopathological liver injury was also induced by MR1-indpendent activation of MAIT cells during acute HAV infection. IL-15 activates intraepithelial intestinal CD8+ T cells independenly of TCR signaling to exert inestinal tissue damage through NKG2D-dependent cytotoxicity during celiac disease. IL-15 activates CD44s-hiCD49dlo TVM cells to damage hair follicle cells through NKG2D-dependent cytotoxicity, contributing to alopecia areata pathogenesis. IL-15 upregulates NK-like feuatures of γδ intestinal intraepithelial lymphocytes that contribute to the pathogenesis of colitis. IL-15 drives hepatic accumulation of CXCR6+CD8+ auto-aggressive T cells, which contribute to liver damage by killing hepatocytes in an MHC-I-independent and Fas ligand-dependent manner during NASH.
ACKNOWLEDGEMENTS
This work was supported by the Institute for Basic Science (IBS), Republic of Korea, under project code IBS-R801-D2.
Abbreviations
- CAR-T
chimeric Ag receptor T cell
- CD44s-hiCD49dlo
CD44 super-high CD49d low
- CMV
cytomegalovirus
- EBV
epstein-Barr virus
- FOXO1
forkhead box O1
- γc
common gamma chain
- HAV
hepatitis A virus
- IAV
influenza A virus
- IEL
intraepithelial lymphocyte
- IL-15Rα
IL-15 receptor alpha
- IRF1
IFN regulatory factor 1
- KIR
killer cell immunoglobulin-like receptor
- MAIT
mucosal-associated invariant T cells
- MR1
class I-related molecule 1
- NASH
non-alcoholic steatohepatitis
- NKR
NK receptor
- NSCLC
non-small cell lung cancer
- SOCS
suppressor of cytokine signaling
- TRM
tissue resident memory CD8+ T cells
- TSCM
stem cell-like memory T cells
- TVM
virtual memory T cells
Footnotes
Conflict of Interest: The authors declare no potential conflicts of interest.
- Conceptualization: Lee H, Park SH, Shin EC.
- Data curation: Lee H, Park SH, Shin EC.
- Investigation: Lee H.
- Methodology: Lee H, Park SH, Shin EC.
- Supervision: Shin EC.
- Writing - original draft: Lee H, Shin EC.
- Writing - review & editing: Lee H, Park SH, Shin EC.
References
- 1.Grabstein KH, Eisenman J, Shanebeck K, Rauch C, Srinivasan S, Fung V, Beers C, Richardson J, Schoenborn MA, Ahdieh M, et al. Cloning of a T cell growth factor that interacts with the beta chain of the interleukin-2 receptor. Science. 1994;264:965–968. doi: 10.1126/science.8178155. [DOI] [PubMed] [Google Scholar]
- 2.Giri JG, Ahdieh M, Eisenman J, Shanebeck K, Grabstein K, Kumaki S, Namen A, Park LS, Cosman D, Anderson D. Utilization of the beta and gamma chains of the IL-2 receptor by the novel cytokine IL-15. EMBO J. 1994;13:2822–2830. doi: 10.1002/j.1460-2075.1994.tb06576.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Leonard WJ, Lin JX, O’Shea JJ. The gamma(c) family of cytokines: basic biology to therapeutic ramifications. Immunity. 2019;50:832–850. doi: 10.1016/j.immuni.2019.03.028. [DOI] [PubMed] [Google Scholar]
- 4.Bamford RN, Grant AJ, Burton JD, Peters C, Kurys G, Goldman CK, Brennan J, Roessler E, Waldmann TA. The interleukin (IL) 2 receptor beta chain is shared by IL-2 and a cytokine, provisionally designated IL-T, that stimulates T-cell proliferation and the induction of lymphokine-activated killer cells. Proc Natl Acad Sci U S A. 1994;91:4940–4944. doi: 10.1073/pnas.91.11.4940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kennedy MK, Glaccum M, Brown SN, Butz EA, Viney JL, Embers M, Matsuki N, Charrier K, Sedger L, Willis CR, et al. Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice. J Exp Med. 2000;191:771–780. doi: 10.1084/jem.191.5.771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hu Q, Ye X, Qu X, Cui D, Zhang L, Xu Z, Wan H, Zhang L, Tao W. Discovery of a novel IL-15 based protein with improved developability and efficacy for cancer immunotherapy. Sci Rep. 2018;8:7675. doi: 10.1038/s41598-018-25987-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Jabri B, Abadie V. IL-15 functions as a danger signal to regulate tissue-resident T cells and tissue destruction. Nat Rev Immunol. 2015;15:771–783. doi: 10.1038/nri3919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lee H, Jeong S, Shin EC. Significance of bystander T cell activation in microbial infection. Nat Immunol. 2022;23:13–22. doi: 10.1038/s41590-021-00985-3. [DOI] [PubMed] [Google Scholar]
- 9.Yomogida K, Trsan T, Sudan R, Rodrigues PF, Ulezko Antonova A, Ingle H, Luccia BD, Collins PL, Cella M, Gilfillan S, et al. The transcription factor Aiolos restrains the activation of intestinal intraepithelial lymphocytes. Nat Immunol. 2024;25:77–87. doi: 10.1038/s41590-023-01693-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ma S, Caligiuri MA, Yu J. Harnessing IL-15 signaling to potentiate NK cell-mediated cancer immunotherapy. Trends Immunol. 2022;43:833–847. doi: 10.1016/j.it.2022.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Quinn LS, Haugk KL, Grabstein KH. Interleukin-15: a novel anabolic cytokine for skeletal muscle. Endocrinology. 1995;136:3669–3672. doi: 10.1210/endo.136.8.7628408. [DOI] [PubMed] [Google Scholar]
- 12.Musso T, Calosso L, Zucca M, Millesimo M, Ravarino D, Giovarelli M, Malavasi F, Ponzi AN, Paus R, Bulfone-Paus S. Human monocytes constitutively express membrane-bound, biologically active, and interferon-gamma-upregulated interleukin-15. Blood. 1999;93:3531–3539. [PubMed] [Google Scholar]
- 13.Rückert R, Asadullah K, Seifert M, Budagian VM, Arnold R, Trombotto C, Paus R, Bulfone-Paus S. Inhibition of keratinocyte apoptosis by IL-15: a new parameter in the pathogenesis of psoriasis? J Immunol. 2000;165:2240–2250. doi: 10.4049/jimmunol.165.4.2240. [DOI] [PubMed] [Google Scholar]
- 14.Mattei F, Schiavoni G, Belardelli F, Tough DF. IL-15 is expressed by dendritic cells in response to type I IFN, double-stranded RNA, or lipopolysaccharide and promotes dendritic cell activation. J Immunol. 2001;167:1179–1187. doi: 10.4049/jimmunol.167.3.1179. [DOI] [PubMed] [Google Scholar]
- 15.Shinozaki M, Hirahashi J, Lebedeva T, Liew FY, Salant DJ, Maron R, Kelley VR. IL-15, a survival factor for kidney epithelial cells, counteracts apoptosis and inflammation during nephritis. J Clin Invest. 2002;109:951–960. doi: 10.1172/JCI14574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Reinecker HC, MacDermott RP, Mirau S, Dignass A, Podolsky DK. Intestinal epithelial cells both express and respond to interleukin 15. Gastroenterology. 1996;111:1706–1713. doi: 10.1016/s0016-5085(96)70036-7. [DOI] [PubMed] [Google Scholar]
- 17.Dubois S, Mariner J, Waldmann TA, Tagaya Y. IL-15Ralpha recycles and presents IL-15 In trans to neighboring cells. Immunity. 2002;17:537–547. doi: 10.1016/s1074-7613(02)00429-6. [DOI] [PubMed] [Google Scholar]
- 18.Mortier E, Woo T, Advincula R, Gozalo S, Ma A. IL-15Ralpha chaperones IL-15 to stable dendritic cell membrane complexes that activate NK cells via trans presentation. J Exp Med. 2008;205:1213–1225. doi: 10.1084/jem.20071913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ota N, Takase M, Uchiyama H, Olsen SK, Kanagawa O. No requirement of trans presentations of IL-15 for human CD8 T cell proliferation. J Immunol. 2010;185:6041–6048. doi: 10.4049/jimmunol.0901834. [DOI] [PubMed] [Google Scholar]
- 20.Bergamaschi C, Bear J, Rosati M, Beach RK, Alicea C, Sowder R, Chertova E, Rosenberg SA, Felber BK, Pavlakis GN. Circulating IL-15 exists as heterodimeric complex with soluble IL-15Rα in human and mouse serum. Blood. 2012;120:e1–e8. doi: 10.1182/blood-2011-10-384362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lucas M, Schachterle W, Oberle K, Aichele P, Diefenbach A. Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity. 2007;26:503–517. doi: 10.1016/j.immuni.2007.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Colpitts SL, Stoklasek TA, Plumlee CR, Obar JJ, Guo C, Lefrançois L. Cutting edge: the role of IFN-α receptor and MyD88 signaling in induction of IL-15 expression in vivo. J Immunol. 2012;188:2483–2487. doi: 10.4049/jimmunol.1103609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Colpitts SL, Stonier SW, Stoklasek TA, Root SH, Aguila HL, Schluns KS, Lefrançois L. Transcriptional regulation of IL-15 expression during hematopoiesis. J Immunol. 2013;191:3017–3024. doi: 10.4049/jimmunol.1301389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Domínguez-Andrés J, Feo-Lucas L, Minguito de la Escalera M, González L, López-Bravo M, Ardavín C. Inflammatory Ly6C(high) monocytes protect against candidiasis through IL-15-driven NK cell/neutrophil activation. Immunity. 2017;46:1059–1072.e4. doi: 10.1016/j.immuni.2017.05.009. [DOI] [PubMed] [Google Scholar]
- 25.Kim TS, Rha MS, Shin EC. IFN-gamma induces IL-15 trans-presentation by epithelial cells via IRF1. J Immunol. 2022;208:338–346. doi: 10.4049/jimmunol.2100057. [DOI] [PubMed] [Google Scholar]
- 26.Lee N, Shin MS, Kang KS, Yoo SA, Mohanty S, Montgomery RR, Shaw AC, Kang I. Human monocytes have increased IFN-γ-mediated IL-15 production with age alongside altered IFN-γ receptor signaling. Clin Immunol. 2014;152:101–110. doi: 10.1016/j.clim.2014.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Xie CB, Jiang B, Qin L, Tellides G, Kirkiles-Smith NC, Jane-Wit D, Pober JS. Complement-activated interferon-γ-primed human endothelium transpresents interleukin-15 to CD8+ T cells. J Clin Invest. 2020;130:3437–3452. doi: 10.1172/JCI135060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Miyazaki T, Kawahara A, Fujii H, Nakagawa Y, Minami Y, Liu ZJ, Oishi I, Silvennoinen O, Witthuhn BA, Ihle JN, et al. Functional activation of Jak1 and Jak3 by selective association with IL-2 receptor subunits. Science. 1994;266:1045–1047. doi: 10.1126/science.7973659. [DOI] [PubMed] [Google Scholar]
- 29.Lin JX, Migone TS, Tsang M, Friedmann M, Weatherbee JA, Zhou L, Yamauchi A, Bloom ET, Mietz J, John S, et al. The role of shared receptor motifs and common Stat proteins in the generation of cytokine pleiotropy and redundancy by IL-2, IL-4, IL-7, IL-13, and IL-15. Immunity. 1995;2:331–339. doi: 10.1016/1074-7613(95)90141-8. [DOI] [PubMed] [Google Scholar]
- 30.Carson WE, Fehniger TA, Haldar S, Eckhert K, Lindemann MJ, Lai CF, Croce CM, Baumann H, Caligiuri MA. A potential role for interleukin-15 in the regulation of human natural killer cell survival. J Clin Invest. 1997;99:937–943. doi: 10.1172/JCI119258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lord JD, McIntosh BC, Greenberg PD, Nelson BH. The IL-2 receptor promotes lymphocyte proliferation and induction of the c-myc, bcl-2, and bcl-x genes through the trans-activation domain of Stat5. J Immunol. 2000;164:2533–2541. doi: 10.4049/jimmunol.164.5.2533. [DOI] [PubMed] [Google Scholar]
- 32.Huntington ND, Puthalakath H, Gunn P, Naik E, Michalak EM, Smyth MJ, Tabarias H, Degli-Esposti MA, Dewson G, Willis SN, et al. Interleukin 15-mediated survival of natural killer cells is determined by interactions among Bim, Noxa and Mcl-1. Nat Immunol. 2007;8:856–863. doi: 10.1038/ni1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gu H, Maeda H, Moon JJ, Lord JD, Yoakim M, Nelson BH, Neel BG. New role for Shc in activation of the phosphatidylinositol 3-kinase/Akt pathway. Mol Cell Biol. 2000;20:7109–7120. doi: 10.1128/mcb.20.19.7109-7120.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Intlekofer AM, Takemoto N, Wherry EJ, Longworth SA, Northrup JT, Palanivel VR, Mullen AC, Gasink CR, Kaech SM, Miller JD, et al. Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nat Immunol. 2005;6:1236–1244. doi: 10.1038/ni1268. [DOI] [PubMed] [Google Scholar]
- 35.Li D, Wang Y, Yang M, Dong Z. mTORC1 and mTORC2 coordinate early NK cell development by differentially inducing E4BP4 and T-bet. Cell Death Differ. 2021;28:1900–1909. doi: 10.1038/s41418-020-00715-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang Y, Zhang Y, Yi P, Dong W, Nalin AP, Zhang J, Zhu Z, Chen L, Benson DM, Mundy-Bosse BL, et al. The IL-15-AKT-XBP1s signaling pathway contributes to effector functions and survival in human NK cells. Nat Immunol. 2019;20:10–17. doi: 10.1038/s41590-018-0265-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Miyazaki T, Liu ZJ, Kawahara A, Minami Y, Yamada K, Tsujimoto Y, Barsoumian EL, Permutter RM, Taniguchi T. Three distinct IL-2 signaling pathways mediated by bcl-2, c-myc, and lck cooperate in hematopoietic cell proliferation. Cell. 1995;81:223–231. doi: 10.1016/0092-8674(95)90332-1. [DOI] [PubMed] [Google Scholar]
- 38.Bianchi T, Gasser S, Trumpp A, MacDonald HR. c-Myc acts downstream of IL-15 in the regulation of memory CD8 T-cell homeostasis. Blood. 2006;107:3992–3999. doi: 10.1182/blood-2005-09-3851. [DOI] [PubMed] [Google Scholar]
- 39.Lin JX, Li P, Liu D, Jin HT, He J, Ata Ur Rasheed M, Rochman Y, Wang L, Cui K, Liu C, et al. Critical role of STAT5 transcription factor tetramerization for cytokine responses and normal immune function. Immunity. 2012;36:586–599. doi: 10.1016/j.immuni.2012.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lin JX, Du N, Li P, Kazemian M, Gebregiorgis T, Spolski R, Leonard WJ. Critical functions for STAT5 tetramers in the maturation and survival of natural killer cells. Nat Commun. 2017;8:1320. doi: 10.1038/s41467-017-01477-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Delconte RB, Kolesnik TB, Dagley LF, Rautela J, Shi W, Putz EM, Stannard K, Zhang JG, Teh C, Firth M, et al. CIS is a potent checkpoint in NK cell-mediated tumor immunity. Nat Immunol. 2016;17:816–824. doi: 10.1038/ni.3470. [DOI] [PubMed] [Google Scholar]
- 42.Wang X, Sun R, Hao X, Lian ZX, Wei H, Tian Z. IL-17 constrains natural killer cell activity by restraining IL-15-driven cell maturation via SOCS3. Proc Natl Acad Sci U S A. 2019;116:17409–17418. doi: 10.1073/pnas.1904125116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tan S, Guo X, Li M, Wang T, Wang Z, Li C, Wu Z, Li N, Gao L, Liang X, et al. Transcription factor Zhx2 restricts NK cell maturation and suppresses their antitumor immunity. J Exp Med. 2021;218:e20210009. doi: 10.1084/jem.20210009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhou X, Yu J, Cheng X, Zhao B, Manyam GC, Zhang L, Schluns K, Li P, Wang J, Sun SC. The deubiquitinase Otub1 controls the activation of CD8+ T cells and NK cells by regulating IL-15-mediated priming. Nat Immunol. 2019;20:879–889. doi: 10.1038/s41590-019-0405-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bi J, Cheng C, Zheng C, Huang C, Zheng X, Wan X, Chen YH, Tian Z, Sun H. TIPE2 is a checkpoint of natural killer cell maturation and antitumor immunity. Sci Adv. 2021;7:eabi6515. doi: 10.1126/sciadv.abi6515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Deng Y, Kerdiles Y, Chu J, Yuan S, Wang Y, Chen X, Mao H, Zhang L, Zhang J, Hughes T, et al. Transcription factor Foxo1 is a negative regulator of natural killer cell maturation and function. Immunity. 2015;42:457–470. doi: 10.1016/j.immuni.2015.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Becker TC, Wherry EJ, Boone D, Murali-Krishna K, Antia R, Ma A, Ahmed R. Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells. J Exp Med. 2002;195:1541–1548. doi: 10.1084/jem.20020369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, Almeida JR, Gostick E, Yu Z, Carpenito C, et al. A human memory T cell subset with stem cell-like properties. Nat Med. 2011;17:1290–1297. doi: 10.1038/nm.2446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yajima T, Yoshihara K, Nakazato K, Kumabe S, Koyasu S, Sad S, Shen H, Kuwano H, Yoshikai Y. IL-15 regulates CD8+ T cell contraction during primary infection. J Immunol. 2006;176:507–515. doi: 10.4049/jimmunol.176.1.507. [DOI] [PubMed] [Google Scholar]
- 50.Kaech SM, Wherry EJ, Ahmed R. Effector and memory T-cell differentiation: implications for vaccine development. Nat Rev Immunol. 2002;2:251–262. doi: 10.1038/nri778. [DOI] [PubMed] [Google Scholar]
- 51.Deshpande P, Cavanagh MM, Le Saux S, Singh K, Weyand CM, Goronzy JJ. IL-7- and IL-15-mediated TCR sensitization enables T cell responses to self-antigens. J Immunol. 2013;190:1416–1423. doi: 10.4049/jimmunol.1201620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Banerjee A, Li D, Guo Y, Mei Z, Lau C, Chen K, Westwick J, Klauda JB, Schrum A, Lazear ER, et al. A reengineered common chain cytokine augments CD8+ T cell-dependent immunotherapy. JCI Insight. 2022;7:e158889. doi: 10.1172/jci.insight.158889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Richer MJ, Pewe LL, Hancox LS, Hartwig SM, Varga SM, Harty JT. Inflammatory IL-15 is required for optimal memory T cell responses. J Clin Invest. 2015;125:3477–3490. doi: 10.1172/JCI81261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Setoguchi R. IL-15 boosts the function and migration of human terminally differentiated CD8+ T cells by inducing a unique gene signature. Int Immunol. 2016;28:293–305. doi: 10.1093/intimm/dxw004. [DOI] [PubMed] [Google Scholar]
- 55.Sharif-Askari E, Fawaz LM, Tran P, Ahmad A, Menezes J. Interleukin 15-mediated induction of cytotoxic effector cells capable of eliminating Epstein-Barr virus-transformed/immortalized lymphocytes in culture. J Natl Cancer Inst. 2001;93:1724–1732. doi: 10.1093/jnci/93.22.1724. [DOI] [PubMed] [Google Scholar]
- 56.Schluns KS, Williams K, Ma A, Zheng XX, Lefrançois L. Cutting edge: requirement for IL-15 in the generation of primary and memory antigen-specific CD8 T cells. J Immunol. 2002;168:4827–4831. doi: 10.4049/jimmunol.168.10.4827. [DOI] [PubMed] [Google Scholar]
- 57.Mueller YM, Bojczuk PM, Halstead ES, Kim AH, Witek J, Altman JD, Katsikis PD. IL-15 enhances survival and function of HIV-specific CD8+ T cells. Blood. 2003;101:1024–1029. doi: 10.1182/blood-2002-07-1957. [DOI] [PubMed] [Google Scholar]
- 58.Saito K, Yajima T, Kumabe S, Doi T, Yamada H, Sad S, Shen H, Yoshikai Y. Impaired protection against Mycobacterium bovis bacillus Calmette-Guerin infection in IL-15-deficient mice. J Immunol. 2006;176:2496–2504. doi: 10.4049/jimmunol.176.4.2496. [DOI] [PubMed] [Google Scholar]
- 59.Yajima T, Nishimura H, Ishimitsu R, Yamamura K, Watase T, Busch DH, Pamer EG, Kuwano H, Yoshikai Y. Memory phenotype CD8(+) T cells in IL-15 transgenic mice are involved in early protection against a primary infection with Listeria monocytogenes. Eur J Immunol. 2001;31:757–766. doi: 10.1002/1521-4141(200103)31:3<757::aid-immu757>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
- 60.Khan IA, Moretto M, Wei XQ, Williams M, Schwartzman JD, Liew FY. Treatment with soluble interleukin-15Ralpha exacerbates intracellular parasitic infection by blocking the development of memory CD8+ T cell response. J Exp Med. 2002;195:1463–1470. doi: 10.1084/jem.20011915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Henson SM, Lanna A, Riddell NE, Franzese O, Macaulay R, Griffiths SJ, Puleston DJ, Watson AS, Simon AK, Tooze SA, et al. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8+ T cells. J Clin Invest. 2014;124:4004–4016. doi: 10.1172/JCI75051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Plunkett FJ, Franzese O, Finney HM, Fletcher JM, Belaramani LL, Salmon M, Dokal I, Webster D, Lawson AD, Akbar AN. The loss of telomerase activity in highly differentiated CD8+CD28-CD27- T cells is associated with decreased Akt (Ser473) phosphorylation. J Immunol. 2007;178:7710–7719. doi: 10.4049/jimmunol.178.12.7710. [DOI] [PubMed] [Google Scholar]
- 63.Romero P, Zippelius A, Kurth I, Pittet MJ, Touvrey C, Iancu EM, Corthesy P, Devevre E, Speiser DE, Rufer N. Four functionally distinct populations of human effector-memory CD8+ T lymphocytes. J Immunol. 2007;178:4112–4119. doi: 10.4049/jimmunol.178.7.4112. [DOI] [PubMed] [Google Scholar]
- 64.Chiu WK, Fann M, Weng NP. Generation and growth of CD28nullCD8+ memory T cells mediated by IL-15 and its induced cytokines. J Immunol. 2006;177:7802–7810. doi: 10.4049/jimmunol.177.11.7802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Goronzy JJ, Li G, Yu M, Weyand CM. Signaling pathways in aged T cells - a reflection of T cell differentiation, cell senescence and host environment. Semin Immunol. 2012;24:365–372. doi: 10.1016/j.smim.2012.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Choi YJ, Lee H, Kim JH, Kim SY, Koh JY, Sa M, Park SH, Shin EC. CD5 suppresses IL-15-induced proliferation of human memory CD8(+) T cells by inhibiting mTOR pathways. J Immunol. 2022;209:1108–1117. doi: 10.4049/jimmunol.2100854. [DOI] [PubMed] [Google Scholar]
- 67.Morris SR, Chen B, Mudd JC, Panigrahi S, Shive CL, Sieg SF, Cameron CM, Zidar DA, Funderburg NT, Younes SA, et al. Inflammescent CX3CR1+CD57+CD8+ T cells are generated and expanded by IL-15. JCI Insight. 2020;5:e132963. doi: 10.1172/jci.insight.132963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Herndler-Brandstetter D, Brunner S, Weiskopf D, van Rijn R, Landgraf K, Dejaco C, Duftner C, Schirmer M, Kloss F, Gassner R, et al. Post-thymic regulation of CD5 levels in human memory T cells is inversely associated with the strength of responsiveness to interleukin-15. Hum Immunol. 2011;72:627–631. doi: 10.1016/j.humimm.2011.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kim YJ, Rho KN, Jeong S, Lee GW, Kim HO, Cho HJ, Bae WK, Oh IJ, Lee SW, Cho JH. CD5 expression dynamically changes during the differentiation of human CD8(+) T cells predicting clinical response to immunotherapy. Immune Netw. 2023;23:e35. doi: 10.4110/in.2023.23.e35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Correia MP, Costa AV, Uhrberg M, Cardoso EM, Arosa FA. IL-15 induces CD8+ T cells to acquire functional NK receptors capable of modulating cytotoxicity and cytokine secretion. Immunobiology. 2011;216:604–612. doi: 10.1016/j.imbio.2010.09.012. [DOI] [PubMed] [Google Scholar]
- 71.Balin SJ, Pellegrini M, Klechevsky E, Won ST, Weiss DI, Choi AW, Hakimian J, Lu J, Ochoa MT, Bloom BR, et al. Human antimicrobial cytotoxic T lymphocytes, defined by NK receptors and antimicrobial proteins, kill intracellular bacteria. Sci Immunol. 2018;3:eaat7668. doi: 10.1126/sciimmunol.aat7668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Correia MP, Stojanovic A, Bauer K, Juraeva D, Tykocinski LO, Lorenz HM, Brors B, Cerwenka A. Distinct human circulating NKp30+FcεRIγ+CD8+ T cell population exhibiting high natural killer-like antitumor potential. Proc Natl Acad Sci U S A. 2018;115:E5980–E5989. doi: 10.1073/pnas.1720564115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Laphanuwat P, Gomes DCO, Akbar AN. Senescent T cells: beneficial and detrimental roles. Immunol Rev. 2023;316:160–175. doi: 10.1111/imr.13206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lanna A, Gomes DC, Muller-Durovic B, McDonnell T, Escors D, Gilroy DW, Lee JH, Karin M, Akbar AN. A sestrin-dependent Erk-Jnk-p38 MAPK activation complex inhibits immunity during aging. Nat Immunol. 2017;18:354–363. doi: 10.1038/ni.3665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Pereira BI, De Maeyer RPH, Covre LP, Nehar-Belaid D, Lanna A, Ward S, Marches R, Chambers ES, Gomes DCO, Riddell NE, et al. Sestrins induce natural killer function in senescent-like CD8+ T cells. Nat Immunol. 2020;21:684–694. doi: 10.1038/s41590-020-0643-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Seok J, Cho SD, Seo SJ, Park SH. Roles of virtual memory T cells in diseases. Immune Netw. 2023;23:e11. doi: 10.4110/in.2023.23.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.White JT, Cross EW, Kedl RM. Antigen-inexperienced memory CD8+ T cells: where they come from and why we need them. Nat Rev Immunol. 2017;17:391–400. doi: 10.1038/nri.2017.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sosinowski T, White JT, Cross EW, Haluszczak C, Marrack P, Gapin L, Kedl RM. CD8α+ dendritic cell trans presentation of IL-15 to naive CD8+ T cells produces antigen-inexperienced T cells in the periphery with memory phenotype and function. J Immunol. 2013;190:1936–1947. doi: 10.4049/jimmunol.1203149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.White JT, Cross EW, Burchill MA, Danhorn T, McCarter MD, Rosen HR, O’Connor B, Kedl RM. Virtual memory T cells develop and mediate bystander protective immunity in an IL-15-dependent manner. Nat Commun. 2016;7:11291. doi: 10.1038/ncomms11291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Jacomet F, Cayssials E, Basbous S, Levescot A, Piccirilli N, Desmier D, Robin A, Barra A, Giraud C, Guilhot F, et al. Evidence for eomesodermin-expressing innate-like CD8(+) KIR/NKG2A(+) T cells in human adults and cord blood samples. Eur J Immunol. 2015;45:1926–1933. doi: 10.1002/eji.201545539. [DOI] [PubMed] [Google Scholar]
- 81.Quinn KM, Fox A, Harland KL, Russ BE, Li J, Nguyen THO, Loh L, Olshanksy M, Naeem H, Tsyganov K, et al. Age-related decline in primary CD8(+) T cell responses is associated with the development of senescence in virtual memory CD8(+) T cells. Cell Rep. 2018;23:3512–3524. doi: 10.1016/j.celrep.2018.05.057. [DOI] [PubMed] [Google Scholar]
- 82.Choi SJ, Koh JY, Rha MS, Seo IH, Lee H, Jeong S, Park SH, Shin EC. KIR+CD8+ and NKG2A+CD8+ T cells are distinct innate-like populations in humans. Cell Rep. 2023;42:112236. doi: 10.1016/j.celrep.2023.112236. [DOI] [PubMed] [Google Scholar]
- 83.Kok L, Masopust D, Schumacher TN. The precursors of CD8+ tissue resident memory T cells: from lymphoid organs to infected tissues. Nat Rev Immunol. 2022;22:283–293. doi: 10.1038/s41577-021-00590-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Schenkel JM, Fraser KA, Beura LK, Pauken KE, Vezys V, Masopust D. T cell memory. Resident memory CD8 T cells trigger protective innate and adaptive immune responses. Science. 2014;346:98–101. doi: 10.1126/science.1254536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ariotti S, Hogenbirk MA, Dijkgraaf FE, Visser LL, Hoekstra ME, Song JY, Jacobs H, Haanen JB, Schumacher TN. T cell memory. Skin-resident memory CD8+ T cells trigger a state of tissue-wide pathogen alert. Science. 2014;346:101–105. doi: 10.1126/science.1254803. [DOI] [PubMed] [Google Scholar]
- 86.Mackay LK, Rahimpour A, Ma JZ, Collins N, Stock AT, Hafon ML, Vega-Ramos J, Lauzurica P, Mueller SN, Stefanovic T, et al. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol. 2013;14:1294–1301. doi: 10.1038/ni.2744. [DOI] [PubMed] [Google Scholar]
- 87.Holz LE, Prier JE, Freestone D, Steiner TM, English K, Johnson DN, Mollard V, Cozijnsen A, Davey GM, Godfrey DI, et al. CD8(+) T cell activation leads to constitutive formation of liver tissue-resident memory T cells that seed a large and flexible niche in the liver. Cell Rep. 2018;25:68–79.e4. doi: 10.1016/j.celrep.2018.08.094. [DOI] [PubMed] [Google Scholar]
- 88.Mackay LK, Wynne-Jones E, Freestone D, Pellicci DG, Mielke LA, Newman DM, Braun A, Masson F, Kallies A, Belz GT, et al. T-box transcription factors combine with the cytokines TGF-beta and IL-15 to control tissue-resident memory T cell fate. Immunity. 2015;43:1101–1111. doi: 10.1016/j.immuni.2015.11.008. [DOI] [PubMed] [Google Scholar]
- 89.Schenkel JM, Fraser KA, Casey KA, Beura LK, Pauken KE, Vezys V, Masopust D. IL-15-independent maintenance of tissue-resident and boosted effector memory CD8 T cells. J Immunol. 2016;196:3920–3926. doi: 10.4049/jimmunol.1502337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Adachi T, Kobayashi T, Sugihara E, Yamada T, Ikuta K, Pittaluga S, Saya H, Amagai M, Nagao K. Hair follicle-derived IL-7 and IL-15 mediate skin-resident memory T cell homeostasis and lymphoma. Nat Med. 2015;21:1272–1279. doi: 10.1038/nm.3962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Jarjour NN, Wanhainen KM, Peng C, Gavil NV, Maurice NJ, Borges da Silva H, Martinez RJ, Dalzell TS, Huggins MA, Masopust D, et al. Responsiveness to interleukin-15 therapy is shared between tissue-resident and circulating memory CD8+ T cell subsets. Proc Natl Acad Sci U S A. 2022;119:e2209021119. doi: 10.1073/pnas.2209021119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Koh JY, Kim DU, Moon BH, Shin EC. Human CD8(+) T-cell populations that express natural killer receptors. Immune Netw. 2023;23:e8. doi: 10.4110/in.2023.23.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Arlettaz L, Villard J, de Rham C, Degermann S, Chapuis B, Huard B, Roosnek E. Activating CD94:NKG2C and inhibitory CD94:NKG2A receptors are expressed by distinct subsets of committed CD8+ TCR alphabeta lymphocytes. Eur J Immunol. 2004;34:3456–3464. doi: 10.1002/eji.200425210. [DOI] [PubMed] [Google Scholar]
- 94.Gumá M, Busch LK, Salazar-Fontana LI, Bellosillo B, Morte C, García P, López-Botet M. The CD94/NKG2C killer lectin-like receptor constitutes an alternative activation pathway for a subset of CD8+ T cells. Eur J Immunol. 2005;35:2071–2080. doi: 10.1002/eji.200425843. [DOI] [PubMed] [Google Scholar]
- 95.Kefalakes H, Horgan XJ, Jung MK, Amanakis G, Kapuria D, Bolte FJ, Kleiner DE, Koh C, Heller T, Rehermann B. Liver-resident bystander CD8+ T cells contribute to liver disease pathogenesis in chronic hepatitis D virus infection. Gastroenterology. 2021;161:1567–1583.e9. doi: 10.1053/j.gastro.2021.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Koh JY, Rha MS, Choi SJ, Lee HS, Han JW, Nam H, Kim DU, Lee JG, Kim MS, Park JY, et al. Identification of a distinct NK-like hepatic T-cell population activated by NKG2C in a TCR-independent manner. J Hepatol. 2022;77:1059–1070. doi: 10.1016/j.jhep.2022.05.020. [DOI] [PubMed] [Google Scholar]
- 97.Ruf B, Greten TF, Korangy F. Innate lymphoid cells and innate-like T cells in cancer - at the crossroads of innate and adaptive immunity. Nat Rev Cancer. 2023;23:351–371. doi: 10.1038/s41568-023-00562-w. [DOI] [PubMed] [Google Scholar]
- 98.Godfrey DI, Koay HF, McCluskey J, Gherardin NA. The biology and functional importance of MAIT cells. Nat Immunol. 2019;20:1110–1128. doi: 10.1038/s41590-019-0444-8. [DOI] [PubMed] [Google Scholar]
- 99.Tao H, Pan Y, Chu S, Li L, Xie J, Wang P, Zhang S, Reddy S, Sleasman JW, Zhong XP. Differential controls of MAIT cell effector polarization by mTORC1/mTORC2 via integrating cytokine and costimulatory signals. Nat Commun. 2021;12:2029. doi: 10.1038/s41467-021-22162-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.van Wilgenburg B, Scherwitzl I, Hutchinson EC, Leng T, Kurioka A, Kulicke C, de Lara C, Cole S, Vasanawathana S, Limpitikul W, et al. MAIT cells are activated during human viral infections. Nat Commun. 2016;7:11653. doi: 10.1038/ncomms11653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sattler A, Dang-Heine C, Reinke P, Babel N. IL-15 dependent induction of IL-18 secretion as a feedback mechanism controlling human MAIT-cell effector functions. Eur J Immunol. 2015;45:2286–2298. doi: 10.1002/eji.201445313. [DOI] [PubMed] [Google Scholar]
- 102.Silva-Santos B, Mensurado S, Coffelt SB. γδ T cells: pleiotropic immune effectors with therapeutic potential in cancer. Nat Rev Cancer. 2019;19:392–404. doi: 10.1038/s41568-019-0153-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lee HW, Chung YS, Kim TJ. Heterogeneity of human gammadelta T cells and their role in cancer immunity. Immune Netw. 2020;20:e5. doi: 10.4110/in.2020.20.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Hu Y, Hu Q, Li Y, Lu L, Xiang Z, Yin Z, Kabelitz D, Wu Y. γδ T cells: origin and fate, subsets, diseases and immunotherapy. Signal Transduct Target Ther. 2023;8:434. doi: 10.1038/s41392-023-01653-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Baccala R, Witherden D, Gonzalez-Quintial R, Dummer W, Surh CD, Havran WL, Theofilopoulos AN. Gamma delta T cell homeostasis is controlled by IL-7 and IL-15 together with subset-specific factors. J Immunol. 2005;174:4606–4612. doi: 10.4049/jimmunol.174.8.4606. [DOI] [PubMed] [Google Scholar]
- 106.Wang H, Wang X, Wang W, Chai W, Song W, Zhang H, Mou W, Pei M, Su Y, Ma X, et al. Interleukin-15 enhanced the survival of human γδT cells by regulating the expression of Mcl-1 in neuroblastoma. Cell Death Dis. 2022;8:139. doi: 10.1038/s41420-022-00942-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Van Acker HH, Anguille S, Willemen Y, Van den Bergh JM, Berneman ZN, Lion E, Smits EL, Van Tendeloo VF. Interleukin-15 enhances the proliferation, stimulatory phenotype, and antitumor effector functions of human gamma delta T cells. J Hematol Oncol. 2016;9:101. doi: 10.1186/s13045-016-0329-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Waldmann TA. The biology of interleukin-2 and interleukin-15: implications for cancer therapy and vaccine design. Nat Rev Immunol. 2006;6:595–601. doi: 10.1038/nri1901. [DOI] [PubMed] [Google Scholar]
- 109.Liu RB, Engels B, Schreiber K, Ciszewski C, Schietinger A, Schreiber H, Jabri B. IL-15 in tumor microenvironment causes rejection of large established tumors by T cells in a noncognate T cell receptor-dependent manner. Proc Natl Acad Sci U S A. 2013;110:8158–8163. doi: 10.1073/pnas.1301022110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Mathios D, Park CK, Marcus WD, Alter S, Rhode PR, Jeng EK, Wong HC, Pardoll DM, Lim M. Therapeutic administration of IL-15 superagonist complex ALT-803 leads to long-term survival and durable antitumor immune response in a murine glioblastoma model. Int J Cancer. 2016;138:187–194. doi: 10.1002/ijc.29686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kim KH, Kim HK, Kim HD, Kim CG, Lee H, Han JW, Choi SJ, Jeong S, Jeon M, Kim H, et al. PD-1 blockade-unresponsive human tumor-infiltrating CD8+ T cells are marked by loss of CD28 expression and rescued by IL-15. Cell Mol Immunol. 2021;18:385–397. doi: 10.1038/s41423-020-0427-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lee J, Lee K, Bae H, Lee K, Lee S, Ma J, Jo K, Kim I, Jee B, Kang M, et al. IL-15 promotes self-renewal of progenitor exhausted CD8 T cells during persistent antigenic stimulation. Front Immunol. 2023;14:1117092. doi: 10.3389/fimmu.2023.1117092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Wrangle JM, Velcheti V, Patel MR, Garrett-Mayer E, Hill EG, Ravenel JG, Miller JS, Farhad M, Anderton K, Lindsey K, et al. ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. Lancet Oncol. 2018;19:694–704. doi: 10.1016/S1470-2045(18)30148-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Leidner R, Conlon K, McNeel DG, Wang-Gillam A, Gupta S, Wesolowski R, Chaudhari M, Hassounah N, Lee JB, Ho Lee L, et al. First-in-human phase I/Ib study of NIZ985, a recombinant heterodimer of IL-15 and IL-15Rα, as a single agent and in combination with spartalizumab in patients with advanced and metastatic solid tumors. J Immunother Cancer. 2023;11:e007725. doi: 10.1136/jitc-2023-007725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Miyazaki T, Maiti M, Hennessy M, Chang T, Kuo P, Addepalli M, Obalapur P, Sheibani S, Wilczek J, Pena R, et al. NKTR-255, a novel polymer-conjugated rhIL-15 with potent antitumor efficacy. J Immunother Cancer. 2021;9:e002024. doi: 10.1136/jitc-2020-002024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Rhode PR, Egan JO, Xu W, Hong H, Webb GM, Chen X, Liu B, Zhu X, Wen J, You L, et al. Comparison of the superagonist complex, ALT-803, to IL15 as cancer immunotherapeutics in animal models. Cancer Immunol Res. 2016;4:49–60. doi: 10.1158/2326-6066.CIR-15-0093-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Kim PS, Kwilas AR, Xu W, Alter S, Jeng EK, Wong HC, Schlom J, Hodge JW. IL-15 superagonist/IL-15RαSushi-Fc fusion complex (IL-15SA/IL-15RαSu-Fc; ALT-803) markedly enhances specific subpopulations of NK and memory CD8+ T cells, and mediates potent anti-tumor activity against murine breast and colon carcinomas. Oncotarget. 2016;7:16130–16145. doi: 10.18632/oncotarget.7470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Knudson KM, Hodge JW, Schlom J, Gameiro SR. Rationale for IL-15 superagonists in cancer immunotherapy. Expert Opin Biol Ther. 2020;20:705–709. doi: 10.1080/14712598.2020.1738379. [DOI] [PubMed] [Google Scholar]
- 119.Chamie K, Lee JH, Rock A, Rhode PR, Soon-Shiong P. Preliminary phase 2 clinical results of IL-15RαFc superagonist N-803 with BCG in BCG-unresponsive non-muscle invasive bladder cancer (NMIBC) patients. J Clin Oncol. 2019;37:4561. [Google Scholar]
- 120.Alizadeh D, Wong RA, Yang X, Wang D, Pecoraro JR, Kuo CF, Aguilar B, Qi Y, Ann DK, Starr R, et al. IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol Res. 2019;7:759–772. doi: 10.1158/2326-6066.CIR-18-0466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Singh N, Perazzelli J, Grupp SA, Barrett DM. Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies. Sci Transl Med. 2016;8:320ra3. doi: 10.1126/scitranslmed.aad5222. [DOI] [PubMed] [Google Scholar]
- 122.Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, Xue A, Goff SL, Yang JC, Sherry RM, et al. Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels. J Clin Oncol. 2017;35:1803–1813. doi: 10.1200/JCO.2016.71.3024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hurton LV, Singh H, Najjar AM, Switzer KC, Mi T, Maiti S, Olivares S, Rabinovich B, Huls H, Forget MA, et al. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc Natl Acad Sci U S A. 2016;113:E7788–E7797. doi: 10.1073/pnas.1610544113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Simoni Y, Becht E, Fehlings M, Loh CY, Koo SL, Teng KWW, Yeong JPS, Nahar R, Zhang T, Kared H, et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature. 2018;557:575–579. doi: 10.1038/s41586-018-0130-2. [DOI] [PubMed] [Google Scholar]
- 125.Tietze JK, Wilkins DE, Sckisel GD, Bouchlaka MN, Alderson KL, Weiss JM, Ames E, Bruhn KW, Craft N, Wiltrout RH, et al. Delineation of antigen-specific and antigen-nonspecific CD8(+) memory T-cell responses after cytokine-based cancer immunotherapy. Blood. 2012;119:3073–3083. doi: 10.1182/blood-2011-07-369736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Liu K, Catalfamo M, Li Y, Henkart PA, Weng NP. IL-15 mimics T cell receptor crosslinking in the induction of cellular proliferation, gene expression, and cytotoxicity in CD8+ memory T cells. Proc Natl Acad Sci U S A. 2002;99:6192–6197. doi: 10.1073/pnas.092675799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Maurice NJ, Taber AK, Prlic M. The ugly duckling turned to swan: a change in perception of bystander-activated memory CD8 T cells. J Immunol. 2021;206:455–462. doi: 10.4049/jimmunol.2000937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Younes SA, Freeman ML, Mudd JC, Shive CL, Reynaldi A, Panigrahi S, Estes JD, Deleage C, Lucero C, Anderson J, et al. IL-15 promotes activation and expansion of CD8+ T cells in HIV-1 infection. J Clin Invest. 2016;126:2745–2756. doi: 10.1172/JCI85996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Chu T, Tyznik AJ, Roepke S, Berkley AM, Woodward-Davis A, Pattacini L, Bevan MJ, Zehn D, Prlic M. Bystander-activated memory CD8 T cells control early pathogen load in an innate-like, NKG2D-dependent manner. Cell Rep. 2013;3:701–708. doi: 10.1016/j.celrep.2013.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Yajima T, Nishimura H, Sad S, Shen H, Kuwano H, Yoshikai Y. A novel role of IL-15 in early activation of memory CD8+ CTL after reinfection. J Immunol. 2005;174:3590–3597. doi: 10.4049/jimmunol.174.6.3590. [DOI] [PubMed] [Google Scholar]
- 131.Soudja SM, Ruiz AL, Marie JC, Lauvau G. Inflammatory monocytes activate memory CD8(+) T and innate NK lymphocytes independent of cognate antigen during microbial pathogen invasion. Immunity. 2012;37:549–562. doi: 10.1016/j.immuni.2012.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Bastidas S, Graw F, Smith MZ, Kuster H, Günthard HF, Oxenius A. CD8+ T cells are activated in an antigen-independent manner in HIV-infected individuals. J Immunol. 2014;192:1732–1744. doi: 10.4049/jimmunol.1302027. [DOI] [PubMed] [Google Scholar]
- 133.Kim J, Chang DY, Lee HW, Lee H, Kim JH, Sung PS, Kim KH, Hong SH, Kang W, Lee J, et al. Innate-like cytotoxic function of bystander-activated CD8(+) T cells is associated with liver injury in acute hepatitis a. Immunity. 2018;48:161–173.e5. doi: 10.1016/j.immuni.2017.11.025. [DOI] [PubMed] [Google Scholar]
- 134.Seo IH, Eun HS, Kim JK, Lee H, Jeong S, Choi SJ, Lee J, Lee BS, Kim SH, Rou WS, et al. IL-15 enhances CCR5-mediated migration of memory CD8+ T cells by upregulating CCR5 expression in the absence of TCR stimulation. Cell Rep. 2021;36:109438. doi: 10.1016/j.celrep.2021.109438. [DOI] [PubMed] [Google Scholar]
- 135.Huang CH, Fan JH, Jeng WJ, Chang ST, Yang CK, Teng W, Wu TH, Hsieh YC, Chen WT, Chen YC, et al. Innate-like bystander-activated CD38+ HLA-DR+ CD8+ T cells play a pathogenic role in patients with chronic hepatitis C. Hepatology. 2022;76:803–818. doi: 10.1002/hep.32349. [DOI] [PubMed] [Google Scholar]
- 136.Freeman CM, Han MK, Martinez FJ, Murray S, Liu LX, Chensue SW, Polak TJ, Sonstein J, Todt JC, Ames TM, et al. Cytotoxic potential of lung CD8(+) T cells increases with chronic obstructive pulmonary disease severity and with in vitro stimulation by IL-18 or IL-15. J Immunol. 2010;184:6504–6513. doi: 10.4049/jimmunol.1000006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Sacramento LA, Farias Amorim C, Campos TM, Saldanha M, Arruda S, Carvalho LP, Beiting DP, Carvalho EM, Novais FO, Scott P. NKG2D promotes CD8 T cell-mediated cytotoxicity and is associated with treatment failure in human cutaneous leishmaniasis. PLoS Negl Trop Dis. 2023;17:e0011552. doi: 10.1371/journal.pntd.0011552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Shin EC, Sung PS, Park SH. Immune responses and immunopathology in acute and chronic viral hepatitis. Nat Rev Immunol. 2016;16:509–523. doi: 10.1038/nri.2016.69. [DOI] [PubMed] [Google Scholar]
- 139.Wilkinson PC, Liew FY. Chemoattraction of human blood T lymphocytes by interleukin-15. J Exp Med. 1995;181:1255–1259. doi: 10.1084/jem.181.3.1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Verbist KC, Cole CJ, Field MB, Klonowski KD. A role for IL-15 in the migration of effector CD8 T cells to the lung airways following influenza infection. J Immunol. 2011;186:174–182. doi: 10.4049/jimmunol.1002613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Nolz JC, Harty JT. IL-15 regulates memory CD8+ T cell O-glycan synthesis and affects trafficking. J Clin Invest. 2014;124:1013–1026. doi: 10.1172/JCI72039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Rha MS, Han JW, Kim JH, Koh JY, Park HJ, Kim SI, Kim MS, Lee JG, Lee HW, Lee DH, et al. Human liver CD8+ MAIT cells exert TCR/MR1-independent innate-like cytotoxicity in response to IL-15. J Hepatol. 2020;73:640–650. doi: 10.1016/j.jhep.2020.03.033. [DOI] [PubMed] [Google Scholar]
- 143.Lee H, Jung MK, Noh JY, Park SH, Chung Y, Ha SJ, Shin EC. Better understanding CD8+ T cells in cancer and viral infections. Nat Immunol. 2023;24:1794–1796. doi: 10.1038/s41590-023-01630-x. [DOI] [PubMed] [Google Scholar]
- 144.Jeong S, Jeon M, Lee H, Kim SY, Park SH, Shin EC. IFITM3 is upregulated characteristically in IL-15-mediated bystander-activated CD8(+) T cells during influenza infection. J Immunol. 2022;208:1901–1911. doi: 10.4049/jimmunol.2100629. [DOI] [PubMed] [Google Scholar]
- 145.Meresse B, Chen Z, Ciszewski C, Tretiakova M, Bhagat G, Krausz TN, Raulet DH, Lanier LL, Groh V, Spies T, et al. Coordinated induction by IL15 of a TCR-independent NKG2D signaling pathway converts CTL into lymphokine-activated killer cells in celiac disease. Immunity. 2004;21:357–366. doi: 10.1016/j.immuni.2004.06.020. [DOI] [PubMed] [Google Scholar]
- 146.Hüe S, Mention JJ, Monteiro RC, Zhang S, Cellier C, Schmitz J, Verkarre V, Fodil N, Bahram S, Cerf-Bensussan N, et al. A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity. 2004;21:367–377. doi: 10.1016/j.immuni.2004.06.018. [DOI] [PubMed] [Google Scholar]
- 147.Hoytema van Konijnenburg DP, Reis BS, Pedicord VA, Farache J, Victora GD, Mucida D. Intestinal epithelial and intraepithelial T cell crosstalk mediates a dynamic response to infection. Cell. 2017;171:783–794.e13. doi: 10.1016/j.cell.2017.08.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Zhao H, Nguyen H, Kang J. Interleukin 15 controls the generation of the restricted T cell receptor repertoire of gamma delta intestinal intraepithelial lymphocytes. Nat Immunol. 2005;6:1263–1271. doi: 10.1038/ni1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Mayassi T, Barreiro LB, Rossjohn J, Jabri B. A multilayered immune system through the lens of unconventional T cells. Nature. 2021;595:501–510. doi: 10.1038/s41586-021-03578-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Simpson SJ, Holländer GA, Mizoguchi E, Allen D, Bhan AK, Wang B, Terhorst C. Expression of pro-inflammatory cytokines by TCR alpha beta+ and TCR gamma delta+ T cells in an experimental model of colitis. Eur J Immunol. 1997;27:17–25. doi: 10.1002/eji.1830270104. [DOI] [PubMed] [Google Scholar]
- 151.Kawaguchi-Miyashita M, Shimada S, Kurosu H, Kato-Nagaoka N, Matsuoka Y, Ohwaki M, Ishikawa H, Nanno M. An accessory role of TCRgammadelta (+) cells in the exacerbation of inflammatory bowel disease in TCRalpha mutant mice. Eur J Immunol. 2001;31:980–988. doi: 10.1002/1521-4141(200104)31:4<980::aid-immu980>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
- 152.Xing L, Dai Z, Jabbari A, Cerise JE, Higgins CA, Gong W, de Jong A, Harel S, DeStefano GM, Rothman L, et al. Alopecia areata is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition. Nat Med. 2014;20:1043–1049. doi: 10.1038/nm.3645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Seok J, Cho SD, Lee J, Choi Y, Kim SY, Lee SM, Kim SH, Jeong S, Jeon M, Lee H, et al. A virtual memory CD8+ T cell-originated subset causes alopecia areata through innate-like cytotoxicity. Nat Immunol. 2023;24:1308–1317. doi: 10.1038/s41590-023-01547-5. [DOI] [PubMed] [Google Scholar]
- 154.Masle-Farquhar E, Jackson KJL, Peters TJ, Al-Eryani G, Singh M, Payne KJ, Rao G, Avery DT, Apps G, Kingham J, et al. STAT3 gain-of-function mutations connect leukemia with autoimmune disease by pathological NKG2Dhi CD8+ T cell dysregulation and accumulation. Immunity. 2022;55:2386–2404.e8. doi: 10.1016/j.immuni.2022.11.001. [DOI] [PubMed] [Google Scholar]
- 155.Dudek M, Pfister D, Donakonda S, Filpe P, Schneider A, Laschinger M, Hartmann D, Hüser N, Meiser P, Bayerl F, et al. Auto-aggressive CXCR6+ CD8 T cells cause liver immune pathology in NASH. Nature. 2021;592:444–449. doi: 10.1038/s41586-021-03233-8. [DOI] [PubMed] [Google Scholar]



