Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2019 Jul 30;93(16):e00617-19. doi: 10.1128/JVI.00617-19

Cytomegalovirus Evades TRAIL-Mediated Innate Lymphoid Cell 1 Defenses

Gaelle Picarda a, Raima Ghosh a, Bryan McDonald a, Shilpi Verma a, Nicolas Thiault b, Rachid El Morabiti a, Thomas S Griffith c, Chris A Benedict a,
Editor: Richard M Longneckerd
PMCID: PMC6675903  PMID: 31142671

Cytomegalovirus (a betaherpesvirus) is a master at manipulating immune responses to promote its lifelong persistence, a result of millions of years of coevolution with its host. Using a one-of-a-kind MCMV mutant unable to restrict expression of the TNF-related apoptosis-inducing ligand death receptors (TRAIL-DR), we show that TRAIL-DR signaling significantly restricts both early and persistent viral replication. Our results also reveal that these defenses are employed by TRAIL-expressing innate lymphoid type I cells (ILC1) but not conventional NK cells. Overall, our results are significant because they show the key importance of viral counterstrategies specifically neutralizing TRAIL effector functions mediated by a specific, tissue-resident subset of group I ILCs.

KEYWORDS: ILC1, MCMV, NK, TRAIL, group I ILCs

ABSTRACT

Cytomegalovirus (CMV) establishes a lifelong infection facilitated, in part, by circumventing immune defenses mediated by tumor necrosis factor (TNF)-family cytokines. An example of this is the mouse CMV (MCMV) m166 protein, which restricts expression of the TNF-related apoptosis-inducing ligand (TRAIL) death receptors, promoting early-phase replication. We show here that replication of an MCMV mutant lacking m166 is also severely attenuated during viral persistence in the salivary glands (SG). Depleting group I innate lymphoid cells (ILCs) or infecting Trail−/− mice completely restored persistent replication of this mutant. Group I ILCs are comprised of two subsets, conventional natural killer cells (cNK) and tissue-resident cells often referred to as innate lymphoid type I cells (ILC1). Using recently identified phenotypic markers to discriminate between these two cell types, their relative expression of TRAIL and gamma interferon (IFN-γ) was assessed during both early and persistent infection. ILC1 were found to be the major TRAIL expressers during both of these infection phases, with cNK expressing very little, indicating that it is ILC1 that curtail replication via TRAIL in the absence of m166-imposed countermeasures. Notably, despite high TRAIL expression by SG-resident ILC1, IFN-γ production by both ILC1 and cNK was minimal at this site of viral persistence. Together these results highlight TRAIL as a key ILC1-utilized effector molecule that can operate in defense against persistent infection at times when other innate control mechanisms may be muted and highlight the importance for the evolution of virus-employed countermeasures.

IMPORTANCE Cytomegalovirus (a betaherpesvirus) is a master at manipulating immune responses to promote its lifelong persistence, a result of millions of years of coevolution with its host. Using a one-of-a-kind MCMV mutant unable to restrict expression of the TNF-related apoptosis-inducing ligand death receptors (TRAIL-DR), we show that TRAIL-DR signaling significantly restricts both early and persistent viral replication. Our results also reveal that these defenses are employed by TRAIL-expressing innate lymphoid type I cells (ILC1) but not conventional NK cells. Overall, our results are significant because they show the key importance of viral counterstrategies specifically neutralizing TRAIL effector functions mediated by a specific, tissue-resident subset of group I ILCs.

INTRODUCTION

Cytomegalovirus (CMV; a betaherpesvirus) infects the majority of the world’s population, with the rate of infection varying from ∼40 to 90% and with infection being based on age, race, geography, and socioeconomic conditions (1). Primary infection is almost always asymptomatic if the subject is healthy but can be life threatening if immunity is compromised or immature (e.g., if the infected individual has had a transplant or congenital infections), and vaccine development is a top priority (2, 3). Like all the Herpesviridae, CMV escapes complete immune clearance, leading to a lifelong persistence/latency. Distinct from its viral cousins, however, CMV establishes a sleepless latency (4) in multiple tissues and cell types, commensurate with significant levels of viral gene expression (5). Consequently, even in healthy individuals, CMV has an enormous impact on shaping the immune system over time (6, 7).

CMV dedicates >50% of its ∼230-kb genome to subverting immune defenses (8), facilitating latency and contributing to its broad impact on immunity. Notably, much of this genomic currency is targeted at the tumor necrosis factor (TNF) superfamily of receptors and ligands (9). The TNF-related apoptosis-inducing ligand (TRAIL) signaling system has been extensively studied for its ability to selectively kill transformed cells, and multiple clinical trials have evaluated the anticancer efficacy of TRAIL death receptor (TRAIL-DR) agonists (10, 11). However, TRAIL is also emerging as an important regulator of antiviral defenses (12). Supporting this, we have reported previously that the UL141 and m166 proteins in human CMV (HCMV) and mouse CMV (MCMV) restrict cell surface expression of the TRAIL-DRs (13, 14). Abolishing m166 expression by inserting two stop codons within the m166 genomic open reading frame (MCMV-m166stop) revealed that this viral immunomodulatory protein promotes acute infection in multiple organs, most dramatically, in the liver, where replication of this mutant was undetectable (14). Depletion with anti-asialo-GM1 or infection of Trail-dr−/− mice completely restored the acute replication of MCMV-m166stop, indicating that m166 restriction of TRAIL-DR expression is critical to block the innate control of early infection by group I innate lymphoid cells (ILCs) (14).

Recently, work by several groups has revealed that group I ILCs are composed not only of conventional natural killer cells (cNK) but also a subset of tissue-resident cells often referred to as innate lymphoid type I cells (ILC1) (15, 16). Both ILC1 and cNK express the transcription factor T-bet, display the NK1.1 and NKp46 cell surface receptors, and can produce gamma interferon (IFN-γ) when activated by various stimuli (15, 16). Consequently, prior to the very recent identification of unique markers segregating ILC1 and cNK, studies of group I ILC antiviral defenses have not assessed the relative importance of these two cellular subsets. This is especially relevant in the case of TRAIL-mediated immunity, as ILC1 have been shown to highly express this TNF-family cytokine under certain conditions (1619). However, despite several groups showing that TRAIL can be produced by ILC1, virtually nothing is known regarding its importance for their role as immune effectors.

Here we have examined TRAIL expression in total group I ILCs during both the acute and persistent phases of MCMV infection, seeking to determine the relative importance of ILC1 and cNK in TRAIL-dependent innate defense. Our data show that in multiple tissues, ILC1 are by far the major cell type to express TRAIL, while cNK produce almost none. In the absence of m166 blockade, these TRAIL-positive (TRAIL+) ILC1 can potently restrict both early and persistent MCMV replication levels. Together, these results highlight ILC1 as key regulators of TRAIL-mediated antiviral immunity.

RESULTS

MCMV m166 inhibits TRAIL-mediated ILC1 defenses during early infection in the liver.

Our past work using a viral mutant lacking expression of the m166 protein (m166stop) showed that early MCMV replication is severely compromised, as m166 blocks cell surface expression of the TRAIL-DR and subverts immune control by cells sensitive to depletion with anti-asialo-GM1 antibody (14). MCMV-m166stop replication is normal in the liver of Trail-dr−/− mice (14), proving the singular importance for restriction of this TNF receptor protein by m166. Together these results suggest that liver-localized, TRAIL+ asialo-GM1-positive (asialo-GM1+) cells can trigger TRAIL-DR signaling in MCMV-m166stop-infected cells, killing them and curtailing viral replication.

Natural killer (NK) cells have long been thought to be the primary innate cell subset mobilized to control early MCMV replication (17). However, mature NK cells express very little TRAIL (18), suggesting that they may not be the key regulators of MCMV-m166stop replication. In the past, liver NK cells have been partitioned into immature and mature subsets based on their expression of CD11b and CD27 (19), and we observed that immature NKp46-positive (NKp46+) cells from naive mice (e.g., CD11b cells) express the highest levels of TRAIL (Fig. 1A and B). More recently, it has become clear that NKp46+ cells are comprised of at least two distinct innate cell types, conventional NK cells (cNK) and innate lymphoid type I cells (ILC1), which together comprise the group I ILCs (16). In the liver, ILC1 and cNK can be readily distinguished by the differential expression of the transcription factor Eomes and integrin α2 (CD49b) (20, 21) (Fig. 1C). We found that more than 90% of liver ILC1 (CD19 T cell receptor β-negative [TCRβ] F4/80 CD3 NKp46+ Eomes CD49b) were CD11b, with only ∼50% of cNK (CD19 TCRβ F4/80 CD3 NKp46+ Eomes+ CD49b+) displaying this phenotype, indicating that the majority of TRAIL-expressing group I ILCs in naive mice were ILC1 and not immature cNK (Fig. 1D).

FIG 1.

FIG 1

TRAIL expression by group I ILCs in the liver. The panels present the results of analysis of total liver mononuclear cells isolated at the indicated times. (A) Total group I ILCs (F4/80 TCRβ CD19 CD3 NKp46+) from naive WT mice were analyzed for expression of CD27 and CD11b. (B) The 4 subsets for which the results are shown in panel A were analyzed for TRAIL expression. (C) Gating strategy for the detection of liver group I ILCs among living CD19 TCRβ F4/80 cells (left) and of ILC1 and cNK among liver group I ILCs (right). (D) ILC1 (F4/80 TCRβ CD19 CD3 NKp46+ Eomes CD49b) and cNK (F4/80 TCRβ CD19 CD3 NKp46+ Eomes CD49b) were analyzed for the expression of CD27 and CD11b. (E, F) Mock- or MCMV-infected mice were analyzed 36 h or 4 days later for the percentage and absolute numbers of ILC1 (E) and cNK (F) among total group I ILCs. Results are representative of those from three independent experiments with at least five mice per group. Graph lines depict the mean, and statistical significance was determined by the two-tailed unpaired Mann-Whitney U test in this and all figures unless otherwise noted. **, P < 0.01; ***, P < 0.001.

Next, we examined the impact of MCMV infection on liver group I ILCs. No changes in the proportions or numbers of ILC1 and cNK were observed at 36 h of infection compared to those in uninfected mice. In stark contrast, the proportion of ILC1 decreased dramatically at day 4 of infection, albeit their absolute numbers did not significantly change, due to an ∼9-fold increase in liver cNK numbers (Fig. 1E and F). TRAIL expression by cNK and ILC1 was also assessed, and we found that the proportion of TRAIL+ ILC1 increased from an average of 58% in naive mice to 87% after 36 h of infection, with a commensurate increase in the levels of ILC1 TRAIL expression (Fig. 2A). Interestingly, MCMV-m166stop induced an increase in the proportions and levels of TRAIL expression by ILC1 similar to that induced by WT virus at 36 h (Fig. 2B). In naive mice, only ∼3% of the liver cNK expressed any detectable TRAIL, and only a slight increase in the proportion was seen in WT or m166stop-infected mice at 36 h or 4 days postinfection (Fig. 2A and C). In addition, >90% of TRAIL+ group I ILCs in both naive and infected mice were Eomes CD49b, indicating that the vast majority of TRAIL-producing group I ILCs in the liver were ILC1 (Fig. 2D and E).

FIG 2.

FIG 2

ILC1 are the major TRAIL-expressing cells in MCMV-infected livers. The panels present the results of analysis of total liver mononuclear cells isolated at the indicated times from naive or MCMV-infected mice. (A) TRAIL expression by total group I ILCs and ILC1 and cNK subsets at 36 h of MCMV infection. (B) TRAIL expression by cNK and ILC1 subsets at 36 h of infection with either the MCMV WT or m166stop. MFI, median fluorescence intensity. (C) TRAIL expression by total group I ILCs, ILC1, and cNK at day 4 of infection. (D) ILC1/cNK distribution for TRAIL+ and TRAIL group I ILCs according to their expression of Eomes and CD49b. (E) Percentage of ILC1 among TRAIL+ group I ILCs from naive mice and mice infected for 8 days. **, P < 0.01.

To further examine whether TRAIL-expressing group I ILCs were composed largely of ILC1 during MCMV infection, expression of 6 additional markers whose differential expression segregates ILC1 and cNK was assessed (Fig. 3A). At 36 h after infection, TRAIL+ group I ILCs expressed higher levels of the tissue residency markers CD69 and CD49a (20, 21) than cNK did, adding further assurance that these cells were ILC1. In addition, the level of expression of CD61, which is selectively expressed by ILC1 from various tissues (22), was also much higher in TRAIL+ group I ILCs than cNK. In turn, the cNK-expressed markers CD62L, KLRG1, and CD11b (21) were absent in TRAIL+ group I ILCs, similar to the phenotype of ILC1 (Fig. 3A). Both uninfected and MCMV-m166stop-infected mice showed phenotypes for TRAIL+ group I ILCs similar to the phenotype seen in WT virus-infected mice (Fig. 3B and C). Taken together, these results strongly support the conclusion that virtually all liver-resident, TRAIL-expressing group I ILCs that participate in defense against MCMV infection are ILC1 and not cNK.

FIG 3.

FIG 3

TRAIL-expressing group I ILCs display characteristic phenotypic markers of ILC1. (A) Expression of CD49a, CD69, CD61, CD62L, CD11b, and KLRG1 was assessed in total TRAIL+ group I ILCs, ILC1, and cNK at 36 h after infection. (B, C) Expression of CD49a, CD69, CD61, CD62L, CD11b, and KLRG1. (B) Expression of each marker by ILC1, cNK, and TRAIL+ group I ILC subsets from naive mice. (C) Expression of each marker by cNK and TRAIL+ group I ILCs at 36 h after infection with the MCMV WT or m166stop. (D) ILC1 and cNK from naive mice and mice infected for 36 h were analyzed directly ex vivo for their expression of IFN-γ after 5 h of incubation with brefeldin A and monensin with or without PMA and ionomycin. No Stim, no stimulation; FSCA, forward scatter area. *, P < 0.1; **, P < 0.01.

ILC1 and cNK function distinctly during MCMV infection.

One unifying characteristic of group I ILCs is thought to be their ability to produce IFN-γ upon appropriate stimulation (21, 23), and this was assessed in liver cNK and ILC1 at 36 h after MCMV infection (Fig. 3D). While no significant IFN-γ production by cNK or ILC1 could be detected in the liver of naive mice, a detectable percentage of IFN-γ-positive (IFN-γ+) cells was observed for both subsets in MCMV-infected livers when analyzed straight ex vivo without restimulation (Fig. 3D), and this was also true at 4 days following infection (data not shown). For comparison, ∼10 to 15% of splenic cNK produced IFN-γ at this time (data not shown). In contrast, ex vivo restimulation with phorbol myristate acetate (PMA) and ionomycin (Iono) revealed that ∼6% of cNK and ∼30% ILC1 from naive mice were capable of producing IFN-γ, with these proportions increasing to ∼38% and 67%, respectively, 36 h after MCMV infection (Fig. 3D).

MCMV m166 inhibits TRAIL-mediated group I ILC defenses in the salivary gland during viral persistence.

After host defenses resolve the initial, systemic replication phase of MCMV within ∼7 days, the virus disseminates to the salivary glands (SG), where it replicates persistently for several more weeks. To determine the extent to which m166 might also impact TRAIL-mediated control of MCMV persistence by group I ILCs, the SG replication levels of WT and m166stop were compared (Fig. 4A). A dramatic reduction in replication of m166stop was observed at all times between days 4 and 15 (e.g., ∼70-fold at day 8 and day 10). We postulated that this might result from reduced early replication, which could potentially restrict seeding of the SG, and tested this directly by varying the dose of initial infection. A low-dose infection (103 PFU) yielded undetectable replication in the liver and spleen at day 4, while a higher-dose infection (105 PFU) yielded readily detectable levels by plaque assay (Fig. 4B). In contrast, both doses resulted in identical SG replication at day 12 (Fig. 4C), indicating that persistent replication levels are largely independent of early-phase levels and that reduced m166stop replication in the SG is likely due to direct immune control at this mucosal site and not decreased dissemination to it.

FIG 4.

FIG 4

M166 restricts TRAIL-dependent control of MCMV by group I ILCs during viral persistence in the salivary gland. MCMV replication levels in various organs are shown as the number of PFU determined in NIH 3T3 cells. (A) Mice were infected with the MCMV WT or m166stop, and SG replication levels were determined 4 to 15 days later. (B, C) Mice were infected with 103 PFU (black dots) or 105 PFU (gray dots) of WT MCMV, and replication was determined at day 4 in the liver and spleen (B) or day 12 in the SG (C). (D) Mice were infected with either the WT (black) or m166stop (red), and replication in the SG was assessed 8 days later. WT mice were depleted (or not) of CD4 T cells prior to infection. (E) Proportion of group I ILCs in the SG 9 days after mice were injected with either PBS or anti-asialo-GM1 antibody. (F, G) Mice were infected with either the WT (black) or m166stop (red), and replication in the SG was assessed 8 days later. (F) WT mice were depleted (or not) of group I ILCs prior to infection. (G) WT and Trail−/− mice were infected with either WT or m166stop MCMV. Results are representative of those from three independent experiments with at least four mice per group. The dashed line denotes the limit of assay detection. *, P < 0.1; **, P < 0.01; ****, P < 0.0001.

CD4 T cells are critical for controlling MCMV persistent replication in the SG (24). To determine to what extent the reduced persistence of m166stop was dependent on CD4 T cells, we depleted CD4 T cells using the anti-CD4 monoclonal antibody GK1.5 (which produces >95% depletion in SG), and MCMV replication was then measured at day 8 (Fig. 4D). This depletion enhanced the replication of both the WT and m166stop, highlighting the known requirement for CD4 T cell control in the SG, but indicated that these cells are not selectively responsible for the enhanced control of m166stop. Very little is known regarding how group I ILCs contribute to MCMV SG defenses, particularly with regard to the relative roles of ILC1 and cNK. To probe this question, mice were injected with anti-asialo-GM1 prior to infection with either the WT or m166stop. Approximately 80% of group I ILCs were depleted in the SG (Fig. 4E), and this completely restored the replication of m166stop to WT levels (Fig. 4F). In addition, m166stop replication was also fully normalized in Trail−/− mice (Fig. 4G). Together these data show that group I ILCs can mediate TRAIL-dependent antiviral defenses to restrict viral persistence in the salivary gland and that MCMV m166 can subvert this mechanism of host immune control.

MCMV induces specific, high-level TRAIL expression by ILC1 in the salivary gland.

Results of assays in which group I ILCs were depleted indicated that either cNK or ILC1 in the SG were responsible for enhanced control of the m166stop mutant. Therefore, TRAIL expression in total NKp46+ SG-resident cells was assessed at day 8 after infection. Notably, unlike the liver, TRAIL expression was virtually undetectable in total group I ILCs isolated from the SG of naive mice but was significantly upregulated following both MCMV WT and m166stop infection (Fig. 5A), albeit to a lower extent with m166stop. Additionally, MCMV induced a significant increase in both the absolute number and the proportion of TRAIL+ group I ILCs at day 8 (Fig. 5A). We then determined whether these TRAIL+ SG cells were ILC1 or cNK. Recent studies in C57BL/6 (B6) mice have shown that, unlike in the liver, these two subsets of group I ILCs cannot be differentiated by their expression of Eomes and CD49b (25, 26). To determine if this was also true in BALB/c mice, TRAIL expression by total NKp46+ SG-resident cells was measured. Unlike in the liver, TRAIL+ group I ILCs distributed equally between Eomes- and CD49b-expressing subsets (Fig. 5B). Additionally, expression of the tissue residency markers CD69 and CD49a was equivalent in Eomes+ CD49b+ and Eomes CD49b SG group I ILCs, again distinct from the findings for the liver (Fig. 5C) (27, 28). These data indicate that, like in B6 mice, Eomes and CD49b expression does not discriminate between ILC1 and cNK in BALB/c mouse SGs.

FIG 5.

FIG 5

TRAIL-expressing group I ILCs in MCMV-infected salivary glands express the classical ILC1 phenotypic markers. The panels present the results of analysis of total SG mononuclear cells isolated from naive WT or Trail−/− mice and at 8 days postinfection with MCMV WT or m166stop. (A) TRAIL expression by total group I ILCs from naive mice or mice infected for 8 days with the MCMV WT or m166stop. (B) Distribution of TRAIL+ and TRAIL group I ILCs at 8 days after infection with the MCMV WT according to their expression of Eomes and CD49b. (C) Expression of CD49a and CD69 by Eomes+ CD49b+ and Eomes CD49b group I ILCs at 8 days after infection with the MCMV WT. Gray histograms represent the liver cNK of mice infected for 36 h. (D) Expression of CD49a, CD69, CD61, CD62L, CD11b, and KLRG1 by the TRAIL+ (red) and TRAIL (blue) group I ILC subsets of mice infected for 8 days. Gray histograms represent liver cNK from mice infected for 8 days. (E) Expression of CD49a, CD69, CD61, CD62L, CD11b, and KLRG1 by TRAIL+ group I ILCs at 8 days after infection with MCMV WT or m166stop. Gray histograms represent liver cNK of mice infected for 36 h. **, P < 0.01.

It has been reported that, similar to the liver, SG-resident ILC1 express low/no CD62L, CD11b, and KLRG1 and also highly express CD61 (22, 25, 27). Therefore, expression of these molecules by TRAIL+ and TRAIL-negative (TRAIL) group I ILCs in the SG was compared to that by liver cNK after infection with the MCMV WT and m166stop (Fig. 5D and E). After infection with both the WT and m166stop, TRAIL+ group I ILCs highly expressed the canonical ILC1 markers (CD69, CD49a, CD61) but not the cNK markers (CD62L, CD11b, KLRG1) (Fig. 5D and E). Interestingly, TRAIL group I ILCs displayed an intermediate phenotype between the phenotypes of the TRAIL+ subset and liver cNK, suggesting that in the SG they may be composed of both cNK and ILC1 (Fig. 5D). Nevertheless, these results strongly suggest that the vast majority of TRAIL+ group I ILCs in the SG are ILC1. As a second approach, we first subdivided total group I ILCs based on their CD61 expression, followed by analysis of the same canonical cNK and ILC1 markers. This analysis revealed that TRAIL+ group I ILCs in the SG show a phenotype very similar to that of total CD61+ cells, while both differed significantly from CD61 group I ILCs, which more closely resembled liver cNK (Fig. 6A). These data strongly suggest that CD61 expression can be used to identify ILC1 (CD61+) and cNK (CD61) in the SG of BALB/c mice. Interestingly, the absolute numbers of both ILC1 and cNK increased significantly in the SG after infection with both WT and m166stop (Fig. 6B). Of these, 72% of ILC1 and 27% of cNK expressed TRAIL in WT-infected mice, whereas 43% and 12%, respectively, expressed TRAIL in m166stop-infected mice (Fig. 6C and D). Overall, our results support the conclusions that (i) CD61 expression identifies SG-resident ILC1, (ii) these CD61+ ILC1 are the major subset of group I ILCs producing TRAIL during MCMV persistence, and (iii) this innate cell subset can control MCMV persistent replication if the virus is unable to restrict TRAIL-DR expression.

FIG 6.

FIG 6

ILC1 are the major TRAIL-expressing cells in MCMV-infected salivary glands. The panels present the results of analysis of total SG mononuclear cells isolated from naive WT or Trail−/− mice and at 8 days postinfection with MCMV WT or m166stop. (A) Group I ILCs from mice infected with the MCMV WT for 8 days were divided according to their expression of CD61. Cell surface expression of CD49a, CD69, CD62L, CD11b, and KLRG1 was assessed on CD61+, CD61, and TRAIL+ total group I ILCs. (B) Absolute number of ILC1 (CD61+) and cNK (CD61) group I ILCs from naive mice (yellow) and at 8 days after infection with either the MCMV WT (purple) or m166stop (green). (C) TRAIL expression by cNK (CD61 group I ILCs) and ILC1 (CD61+ group I ILCs) from naive mice and mice infected for 8 days. (D) The proportion of TRAIL+ ILC1 and cNK from naive mice (yellow) and at 8 days after infection with MCMV WT (purple) or m166stop (green). (E) ILC1 (CD61+ group I ILCs, blue) and cNK (CD61 group I ILCs, gray) from naive mice and mice infected for 8 days were analyzed ex vivo for their expression of IFN-γ after 5 h of incubation with brefeldin A and monensin with or without PMA and ionomycin. *, P < 0.1; **, P < 0.01.

Finally, we assessed the relative expression of IFN-γ by SG-resident group I ILCs in naive and MCMV-infected BALB/c mice (Fig. 6E), as they produce low levels of this effector cytokine in B6 mice compared to their splenic counterparts (29). At day 8 of infection, neither cNK nor ILC1 produced detectable IFN-γ (Fig. 6E). Even following ex vivo stimulation with PMA and ionomycin, only a small proportion of both cNK and ILC1 was able to produce IFN-γ (∼5%).

Together these results reveal that SG-resident ILC1 produce low levels of the canonical effector cytokine IFN-γ in response to MCMV but express high levels of TRAIL, suggesting that TRAIL-mediated defenses may play a particularly important role in regulating host immune responses at mucosal sites of viral persistence.

DISCUSSION

ILCs are important first-line defenders against viral infection due to their ability to mount fast and robust responses in an antigen-independent manner (30, 31). NK cells were the first described and prototypical ILCs (32, 33). Since then, additional subsets have been defined and subdivided into 3 groups based largely on distinct transcription factor requirements for development and unique cytokine production (15, 16, 20). NK cells fall within the group I ILCs and were long believed to be a largely homogeneous cell population expressing NK1.1 and NKp46, with CD11b and CD27 expression varying based on their activation/maturation state (19). Very recently, however, work from several groups has revealed that NK cells defined by these parameters are composed of at least 2 developmentally distinct subsets, cNK and ILC1, together comprising the group I ILCs (16, 20, 22). Consequently, the relative importance of ILC1 and cNK in mounting specific antiviral responses is only beginning to be deciphered, especially with regard to their key effector mechanisms. The fact that ILC1 reside permanently within multiple tissues suggests that they may play a particularly important role in frontline defense against infection, perhaps partnering with tissue-resident memory T cells to provide a rapid one-two punch. ILC1 can rapidly produce IFN-γ in response to both viruses and bacteria (22, 23, 34), but the roles that other immune effector molecules may play is understudied, and our data suggest that their expression of TRAIL may be particularly important. Using the most recently described markers to differentiate ILC1 and cNK, we investigated the phenotype and function of these 2 subsets in naive and MCMV-infected mice. Consistent with what has been reported for B6 mice (16), we found that SG-resident ILC1 have major phenotypic differences compared to their liver counterparts in BALB/c mice. Illustrating this, liver-resident ILC1 do not express Eomes or CD49b, while a significant proportion of ILC1 in the SG do. This is consistent with the findings of several studies showing that the ILC1 phenotype can vary significantly based upon the specific tissue. However, these differences seem to be particularly notable for SG-resident ILC1, prompting them to be proposed as a unique ILC1 subset (16).

CMV has evolved a multitude of strategies to avoid immune detection, with restriction of TRAIL-DR expression by m166 being one example (13, 14). TRAIL can be induced in multiple innate and adaptive immune cells, with its death receptors also being broadly expressed (35). The use of MCMV-m166stop and several recently described phenotypic markers for group I ILCs allowed us to assess the relative role of cNK and ILC1 in TRAIL-mediated antiviral defense. Additionally, as this work was performed in BALB/c mice, we have confirmed that the cNK and ILC1 phenotypes are largely conserved between these and the widely used B6 strain. Notably, BALB/c mice depleted of group I ILCs showed no increase in persistent replication levels of WT MCMV in the SG, similar to the findings seen in the liver and spleen (14), indicating that this virus can completely neutralize all effector mechanisms used by these innate cells, including TRAIL. This raises interesting questions regarding the relative importance of TRAIL and other group I ILC effector pathways dampened by CMV, such as NKG2D activation (17). It would have been desirable to specifically assess the capacity of ILC1 and cNK to control m166stop in the absence of the other subset, as opposed to depleting all group I ILCs with anti-asialo-GM1. However, to the best of our knowledge, no currently available antibody/chemical reagents can achieve this. A second approach would be to use genetically deficient mice specifically lacking either subset. Potentially suitable mice do exist on the B6 genetic background, as Zfp683−/− mice are reported to lack liver-resident ILC1, and cNK are absent or reduced in the SG of Nfil3−/− mice and in the liver of NCR1CreEomes−/− mice (22, 26, 36). Unfortunately, none of these knockout strains are currently available on the BALB/c background, and the presence of Ly49H+ group I ILCs in B6 mice abrogates the ability to study m166 function in the absence of m157-mediated hyperactivation of these cells (3739).

Past work has shown that MCMV can induce TRAIL expression by group I ILCs in the liver and SG, but whether this was by cNK or ILC1 was not explored at that time (14, 40). More recently, splenic cNK have been shown to be capable of expressing TRAIL in response to transforming growth factor β signaling both in vitro and in vivo (25, 41). Taking advantage of recently identified markers differentiating ILC1 and cNK, we have found that liver-resident cNK express virtually no TRAIL, irrespective of MCMV infection. In contrast, ∼60% of ILC1 are TRAIL+ in naive livers, with this proportion increasing to >80% by 36 h of infection. While ILC1 from multiple tissues can upregulate TRAIL upon stimulation, liver ILC1 uniquely express very high constitutive levels (20), perhaps emanating from basal IFN-γ signaling in this organ (18). Using CD61 to identify SG ILC1, 34% of ILC1 in naive mice were seen to express TRAIL, while only 3% of SG cNK were TRAIL+. Notably, if CD61 was used to segregate liver-resident ILC1 and cNK, results similar to those seen when CD49b and Eomes were used as discriminating markers were seen (Fig. 7). Our data reveal that downregulation of TRAIL-DR cell surface expression by m166 in MCMV-infected cells prevents TRAIL-expressing ILC1 from inducing their apoptosis. Interestingly, MCMV-m166stop is essentially dead in the liver, while in the spleen and SG, replication is reduced ∼10- to 100-fold but remains detectable (14). Perhaps this TRAIL poised state in the liver (∼60% of ILC1 from naive liver express TRAIL) allows ILC1 to respond even more rapidly than other organs with lower constitutive TRAIL expression.

FIG 7.

FIG 7

CD61 expression segregates ILC1 and cNK in the liver and SG. TRAIL expression by liver ILC1 (F4/80 TCRβ CD19 CD3 NKp46+ CD61+) and cNK (F4/80 TCRβ CD19 CD3 NKp46+ CD61) at 36 h of MCMV WT infection. **, P < 0.01.

In contrast to MCMV inducing high numbers of TRAIL-expressing ILC1 in both the liver and SG, <5% of both ILC1 and cNK produced IFN-γ (without ex vivo restimulation). This may be due to a general tolerogenic environment of these organs compared to the spleen, where a much higher proportion of cNK are IFN-γ+ at similar infection times (42, 43). However, a significant proportion of both ILC1 and cNK isolated from infected livers were capable of producing IFN-γ following PMA-Iono restimulation, with ILC1 producing more than cNK (∼70% and 40%, respectively). In contrast, only ∼5% of both subsets isolated from infected SG were IFN-γ+ following this strong ex vivo restimulation. This is consistent with past results showing that SG-resident group I ILCs are hyporesponsive compared to their liver counterparts (29). Notably, at the same day 8 time point, T cells produced high IFN-γ levels following PMA-Iono restimulation (Fig. 8), indicating that not all IFN-γ-dependent immune cell responses are dampened in this mucosal organ.

FIG 8.

FIG 8

IFN-γ hyporesponsiveness in the SG is group I ILC specific. T cells (CD3+ TCRβ+) isolated from the SG of naive mice and mice infected for 8 days were analyzed ex vivo for their expression of IFN-γ after 5 h of incubation with brefeldin A and monensin with or without PMA and ionomycin.

The existence of an HCMV protein, UL141, exerting the same function as m166 strongly suggests that blocking group I ILC-mediated TRAIL-induced apoptosis is critical for viral replication in various hosts (13). Whether analogous populations of mouse ILC1 and cNK exist in humans is an important question. Phenotypically distinct NK cells are present in human peripheral blood and tissues, but whether these represent ILC1 and cNK remains controversial due to overlapping phenotypes and conflicting reports (44, 45). HCMV-infected humans often show expansions of CD57+ NKG2Chigh NK cells in blood, and these cells play an important role in controlling viremia in transplant patients (46). These NK cells have been postulated to be counterparts to memory-like, Ly49H+ cNK that expand in response to m157-mediated activation (46). Distinct populations of CD56bright human NK cells exist and are present at higher levels in tissues than in blood, and whether these CD56bright human NK cells are analogous to ILC1 in mice is being explored (47, 48). Interestingly, a subset of CCR6+ CD56bright NK cells residing in human livers expresses TRAIL when appropriately stimulated, and despite showing phenotypic differences from mouse ILC1, they also highly express the tissue residency marker CD69 (48). Similar to m166, the HCMV UL141 protein inhibits cell surface expression of the human TRAIL-DRs and blocks TRAIL-mediated killing of infected cells (13). These data support the hypothesis that CMV has evolved to block TRAIL-mediated ILC1 defenses in both people and mice, a model that can be directly tested once human group I ILC subsets are better characterized.

MATERIALS AND METHODS

Mice.

All the mice used in the study were generated on a BALB/c background. BALB/c mice were originally purchased from The Jackson Laboratory (Bar Harbor, ME) and subsequently bred in-house. They are defined throughout the article as WT mice. Trail−/− BALB/c mice were obtained from Tom Griffith (University of Minnesota, Minneapolis, MN, USA). They are defined in the article as Trail−/− mice. The Institutional Animal Care and Use Committee (IACUC) of the La Jolla Institute for Immunology approved all the animal experiments.

Reagents and antibodies.

Anti-asialo-GM1 antibody, purchased from Wako, was injected 24 h before infection to deplete group I ILCs. GK1.5, purchased from Bio X Cell, was injected at day 3 preinfection and day 2 postinfection to deplete CD4 T cells. Cellular depletion was verified in spleen and organs by flow cytometry. The following antibodies were purchased from BioLegend: anti-CD3-Alexa Fluor 700 (clone 17A2), anti-CD11b-Brilliant Violet 650 (clone M1/70), anti-CD19-Brilliant Violet 570 (clone 6D5), anti-CD49a-phycoerythrin (PE; clone HMα1), anti-CD49b-fluorescein isothiocyanate (clone DX5), anti-CD61-PE-Cy7 (antibody 2C9.G2; clone HMβ3-1), anti-CD62L-allophycocyanin-Cy7 (clone MEL-14), anti-TCRβ-Brilliant Violet 570 (clone H57-597), anti-TRAIL-biotin (clone N2B2), and anti-streptavidin-Brilliant Violet 605. The following antibodies were purchased from BD Horizon: anti-CD69-PE-CF594 (clone H1.2F3), anti-KLRG1-Brilliant Violet 786 (clone 2F1), and anti-F4/80-Brilliant Violet 570 (clone T45-2342). The following antibodies and reagents were purchased from eBiosciences: anti-IFN-γ-ef450 (clone XMG1.2), brefeldin A, and red blood cell (RBC) lysis buffer. The following antibodies and reagents were purchased from Invitrogen: anti-Eomes-ef450 (clone Dan11mag), Yellow LIVE/DEAD, fixation/permeabilization buffer, and permeabilization buffer. The following antibodies and reagents were purchased from BD Biosciences: anti-NKp46-Brilliant Violet 711 (clone 29A1.4) and GolgiStop protein transport inhibitor. PMA and ionomycin were purchased from Sigma-Aldrich. Percoll was purchased from GE Healthcare. The following reagents were purchased from Gibco: Dulbecco's modified Eagle’s medium (DMEM), 200 mM l-glutamine, penicillin (Pen; 10,000 units/ml), streptomycin (Strep; 10,000 μg/ml), 0.01 M HEPES buffer, phosphate-buffered saline (PBS), and RPMI 1640. The fetal bovine serum (FBS) was purchased from Gemini. The newborn calf serum (NCS) was purchased from Omega Scientific. Collagenase D and DNase were purchased from Sigma. The sodium azide (NaN3) was purchased from Fisher Scientific.

Cell culture.

NIH 3T3 cells were obtained from the ATCC (CRL1658) and were cultured in Dulbecco's modified Eagle’s medium (DMEM) supplemented with 10% NCS, 1% 200 mM l-glutamine, 1% Pen-Strep, and 1% 0.01 M HEPES buffer. All cell cultures were verified to be mycoplasma free. All plaque assays used NIH 3T3 cells.

Generation of CMV stock preparation.

The K181 m166stop mutant virus was generated as described in previous work (14). K181 WT, referred to in this article as MCMV WT, and m166stop, referred to in this article as m166stop, bacterial artificial chromosome-derived viral stocks were generated in NIH 3T3 cells (49), and stock titers were determined in NIH 3T3 cells.

Tissue cell isolation.

Harvested livers were perfused with phosphate-buffered saline (PBS), mashed though a 70-μm-pore-size cell strainer, washed with 1× PBS, and pelleted. Cell slurries were resuspended in 33.75% Percoll solution (in 1× PBS) and centrifuged at room temperature for 12 min (700 × g) to pellet the mononuclear cells. The cells were washed, treated with red blood cell (RBC) lysis buffer for 5 min, washed again, and then counted. Spleens were mashed through a 70-μm-pore-size cell strainer and washed with complete medium prior to RBC lysis, washed again, and counted. Salivary gland (SG) lymphocytes were isolated by mincing submaxillary glands devoid of contaminating sublingual, parotid, or fat tissue, prior to incubating at 37°C with rotation (85 rpm) in complete medium (RPMI 1640 supplemented with 10% fetal bovine serum [FBS], 1% 200 mM l-glutamine, 1% Pen-Strep) with collagenase D (100 μg/ml) and DNase (10 μg/ml) for 20 min. Cells were then mashed through a 70-μm-pore-size strainer, washed, pelleted, and treated with RBC lysis buffer for 5 min, washed again, and counted. To detect cytokine expression, cells isolated from various tissues were incubated in a 96-well plate in complete medium with brefeldin A and GolgiStop protein transport inhibitor (BD) with or without PMA (0.1 μg/ml) and ionomycin (0.5 μg/ml) for 5 h at 37°C in 5% CO2.

Determination of virus replication level in mouse organs.

Age- and sex-matched mice were infected with 103 to 106 PFU of the K181 WT or K181 m166stop virus intraperitoneally (i.p.). The numbers of PFU from the infected spleen, liver, or total SG were quantified at different days following the infection by doing a standard plaque assay in NIH 3T3 cells as described previously (50).

Flow cytometry staining.

Cells isolated from tissues were washed in fluorescence-activated cell sorting (FACS) buffer (1× PBS, 2% FBS, 0.1% NaN3), incubated at 4°C for 20 min with antibodies, and washed again. If streptavidin addition was needed, a second similar incubation step was performed. Cells were then fixed with fixation/permeabilization buffer at 4°C for 20 min, washed twice with FACS buffer, washed once with permeabilization buffer, and then incubated at 4°C for 30 min with the antibodies diluted in permeabilization buffer to detect intracellular cytokine and transcription factor expression. Finally, cells were collected in FACS buffer prior to analysis by flow cytometry.

Statistical analysis.

Acquisition of flow cytometric data was done with an LSR II flow cytometer (BD Biosciences), with data analysis being performed using FlowJo (version X) software (TreeStar). GraphPad Prism (version 7.0c) software (GraphPad Software) was used to create graphs and perform statistical analyses. Differences between groups were evaluated for statistical significance by the two-tailed unpaired Mann-Whitney U test. Results are expressed as the mean ± standard deviation (SD).

ACKNOWLEDGMENTS

This work was supported by NIH grants AI101423 and AI113349 to C.A.B. and NIH grant S10RR027366 to the LJI Flow Cytometry Core.

We thank the staff of the Flow Cytometry Core and the Department of Laboratory Animal Care (DLAC) at LJI for excellent technical assistance. We also thank Mick Croft, Ricardo Da Silva Antunes, and Laurent Brossay for productive discussions.

We declare that we have no conflict of interest with the contents of this article.

G.P., S.V., and C.A.B. conceived, conducted, and performed the experiments. R.G., B.M., and R.E.M. performed the experiments. G.P. and C.A.B. wrote the manuscript. N.T. and T.S.G. provided key input for experimental design and interpretation of the results. C.A.B. acquired the funding, provided resources, and served as the principal investigator.

REFERENCES

  • 1.Cannon MJ, Schmid DS, Hyde TB. 2010. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev Med Virol 20:202–213. doi: 10.1002/rmv.655. [DOI] [PubMed] [Google Scholar]
  • 2.Dasari V, Smith C, Khanna R. 2013. Recent advances in designing an effective vaccine to prevent cytomegalovirus-associated clinical diseases. Expert Rev Vaccines 12:661–676. doi: 10.1586/erv.13.46. [DOI] [PubMed] [Google Scholar]
  • 3.Benedict CA. 2013. A CMV vaccine: TREATing despite the TRICKs. Expert Rev Vaccines 12:1235–1237. doi: 10.1586/14760584.2013.844653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Poole E, Sinclair J. 2015. Sleepless latency of human cytomegalovirus. Med Microbiol Immunol 204:421–429. doi: 10.1007/s00430-015-0401-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cheng S, Caviness K, Buehler J, Smithey M, Nikolich-Žugich J, Goodrum F. 2017. Transcriptome-wide characterization of human cytomegalovirus in natural infection and experimental latency. Proc Natl Acad Sci U S A 114:E10586–E10595. doi: 10.1073/pnas.1710522114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Brodin P, Jojic V, Gao T, Bhattacharya S, Angel CJL, Furman D, Shen-Orr S, Dekker CL, Swan GE, Butte AJ, Maecker HT, Davis MM. 2015. Variation in the human immune system is largely driven by non-heritable influences. Cell 160:37–47. doi: 10.1016/j.cell.2014.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Picarda G, Benedict CA. 2018. Cytomegalovirus: shape-shifting the immune system. J Immunol 200:3881–3889. doi: 10.4049/jimmunol.1800171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yu D, Silva MC, Shenk T. 2003. Functional map of human cytomegalovirus AD169 defined by global mutational analysis. Proc Natl Acad Sci U S A 100:12396–12401. doi: 10.1073/pnas.1635160100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Benedict CA. 2003. Viruses and the TNF-related cytokines, an evolving battle. Cytokine Growth Factor Rev 14:349–357. doi: 10.1016/S1359-6101(03)00030-3. [DOI] [PubMed] [Google Scholar]
  • 10.Croft M, Benedict CA, Ware CF. 2013. Clinical targeting of the TNF and TNFR superfamilies. Nat Rev Drug Discov 12:147–168. doi: 10.1038/nrd3930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.von Karstedt S, Montinaro A, Walczak H. 2017. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer 17:352–366. doi: 10.1038/nrc.2017.28. [DOI] [PubMed] [Google Scholar]
  • 12.Gyurkovska V, Ivanovska N. 2016. Distinct roles of TNF-related apoptosis-inducing ligand (TRAIL) in viral and bacterial infections: from pathogenesis to pathogen clearance. Inflamm Res 65:427–437. doi: 10.1007/s00011-016-0934-1. [DOI] [PubMed] [Google Scholar]
  • 13.Smith W, Tomasec P, Aicheler R, Loewendorf A, Nemčovičova I, Wang ECY, Stanton RJ, Macauley M, Norris P, Willen L, Ruckova E, Nomoto A, Schneider P, Hahn G, Zajonc DM, Ware CF, Wilkinson GWG, Benedict CA. 2013. Human cytomegalovirus glycoprotein UL141 targets the TRAIL death receptors to thwart host innate antiviral defenses. Cell Host Microbe 13:324–335. doi: 10.1016/j.chom.2013.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Verma S, Loewendorf A, Wang Q, McDonald B, Redwood A, Benedict CA. 2014. Inhibition of the TRAIL death receptor by CMV reveals its importance in NK cell-mediated antiviral defense. PLoS Pathog 10:e1004268. doi: 10.1371/journal.ppat.1004268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie ANJ, Mebius RE, Powrie F, Vivier E. 2013. Innate lymphoid cells—a proposal for uniform nomenclature. Nat Rev Immunol 13:145–149. doi: 10.1038/nri3365. [DOI] [PubMed] [Google Scholar]
  • 16.Cortez VS, Colonna M. 2016. Diversity and function of group 1 innate lymphoid cells. Immunol Lett 179:19–24. doi: 10.1016/j.imlet.2016.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lisnić B, Lisnić VJ, Jonjić S. 2015. NK cell interplay with cytomegaloviruses. Curr Opin Virol 15:9–18. doi: 10.1016/j.coviro.2015.07.001. [DOI] [PubMed] [Google Scholar]
  • 18.Takeda K, Cretney E, Hayakawa Y, Ota T, Akiba H, Ogasawara K, Yagita H, Kinoshita K, Okumura K, Smyth MJ. 2005. TRAIL identifies immature natural killer cells in newborn mice and adult mouse liver. Blood 105:2082–2089. doi: 10.1182/blood-2004-08-3262. [DOI] [PubMed] [Google Scholar]
  • 19.Chiossone L, Chaix J, Fuseri N, Roth C, Vivier E, Walzer T. 2009. Maturation of mouse NK cells is a 4-stage developmental program. Blood 113:5488–5496. doi: 10.1182/blood-2008-10-187179. [DOI] [PubMed] [Google Scholar]
  • 20.Peng H, Tian Z. 2017. Diversity of tissue-resident NK cells. Semin Immunol 31:3–10. doi: 10.1016/j.smim.2017.07.006. [DOI] [PubMed] [Google Scholar]
  • 21.Sojka DK, Plougastel-Douglas B, Yang L, Pak-Wittel MA, Artyomov MN, Ivanova Y, Zhong C, Chase JM, Rothman PB, Yu J, Riley JK, Zhu J, Tian Z, Yokoyama WM. 2014. Tissue-resident natural killer (NK) cells are cell lineages distinct from thymic and conventional splenic NK cells. Elife 3:e01659. doi: 10.7554/eLife.01659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Weizman O-E, Adams NM, Schuster IS, Krishna C, Pritykin Y, Lau C, Degli-Esposti MA, Leslie CS, Sun JC, O'Sullivan TE. 2017. ILC1 confer early host protection at initial sites of viral infection. Cell 171:795–808.e12. doi: 10.1016/j.cell.2017.09.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Daussy C, Faure F, Mayol K, Viel S, Gasteiger G, Charrier E, Bienvenu J, Henry T, Debien E, Hasan UA, Marvel J, Yoh K, Takahashi S, Prinz I, de Bernard S, Buffat L, Walzer T. 2014. T-bet and Eomes instruct the development of two distinct natural killer cell lineages in the liver and in the bone marrow. J Exp Med 211:563–577. doi: 10.1084/jem.20131560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jonjic S, Mutter W, Weiland F, Reddehase MJ, Koszinowski UH. 1989. Site-restricted persistent cytomegalovirus infection after selective long-term depletion of CD4+ T lymphocytes. J Exp Med 169:1199–1212. doi: 10.1084/jem.169.4.1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cortez VS, Fuchs A, Cella M, Gilfillan S, Colonna M. 2014. Cutting edge: salivary gland NK cells develop independently of Nfil3 in steady-state. J Immunol 192:4487–4491. doi: 10.4049/jimmunol.1303469. [DOI] [PubMed] [Google Scholar]
  • 26.Erick TK, Anderson CK, Reilly EC, Wands JR, Brossay L. 2016. NFIL3 expression distinguishes tissue-resident NK cells and conventional NK-like cells in the mouse submandibular glands. J Immunol 197:2485–2491. doi: 10.4049/jimmunol.1601099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cortez VS, Cervantes-Barragan L, Robinette ML, Bando JK, Wang Y, Geiger TL, Gilfillan S, Fuchs A, Vivier E, Sun JC, Cella M, Colonna M. 2016. Transforming growth factor-β signaling guides the differentiation of innate lymphoid cells in salivary glands. Immunity 44:1127–1139. doi: 10.1016/j.immuni.2016.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cortez VS, Ulland TK, Cervantes-Barragan L, Bando JK, Robinette ML, Wang Q, White AJ, Gilfillan S, Cella M, Colonna M. 2017. SMAD4 impedes the conversion of NK cells into ILC1-like cells by curtailing non-canonical TGF-β signaling. Nat Immunol 18:995–1003. doi: 10.1038/ni.3809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tessmer MS, Reilly EC, Brossay L. 2011. Salivary gland NK cells are phenotypically and functionally unique. PLoS Pathog 7:e1001254. doi: 10.1371/journal.ppat.1001254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Artis D, Spits H. 2015. The biology of innate lymphoid cells. Nature 517:293–301. doi: 10.1038/nature14189. [DOI] [PubMed] [Google Scholar]
  • 31.Adams NM, Sun JC. 2018. Spatial and temporal coordination of antiviral responses by group 1 ILCs. Immunol Rev 286:23–36. doi: 10.1111/imr.12710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Herberman RB, Nunn ME, Lavrin DH. 1975. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic acid allogeneic tumors. I. Distribution of reactivity and specificity. Int J Cancer 16:216–229. doi: 10.1002/ijc.2910160204. [DOI] [PubMed] [Google Scholar]
  • 33.Kiessling R, Klein E, Wigzell H. 1975. “Natural” killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. Eur J Immunol 5:112–117. doi: 10.1002/eji.1830050208. [DOI] [PubMed] [Google Scholar]
  • 34.Vashist N, Trittel S, Ebensen T, Chambers BJ, Guzmán CA, Riese P. 2018. Influenza-activated ILC1s contribute to antiviral immunity partially influenced by differential GITR expression. Front Immunol 9:505. doi: 10.3389/fimmu.2018.00505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Amarante-Mendes GP, Griffith TS. 2015. Therapeutic applications of TRAIL receptor agonists in cancer and beyond. Pharmacol Ther 155:117–131. doi: 10.1016/j.pharmthera.2015.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mackay LK, Minnich M, Kragten NAM, Liao Y, Nota B, Seillet C, Zaid A, Man K, Preston S, Freestone D, Braun A, Wynne-Jones E, Behr FM, Stark R, Pellicci DG, Godfrey DI, Belz GT, Pellegrini M, Gebhardt T, Busslinger M, Shi W, Carbone FR, van Lier RAW, Kallies A, van Gisbergen K. 2016. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352:459–463. doi: 10.1126/science.aad2035. [DOI] [PubMed] [Google Scholar]
  • 37.Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. 2002. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science 296:1323–1326. doi: 10.1126/science.1070884. [DOI] [PubMed] [Google Scholar]
  • 38.Brown MG, Dokun AO, Heusel JW, Smith HR, Beckman DL, Blattenberger EA, Dubbelde CE, Stone LR, Scalzo AA, Yokoyama WM. 2001. Vital involvement of a natural killer cell activation receptor in resistance to viral infection. Science 292:934–937. doi: 10.1126/science.1060042. [DOI] [PubMed] [Google Scholar]
  • 39.Scalzo AA, Fitzgerald NA, Simmons A, La Vista AB, Shellam GR. 1990. Cmv-1, a genetic locus that controls murine cytomegalovirus replication in the spleen. J Exp Med 171:1469–1483. doi: 10.1084/jem.171.5.1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Schuster IS, Wikstrom ME, Brizard G, Coudert JD, Estcourt MJ, Manzur M, O'Reilly LA, Smyth MJ, Trapani JA, Hill GR, Andoniou CE, Degli-Esposti MA. 2014. TRAIL+ NK cells control CD4+ T cell responses during chronic viral infection to limit autoimmunity. Immunity 41:646–656. doi: 10.1016/j.immuni.2014.09.013. [DOI] [PubMed] [Google Scholar]
  • 41.Gao Y, Souza-Fonseca-Guimaraes F, Bald T, Ng SS, Young A, Ngiow SF, Rautela J, Straube J, Waddell N, Blake SJ, Yan J, Bartholin L, Lee JS, Vivier E, Takeda K, Messaoudene M, Zitvogel L, Teng MWL, Belz GT, Engwerda CR, Huntington ND, Nakamura K, Hölzel M, Smyth MJ. 2017. Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat Immunol 18:1004–1015. doi: 10.1038/ni.3800. [DOI] [PubMed] [Google Scholar]
  • 42.Commins SP. 2015. Mechanisms of oral tolerance. Pediatr Clin North Am 62:1523–1529. doi: 10.1016/j.pcl.2015.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Dou L, Ono Y, Chen Y-F, Thomson AW, Chen X-P. 2018. Hepatic dendritic cells, the tolerogenic liver environment, and liver disease. Semin Liver Dis 38:170–180. doi: 10.1055/s-0038-1646949 (Erratum, 38: 298, doi:.) [DOI] [PubMed] [Google Scholar]
  • 44.Ealey KN, Koyasu S. 2017. How many subsets of innate lymphoid cells do we need? Immunity 46:10–13. doi: 10.1016/j.immuni.2016.12.018. [DOI] [PubMed] [Google Scholar]
  • 45.Simoni Y, Newell EW. 2018. Dissecting human ILC heterogeneity: more than just three subsets. Immunology 153:297–303. doi: 10.1111/imm.12862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lopez-Vergès S, Milush JM, Schwartz BS, Pando MJ, Jarjoura J, York VA, Houchins JP, Miller S, Kang S-M, Norris PJ, Nixon DF, Lanier LL. 2011. Expansion of a unique CD57+NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc Natl Acad Sci U S A 108:14725–14732. doi: 10.1073/pnas.1110900108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Allan DSJ, Cerdeira AS, Ranjan A, Kirkham CL, Aguilar OA, Tanaka M, Childs RW, Dunbar CE, Strominger JL, Kopcow HD, Carlyle JR. 2017. Transcriptome analysis reveals similarities between human blood CD3 CD56bright cells and mouse CD127+ innate lymphoid cells. Sci Rep 7:3501. doi: 10.1038/s41598-017-03256-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Stegmann KA, Robertson F, Hansi N, Gill U, Pallant C, Christophides T, Pallett LJ, Peppa D, Dunn C, Fusai G, Male V, Davidson BR, Kennedy P, Maini MK. 2016. CXCR6 marks a novel subset of T-betloEomeshi natural killer cells residing in human liver. Sci Rep 6:26157. doi: 10.1038/srep26157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lio C-W, McDonald B, Takahashi M, Dhanwani R, Sharma N, Huang J, Pham E, Benedict CA, Sharma S. 2016. cGAS-STING signaling regulates initial innate control of cytomegalovirus infection. J Virol 90:7789–7797. doi: 10.1128/JVI.01040-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Reddehase MJ, Weiland F, Münch K, Jonjic S, Lüske A, Koszinowski UH. 1985. Interstitial murine cytomegalovirus pneumonia after irradiation: characterization of cells that limit viral replication during established infection of the lungs. J Virol 55:264–273. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES