key: cord-0856520-8ed8qf0d authors: Choi, Hanbyeul; Song, Heonju; Jung, Yong Woo title: The Roles of CCR7 for the Homing of Memory CD8+ T Cells into Their Survival Niches date: 2020-05-20 journal: Immune Netw DOI: 10.4110/in.2020.20.e20 sha: 9d6ebfd4cae03d2d42deddfca140cbe932db49bb doc_id: 856520 cord_uid: 8ed8qf0d Memory CD8+ T cells in the immune system are responsible for the removal of external Ags for a long period of time to protect against re-infection. Naïve to memory CD8+ T cell differentiation and memory CD8+ T cell maintenance require many different factors including local environmental factors. Thus, it has been suggested that the migration of memory CD8+ T cells into specific microenvironments alters their longevity and functions. In this review, we have summarized the subsets of memory CD8+ T cells based on their migratory capacities and described the niche hypothesis for their survival. In addition, the basic roles of CCR7 in conjunction with the migration of memory CD8+ T cells and recent understandings of their survival niches have been introduced. Finally, the applications of altering CCR7 signaling have been discussed. The defense against pathogens is composed of innate and adaptive immunities. While innate response mounts in a few hours by the recognition of molecular patterns, adaptive immune system takes a few days to initiate Ag-dependent specific response. The adaptive immune system employs B cells, which produce antibodies, and T cells to mediate cellular immunity. One cardinal feature of the adaptive immune system is "memory" response to the same Ag. Once the adaptive immune system has been stimulated with vaccines or pathogens, challenges with the same Ag induce a faster and stronger response than the primary response. These memory responses are mainly mediated by Ab-producing plasma cells and memory T (T M ) cells. Through these memory responses, vaccines can protect our body against fatal or serious infectious agents. Although vaccination is the most efficient way to prevent infectious disease, few vaccines have been developed to induce functional T M cells for certain infectious diseases, which is probably because of the lack of knowledge about T M cell development and homeostasis. Migratory capacity is another important feature of the adaptive immunity, which enables adaptive immune cells to circulate constantly among tissues. Depending on their activation status, these cells generally circulate throughout the body to search for their cognate Ags. For example, naïve T cells are activated with Ags in the secondary lymphoid organs (SLOs) where adaptive immune responses initiate. When T cells are properly activated, they develop into effector T (T E ) cells and migrate into infected sites. While effector CD4+ T cells provide "help" to other immune cells, effector CD8+ T cells, also called CTLs, kill infected or damaged cells. Hence, T cell homing is crucial to fight against invading pathogens so that their migration is tightly regulated. It has been well documented that stimulation of chemokine receptors and adhesion molecules on T cell surface allows for an orderly access to a specific microenvironment. Therefore, it is known that various chemokine receptors are sequentially expressed in the course of immune response to pathogens. Recently, the migratory capacities of T M cells have been highlighted because their recall response and survival are affected by their localization. In this review, we discussed memory CD8+ T cells and their migratory capacities, particularly the C-C chemokine receptor 7 (CCR7)-dependent pathway, for their survival and longevity. We also highlighted recent studies describing the expression of CCR7 and its ligands, CCL19 and CCL21. Naïve CD8+ T cells constantly circulate throughout the body through SLOs including the lymph nodes (LNs), spleen, and Peyer's patches (PPs) (1) (2) (3) (4) . The activation of T cells was first reported in 1970s using a mouse model of lymphocytic choriomeningitis virus (LCMV) infection (5, 6) . During viral infections, antigen presenting cells (APCs) such as dendritic cells (DCs) obtain viruses and migrate into the SLOs where naïve T cells search for their cognate Ags. Activated CD8+ T cells by these APCs undergo several pathways to proliferate and gain effector functions important to fight against infectious agents. In addition, CD8+ T E cells modulate the expression of their homing receptors to egress from the SLOs and move into the infected area to induce protective immunity (7, 8) . After the pathogens are cleared, majority (up to 90%) of the activated CD8+ T cells dies, while only a fraction (up to 10%) survives and are maintained as T M (9) cells. These T M cells survive for an extended period of time and provide rapid and robust secondary response to the same Ag. In addition, they undergo self-renewal without Ag exposure and subsets of T M cells undergo a series of steps to re-circulate the whole body searching for re-infections. These prominent characteristics enable T M cells to fight against secondary infections with the same pathogens efficiently (10). CD8+ T M cells with these cardinal characteristics can be categorized into diverse subsets including effector memory T (T EM ) (11) , central memory T (T CM ), and tissue-resident memory T (T RM ) cells ( Fig. 1) (12) . These subsets were initially identified based on their differential localizations by the expressions of homing receptors, but they also differ from each other in terms of their cytokine production and proliferation capacities. These abilities are regulated by a series of expression of their receptors, signaling molecules, transcription factors, and other important factors. Of note, different subsets of T M cells form depending on the tropism and characteristics of infectious agents and these subsets cooperate to eradicate infections. T CM cells monitor infection in the body by circulating in the SLOs. In contrast, T EM and T RM cells do not survey SLOs and are located in peripheral tissues (Fig. 1) . However, T EM cells can return to the SLOs if the Ags exist in the SLOs even after Ag is cleared in the infected target area (13) (14) (15) (16) . In 2009, Gebhardt et al. (17) reported that T RM cells stay in the non-lymphoid tissues (NLTs) without returning to the SLOs. They prepare to fight against local infections and are found in many different organs including the brain, lung, gut, skin, and other peripheral organs (18) (19) (20) . The generation of specific subsets of CD8+ T M cells has been attempted using different animal models. It is generally assumed that systemic infections or vaccinations can induce T CM and T RM cell formation, while local infections induce T RM cell development (21) . Thus, Ag tropism or location determines the fate of T M cells. In addition, the generation of certain subsets may be crucial to protect against specific pathogens (22) . For example, the presence of T RM cells in the liver substantially enhances protection against Plasmodium (23) . Altogether, the modulation of T M cell mobility would benefit our body to fight properly against pathogens by placing T M cells in proper positions. A remarkable aspect of T M cells is their longevity and homeostasis without further antigenic stimulation. The underlying mechanisms of their homeostasis are based on the exposure to the homeostatic cytokines such as IL-7 and -15 (24) (25) (26) (27) (28) (29) (30) (31) . IL-7 has been well documented as a survival cytokine of naïve, memory precursor (MP) and T M cells. This cytokine is provided by stromal cells including fibroblastic reticular cells (FRCs) in the spleen and LNs (32) (33) (34) . In conjunction with IL-7, IL-15 can induce homeostatic proliferation of T M cells. IL-15 also helps for the survival of KLRG1 hi terminally differentiated T E and T M cells. Therefore, it is crucial for T M cells to "see" these cytokines in order to develop and maintain homeostasis. These subsets are identified on the basis of differential expressions in several chemokine receptors and adhesion molecules. While T CM cells express high levels of CCR7 and CD62L (L-selectin) (CCR7 hi CD62L hi ), T EM cells express low levels of CCR7 and CD62L (CCR7 lo CD62L lo ). These phenotypes indicate that T CM cells circulate to SLOs, whereas T EM cells circulate to peripheral organs. T RM cells express low levels of CCR7 and high levels of CD69 and CD103 (CCR7 lo CD69 hi CD103 hi ) to remain in peripheral organs. T M cells develop and maintain in multiple organs including the spleen, LNs, liver, lung, and bone marrow (BM) (35). After systemic infection, T M cells can survive and proliferate in these organs, particularly in the BM (36). However, different T M cell subsets are differentially localized within different organs, suggesting that these cells may be exposed to different survival factors depending on their location (37,38). Since leukocyte recruitment is tightly regulated, it is interesting to understand the homing of each subset. CCR7 is a lymphocyte-specific G-protein-coupled receptor with 7 transmembrane spanning alpha helices for CCL19 and CCL21 as ligands. It was first named Epstein-Barr virus (EBV)indicted gene 1, a gene induced by EBV and Burkitt's lymphoma cells in B-lymphocytes. In the same study, it was shown that it plays an important role in response to virus infection and is detected only in B-and T-lymphocytes (39,40). In the late 1990s, a study using CCR7-deficient mice showed that CCR7 plays an important role in controlling T cell movement to SLOs, particularly LNs and PPs. In addition, the formation of T cell zone was abolished due to abnormal T cell migration. After immunization, the migration of mature skin DCs into the LNs resulted in delayed immune response to injected Ags (41,42). Based on this observation, CCR7 has been established as one of the crucial receivers responsible for lymphocyte homing (41). Among the CD8+ T cells, naïve and T CM cells generally express high levels of CCR7 (3,12,43,44), hence they can migrate to the T cell zone of the LNs and spleen. These T cells can be activated in the T cell zone by the APCs and developed into T E cells. During this process, T E cells can move from the T cell zone to the red pulp and the infected area by the downregulation of CCR7 expression (45). Through this regulation of CCR7 expression, CD8+ T cells can find their cognate Ag in the SLOs to be activated and migrated into infected locus. After infections are cleared, T M cells form and circulate to different parts of the body based on the levels of CCR7 expression (45-47). During T E -T M cell transition, CCR7 expression influences the fate of these cells. It was reported that the mRNA levels of CCR7 were more pronounced in memory precursor T cells (MPECs) than in short-lived effector cells (SLECs) (48). In addition, the T CM and T EM cells were found in different locations of the SLOs depending on CCR7 concentration. CCR7 expression was inhibited in T RM cells, the recently identified T M cell subset, making it possible for T RM cells to act as the first line of defense within peripheral tissues (49). Altogether, the regulation of CCR7 expression controls the recruitment and release of CD8+ T cells from SLOs, determining the CD8+ T cell response outcome. The expression of CCR7 on CD8+ T cells is regulated by several transcription factors. In the CCR7 promoter region, there are 3 binding sites specific for protein 1 (Sp1) and one Ets-1binding site (50), which suggests that the increased expression of CCR7 is mediated at least 4/15 https://doi.org/10.4110/in.2020.20.e20 CCR7 Influences the Fate of Memory CD8+ T Cells https://immunenetwork.org partially by transcription factors such as Sp1 and Ets-1 (Fig. 2) (35,50). AP1 and NF-κB were also reported to upregulate CCR7 expression via binding to the CCR7 promoter locus in various cancer cell lines (51-53). In addition, Krüppel-like factor 2 (KLF2) and T cell factor 1 (TCF1) are significantly upregulated in naïve and T CM cells and regulate the expression of several molecules including CCR7, CD62L (L-selectin; encoded by Sell), and sphingosine-1-phosphate receptor 1 in order to modulate the migration into the SLOs (54) . In contrast, CD8+ T EM and T RM cells suppress KLF2 and TCF1, while simultaneously expressing Blimp-1 and suppressing the transcription of CCR7 independently (55) (56) (57) . It was also reported that Forkhead O 1 (FOXO1), another transcription factor, regulates CCR7 transcription during T E -T M cell transition, and not during naïve-CD8+ T E cell transition. A study using FOXO1 knockout (KO) mice demonstrated that FOXO1 promoted enrichment of MP cells, thus wild-type T E cells highly proliferated upon secondary infections in CD8+ T cells compared to FOXO1-deficient T E cells (58) . In a related study, it was observed that FOXO1 expression is significantly higher in MPECs than in SLECs, and it increase the levels of IL-7Ra and TCF7 expression to help create and maintain T CM cells which were derived from MPECs (59). CCR7 expression is also controlled by miRNAs. MiRNAs consisting of small RNA fragments under 23 nucleotides generally bind to 3' UTRs of complementary target sequences on target mRNAs to decay or stabilize the mRNA (60). Using big data analysis, several T-cell-associated genes including CCR7 were discovered to be targeted by miR-21, an anti-apoptotic factor (61-63). MiR-21, a miRNA inhibiting post-transcription levels of CCR7, led to a significant reduction of CCR7 protein expression when naïve and CD4+ T M cells are activated. Indeed, it is activated in inverse correlation to the amount of CCR7 protein (Fig. 2) (64) . Further, it was reported that the expression of CCR7 was adjusted by miRNAs in several cancers, such as let-7α (in breast cancer cells and patients) (65), miR-320d (in oral squamous cell carcinoma) (66), miR-532-3p (in tongue squamous cell carcinoma) (67) and head and neck squamous cell carcinoma (68) , and miR-199a (in human mantle cell lymphoma) (69). Although there is little report of epigenetic regulators for CCR7, it has been documented based on CCR7 gene methylation using big data analysis. CCR7 methylation in human CD8+ T M cells increased in the order of CD8+ T M cell differentiation state: naïve