Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 May 1.
Published in final edited form as: Immunol Lett. 2017 Mar 6;185:32–39. doi: 10.1016/j.imlet.2017.02.012

Understanding Memory CD8+ T cells

Tasleem Samji 1, Kamal M Khanna 1,2,*
PMCID: PMC5508124  NIHMSID: NIHMS862091  PMID: 28274794

Abstract

Memory CD8+ T cells were originally thought to exist as two populations (effector and central memory). In recent years, a third population called resident memory T cells has been discovered and further to this these populations are being divided into different subtypes. Understanding the function and developmental pathways of memory CD8+ T cells is key to developing effective therapies against cancer and infectious diseases. Here we have reviewed what is currently known about all three subsets of memory CD8+ T populations and as to how each population was originally discovered and the developmental pathways of each subpopulation. Each memory population appears to play a distinct role in adaptive immune responses but we are still a long way from understanding how the populations are generated and what roles they play in protection against invading pathogens and if they contribute to the pathogenesis of inflammatory diseases.

Introduction

Broadly speaking, the immune system is divided into the innate and adaptive components. With innate immunity encompassing the initial rapid response of the body to a pathogen and the adaptive response following soon after, which ultimately leads to a long lasting immunological memory. In this review we will focus on the different types of CD8+ T memory subsets.

The first evidence for differentiating the two distinct populations of CD8+ T cells, effector and memory, was demonstrated through studies using lymphocyte choriomeningitis virus (LCMV) infection in mice [1,2]. Here the authors discovered that a large population of antigen specific effector CD8+ T cell population could be found in lymphoid organs early between 7 and 9 days after LCMV infection. They also identified another population that was found to survive long after the infection had cleared. This second memory population was shown to have an increased ability to restrict viral replication upon secondary infection, and thus indicated the importance and potency of memory T cells compared to naive or effector T cells [2]. These seminal studies led the way for investigations to understand how T cell memory is generated.

Naive CD8+ T cells are continuously circulating throughout the body migrating through the blood and secondary lymphoid tissues, such as the lymph nodes, spleen and the Peyer’s Patches, and one school of thought indicates that naïve cells may also migrate through non-lymphoid organs and then back into the blood via the afferent lymph [36]. Once a naive T cell encounters its cognate antigen on an antigen presenting cells (APC) within a secondary lymphoid organ (SLO), the CD8+ T cell undergoes a number of molecular changes leading to the T cell being in an activated state and differentiating into a number of different subpopulations [7,8]. Upon activation, antigen specific CD8 T cells undergo three distinct phases: 1) clonal expansion during which the cells acquire effector functions, 2) contraction of the effector population through activation induced cell death (ACID) with the concomitant survival of a small percentage of memory population, and 3) maturation of the memory population [912]. It should be noted that contraction and the formation of memory are independent of each other because in the absence of contraction, memory cell still form. The contraction phase is controlled by early inflammation [13].

The definition of immunological memory remains contested [14]. In general, memory CD8+ T cells are a long-lived population that are antigen specific and provide an enhanced protective response when the same antigen is encountered again [15,16]. These cells persist in the absence of antigen but maintain a distinct phenotype, and elevated precursor frequency, which is one way of distinguishing them from the naive CD8+ T cell population [12], thus indicating that this population of cells is ready and waiting to act rapidly upon reencountering the antigen. Memory T cells respond consistently to stimulation to distinguish them from the random manner in which a host may respond to a given antigen and this response thus provides an evolutionary advantage to the host in that the host is less likely to fall sick or die upon re-encountering the same antigen and this important trait is passed down through the germline [14]. Recently, this view of immunological memory being a hallmark of the adaptive immune response is being broadened to encompass some innate immune cell populations, such as NK cells, γδ T cells [17,18] or myeloid cells [14], therefore with this in mind it ought to be stated that in this review we will focus on the adaptive immunological memory with specific reference to the CD8+ αβ T cell population.

Identifying CD8+ T Memory Cells

Since the advent of technologies such as monoclonal antibodies, MHC class I tetramers, flow cytometry, fluorescence-activated cell sorting (FACS), and RNA-Sequencing (RNA-Seq), investigators have been able to identify unique subsets of CD8+ T cell memory populations since they express different cell surface markers. It was not until two decades ago when multiple varieties of memory CD8+ T cells were identified [19]. In mice and in humans certain distinct cell surface markers can be used to distinguish memory T cells from effector (TEFF) or naive cells. In mice, typically increased expression of adhesion molecules such as CD11a and CD44 distinguishes effector and memory CD8 T cells from naive CD8 T cells (which express lower levels of CD11a and CD44). Naive CD8 T cells express higher levels of CD62L, CCR7 and CD127 (IL-7Rα). However, activated effector CD8 T cells downregulate these three proteins. CD127 is an important protein that is used to distinguish effector cells from memory T cells [20]. During the early phase of an antimicrobial immune response the majority of effector CD8 T cells express high levels of killer cell lectin-like receptor G1 (KLRG1) and low levels of CD127, which marks the cells as terminally differentiated and are called short lived effector cells (SLECs). Conversely, a small population of TEFF cells retain CD127 expression, but fail to express KLRG1, and survive the contraction phase to become memory T cells, and are labeled as memory precursor effector cells (MPECs).

Studies in humans indicated the importance of CD45RA and CD45RO, as well as a few other markers, as a way of distinguishing circulating effector and memory T cells. In a study by Hamann et al., the authors identified circulating memory population as CD45RA CD27+ and expressed high levels of CD95 (Fas Ligand), CD11a, CD18, CD29, and secreted interleukin-2 (IL-2) and interferon gamma (IFN-γ). In contrast TFFF cells were identified as being CD45RA+ CD27 CD62L but still expressed high levels of CD11a, CD11b, CD18, CD49d, CD95, perforin and granzyme B and were shown to produce IFN-γ and tumor necrosis factor alpha (TNF-α). Interestingly, the effector population had high cytolytic activity without requiring stimulation thus setting one of the differences between effector and memory T cells [21].

Pioneering work by Sallusto et al. showed that the expression of CCR7, a chemokine receptor that regulates homing of CD8+ T cells to SLOs, revealed the difference between naive, central and effector memory T cell subsets. Their work showed that in humans naive CD8+ T cells were CD45RA+ CCR7+, TEFF cells were CD45RA+CCR7, central memory cells (TCM) were CD45RACCR7+ and effector memory cells (TEM) were CD45RA CCR7. This highlighted that there was a difference in memory CD8+ T cells that were found to circulate in the blood and readily available to act versus those that remained in the SLOs and once stimulated are able to efficiently generate a new population of effector cells [19]. Following this study, these two distinct sets of memory CD8+ T cells were identified in the blood of Epstein Barr virus infected patients. In this study, Tussey et al. identified CD8+ T memory cells that were CD62L+CCR7+ but were unable to immediate produce IFN-γ, similar to TCM, and another set that were CD62LCCR7 and were able rapidly produce IFN-γ, similar to TEM cells [22]. More recently, other markers, such as CXC3R1, have been identified that could further distinguish between different CD8+ T cells populations thus indicating that there are a variety of ways to distinguish between the different subsets [23,24].

For some time, it was hypothesized that memory cells existed only in the blood and SLOs. The study by Kim et al. provided the first evidence that memory cells were found in non-lymphoid tissues. Using an OT-I TCR transgenic CD8+ T cell adoptive transfer system, the authors showed that memory cells were localized within the lamina propria and intraepithelial compartments of the intestinal mucosa [25]. Masopust et al. showed that memory cells in mice were present in both lymphoid and non-lymphoid tissues, with a preference of residing in non-lymphoid tissues. The antigen specific CD8+ T cells that were residing in non-lymphoid tissues were able to exert immediate effector functions, similar to TEM, whereas those that were in SLOs, such as the spleen, were not [26].

Soon after the understanding that memory cells resided in non-lymphoid tissues, it became clear that these memory cells were not all the same. A mouse study that focused on T cells found in the lamina propria (LP), indicated that memory cells that resided in this tissue were made up of a distinct set of memory cells that were not found elsewhere and that did not migrate elsewhere, indicating the presence of a different type of memory that is similar to TEM but distinct to TCM [27]. The TEM cells identified in the LP expressed high levels of CD69 which was not seen in TEM cells found in the spleen or blood [28]. Similar high expression of CD69 on memory cells was also found in cells in the lung following a respiratory virus infection [29,30]. Unlike splenic memory CD8 T cells (which were a combination of TCM and TEM cells), those found in the small intestines expressed high levels of granzyme B, CD69, CD103 and β7. Splenic memory CD8 T cells expressed high levels of CD27, CD62L, IL-15Rβ and Ly6C, while the intraepithelial mucosal cells (lELs) expressed low levels of these proteins. Both populations of memory cells expressed high levels of TNF-α and IFN-γ, although surprisingly, IEL memory cells produced much lower levels of IL-2 when compared to splenic memory cells [31].

Further mouse studies showed that memory cells were found in all non-lymphoid tissues irrespective of the site of initial infection. When memory cells from distinct non-lymphoid tissues were transferred into naïve mice they were found to migrate to most tissues, except the LP, with some preference to their original location. This indicated that a number of memory cells in these non-lymphoid tissues are made up of a pool of circulating memory cells whereas others home to a specific tissue, with the exception of LP memory cells that appeared not to circulate outside of the intestinal mucosa [28]. Parabiosis experiments reinforced the idea that some memory cells were able to migrate to lymphoid and some non-lymphoid tissues (such as lung and liver) but other memory cells found in some non-lymphoid tissues, such as the brain, peritoneal cavity and the LP, were less likely to migrate to out of their resident organs indicating the presence of distinct sets of memory cells [32]. Studies using Herpes Simplex Virus type 1 (HSV-1) indicated the presence of a similar type of antigen specific CD8+ T cells that expressed CD69 in the trigeminal ganglion that had the ability to suppress HSV-1 reactivation from latency without killing the neurons harboring the virus [33].

Interestingly, cytotoxic chemotherapy leads to the deletion of circulating T cells, leading to the thought that patients undergoing this type of therapy would be vulnerable to severe infections as their circulating memory T cells would be absent. A study that followed such patients found that they were rarely susceptible to these diseases thus indicating that a subset of non-circulating memory T cells was able to provide an effective immune response to protect these patients from succumbing to infection [34].

This subset of memory T cells that resided permanently within the mucosal tissues and failed to enter the circulation but were similar to, yet distinct from TEM cells were eventually labeled as tissue resident memory T cells (TRM) [35,36].

Since these studies, TRMs have been identified in human tissues as well as mice. The study by Sathaliyawala et al. showed that CD45RO, CD69 and CCR7 expression can be used to identify different populations of TEM, TCM and TRMs, whereas CD45RO and CD103 expression can identify mucosal memory CD8+ T cells in humans [37]. Hombrink et al. identified lung resident memory CD45RA CD8+ T cells from healthy lung tissues from patients undergoing lung resection [38]. Circulating memory cells were in the blood were CD103CD27+ (TCM cells), and CD103CD27+CCR7 (TEM cells), while in the lung, the majority of the cells were CD69+CD103; with a smaller proportion that were CD69+CD103+. In this study the authors chose to focus on the CD69+CD103+ population and used the TEM population found in the blood as a baseline for the transcriptomic comparison. From their analysis they showed that the differences between these two populations were found in genes associated with cytokine signaling, chemokine receptor signaling, active transport of ions and small molecules, apoptosis, TNF receptor signaling, the proinflammatory immune response, glucose deprivation and hypoxia. Migratory receptors that distinguished TRMs from TEMs were high expression of CXCR3, CXCR6, CCR5 and CCR6 and low expression of CX3CR1. High expression of CCR6 and CXCR6 in lung TRMs was not unexpected given that this receptor recognizes CCL20 and CXCL16, respectively, produced by lung epithelial cells. Lung TRMs also expressed high levels of adhesion molecules: CD103, CD51 (also known as VTNR), CD44, CD97, VCAM and Claudin19; as well as chemokines: CCL3 (important for attracting macrophages, CD8+ T cells and CD4+ T cells), CCL4 (important for attracting NK cells and monocytes), CCL20 and XCL1. After in vitro stimulation with phorbol ester, PMA, and ionomycin lung TRMs secreted IFN-γ more rapidly than blood TEM cells reflecting data derived from mouse models. Interestingly, Hombrink et al. showed that T-bet and eomesodermin (EOMES) were not expressed in the human lung CD69+CD103+ TRM population, which is the opposite of what has been observed in mouse skin TRM cells [39]. Along similar lines, the authors identified Notch signaling, through Notch1 and Notch2 receptors, as a key component to maintaining constitutive expression of IFN-γ in human lung TRM’s independent of TCR stimulation, whereas inhibition of Notch signaling in mice did not reduce expression of IFN-γ in mouse lung TRMs.

Even with these differences between human and mouse lungs, the core gene signature of CD103+ TRM versus TEM in human was similar to the core gene signature of CD69+ TRM versus CD69 TRM in mouse lungs. The authors showed that Notch signaling in TRM cells is required for the control of metabolism [38].

Another human study by Wong et al. analyzed the expression profiles of CD8+CD69CD103, CD8+CD69+CD103 and CD8+CD69+CD103+ across several tissues. As expected CD69+CD103 TRMs were present in all non-lymphoid tissues but the blood. Using bioinformatics algorithm on total CD8+ T cells from several different tissues, the authors identified four subtypes of CD69+CD103+ TRMs. CCR5+CD49a+CD45RA+ and CCR5+CD49a+CD45RO+ populations were found at high frequencies in the colon and lung likely due to the high expression of CCL5 (a chemokine that is highly expressed in the lungs and colon). CCR5+CD45RO+CD161+ TRMs that are found in the colon; these are likely to be the mucosal-associated invariant T (MAIT)-like cells. The final subtype was mainly present in the tonsil and spleen and was CXCR5+PD-1+, which are a subtype of cells that were recently identified in the spleens of mice infected with LCMV clone 13 that respond rapidly to treatment with anti-PD-L1 blocking antibody [40]. This study highlighted the diversity between the CD8+ TRM populations in various tissues in humans indicating the necessity of utilizing multiple surface markers to identify specific populations [41].

Similarities and differences between the different types of memory CD8+ T cells

Physiologically, having these three different flavors of memory CD8+ T cells is very advantageous. Due to the presence of very few naive CD8+ T cells that can respond to a given pathogen [42], the time in which a primary response is generated in the draining lymph nodes is large enough the infecting pathogen can cause serious damage to the host. Epithelial and mucosal tissues represent those most vulnerable to attack by pathogens, therefore the presence of resident memory CD8+ T cells that can respond efficiently and rapidly to infection are necessary for protecting animals against debilitating infections. Some pathogens move directly into lymphatic vessels and draining lymph nodes and therefore the presence of memory cells that are either circulating (TEM) or that reside within the lymphoid organs (TCM) are a necessary component of the immune response to previous infections. Thus, TRM appear to exclusively reside within non-lymphoid tissues and do not reenter circulation whereas TCM while mostly residing in SLOs are seen within circulation. TEM cells are motile and recirculate throughout the body. These cells were previously conflated as non-lymphoid tissue resident cells, however, it was later discovered that these cells were in fact present in blood vessels and not in tissue parenchyma [43]. While both TEM and TCM can be found in the spleen TEM are primarily located with the red pulp whereas TCM are mostly located within the white pulp [44].

All types of memory CD8+ T cells respond rapidly to reinfection and are all CD127hi CD44hi CD11ahi, but they are distinct from each other with respect to the expression of the transcriptome and cell surface proteins [45]. TEM and TCM cells are known to be CD69 CD103, where as TRM CD8+ T cells are mostly CD69+ CD103+, although there is some evidence of TRM CD8+ T cells that are CD103 [46]. It should also be noted that there are some TRM cells that have been found to be both CD69 CD103 and so while these prove to be useful markers for TRMs, it is critical to identify TRMs based on their inability to recirculate [47]. CD103 (α chain of α4β7 integrin) binds to E-cadherin, which is mainly expressed on epithelial cells, and has been implicated in mediating survival, retention and localization of TRM cells [36,45,4851]. Both TEM and TRM are CD62L CCR7 due to their exclusion from the lymph nodes and therefore it is only TCM that are CD62L+ CCR7+, which remain in the lymph nodes. One study also identified another population of migratory memory T cells, termed TMM, that were defined as CD62L CCR7+. These were found to be circulating in healthy human skin and were confined to the dermis. TMMs produced an intermediate levels of cytokines compared to TEM and TCM cells and appeared to recirculate more slowly than TCMs [52]. TCM have a much higher proliferative potential than TEM and express CD27 whereas TEM do not. Furthermore, TCM need a much longer period of reactivation in order to express cytolytic markers such as perforin, whereas TEM respond immediately and contain high levels of perforin [53]. After TCR activation, TCM express IL-2 but once they have differentiated into effectors cells they stop producing IL-2 [53]. Interestingly in vitro when activated CD8+ T cells are exposed to IL-2 they are more likely to become TEM, whereas if the activated CD8+ T cells are exposed to IL-15 they acquire a TCM phenotype [3]. CXCR3 is required for the correct amount of TEM formation, in its absence reduced numbers of TEM cells are generated but the recall response is better due to greater numbers of TCM [54]. It has also been shown that TCM express CXCR3 which is important in the recall response and distinguishes them from naïve T cells [55].

A number of transcription factors have been implicated in differentiating memory T cell populations from effector T cells (Table 1). For example, MPECs (which are the precursors to memory cells) are associated with high expression of EOMES ([5658]), inhibitor of DNA binding 3 (ID3, [59,60]), B cell lymphoma 6 (BCL-6, [61]), Signal transducer and Activator of Transcription 3 (STAT3, [62,63]), forehead box protein O1 (FOXO1, [64,65]), serine protease inhibitor 2A (Spi2A, [66]) and transcription factor 1 (TCF-1, [67]). It has also been shown that high expression of Fas ligand (FasL) leads to formation of TCM over TEM [68]. Similarly the differential expression of ID2 and ID3 leads to the formation of either TEM or TCM [60]. Expression of the transcription factor Blimp-1 is required for TEM formation over TCM, although the study by Kallies et al. showed that Blimp-1 expression is more important for efficient recall responses. One possibility is that Blimp-1 maybe important for early programming of memory T cells as its expression is control by IL-2 [69]. Eomes and TCF-1 is required for TCM cells formation over TEM, with TCF-1-Wnt signaling pathway inducing expression of Eomes [56,67]. TRM cells express low levels of Kruppel Like Factor 2 (KLF2) leading to downregulation of the sphingosine 1-phosphate receptor-1 (S1PR1) [70]. The downregulation of KLF2 in TRMs is due to the phosphatidylinositol-3-OH kinase - Akt pathway [70]. Downregulation of S1PR1 allows for increased expression of CD69 and thus, prevents egress of TRM cells from non-lymphoid tissues [71]1. Recently it was shown that the transcription factor Homolog of Blimp1 in T cells (Hobit) is specifically upregulated in all TRM cells [72]. Table 1 provides a list of known transcription factors that are important for the formation of MPECs and memory CD8+ T cells.

Table 1.

Table of important Transcription Factors. Unless indicated the listed transcription factors are highly expressed in the specific populations. The gene name and protein names are how these genes are identified in the mouse genome, unless indicated as human.

Population Gene encoding Transcription Factor Protein Name of Transcription Factor Reference
MPEC (Memory Precursor Effector Cell) Eomes Eomesodermin Banerjee et al. J. I. (2010)
Pearce et al. Science (2003) Intlekofer et al. Nature Immunology (2005)
ID3 ID3 Ji et al. Nature Immunology (2011) Yang et al. Nature Immunology (2011)
BCL-6 BCL-6 Yoshida et al. J. I. (2006)
STAT3 STAT3 Cui et al. Immunity (2011) Siegel et al. Immunity (2011)
FOXO1 FOXO1 Hess Michelini et al. J.E.M. (2013) Kim et al. Immunity (2013)
Serpina3g Spi2A Liu et al. Nature Immunology (2004)
TCF-7 TCF-1 Zhou et al. Immunity (2010)
Memory (Effector and Central Memory combined) Fos Fos Kaech et al. Cell (2002), Wherry et al. Immunity (2007)
ATF-2 ATF-2 Kaech et al. Cell (2002)
ROR-α ROR-α Kaech et al. Cell (2002)
Jun B Jun B Kaech et al. Cell (2002), Wherry et al. Immunity (2007)
BHLHE40 SHARP-2 Wherry et al. Immunity (2007)
Eomes Eomesodermin Wherry et al. Immunity (2007)
Fosb Fosb Wherry et al. Immunity (2007)
Hopx Hop Wherry et al. Immunity (2007)
ID2 ID2 Wherry et al. Immunity (2007)
Klf4 Klf4 Wherry et al. Immunity (2007)
Runx2 Runx2 Wherry et al. Immunity (2007)
Cebpb Cebpb Wherry et al. Immunity (2007)
Myb (downregulated) Myb Wherry et al. Immunity (2007)
TCM (Central Memory) FasL FasL Dudani et al. J.I. (2008)
ID3 ID3 Yang et al. Nature Immunology (2011)
Eomes Eomesodermin Banerjee et al. J.I. (2010)
TCF-7 TCF-1 Zhou et al. Immunity (2010)
Forced expression of Bcl-6 + inactivation of Tbx21, Prdm1 and ID2 T-bet (Tbx21) Blimp-1 (Prdm1) Cannarile, et al (2006 Ichii et al (2004) Intlekofer et al (2007) Rutishuaser et al. (2009)
KLF2 KLF2 Skon et al. Nature Immunology (2013)
TEM (Effector Memory) Prdm1 [not necessary for maintenance of memory but required for efficient recall response; and migration of cells to the lung for influenza infection] Blimp1 Kallies et al. Immunity (2009)
KLF2 KLF2 Skon et al. Nature Immunology (2013)
ASCL2 (human) MASH2 or ASH2 Hombrink et al. Nature Immunology (2016)
RUNX1 (human) Runx1 Hombrink et al. Nature Immunology (2016)
LEF1 (human) TCF10 Hombrink et al. Nature Immunology (2016)
FOXP1 (human) Foxp1 Hombrink et al. Nature Immunology (2016)
KLF12 (human) AP2 repressor Hombrink et al. Nature Immunology (2016)
ZNF511 (human) Znf511 Hombrink et al. Nature Immunology (2016)
IKZF5 (human) Znfn1A5 or PEGASUS Hombrink et al. Nature Immunology (2016)
GTF2H2 (human) TFIIH or BTF2 or P44 Hombrink et al. Nature Immunology (2016)
RERE (human) Arp or Arg Hombrink et al. Nature Immunology (2016)
KLF7 (human) Klf7 Hombrink et al. Nature Immunology (2016)
ZNF318 (human) Zfp318 or Tzf Hombrink et al. Nature Immunology (2016)
TFDP2 (human) Dp2 Hombrink et al. Nature Immunology (2016)
BBX (human) HBP2 or ARTC1 Hombrink et al. Nature Immunology (2016)
HOXB7 (human) Hox2 Hombrink et al. Nature Immunology (2016)
ZNF274 (human) Zf2 or Zscan51 Hombrink et al. Nature Immunology (2016)
ZNF638 (human) Np220 or Zfml Hombrink et al. Nature Immunology (2016)
ZNF365 (human) Talanin or Uan Hombrink et al. Nature Immunology (2016)
EWSR1 (human) Ews Hombrink et al. Nature Immunology (2016)
ZNF18 (human) Zfp535 or Kox11 Hombrink et al. Nature Immunology (2016)
CNOT7 (human) Hcaf-1 Hombrink et al. Nature Immunology (2016)
ZNF22 (human) Zfp422 or Kox15 Hombrink et al. Nature Immunology (2016)
ZNF428 (human) Zfp428 or C19orf37 Hombrink et al. Nature Immunology (2016)
FOXN3 (human) C14orf116 or Ches1 Hombrink et al. Nature Immunology (2016)
ZNF83 (human) Znf816B or HPF1 Hombrink et al. Nature Immunology (2016)
GTF3A (human) TFIIIA or AP2 Hombrink et al. Nature Immunology (2016)
TRM (Resident memory) Litaf Litaf Mackay et al. Nature Immunology (2013)
Nr4a1 Nur77 Mackay et al. Nature Immunology (2013); Hombrink et al. Nature Immunology (2016)
Nr4a2 Nurr1 Mackay et al. Nature Immunology (2013)
KLF2 (low levels) KLF2 Skon et al. Nature Immunology (2013)
Zfp683 Hobit Mackay et al. Science (2015)
ETS1 (human) Ets-1 or EWSR2 Hombrink et al. Nature Immunology (2016)
ZFP36L1 (human) Cmg1 or Brf1 Hombrink et al. Nature Immunology (2016)
GPBP1 (human) Gpbp or Vasculin Hombrink et al. Nature Immunology (2016)
NFE2L2 (human) NRF2 or HEBP1 Hombrink et al. Nature Immunology (2016)
RELA (human) p65 or NFKB3 Hombrink et al. Nature Immunology (2016)
FOS (human) Ap-1 or p55 Hombrink et al. Nature Immunology (2016)
NFAT5 (human) NFAT5 Hombrink et al. Nature Immunology (2016)
ZC3H12A (human) MCPIP Hombrink et al. Nature Immunology (2016)
NOTCH1 (human) Notch1 Hombrink et al. Nature Immunology (2016)
MYB (human) c-Myb Hombrink et al. Nature Immunology (2016)
KLF5 (human) Klf5 OR Bteb2 Hombrink et al. Nature Immunology (2016)
CITED2 (human) Mrg1 or VSD2 Hombrink et al. Nature Immunology (2016)
NR3C1 (human) GRL Hombrink et al. Nature Immunology (2016)
REL (human) c-Rel Hombrink et al. Nature Immunology (2016)
ZNF32 (human) Kox30 Hombrink et al. Nature Immunology (2016)
AHR (human) AhR or BHLHe76 Hombrink et al. Nature Immunology (2016)
TWIST1 (human) Twist or BHLHa38 Hombrink et al. Nature Immunology (2016)
EPAS1 (human) Epas-1 or Hif2alpha Hombrink et al. Nature Immunology (2016)
KLF6 (human) Bcd1 or ST12 or COPEB Hombrink et al. Nature Immunology (2016)
NFKB1 (human) EBP-1 or p50 Hombrink et al. Nature Immunology (2016)
CREM (human) CDREM-2 or ICER Hombrink et al. Nature Immunology (2016)
NR4A3 (human) Nor1 or Chn Hombrink et al. Nature Immunology (2016)
NFKB2 (human) Lyt10 or H2TF1 or p52 Hombrink et al. Nature Immunology (2016)
BATF (human) Batf or SFA2 Hombrink et al. Nature Immunology (2016)
IRF4 (human) Irf4 Hombrink et al. Nature Immunology (2016)
RBPJ (human) RBPJ Kappa or CSL or SUH Hombrink et al. Nature Immunology (2016)
PBX4 (human) Pbx4 Hombrink et al. Nature Immunology (2016)
ATF3 (human) Atf3 Hombrink et al. Nature Immunology (2016)
FOSB (human) FosB or AP-1 Hombrink et al. Nature Immunology (2016)
BACH2 (human) Bach2 Hombrink et al. Nature Immunology (2016)
RUNX3 (human) Runx3 or CBFA3 Hombrink et al. Nature Immunology (2016)
EGR2 (human) Egr2 or Krox20 Hombrink et al. Nature Immunology (2016)

Ultimately expression of the various transcription factors leads to the expression of cytokines such as IL-2, IL-7 and IL-15, or receptors that respond to the presence of specific cytokines, such as CD122, which is the receptor for responding to the presence of IL-15. Indeed, the local milieu provides the instructive signals to cells indicating what type of memory T cell is generated [73,74]. It is known that if CD8+ T cells are exposed to IL-15 they are more likely to form memory T cells versus effector T cells, whereas exposure of cells to IL-2 produces poor memory [3,75]. Memory CD8+ T cells also required IL-7 for survival [76], It has been shown that IL-15, TGF-β, IL-33 and tumor necrosis factor (TNF) are required for the formation and survival of skin TRMs. In vitro, TGF-β and IL-33 combined led to downregulation of KLF2 and interestingly so did the combination of IL-12 and IL-18, overall indicating that these combination of cytokines could be important for the formation of TRMs [45,70], TGF-β is known to signal mainly through the Smad pathway. A recent study shows that signaling via that TGF-β-Smad4 pathway is not required for the formation of TRMs but interestingly, TGF-β is not necessary for development of circulating memory cells but Smad4 is [77]. Furthermore, the downregulation of T-bet and Eomes is necessary for TGF-β signaling to occur for the formation of TRM cells but that a low levels of T-bet expression is still required to enable the TRMs to be responsive to IL-15 [39].

The origin of the different memory cells has been subject of much discussion [73,7883] and so we will briefly cover some of the most recent advances in this manuscript. Recent evidence points towards TCM and TRM having the same clonal origin [80], although it should be noted that this study did not pursue the origin of TEM cells. A microarray study comparing the transcriptional profile of skin, gut and lung TRMs, to splenic naïve, TCM and TEM indicated that these circulating cells have a very different transcriptional profile compared to the TRMs. Even more interesting was the finding that TRMs found in the skin had a transcriptional profile that is different from the gut and lung TRMs. This study identified 37 transcripts that were common to TRMs but were different from the TCM and TEM cells including the transcription factors, such as lipopolysaccharide-induced tumor-necrosis factor (Litaf) and nuclear receptor subfamily 4 group A, member 1 (Nr4a2) were upregulated in TRMs compared to TCM and TEM cells. This study came to the conclusion that TRM cells originated from the same precursors of TCM and TEM and that TRMs develop due their location and environmental milieu [45].

Memory T cells and Disease

To date relatively little is known about how aberrant memory CD8+ T cell responses can lead to specific inflammatory diseases.

Fixed-drug eruption occurs when a patient is treated with a specific drug whose adverse reaction leads to the formation of a cutaneous lesion, which may recur in the same location (as well as new locations) if the patient is treated with the same drug. Interestingly, while a fixed drug eruption manifests as a skin lesion it does not lead to systemic symptoms indicating that it is caused by cells that do not circulate and reside in the skin. Recent studies have indicated that fixed drug eruptions are due to the presence of large numbers of TEM cells that express CD69 and are found in the intraepidermis. Considering the location of these cells in the skin, and that they were distinguished from dermal and circulating CD8+ T cells, indicates that they are actually TRM cells rather than TEM cells. Mizukawa et al. showed that TRMs isolated from coetaneous lesions produced high levels of IFN-γ. This led to localized epidermal injury, thus indicating that the TRM cells are the cause of the fixed drug eruption [84].

Similarly, plaque psoriasis is a chronic inflammatory skin disease, caused by infiltration of T cells into the epidermis. While current treatments can lead to remission of the disease, it can recur in the same location when treatment is stopped. This led to the concept that the disease is due to activated T cells that reside within the areas where the lesions occur. The study by Cheuk et al. showed that a high proportion of the epidermal T cells in diseased lesions are CD103+ CD49a+ CD8+ T compared to those in normal skin. The CD8+ T cells isolated from the epidermis of psoriasis plaques expressed high levels of IL-12, IL-22, IFN-γ, granzyme A, granzyme B and perforin compared to healthy skin, indicating that they were highly active cells. Furthermore, high numbers of these CD103+ CD49a+ CD8+ T cells remained in the same region of the lesions even after the psoriasis was successfully treated, and were capable of being reactivated to produce inflammatory cytokines. This indicated that pathogenic TRM cells survived long-term at the site of the inflammatory disease and were active participants in the pathogenesis and persistence of psoriasis [85]. A similar role for TRM cells was found in the study of patients with cutaneous T cell lymphoma (CTCL) [52]. Together, these studies indicate that pathogenic TRMs may lead to inflammatory disease in humans.

Memory subsets other than TRM cells have also been implicated in human diseases. Patients who receive allogenic hemopoeitic stem cell transplantation to cure hematologic malignancies, such as acute and chronic leukemia and lymphoma, are susceptible to graft versus host disease (GVHD) due to the transplantation of donor T cells. A report by Chen et al. indicated that while donor TCM cells do respond to alloantigens they do not cause GVHD disease in the B6(H-2b) -> BALB/c (H-2d) fully mismatched allogeneic bone marrow transplant model [86]. However, in another study TCM cells were shown to only partially participate in the development of GVHD [87]. Thus, the precise role of these memory subset of T cells in GVHD pathogenesis remains to be fully elucidated.

The role for memory CD8 T cells in the pathogenesis of other inflammatory diseases such as asthma has also been investigated. Several studies have implied a role of IL-6, IL-15, IL-13 and CD8+ T cells in the development of asthma [8890]. Indeed, a distinct population of TEM cells expressing high levels of IL-6Rα (also known as CD126) was found at a greater frequency in asthma patients. Althoug it was unclear whether their increased presence led to higher disease severity [91], these cells were shown to produce high levels of IL-15, IL-13, IFN-γ and GATA3 compared with TEM cells that express low levels of IL-6Rα. Furthermore, this population also expressed higher levels CCR4, which in important in recruiting T cells into the airways of asthma patients [91]. While the biological significance of these subsets of TEMs in asthma patients needs to be fully determined, this study did suggest that TEM cells may have a role in the pathogenesis of asthma.

Conclusion

Over the years, immunologists have identified a number of circulating and resident CD8+ memory T cells. Each subset of memory T cells appears to have a discreet and important role in ensuring that we are protecting from pathogens. With better technologies that are available we are realizing that there may be many more memory populations present than the original two circulating populations that were discovered by Volkert et al in 1974 [1]. While we have identified several important populations (TEM, TCM and TRM) much about them remains a mystery.

It seems surprising that very few diseases have been associated with aberrant memory CD8+ T cells and more studies into this need to be performed. While we know about a number of transcription factors, cytokines and cell surface markers that are associated with each memory subset, much more information needs to be uncovered to help us understand the molecular pathways and the necessary environment that lead to the formation of these T cells. This information is critical to enable us to develop highly effective and long-lived therapeutics.

Highlights.

  • Memory CD8 T cell differentiation is a complex process

  • Several subset of memory CD8 T cells have been identified

  • Here we discuss the most current understanding of different subsets of memory CD8 T cells and their role in antimicrobial immune responses

Acknowledgments

This work was supported by National Institute of Health grant AI097375 and AI041576 to K.M.K.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Bibliography

  • 1.Volkert M, Marker O, Bro-Jorgensen K. Two populations of T lymphocytes immune to the lymphocytic choriomeningitis virus. Journal of Experimental Medicine. 1974;139:1329–1343. doi: 10.1084/jem.139.5.1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Johnson ED, Cole GA. Functional heterogeneity of lymphocytic choriomeningitis virus-specific T lymphocytes. I. Identification of effector and memory subsets. J Exp Med. 1975;141:866–881. [PMC free article] [PubMed] [Google Scholar]
  • 3.Weninger W, Crowley MA, Manjunath N, von Andrian UH. Migratory properties of naive, effector, and memory CD8(+) T cells. J Exp Med. 2001;194:953–966. doi: 10.1084/jem.194.7.953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Butcher EC, Williams M, Youngman K, Rott L, Briskin M. Lymphocyte Trafficking and Regional Immunity. Elsevier; 1999. pp. 209–253. [DOI] [PubMed] [Google Scholar]
  • 5.Campbell JJ, Murphy KE, Kunkel EJ, Brightling CE, Soler D, Shen Z, et al. CCR7 Expression and Memory T Cell Diversity in Humans. J Immunol. 2001;166:877–884. doi: 10.4049/jimmunol.166.2.877. [DOI] [PubMed] [Google Scholar]
  • 6.Cose S, Brammer C, Khanna KM, Masopust D, Lefrançois L. Evidence that a significant number of naive T cells enter non-lymphoid organs as part of a normal migratory pathway. Eur J Immunol. 2006;36:1423–1433. doi: 10.1002/eji.200535539. [DOI] [PubMed] [Google Scholar]
  • 7.Huster KM, Busch V, Schiemann M, Linkemann K, Kerksiek KM, Wagner H, et al. Selective expression of IL-7 receptor on memory T cells identifies early CD40L-dependent generation of distinct CD8+ memory T cell subsets. Proceedings of the National Academy of Sciences. 2004;101:5610–5615. doi: 10.1073/pnas.0308054101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Joshi NS, Cui W, Chandele A, Lee HK, Urso DR, Hagman J, et al. Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity. 2007;27:281–295. doi: 10.1016/j.immuni.2007.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Homann D, Teyton L, Oldstone MB. Differential regulation of antiviral T-cell immunity results in stable CD8+ but declining CD4+ T-cell memory. Nat Med. 2001;7:913–919. doi: 10.1038/90950. [DOI] [PubMed] [Google Scholar]
  • 10.Razvi ES, Welsh RM. Programmed cell death of T lymphocytes during acute viral infection: a mechanism for virus-induced immune deficiency. Journal of Virology. 1993;67:5754–5765. doi: 10.1128/jvi.67.10.5754-5765.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Murali-Krishna K, Altman JD, Suresh M, Sourdive DJ, Zajac AJ, Miller JD, et al. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity. 1998;8:177–187. doi: 10.1016/S1074-7613(00)80470-7. [DOI] [PubMed] [Google Scholar]
  • 12.Lau LL, Jamieson BD, Somasundaram T, Ahmed R. Cytotoxic T-cell memory without antigen. Nature. 1994;369:648–652. doi: 10.1038/369648a0. [DOI] [PubMed] [Google Scholar]
  • 13.Badovinac VP, Porter BB, Harty JT. CD8+ T cell contraction is controlled by early inflammation. Nat Immunol. 2004;5:809–817. doi: 10.1038/ni1098. [DOI] [PubMed] [Google Scholar]
  • 14.Farber DL, Netea MG, Radbruch A, Rajewsky K, Zinkernagel RM. Immunological memory: lessons from the past and a look to the future. Nat Rev Immunol. 2016;16:124–128. doi: 10.1038/nri.2016.13. [DOI] [PubMed] [Google Scholar]
  • 15.Doherty PC, Hou S, Tripp RA. CD8+ T-cell memory to viruses. Curr Opin Immunol. 1994;6:545–552. doi: 10.1016/0952-7915(94)90139-2. [DOI] [PubMed] [Google Scholar]
  • 16.Mackay CR. Naive and memory T cells show distinct pathways of lymphocyte recirculation. J Exp Med. 1990;171:801–817. doi: 10.1084/jem.171.3.801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sheridan BS, Romagnoli PA, Pham Q-M, Fu H-H, Alonzo F, III, Schubert W-D, et al. γδ t Cells Exhibit Multifunctional and Protective Memory in Intestinal Tissues. Immunity. 2013;39:184–195. doi: 10.1016/j.immuni.2013.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Romagnoli PA, Sheridan BS, Pham Q-M, Lefrançois L, Khanna KM. IL-17A-producing resident memory γδ T cells orchestrate the innate immune response to secondary oral Listeria monocytogenes infection. Proc Natl Acad Sci USA. 2016;113:8502–8507. doi: 10.1073/pnas.1600713113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712. doi: 10.1038/44385. [DOI] [PubMed] [Google Scholar]
  • 20.Kaech SM, Tan JT, Wherry EJ, Konieczny BT, Surh CD, Ahmed R. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat Immunol. 2003;4:1191–1198. doi: 10.1038/ni1009. [DOI] [PubMed] [Google Scholar]
  • 21.Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, Klein MR, et al. Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med. 1997;186:1407–1418. doi: 10.1002/eji.1830181102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Tussey L, Speller S, Gallimore A, Vessey R. Functionally distinct CD8+ memory T cell subsets in persistent EBV infection are differentiated by migratory receptor expression. Eur J Immunol. 2000;30:1823–1829. doi: 10.1002/1521-4141(200007)30:7<1823∷AID-IMMU1823>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • 23.Böttcher JP, Beyer M, Meissner F, Abdullah Z, Sander J, Höchst B, et al. Functional classification of memory CD8(+) T cells by CX3CR1 expression. Nat Commun. 2015;6:8306. doi: 10.1038/ncomms9306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gerlach C, Moseman A, Loughhead S. CX3CR1 distinguishes three antigen-experienced CD8 T cell subsets with distinct migratory, functional and homeostatic properties. The Journal of …. 2016 [Google Scholar]
  • 25.Kim SK, Schluns KS, Lefrançois L. Induction and visualization of mucosal memory CD8 T cells following systemic virus infection. J Immunol. 1999;163:4125–4132. doi: 10.1016/S1074-7613(00)80540-3. [DOI] [PubMed] [Google Scholar]
  • 26.Masopust D, Vezys V, Marzo AL, Lefrançois L. Preferential Localization of Effector Memory Cells in Nonlymphoid Tissue. Science. 2001;291:2413–2417. doi: 10.1126/science.1058867. [DOI] [PubMed] [Google Scholar]
  • 27.Masopust D, Jiang J, Shen H, Lefrançois L. Direct analysis of the dynamics of the intestinal mucosa CD8 T cell response to systemic virus infection. J Immunol. 2001;166:2348–2356. doi: 10.4049/jimmunol.166.4.2348. [DOI] [PubMed] [Google Scholar]
  • 28.Masopust D, Vezys V, Usherwood EJ, Cauley LS, Olson S, Marzo AL, et al. Activated primary and memory CD8 T cells migrate to nonlymphoid tissues regardless of site of activation or tissue of origin. J Immunol. 2004;172:4875–4882. doi: 10.4049/jimmunol.172.8.4875. [DOI] [PubMed] [Google Scholar]
  • 29.Marshall DR, Turner SJ, Belz GT, Wingo S, Andreansky S, Sangster MY, et al. Measuring the diaspora for virus-specific CD8+ T cells. Proceedings of the National Academy of Sciences. 2001;98:6313–6318. doi: 10.1073/pnas.101132698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hogan RJ, Usherwood EJ, Zhong W, Roberts AD, Dutton RW, Harmsen AG, et al. Activated Antigen-Specific CD8+ T Cells Persist in the Lungs Following Recovery from Respiratory Virus Infections. J Immunol. 2001;166:1813–1822. doi: 10.4049/jimmunol.166.3.1813. [DOI] [PubMed] [Google Scholar]
  • 31.Masopust D, Vezys V, Wherry EJ, Barber DL, Ahmed R. Cutting Edge: Gut Microenvironment Promotes Differentiation of a Unique Memory CD8 T Cell Population. J Immunol. 2006;176:2079–2083. doi: 10.4049/jimmunol.176.4.2079. [DOI] [PubMed] [Google Scholar]
  • 32.Klonowski KD, Williams KJ, Marzo AL, Blair DA, Lingenheld EG, Lefrançois L. Dynamics of blood-borne CD8 memory T cell migration in vivo. Immunity. 2004;20:551–562. doi: 10.1016/s1074-7613(04)00103-7. [DOI] [PubMed] [Google Scholar]
  • 33.Khanna KM, Bonneau RH, Kinchington PR, Hendricks RL. Herpes simplex virus-specific memory CD8+ T cells are selectively activated and retained in latently infected sensory ganglia. Immunity. 2003;18:593–603. doi: 10.1016/s1074-7613(03)00112-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Turtle CJ, Swanson HM, Fujii N, Estey EH, Riddell SR. A distinct subset of self-renewing human memory CD8+ T cells survives cytotoxic chemotherapy. Immunity. 2009;31:834–844. doi: 10.1016/j.immuni.2009.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wakim LM, Waithman J, van Rooijen N, Heath WR, Carbone FR. Dendritic cell-induced memory T cell activation in nonlymphoid tissues. Science. 2008;319:198–202. doi: 10.1126/science.1151869. [DOI] [PubMed] [Google Scholar]
  • 36.Gebhardt T, Wakim LM, Eidsmo L, Reading PC, Heath WR, Carbone FR. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat Immunol. 2009;10:524–530. doi: 10.1038/ni.1718. [DOI] [PubMed] [Google Scholar]
  • 37.Sathaliyawala T, Kubota M, Yudanin N, Turner D, Camp P, Thome JJC, et al. Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity. 2013;38:187–197. doi: 10.1016/j.immuni.2012.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hombrink P, Helbig C, Backer RA, Piet B, Oja AE, Stark R, et al. Programs for the persistence, vigilance and control of human CD8(+) lung-resident memory T cells. Nat Immunol. 2016;17:1467–1478. doi: 10.1038/ni.3589. [DOI] [PubMed] [Google Scholar]
  • 39.Mackay LK, Wynne-Jones E, Freestone D, Pellicci DG, Mielke LA, Newman DM, et al. T-box Transcription Factors Combine with the Cytokines TGF-β 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]
  • 40.Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, et al. Defining CD8(+) T cells that provide the proliferative burst after PD-1 therapy. Nature. 2016;537:417–421. doi: 10.1038/nature19330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wong MT, Ong DEH, Lim FSH, Teng KWW, McGovern N, Narayanan S, et al. A High-Dimensional Atlas of Human T Cell Diversity Reveals Tissue-Specific Trafficking and Cytokine Signatures. Immunity. 2016;45:442–456. doi: 10.1016/j.immuni.2016.07.007. [DOI] [PubMed] [Google Scholar]
  • 42.Obar JJ, Khanna KM, Lefrançois L. Endogenous naive CD8+ T cell precursor frequency regulates primary and memory responses to infection. Immunity. 2008;28:859–869. doi: 10.1016/j.immuni.2008.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Anderson KG, Sung H, Skon CN, Lefrançois L, Deisinger A, Vezys V, et al. Cutting edge: intravascular staining redefines lung CD8 T cell responses. The Journal of Immunology. 2012;189:2702–2706. doi: 10.4049/jimmunol.1201682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jung YW, Rutishauser RL, Joshi NS, Haberman AM, Kaech SM. Differential localization of effector and memory CD8 T cell subsets in lymphoid organs during acute viral infection. The Journal of Immunology. 2010;185:5315–5325. doi: 10.4049/jimmunol.1001948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mackay LK, Rahimpour A, Ma JZ, Collins N, Stock AT, Hafon ML, 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]
  • 46.Mueller SN, Mackay LK. Tissue-resident memory T cells: local specialists in immune defence. Nat Rev Immunol. 2015 doi: 10.1038/nri.2015.3. [DOI] [PubMed] [Google Scholar]
  • 47.Steinert EM, Schenkel JM, Fraser KA, Beura LK, Manlove LS, Igyártó BZ, et al. Quantifying Memory CD8 T Cells Reveals Regionalization of Immunosurveillance. Cell. 2015;161:737–749. doi: 10.1016/j.cell.2015.03.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cepek KL, Shaw SK, Parker CM, Russell GJ, Morrow JS, Rimm DL, et al. Adhesion between epithelial cells and T lymphocytes mediated by E-cadherin and the alpha E beta 7 integrin. Nature. 1994;372:190–193. doi: 10.1038/372190a0. [DOI] [PubMed] [Google Scholar]
  • 49.Pauls K, Schön M, Kubitza RC, Homey B, Wiesenborn A, Lehmann P, et al. Role of integrin alphaE(CD103)beta7 for tissue-specific epidermal localization of CD8+ T lymphocytes. J Invest Dermatol. 2001;117:569–575. doi: 10.1046/j.0022-202x.2001.01481.x. [DOI] [PubMed] [Google Scholar]
  • 50.Hofmann M, Pircher H. E-cadherin promotes accumulation of a unique memory CD8 T-cell population in murine salivary glands. Proc Natl Acad Sci USA. 2011;108:16741–16746. doi: 10.1073/pnas.1107200108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Sheridan BS, Pham Q-M, Lee Y-T, Cauley LS, Puddington L, Lefrançois L. Oral infection drives a distinct population of intestinal resident memory CD8(+) T cells with enhanced protective function. Immunity. 2014;40:747–757. doi: 10.1016/j.immuni.2014.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Watanabe R, Gehad A, Yang C, Scott LL, Teague JE, Schlapbach C, et al. Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci Transl Med. 2015;7:279ra39–279ra39. doi: 10.1126/scitranslmed.3010302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–763. doi: 10.1146/annurev.immunol.22.012703.104702. [DOI] [PubMed] [Google Scholar]
  • 54.Hu JK, Kagari T, Clingan JM. Expression of chemokine receptor CXCR3 on T cells affects the balance between effector and memory CD8 T-cell generation. 2011 doi: 10.1073/pnas.1101881108/-/DCSupplemental. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sung JH, Zhang H, Moseman EA, Alvarez D, Iannacone M, Henrickson SE, et al. Chemokine guidance of central memory T cells is critical for antiviral recall responses in lymph nodes. Cell. 2012;150:1249–1263. doi: 10.1016/j.cell.2012.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Banerjee A, Gordon SM, Intlekofer AM, Paley MA, Mooney EC, Lindsten T, et al. Cutting edge: The transcription factor eomesodermin enables CD8+ T cells to compete for the memory cell niche. The Journal of Immunology. 2010;185:4988–4992. doi: 10.4049/jimmunol.1002042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Li Q, Eppolito C, Odunsi K, Shrikant PA. IL-12-programmed long-term CD8+ T cell responses require STAT4. J Immunol. 2006;177:7618–7625. doi: 10.4049/jimmunol.177.11.7618. [DOI] [PubMed] [Google Scholar]
  • 58.Intlekofer AM, Takemoto N, Wherry EJ, Longworth SA, Northrup JT, Palanivel VR, 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]
  • 59.Ji Y, Pos Z, Rao M, Klebanoff CA, Yu Z, Sukumar M, et al. Repression of the DNA-binding inhibitor Id3 by Blimp-1 limits the formation of memory CD8+ T cells. Nat Immunol. 2011;12:1230–1237. doi: 10.1038/ni.2153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yang CY, Best JA, Knell J, Yang E, Sheridan AD, Jesionek AK, et al. The transcriptional regulators Id2 and Id3 control the formation of distinct memory CD8+ T cell subsets. Nat Immunol. 2011;12:1221–1229. doi: 10.1038/ni.2158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Yoshida K, Sakamoto A, Yamashita K, Arguni E, Horigome S, Arima M, et al. Bcl6 controls granzyme B expression in effector CD8+ T cells. Eur J Immunol. 2006;36:3146–3156. doi: 10.1002/eji.200636165. [DOI] [PubMed] [Google Scholar]
  • 62.Cui W, Liu Y, Weinstein JS, Craft J, Kaech SM. An interleukin-21-interleukin-10-STAT3 pathway is critical for functional maturation of memory CD8+ T cells. Immunity. 2011;35:792–805. doi: 10.1016/j.immuni.2011.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Siegel AM, Heimall J, Freeman AF, Hsu AP, Brittain E, Brenchley JM, et al. A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory. Immunity. 2011;35:806–818. doi: 10.1016/j.immuni.2011.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hess Michelini R, Doedens AL, Goldrath AW, Hedrick SM. Differentiation of CD8 memory T cells depends on Foxo1. J Exp Med. 2013;210:1189–1200. doi: 10.1084/jem.20130392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kim MV, Ouyang W, Liao W, Zhang MQ, Li MO. The transcription factor Foxo1 controls central-memory CD8+ T cell responses to infection. Immunity. 2013;39:286–297. doi: 10.1016/j.immuni.2013.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Liu N, Phillips T, Zhang M, Wang Y, Opferman JT, Shah R, et al. Serine protease inhibitor 2A is a protective factor for memory T cell development. Nat Immunol. 2004;5:919–926. doi: 10.1038/ni1107. [DOI] [PubMed] [Google Scholar]
  • 67.Zhou X, Yu S, Zhao DM, Harty JT, Badovinac VP, Xue HH. Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1. Immunity. 2010;33:229–240. doi: 10.1016/j.immuni.2010.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Dudani R, Russell M, van Faassen H, Krishnan L, Sad S. Mutation in the Fas Pathway Impairs CD8+ T Cell Memory. J Immunol. 2008;180:2933–2941. doi: 10.4049/jimmunol.180.5.2933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kallies A, Xin A, Belz GT, Nutt SL. Blimp-1 transcription factor is required for the differentiation of effector CD8(+) T cells and memory responses. Immunity. 2009;31:283–295. doi: 10.1016/j.immuni.2009.06.021. [DOI] [PubMed] [Google Scholar]
  • 70.Skon CN, Lee JY, Anderson KG, Masopust D, Hogquist KA, Jameson SC. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat Immunol. 2013;14:1285–1293. doi: 10.1038/ni.2745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature. 2004;427:355–360. doi: 10.1038/nature02284. [DOI] [PubMed] [Google Scholar]
  • 72.Mackay LK, Minnich M, Kragten NAM, Liao Y, Nota B, Seillet C, et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science. 2016;352:459–463. doi: 10.1126/science.aad2035. [DOI] [PubMed] [Google Scholar]
  • 73.Plumlee CR, Sheridan BS, Cicek BB, Lefrançois L. Environmental cues dictate the fate of individual CD8+ T cells responding to infection. Immunity. 2013;39:347–356. doi: 10.1016/j.immuni.2013.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Schluns KS, Lefrançois L. Cytokine control of memory T-cell development and survival. Nat Rev Immunol. 2003;3:269–279. doi: 10.1038/nri1052. [DOI] [PubMed] [Google Scholar]
  • 75.Schluns KS, Williams K, Ma A, Zheng XX. Cutting edge: requirement for IL-15 in the generation of primary and memory antigen-specific CD8 T cells. The Journal of …. 2002 doi: 10.4049/jimmunol.168.10.4827. [DOI] [PubMed] [Google Scholar]
  • 76.Schluns KS, Kieper WC, Jameson SC, Lefrançois L. Interleukin-7 mediates the homeostasis of naïve and memory CD8 T cells in vivo. Nat Immunol. 2000;1:426–432. doi: 10.1038/80868. [DOI] [PubMed] [Google Scholar]
  • 77.Hu Y, Lee YT, Kaech SM, Garvy B, Cauley LS. Smad4 promotes differentiation of effector and circulating memory CD8 T cells but is dispensable for tissue-resident memory CD8 T cells. The Journal of Immunology. 2015;194:2407–2414. doi: 10.4049/jimmunol.1402369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Schenkel JM, Masopust D. Tissue-resident memory T cells. Immunity. 2014;41:886–897. doi: 10.1016/j.immuni.2014.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol. 2014;14:24–35. doi: 10.1038/nri3567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Gaide O, Emerson RO, Jiang X, Gulati N, Nizza S, Desmarais C, et al. Common clonal origin of central and resident memory T cells following skin immunization. Nat Med. 2015;21:647–653. doi: 10.1038/nm.3860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lefrançois L, Marzo AL. The descent of memory T-cell subsets. Nat Rev Immunol. 2006;6:618–623. doi: 10.1038/nri1866. [DOI] [PubMed] [Google Scholar]
  • 82.Stemberger C, Neuenhahn M, Buchholz VR, Busch DH. Origin of CD8+ effector and memory T cell subsets. Cell Mol Immunol. 2007;4:399–405. [PubMed] [Google Scholar]
  • 83.D’Cruz LM, Rubinstein MP, Goldrath AW. Surviving the crash: transitioning from effector to memory CD8+ T cell. Semin Immunol. 2009;21:92–98. doi: 10.1016/j.smim.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Mizukawa Y, Yamazaki Y, Teraki Y, Hayakawa J. Direct evidence for interferon-γ production by effector-memory-type intraepidermal T cells residing at an effector site of immunopathology in fixed drug eruption. Ajp. 2002;161:1337–1347. doi: 10.1016/S0002-9440(10)64410-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Cheuk S, Wikén M, Blomqvist L, Nylen S, Talme T, Stahle M, et al. Epidermal Th22 and Tc17 cells form a localized disease memory in clinically healed psoriasis. The Journal of Immunology. 2014;192:3111–3120. doi: 10.4049/jimmunol.1302313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Chen BJ, Deoliveira D, Cui X, Le NT, Son J, Whitesides JF, et al. Inability of memory T cells to induce graft-versus-host disease is a result of an abortive alloresponse. Blood. 2007;109:3115–3123. doi: 10.1182/blood-2006-04-016410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Zheng H, Matte-Martone C, Jain D, McNiff J, Shlomchik WD. Central memory CD8+ T cells induce graft-versus-host disease and mediate graft-versus-leukemia. The Journal of Immunology. 2009;182:5938–5948. doi: 10.4049/jimmunol.0802212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Yokoyama A, Kohno N, Fujino S, Hamada H, Inoue Y, Fujioka S, et al. Circulating interleukin-6 levels in patients with bronchial asthma. Am J Respir Crit Care Med. 1995;151:1354–1358. doi: 10.1164/ajrccm.151.5.7735584. [DOI] [PubMed] [Google Scholar]
  • 89.Miyahara N, Swanson BJ, Takeda K, Taube C, Miyahara S, Kodama T, et al. Effector CD8+ T cells mediate inflammation and airway hyper-responsiveness. Nat Med. 2004;10:865–869. doi: 10.1038/nm1081. [DOI] [PubMed] [Google Scholar]
  • 90.Ying S, Humbert M, Barkans J, Corrigan CJ, Pfister R, Menz G, et al. Expression of IL-4 and IL-5 mRNA and protein product by CD4+ and CD8+ T cells, eosinophils, and mast cells in bronchial biopsies obtained from atopic and nonatopic (intrinsic) asthmatics. J Immunol. 1997;158:3539–3544. [PubMed] [Google Scholar]
  • 91.Lee N, You S, Shin MS, Lee WW, Kang KS, Kim SH, et al. IL-6 receptor α defines effector memory CD8+ T cells producing Th2 cytokines and expanding in asthma. Am J Respir Crit Care Med. 2014;190:1383–1394. doi: 10.1164/rccm.201403-0601OC. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES