Next Article in Journal
Role of Endoplasmic Reticulum-Associated Proteins in Flavivirus Replication and Assembly Complexes
Next Article in Special Issue
Role of ARP2/3 Complex-Driven Actin Polymerization in RSV Infection
Previous Article in Journal
Janthinobacterium lividum as An Emerging Pathogenic Bacterium Affecting Rainbow Trout (Oncorhynchus mykiss) Fisheries in Korea
Previous Article in Special Issue
Airway Epithelial Derived Cytokines and Chemokines and Their Role in the Immune Response to Respiratory Syncytial Virus Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Contribution of Resident Memory CD8+ T Cells to Protective Immunity against Respiratory Syncytial Virus and Their Impact on Vaccine Design

by
Angello Retamal-Díaz
1,
Camila Covián
1,
Gaspar A. Pacheco
1,
Angelo T. Castiglione-Matamala
1,
Susan M. Bueno
1,
Pablo A. González
1 and
Alexis M. Kalergis
1,2,*
1
Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
2
Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
*
Author to whom correspondence should be addressed.
Pathogens 2019, 8(3), 147; https://doi.org/10.3390/pathogens8030147
Submission received: 25 July 2019 / Revised: 6 September 2019 / Accepted: 7 September 2019 / Published: 11 September 2019
(This article belongs to the Special Issue Virus-Host Interactions of Respiratory Syncytial Virus (RSV))

Abstract

:
Worldwide, human respiratory syncytial virus (RSV) is the most common etiological agent for acute lower respiratory tract infections (ALRI). RSV-ALRI is the major cause of hospital admissions in young children, and it can cause in-hospital deaths in children younger than six months old. Therefore, RSV remains one of the pathogens deemed most important for the generation of a vaccine. On the other hand, the effectiveness of a vaccine depends on the development of immunological memory against the pathogenic agent of interest. This memory is achieved by long-lived memory T cells, based on the establishment of an effective immune response to viral infections when subsequent exposures to the pathogen take place. Memory T cells can be classified into three subsets according to their expression of lymphoid homing receptors: central memory cells (TCM), effector memory cells (TEM) and resident memory T cells (TRM). The latter subset consists of cells that are permanently found in non-lymphoid tissues and are capable of recognizing antigens and mounting an effective immune response at those sites. TRM cells activate both innate and adaptive immune responses, thus establishing a robust and rapid response characterized by the production of large amounts of effector molecules. TRM cells can also recognize antigenically unrelated pathogens and trigger an innate-like alarm with the recruitment of other immune cells. It is noteworthy that this rapid and effective immune response induced by TRM cells make these cells an interesting aim in the design of vaccination strategies in order to establish TRM cell populations to prevent respiratory infectious diseases. Here, we discuss the biogenesis of TRM cells, their contribution to the resolution of respiratory viral infections and the induction of TRM cells, which should be considered for the rational design of new vaccines against RSV.

1. Introduction

RSV is the most common responsible pathogen for acute lower respiratory tract infections (ALRI) [1]. RSV causes respiratory distress mostly in young infants, immuno-compromised patients and senior adults [2]. ALRI is the global leader in respiratory tract infections alongside RSV, which causes 45% of young children hospitalizations and in-hospital deaths in infants younger than six months old [3]. Therefore, RSV remains one of the pathogens deemed most important for vaccine development.
RSV is an enveloped, cytoplasmic virus with a single-stranded, non-segmented, negative-sense RNA genome. This virus belongs to the Pneumoviridae family, genus Orthopneumovirus, which includes three viral species: bovine orthopneumovirus, murine orthopneumovirus and human orthopneumovirus [4]. The RSV genome is approximately 15.2 kb in size and encodes ten genes in the following order: 3′-NS1-NS2-N-P-M-SH-G-F-M2-L-5′. The M2 gene is transcribed in 2 different ORFs, coding for two proteins, M2-1 and M2-2. The genome is encapsidated by multiple copies of the nucleoprotein (N), forming a helical nucleocapsid [5]. Phosphoprotein (P) copies interact with N and recruit the L protein, an RNA-dependent RNA polymerase and the cofactor M2-1 to the viral nucleocapsid complex. Interestingly, the N protein (a restricted cytosolic protein) can also be expressed on the surface of RSV-infected dendritic cells (DCs). Particularly, the N protein interferes with the interaction of the T cell receptor (TCR) with peptides presented on major histocompatibility complex molecules (pMHC) and inhibits the immunological synapse assembly [6,7]. The RSV genome also encodes for two non-structural proteins, NS-1 and NS-2, which display immunomodulatory properties by inhibiting the induction of interferon-alpha/beta (IFNs α/β) in lung epithelial cells and macrophages [8].
The immune system has the capacity to “remember” pathogens, which is the reason why upon pathogen reencounter, a faster and effective response takes place to establish an adaptive and protective immunological response, which is known as immunological memory [9]. During primary respiratory virus infections, antigen-specific CD8+ T cells play fundamental roles in the resolution of infections generated by this type of intracellular pathogens. The adaptive immune response also contributes by conferring protection against subsequent reinfections via immune T and B cell memory development [10]. Thus, frequent reinfections caused by RSV suggest the absence of long-lasting protective immune memory in the host [2,11]. Memory T cells have been classified based on their trafficking patterns and the expression of lymphoid homing receptors CCR7 (chemokine receptor type 7) and CD62L (L-selectin); central memory T cells (TCM; CCR7high/CD62Lhigh) recirculate through the blood and secondary lymphoid organs (SLO), while effector memory T cells (TEM; CCR7low/CD62Llow) do not express homing molecules to lymphoid organs, but instead express migratory receptors with the potential of transit through the blood, lymphoid and peripheral tissues (non-lymphoid tissues) [12]. Nonetheless, parabiosis studies have shown that some tissues, namely the gut and brain, are only under immunosurveillance by effector T cells, but not memory T cells, because the circulation of these cells through peripheral tissue is different for each T cell phenotype [13,14,15,16]. The method as to how these tissues are protected was assessed in subsequent studies where a third subset of memory T cells was identified. This subset corresponds to resident memory T cells (TRM; CCR7+/CD62L+) that are permanently present in non-lymphoid tissues and can mediate innate and adaptive immune responses against reinfections with pathogens [17,18,19]. TRM cells constitutively express high levels of the sphingosine-1-phosphate receptor 1 (S1PR1) antagonist CD69 and hyaluronic acid (HA), which binds CD44, collagen-binding CD49a and CD103 (integrin αE paired with integrin chain β7) [18]. Importantly, the role of CD8+ TRM cells in tissues has been studied more than that of CD4+ TRM lymphocytes. Nevertheless, both cell types participate in the development of the immune response near the locations they occupy and secrete cytokines that promote the recruitment of other immune cells. Several studies have provided evidence for the importance of TRM during protective immune response against RSV [20,21,22,23]. Thus, this response constitutes a critical mechanism that confers immune protection to the host from infections and may also be crucial for the proper function of vaccines, especially for the case of RSV infection.

2. Immunological Synapse and Memory T cells

When initiating an immune response, professional antigen presenting cells (APCs), such as DCs, play crucial roles in the intercommunication between innate and adaptive immunity, as well as the modulation of this response [24]. DCs and macrophages are the major APCs in various mucosal tissues, such as the intestine, lung, genital tract and mouth, where they sense and capture microbes and particles that are potentially harmful to the host [25]. Once captured, antigens derived from microbes are processed and presented to naïve T cells at the regional draining lymph nodes [26]. Activation of T cells requires the assembly of an immunological synapse that provides three types of signals: interaction between the TCR and a peptide-loaded MHC class I or class II molecule (pMHC), co-stimulatory molecule signaling and specific cytokines (Figure 1a). If a cell does not receive a full set of signals, it will neither divide nor become activated, but instead may become anergic. The activation of T cells is triggered by TCR-transduced signals upon engagement with a cognate pMHC complex displayed at the cell surface of an APC. Naïve T cells are activated after their encounter with antigens through TCR signaling, inducing clonal expansion. This process is known as priming. Moreover, T cells can differentiate into distinct classes of effector T cells. After pathogen elimination, the existing T cell populations undergo an inflammation-mediated contraction phase, the survivors of which will serve as the initial memory pool [27,28,29]. Furthermore, although the establishment of effective and long-lasting protective immunity has been historically associated with antigen-specific antibody titers, it may also depend on the generation of specific multifunctional effector memory T cells, which are able to eradicate intracellular infections with the pathogen when accompanied by poor or non-effective humoral responses, as seen for hepatitis C virus, tuberculosis, vesicular stomatitis virus, varicella zoster virus, dengue virus and respiratory syncytial virus [6,7,23,30,31,32,33,34,35]. In contrast, the requirement of both antibodies and multifunctional effector memory T cells has been described during infection with malaria [36,37], simian/human immunodeficiency virus [38], Salmonella [39] and influenza virus [40,41].
TCR signaling leads to the expression of many genes and the entry of T cells into the cell cycle. There are three significant variables that determine the effects of TCR stimulation in T cells: the affinity and half-life of interaction between the TCR and the pMHC, as well as the phenotype of the APC [6,7,24,42,43,44,45,46]. T cell activation induces T cell proliferation to clonally select and expand antigen-specific T cells (Figure 1b). The transition from naïve T cells into effector cells occurs with changes in cell function. CD8+ cytotoxic T cell (CTL) effector functions are stimulated by antigen recognition of a cognate MHC class I–peptide complex by the TCR, inducing apoptosis in the target cell [10]. Since many viruses have developed mechanisms to prevent presentation of their antigens on MHC-I molecules in order to prevent the activation of CD8+ T cells, the development, abundance and antiviral effector functions of TRM cells are usually dampened during viral infections [47]. Along these lines, several reports have shown that RSV prevents the development of protective immunological memory by interfering with the capacity of the DCs to activate and elicit effector activities of virus-specific CD4+ and CD8+ T cells [6,7,48,49]. RSV-infected DCs dampen the activation of naïve CD4+ T cells, which downregulate the TCR-driven activation marker CD69 and show reduced secretion of IL-2 [6]. Similarly, immunization of mice with formalin-inactivated RSV (FI-RSV) leads to a pathology known as vaccine-enhanced respiratory disease (ERD), which elicits a pathological immune reaction after vaccination when the individual is exposed to RSV. This disease is characterized by exacerbated eosinophil infiltration and the development of a TH2-like response in lungs [50,51]. FI-RSV immunization does not activate a CD8+ T cell response that contributes to the eosinophil infiltration in lungs and increased pathology [52]. It is thought that this immune modulatory mechanism could in part be responsible for the lack of generation of long-lasting immunological memory after RSV infection.

3. Tissue-Resident Memory T Cells: Biogenesis, Transcription Factors and Surface Markers

Among the cellular components that constitute the first line of defense is a population of memory T cells called tissue-resident memory CD8+ T cells (TRM). These cells are found in mucosal tissues such as the lungs (both airways and parenchyma), digestive [15] and urogenital tracts [59]. TRM cells have been characterized in elegant parabiosis experiments, which show that tissue surveillance is a function performed by a subset of memory T cells with different characteristics and phenotype behavior than TEM cells, to which this function was previously attributed [59,60]. Besides, once generated, TRM cells can persist at the site of both the infection and vaccination [61].
After respiratory virus infection, CD8+ T cell priming takes place in the regional draining lymph nodes after lung-resident DCs have transported viral antigens to that site. Interestingly, CD103+ DCs have the ability to cross-present foreign antigens to CD8+ T cells, conferring protection against infection [62]. Two migratory DC subsets have been described in the mouse lung tissue, namely CD103+-DCs (CD11c+/CD11blow/CD103+ DC) and CD11bhigh-DCs (CD11c+/CD11bhigh/CD103). These DC-subsets show differential T cell-stimulatory functions, resulting in a range of distinct memory CD8+ T cell subsets [30,63]. CD8+ T cells activated by CD103+ DCs proliferate vigorously and acquire potent effector properties that, among others, bring them back to the lung to eliminate virus-infected cells. Importantly, subsequent differentiation into TRM cells after influenza virus infection was reported to require priming by CD103+ respiratory DCs for the development of resident memory T cell phenotypes [64]. Interestingly, differences in the migration and antigen presentation times between CD103+- and CD11bhigh-DCs have been observed, with CD103+ DCs cross-presenting viral antigens in the mediastinal lymph node (MLN) at early time points of 2–4 days post-infection, as compared to CD11bhighDCs at 5–7 days post-infection, possibly contributing to the development of different T cells subsets [30]. Moreover, the selective loss of CD103+ respiratory DCs reduces antigen-specific CD8+ T cell responses and only CD103+ DCs are capable of cross-presenting antigens in the lymph nodes [65], which suggests that CD103+ respiratory DCs contribute to the development of antiviral CD8+ T cell immunity.
To discriminate between memory precursor cells and effector T cells, killer-cell lectin-like receptor G1 (KLRG1) has been used in mice; short-lived effector CD8+ T cells are generated from KLRG1high precursors, and all memory CD8+ T cells are from KLRG1low precursors [66,67,68] (Figure 1). The TCR sequences used by these T cells showed an overlap between TCM and TRM, suggesting that they could derive from a single naïve T cell [69]. However, it is unclear whether an individual memory precursor has the potential to differentiate into both TRM and TCM cells [70].
The generation and establishment of TRM cells in each tissue can significantly differ [71], indicating that tissue-specific environments play pivotal roles in these processes [72]. Moreover, the transcriptional regulation of these T cells poses specific requirements that vary between mice and humans [73,74]. However, both effector and memory T cells exhibit a similar pattern of chromatin accessibility, as compared to naïve CD8+ T cells [75]. Particularly, the transcription factors (TFs) Blimp-1, ID2, T-bet and STAT4 promote the development of TEM [67,68,69,70,71,72,73,74,75,76,77,78,79], while the expression of BCL-6, Eomes, ID3, TCF-1 and STAT3 are associated with the generation of TCM (Figure 1c, blue boxes) [80,81,82,83,84]. In mice, TRM cells show an elevated expression level of the homolog of B lymphocyte-induced maturation protein-1 in T cells (Hobit), B lymphocyte-induced maturation protein-1 (Blimp-1) and Runx3 [85]. Together these TFs contribute to differentiation, residence and survival of TRM cells in the tissue [86], and negatively regulate the expression of genes associated with tissue egress from non-lymphoid organs [87]. Nevertheless, at the protein level, Blimp-1 expression increased only during the effector stage, while Hobit mRNA and protein expression was constitutive during quiescence and downregulated after activation [88,89]. Both transcriptional factors Hobit and Blimp-1 have overlapping target genes that include cytotoxic molecules, such as granzyme B. During effector differentiation, CD8+ T cells acquire these cytotoxic molecules. Blimp-1 initiates the cytotoxic effector function because it is required for expression of granzyme B in murine effector T cells and TRM cells, while Hobit maintains granzyme B expression in murine TRM cells during their memory phase [88]. After influenza virus infection, CD8+ TRM require continuous recruitment from the circulating memory pool to establish tissue resident cells, and this viral infection shows that Blimp-1, rather than Hobit, mediates the formation of TRM cells within the lungs [86], potentially through control of the lineage choice between TCM and TRM cells during the differentiation of influenza-specific CD8+ T cells [87]. These data suggest that TRM cell differentiation towards a unique program of transcriptional regulation occurs in a pathogen-specific manner. Besides, the differentiation of TRM cells is highly dependent on local cues from the microenvironment and it has been shown that the expression of CD103 on T cells is dependent on TGF-β, which is required for TRM cell establishment in some tissues. Indeed, the best-characterized signal required for TRM cell development is mediated by TGF-β [90]; thus, CD8+ T cells that lack TGFβR fail to upregulate the expression of CD103 after antigen exposure and subsequently fail to differentiate into TRM cells [89,90,91,92]. The downregulation of transcription factors KLF2, Eomes and the low expression of T-bet and TCF 1 is needed for proper TRM cell residency [93]. Both T-bet and Eomes transcription factors control and down-regulate the signaling of TGF-β and upregulate CD103 [91]. Also, low levels of T-bet are required to maintain cell surface expression of the IL-15 receptor β-chain (CD122), which has been shown to be critical for the rapid proliferation of memory CD8+ T cells. In response to viral infection, type-I IFNs trigger the expression of IL-15 that prepares memory T cells for rapid division, independently of antigen re-exposure, by transiently inducing cell-cycle progression by the mTOR complex-1 (mTORC1) signaling pathway [92,94,95].
The type II C-lectin CD69 was initially related as an early T cell activation marker because its expression is rapidly induced on the surface of activated T cells [96]. Besides, CD69 is an antagonist of the sphingosine-1-phosphate receptor (S1PR1) that limits egress by blocking responsiveness to sphingosine-1-phosphate (SP1) gradients. The S1PR1 expression is commanded by the transcription factor Krüppel-like factor 2 (KLF2), and its downregulation is needed for TRM cell formation in mice [97,98]. The constitutive expression of CD69 is essential for TRM cell formation and is a typical marker of peripheral TRM cells [93,98].
Another relevant surface molecule is CD44, a C-lectin that can act as a receptor for hyaluronic acid (HA). This molecule is maintained at high levels in TRM cells populations, but CD44 alone does not single out TRM cells from other CD8+ T cell populations [99]. The most relevant function of CD44 is the maintaining of cell structure through adherence to the extracellular matrix, mainly composed by HA [100,101].
The very late antigen 1 (VLA-1), also known as CD49a, is the α-subunit of the α1β1 integrin receptor that pairs with CD29 (integrin β1) [102]. CD49a is an adhesion molecule that binds to collagen IV (Coll IV) present in the basement membrane of epithelium [103]. Functionally, the blocking or genetic deletion of CD49a results in smaller TRM cell population at the mucosal site, which becomes susceptible to secondary infections. These data suggest that CD49a plays a striking role in keeping these cells in the lungs [104,105].
CD103 is α-subunit of the integrin αEβ7. It is involved in tissue retention by its interaction with the ligand E-cadherin and is upregulated on TRM cells upon exposure to TGF-β [106]. In a mouse model of influenza infection, CD103 contributes to the retention of pathogen-specific cytotoxic T lymphocytes in the lungs and CD103 deficiency results in a reduction of CD8+ TRM cells in the lungs [107,108]. Also, the expression of CD103 is essential for TRM cell formation and constitutes a typical surface marker of TRM cells.
TRM cells in each tissue are maintained by different microenvironments, implicating that the process of the establishment and renewal of these cells varies between tissues (reviewed in [109] and [110]). This also implies that the signals and mechanisms of action are also dependent on the tissue milieu and, eventually, in the formation of particular structures for their maintenance [111,112,113]. For instance, in the lung tissue, an injury promotes the temporary generation of spatial niches called Repair-Associated Memory Depots (RAMDs) that contribute to tissue regeneration and constitute a niche that supports TRM cell maintenance in the lungs [114]. Furthermore, the time for which TRM cells persist in each tissue over time may vary. Several studies have shown that the dynamics of the establishment/renewal of TRM cells in the lungs is different as compared to the female reproductive tract (FRT) or the skin [110]. Particularly, the establishment of TRM cells in the FRT and skin are trigged by antigen-independent inflammation that recruits memory CD8+ T cells [115]. However, in the lung tissue, TRM establishment depends on cognate-antigen and inflammation that promotes the formation of RAMDs [116]. On the other hand, the presence of memory T cells in the lung airways is maintained by continual recruitment from circulation [117], and presumably TRM cells in the airways could be supported by RAMDs. Nonetheless, the transient formation of RAMDs may account for the shorter longevity of CD8+ TRM cells in the lung as compared to the gut and skin [22]. In addition, another essential aspect of T cells proliferation and differentiation is the exposure to damage/danger-associated molecular patterns (DAMPs), such as the nucleotides ATP and NAD+ [118]. CD8+ TRM cells express the P2RX7 receptor, but not TCM cells. Thereby, during infection or tissue damage, the release of DAMPs contributes to depleting TRM locally [119]. For example, P2RX7 activation in vivo by exogenous NAD+ led to a specific depletion of TRM while retaining circulating T cells, suggesting that the P2RX7 pathway contributes to the regulation of TRM maintenance [120].
TRM elicits a faster and more potent response after antigen recognition than circulating memory T cells [121]. TRM triggers an innate-like alarm characterized by the production of massive amounts of effector molecules, such as: IFN-γ, TNF-α and granzyme B, as well as pro-inflammatory cytokines, chemokines and antimicrobial molecules [122]. The contribution of TRM cells in mediating protective responses has attracted the attention for developing vaccination strategies that exploit the ability of these cells to establish long-lasting residency, which would prevent infections, particularly respiratory infectious diseases [22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124].

4. TRM cells in Response to RSV Infection

Both in mice and humans, natural infection with RSV induces the generation of virus-specific CD8+ TRM cells that provide protection against RSV infection and contribute to reducing the severity of the disease. In the murine model, the adoptive transfer of airway CD8+ TRM from RSV-infected mice into naïve recipients diminished the disease severity upon viral challenge. Additionally, RSV infection in mice induced TRM recruitment in the airways and lung parenchyma [23]. Moreover, a study performed with humans showed that adults with higher frequencies of RSV-specific CD8+ T cells in the airways (many of which show a TRM phenotype), developed less severe lower respiratory tract symptoms and reduced viral loads upon RSV challenge. It is noteworthy that increased protection against RSV did not correlate with an increase in circulating CD8+ T cells in the blood, but instead correlated with increased abundance of a preexisting TRM cell population in the airways prior to infection [125]. Interestingly, during acute infection, RSV-specific CD8+ TCM cells were temporarily accompanied by a CD103+/CD69+ population that could represent a cell population that migrates from the blood to the airways, which could represent TRM cells [125].
In a non-human primate model, the infection of juvenile naïve African green monkeys with RSV showed that the peak of abundance of virus-specific CD8+ T cells emerging in peripheral blood and bronchoalveolar lavage (BAL) coincided with the declining of viral titers, suggesting that virus-specific cellular responses contribute to the clearance of RSV infection [21]. Moreover, this study showed an abundant CD103+ CD69+ population in BALs among the T cell population of these monkeys.
Influenza virus is another major respiratory pathogen, and CD8+ TRM cells in nasal epithelia reduce viral transmission to the lungs, preventing pulmonary infection [126]. During secondary influenza virus infection, TRM cells were shown to limit early viral replication, attenuating the duration of inflammation in an IFN-γ-dependent fashion. Additionally, specific TRM showed an indispensable role in cross-protection against different influenza virus strains. These data suggest that TRM plays a critical role in protection against respiratory infections in the upper and lower respiratory tracts [40,127].
In a mouse model of influenza and Sendai virus infection, the intratracheal transfer of antigen-specific CD8+ TRM cells from immunized into naïve mice was sufficient to confer protection against respiratory virus infection. Interestingly, upon antigen exposure, the TRM secreted IFN-γ quickly enough to limit viral replication in the lungs [128]. Additionally, the transfer of airway-resident IFN-γ-deficient CD8+ TRM cells had a reduced protective potential. These findings suggest that airway-resident CD8+ TRM cells contribute to the generation of protective immunity against respiratory pathogens and to the rapid secretion of IFN-γ, which is essential for achieving a successful viral clearance [128,129].

5. Tissue-Resident T Cells as a Target for Vaccine Development

While B cells mount responses against pathogens’ surface epitopes, CD8+ T cells attack mainly the internal epitopes of pathogens, which are more conserved and therefore prevent the pathogen’s escape of the immune system via antigenic drift. Therefore, the remarkable ability of CD8+ TRM cells to serve as a first line of defense against reinfecting pathogens has become attractive so as to develop vaccination strategies that take advantage of the ability of TRM cells to prevent respiratory infectious diseases [6,26,30]. Moreover, intranasal RSV infection in mice leads to the generation of antigen-specific CD8+ TRM cells (CD103+/CD69+) in the lungs and airways. It has been shown that intranasal transfer of CD8+ TRM cells from the airways of previously infected animals to naïve animals reduces weight loss upon infection. Additionally, the transfer of airway pan-CD8+ cells protected the body upon infection, ameliorating illness, reducing viral load and increasing IFN-γ secretion in the airways of the recipient mice [23].
Experimental RSV vaccines are capable of inducing protective memory T cells, such as the recombinant Mycobacterium bovis bacillus Calmette-Guèrin (BCG) expressing the RSV-N protein (rBCG-N-hRSV vaccine expressing RSV-nucleoprotein N), which is capable of eliciting protective immune responses [130,131]. This promising formulation developed under good manufacturing practices (GMP) has been shown to be safe in preclinical models and induces a strong antiviral TH1/TH17 T-cell memory response [132]. Additionally, some formulations are capable of generating TRM cells. For example, intranasal administration of virus-like particles containing RSV M and M2 proteins has also been shown to induce TRM cell development in the lung [133]. Moreover, DCs pulsed with Listeria monocytogenes as an immunization strategy were able to avoid the immunopathology caused by circulating T cells, and protect against a subsequent RSV challenge by promoting the generation of TRM cells when administered locally [134].
The route of antigen delivery is relevant for gathering TRM to the target tissue. For example, immunization with recombinant cytomegalovirus expressing the RSV M protein generated TRM and TEM CD8+ T cells populations when administered intranasally. However, when the vaccine was administered intraperitoneally, these cell populations were undetectable [21]. These findings support the notion that the route of antigen delivery in a vaccination context is a crucial determinant of immune priming at the infection site. Controversially, the immunization with a vaccine candidate G1F/M2 plus CpG as an adjuvant by the intraperitoneal route elicits high levels of TCM cells and TH1 type of TEM cells in the spleen, which may contribute to inhibition of lung inflammation. Nevertheless, the intranasal immunization recruits high levels of TRM cells that lack or possess a weak TH1 type immune memory that transfers from the spleen into the lung, which might promote lung inflammation following RSV infection [135].
An interesting approach for generating TRM cells is designated “prime and pull,” which combines vaccination (prime) with local administration of chemokines, adjuvants or antigens to recruit TRM precursors to target tissues (pull) [136]. For example, zymosan can promote differentiation of effector CD8+ T cells to their differentiation in lung TRM cells when administered as an adjuvant into the airways without any antigen [137].
As previously described, the formalin-inactivated RSV vaccine developed in the sixties (FI-RSV) caused vaccine-enhanced respiratory disease (ERD). Recently, it has been described that modulation of the immune response via TLR agonists can help to regulate the exacerbated immune response elicited by FI-RSV. TLR3 agonist CpG together with Notch receptor inhibitor L685458 markedly reduced FI-RSV-enhanced airway pathology symptoms, such as weight loss, lung inflammation and airway hyperresponsiveness. Moreover, the combined treatment promoted protective CD8+ TRM cells in the lungs. Interestingly, none of the treatments by themselves alone reduced ERD. The proposed mechanism seems to involve the suppression of TH17 memory responses and the promotion of tissue-resident memory cells. Additionally, these results imply that the modulation of lung immune memory with adjuvants might be a good strategy to generate TRM cells with antiviral properties [138].
The migration of antigen-specific T cells into the lungs, among other non-lymphoid tissues, is key for containing peripheral infections, and its modulation is an important strategy for the development of prophylactic tools [139]. The homing of T cells can be taught and would be a strategy for TRM cell establishment after immunization. Tissue-associated DCs are relevant for imprinting the tropism of T cells during the priming phase. For example, T cell activation by intestinal DCs generates a gut-tropism because these cells are able to suppress in T cells the expression of skin-homing receptors once the T cell is activated [140]. Moreover, the synthesis of specific metabolites by DCs in each tissue contributes to particular T cell migration programs defined by DC priming [141]. Therefore, repeated intranasal immunizations, which promote the generation of TRM cells through the development of a precursor cell pool, may be a promising strategy for the establishment of long-lasting lung TRM-mediated immunity against respiratory pathogens.

6. Concluding Remarks

In summary, CD8+ TRM cells are noncirculating adaptive immune cells that are localized as sentinels at the physical barriers that are susceptible to the entry pathogens. Located at such places, they are key for the avoidance of reinfections by previously encountered microbial agents. Their placing at these hotspots of pathogen encounters provides rapid protection against viral infections. However, the signals that stabilize the TRM population in the lung have yet to be identified.
Considering that RSV is a respiratory viral pathogen that infects the respiratory epithelia in order to replicate, we consider that vaccination strategies against RSV should promote cell-based immunity that relies on the contribution of CD8+ TRM response in the lungs. This claim is supported by the fact that CD8+ T cells in mice infected with RSV are essential for the clearance of this virus upon secondary infection. However, pre-existing RSV-specific memory CD8+ T cells migrating into the lungs lead to exacerbated disease severity and promote lethal immunopathology, mediated by the abundant secretion of IFN-γ in the lungs [142]. Therefore, CD8+ T cells are a critical subset that can positively contribute to viral resolution, or negatively contribute to vaccine-enhanced respiratory disease [134,143]. In addition, the establishment of a protective immune response against RSV requires neutralizing antibodies that reduce the infectivity of virions [144]. Thus, a cell-based immunity that recognizes virus-infected cells and neutralizing antibodies that prevent the propagation of virus is desirable in an effective vaccine.
An attractive approach for generating TRM cells was recently described and named “prime and pull,” which combines vaccination (prime) with the local administration of chemokines, adjuvants or antigens to recruit TRM precursors that target particular tissues (pull) [136]. This strategy has the ability to recruit circulating effector T cells and memory T cells to sites where pathogens may enter, resulting in the generation of local TRM that may significantly contribute to protection against viral infections caused by viruses, such as herpes simplex virus type 2 [18], malaria at the liver-stage of infection [145,146], human papillomavirus [147,148], influenza and even for developing cancer vaccines [149]. The main characteristics of these cells are their long permanence in the tissues, as well as their rapid recall responses that release molecules that mediate both antiviral and anti-tumor immunity. These attributes are desirable in the development of vaccines against viral pathogens and for cancer vaccines [150]. Thus, a detailed characterization of tissue-specific factors that regulate TRM biology and their contribution both in pathology and protection could be crucial to new therapeutic designs.

Funding

This research was funded by FONDECYT grant number 1190830, 1170694 and 1190864 from CONICYT Chile, as well as the Millennium Institute on Immunology and Immunotherapy grant number P09/016-F. AMK is a Helen C. Levitt visiting professor at the Department of Microbiology and Immunology of the University of Iowa.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Tregoning, J.S.; Schwarze, J. Respiratory viral infections in infants: Causes, clinical symptoms, virology, and immunology. Clin. Microbiol. Rev. 2010, 23, 74–98. [Google Scholar] [CrossRef] [PubMed]
  2. Borchers, A.T.; Chang, C.; Gershwin, M.E.; Gershwin, L.J. Respiratory syncytial virus—A comprehensive review. Clin. Rev. Allergy Immunol. 2013, 45, 331–379. [Google Scholar] [CrossRef] [PubMed]
  3. Shi, T.; McAllister, D.A.; O’Brien, K.L.; Simoes, E.A.; Madhi, S.A.; Gessner, B.D.; Alassani, I. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in young children in 2015: A systematic review and modelling study. Lancet 2017, 390, 946–958. [Google Scholar] [CrossRef]
  4. King, A.M.Q.; Lefkowitz, E.J.; Mushegian, A.R.; Adams, M.J.; Dutilh, B.E.; Gorbalenya, A.E.; Kropinski, A.M. Changes to taxonomy and the International Code of Virus Classification and Nomenclature ratified by the International Committee on Taxonomy of Viruses (2018). Arch. Virol. 2018, 163, 2601–2631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Malboeuf, C.M.; Yang, X.; Charlebois, P.; Qu, J.; Berlin, A.M.; Casali, M.; Allen, T.M. Complete viral RNA genome sequencing of ultra-low copy samples by sequence-independent amplification. Nucleic Acids Res. 2013, 41, e13. [Google Scholar] [CrossRef]
  6. Gonzalez, P.A.; Prado, C.E.; Leiva, E.D.; Carreño, L.J.; Bueno, S.M.; Riedel, C.A.; Kalergis, A.M. Respiratory syncytial virus impairs T cell activation by preventing synapse assembly with dendritic cells. Proc. Natl. Acad. Sci. USA 2008, 105, 14999–15004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Cespedes, P.F.; Bueno, S.M.; Ramírez, B.A.; Gomez, R.S.; Riquelme, S.A.; Palavecino, C.E.; Cammer, M. Surface expression of the hRSV nucleoprotein impairs immunological synapse formation with T cells. Proc. Natl. Acad. Sci. USA 2014, 111, E3214–E3223. [Google Scholar] [CrossRef] [Green Version]
  8. Spann, K.M.; Tran, K.C.; Collins, P.L. Effects of nonstructural proteins NS1 and NS2 of human respiratory syncytial virus on interferon regulatory factor 3, NF-kappaB, and proinflammatory cytokines. J. Virol. 2005, 79, 5353–5362. [Google Scholar] [CrossRef]
  9. Zinkernagel, R.M. On immunological memory. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2000, 355, 369–371. [Google Scholar] [CrossRef]
  10. Zhang, N.; Bevan, M.J. CD8(+) T cells: Foot soldiers of the immune system. Immunity 2011, 35, 161–168. [Google Scholar] [CrossRef]
  11. Lambert, L.; Sagfors, A.M.; Openshaw, P.J.; Culley, F.J. Immunity to RSV in Early-Life. Front. Immunol. 2014, 5, 466. [Google Scholar] [CrossRef] [PubMed]
  12. 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. [Google Scholar] [CrossRef]
  13. Klonowski, K.D.; Williams, K.J.; Marzo, A.L.; Blair, D.A.; Lingenheld, E.G.; Lefrançois, L. Dynamics of blood-borne CD8 memory T cell migration in vivo. Immunity 2004, 20, 551–562. [Google Scholar] [CrossRef]
  14. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Masopust, D.; Choo, D.; Vezys, V.; Wherry, E.J.; Duraiswamy, J.; Akondy, R.; Fraser, K.A. Dynamic T cell migration program provides resident memory within intestinal epithelium. J. Exp. Med. 2010, 207, 553–564. [Google Scholar] [CrossRef] [Green Version]
  16. Wakim, L.M.; Woodward-Davis, A.; Bevan, M.J. Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence. Proc. Natl. Acad. Sci. USA 2010, 107, 17872–17879. [Google Scholar] [CrossRef] [Green Version]
  17. Masopust, D.; Vezys, V.; Marzo, A.L.; Lefrançois, L. Preferential localization of effector memory cells in nonlymphoid tissue. Science 2001, 291, 2413–2417. [Google Scholar] [CrossRef]
  18. Gebhardt, T.; Wakim, L.M.; Eidsmo, L.; Reading, P.C.; Heath, W.R.; Carbone, F.R. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 2009, 10, 524–530. [Google Scholar] [CrossRef]
  19. Jiang, X.; Clark, R.A.; Liu, L.; Wagers, A.J.; Fuhlbrigge, R.C.; Kupper, T.S. Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature 2012, 483, 227–231. [Google Scholar] [CrossRef]
  20. Morabito, K.M.; Ruckwardt, T.R.; Redwood, A.J.; Moin, S.M.; Price, D.A.; Graham, B.S. Intranasal administration of RSV antigen-expressing MCMV elicits robust tissue-resident effector and effector memory CD8+ T cells in the lung. Mucosal Immunol. 2017, 10, 545–554. [Google Scholar] [CrossRef]
  21. Li, H.; Callahan, C.; Citron, M.; Wen, Z.; Touch, S.; Monslow, M.A.; Espeseth, A. Respiratory syncytial virus elicits enriched CD8+ T lymphocyte responses in lung compared with blood in African green monkeys. PLoS ONE 2017, 12, e0187642. [Google Scholar] [CrossRef] [PubMed]
  22. Wu, T.; Hu, Y.; Lee, Y.T.; Bouchard, K.R.; Benechet, A.; Khanna, K.; Cauley, L.S. Lung-resident memory CD8 T cells (TRM) are indispensable for optimal cross-protection against pulmonary virus infection. J. Leukoc. Biol. 2014, 95, 215–224. [Google Scholar] [CrossRef] [PubMed]
  23. Kinnear, E.; Lambert, L.; McDonald, J.U.; Cheeseman, H.M.; Caproni, L.J.; Tregoning, J.S. Airway T cells protect against RSV infection in the absence of antibody. Mucosal Immunol. 2018, 11, 290. [Google Scholar] [CrossRef] [PubMed]
  24. Gonzalez, P.A.; Carreno, L.J.; Figueroa, C.A.; Kalergis, A. Modulation of immunological synapse by membrane-bound and soluble ligands. Cytokine Growth Factor Rev. 2007, 18, 19–31. [Google Scholar] [CrossRef] [PubMed]
  25. Soloff, A.C.; Barratt-Boyes, S.M. Enemy at the gates: Dendritic cells and immunity to mucosal pathogens. Cell Res. 2010, 20, 872–885. [Google Scholar] [CrossRef] [PubMed]
  26. Iijima, N.; Thompson, J.M.; Iwasaki, A. Dendritic cells and macrophages in the genitourinary tract. Mucosal Immunol. 2008, 1, 451–459. [Google Scholar] [CrossRef] [Green Version]
  27. Badovinac, V.P.; Porter, B.B.; Harty, J.T. CD8+ T cell contraction is controlled by early inflammation. Nat. Immunol. 2004, 5, 809–817. [Google Scholar] [CrossRef]
  28. Murali-Krishna, K.; Altman, J.D.; Suresh, M.; Sourdive, D.J.; Zajac, A.J.; Miller, J.D.; Ahmed, R. Counting antigen-specific CD8 T cells: A reevaluation of bystander activation during viral infection. Immunity 1998, 8, 177–187. [Google Scholar] [CrossRef]
  29. Razvi, E.S.; Welsh, R.M. Programmed cell death of T lymphocytes during acute viral infection: A mechanism for virus-induced immune deficiency. J. Virol. 1993, 67, 5754–5765. [Google Scholar]
  30. Kim, T.S.; Braciale, T.J. Respiratory dendritic cell subsets differ in their capacity to support the induction of virus-specific cytotoxic CD8+ T cell responses. PLoS ONE 2009, 4, e4204. [Google Scholar] [CrossRef]
  31. Moseman, E.A.; Iannacone, M.; Bosurgi, L.; Tonti, E.; Chevrier, N.; Tumanov, A.; von Andrian, U.H. B cell maintenance of subcapsular sinus macrophages protects against a fatal viral infection independent of adaptive immunity. Immunity 2012, 36, 415–426. [Google Scholar] [CrossRef]
  32. Levin, M.J.; Oxman, M.N.; Zhang, J.H.; Johnson, G.R.; Stanley, H.; Hayward, A.R.; Chan, I.F. Varicella-zoster virus-specific immune responses in elderly recipients of a herpes zoster vaccine. J. Infect. Dis. 2008, 197, 825–835. [Google Scholar] [CrossRef] [PubMed]
  33. Amorim, J.H.; dos Santos Alves, R.P.; Bizerra, R.; Pereira, S.A.; Pereira, L.R.; Fabris, D.L.N.; de Souza Ferreira, L.C. Antibodies are not required to a protective immune response against dengue virus elicited in a mouse encephalitis model. Virology 2016, 487, 41–49. [Google Scholar] [CrossRef] [PubMed]
  34. Orme, I.M.; Collins, F.M. Adoptive protection of the Mycobacterium tuberculosis-infected lung. Dissociation between cells that passively transfer protective immunity and those that transfer delayed-type hypersensitivity to tuberculin. Cell. Immunol. 1984, 84, 113–120. [Google Scholar] [CrossRef]
  35. Lechner, F.; Wong, D.K.; Dunbar, P.R.; Chapman, R.; Chung, R.T.; Dohrenwend, P.; Walker, B.D. Analysis of successful immune responses in persons infected with hepatitis C virus. J. Exp. Med. 2000, 191, 1499–1512. [Google Scholar] [CrossRef] [PubMed]
  36. Kaba, S.A.; McCoy, M.E.; Doll, T.A.; Brando, C.; Guo, Q.; Dasgupta, D.; Burkhard, P. Protective antibody and CD8+ T-cell responses to the Plasmodium falciparum circumsporozoite protein induced by a nanoparticle vaccine. PLoS ONE 2012, 7, e48304. [Google Scholar] [CrossRef] [PubMed]
  37. Walker, K.M.; Okitsu, S.; Porter, D.W.; Duncan, C.; Amacker, M.; Pluschke, G.; Todryk, S.M. Antibody and T-cell responses associated with experimental human malaria infection or vaccination show limited relationships. Immunology 2015, 145, 71–81. [Google Scholar] [CrossRef]
  38. Petitdemange, C.; Kasturi, S.P.; Kozlowski, P.A.; Nabi, R.; Quarnstrom, C.F.; Reddy, P.B.J.; Kovalenkov, Y.O. Vaccine induction of antibodies and tissue-resident CD8+ T cells enhances protection against mucosal SHIV-infection in young macaques. JCI Insight 2019, 4, 126047. [Google Scholar] [CrossRef]
  39. Benoun, J.M.; Peres, N.G.; Wang, N.; Pham, O.H.; Rudisill, V.L.; Fogassy, Z.N.; Puddington, L. Optimal protection against Salmonella infection requires noncirculating memory. Proc. Natl. Acad. Sci. USA 2018, 115, 10416–10421. [Google Scholar] [CrossRef]
  40. Wakim, L.M.; Smith, J.; Caminschi, I.; Lahoud, M.H.; Villadangos, J.A. Antibody-targeted vaccination to lung dendritic cells generates tissue-resident memory CD8 T cells that are highly protective against influenza virus infection. Mucosal Immunol. 2015, 8, 1060–1071. [Google Scholar] [CrossRef] [Green Version]
  41. Lee, Y.N.; Lee, Y.T.; Kim, M.C.; Gewirtz, A.T.; Kang, S.M. A Novel Vaccination Strategy Mediating the Induction of Lung-Resident Memory CD8 T Cells Confers Heterosubtypic Immunity against Future Pandemic Influenza Virus. J. Immunol. 2016, 196, 2637–2645. [Google Scholar] [CrossRef] [PubMed]
  42. Gonzalez, P.A.; Carreno, L.J.; Coombs, D.; Mora, J.E.; Palmieri, E.; Goldstein, B.; Kalergis, A.M. T cell receptor binding kinetics required for T cell activation depend on the density of cognate ligand on the antigen-presenting cell. Proc. Natl. Acad. Sci. USA 2005, 102, 4824–4829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Carreno, L.J.; Riquelme, E.M.; González, P.A.; Espagnolle, N.; Riedel, C.A.; Valitutti, S.; Kalergis, A.M. T-cell antagonism by short half-life pMHC ligands can be mediated by an efficient trapping of T-cell polarization toward the APC. Proc. Natl. Acad. Sci. USA 2010, 107, 210–215. [Google Scholar] [CrossRef] [PubMed]
  44. Riquelme, E.; Carreno, L.J.; González, P.A.; Kalergis, A.M. The duration of TCR/pMHC interactions regulates CTL effector function and tumor-killing capacity. Eur. J. Immunol. 2009, 39, 2259–2269. [Google Scholar] [CrossRef] [PubMed]
  45. Carreno, L.J.; Bueno, S.M.; Bull, P.; Nathenson, S.G.; Kalergis, A.M. The half-life of the T-cell receptor/peptide-major histocompatibility complex interaction can modulate T-cell activation in response to bacterial challenge. Immunology 2007, 121, 227–237. [Google Scholar] [CrossRef]
  46. Carreno, L.J.; Gonzalez, P.A.; Kalergis, A.M. Modulation of T cell function by TCR/pMHC binding kinetics. Immunobiology 2006, 211, 47–64. [Google Scholar] [CrossRef] [PubMed]
  47. Lauron, E.J.; Yang, L.; Harvey, I.B.; Sojka, D.K.; Williams, G.D.; Paley, M.A.; Yokoyama, W.M. Viral MHCI inhibition evades tissue-resident memory T cell formation and responses. J. Exp. Med. 2019, 216, 117–132. [Google Scholar] [CrossRef]
  48. Connors, M.; Kulkarni, A.B.; Firestone, C.Y.; Holmes, K.L.; Morse, H.; Sotnikov, A.V.; Murphy, B.R. Pulmonary histopathology induced by respiratory syncytial virus (RSV) challenge of formalin-inactivated RSV-immunized BALB/c mice is abrogated by depletion of CD4+ T cells. J. Virol. 1992, 66, 7444–7451. [Google Scholar] [Green Version]
  49. Sallusto, F. Heterogeneity of Human CD4(+) T Cells Against Microbes. Annu. Rev. Immunol. 2016, 34, 317–334. [Google Scholar] [CrossRef] [PubMed]
  50. Kruijsen, D.; Schijf, M.A.; Lukens, M.V.; van Uden, N.O.; Kimpen, J.L.; Coenjaerts, F.E.; van Bleek, G.M. Local innate and adaptive immune responses regulate inflammatory cell influx into the lungs after vaccination with formalin inactivated RSV. Vaccine 2011, 29, 2730–2741. [Google Scholar] [CrossRef] [PubMed]
  51. Christiaansen, A.F.; Knudson, C.J.; Weiss, K.A.; Varga, S.M. The CD4 T cell response to respiratory syncytial virus infection. Immunol. Res. 2014, 59, 109–117. [Google Scholar] [CrossRef] [PubMed]
  52. Olson, M.R.; Varga, S.M. CD8 T cells inhibit respiratory syncytial virus (RSV) vaccine-enhanced disease. J. Immunol. 2007, 179, 5415–5424. [Google Scholar] [CrossRef] [PubMed]
  53. Schluns, K.S.; Lefrançois, L. Cytokine control of memory T-cell development and survival. Nat. Rev. Immunol. 2003, 3, 269–279. [Google Scholar] [CrossRef] [PubMed]
  54. Kaneko, S.; Mastaglio, S.; Bondanza, A.; Ponzoni, M.; Sanvito, F.; Aldrighetti, L.; Nagasawa, T. IL-7 and IL-15 allow the generation of suicide gene-modified alloreactive self-renewing central memory human T lymphocytes. Blood 2009, 113, 1006–1015. [Google Scholar] [CrossRef] [PubMed]
  55. Schluns, K.S.; Kieper, W.C.; Jameson, S.C.; Lefrançois, L. Interleukin-7 mediates the homeostasis of naïve and memory CD8 T cells in vivo. Nat. Immunol. 2000, 1, 426–432. [Google Scholar] [CrossRef] [PubMed]
  56. Tan, J.T.; Ernst, B.; Kieper, W.C.; LeRoy, E.; Sprent, J.; Surh, C.D. Interleukin (IL)-15 and IL-7 jointly regulate homeostatic proliferation of memory phenotype CD8+ cells but are not required for memory phenotype CD4+ cells. J. Exp. Med. 2002, 195, 1523–1532. [Google Scholar] [CrossRef] [PubMed]
  57. Verbist, K.C.; Field, M.B.; Klonowski, K.D. Cutting edge: IL-15-independent maintenance of mucosally generated memory CD8 T cells. J. Immunol. 2011, 186, 6667–6671. [Google Scholar] [CrossRef]
  58. Ku, C.C.; Murakami, M.; Sakamoto, A.; Kappler, J.; Marrack, P. Control of homeostasis of CD8+ memory T cells by opposing cytokines. Science 2000, 288, 675–678. [Google Scholar] [CrossRef]
  59. Ariotti, S.; Hogenbirk, M.A.; Dijkgraaf, F.E.; Visser, L.L.; Hoekstra, M.E.; Song, J.Y.; Schumacher, T.N. T cell memory. Skin-resident memory CD8(+) T cells trigger a state of tissue-wide pathogen alert. Science 2014, 346, 101–105. [Google Scholar] [CrossRef]
  60. Steinert, E.M.; Schenkel, J.M.; Fraser, K.A.; Beura, L.K.; Manlove, L.S.; Igyártó, B.Z.; Masopust, D. Quantifying Memory CD8 T Cells Reveals Regionalization of Immunosurveillance. Cell 2015, 161, 737–749. [Google Scholar] [CrossRef] [Green Version]
  61. Schenkel, J.M.; Fraser, K.A.; Beura, L.K.; Pauken, K.E.; Vezys, V.; Masopust, D. T cell memory. Resident memory CD8 T cells trigger protective innate and adaptive immune responses. Science 2014, 346, 98–101. [Google Scholar] [CrossRef] [PubMed]
  62. Cao, Q.; Lu, J.; Li, Q.; Wang, C.; Wang, X.M.; Lee, V.W.; Alexander, S.I. CD103+ Dendritic Cells Elicit CD8+ T Cell Responses to Accelerate Kidney Injury in Adriamycin Nephropathy. J. Am. Soc. Nephrol. 2016, 27, 1344–1360. [Google Scholar] [CrossRef] [PubMed]
  63. Beaty, S.R.; Rose, C.E., Jr.; Sung, S.S. Diverse and potent chemokine production by lung CD11bhigh dendritic cells in homeostasis and in allergic lung inflammation. J. Immunol. 2007, 178, 1882–1895. [Google Scholar] [CrossRef] [PubMed]
  64. Ng, S.L.; Teo, Y.J.; Setiagiani, Y.A. Type 1 Conventional CD103(+) Dendritic Cells Control Effector CD8(+) T Cell Migration, Survival, and Memory Responses During Influenza Infection. Front. Immunol. 2018, 9, 3043. [Google Scholar] [CrossRef] [PubMed]
  65. Desch, A.N.; Randolph, G.J.; Murphy, K.; Gautier, E.L.; Kedl, R.M.; Lahoud, M.H.; Jakubzick, C.V. CD103+ pulmonary dendritic cells preferentially acquire and present apoptotic cell-associated antigen. J. Exp. Med. 2011, 208, 1789–1797. [Google Scholar] [CrossRef] [PubMed]
  66. Mousavi, S.F.; Soroosh, P.; Takahashi, T.; Yoshikai, Y.; Shen, H.; Lefrançois, L.; Ishii, N. OX40 costimulatory signals potentiate the memory commitment of effector CD8+ T cells. J. Immunol. 2008, 181, 5990–6001. [Google Scholar] [CrossRef] [PubMed]
  67. Joshi, N.S.; Cui, W.; Chandele, A.; Lee, H.K.; Urso, D.R.; Hagman, J.; Kaech, S.M. 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. [Google Scholar] [CrossRef] [PubMed]
  68. Herndler-Brandstetter, D.; Ishigame, H.; Shinnakasu, R.; Plajer, V.; Stecher, C.; Zhao, J.; Inoue, T. KLRG1(+) Effector CD8(+) T Cells Lose KLRG1, Differentiate into All Memory T Cell Lineages, and Convey Enhanced Protective Immunity. Immunity 2018, 48, 716–729. [Google Scholar] [CrossRef] [PubMed]
  69. Gaide, O.; Emerson, R.O.; Jiang, X.; Gulati, N.; Nizza, S.; Desmarais, C.; Kupper, T.S. Common clonal origin of central and resident memory T cells following skin immunization. Nat. Med. 2015, 21, 647–653. [Google Scholar] [CrossRef] [PubMed]
  70. Mackay, L.K.; Rahimpour, A.; Ma, J.Z.; Collins, N.; Stock, A.T.; Hafon, M.L.; Tscharke, D.C. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat. Immunol. 2013, 14, 1294–1301. [Google Scholar] [CrossRef] [PubMed]
  71. Mami-Chouaib, F.; Blanc, C.; Corgnac, S.; Hans, S.; Malenica, I.; Granier, C.; Tartour, E. Resident memory T cells, critical components in tumor immunology. J. Immunother. Cancer 2018, 6, 87. [Google Scholar] [CrossRef] [PubMed]
  72. Kumar, B.V.; Kratchmarov, R.; Miron, M.; Carpenter, D.J.; Senda, T.; Lerner, H.; Farber, D.L. Functional heterogeneity of human tissue-resident memory T cells based on dye efflux capacities. JCI Insight 2018, 3, 123568. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, Y.; Zander, R.; Khatun, A.; Schauder, D. Transcriptional and Epigenetic Regulation of Effector and Memory CD8 T Cell Differentiation. Front. Immunol. 2018, 9, 2826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kumar, B.V.; Ma, W.; Miron, M.; Granot, T.; Guyer, R.S.; Carpenter, D.J.; Friedman, A.L. Human Tissue-Resident Memory T Cells Are Defined by Core Transcriptional and Functional Signatures in Lymphoid and Mucosal Sites. Cell Rep. 2017, 20, 2921–2934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Scott-Browne, J.P.; Lopez-Moyado, I.F.; Trifari, S.; Wong, V.; Chavez, L.; Rao, A.; Pereira, R.M. Dynamic Changes in Chromatin Accessibility Occur in CD8(+) T Cells Responding to Viral Infection. Immunity 2016, 45, 1327–1340. [Google Scholar] [CrossRef] [PubMed]
  76. Yang, C.Y.; Best, J.A.; Knell, J.; Yang, E.; Sheridan, A.D.; Jesionek, A.K.; Watowich, S.S. The transcriptional regulators Id2 and Id3 control the formation of distinct memory CD8+ T cell subsets. Nat. Immunol. 2011, 12, 1221–1229. [Google Scholar] [CrossRef] [PubMed]
  77. Cannarile, M.A.; Lind, N.A.; Rivera, R.; Sheridan, A.D.; Camfield, K.A.; Wu, B.B.; Goldrath, A.W. Transcriptional regulator Id2 mediates CD8+ T cell immunity. Nat. Immunol. 2006, 7, 1317–1325. [Google Scholar] [CrossRef]
  78. Rutishauser, R.L.; Martins, G.A.; Kalachikov, S.; Chandele, A.; Parish, I.A.; Meffre, E.; Kaech, S.M. Transcriptional repressor Blimp-1 promotes CD8(+) T cell terminal differentiation and represses the acquisition of central memory T cell properties. Immunity 2009, 31, 296–308. [Google Scholar] [CrossRef]
  79. Intlekofer, A.M.; Takemoto, N.; Wherry, E.J.; Longworth, S.A.; Northrup, J.T.; Palanivel, V.R.; Gapin, L. Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nat. Immunol. 2005, 6, 1236–1244. [Google Scholar] [CrossRef]
  80. Ji, Y.; Pos, Z.; Rao, M.; Klebanoff, C.A.; Yu, Z.; Sukumar, M.; Wang, E. Repression of the DNA-binding inhibitor Id3 by Blimp-1 limits the formation of memory CD8+ T cells. Nat. Immunol. 2011, 12, 1230–1237. [Google Scholar] [CrossRef]
  81. Zhou, X.; Yu, S.; Zhao, D.M.; Harty, J.T.; Badovinac, V.P.; Xue, H.H. Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1. Immunity 2010, 33, 229–240. [Google Scholar] [CrossRef] [PubMed]
  82. Ichii, H.; Sakamoto, A.; Kuroda, Y.; Tokuhisa, T. Bcl6 acts as an amplifier for the generation and proliferative capacity of central memory CD8+ T cells. J. Immunol. 2004, 173, 883–891. [Google Scholar] [CrossRef] [PubMed]
  83. Cui, W.; Liu, Y.; Weinstein, J.S.; Craft, J.; Kaech, S.M. An interleukin-21-interleukin-10-STAT3 pathway is critical for functional maturation of memory CD8+ T cells. Immunity 2011, 35, 792–805. [Google Scholar] [CrossRef] [PubMed]
  84. Banerjee, A.; Gordon, S.M.; Intlekofer, A.M.; Paley, M.A.; Mooney, E.C.; Lindsten, T.; Reiner, S.L. Cutting edge: The transcription factor eomesodermin enables CD8+ T cells to compete for the memory cell niche. J. Immunol. 2010, 185, 4988–4992. [Google Scholar] [CrossRef] [PubMed]
  85. Milner, J.J.; Toma, C.; Yu, B.; Zhang, K.; Omilusik, K.; Phan, A.T.; Wang, W. Runx3 programs CD8(+) T cell residency in non-lymphoid tissues and tumours. Nature 2017, 552, 253–257. [Google Scholar] [CrossRef]
  86. Kallies, A.; Xin, A.; Belz, G.T.; Nutt, S.L. Blimp-1 transcription factor is required for the differentiation of effector CD8(+) T cells and memory responses. Immunity 2009, 31, 283–295. [Google Scholar] [CrossRef] [PubMed]
  87. Behr, F.M.; Kragten, N.A.; Wesselink, T.H.; Nota, B.; Van Lier, R.A.; Amsen, D.; Van Gisbergen, K.P. Blimp-1 Rather Than Hobit Drives the Formation of Tissue-Resident Memory CD8(+) T Cells in the Lungs. Front. Immunol. 2019, 10, 400. [Google Scholar] [CrossRef]
  88. Kragten, N.A.; Behr, F.M.; Vieira Braga, F.A.; Remmerswaal, E.B.; Wesselink, T.H.; Oja, A.E.; van Gisbergen, K.P. Blimp-1 induces and Hobit maintains the cytotoxic mediator granzyme B in CD8 T cells. Eur. J. Immunol. 2018, 48, 1644–1662. [Google Scholar] [CrossRef] [Green Version]
  89. Mackay, L.K.; Minnich, M.; Kragten, N.A.; Liao, Y.; Nota, B.; Seillet, C.; Braun, A. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 2016, 352, 459–463. [Google Scholar] [CrossRef] [Green Version]
  90. Nath, A.P.; Braun, A.; Ritchie, S.C.; Carbone, F.R.; Mackay, L.K.; Gebhardt, T.; Inouye, M. Comparative analysis reveals a role for TGF-beta in shaping the residency-related transcriptional signature in tissue-resident memory CD8+ T cells. PLoS ONE 2019, 14, e0210495. [Google Scholar] [CrossRef]
  91. Mohammed, J.; Beura, L.K.; Bobr, A.; Astry, B.; Chicoine, B.; Kashem, S.W.; Matte, C. Stromal cells control the epithelial residence of DCs and memory T cells by regulated activation of TGF-beta. Nat. Immunol. 2016, 17, 414–421. [Google Scholar] [CrossRef] [PubMed]
  92. Mackay, L.K.; Wynne-Jones, E.; Freestone, D.; Pellicci, D.G.; Mielke, L.A.; Newman, D.M.; Carbone, F.R. T-box Transcription Factors Combine with the Cytokines TGF-beta and IL-15 to Control Tissue-Resident Memory T Cell Fate. Immunity 2015, 43, 1101–1111. [Google Scholar] [CrossRef] [PubMed]
  93. Mackay, L.K.; Braun, A.; Macleod, B.L.; Collins, N.; Tebartz, C.; Bedoui, S.; Gebhardt, T. Cutting edge: CD69 interference with sphingosine-1-phosphate receptor function regulates peripheral T cell retention. J. Immunol. 2015, 194, 2059–2063. [Google Scholar] [CrossRef] [PubMed]
  94. Schenkel, J.M.; Fraser, K.A.; Casey, K.A.; Beura, L.K.; Pauken, K.E.; Vezys, V.; Masopust, D. IL-15-Independent Maintenance of Tissue-Resident and Boosted Effector Memory CD8 T Cells. J. Immunol. 2016, 196, 3920–3926. [Google Scholar] [CrossRef] [PubMed]
  95. Richer, M.J.; Pewe, L.L.; Hancox, L.S.; Hartwig, S.M.; Varga, S.M.; Harty, J.T. Inflammatory IL-15 is required for optimal memory T cell responses. J. Clin. Investig. 2015, 125, 3477–3490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Ziegler, S.F.; Ramsdell, F.; Hjerrild, K.A.; Armitage, R.J.; Grabstein, K.H.; Hennen, K.B.; Alderson, M.R. Molecular characterization of the early activation antigen CD69: A type II membrane glycoprotein related to a family of natural killer cell activation antigens. Eur. J. Immunol. 1993, 23, 1643–1648. [Google Scholar] [CrossRef] [PubMed]
  97. Shiow, L.R.; Rosen, D.B.; Brdičková, N.; Xu, Y.; An, J.; Lanier, L.L.; Matloubian, M. CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 2006, 440, 540–544. [Google Scholar] [CrossRef] [PubMed]
  98. Skon, C.N.; Lee, J.Y.; Anderson, K.G.; Masopust, D.; Hogquist, K.A.; Jameson, S.C. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 2013, 14, 1285–1293. [Google Scholar] [CrossRef] [Green Version]
  99. Kennel, S.J.; Lankford, T.K.; Foote, L.J.; Shinpock, S.G.; Stringer, C. CD44 expression on murine tissues. J. Cell Sci. 1993, 104, 373–382. [Google Scholar]
  100. Lesley, J.; Howes, N.; Perschl, A.; Hyman, R. Hyaluronan binding function of CD44 is transiently activated on T cells during an in vivo immune response. J. Exp. Med. 1994, 180, 383–387. [Google Scholar] [CrossRef]
  101. Mikecz, K.; Brennan, F.R.; Kim, J.H.; Glant, T.T. Anti-CD44 treatment abrogates tissue oedema and leukocyte infiltration in murine arthritis. Nat. Med. 1995, 1, 558–563. [Google Scholar] [CrossRef] [PubMed]
  102. Bank, I.; Book, M.; Ware, R. Functional role of VLA-1 (CD49A) in adhesion, cation-dependent spreading, and activation of cultured human T lymphocytes. Cell. Immunol. 1994, 156, 424–437. [Google Scholar] [CrossRef] [PubMed]
  103. Roberts, A.I.; Brolin, R.E.; Ebert, E.C. Integrin alpha1beta1 (VLA-1) mediates adhesion of activated intraepithelial lymphocytes to collagen. Immunology 1999, 97, 679–685. [Google Scholar] [CrossRef] [PubMed]
  104. Haddadi, S.; Thanthrige-Don, N.; Afkhami, S.; Khera, A.; Jeyanathan, M.; Xing, Z. Expression and role of VLA-1 in resident memory CD8 T cell responses to respiratory mucosal viral-vectored immunization against tuberculosis. Sci. Rep. 2017, 7, 9525. [Google Scholar] [CrossRef] [PubMed]
  105. Cheuk, S.; Schlums, H.; Sérézal, I.G.; Martini, E.; Chiang, S.C.; Marquardt, N.; Höög, C. CD49a Expression Defines Tissue-Resident CD8(+) T Cells Poised for Cytotoxic Function in Human Skin. Immunity 2017, 46, 287–300. [Google Scholar] [CrossRef] [PubMed]
  106. Hadley, G.A.; Bartlett, S.T.; Via, C.S.; Rostapshova, E.A.; Moainie, S. The epithelial cell-specific integrin, CD103 (alpha E integrin), defines a novel subset of alloreactive CD8+ CTL. J. Immunol. 1997, 159, 3748–3756. [Google Scholar] [PubMed]
  107. Pizzolla, A.; Nguyen, T.H.; Sant, S.; Jaffar, J.; Loudovaris, T.; Mannering, S.I.; Wakim, L.M. Influenza-specific lung-resident memory T cells are proliferative and polyfunctional and maintain diverse TCR profiles. J. Clin. Investig. 2018, 128, 721–733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Lee, Y.T.; Suarez-Ramirez, J.E.; Wu, T.; Redman, J.M.; Bouchard, K.; Hadley, G.A.; Cauley, L.S. Environmental and antigen receptor-derived signals support sustained surveillance of the lungs by pathogen-specific cytotoxic T lymphocytes. J. Virol. 2011, 85, 4085–4094. [Google Scholar] [CrossRef]
  109. Takamura, S. Persistence in Temporary Lung Niches: A Survival Strategy of Lung-Resident Memory CD8(+) T Cells. Viral Immunol. 2017, 30, 438–450. [Google Scholar] [CrossRef]
  110. Takamura, S. Niches for the Long-Term Maintenance of Tissue-Resident Memory T Cells. Front. Immunol. 2018, 9, 1214. [Google Scholar] [CrossRef]
  111. Zaid, A.; Mackay, L.K.; Rahimpour, A.; Braun, A.; Veldhoen, M.; Carbone, F.R.; Mueller, S.N. Persistence of skin-resident memory T cells within an epidermal niche. Proc. Natl. Acad. Sci. USA 2014, 111, 5307–5312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Bergsbaken, T.; Bevan, M.J.; Fink, P.J. Local Inflammatory Cues Regulate Differentiation and Persistence of CD8(+) Tissue-Resident Memory T Cells. Cell Rep. 2017, 19, 114–124. [Google Scholar] [CrossRef] [PubMed]
  113. Iijima, N.; Iwasaki, A. T cell memory. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science 2014, 346, 93–98. [Google Scholar] [CrossRef] [PubMed]
  114. Takamura, S.; Yagi, H.; Hakata, Y.; Motozono, C.; McMaster, S.R.; Masumoto, T.; Umemura, M. Specific niches for lung-resident memory CD8+ T cells at the site of tissue regeneration enable CD69-independent maintenance. J. Exp. Med. 2016, 213, 3057–3073. [Google Scholar] [CrossRef] [PubMed]
  115. Mackay, L.K.; Stock, A.T.; Ma, J.Z.; Jones, C.M.; Kent, S.J.; Mueller, S.N.; Gebhardt, T. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc. Natl. Acad. Sci. USA 2012, 109, 7037–7042. [Google Scholar] [CrossRef] [PubMed]
  116. McMaster, S.R.; Wein, A.N.; Dunbar, P.R.; Hayward, S.L.; Cartwright, E.K.; Denning, T.L.; Kohlmeier, J.E. Pulmonary antigen encounter regulates the establishment of tissue-resident CD8 memory T cells in the lung airways and parenchyma. Mucosal Immunol. 2018, 11, 1071–1078. [Google Scholar] [CrossRef] [Green Version]
  117. Ely, K.H.; Cookenham, T.; Roberts, A.D.; Woodland, D.L. Memory T cell populations in the lung airways are maintained by continual recruitment. J. Immunol. 2006, 176, 537–543. [Google Scholar] [CrossRef]
  118. Di Virgilio, F.; Dal Ben, D.; Sarti, A.C.; Giuliani, A.L.; Falzoni, S. The P2X7 Receptor in Infection and Inflammation. Immunity 2017, 47, 15–31. [Google Scholar] [CrossRef] [Green Version]
  119. Borges da Silva, H.; Beura, L.K.; Wang, H.; Hanse, E.A.; Gore, R.; Scott, M.C.; Hippen, K.L. The purinergic receptor P2RX7 directs metabolic fitness of long-lived memory CD8(+) T cells. Nature 2018, 559, 264–268. [Google Scholar] [CrossRef]
  120. Stark, R.; Wesselink, T.H.; Behr, F.M.; Kragten, N.A.; Arens, R.; Koch-Nolte, F.; van Lier, R.A. T RM maintenance is regulated by tissue damage via P2RX7. Sci. Immunol. 2018, 3. [Google Scholar] [CrossRef]
  121. Clark, R.A. Resident memory T cells in human health and disease. Sci. Transl. Med. 2015, 7, 269rv1. [Google Scholar] [CrossRef] [PubMed]
  122. Sun, H.; Sun, C.; Xiao, W.; Sun, R. Tissue-resident lymphocytes: From adaptive to innate immunity. Cell. Mol. Immunol. 2019, 16, 205–215. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, K.; Kolls, J.K. T cell-mediated host immune defenses in the lung. Annu. Rev. Immunol. 2013, 31, 605–633. [Google Scholar] [CrossRef] [PubMed]
  124. Jiang, J.; Fisher, E.M.; Murasko, D.M. CD8 T cell responses to influenza virus infection in aged mice. Ageing Res. Rev. 2011, 10, 422–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Jozwik, A.; Habibi, M.S.; Paras, A.; Zhu, J.; Guvenel, A.; Dhariwal, J.; Del Rosario, J. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nat. Commun. 2015, 6, 10224. [Google Scholar] [CrossRef] [PubMed]
  126. Pizzolla, A.; Nguyen, T.H.; Smith, J.M.; Brooks, A.G.; Kedzierska, K.; Heath, W.R.; Wakim, L.M. Resident memory CD8(+) T cells in the upper respiratory tract prevent pulmonary influenza virus infection. Sci. Immunol. 2017, 2, 6970. [Google Scholar] [CrossRef] [PubMed]
  127. Piet, B.; de Bree, G.J.; Smids-Dierdorp, B.S.; van der Loos, C.M.; Remmerswaal, E.B.; Jan, H.; Timens, W. CD8(+) T cells with an intraepithelial phenotype upregulate cytotoxic function upon influenza infection in human lung. J. Clin. Investig. 2011, 121, 2254–2263. [Google Scholar] [CrossRef]
  128. McMaster, S.R.; Wilson, J.J.; Wang, H.; Kohlmeier, J.E. Airway-Resident Memory CD8 T Cells Provide Antigen-Specific Protection against Respiratory Virus Challenge through Rapid IFN-gamma Production. J. Immunol. 2015, 195, 203–209. [Google Scholar] [CrossRef]
  129. Aguilar-Valenzuela, R.; Netland, J.; Seo, Y.J.; Bevan, M.J.; Grakoui, A.; Suthar, M.S. Dynamics of Tissue-Specific CD8(+) T Cell Responses during West Nile Virus Infection. J. Virol. 2018, 92. [Google Scholar] [CrossRef]
  130. Cautivo, K.M.; Bueno, S.M.; Cortes, C.M.; Wozniak, A.; Riedel, C.A.; Kalergis, A.M. Efficient lung recruitment of respiratory syncytial virus-specific Th1 cells induced by recombinant bacillus Calmette-Guerin promotes virus clearance and protects from infection. J. Immunol. 2010, 185, 7633–7645. [Google Scholar] [CrossRef]
  131. Bueno, S.M.; Gonzalez, P.A.; Cautivo, K.M.; Mora, J.E.; Leiva, E.D.; Tobar, H.E.; Kalergis, A.M. Protective T cell immunity against respiratory syncytial virus is efficiently induced by recombinant BCG. Proc. Natl. Acad. Sci. USA 2008, 105, 20822–20827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Cespedes, P.F.; Rey-Jurado, E.; Espinoza, J.A.; Rivera, C.A.; Canedo-Marroquín, G.; Bueno, S.M.; Kalergis, A.M. A single, low dose of a cGMP recombinant BCG vaccine elicits protective T cell immunity against the human respiratory syncytial virus infection and prevents lung pathology in mice. Vaccine 2017, 35, 757–766. [Google Scholar] [CrossRef] [PubMed]
  133. Schwarz, B.; Morabito, K.M.; Ruckwardt, T.J.; Patterson, D.P.; Avera, J.; Miettinen, H.M.; Douglas, T. Viruslike Particles Encapsidating Respiratory Syncytial Virus M and M2 Proteins Induce Robust T Cell Responses. ACS Biomater. Sci. Eng. 2016, 2, 2324–2332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Schmidt, M.E.; Knudson, C.J.; Hartwig, S.M.; Pewe, L.L.; Meyerholz, D.K.; Langlois, R.A.; Varga, S.M. Memory CD8 T cells mediate severe immunopathology following respiratory syncytial virus infection. PLoS Pathog. 2018, 14, e1006810. [Google Scholar] [CrossRef] [PubMed]
  135. Li, N.; Zhang, L.; Zheng, B.; Li, W.; Liu, J.; Zhang, H.; Zeng, R. RSV recombinant candidate vaccine G1F/M2 with CpG as an adjuvant prevents vaccine-associated lung inflammation, which may be associated with the appropriate types of immune memory in spleens and lungs. Hum. Vaccin. Immunother. 2019, 15, 1–11. [Google Scholar] [CrossRef]
  136. Shin, H.; Iwasaki, A. A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature 2012, 491, 463–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Caminschi, I.; Lahoud, M.H.; Pizzolla, A.; Wakim, L.M. Zymosan by-passes the requirement for pulmonary antigen encounter in lung tissue-resident memory CD8(+) T cell development. Mucosal Immunol. 2019, 12, 403–412. [Google Scholar] [CrossRef] [PubMed]
  138. Zhang, L.; Li, H.; Hai, Y.; Yin, W.; Li, W.; Zheng, B.; Zeng, R. CpG in Combination with an Inhibitor of Notch Signaling Suppresses Formalin-Inactivated Respiratory Syncytial Virus-Enhanced Airway Hyperresponsiveness and Inflammation by Inhibiting Th17 Memory Responses and Promoting Tissue-Resident Memory Cells in Lungs. J. Virol. 2017, 91, e02111–e02116. [Google Scholar] [CrossRef]
  139. Van Braeckel-Budimir, N.; Varga, S.M.; Badovinac, V.P.; Harty, J.T. Repeated Antigen Exposure Extends the Durability of Influenza-Specific Lung-Resident Memory CD8(+) T Cells and Heterosubtypic Immunity. Cell Rep. 2018, 24, 3374–3382. [Google Scholar] [CrossRef]
  140. Mora, J.R.; Cheng, G.; Picarella, D.; Briskin, M.; Buchanan, N.; von Andrian, U.H. Reciprocal and dynamic control of CD8 T cell homing by dendritic cells from skin- and gut-associated lymphoid tissues. J. Exp. Med. 2005, 201, 303–316. [Google Scholar] [CrossRef] [Green Version]
  141. Iwata, M.; Hirakiyama, A.; Eshima, Y.; Kagechika, H.; Kato, C.; Song, S.Y. Retinoic acid imprints gut-homing specificity on T cells. Immunity 2004, 21, 527–538. [Google Scholar] [CrossRef] [PubMed]
  142. Graham, B.S.; Bunton, L.A.; Wright, P.F.; Karzon, D.T. Role of T lymphocyte subsets in the pathogenesis of primary infection and rechallenge with respiratory syncytial virus in mice. J. Clin. Investig. 1991, 88, 1026–1033. [Google Scholar] [CrossRef] [PubMed]
  143. Cannon, M.J.; Openshaw, P.J.; Askonas, B.A. Cytotoxic T cells clear virus but augment lung pathology in mice infected with respiratory syncytial virus. J. Exp. Med. 1988, 168, 1163–1168. [Google Scholar] [CrossRef] [PubMed]
  144. Mazur, N.I.; Horsley, N.M.; Englund, J.A.; Nederend, M.; Magaret, A.; Kumar, A.; Steinhoff, M.C. Breast Milk Prefusion F Immunoglobulin G as a Correlate of Protection Against Respiratory Syncytial Virus Acute Respiratory Illness. J. Infect. Dis. 2019, 219, 59–67. [Google Scholar] [CrossRef] [PubMed]
  145. Fernandez-Ruiz, D.; Ng, W.Y.; Holz, L.E.; Ma, J.Z.; Zaid, A.; Wong, Y.C.; Li, J. Liver-Resident Memory CD8(+) T Cells Form a Front-Line Defense against Malaria Liver-Stage Infection. Immunity 2016, 45, 889–902. [Google Scholar] [CrossRef] [PubMed]
  146. Olsen, T.M.; Stone, B.C.; Chuenchob, V.; Murphy, S.C. Prime-and-Trap Malaria Vaccination To Generate Protective CD8(+) Liver-Resident Memory T Cells. J. Immunol. 2018, 201, 1984–1993. [Google Scholar] [CrossRef] [PubMed]
  147. Cuburu, N.; Kim, R.; Guittard, G.C.; Thompson, C.D.; Day, P.M.; Hamm, D.E.; Schiller, J.T. A Prime-Pull-Amplify Vaccination Strategy To Maximize Induction of Circulating and Genital-Resident Intraepithelial CD8(+) Memory T Cells. J. Immunol. 2019, 202, 1250–1264. [Google Scholar] [CrossRef] [PubMed]
  148. Cuburu, N.; Khan, S.; Thompson, C.D.; Kim, R.; Vellinga, J.; Zahn, R.; Schiller, J.T. Adenovirus vector-based prime-boost vaccination via heterologous routes induces cervicovaginal CD8(+) T cell responses against HPV16 oncoproteins. Int. J. Cancer 2018, 142, 1467–1479. [Google Scholar] [CrossRef]
  149. Galvez-Cancino, F.; Lopez, E.; Menares, E.; Díaz, X.; Flores, C.; Cáceres, P.; Varas-Godoy, M. Vaccination-induced skin-resident memory CD8(+) T cells mediate strong protection against cutaneous melanoma. Oncoimmunology 2018, 7, e1442163. [Google Scholar] [CrossRef]
  150. Dumauthioz, N.; Labiano, S.; Romero, P. Tumor Resident Memory T Cells: New Players in Immune Surveillance and Therapy. Front. Immunol. 2018, 9, 2076. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Steps occurring during the activation and differentiation of T lymphocytes. (a). Antigen Presentation. Activation of T cells requires the assembly of an immunological synapse with APCs, which provides three types of signals: i) interaction between the TCR and a peptide-loaded MHC class I or class II molecule, ii) co-stimulatory molecule signaling and iii) specific cytokines. (b). Proliferation/Differentiation. T cell activation induces T cell proliferation to clonally select and expand antigen-specific T cells. (c). Effector functions, surface markers and some cytokines required for homeostasis [53,54,55,56,57,58] and transcription factors that characterize each subpopulation of memory T cells. Central memory T cells (TCM) express lymphoid homing markers and circulate through secondary lymphoid organs (SLO). The effector memory T cells (TEM) lack expression of lymphoid homing markers, but express other migratory receptors with the potential to migrate through non-lymphoid tissues. The third subset of memory T cells, which consists of resident memory T cells (TRM), stably reside in non-lymphoid tissues and contribute to enhancing innate and adaptive immunity against pathogens.
Figure 1. Steps occurring during the activation and differentiation of T lymphocytes. (a). Antigen Presentation. Activation of T cells requires the assembly of an immunological synapse with APCs, which provides three types of signals: i) interaction between the TCR and a peptide-loaded MHC class I or class II molecule, ii) co-stimulatory molecule signaling and iii) specific cytokines. (b). Proliferation/Differentiation. T cell activation induces T cell proliferation to clonally select and expand antigen-specific T cells. (c). Effector functions, surface markers and some cytokines required for homeostasis [53,54,55,56,57,58] and transcription factors that characterize each subpopulation of memory T cells. Central memory T cells (TCM) express lymphoid homing markers and circulate through secondary lymphoid organs (SLO). The effector memory T cells (TEM) lack expression of lymphoid homing markers, but express other migratory receptors with the potential to migrate through non-lymphoid tissues. The third subset of memory T cells, which consists of resident memory T cells (TRM), stably reside in non-lymphoid tissues and contribute to enhancing innate and adaptive immunity against pathogens.
Pathogens 08 00147 g001

Share and Cite

MDPI and ACS Style

Retamal-Díaz, A.; Covián, C.; Pacheco, G.A.; Castiglione-Matamala, A.T.; Bueno, S.M.; González, P.A.; Kalergis, A.M. Contribution of Resident Memory CD8+ T Cells to Protective Immunity against Respiratory Syncytial Virus and Their Impact on Vaccine Design. Pathogens 2019, 8, 147. https://doi.org/10.3390/pathogens8030147

AMA Style

Retamal-Díaz A, Covián C, Pacheco GA, Castiglione-Matamala AT, Bueno SM, González PA, Kalergis AM. Contribution of Resident Memory CD8+ T Cells to Protective Immunity against Respiratory Syncytial Virus and Their Impact on Vaccine Design. Pathogens. 2019; 8(3):147. https://doi.org/10.3390/pathogens8030147

Chicago/Turabian Style

Retamal-Díaz, Angello, Camila Covián, Gaspar A. Pacheco, Angelo T. Castiglione-Matamala, Susan M. Bueno, Pablo A. González, and Alexis M. Kalergis. 2019. "Contribution of Resident Memory CD8+ T Cells to Protective Immunity against Respiratory Syncytial Virus and Their Impact on Vaccine Design" Pathogens 8, no. 3: 147. https://doi.org/10.3390/pathogens8030147

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

Article Metrics

Back to TopTop