Skip to main content
Pathogens and Disease logoLink to Pathogens and Disease
. 2021 Mar 9;79(4):ftab014. doi: 10.1093/femspd/ftab014

T cell responses to Chlamydia

Jennifer D Helble 1, Michael N Starnbach 2,
PMCID: PMC8012111  PMID: 33693620

ABSTRACT

Chlamydia trachomatis is the most commonly reported sexually transmitted infection in the United States. The high prevalence of infection and lack of a vaccine indicate a critical knowledge gap surrounding the host's response to infection and how to effectively generate protective immunity. The immune response to C. trachomatis is complex, with cells of the adaptive immune system playing a crucial role in bacterial clearance. Here, we discuss the CD4+ and CD8+ T cell response to Chlamydia, the importance of antigen specificity and the role of memory T cells during the recall response. Ultimately, a deeper understanding of protective immune responses is necessary to develop a vaccine that prevents the inflammatory diseases associated with Chlamydia infection.

Keywords: T cells, Chlamydia trachomatis, immunity, immune evasion, T cell functions, interferon gamma


Here, the authors review the functions by which T cells limit Chlamydia trachomatis infections as well as the ways this pathogen inhibits T cell immunity.

INTRODUCTION

Chlamydia trachomatis is an obligate intracellular Gram-negative bacterium that primarily infects mucosal epithelial cells. Chlamydia species grow inside a membrane-bound vacuole called an inclusion and have a biphasic developmental cycle, where the bacteria alternate between the elementary body and the reticulate body (Ward 1983; Bastidas et al. 2013; Elwell, Mirrashidi and Engel 2016). Genital Chlamydia infection is the most commonly reported bacterial sexually transmitted infection in the United States with an estimated 3 million new cases each year (Wiesenfeld 2017; Centers for Disease Control and Prevention 2018). While the numbers are striking—539.9 cases per 100 000 people—the overall reported rate of Chlamydia infection in the United States drastically underscores the higher rates of infection in women, which are about twice as high compared with men (Centers for Disease Control and Prevention 2018). Young women in particular have extremely high rates of infection—in 2018, 4% of women aged 20–24 tested positive for Chlamydia infection (Centers for Disease Control and Prevention 2018).

Despite these high rates of infection, there is no vaccine against Chlamydia. While readily treated with antibiotics, around 75% of genital infections are asymptomatic and are therefore at a high risk for being left untreated (Farley, Cohen and Elkins 2003; Mylonas 2012; CDC 2017). Additionally, antibiotic treatment does not preclude women from subsequent infections, and repeated or untreated genital infection can lead to excessive inflammation in the upper genital tract resulting in multiple disease sequelae, including pelvic inflammatory disease, which can have long-lasting consequences on reproductive health (Risser and Risser 2007; Hosenfeld et al. 2009; Brunham, Gottlieb and Paavonen 2015). Ultimately, an ideal prophylactic vaccine would prevent not only infection but also the downstream inflammatory diseases associated with Chlamydia infection (Starnbach 2018). In order to generate protection against future infections, most vaccines aim to target the adaptive immune system, as it is the arm of the immune system that is critical for forming immune memory. Indeed, the adaptive immune response to C. trachomatis is absolutely essential for generating future protection. Much of the work on immunity to C. trachomatis has been conducted in mice, and these studies will be the focus of this review. Here, we will focus on the importance of T cell-driven immunity and provide perspective on the different hurdles that need to be overcome in order to generate protective immunity against C. trachomatis.

OVERVIEW OF MURINE ADAPTIVE IMMUNITY AGAINST CHLAMYDIA

Components of the innate immune system are often critical to jump-start adaptive immunity. During Chlamydia infection, natural killer cells (NK cells) traffic to the infected tissue early during infection and are the primary early producers of interferon-γ (IFNγ), which helps to shape the adaptive immune response (Tseng and Rank 1998). Other innate immune cells, including CD103 dendritic cells, have been shown to acquire C. trachomatis antigen in the genital tract and then traffic to the draining lymph node to activate T cells (Stary et al. 2015).

While the innate immune system can rapidly detect and respond to Chlamydia via various pattern recognition receptors and resulting cytokine production (Darville et al. 2003; O'Connell et al. 2006; Derbigny et al. 2012; Barker et al. 2013; Massari et al. 2013), the adaptive immune system is required to mediate Chlamydia clearance and protection against reinfection. Despite the fact that Chlamydia are obligate intracellular bacteria, they can activate multiple arms of the adaptive immune system. Humoral immunity, mediated by B cells, can lead to the production of antibodies. In the context of C. trachomatis infection, B cells appear to be dispensable for clearance during primary infection (Morrison, Feilzer and Tumas 1995). There is some evidence to suggest that B cells may play an important role in clearing secondary C. trachomatis infection, although the mechanism for how this occurs has not yet been elucidated (Su et al. 1997) and is therefore not a focus of this review.

The cellular part of the adaptive immune response is mediated by T cells, which recognize antigen via the membrane-bound T cell receptor (TCR) (reviewed in Davis and Bjorkman 1988; Alcover, Alarcón and Di Bartolo 2018). There are two main subsets of T cells, characterized by the type of TCR present—αβ T cells and γδ T cells—the latter of which are thought to be less critical during Chlamydia infection (Davis and Bjorkman 1988; Perry, Feilzer and Caldwell 1997; Yang, Hayglasst and Brunham 1998). In order for the TCR to recognize foreign Chlamydia antigen, the proteins need to be processed into peptides and presented on major histocompatibility complex (MHC) class I or II. Below, we detail the differences in antigen recognition between MHC I and MHC II, and the importance of antigen-specific responses in combating Chlamydia infection.

CD8+ T CELL-MEDIATED IMMUNITY AGAINST CHLAMYDIA

MHC I is expressed on all nucleated cells and classically presents antigen originating from the cytosol to T cells. These cytosolic proteins, which can originate from intracellular pathogens, are degraded by the proteasome and peptides are funneled into the endoplasmic reticulum by the protein TAP. Peptides are then loaded into the binding groove of MHC I by the chaperone protein tapasin and the fully loaded peptide:MHC I complex is exported to the surface of the cell (reviewed in Neefjes et al. 2011). Typically, the TCR on CD8+ T cells recognizes antigen presented on MHC I. This recognition results in the upregulation of various effector cytokines, including IFNγ, as well as the release of perforin and granzyme cytotoxic molecules that can result in target cell death (Halle, Halle and Förster 2017). CD8+ T cells are therefore thought to play a major role in the immune response to intracellular pathogens, as they can directly deplete the intracellular niche required for pathogen replication.

Indeed, it has been shown that CD8+ T cells recognizing C. trachomatis proteins class I accessible protein-1 (Cap1) and cysteine-rich protein A (CrpA) can kill target cells in an antigen-dependent manner (Fling et al. 2000; Starnbach et al. 2003). Despite the ability of CD8+ T cells to target infected cells in vitro, CD8+ T cells are dispensable for clearance of Chlamydia in mice, as CD8−/− and perforin-deficient animals clear infection at the same rate as wild-type (WT) mice (Johansson et al. 1997; Perry et al. 1999). Despite this, IFNγ production by CD8+ T cells is critical for resolution of Chlamydia infection, as CD8+ T cell lines derived from Chlamydia-infected mice are only protective against infection if they can produce IFNγ (Starnbach, Bevan and Lampe 1994; Lampe et al. 1998).

In addition, antigen-specific CD8+ T cells can be programmed to be protective against Chlamydia infection. Transgenic NR23.4 T cells that recognize CrpA can protect mice against C. trachomatis when adoptively transferred into mice prior to bacterial challenge (Roan and Starnbach 2006). Additionally, mice that are vaccinated with recombinant vaccinia virus expressing CrpA (VacCrpA) develop a CD8+ T cell population specific for CrpA that is protective upon infection with C. trachomatis (Loomis and Starnbach 2006; Zhang and Starnbach 2015). Using MHC I tetramers, it is also possible to track endogenous CrpA-specific CD8+ T cells to monitor the kinetics of the C. trachomatis-specific CD8+ T cell response over time. For many pathogens, the resolution of primary infection results in the formation of a stable memory T cell population. Following secondary infection, these memory T cells expand more rapidly and to a higher extent compared with primary infection. However, the CD8+ T cell response to C. trachomatis infection does not follow this same pattern, as mice that are infected and then re-challenged with C. trachomatis have fewer numbers of CrpA CD8+ T cells during the secondary response, suggesting some impairment with natural memory formation (Loomis and Starnbach 2006; Fankhauser and Starnbach 2014).

Recent work has demonstrated that the immunoinhibitory molecule PD-L1 (programmed death-ligand 1) is upregulated on uterine epithelial cells and dendritic cells in the draining lymph node after primary and secondary C. trachomatis infection (Fig. 1). This upregulation skews CrpA-specific CD8+ T cells toward a central memory (Tcm) phenotype (Fankhauser and Starnbach 2014). Tcm cells typically circulate through the secondary lymphoid organs (SLOs) and lymphatic vessels but are not well equipped to home to or exert effector functions in peripheral tissues. Effector memory T cells (Tem) are able to circulate from SLOs to peripheral tissues and are thought to aid in clearance of pathogens that infect these tissues (Mueller et al. 2013). When PD-L1 is blocked or deleted in mice during C. trachomatis infection, the T cell population shifts toward a Tem phenotype and C. trachomatis is cleared more rapidly. Additionally, PD-L1 deficiency increases the number of IFNγ producing CD8+ T cells during secondary infection and the IFNγ produced by CD8+ T cells is sufficient to protect against C. trachomatis infection (Fig. 1) (Fankhauser and Starnbach 2014). However, it is important to consider that immunoinhibitory molecules often act to regulate inflammation by preventing T cell activation and consequent cytokine production. In Chlamydia muridarum-infected mice, blockade of the immunoinhibitory molecules PD-L1 and TIM3 enhances uterine and oviduct pathology (Peng et al. 2011).

Figure 1.

Figure 1.

Impact of PD-L1 upregulation on the CD8+ T cell response to C. trachomatis. Left: During infection, uterine epithelial cells and lymph node dendritic cells upregulate the immunoinhibitory molecule PD-L1. MHC I presentation of the C. trachomatis antigen CrpA in conjunction with PD-L1 expression results in antigen-specific CD8+ T cells being programmed to the Tcm phenotype. Right: When PD-L1 is blocked, antigen-specific CD8+ T cells are skewed to the Tem phenotype, resulting in enhanced IFNγ production and Chlamydia clearance.

Although immunoinhibitory molecules have been extensively studied during chronic viral infections and cancer, the upregulation of these molecules during chronic bacterial infections suggests that potentially targeting these pathways to promote T cell reactivation may aid in bacterial clearance but could also contribute to enhanced downstream inflammation. Several studies have asserted that the C. muridarum CD8+ T cell response contributes to pathology (Manam, Nicholson and Murthy 2013; Vlcek et al. 2016). However, the basis of these findings is that WT mice have enhanced pathology following infection compared with transgenic mice with fixed CD8+ T cell reactivity to ovalbumin (OT-I mice). A comparison of the responses in WT mice versus OT-I mice reflects only the immunopathology caused by OT-I cells and may not reflect the diversity of clonal responses (whether antigen-specific or bystander) stimulated by infection of WT mice. When designing a vaccine against C. trachomatis, it is important to take both of these factors into account in order to adequately generate a long-lasting, effective memory population without excess inflammation that could cause downstream disease.

CD4+ T CELL-MEDIATED IMMUNITY AGAINST CHLAMYDIA

While CD8+ T cells classically recognize endogenous antigen presented on MHC I, CD4+ T cells recognize exogenous antigen presented on MHC II. In contrast to MHC I, MHC II expression is restricted to professional antigen presenting cells (APCs), including dendritic cells, macrophages and B cells (Neefjes et al. 2011). Typically, MHC II presents peptides derived from extracellular proteins that have been endocytosed by APCs and degraded by early endosome proteases. During Chlamydia infection, this can occur through engulfment of extracellular elementary bodies or capture of dying infected epithelial cells. Once the peptide:MHC II complex has assembled, it is then transported to the cell surface to be recognized by the TCR on CD4+ T cells (reviewed in Neefjes et al. 2011). Recognition of antigen leads to CD4+ T cell differentiation into distinct T cell subtypes (reviewed in O'Shea and Paul 2010), characterized by the upregulation of a master transcription factor that culminates in the production of specific cytokines. CD4+ T cell subtype cytokine production ultimately helps to resolve specific types of infection or disease. For example, Th1 cells, which are characterized by their large production of proinflammatory cytokines like IFNγ, are particularly important for clearing viral infections and intracellular bacterial pathogens (Zhu 2018). Indeed, Th1 cells are thought to be the predominant T cell subset in the context of Chlamydia infection. These T cells differentiate following IFNγ and IL-12 production by innate immune cells early during infection (Johansson et al. 1997; Tseng and Rank 1998; Gondek, Roan and Starnbach 2009; Jiao et al. 2011). While there are several other CD4+ T cell subtypes, including Th2, Th17, Th22 and Th9 cells (Zhu 2018), it is unclear what role they play during C. trachomatis infection.

CD4+ T cells play a critical role during Chlamydia infection. Previous work has demonstrated that CD4+ T cells are necessary and sufficient for Chlamydia clearance and protection against reinfection (Su and Caldwell 1995; Gondek et al. 2012). The identification of the CD4+ T cell antigen Cta1 (Chlamydia-specific T cell antigen 1) has allowed for further discovery of how these CD4+ T cells are capable of clearing infection (Roan et al. 2006). CD4+ T cells from TCR transgenic mice that recognize a specific 20-mer peptide from Cta1 (Cta1133–152), termed NR1 T cells, have been critical for determining the effects of antigen-specific CD4+ T cells during C. trachomatis infection. Following genital tract infection with C. trachomatis, NR1 T cells are activated in the genital tract draining iliac lymph nodes (Fig. 2). Upon antigen stimulation, these T cells upregulate the activation marker CD44 and downregulate the lymph node retention marker, CD62L (Roan et al. 2006). In order to traffic to the genital tract, the T cells upregulate the chemokine receptors CXCR3 and CCR5 (Olive, Gondek and Starnbach 2011) and the integrin α4β1 (Fig. 2) (Davila, Olive and Starnbach 2014). These molecules are critical for driving T cell-mediated protection in the genital tract—blockade or deletion of either the chemokine receptors or integrins prevents antigen-specific CD4+ T cells from homing to the genital tract, resulting in higher bacterial burden.

Figure 2.

Figure 2.

Overview of antigen-specific CD4+ T cell immunity to C. trachomatis. Upon adoptive transfer of transgenic NR1 T cells into mice and subsequent transcervical infection with C. trachomatis, NR1 T cells are activated in the draining lymph node. This results in downregulation of the lymph node retention marker CD62L and upregulation of the activation marker CD44. To travel to the genital tract, NR1 T cells also upregulate the integrin α4β1 and chemokine receptors CXCR3 and CCR5. Upon entry into the genital tract, NR1 T cells cluster in specific sections of the genital tract that correlate with where C. trachomatis is located.

T cell activation and trafficking to the site of infection also occur in an antigen-dependent manner. NR1 T cells do not proliferate or become activated in mice infected with Salmonella enterica or Listeria monocytogenes. Similarly, OT-II T cells, which are specific for a peptide fragment in the protein ovalbumin (OVA323–339) that is not found in C. trachomatis, are unable to become activated and traffic to the genital tract following C. trachomatis infection (Roan et al. 2006). Two-photon microscopy has become a valuable tool for visualizing these CD4+ T cell populations in the genital tract in response to C. trachomatis. Using fluorescent NR1 T cells, our lab has shown that the antigen-specific CD4+ T cell response to C. trachomatis in the genital tract is not evenly distributed (Helble et al. 2020). Rather, there are distinct sections within the genital tract that contain greater numbers of NR1 T cells (Fig. 2). Sections enriched with NR1 T cells correspond to higher C. trachomatis burdens, suggesting that NR1 T cells are able to home specifically to sections in the genital tract that contain bacteria (Helble et al. 2020). However, it is unknown what drives these gradients and how the NR1 T cells are able to home to specific sections within the genital tract. It is possible that these gradients of antigen-specific CD4+ T cells are also reflective of the adaptive immune response to Chlamydia infection in the human female genital tract. There have been a limited number of human studies examining infection and immune responses in the genital tract using stained tissue sections. However, these studies have indicated that lymphoid follicles (similar to germinal centers) do exist in the genital tract tissue (Kiviat et al. 1990b), as well as CD4+ T cell clustering in the endometrial tissue (Vicetti Miguel et al. 2013). It is unclear if these patches of immune cells correlate with where the bacteria are localized, but it does provide evidence that the observed NR1 T cell clustering in mice is reminiscent of the immune response in humans. While there has been no direct correlation of adaptive immune cells and C. trachomatis burden in human samples, Kiviat et al. did provide evidence for neutrophil aggregation surrounding C. trachomatis-infected cells (Kiviat et al. 1990a), suggesting that certain immune responses to C. trachomatis may be targeted to specific areas in the genital tract in humans.

Once in the genital tract, NR1 T cells have been shown to be sufficient to protect mice against reinfection (Gondek et al. 2012). It is thought that CD4+ T cell-mediated clearance of Chlamydia is driven by IFNγ production from Th1 CD4+ T cells. Indeed, NR1 T cells that are pre-skewed as Th2 or Th17 prior to adoptive transfer do not confer protection to mice infected with C. trachomatis (Gondek, Roan and Starnbach 2009). While Th1 T cells can produce other cytokines in addition to IFNγ, the importance of IFNγ production was reenforced when it was discovered that CD4+ T cell-mediated clearance of C. trachomatis requires the host to sense IFNγ. Mice that are deficient in the IFNγ receptor (IFNγR−/− mice) are unable to clear C. trachomatis infection, even in the presence of IFNγ producing CD4+ T cells (Gondek, Roan and Starnbach 2009). Additionally, NR1 T cell-mediated IFNγ production but not IFNγ sensing is required for C. trachomatis clearance and preventing NR1 T cell accumulation in the genital tract (Helble et al. 2020).

While the use of NR1 transgenic T cells has been essential to studying antigen-specific CD4+ T cells in mice in response to C. trachomatis, the T cells are not cross-reactive to C. muridarum. Although C. muridarum contains an analogous Cta1 protein, there exist several amino acid differences in the 20-mer peptide that NR1 T cells recognize. The differences in these residues may be critical for TCR contact, which could explain why they are not effective against C. muridarum. It is also possible Cta1 is not an effective T cell antigen in C. muridarum. Despite the inability of NR1 T cells to recognize Cta1 in C. muridarum, antigen-specific CD4+ T cells are still important for clearance of C. muridarum in mice. Peptides derived from multiple C. muridarum proteins have been shown to bind to MHC II and elicit a CD4+ T cell response (Karunakaran et al. 2008, 2015; Yu et al. 2011). Adoptive transfer of dendritic cells pulsed with these peptides also have the potential to protect mice against genital infection, and represent an intriguing approach toward vaccine design (Karunakaran et al. 2008, 2015). In addition, MHC II tetramers against C. muridarum peptides have been useful for studying the kinetics of the CD4+ T cell response to infection and the potential for B cell help in shaping protective T cell immunity (Li and McSorley 2013).

In 2017, the development of a new CD4+ TCR transgenic mouse specific for C. muridarum has allowed for mechanistic studies on antigen-specific CD4+ T cells in this mouse model of infection (Poston et al. 2017). These CD4+ T cells become activated in the draining iliac lymph nodes where they upregulate CD44 and downregulate CD62L. Following activation, the T cells traffic to the genital tract using currently unknown chemokine receptors and integrins. Once in the genital tract, these T cells produce IFNγ and can subsequently reduce bacterial burden in mice infected intravaginally with C. muridarum (Poston et al. 2017). Although it is unknown which C. muridarum antigen these T cells recognize, these transgenic T cells are cross-reactive to C. trachomatis (Poston et al. 2017).

The specific requirements for what makes an effective CD4+ T cell antigen in the context of Chlamydia infection are unknown. Currently, Cta1 is the most well-characterized CD4+ T cell antigen in C. trachomatis (Roan et al. 2006), and very little is known about this protein's function within the infected cell or how it gets processed for antigen presentation on MHC II. Genetic manipulation of Chlamydia is paramount in order to address these questions (Wang et al. 2011; Agaisse and Derré 2013; Johnson and Fisher 2013; Mueller, Wolf and Fields 2016). To assess the role of Cta1 during infection and eliciting a CD4+ T cell response, an attractive option would be to simply create a Cta1 deficient C. trachomatis strain. This is theoretically possible using either TargeTron (Johnson and Fisher 2013), a technique involving the insertion of mobile group II introns into the coding sequence of Cta1 and thereby gene inactivation, or FRAEM (fluorescence-reported allelic exchange mutagenesis) (Mueller, Wolf and Fields 2016), using allelic exchange to replace Cta1 with a fluorescent marker, thereby eliminating it from the genome. Alternative approaches involve introducing heterologous antigens that are localized to different sections in the infected cell (host cell cytoplasm, inclusion membrane, etc.) or under different promoters to induce expression at specific points during the developmental cycle in order to determine what role these factors have in proper antigen presentation and stimulation of a protective CD4+ T cell response. We have started to address these questions, with the introduction of the MHC II restricted ovalbumin peptide, OVA323–339, to the cytoplasm of C. trachomatis (Helble and Starnbach 2019). While OT-II CD4+ T cells were stimulated in response to C. trachomatis-OVA, these cells could not be programmed to be protective and underscore the complexities surrounding CD4+ antigen presentation of a vacuolar pathogen and effective protection against a mucosal pathogen. Future studies should aim to determine when Cta1 is expressed across the developmental cycle, where within the infected Cta1 is expressed, and how this changes between C. trachomatis and C. muridarum in order to identify specific characteristics necessary for appropriate CD4+ T cell activation.

IFNγ-MEDIATED CONTROL OF CHLAMYDIA INFECTION

IFNγ produced by T cells can lead to a downstream signaling cascade in infected epithelial cells, culminating in the upregulation of several interferon-stimulated genes (ISGs) that can help control intracellular bacterial replication. Secreted IFNγ binds to the IFNγR complex, comprising two distinct chains (IFNγR1 and IFNγR2) (Pestka, Krause and Walter 2004). This binding event induces the phosphorylation of the two cytoplasmic associated Janus-kinases (JAK), JAK1 and JAK2. JAK2 phosphorylation leads to STAT1 (signal transducer and activator of transcription 1) recruitment and eventual phosphorylation. Dimerized phosphorylated STAT1 can then translocate into the nucleus to act as a transcription factor to upregulate ISGs and other interferon responsive transcription factors (Pestka, Krause and Walter 2004; Bhat et al. 2018).

During C. trachomatis infection in humans, this IFNγ signaling cascade leads to the upregulation of the protein indoleamine 2,3-dioxygenase (IDO) in genital tract epithelial cells (Fig. 3) (Thomas et al. 1993; Chon, Hassanain and Gupta 1996). Once expressed, IDO can degrade intracellular tryptophan stores by catalyzing the breakdown of tryptophan into N-formylkynurenine and kynurenine. Through this, IDO can act to starve C. trachomatis of this essential nutrient, inducing C. trachomatis to enter a state of persistence (Fig. 3) (Beatty et al. 1994). While C. trachomatis replication is limited in the absence of tryptophan, removal of IFNγ and subsequent decreased expression of IDO can cause persistent inclusions to become viable once more (Beatty, Byrne and Morrison 1993). Certain genital tract strains are able to overcome tryptophan depletion, as they express tryptophan synthase and can use exogenous indole to produce their own tryptophan (Aiyar et al. 2014).

Figure 3.

Figure 3.

IFNγ signaling induces upregulation of IDO and can impair the CD4+ T cell response. In humans, IFNγ production by CD4+ T cells can upregulate IDO. IDO breaks down tryptophan into N-formylkynurenine and kynurenine. Because C. trachomatis requires tryptophan to survive, IDO-mediated depletion of this amino acid results in a dormant, persistent inclusion. Kynurenines can go on to inhibit the CD4+ T cell response through a variety of mechanisms. Together, upregulation of IDO and inhibition of the CD4+ T cell response contribute to the persistence of C. trachomatis in humans.

The by-product of tryptophan catabolism, kynurenine, can have a detrimental effect on the host CD4+ T cell response (Fig. 3). Kynurenines can directly and indirectly inhibit the CD4+ T cell response through a variety of mechanisms, including limiting their proliferation, promoting the differentiation of naïve T cells into regulatory T cells and enhancing the immunoregulatory function of antigen presenting cells (Terness et al. 2002; Belladonna et al. 2007; Baban et al. 2009; Mezrich et al. 2010). Limited T cell proliferation and regulatory T cell formation would lead to limited IFNγ production. Similarly, enhancing immunoregulatory capabilities of antigen presenting cells could limit T cell activation and proper Th1 T cell formation. Given that CD4+ T cells are potent producers of IFNγ, inhibiting this pathway through IDO catabolism of tryptophan can result in the decreased production of IFNγ and ultimately the reactivation of C. trachomatis.

In mice, IDO is neither upregulated by IFNγ nor is it upregulated to sufficient amounts to control pathogen replication during C. trachomatis infection (Roshick et al. 2006). Instead, IFNγ induces the upregulation of a family of interferon-inducible p47GTPases called the immunity-related GTPases (IRGs) (Bernstein-Hanley et al. 2006). The IRG family consists of ∼20 different proteins that all exert cell autonomous control over intracellular pathogen growth. On a molecular level during C. trachomatis infection, murine IRGs work cooperatively to identify the location of the inclusion within the host cell and can then promote the ubiquitination of unknown substrates on the inclusion membrane. As a result, the inclusion membrane breaks, releasing C. trachomatis into the host cell cytosol and ultimately into autolysosomes where the bacteria are degraded (Haldar et al. 2015). In contrast, C. muridarum inclusions in murine cells are not ubiquitinated or targeted for destruction by IRGs (Haldar et al. 2015). This host cell tropism is indicative of the evolution of Chlamydia species that have built up resistance mechanisms against species-specific cell autonomous immunity in host cells.

Species-specific cell autonomous immunity can also account for differences in mouse infection models of Chlamydia. Intravaginal infection with C. muridarum yields a robust infection that is able to ascend to the upper genital tract of mice, in stark contrast to the rapid clearance of C. trachomatis in mice infected intravaginally (Gondek et al. 2012). It is likely that the murine IRGs, to which C. trachomatis is susceptible but C. muridarum is not, play a role in clearing C. trachomatis intravaginally before it can ascend to the upper genital tract of mice. Through removal of these mouse-specific innate immune responses, we can start to build a more accurate mouse model of chronic human C. trachomatis infection. Indeed, when mice lack two important IRG regulatory proteins, Irgm1 and Irgm3, transcervical infection with C. trachomatis results in exacerbated bacterial burden compared with WT mice (Coers et al. 2011). It is unclear if C. trachomatis deposited intravaginally in Irgm1/3−/− mice would be able to ascend to the upper genital tract, but the removal of these mouse-specific innate immune responses does allow for a better representation of modeled human infection. Species-specific immunity can also be seen with C. muridarum infection in human cells. Although there have been no reports of humans becoming infected with C. muridarum, when human cells are infected with C. muridarum in vitro, C. muridarum can overcome tryptophan starvation caused by IDO expression (Roshick et al. 2006). In summary, IFNγ production by CD4+ T cells can bind to the IFNγR on epithelial cells, leading to a downstream signaling cascade culminating in ISG upregulation. In turn, this leads to initial cell autonomous control of Chlamydia infection.

TISSUE RESIDENT MEMORY T CELLS AND MUCOSAL PRIMING

Both naïve and memory antigen-specific CD4+ T cells have been studied independently; however, direct comparisons of the two populations in the same animal can determine the specific contributions of each cell type during secondary C. trachomatis infection. Using RNA-sequencing, work from our lab has shown that naïve antigen-specific CD4+ T cells in the genital tract draining lymph nodes were more abundant and more proliferative than memory antigen-specific CD4+ T cells (Helble, Mann and Starnbach 2020). While these data were surprising, as memory T cells have been shown to proliferate faster in response to antigen (Rogers, Dubey and Swain 2000; Veiga-Fernandes et al. 2000; Hu et al. 2001; Whitmire, Eam and Whitton 2008), it is possible that these results were simply due to the time point and anatomical site investigated. A similar experimental setup can be used to identify transcriptional differences between other T cell populations, and future experiments should aim to compare differences between bulk endogenous CD4+ T cells and antigen-specific CD4+ T cells in order to identify other genes important for generating protection.

While described above in the context of CD8+ T cells, memory CD4+ T cells have been traditionally grouped into two separate categories: central memory (Tcm) and effector memory (Tem). Like their CD8+ T cell counterparts, CD4+ Tcm cells circulate primarily within the lymphatics, while CD4+ Tem cells are able to home to peripheral, nonlymphoid tissues (Mueller et al. 2013). As such, it is thought that CD4+ Tem cells play a predominant role in clearing genital Chlamydia infections. Recently, a third subset of memory T cells, termed tissue resident memory T cells (Trms), has been discovered. In contrast to Tem cells, which can recirculate into the lymphatics and blood following pathogen clearance, Trm cells remain in nonlymphoid tissue after the pathogen has been cleared and can be maintained at peripheral tissues for long periods of time even in the absence of persisting antigen (Mueller et al. 2013; Shin and Iwasaki 2013; Schenkel and Masopust 2014). Upon re-exposure to pathogens in peripheral tissues, Trm cells act as a first line of defense. These cells are able to respond more rapidly to the pathogenic insult than other memory T cell subsets that need to traffic to the tissue in order to respond.

Similar to Tem cells, Trm cells downregulate the lymph node homing marker CD62L in order to traffic to nonlymphoid organs. In contrast to Tem cells, Trm cells upregulate the C-type lectin CD69 (Schenkel and Masopust 2014). CD69 is briefly expressed on all T cells following initial activation but is then immediately downregulated for Tem and Tcm cells. The role of CD69 in Trm cells is still unclear, but it is known that CD69 expression is also associated with downregulation of sphingosine-1-phosophate receptor 1 (S1P1) (Shiow et al. 2006; Shin and Iwasaki 2013). S1P1 expression on T cells is important for T cell egress from the lymphatics, and it is thought that S1P1 downregulation may be an important step for driving Trm cell formation through preventing recirculation through the lymphatics (Schenkel and Masopust 2014). While less well characterized for CD4+ Trm cells, CD8+ Trm cells have also been shown to upregulate the αEβ7 integrin (CD103) (Mueller et al. 2013) that plays an important role in Trm cell retention in peripheral tissues. The ligand for CD103, E-cadherin, is expressed on epithelial cells in nonlymphoid tissues (Cepek et al. 1994). As such, it is thought that CD103 expressed on Trm cells can interact with E-cadherin on epithelial cells, leading to Trm cell maintenance in the tissue.

While CD69 (and in some T cell subsets CD103) can be a useful marker to identify Trm cells in the tissue, one of the most definitive methods of proving tissue residence and the importance of Trm cells in clearing infection is through parabiosis studies. Parabiosis is the surgical joining of two organisms resulting in shared circulatory systems in the two conjoined partners (Kamran et al. 2013). Migratory memory T cells (Tem and Tcm cells) are able to circulate between both organisms equally. However, memory T cells that do not travel to the conjoined partner but rather are maintained in the tissue of the host are be considered true Trm cells. Indeed, the discovery of CD4+ Trm cells during C. trachomatis infection was established through parabiosis (Stary et al. 2015). Optimal clearance of secondary C. trachomatis infection in mice is dependent on both the establishment of CD4+ Trm cells and the recruitment of circulating memory T cells to the upper genital tract mucosa. Despite the fact that circulating memory T cells can contribute to secondary C. trachomatis clearance, mice that only receive circulating memory T cells but not Trm cells are unable to clear secondary C. trachomatis infection as effectively, highlighting the importance of these resident T cells (Stary et al. 2015). The generation of genital tract Trm cells following C. trachomatis infection is dependent on a mucosal priming event. Mice that are infected intranasally or transcervically but not subcutaneously with C. trachomatis are able to establish Trm cells in the genital tract. The importance of Trm cells is not limited to C. trachomatis infection. Protective Trm cells have been well documented during infection with pathogens that infect mucosal peripheral tissues, including influenza virus (lung), herpes simplex virus (skin, genital tract) and Mycobacterium tuberculosis (lung) (Gebhardt et al. 2009; Mackay et al. 2012; Shin and Iwasaki 2012; Iijima and Iwasaki 2014; Perdomo et al. 2016; Pizzolla et al. 2017).

CROSS-MUCOSAL PROTECTION

Mucosal sites are often where pathogens first encounter the host and where pathogens leave the host to infect other individuals. Despite being located in completely different sections of the body, infection or vaccination at one mucosal surface can elicit protection at another, suggesting shared elements of immunity between these two distal mucosal sites. Aside from inducing a Trm population, intranasal infection with C. trachomatis induces both a CD4+ and a CD8+ T cell response that together can reduce burden in the genital tract (Nogueira et al. 2015). IFNγ also plays a role in eliciting this cross-mucosal protection: mice deficient in IFNγ or treated with an IFNγ depleting antibody are unable to clear transcervical C. trachomatis infection after intranasal priming. The ability of the host to respond to IFNγ is also necessary in order for cross-mucosal protection to occur (Nogueira et al. 2015). This effect has also been demonstrated with other Chlamydia antigens. Mice that are primed with live C. muridarum intranasally are protected against intravaginal C. muridarum infection (Yu et al. 2011). Similarly, mice vaccinated intranasally with recombinant C. muridarum protein CPAF (Chlamydia protease-like activity factor) and then challenged intravaginally had less bacterial shedding and reduced pathology (Murthy et al. 2007). Ultimately, it is clear that Trm and mucosal priming are necessary to generate protection against Chlamydia.

CONCLUDING REMARKS

The prevalence of Chlamydia infections has been steadily on the rise and poses an important public health problem both in the United States and worldwide. The inability of humans to develop protective immunity against Chlamydia has significant ramifications—repeat infections are common and can lead to excessive inflammation, resulting in multiple disease sequelae (Risser and Risser 2007; Hosenfeld et al. 2009). In order to develop an effective vaccine against Chlamydia, it is necessary to understand what constitutes protective immunity versus inflammatory pathology. By enhancing protective immune populations, we can hopefully build an ideal vaccine against Chlamydia, ultimately protecting against both Chlamydia pathogenesis and the associated inflammatory diseases. While this review has largely focused on the protective aspects of antigen-specific T cells following Chlamydia infection, it is important to note that an influx of antigen non-specific bystander CD4+ and CD8+ T cells has been shown to induce excessive immunopathology in the genital tract of mice (Lijek et al. 2018). Preventing these bulk T cell populations from entering the genital tract has no impact on antigen-specific T cell populations, and can significantly reduce genital tract pathology (Lijek et al. 2018).

Novel imaging techniques and high-throughput sequencing can provide valuable insight into host responses to genital Chlamydia infection. Two-photon microscopy of genital tract tissues allows for visualization of adaptive immune cells in whole tissues. There are multiple applications to this technology, including identifying interactions between antigen-specific T cells and other cells of the genital tract, such as epithelial cells or innate immune cells. Using this in combination with RNA-sequencing, researchers can identify specific components of the adaptive immune response that are critical for promoting protection against Chlamydia infection while simultaneously preventing pathology. Armed with this knowledge, we can build a better model of Chlamydia infection with the ultimate goal of designing a vaccine against this important mucosal pathogen.

ACKNOWLEDGMENT

We wish to apologize to those colleagues whose work we could not cite or discuss due to space limitations.

Supplementary Material

ftab014_Supplemental_Files

Contributor Information

Jennifer D. Helble, Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.

Michael N. Starnbach, Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.

FUNDING

This work was supported by the National Institutes of Health (AI39558 to MNS).

Conflict of interest

None declared.

REFERENCES

  1. Agaisse H, Derré I.. A C. trachomatis cloning vector and the generation of C. trachomatis strains expressing fluorescent proteins under the control of a C. trachomatis promoter. PLoS One. 2013;8:e57090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aiyar A, Quayle AJ, Buckner LRet al. . Influence of the tryptophan-indole-IFNγ axis on human genital Chlamydia trachomatis infection: role of vaginal co-infections. Front Cell Infect Microbiol. 2014;4:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alcover A, Alarcón B, Di Bartolo V.. Cell biology of T cell receptor expression and regulation. Annu Rev Immunol. 2018;36:103–25. [DOI] [PubMed] [Google Scholar]
  4. Baban B, Chandler PR, Sharma MDet al. . IDO activates regulatory T cells and blocks their conversion into Th17-like T cells. J Immunol. 2009;183:2475–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Barker JR, Koestler BJ, Carpenter VKet al. . STING-dependent recognition of cyclic di-AMP mediates type I interferon responses during Chlamydia trachomatis infection. mBio. 2013;4:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bastidas RJ, Elwell CA, Engel JNet al. . Chlamydial intracellular survival strategies. Cold Spring Harb Perspect Med. 2013;3:a010256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Beatty WL, Belanger TA, Desai AAet al. . Tryptophan depletion as a mechanism of gamma interferon-mediated chlamydial persistence. Infect Immun. 1994;62:3705–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Beatty WL, Byrne GI, Morrison RP.. Morphologic and antigenic characterization of interferon gamma-mediated persistent Chlamydia trachomatis infection in vitro. Proc Natl Acad Sci USA. 1993;90:3998–4002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Belladonna ML, Puccetti P, Orabona Cet al. . Immunosuppression via tryptophan catabolism: the role of kynurenine pathway enzymes. Transplantation. 2007;84:17–20. [DOI] [PubMed] [Google Scholar]
  10. Bernstein-Hanley I, Coers J, Balsara ZRet al. . The p47 GTPases Igtp and Irgb10 map to the Chlamydia trachomatis susceptibility locus Ctrq-3 and mediate cellular resistance in mice. Proc Natl Acad Sci USA. 2006;103:14092–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bhat MY, Solanki HS, Advani Jet al. . Comprehensive network map of interferon gamma signaling. J Cell Commun Signal. 2018;12:745–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Brunham RC, Gottlieb SL, Paavonen J.. Pelvic inflammatory disease. N Engl J Med. 2015;372:2039–48. [DOI] [PubMed] [Google Scholar]
  13. CDC . Chlamydia—CDC fact sheet. 2017. [Google Scholar]
  14. Centers for Disease Control and Prevention. Sexually Transmitted Disease Surveillance 2018. 2018. [Google Scholar]
  15. Cepek KL, Shaw SK, Parker CMet al. . Adhesion between epithelial cells and T lymphocytes mediated by E-cadherin and the ⍺eβ7 integrin. Nature. 1994;372:190–3. [DOI] [PubMed] [Google Scholar]
  16. Chon SY, Hassanain HH, Gupta SL.. Cooperative role of interferon regulatory factor 1 and p91 (STAT1) response elements in interferon-γ-inducible expression of human indoleamine 2,3-dioxygenase gene. J Biol Chem. 1996;271:17247–52. [DOI] [PubMed] [Google Scholar]
  17. Coers J, Gondek DC, Olive AJet al. . Compensatory T cell responses in IRG-deficient mice prevent sustained Chlamydia trachomatis infections. PLoS Pathog. 2011;7:e1001346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Darville T, O'Neill JM, Andrews CWet al. . Toll-like receptor-2, but not Toll-like receptor-4, is essential for development of oviduct pathology in chlamydial genital tract infection. J Immunol. 2003;171:6187–97. [DOI] [PubMed] [Google Scholar]
  19. Davila SJ, Olive AJ, Starnbach MN.. Integrin α4β1 is necessary for CD4+ T cell−mediated protection against genital Chlamydia trachomatis infection. J Immunol. 2014;192:4284–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Davis MM, Bjorkman PJ.. The T cell receptor genes and T cell recognition. Nature. 1988;334:395–402. [DOI] [PubMed] [Google Scholar]
  21. Derbigny WA, Shobe LR, Kamran JCet al. . Identifying a role for Toll-like receptor 3 in the innate immune response to Chlamydia muridarum infection in murine oviduct epithelial cells. Infect Immun. 2012;80:254–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Elwell C, Mirrashidi K, Engel J.. Chlamydia cell biology and pathogenesis. Nat Publ Gr. 2016;14:385–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Fankhauser SC, Starnbach MN.. PD-L1 limits the mucosal CD8+ T cell response to Chlamydia trachomatis. J Immunol. 2014;192:1079–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Farley TA, Cohen DA, Elkins W.. Asymptomatic sexually transmitted diseases: the case for screening. Prev Med. 2003;36:502–9. [DOI] [PubMed] [Google Scholar]
  25. Fling SP, Sutherland RA, Steele LNet al. . CD8+ T cells recognize an inclusion membrane-associated protein from the vacuolar pathogen Chlamydia trachomatis. Proc Natl Acad Sci USA. 2000;98:1160–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Gebhardt T, Wakim LM, Eidsmo Let al. . Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat Immunol. 2009;10:524–30. [DOI] [PubMed] [Google Scholar]
  27. Gondek DC, Olive AJ, Stary Get al. . CD4+ T cells are necessary and sufficient to confer protection against Chlamydia trachomatis infection in the murine upper genital tract. J Immunol. 2012;189:2441–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Gondek DC, Roan NR, Starnbach MN.. T cell responses in the absence of IFN-γ exacerbate uterine infection with Chlamydia trachomatis. J Immunol. 2009;183:1313–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Haldar AK, Foltz C, Finethy Ret al. . Ubiquitin systems mark pathogen-containing vacuoles as targets for host defense by guanylate binding proteins. Proc Natl Acad Sci USA. 2015;112:E5628–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Halle S, Halle O, Förster R.. Mechanisms and dynamics of T cell-mediated cytotoxicity in vivo. Trends Immunol. 2017;38:432–43. [DOI] [PubMed] [Google Scholar]
  31. Helble JD, Gonzalez RJ, von Andrian UHet al. . Gamma interferon is required for Chlamydia clearance but is dispensable for T Cell homing to the genital tract. mBio. 2020;11:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Helble JD, Mann AO, Starnbach MN.. Antigen-specific memory and naïve CD4+ T cells following secondary Chlamydia trachomatis infection. PLoS One. 2020;15:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Helble JD, Starnbach MN.. A Chlamydia trachomatis strain expressing ovalbumin stimulates an antigen-specific CD4+ T cell response in mice. Infect Immun. 2019;87:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Hosenfeld CB, Workowski KA, Berman Set al. . Repeat infection with Chlamydia and gonorrhea among females: a systematic review of the literature. Sex Transm Dis. 2009;36:478–89. [DOI] [PubMed] [Google Scholar]
  35. Hu H, Huston G, Duso Det al. . CD4+ T cell effectors can become memory cells with high efficiency and without further division. Nat Immunol. 2001;2:705–10. [DOI] [PubMed] [Google Scholar]
  36. Iijima N, Iwasaki A.. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science. 2014;346:93–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Jiao L, Gao X, Joyee AGet al. . NK cells promote type 1 T cell immunity through modulating the function of dendritic cells during intracellular bacterial infection. J Immunol. 2011;187:401–11. [DOI] [PubMed] [Google Scholar]
  38. Johansson M, Schön K, Ward Met al. . Studies in knockout mice reveal that anti-chlamydial protection requires TH1 cells producing IFN-γ: is this true for humans?. Scand J Immunol. 1997;46:546–52. [DOI] [PubMed] [Google Scholar]
  39. Johnson CM, Fisher DJ.. Site-specific, insertional inactivation of incA in Chlamydia trachomatis using a group II intron. PLoS One. 2013;8:e83989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Kamran P, Sereti KI, Zhao Pet al. . Parabiosis in mice: a detailed protocol. J Vis Exp. 2013:50556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Karunakaran KP, Rey-Ladino J, Stoynov Net al. . Immunoproteomic discovery of novel T cell antigens from the obligate intracellular pathogen Chlamydia. J Immunol. 2008;180:2459–65. [DOI] [PubMed] [Google Scholar]
  42. Karunakaran KP, Yu H, Jiang Xet al. . Outer membrane proteins preferentially load MHC class II peptides: implications for a Chlamydia trachomatis T cell vaccine. Vaccine. 2015;33:2159–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Kiviat NB, Paavonen JA, Wolner-Hanssen Pet al. . Histopathology of endocervical infection caused by Chlamydia trachomatis, herpes simplex virus, Trichomonas vaginalis, and Neisseria gonorrhoeae. Hum Pathol. 1990a;21:831–7. [DOI] [PubMed] [Google Scholar]
  44. Kiviat NB, Wolner-Hanssen P, Eschenbach DAet al. . Endometrial histopathology in patients with culture-proved upper genital tract infection and laparoscopically diagnosed acute salpingitis. Am J Surg Pathol. 1990b;14:167–75. [DOI] [PubMed] [Google Scholar]
  45. Lampe MF, Wilson CB, Bevan MJet al. . Gamma interferon production by cytotoxic T lymphocytes is required for resolution of Chlamydia trachomatis infection. Infect Immun. 1998;66:5457–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Lijek RS, Helble JD, Olive AJet al. . Pathology after Chlamydia trachomatis infection is driven by nonprotective immune cells that are distinct from protective populations. Proc Natl Acad Sci USA. 2018;115:2216–2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Li L-X, McSorley SJ.. B cells enhance antigen-specific CD4 T cell priming and prevent bacteria dissemination following Chlamydia muridarum genital tract infection. PLoS Pathog. 2013;9:e1003707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Loomis WP, Starnbach MN.. Chlamydia trachomatisinfection alters the development of memory CD8+ T cells. J Immunol. 2006;177:4021–7. [DOI] [PubMed] [Google Scholar]
  49. Mackay LK, Stock AT, Ma JZet al. . 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;109:7037–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Manam S, Nicholson BJ, Murthy AK.. OT-1 mice display minimal upper genital tract pathology following primary intravaginal Chlamydia muridarum infection. Pathog Dis. 2013;67:221–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Massari P, Toussi DN, Tifrea DFet al. . Toll-like receptor 2-dependent activity of native major outer membrane protein proteosomes of Chlamydia trachomatis. Infect Immun. 2013;81:303–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Mezrich JD, Fechner JH, Zhang Xet al. . An Interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol. 2010;185:3190–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Morrison RP, Feilzer K, Tumas DB.. Gene knockout mice establish a primary protective role for major histocompatibility complex class II-restricted responses in Chlamydia trachomatis genital tract infection. Infect Immun. 1995;63:4661–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Mueller KE, Wolf K, Fields KA.. Gene deletion by fluorescence-reported allelic exchange mutagenesis in Chlamydia trachomatis. mBio. 2016;7:e01817–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Mueller SN, Gebhardt T, Carbone FRet al. . Memory T cell subsets, migration patterns, and tissue residence. Annu Rev Immunol. 2013;31:137–61. [DOI] [PubMed] [Google Scholar]
  56. Murthy AK, Chambers JP, Meier PAet al. . Intranasal vaccination with a secreted chlamydial protein enhances resolution of genitalChlamydia muridarum infection, protects against oviduct pathology, and is highly dependent upon endogenous gamma interferon production. Infect Immun. 2007;75:666–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Mylonas I. Female genital Chlamydia trachomatis infection: where are we heading?. Arch Gynecol Obstet. 2012;285:1271–85. [DOI] [PubMed] [Google Scholar]
  58. Neefjes J, Jongsma MLM, Paul Pet al. . Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol. 2011;11:823–36. [DOI] [PubMed] [Google Scholar]
  59. Nogueira C V., Zhang X, Giovannone Net al. . Protective immunity against Chlamydia trachomatis can engage both CD4+ and CD8+ T cells and bridge the respiratory and genital mucosae. J Immunol. 2015;194:2319–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. O'Connell CM, Ionova IA, Quayle AJet al. . Localization of TLR2 and MyD88 to Chlamydia trachomatis inclusions. J Biol Chem. 2006;281:1652–9. [DOI] [PubMed] [Google Scholar]
  61. O'Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science. 2010;327:1098–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Olive AJ, Gondek DC, Starnbach MN.. CXCR3 and CCR5 are both required for T cell-mediated protection against C. trachomatis infection in the murine genital mucosa. Mucosal Immunol. 2011;4:208–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Peng B, Lu C, Tang Let al. . Enhanced upper genital tract pathologies by blocking Tim-3 and PD-L1 signaling pathways in mice intravaginally infected with Chlamydia muridarum. BMC Infect Dis. 2011;11:347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Perdomo C, Zedler U, Kühl AAet al. . Mucosal BCG vaccination induces protective lung-resident memory T cell populations against tuberculosis. mBio. 2016;7:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Perry LL, Feilzer K, Caldwell HD.. Immunity to Chlamydia trachomatis is mediated by T helper 1 cells through IFN-gamma-dependent and -independent pathways. J Immunol. 1997;158:3344–52. [PubMed] [Google Scholar]
  66. Perry LL, Feilzer K, Hughes Set al. . Clearance of Chlamydia trachomatis from the murine genital mucosa does not require perforin-mediated cytolysis or Fas-mediated apoptosis. 1999;67:1379–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Pestka S, Krause CD, Walter MR.. Interferons, interferon-like cytokines, and their receptors. Immunol Rev. 2004;202:8–32. [DOI] [PubMed] [Google Scholar]
  68. Pizzolla A, Nguyen THO, Smith JMet al. . Resident memory CD8+ T cells in the upper respiratory tract prevent pulmonary influenza virus infection. Sci Immunol. 2017;2:eaam6970. [DOI] [PubMed] [Google Scholar]
  69. Poston TB, Qu Y, Girardi Jet al. . A Chlamydia-specific TCR-transgenic mouse demonstrates Th1 polyfunctionality with enhanced effector function. J Immunol. 2017;199:2845–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Risser WL, Risser JMH.. The incidence of pelvic inflammatory disease in untreated women infected with Chlamydia trachomatis: a structured review. Int J STD AIDS. 2007;18:727–31. [DOI] [PubMed] [Google Scholar]
  71. Roan NR, Gierahn TM, Higgins DEet al. . Monitoring the T cell response to genital tract infection. Proc Natl Acad Sci USA. 2006;103:12069–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Roan NR, Starnbach MN.. Antigen-specific CD8+ T cells respond to Chlamydia trachomatis in the genital mucosa. J Immunol. 2006;177:7974–9. [DOI] [PubMed] [Google Scholar]
  73. Rogers PR, Dubey C, Swain SL.. Qualitative changes accompany memory T cell generation: faster, more effective responses at lower doses of antigen. J Immunol. 2000;164:2338–46. [DOI] [PubMed] [Google Scholar]
  74. Roshick C, Wood H, Caldwell HDet al. . Comparison of gamma interferon-mediated antichlamydial defense mechanisms in human and mouse cells. Infect Immun. 2006;74:225–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Schenkel JM, Masopust D.. Tissue-resident memory T cells. Immunity. 2014;41:886–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Shin H, Iwasaki A.. A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature. 2012;491:463–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Shin H, Iwasaki A.. Tissue-resident memory T cells. 2013;255:165–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Shiow LR, Rosen DB, Brdičková Net al. . CD69 acts downstream of interferon-α/β to inhibit S1P 1 and lymphocyte egress from lymphoid organs. Nature. 2006;440:540–4. [DOI] [PubMed] [Google Scholar]
  79. Starnbach MN, Bevan MJ, Lampe MF.. Protective cytotoxic T lymphocytes are induced during murine infection with Chlamydia trachomatis. J Immunol. 1994;153:5183–9. [PubMed] [Google Scholar]
  80. Starnbach MN, Loomis WP, Ovendale Pet al. . An inclusion membrane protein from Chlamydia trachomatis enters the MHC class I pathway and stimulates a CD8+ T cell response. J Immunol. 2003;171:4742–9. [DOI] [PubMed] [Google Scholar]
  81. Starnbach MN. Action needed on Chlamydia vaccines. Trends Microbiol. 2018;26:639–40. [DOI] [PubMed] [Google Scholar]
  82. Stary G, Olive A, Radovic-Moreno AFet al. . A mucosal vaccine against Chlamydia trachomatis generates two waves of protective memory T cells. Science. 2015;348, DOI: 10.1126/science.aaa8205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Su H, Caldwell HD.. CD4+ T cells play a significant role in adoptive immunity to Chlamydia trachomatis infection of the mouse genital tract. Infect Immun. 1995;63:3302–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Su H, Feilzer K, Caldwell HDet al. . Chlamydia trachomatis genital tract infection of antibody-deficient gene knockout mice. Infect Immun. 1997;65:1993–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Terness P, Bauer TM, Röse Let al. . Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. J Exp Med. 2002;196:447–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Thomas SM, Garrity LF, Brandt CRet al. . Indoleamine 2,3-dioxygenase-deficient mutant host cells no longer inhibit intracellular Chlamydia spp. or toxoplasma growth. J Immunol. 1993;150:5529–34. [PubMed] [Google Scholar]
  87. Tseng CK, Rank RG.. Role of NK cells in early host response to Chlamydial genital infection. 1998;66:5867–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Veiga-Fernandes H, Walter U, Bourgeois Cet al. . Response of naïve and memory CD8+ T cells to antigen stimulation in vivo. Nat Immunol. 2000;1:47–53. [DOI] [PubMed] [Google Scholar]
  89. Vicetti Miguel RD, Harvey SAK, LaFramboise WAet al. . Human female genital tract infection by the obligate intracellular bacterium Chlamydia trachomatis elicits robust type 2 immunity. PLoS One. 2013;8:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Vlcek KR, Li W, Manam Set al. . The contribution of Chlamydia-specific CD8+ T cells to upper genital tract pathology. Immunol Cell Biol. 2016;94:208–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Wang Y, Kahane S, Cutcliffe LTet al. . Development of a transformation system for Chlamydia trachomatis: restoration of glycogen biosynthesis by acquisition of a plasmid shuttle vector. PLoS Pathog. 2011;7:e1002258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Ward ME. Chlamydial classification, development and structure. Br Med Bull. 1983;39:109–15. [DOI] [PubMed] [Google Scholar]
  93. Whitmire JK, Eam B, Whitton JL.. Tentative T cells: memory cells are quick to respond, but slow to divide. PLoS Pathog. 2008;4:e1000041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Wiesenfeld HC. Screening for Chlamydia trachomatis infections in women. N Engl J Med. 2017;376:765–73. [DOI] [PubMed] [Google Scholar]
  95. Yang X, Hayglasst KT, Brunham RC.. Different roles are played by ⍺β and γδ T cells in acquired immunity to Chlamydia trachomatis pulmonary infection. Immunology. 1998;94:469–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Yu H, Karunakaran KP, Kelly Iet al. . Immunization with live and dead Chlamydia muridarum induces different levels of protective immunity in a murine genital tract model: correlation with MHC class II peptide presentation and multifunctional Th1 cells. J Immunol. 2011;186:3615–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Zhang X, Starnbach MN.. An excess of the proinflammatory cytokines IFN-γ and IL-12 impairs the development of the memory CD8+ T cell response to Chlamydia trachomatis. J Immunol. 2015;195:1665–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Zhu J. T helper cell differentiation, heterogeneity, and plasticity. Cold Spring Harb Perspect Biol. 2018;10:1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ftab014_Supplemental_Files

Articles from Pathogens and Disease are provided here courtesy of Oxford University Press

RESOURCES