Elsevier

Cytokine

Volume 63, Issue 2, August 2013, Pages 151-165
Cytokine

Chlamydia trachomatis infection results in a modest pro-inflammatory cytokine response and a decrease in T cell chemokine secretion in human polarized endocervical epithelial cells

https://doi.org/10.1016/j.cyto.2013.04.022Get rights and content

Highlights

  • A polarized endocervical epithelial cell model was infected with Chlamydia trachomatis.

  • C. trachomatis infection failed to yield a robust pro-inflammatory cytokine response.

  • Constitutive RANTES and IP10 were decreased by C. trachomatis infection.

  • C. trachomatis infected primary epithelial cells yielded similar cytokine responses.

  • Results suggest C. trachomatis uses evasive strategies to survive in the endocervix.

Abstract

The endocervical epithelium is a major reservoir for Chlamydia trachomatis in women, and genital infections are extended in their duration. Epithelial cells act as mucosal sentinels by secreting cytokines and chemokines in response to pathogen challenge and infection. We therefore determined the signature cytokine and chemokine response of primary-like endocervix-derived epithelial cells in response to a common genital serovar (D) of C. trachomatis. For these studies, we used a recently-established polarized, immortalized, endocervical epithelial cell model (polA2EN) that maintains, in vitro, the architectural and functional characteristics of endocervical epithelial cells in vivo including the production of pro-inflammatory cytokines. PolA2EN cells were susceptible to C. trachomatis infection, and chlamydiae in these cells underwent a normal developmental cycle as determined by a one-step growth curve. IL1α protein levels were increased in both apical and basolateral secretions of C. trachomatis infected polA2EN cells, but this response did not occur until 72 h after infection. Furthermore, protein levels of the pro-inflammatory cytokines and chemokines IL6, TNFα and CXCL8 were not significantly different between C. trachomatis infected polA2EN cells and mock infected cells at any time during the chlamydial developmental cycle up to 120 h post-infection. Intriguingly, C. trachomatis infection resulted in a significant decrease in the constitutive secretion of T cell chemokines IP10 and RANTES, and this required a productive C. trachomatis infection. Examination of anti-inflammatory cytokines revealed a high constitutive apical secretion of IL1ra from polA2EN cells that was not significantly modulated by C. trachomatis infection. IL-11 was induced by C. trachomatis, although only from the basolateral membrane. These results suggest that C. trachomatis can use evasion strategies to circumvent a robust pro-inflammatory cytokine and chemokine response. These evasion strategies, together with the inherent immune repertoire of endocervical epithelial cells, may aid chlamydiae in establishing, and possibly sustaining, an intracellular niche in microenvironments of the endocervix in vivo.

Introduction

Genital serovariants (D-K) of the obligate intracellular bacterium Chlamydia trachomatis are the world’s most common sexually transmitted bacterial pathogens, accounting for an estimated 90 million new cases reported annually [1]. C. trachomatis exhibits a tropism for the columnar epithelial cells of the genital mucosae, with the endocervix being the most commonly infected site in women. In a proportion of infected women, organisms also ascend into the endometrium and Fallopian tubes where chronic infection can lead to devastating reproductive consequences, including pelvic inflammatory disease (PID), tubal infertility, and ectopic pregnancy, all of which result from immune mediated damage [1]. The reason why C. trachomatis can cause extended infections, lasting months to years in the face of an immune response [2], [3], [4], [5], [6], is not well understood, but does suggest the organism can adapt to, or evade, elements of the local host immune response.

Chlamydiae have a biphasic developmental cycle that begins when non-metabolically active, infectious, elementary bodies (EBs) encounter the apical surface of polarized epithelial cells. Following entry into the host cell, EBs escape lysosomal fusion, and endosomes containing EBs fuse to form the membrane bound vacuole termed an inclusion. EBs differentiate into metabolically active, non-infectious reticulate bodies (RBs) that undergo DNA replication and binary fission. RBs then re-differentiate into EBs that may then escape the host cell through lysis or extrusion mechanisms [7], [8].

Traditional methods for culturing C. trachomatis in vitro utilize either murine fibroblast cell lines or the ectocervix derived cervical carcinoma cell line (HeLa). Recent studies, however, have highlighted the importance of the cell type in which chlamydiae are grown, as cell lines derived from different anatomical sites yield different growth rates and infectious yields [9], [10]. Neither HeLa cells nor murine fibroblast cells accurately represent the target cells in vivo, the polarized columnar epithelial cells, with respect to morphology, expression of innate immune mediators or responsiveness to TLR agonists [11], [12]. Furthermore, innate immune signaling pathways have been altered over the years during the time that HeLa cells have been grown in vitro [13], [14]. Alterations in innate immune pathways are significant, as it has been hypothesized that the epithelial cells, generally believed to be the only sites of chlamydial replication in the female reproductive tract (FRT), are responsible for eliciting and sustaining the inflammatory immune cascade associated with chlamydial disease via the release of intracellular IL1α upon chlamydiae induced host cell lysis [15], [16]. Therefore, traditional cell lines may not be the optimal model systems in which to investigate the innate immune cascade elicited by C. trachomatis infected epithelial cells.

In recent years the orientation of the cells used to culture chlamydiae has been shown to influence chlamydial biology. Columnar epithelial cells, the target cells for chlamydial infection, maintain functionally distinct apical and basolateral membrane domains that are separated by tight junctions. Epithelial cells grown in a polarized orientation contain greater nutrient pools that are important for chlamydial growth, such as tryptophan, than their traditionally-grown submerged cell counterparts grown on plastic surfaces [17]. The use of polarized epithelial cell culture models for chlamydial studies, pioneered by Wyrick, has also revealed differences in chlamydial entry and exit mechanisms, infectious progeny, duration of the developmental cycle, infectivity, duration of the persistent growth state, reactivity to antibiotics, responsiveness to female sex steroid hormones, and innate inflammatory responses (reviewed in [18]). All of these parameters of chlamydial biology may also influence the subsequent innate epithelial immune response to the bacteria as well, although this has not yet been investigated in more primary-like genital epithelial cells.

We recently developed an epithelial cell model derived from human endocervical tissue (A2EN cells). A2EN cells polarize and appropriately express many of the functional proteins of the endocervical epithelium in vivo such as hormone receptors, mucins, anti-microbial peptides, and cytokines [11]. The aim of this study was to determine the characteristics of, and the cytokine response to, C. trachomatis infection in polarized A2EN (polA2EN) cells in order to examine the potential role of the endocervical epithelium in initiating, sustaining and amplifying a proinflammatory immune response to this organism.

Section snippets

Epithelial cell culture

The A2EN human endocervical epithelial cell line was originally generated in our laboratory from primary epithelial cells grown out from an endocervical explant and immortalized using human papilloma virus genes E6 and E7, as recently described [11], [12]. A2EN cells were grown in a phenol red-free serum-free, medium (EpiLife; Cascade Biologics) with a defined growth supplement, and seeded onto 0.4 μm transwell inserts under differentiation conditions to induce polarization, as previously

Polarized A2EN cells are susceptible to C. trachomatis infection

We recently developed a polarized, immortalized, endocervical epithelial cell model (polA2EN) that generates many innate immune mediators including mucin, anti-microbial peptides, cytokines and chemokines [11]. Since constitutively made innate immune mediators might interfere with C. trachomatis infection of polA2EN cells, the first objective of this study was to determine whether or not polA2EN cells could be infected with C. trachomatis. First, we utilized two methods to infect polA2EN cells

Acknowledgments

This study was supported by NIH Grant AI087899. The authors thank Dr. Wandy Beatty for EM imaging and Drs. Chris McGowin, Danny Schust and Priscilla Wyrick for critical review of the manuscript.

References (95)

  • A.J. Quayle

    The innate and early immune response to pathogen challenge in the female genital tract and the pivotal role of epithelial cells

    J Reprod Immunol

    (2002)
  • P.B. Kavathas et al.

    Nod1, but not the ASC inflammasome, contributes to induction of IL-1beta secretion in human trophoblasts after sensing of Chlamydia trachomatis

    Mucosal Immunol

    (2013)
  • S. Ying et al.

    Chlamydia trachomatis can protect host cells against apoptosis in the absence of cellular inhibitor of apoptosis proteins and Mcl-1

    Microbes Infect

    (2008)
  • C. Fehlner-Gardiner et al.

    Molecular basis defining human Chlamydia trachomatis tissue tropism. A possible role for tryptophan synthase

    J Biol Chem

    (2002)
  • G. McClarty et al.

    Chlamydial interferon gamma immune evasion influences infection tropism

    Curr Opin Microbiol

    (2007)
  • L. Koehler et al.

    Ultrastructural and molecular analyses of the persistence of Chlamydia trachomatis (serovar K) in human monocytes

    Microb Pathog

    (1997)
  • N.V. Guseva et al.

    Characterization of estrogen-responsive epithelial cell lines and their infectivity by genital Chlamydia trachomatis

    Microbes Infect

    (2005)
  • A.S. Maslow et al.

    Estrogen enhances attachment of Chlamydia trachomatis to human endometrial epithelial cells in vitro

    Am J Obstet Gynecol

    (1988)
  • S.A. Robertson et al.

    Transforming growth factor beta–a mediator of immune deviation in seminal plasma

    J Reprod Immunol

    (2002)
  • R.C. Brunham et al.

    Immunology of Chlamydia infection: implications for a Chlamydia trachomatis vaccine

    Nat Rev Immunol

    (2005)
  • M.R. Golden et al.

    Duration of untreated genital infections with Chlamydia trachomatis: a review of the literature

    Sex Transm Dis

    (2000)
  • J.L. Joyner et al.

    Persistence of Chlamydia trachomatis infection detected by polymerase chain reaction in untreated patients

    Sex Transm Dis

    (2002)
  • M. Molano et al.

    The natural course of Chlamydia trachomatis infection in asymptomatic Colombian women: a 5-year follow-up study

    J Infect Dis

    (2005)
  • S.A. Morre et al.

    The natural course of asymptomatic Chlamydia trachomatis infections: 45% clearance and no development of clinical PID after one-year follow-up

    Int J STD AIDS

    (2002)
  • K.S. Parks et al.

    Spontaneous clearance of Chlamydia trachomatis infection in untreated patients

    Sex Transm Dis

    (1997)
  • K. Hybiske et al.

    Mechanisms of host cell exit by the intracellular bacterium Chlamydia

    Proc Natl Acad Sci USA

    (2007)
  • J.W. Moulder

    Interaction of chlamydiae and host cells in vitro

    Microbiol Rev

    (1991)
  • M.M. Herbst-Kralovetz et al.

    Quantification and comparison of toll-like receptor expression and responsiveness in primary and immortalized human female lower genital tract epithelia

    Am J Reprod Immunol

    (2008)
  • A. Nair et al.

    NF-kappaB is constitutively activated in high-grade squamous intraepithelial lesions and squamous cell carcinomas of the human uterine cervix

    Oncogene

    (2003)
  • Landry J, Pyl PT, Rausch T, Tekkedil MM, Stuetz AM, Jauch A, Aiyar RS, Pau G, Delhomme N, Gagneur J, Korbel JO, Huber...
  • S.J. Rasmussen et al.

    Secretion of proinflammatory cytokines by epithelial cells in response to Chlamydia infection suggests a central role for epithelial cells in chlamydial pathogenesis

    J Clin Invest

    (1997)
  • C.D. Kane et al.

    Intracellular tryptophan pool sizes may account for differences in gamma interferon-mediated inhibition and persistence of chlamydial growth in polarized and nonpolarized cells

    Infect Immun

    (1999)
  • Wyrick P. Polarized epithelial cell culture for Chlamydia trachomatis. In: Wyrick P, Bavoil P, editors. Chlamydia...
  • R.J. Belland et al.

    Transcriptome analysis of chlamydial growth during IFN-gamma-mediated persistence and reactivation

    Proc Natl Acad Sci USA

    (2003)
  • W.L. Beatty

    Trafficking from CD63-positive late endocytic multivesicular bodies is essential for intracellular development of Chlamydia trachomatis

    J Cell Sci

    (2006)
  • K.P. Tremellen et al.

    Seminal transforming growth factor beta1 stimulates granulocyte-macrophage colony-stimulating factor production and inflammatory cell recruitment in the murine uterus

    Biol Reprod

    (1998)
  • E. de la Torre et al.

    Chlamydia trachomatis infection modulates trophoblast cytokine/chemokine production

    J Immunol

    (2009)
  • K.R. Buchholz et al.

    The extracellular signal-regulated kinase/mitogen-activated protein kinase pathway induces the inflammatory factor interleukin-8 following Chlamydia trachomatis infection

    Infect Immun

    (2007)
  • P. Mpiga et al.

    Sustained interleukin-6 and interleukin-8 expression following infection with Chlamydia trachomatis serovar L2 in a HeLa/THP-1 cell co-culture model

    Scand J Immunol

    (2006)
  • S. Dessus-Babus et al.

    Differences in innate immune responses (in vitro) to HeLa cells infected with nondisseminating serovar E and disseminating serovar L2 of Chlamydia trachomatis

    Infect Immun

    (2002)
  • J.V. Fahey et al.

    Secretion of cytokines and chemokines by polarized human epithelial cells from the female reproductive tract

    Hum Reprod

    (2005)
  • R.N. Fichorova et al.

    Differential expression of immunobiological mediators by immortalized human cervical and vaginal epithelial cells

    Biol Reprod

    (1999)
  • L. Eckmann et al.

    Epithelial cells secrete the chemokine interleukin-8 in response to bacterial entry

    Infect Immun

    (1993)
  • W.L. Beatty et al.

    Morphologic and antigenic characterization of interferon gamma-mediated persistent Chlamydia trachomatis infection in vitro

    Proc Natl Acad Sci USA

    (1993)
  • J.A. Ibana et al.

    Chlamydia trachomatis immune evasion via downregulation of MHC class I surface expression involves direct and indirect mechanisms

    Infect Dis Obstet Gynecol

    (2011)
  • S.P. Lad et al.

    Cleavage of p65/RelA of the NF-kappaB pathway by Chlamydia

    Proc Natl Acad Sci USA

    (2007)
  • G. Le Negrate et al.

    ChlaDub1 of Chlamydia trachomatis suppresses NF-kappaB activation and inhibits IkappaBalpha ubiquitination and degradation

    Cell Microbiol

    (2008)
  • Cited by (37)

    • Mucus secretions from a conditionally reprogrammed primary endocervical cell culture

      2022, F and S Science
      Citation Excerpt :

      These applications could extend to immune studies as well given the high quantity of immune proteins found in CREC secretions. Previous in vitro studies examining immune responses to common pathogens could be repeated in CRECs to measure the variability of inflammatory response (20, 21). Finally, one of the key benefits of using conditionally reprogrammed cultures is that they maintain genetic similarity to parent cells even after 30+ doublings, suggesting that in vitro experiments with CRECs would faithfully recapitulate their in vivo variations (8).

    • Human genetic diversity regulating the TLR10/TLR1/TLR6 locus confers increased cytokines in response to Chlamydia trachomatis

      2022, Human Genetics and Genomics Advances
      Citation Excerpt :

      Because a fine-tuned balance of cytokine production is necessary for effective pathogen clearance without excessive inflammation and damage to host tissues, levels of cytokines are a particular aspect of host response that may be crucial to susceptibility and severity. C. trachomatis induces the production of pro-inflammatory cytokines including IL-1β, IL-6, IL-8, IFNγ, and TNFα.42–45 While these cytokines help eradicate infection, a prolonged cytokine response may promote tissue damage.42

    • 5-HT<inf>2</inf> receptor activation alleviates airway inflammation and structural remodeling in a chronic mouse asthma model

      2019, Life Sciences
      Citation Excerpt :

      BALF from treatment groups was harvested in 1 ml of PBS containing 2% BSA (Boston BioProducts, Ashland, MA). 50 μl of cell-free BALF was analyzed by cytometric bead array for pro-inflammatory cytokines and chemokines using the Milliplex cytometric bead array kit (Millipore Sigma, Burlington MA Cat # MCYTOMAG-70K-PMX) according to the manufacturer's instructions [38]. The samples were run on a BioRad Bio-Plex 200 system and data was analyzed relative to a 5-parameter logistical standard curve for each corresponding cytokine/chemokine using Bio-Plex Manager 6.1.1.

    • Embryotoxic cytokines—Potential roles in embryo loss and fetal programming

      2018, Journal of Reproductive Immunology
      Citation Excerpt :

      Reproductive tract cytokine patterns are susceptible to interference from the pro-inflammatory effects of infection or dysbiosis. Chlamydia bind to Toll-like receptors (TLRs) expressed by epithelial cells in the rat and human uterus and oviduct, and alter the cytokine synthesis profile, notably inducing elevated TNF and IL1RA (Rasmussen et al., 1997; Kaushic et al., 2000; Buckner et al., 2013). Meta-analysis of inflammatory cytokine responses in various reproductive tract infections identifies known embryotoxic cytokines as induced locally and in peripheral blood by infection.

    View all citing articles on Scopus
    View full text