Article

Chlamydial infection of polarized HeLa cells induces PMN chemotaxis but the cytokine profile varies between disseminating and non-disseminating strains

Wiley
Cellular Microbiology
Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

While genital infections caused by Chlamydia trachomatis are generally asymptomatic, the density and pattern of inflammation varies considerably. The purpose of this study was to try to dissect the signalling in chlamydiae-infected epithelial cells that triggers innate responses and regulates polymorphonuclear neutrophil (PMN) chemotaxis. Polarized endocervical epithelial HeLa cells, grown in commercial inserts, were inoculated either with the non-disseminating (luminal) serovar E or the disseminating serovar L2. At 12-48 h after infection, the chambers were used in a quantitative chemotaxis assay, and cytokine production by infected cells was examined using cDNA microarray technology and confirmed by enzyme-linked immunosorbent assay (ELISA). Infection of HeLa cells with C. trachomatis E or L2 induced a strong and similar PMN chemotactic response, but larger amounts of interleukin (IL)-8 and IL-11 were released after infection with serovar L2. IL-6 was also produced in modest amounts after infection with either strain, but no IL-1alpha or tumour necrosis factor (TNF)-alpha was detected in any of the culture supernatants tested. IL-11 did not appear to influence the PMN response to chlamydial infection, but secretion of large amounts of this anti-inflammatory cytokine, mainly active on macrophages, in the very early stages of the infection may allow C. trachomatis to escape some innate defences to establish infection.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... This was in accordance with previous results of cytokine regulation under wIRA/VIS irradiation ( Marti et al., 2014). Dessus- Babus et al. (2000) and Leonard et al. (2017) reported increased IL-6 secretion after chlamydial infection under CHX-influence, which we could confirm after comparing CHX-free and CHX-incubated conditions by t-test (data not shown). IL-8 secretion, however, was previously shown to decrease after CHX-treatment of Chlamydia-infected host cells (Dessus- Babus et al., 2000), which was not confirmed in our study. ...
... Dessus- Babus et al. (2000) and Leonard et al. (2017) reported increased IL-6 secretion after chlamydial infection under CHX-influence, which we could confirm after comparing CHX-free and CHX-incubated conditions by t-test (data not shown). IL-8 secretion, however, was previously shown to decrease after CHX-treatment of Chlamydia-infected host cells (Dessus- Babus et al., 2000), which was not confirmed in our study. or Maraviroc (C,D) supplementation and irradiation, monolayers were either fixed with methanol for direct IFA analysis (B,D) or sampled for titration by sub-passage to determine chlamydial infectivity (A,C). ...
... In this study, we did not report significantly increased levels of IL-6, IL-8, or RANTES upon chlamydial infection (alone or in combination with wIRA/VIS irradiation; Figure 3). The induction of IL-6 after C. trachomatis infection (in CHX-free conditions) has been observed by other authors ( Rasmussen et al., 1997;Dessus-Babus et al., 2000;Gervassi et al., 2004;Cheng et al., 2008;Buckner et al., 2013). Similar results were obtained in multiple studies regarding IL-8 secretion ( Rasmussen et al., 1997;Gervassi et al., 2004;Buchholz and Stephens, 2006;Cheng et al., 2008;Buckner et al., 2013). ...
Article
Full-text available
Chlamydia trachomatis is the major cause of infectious blindness and represents the most common bacterial sexually transmitted infection worldwide. Considering the potential side effects of antibiotic therapy and increasing threat of antibiotic resistance, alternative therapeutic strategies are needed. Previous studies showed that water filtered infrared A alone (wIRA) or in combination with visible light (wIRA/VIS) reduced C. trachomatis infectivity. Furthermore, wIRA/VIS irradiation led to secretion of pro-inflammatory cytokines similar to that observed upon C. trachomatis infection. We confirmed the results of previous studies, namely that cytokine secretion (IL-6, IL-8, and RANTES/CCL5) upon wIRA/VIS treatment, and the subsequent reduction of chlamydial infectivity, are independent of the addition of cycloheximide, a host protein synthesis inhibitor. Reproducible cytokine release upon irradiation indicated that cytokines might be involved in the anti-chlamydial mechanism of wIRA/VIS. This hypothesis was tested by inhibiting IL-6, IL-8, and RANTES secretion in C. trachomatis or mock-infected cells by gene silencing or pharmaceutical inhibition. Celastrol, a substance derived from Trypterygium wilfordii, used in traditional Chinese medicine and known for anti-cancer and anti-inflammatory effects, was used for IL-6 and IL-8 inhibition, while Maraviroc, a competitive CCR5 antagonist and anti-HIV drug, served as a RANTES/CCL5 inhibitor. HeLa cell cytotoxicity and impact on chlamydial morphology, size and inclusion number was evaluated upon increasing inhibitor concentration, and concentrations of 0.1 and 1 μM Celastrol and 10 and 20 μM Maraviroc were subsequently selected for irradiation experiments. Celastrol at any concentration reduced chlamydial infectivity, an effect only observed for 20 μM Maraviroc. Triple dose irradiation (24, 36, 40 hpi) significantly reduced chlamydial infectivity regardless of IL-6, IL-8, or RANTES/CCL5 gene silencing, Celastrol or Maraviroc treatment. Neither gene silencing nor pharmaceutical cytokine inhibition provoked the chlamydial stress response. The anti-chlamydial effect of wIRA/VIS is independent of cytokine inhibition under all conditions evaluated. Thus, factors other than host cell cytokines must be involved in the working mechanism of wIRA/VIS. This study gives a first insight into the working mechanism of wIRA/VIS in relation to an integral component of the host immune system and supports the potential of wIRA/VIS as a promising new tool for treatment in trachoma.
... Vermehrte Expression durch C. pneumoniae Zytokine/ Wachstumsfaktoren Epithelzellen IL-1α, IL-6, IL-8, IL-11, Gro-α, GM-CSF [15,16] IL-8 [17] Endothelzellen IFN-γ, IL-1β, IL-6, IL-8, MCP1, TNF-α [18][19][20] Glatte Muskelzellen bFGF, IFN-β, IL-6 [20,21] Synoviozyten (fibroblastenartig) IFN-β, IL-6, TGF-β, GM-CSF [22,23] Makrophagen/ Monozyten/ PBMCs IL-1α, IL-1β, IL-8 [24,25] IFN-α, IL-1β, IL-6, IL-8, IL-10, MCP1, MIP-1α, TNF-α [18,[26][27][28][29][30] Adhäsionsmoleküle Epithelzellen ICAM-1 [17] Endothelzellen ICAM-1, VCAM-1, MAdCAM-1 [31] E-Selektin, ICAM-1, VCAM-1 [32,33] Makrophagen/ Monozyten ICAM-1 [34] Tabelle 2: Publizierte Chlamydien-induzierte Veränderungen der Expression von Adhäsionsmolekülen sowie der Sekretion von Zytokinen und Wachstumsfaktoren bei verschiedenen Wirtszellen ...
... Für die Effekte von C. trachomatis auf Epithelzellen gibt es einige Veröffentlichungen, die schon in Kapitel 1.4.1 erwähnt wurde. So konnte der Anstieg von IL-6, IL-8 und IL-11 ge-zeigt werden [15,16], ebenso wie ein Anstieg der Expression von IL-8, PTGS2 und ICAM1 ...
... In primary uterine cells, the basal expression of these cytokines was much closer to the uninfected nonpolarized cells than to the polarized cells (Fahey et al., 2005). This observation is counterintuitive because embedding in an extracellular matrix (ECM) gel should provide a more physiological setup shown by Dessus-Babus et al. (2000). This difference might be explained by the fact that the cancerous cell line used by Dessus-Babus et al. (2000), HeLa 229, is a subclone of HeLa (CCL2) used by Rasmussen et al. (1997). ...
... This observation is counterintuitive because embedding in an extracellular matrix (ECM) gel should provide a more physiological setup shown by Dessus-Babus et al. (2000). This difference might be explained by the fact that the cancerous cell line used by Dessus-Babus et al. (2000), HeLa 229, is a subclone of HeLa (CCL2) used by Rasmussen et al. (1997). ...
Article
Pathogenicity of Chlamydia and Chlamydia-related bacteria could be partially mediated by an enhanced activation of the innate immune response. The study of this host pathogen interaction has proved challenging due to the restricted in vitro growth of these strict intracellular bacteria and the lack of genetic tools to manipulate their genomes. Despite these difficulties, the interactions of Chlamydiales with the innate immune cells and their effectors have been studied thoroughly. This review aims to point out the role of pattern recognition receptors and signal molecules (cytokines, reactive oxygen species) of the innate immune response in the pathogenesis of chlamydial infection. Besides inducing clearance of the bacteria, some of these effectors may be used by the Chlamydia to establish chronic infections or to spread. Thus, the induced innate immune response seems to be variable depending on the species and/or the serovar, making the pattern more complex. It remains crucial to determine the common players of the innate immune response in order to help define new treatment strategies and to develop effective vaccines. The excellent growth in phagocytic cells of some Chlamydia-related organisms such as Waddlia chondrophila supports their use as model organisms to study conserved features important for interactions between the innate immunity and Chlamydia.
... However, the role of the primarily infected host cells in initiating this pathologic process has not been well defined. Previous studies examining host responses have focused on cytokines and have indicated that cytokine expression is an important part of the host reaction [3][4][5]. To expand the scope of host gene responses examined, a recent study by Hess et al. [6] that used 1176 microarray-screened genes found altered gene expression of cytokines, transcription factors, and antiapoptotic genes. ...
... Cytokines, including IL-1b, IL-6, and IL-16, as well as interferon (IFN)-induced proteins and IFN regulatory factors, were up-regulated. Previous studies have documented that chlamydial infection induces host expression of IL-1b and IL-6 [3][4][5][6]25]. However, in addition to IL-1 and IL-6, a modest up-regulation of IL-16 from the chlamydia-infected host was observed in our study and has not been reported. ...
Article
To study the responses of the host cell to chlamydial infection, differentially transcribed genes of the host cells were examined. Complementary DNA (cDNA) probes were made from messenger RNAs of HeLa cells infected with Chlamydia trachomatis and were hybridized to a high-density human DNA microarray of 15,000 genes and expressed sequence tags. C. trachomatis alters host cell transcription at both the early and middle phases of its developmental cycle. At 2 h after infection, 13 host genes showed mean expression ratios ⩾2-fold. At 16 h after infection, 130 genes were differentially transcribed. These genes encoded factors inhibiting apoptosis and factors regulating cell differentiation, components of the cytoskeleton, transcription factors, and proinflammatory cytokines. This indicates that chlamydial infection, despite its intravacuolar location, alters the transcription of a broad range of host genes in diverse cellular pathways and provides a framework for future studies
... Infection with C. trachomatis has been shown to induce inflammatory cytokines both in vitro and in vivo (Magee et al ., 1992;Rasmussen et al ., 1997;Dessus-Babus et al ., 2000). Epithelial cells are a primary site for C. trachomatis infection in vivo . ...
... IL-8 is an inflammatory chemokine associated with immune-mediated tissue damage and pathology that recruits and activates neutrophils and other immune cells (Mukaida et al., 1998a). IL-8 is produced by both Chlamydia-infected HeLa cells, as well as during infection of primary endocervical epithelial cells (Rasmussen et al., 1997;Dessus-Babus et al., 2000). The mechanism of IL-8 activation during C. trachomatis infection is not known, and remains (Mukaida et al., 1998b;Sansonetti et al., 1999). ...
Article
Diseases associated with Chlamydia are caused by inflammation-associated tissue damage following repeated or chronic infection; however, the mechanism by which the inflammatory response is induced is unknown. The inflammatory cytokine interleukin-8 (IL-8) is produced by C. trachomatis-infected epithelial cells in a bacterial growth-dependent manner. We hypothesized that IL-8 is induced through activation of host signalling pathways within Chlamydia-infected cells. Bacterial protein synthesis occurring after 15 h post infection (hpi) was required for the induction of IL-8, thus, increases in IL-8 mRNA are due to chlamydial growth or a bacterial product produced at 15 hpi. The induction of IL-8 was not dependent on soluble factors in the supernatant of C. trachomatis-infected cells and therefore was associated with an internal cellular signal. The AP-1, NFIL6 (C/EBPbeta) and NFkappaB transcriptional regulatory sites of the IL-8 promoter and the host NFkappaB signalling pathway were necessary for IL-8 induction by C. trachomatis. We conclude that a C. trachomatis growth-dependent factor produced at mid-developmental stage induces IL-8 within the epithelial cell it infects through activation of host signalling pathways.
... Cytokines produced locally by Chlamydia-infected epithelial cells are postulated to be essential for the progression of the inflammatory response that leads to pathology and disease (22). Inflammatory cytokines are induced by Chlamydia trachomatis infection in vitro and in vivo (3,16,20) and include IL-8, a chemotactic attractant and activator of neutrophils that is associated with inflammation-mediated tissue damage (18). The markedly late induction of IL-8 (at 15 h postinfection [hpi]) and its dependence on bacterial growth and protein synthesis implicate direct intracellular interactions of bacteria and the host cell (1). ...
... Rasmussen et al. (20) found that IL-8 was stimulated by C. trachomatis L2 in various cell types, including primary endocervical cells (20). Various species and serovars of Chlamydia have been found to induce IL-8, including C. trachomatis serovars D, E, and I and C. psittaci (3,20). Similar to the response in HeLa cells, activation of the ERK signaling pathway occurs in HeP2 cells (this study) and primary endometrial cells (23). ...
Article
Full-text available
Diseases associated with Chlamydia infection, such as pelvic inflammatory disease and ectopic pregnancy, are due to inflammation-mediated tissue damage and scarring that occur after chronic or repeated infections. The inflammatory chemokine interleukin-8 (IL-8) is produced by Chlamydia-infected cells through an endogenous mechanism of activation, independent of soluble factors in the supernatant. The host signaling pathways necessary for this response are not understood, but the mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase (ERK) has been shown to be activated at similar times as IL-8 mRNA up-regulation. The purpose of this study was to elucidate the MAPK pathways necessary to induce the endogenous IL-8 response to Chlamydia trachomatis infection of epithelial cells. IL-8 induced by infection with C. trachomatis L2 was shown to be dependent on ERK and independent of p38 and Jun N-terminal MAPK by use of chemical inhibitors of the signaling pathways. Persistent ERK activation during IL-8 mRNA production at 24 h postinfection was necessary to maintain the response. C. trachomatis serovar D also induced IL-8 in an ERK-dependent manner. We concluded that IL-8 induced during infection of epithelial cells is dependent on continual activation of ERK by C. trachomatis.
... Others have detected significantly higher levels of IL-11 in C. trachomatis-infected cultures of polarized HeLa cells. Of note, the level of IL-11 was significantly higher in infection involving a disseminating serovar relative to a non-disseminating variant [45]. The immunosuppressive role of IL-11 allows the bacteria to escape from host innate defenses for better dissemination. ...
Article
Full-text available
Background: Burkholderia pseudomallei, the causative agent of melioidosis poses a serious threat to humankind. B. pseudomallei secretes numerous virulence proteins that alter host cell functions to escape from intracellular immune sensors. However, the events underlying disease pathogenesis are poorly understood. Methods: We determined the ability of B. pseudomallei to invade and survive intracellularly in A549 human lung epithelial cells, and also investigated the early transcriptional responses using an Illumina® HumanHT-12 v4 microarray platform, after three hours of exposure to live B. pseudomallei (BCMS) and its secreted proteins (CCMS). Results: We found that the ability of B. pseudomallei to invade and survive intracellularly correlated with increase of multiplicity of infection and duration of contact. Activation of host carbohydrate metabolism and apoptosis as well as suppression of amino acid metabolism and innate immune responses both by live bacteria and its secreted proteins were evident. These early events might be linked to initial activation of host genes directed towards bacterial dissemination from lungs to target organs (via proposed in vivo mechanisms) or to escape potential sensing by macrophages. Conclusion: Understanding the early responses of A549 cells toward B. pseudomallei infection provide preliminary insights into the likely pathogenesis mechanisms underlying melioidosis, and could contribute to development of novel intervention strategies to combat B. pseudomallei infections.
... Subsequent studies argued against a passive epithelial model. Chlamydia-Infected epithelial cells were shown to produce chemokines and inflammatory mediators likely responsible for recruiting innate and adaptive effector cells, and possibly contributing directly to immunopathology (Rasmussen et al. 1997;Wyrick et al. 1999;Dessus-Babus, Knight and Wyrick 2000;Hess et al. 2001;Xia et al. 2003;Johnson 2004). Our lab demonstrated that Chlamydia-specific CD4 T cells directly recognized infected syngeneic upper reproductive tract epithelial cells in an IFN-γ -and CD4-dependent fashion, and showed a correlation between CD4 T cell clones relative ability to be activated by infected epithelial cells and their ability to terminate Chlamydia replication in them (Jayarapu et al. 2009). ...
Article
Chlamydia trachomatis urogenital serovars are intracellular bacteria that parasitize human reproductive tract epithelium. As the principal cell type supporting bacterial replication, epithelial cells are central to Chlamydia immunobiology initially as sentries and innate defenders, and subsequently as collaborators in adaptive immunity-mediated bacterial clearance. In asymptomatic individuals who do not seek medical care a decisive struggle between C. trachomatis and host defenses occurs at the epithelial interface. For this study we modeled the immunobiology of epithelial cells and macrophages lining healthy genital mucosa and inflamed/infected mucosa during the transition from innate to adaptive immunity. Upper reproductive tract epithelial cell line responses were compared to bone marrow-derived macrophages utilizing gene expression microarray technology. Those comparisons showed minor differences in the intrinsic innate defenses of macrophages and epithelial cells. Major lineage-specific differences in immunobiology relate to epithelial collaboration with adaptive immunity including an epithelial requirement for inflammatory cytokines to express MHC class II molecules, and a paucity and imbalance between costimulatory and coinhibitory ligands on epithelial cells that potentially limits sterilizing immunity (replication termination) to Chlamydia-specific T cells activated with limited or unconventional second signals.
... TRAF6 then activates various proteins that ultimately lead to the phosphorylation of inhibitor of kappa B alpha (I-kBa), which subsequently undergoes degradation via ubiquitination. The degradation of I-kB releases the activated nuclear factor-kB (NF-kB), which allows it to translocate into the nucleus and stimulate the expression of pro-inflammatory components, such as interleukin (IL)-8, IL-6, IL-18, IL-1a and granulocyte-macrophage colony-stimulating factor (GM-CSF) that recruit and activate various immune cells [14][15][16][17]. ...
Article
Full-text available
The immune system eliminates Chlamydia trachomatis infection through inflammation. However, uncontrolled inflammation can enhance pathology. In mice, TNF-related apoptosis-inducing ligand receptor (TRAIL-R), known for its effects on apoptosis, also regulates inflammation. In humans, the four homologues of TRAIL-R had never been investigated for effects on inflammation. Here, we examined whether TRAIL-R regulates inflammation during chlamydial infection. We examined TRAIL-R1 single nucleotide polymorphisms (SNPs) in an Ecuadorian cohort with and without C. trachomatis infections. There was a highly significant association for the TRAIL+626 homozygous mutant GG for infection vs no infection in this population. To confirm the results observed in the human population, primary lung fibroblasts and bone marrow-derived macrophages (BMDMs) were isolated from wildtype (WT) and TRAIL-R-deficient mice, and TRAIL-R1 levels in human cervical epithelial cells were depleted by RNA interference. Infection of BMDMs and primary lung fibroblasts with C. trachomatis strain L2, or the murine pathogen C. muridarum, led to higher levels of MIP2 mRNA expression or IL-1β secretion from TRAIL-R-deficient cells than WT cells. Similarly, depletion of TRAIL-R1 expression in human epithelial cells resulted in a higher level of IL-8 mRNA expression and protein secretion during C. trachomatis infection. We conclude that human TRAIL-R1 SNPs and murine TRAIL-R modulate the innate immune response against chlamydial infection. This is the first evidence that human TRAIL-R1 is a negative regulator of inflammation and plays a role in modulating Chlamydia pathogenesis.
... In women, C. trachomatis primarily infects the endocervix (Brunham and Rey-Ladino, 2005), which is also a portal of entry for HIV/SIV (Li et al., 2009; Haase, 2005). C. trachomatis induces robust production of proinflammatory cytokines including IL-8, IL-6, and IL-1 in HeLa cells (an adenocarcinoma cell line from the cervix), but only a very modest induction in endocervical (A2EN cell line) and endometrial epithelial cells (HEC-1B adenocarcinoma cell line) (Rasmussen et al., 1997; Dessus-Babus et al., 2000; Buckner et al., 2011a). In C. trachomatis-positive, infertile women (aged 20–38), the levels of IFN-, TNF-, IL-10, and IL-12 are up-regulated in cervical secretions (Reddy et al., 2004). ...
Article
Full-text available
Chlamydia trachomatis infection is one of the most prevalent bacterial STIs in the USA and worldwide, and women with C. trachomatis infection are at increased risk of acquiring HIV. Because immune activation at the genital mucosa facilitates HIV/SIV infection, C. trachomatis-mediated cytokine induction may contribute to increased HIV transmission in asymptomatic women. To begin to elucidate the mechanisms, we longitudinally analyzed profiles of innate immune factors and HIV infectivity in genital secretions from anatomically specific sites in asymptomatic women during C. trachomatis infection and post-antibiotic treatment. We found higher levels of cytokines and chemokines in endocervical secretions than vaginal secretions. Compared with the convalescent state, G-CSF, IL-1α, and RANTES were elevated in endocervical secretions, IFN-γ and TNF-α were elevated in vaginal secretions, and IFNγ, IL-1β, and MIP1-α were elevated in cervicolavage fluid (CVL), before adjustment of multiple comparisons. Elevated endocervical levels of IP-10 and MCP-1 were associated with the use of hormonal contraception in infected women after successful treatment, suggesting the role of hormonal contraception in inflammation independent of STIs. Importantly, soluble factors found in endocervical secretions during infection enhanced HIV infectivity while no difference in HIV infectivity was found with vaginal secretions or CVL during infection or at convalescence. Taken together, the profiles of immune mediators and in vitro HIV infectivity indicate that the endocervical and vaginal mucosa are immunologically distinct. Our results underscore the importance of considering anatomical site and local sampling methodology when measuring mucosal responses, particularly in the presence of C. trachomatis infection.
... Chlamydia blocks degradation of the NF-kB retention factor IkBa and nuclear translocation Kaukoranta-Tolvanen et al. 1996;Rasmussen et al. 1997;Dessus-Babus et al. 2000;Geng et al. 2000;Netea et al. 2004;Rothfuchs et al. 2004;Rank et al. 2010. ...
Article
Full-text available
Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen and the causative agent of blinding trachoma. Although Chlamydia is protected from humoral immune responses by residing within remodeled intracellular vacuoles, it still must contend with multilayered intracellular innate immune defenses deployed by its host while scavenging for nutrients. Here we provide an overview of Chlamydia biology and highlight recent findings detailing how this vacuole-bound pathogen manipulates host-cellular functions to invade host cells and maintain a replicative niche.
... This idea also applies to the role of cPLA2 in intra-Golgi trafficking in HeLa cells (San Pietro et al., 2009). Although HeLa cells cultured in monolayer are nonpolarized and do not display tight junctions , these epithelial carcinoma cells do form tight junctions and polarize under specific growth conditions (Shimojo et al., 1995; Dessus-Babus et al., 2000). Because these cells have retained the capacity to polarize under specific circumstances , it is possible that they possess polarity-related modes of transport—such as cPLA2-driven trafficking— that are effective even in a nonpolarized cellular context. ...
Article
Full-text available
In endothelial cells specifically, cPLA2alpha translocates from the cytoplasm to the Golgi complex in response to cell confluence. Considering the link between confluence and cell-cell junction formation, and the emerging role of cPLA2alpha in intracellular trafficking, we tested whether Golgi-associated cPLA2alpha is involved in the trafficking of junction proteins. Here, we show that the redistribution of cPLA2alpha from the cytoplasm to the Golgi correlates with adherens junction maturation and occurs before tight junction formation. Disruption of adherens junctions using a blocking anti-VE-cadherin antibody reverses the association of cPLA2alpha with the Golgi. Silencing of cPLA2alpha and inhibition of cPLA2alpha enzymatic activity using various inhibitors result in the diminished presence of the transmembrane junction proteins VE-cadherin, occludin, and claudin-5 at cell-cell contacts, and in their accumulation at the Golgi. Altogether, our data support the idea that VE-cadherin triggers the relocation of cPLA2alpha to the Golgi and that in turn, Golgi-associated cPLA2alpha regulates the transport of transmembrane junction proteins through or from the Golgi, thereby controlling the integrity of endothelial cell-cell junctions.
... This study is a first towards the development of a molecular fingerprint (diagnostic) for brain immunopathogenesis associated with malaria. In this regard, recent studies with infectious agents such as Salmonella, Chlamydia and Trypanosoma using cDNA microarray technology have revealed unique gene-expression profiles [24][25][26] which may be of diagnostic value. ...
Article
Full-text available
Abstract Background Malaria afflicts 300–500 million people causing over 1 million deaths globally per year. The immunopathogenesis of malaria is mediated partly by co mplex cellular and immunomodulator interactions involving co-regulators such as cytokines and adhesion molecules. However, the role of chemokines and their receptors in malaria immunopathology remains unclear. RANTES (Regulated on Activation Normal T-Cell Expressed and Secreted) is a chemokine involved in the generation of inflammatory infiltrates. Recent studies indicate that the degradation of cell-cell junctions, blood-brain barrier dysfunction, recruitment of leukocytes and Plasmodium -infected erythrocytes into and occlusion of microvessels relevant to malaria pathogenesis are associated with RANTES expression. Additionally, activated lymphocytes, platelets and endothelial cells release large quantities of RANTES, thus suggesting a unique role for RANTES in the generation and maintenance of the malaria-induced inflammatory response. The hypothesis of this study is that RANTES and its corresponding receptors (CCR1, CCR3 and CCR5) modulate malaria immunopathogenesis. A murine malaria model was utilized to evaluate the role of this chemokine and its receptors in malaria. Methods The alterations in immunomodulator gene expression in brains of Plasmodium yoelii 17XL-infected mice was analysed using cDNA microarray screening, followed by a temporal comparison of mRNA and protein expression of RANTES and its corresponding receptors by qRT-PCR and Western blot analysis, respectively. Plasma RANTES levels was determined by ELISA and ultrastructural studies of brain sections from infected and uninfected mice was conducted. Results RANTES (p < 0.002), CCR1 (p < 0.036), CCR3 (p < 0.033), and CCR5 (p < 0.026) mRNA were significantly upregulated at peak parasitaemia and remained high thereafter in the experimental mouse model. RANTES protein in the brain of infected mice was upregulated (p < 0.034) compared with controls. RANTES plasma levels were significantly upregulated; two to three fold in infected mice compared with controls (p < 0.026). Some d istal microvascular endothelium in infected cerebellum appeared degraded, but remained intact in controls. Conclusion The upregulation of RANTES, CCR1, CCR3, and CCR5 mRNA, and RANTES protein mediate inflammation and cellular degradation in the cerebellum during P. yoelii 17XL malaria.
... C. trachomatis is able to infect and multiply within a broad range of eukaryotic cells, including macrophages, smooth muscle, epithelial, and endothelial cells [5]. Chlamydial infection of epithelial cells at mucosal surface produces proinflammatory factors such as Interleukin (IL)-1α, IL-6, IL-8, IL-11, GRO-α, and granulocytemacrophage colony-stimulating factor [6][7][8], which can lead to an acute inflammatory response characterized by neutrophil infiltration to the primary sites of infection, followed by a subepithelial accumulation of mononuclear leukocytes during the chronic phase of infection [9][10][11]. These cellular responses promote cellular proliferation and tissue damage of affected organs [12]. ...
Article
Full-text available
Chlamydia trachomatis is a leading cause of sexually transmitted infection worldwide and responsible for myriad of immunopathological changes associated with reproductive health. Delayed secretion of proinflammatory chemokine interleukin (IL)-8 is a hallmark of chlamydial infection and is dependent on chlamydial growth. We examined the effect of iron chelators on IL-8 production in HeLa 229 (cervix epitheloid cell, CCL2) cells infected with C. trachomatis. IL-8 production was induced by Iron chelator DFO and Mimosine, however, synergy with chlamydial infection was obtained with DFO only. Temporal expression of proinflammatory secreted cytokines IL-1beta, TNF-alpha, and IL-8 did not show synchrony in Chlamydia trachomatis infected cells. Secretion of IL-8 from Hela cells infected with C. trachomatis was not dependent on IL-1 beta and TNF- alpha induction. These results indicate towards involvement of iron in chlamydia induced IL-8 production.
... The recent application of oligonucleotide and cDNA microarray technologies toward the study of host-pathogen interactions has permitted rapid and unbiased examination of changes in expression of a large number of genes at the transcript level (2). Microarray analysis of host cell gene expression following infection with viral (3)(4)(5)(6)(7), bacterial (8 -12), fungal (13), and protozoan (14) pathogens has revealed complex and diverse transcriptional responses to these infectious agents. Data bases generated from these, and future, studies will provide an invaluable resource for the functional characterization of host cell pathways required to facilitate pathogen survival and for the further understanding of host defense mechanisms (1,15,16). ...
Article
Full-text available
Host cell infection by the intracellular pathogen, Trypanosoma cruzi, involves activation of signaling pathways, cytoskeletal reorganization, and targeted recruitment of host cell lysosomes. To determine the consequences of T. cruzi invasion on host cell gene expression, high density microarrays consisting of approximately 27,000 human cDNAs were hybridized with fluorescent probes generated from T. cruzi-infected human fibroblasts (HFF) at early time points following infection (2-24 h). Surprisingly, no genes were induced > or =2-fold in HFF between 2 and 6 h post-infection (hpi) in repeated experiments while immediate repression of six host cell transcripts was observed. A significant increase in transcript abundance for 106 host cell genes was observed at 24 hpi. Among the most highly induced is a set of interferon-stimulated genes, indicative of a type I interferon (IFN) response to T. cruzi. In support of this, T. cruzi-infected fibroblasts begin to secrete IFNbeta at 18 hpi following the induction of IFNbeta transcripts. As compared with global transcriptional responses evoked by other intracellular pathogens, T. cruzi is a stealth parasite that elicits few changes in host cell transcription during the initiation of infection.
... In this study, we describe for the first time the induction of CTGF, ETV4, NR4A2, DUSP4, DUSP5, and GAS-1 by C. trachomatis and C. pneumoniae. We show also for the first time that C. pneumoniae induces genes whose induction has so far only been described by us and others for C. trachomatis (Dessus-Babus et al., 2000;Hess et al., 2001): EGR1 LIF, MIP-2a, IER3, MCL-1, and EPHA2. Moreover, we compared host-cell transcriptome changes induced by Chlamydia and Salmonella. ...
Article
Chlamydophila pneumoniae and Chlamydia trachomatis cause infections of the respiratory or urogenital tract. In addition, both species have been associated with atherosclerosis or reactive arthritis respectively. For these intracellular pathogens the interaction with their host-cells is of particular importance. To get insight into this relationship, we conducted a comparative analysis of the host-cell gene regulation of human epithelial cells during infection with Chlamydia. In a screening of HeLa cells by Affymetrix-microchips, numerous regulated host-genes were identified. A detailed expression profile was obtained for 14 genes by real-time RT-PCR - comparing C. pneumoniae, C. trachomatis and intracellular S. typhimurium. The transcriptional responses induced by C. pneumoniae were similar (but usually smaller) compared to C. trachomatis, some were absent. UV-inactivated bacteria induced no differential gene expression suggesting that pathomechanisms other than those associated with innate immunity play here an important role. The expression pattern induced by Salmonella differed substantially. These genus- or group-specific transcriptional response patterns elicited by viable intracellular pathogens may considerably contribute to the different pathologies encountered in the clinic.
... Despite this decrease in cytokine secretion, lower levels of pathology were not observed. Epithelial cell secretion of chemokines in response to Chlamydia infection may be the primary driving force for influx of inflammatory cells262728; thus, inhibition of infection may be required to inhibit pathology. It has been shown that attachment to HeLa cells of invasive C. trachomatis serovar L2, which is a submucosal pathogen that probably uses extracellular matrix GAG to colonize the basolateral surface of mucosal epithelial cells, is inhibited by heparin or heparin sulfate, whereas soluble GAG have no effect on attachment to HeLa cells of noninvasive C. trachomatis serovar E, which enters and exits the apical surface of columnar epithelial cells in the genital tract [29] . ...
Article
Glycosaminoglycans (GAG) efficiently inhibit adherence of several strains of Chlamydia trachomatis to cell lines in vitro, but none of the GAG have been able to inhibit infections in vivo. One possible cause for failure of GAG inhibition in vivo is the inability to deliver a sustained concentration of GAG at the mucosal surface. We tested the possibility of enhancing cell protection by increasing the cell-surface concentration of GAG using membrane-anchored GAG (MAG), composed of phosphatidylethanolamine (PE)-linked GAG. These lipid conjugates were originally designed as extracellular phospholipase A2 (PLA2) inhibitors and exhibit a dual effect: the lipid moiety incorporates into the cell membrane, interfering with the action of PLA2 on cell membranes, and the anchored GAG protects the cell membrane from exogenous inflammatory mediators. We tested the ability of MAG to block chlamydia infection in vitro and in vivo. The MAG blocked infection of epithelial cells in vitro when added to the cells at the same time or before infection, but not if added after the bacteria had already invaded the host cells. One of the MAG led to the production of aberrant Chlamydia vacuoles, suggesting it may inhibit intracellular PLA2 associated with development of the vacuole. Although the MAG did not inhibit vaginal infection of mice, they decreased significantly the level of secretion of the inflammatory cytokines TNF-alpha and IFN-gamma but had no effect on secretion of the neutrophil chemokine, macrophage inflammatory protein-2 (MIP-2). Acute and chronic inflammatory cell infiltrates were not altered by MAG treatment. These findings suggest that lipid conjugation of GAG could be used as a novel approach for increasing cell-surface concentrations of GAG. The inconclusive in vivo results might be due to the physical properties of the tested MAG or an insufficient application protocol, and their improvement might provide the desired inhibitory effects.
... Of the 18, 4 were cytokines: IL-8, IL-11, leukemia inhibitory factor, and macrophage inhibitory protein 2␣ (MIP-2␣; CXCL2) (25). Similarly, analysis of C. trachomatis serovar E-infected polarized HeLa cells showed upregulation of IL-11 mRNA and secretion of IL-6, IL-8, and IL-11 (20). HeLa cells infected by LGV, urogenital serovars, and C. muridarum secreted preformed IL-18 via a posttranslational mechanism dependent on caspase-1 (39). ...
Article
Full-text available
Epithelial cells play an important role in host defense as sentinels for invading microbial pathogens. Chlamydia trachomatis is an intracellular bacterial pathogen that replicates in reproductive tract epithelium. Epithelial cells lining the reproductive tract likely play a key role in triggering inflammation and adaptive immunity during Chlamydia infections. For this report a murine oviduct epithelial cell line was derived in order to determine how epithelial cells influence innate and adaptive immune responses during Chlamydia infections. As expected, oviduct epithelial cells infected by Chlamydia muridarum produced a broad spectrum of chemokines, including CXCL16, and regulators of the acute-phase response, including interleukin-1α (IL-1α), IL-6, and tumor necrosis factor alpha. In addition, infected epithelial cells expressed cytokines that augment gamma interferon (IFN) production, including IFN-α/β and IL-12-p70. To my knowledge this is the first report of a non-myeloid/lymphoid cell type making IL-12-p70 in response to an infection. Equally interesting, infected epithelial cells significantly upregulated transforming growth factor alpha precursor expression, suggesting a mechanism by which they might play a direct role in the pathological scarring seen as a consequence of Chlamydia infections. Data from these in vitro studies predict that infected oviduct epithelium contributes significantly to host innate and adaptive defenses but may also participate in the immunopathology seen with Chlamydia infections.
... Though significant progress through this type of work can be accomplished in vivo through the administration of MMP inhibitors and application of various knockout mice, an in vitro system that replicates the interaction of the various players seen in vivo (e.g., epithelial cells, neutrophils, etc.) and allows delineation of the specific regulators involved in MMP release and activation would greatly assist in this process. Some very useful systems have been developed for the study of the transepithelial migration of human neutrophils, though this system has yet to be used for the study of MMPs (13,54). An analogous murine system is currently being developed in our laboratory to study the interactions between epithelial cells and neutrophils. ...
Article
Full-text available
The central hypothesis of this study was that matrix metalloproteinases (MMPs) would be enhanced following murine chlamydial infection and that their expression would vary in mouse strains that differ in their susceptibility to chronic chlamydia-induced disease. To address this hypothesis, female C3H/HeN and C57BL/6 mice were infected intravaginally with Chlamydia muridarum. Uterine and oviduct tissues were assessed for transcription of MMP genes and their tissue inhibitors. An increased activity of MMP genes relative to preinfection tissues was observed in the C3H/HeN mice when compared to C57BL/6 mice. Using gelatin zymography, we detected constitutive MMP-2 activity in both strains of mice but an increase in MMP-9. Casein zymography indicated the presence of two elastase-like activities consistent with MMP-12 and possibly MMP-7. Western blotting and antigen capture enzyme-linked immunoassay also confirmed an increase in MMP-9 but constitutive MMP-2 expression subsequent to the infection in both strains of mice. In C57BL/6 mice, MMP-9 was present in monomer and dimer form throughout the 56-day monitoring period. C3H/HeN mice produced dimeric MMP-9, but increases in the monomer form were also observed through day 14. Post-translational modification of MMP-9 between the two strains also differed. Immunohistochemistry revealed neutrophils as a prominent source for MMP-9 in both strains of mice. We conclude that differences in the relative expression and activity of MMPs, particularly MMP-9, occur in mice differing in their susceptibility to the development of chronic chlamydial disease. These differences may account for disparate outcomes with regard to chronic sequelae of the disease.
... This study is a first towards the development of a molecular fingerprint (diagnostic) for brain immunopathogenesis associated with malaria. In this regard, recent studies with infectious agents such as Salmonella, Chlamydia and Trypanosoma using cDNA microarray technology have revealed unique gene-expression profiles242526 which may be of diagnostic value. ...
Article
Full-text available
Malaria afflicts 300-500 million people causing over 1 million deaths globally per year. The immunopathogenesis of malaria is mediated partly by co mplex cellular and immunomodulator interactions involving co-regulators such as cytokines and adhesion molecules. However, the role of chemokines and their receptors in malaria immunopathology remains unclear. RANTES (Regulated on Activation Normal T-Cell Expressed and Secreted) is a chemokine involved in the generation of inflammatory infiltrates. Recent studies indicate that the degradation of cell-cell junctions, blood-brain barrier dysfunction, recruitment of leukocytes and Plasmodium-infected erythrocytes into and occlusion of microvessels relevant to malaria pathogenesis are associated with RANTES expression. Additionally, activated lymphocytes, platelets and endothelial cells release large quantities of RANTES, thus suggesting a unique role for RANTES in the generation and maintenance of the malaria-induced inflammatory response. The hypothesis of this study is that RANTES and its corresponding receptors (CCR1, CCR3 and CCR5) modulate malaria immunopathogenesis. A murine malaria model was utilized to evaluate the role of this chemokine and its receptors in malaria. The alterations in immunomodulator gene expression in brains of Plasmodium yoelii 17XL-infected mice was analysed using cDNA microarray screening, followed by a temporal comparison of mRNA and protein expression of RANTES and its corresponding receptors by qRT-PCR and Western blot analysis, respectively. Plasma RANTES levels was determined by ELISA and ultrastructural studies of brain sections from infected and uninfected mice was conducted. RANTES (p < 0.002), CCR1 (p < 0.036), CCR3 (p < 0.033), and CCR5 (p < 0.026) mRNA were significantly upregulated at peak parasitaemia and remained high thereafter in the experimental mouse model. RANTES protein in the brain of infected mice was upregulated (p < 0.034) compared with controls. RANTES plasma levels were significantly upregulated; two to three fold in infected mice compared with controls (p < 0.026). Some distal microvascular endothelium in infected cerebellum appeared degraded, but remained intact in controls. The upregulation of RANTES, CCR1, CCR3, and CCR5 mRNA, and RANTES protein mediate inflammation and cellular degradation in the cerebellum during P. yoelii 17XL malaria.
... For example, HSV infection suppresses dendritic cell function (Pollara et al., 2003). C. trachomatis infection increases IL-11 production, which may suppress local immune responses and aid in establishment of infection (Dessus-Babus et al., 2000). It is easy to envision how an organism that manipulates the local immune response in the genital tract might also suppress or alter the host response to other pathogens that are present at the same time. ...
Article
Epidemiological and clinical studies have shown that double infection with herpes simplex virus type 2 (HSV-2) and Chlamydia trachomatis occurs in vivo. We hypothesized that co-infection would alter replication of these agents. To test this hypothesis, HeLa cells were infected with C. trachomatis serovar E, followed 24 h later by HSV-2 strain 333. Transmission electron microscopic (TEM) analyses indicated that, by 10 h after HSV addition, reticulate bodies (RBs) in co-infected cells were swollen, aberrantly shaped and electron-lucent. In infectious titre assays, HSV-2 co-infection abrogated production of infectious chlamydial progeny. Western blot analyses indicated that accumulation of chlamydial major outer membrane protein (MOMP) was decreased by HSV co-infection while accumulation of chlamydial heat-shock protein 60-1 (HSP60-1) was increased. Polymerase chain reaction (PCR) experiments indicated that chlamydial genome copy number was unaltered by HSV-2 superinfection. Semi-quantitative, reverse transcription PCR (RT-PCR) experiments demonstrated that levels of chlamydial groEL, ftsK, ftsW, dnaA and unprocessed 16S rRNA transcripts were not changed by HSV-2 super-infection. These data indicate that HSV-2 superinfection drives chlamydia into a viable but non-cultivable state, which is the hallmark of persistence. Because chlamydial HSP60-1 has been associated with immunopathology in vivo, these results also suggest that disease severity might be increased in co-infected individuals.
... As regards chlamydial infection, infected epithelial cells have been shown to be intimately involved in the early innate response to infection (5,6,47). We investigated the role of TLR2 and TLR4 in Chlamydia-induced inflammatory cytokine gene transcription in primary murine fibroblasts and found a significant dependence on TLR2 for production of IL-1, MIP-2, and IL-6 in response to infection with MoPn. ...
Article
Full-text available
The roles of Toll-like receptor (TLR) 2 and TLR4 in the host inflammatory response to infection caused by Chlamydia trachomatis have not been elucidated. We examined production of TNF-alpha and IL-6 in wild-type TLR2 knockout (KO), and TLR4 KO murine peritoneal macrophages infected with the mouse pneumonitis strain of C. trachomatis. Furthermore, we compared the outcomes of genital tract infection in control, TLR2 KO, and TLR4 KO mice. Macrophages lacking TLR2 produced significantly less TNF-alpha and IL6 in response to active infection. In contrast, macrophages from TLR4 KO mice consistently produced higher TNF-alpha and IL-6 responses than those from normal mice on in vitro infection. Infected TLR2-deficient fibroblasts had less mRNA for IL-1, IL-6, and macrophage-inflammatory protein-2, but TLR4-deficient cells had increased mRNA levels for these cytokines compared with controls, suggesting that ligation of TLR4 by whole chlamydiae may down-modulate signaling by other TLRs. In TLR2 KO mice, although the course of genital tract infection was not different from that of controls, significantly lower levels of TNF-alpha and macrophage-inflammatory protein-2 were detected in genital tract secretions during the first week of infection, and there was a significant reduction in oviduct and mesosalpinx pathology at late time points. TLR4 KO mice responded to in vivo infection similarly to wild-type controls and developed similar pathology. TLR2 is an important mediator in the innate immune response to C. trachomatis infection and appears to play a role in both early production of inflammatory mediators and development of chronic inflammatory pathology.
... C. (16,17). The latter results are also likely a reflection of the fundamentally different physiological functions between endometrial versus endocervical epithelial cells. ...
Article
Full-text available
In vitro studies of obligate intracellular chlamydia biology and pathogenesis are highly dependent on the use of experimental models and growth conditions that mimic the mucosal architecture and environment these pathogens encounter during natural infections. In this study, the growth of Chlamydia trachomatis genital serovar E was monitored in mouse fibroblast McCoy cells and compared to more relevant host human epithelial endometrium-derived HEC-1B and cervix-derived HeLa cells, seeded and polarized on collagen-coated microcarrier beads, using a three-dimensional culture system. Microscopy analysis of these cell lines prior to infection revealed morphological differences reminiscent of their in vivo architecture. Upon infection, early chlamydial inclusion distribution was uniform in McCoy cells but patchy in both epithelial cell lines. Although no difference in chlamydial attachment to or entry into the two genital epithelial cell lines was noted, active bacterial genome replication and transcription, as well as initial transformation of elementary bodies to reticulate bodies, were detected earlier in HEC-1B than in HeLa cells, suggesting a faster growth, which led to higher progeny counts and titers in HEC-1B cells upon completion of the developmental cycle. Chlamydial development in the less relevant McCoy cells was very similar to that in HeLa cells, although higher progeny counts were obtained. In conclusion, this three-dimensional bead culture system represents an improved model for harvesting large quantities of infectious chlamydia progeny from their more natural polarized epithelial host cells.
Article
Full-text available
Ocular, genital, and anogenital infection by the obligate intracellular pathogen Chlamydia trachomatis have been consistently associated with scar-forming sequelae. In cases of chronic or repeated infection of the female genital tract, infection-associated fibrosis of the fallopian tubes can result in ectopic pregnancy or infertility. In light of this urgent concern to public health, the underlying mechanism of C. trachomatis-associated scarring is a topic of ongoing study. Fibrosis is understood to be an outcome of persistent injury and/or dysregulated wound healing, in which an aberrantly activated myofibroblast population mediates hypertrophic remodeling of the basement membrane via deposition of collagens and other components of the extracellular matrix, as well as induction of epithelial cell proliferation via growth factor signaling. Initial study of infection-associated immune cell recruitment and pro-inflammatory signaling have suggested the cellular paradigm of chlamydial pathogenesis, wherein inflammation-associated tissue damage and fibrosis are the indirect result of an immune response to the pathogen initiated by host epithelial cells. However, recent work has revealed more direct routes by which C. trachomatis may induce scarring, such as infection-associated induction of growth factor signaling and pro-fibrotic remodeling of the extracellular matrix. Additionally, C. trachomatis infection has been shown to induce an epithelial-to-mesenchymal transition in host epithelial cells, prompting transdifferentiation into a myofibroblast-like phenotype. In this review, we summarize the field’s current understanding of Chlamydia-associated fibrosis, reviewing key new findings and identifying opportunities for further research.
Article
In recent years, substantial progress has been made in the understanding of the intracellular lifestyle of Chlamydia trachomatis and how the bacteria establish themselves in the human host. As an obligate intracellular pathogenic bacterium with a strongly reduced coding capacity, C. trachomatis depends on the provision of nutrients from the host cell. In this Review, we summarize the current understanding of how C. trachomatis establishes its intracellular replication niche, how its metabolism functions in the host cell, how it can defend itself against the cell autonomous and innate immune response and how it overcomes adverse situations through the transition to a persistent state. In particular, we focus on those processes for which a mechanistic understanding has been achieved. In this Review, Stelzner, Vollmuth and Rudel summarize current knowledge of Chlamydia trachomatis intracellular replication, its metabolism within the host cell and how it defends against host cell autonomous and innate immune responses, as well as its transition to a persistence state.
Article
Full-text available
The tumoral origin and extensive passaging of HeLa cells, a most commonly used cervical epithelial cell line, raise concerns on their suitability to study the cell responses to infection. The present study was designed to isolate primary epithelial cells from human ectocervix explants and characterize their susceptibility to C. trachomatis infection. We achieved a high purity of isolation, assessed by the expression of E-cadherin and cytokeratin 14. The infectious progeny in these primary epithelial cells was lower than in HeLa cells. We showed that the difference in culture medium, and the addition of serum in HeLa cultures, accounted for a large part of these differences. However, all things considered the primary ectocervical epithelial cells remained less permissive than HeLa cells to C. trachomatis serovar L2 or D development. Finally, the basal level of transcription of genes coding for pro-inflammatory cytokines was globally higher in primary epithelial cells than in HeLa cells. Transcription of several pro-inflammatory genes was further induced by infection with C. trachomatis serovar L2 or serovar D. In conclusion, primary epithelial cells have a strong capacity to mount an inflammatory response to Chlamydia infection. Our simplified purification protocol from human explants should facilitate future studies to understand the contribution of this response to limiting the spread of the pathogen to the upper female genital tract.
Article
Our understanding of how the obligate intracellular bacterial pathogen Chlamydia trachomatis reprograms the function of infected cells in the upper genital tract is largely based on observations made in cell culture with transformed epithelial cell lines. Here we describe a primary organoid system derived from endometrial tissue to recapitulate epithelial cell diversity, polarity, and ensuing responses to Chlamydia infection. Using high-resolution and time-lapse microscopy, we catalogue the infection process in organoids from invasion to egress, including the reorganization of the cytoskeleton and positioning of intracellular organelles. We show this model is amenable to screening C. trachomatis mutants for defects in the fusion of pathogenic vacuoles, the recruitment of intracellular organelles, and inhibition of cell death. Moreover, we reconstructed a primary immune cell response by co-culturing infected organoids with neutrophils, and determined that effectors like CPAF and TepP limit the recruitment of neutrophils to infected organoids. Collectively, our model can be applied to study the cell biology of Chlamydia infections in three dimensional structures that better reflect the diversity of cell types and polarity encountered by Chlamydia in their animal hosts.
Article
Full-text available
Commensals of the human body can shift to a pathogenic phase when the host immune system is impaired. This study aims to investigate the effect of seven yeast and two bacterial commensals and opportunistic pathogens isolated from blood and the female genital tract on the transepithelial electrical resistance (TER) of human cervical epithelial cell cultures (HeLa). The pathogens Candida tropicalis, C. parapsilosis, C. glabrata, C. krusei, C. albicans and Saccharomyces cerevisiae, caused a significant decrease in TER as compared to the controls; Lactobacillus spp caused a significant increase in TER versus the controls and Escherichia coli had no effect on the TER of the cell monolayers. The above data show that Candida spp., S. cerevisiae and Lactobacillus spp. have a non-selective effect on the TER of HeLa cell monolayers. These results are consistent with the in vivo non-selective action of these microorganisms on the various human mucosal epithelia.
Article
Full-text available
The ability of certain species of Chlamydia to inhibit the biogenesis of phagolysosomes permits their survival and replication within macrophages. Survival of macrophage-adapted chlamydiae correlates with the multiplicity of infection (MOI), and optimal chlamydial growth occurs in macrophages infected at MOI ≤ 1. In this study, we examined the replicative capacity of Chlamydia muridarum in the RAW 264.7 murine macrophage cell line at different MOI. C. muridarum productively infected these macrophages at low MOI but yielded few viable EB when macrophages were infected at moderate (10) or high (100) MOI. While high MOI caused cytotoxicity and irreversible host cell death, macrophages infected at a moderate MOI did not show signs of cytotoxicity until late in the infectious cycle. Inhibition of host protein synthesis rescued C. muridarum in macrophages infected at a moderate MOI, implying that chlamydial growth was blocked by activated defense mechanisms. Conditioned medium from these macrophages was anti-chlamydial and contained elevated levels of IL-1β, IL-6, IL-10 and IFN-β. Macrophage activation depended on TLR2 signaling and cytokine production required live, transcriptionally active chlamydiae. A hydroxyl radical scavenger and inhibitors of iNOS and cathepsin B also reversed chlamydial killing. High levels of reactive oxygen species (ROS) led to an increase in cathepsin B activity, and pharmacological inhibition of ROS and cathepsin B reduced iNOS expression. Our data demonstrate that MOI dependent TLR2 activation of macrophages results in iNOS induction via a novel ROS and cathepsin-dependent mechanism to facilitate C. muridarum clearance. Copyright © 2015, American Society for Microbiology. All Rights Reserved.
Article
Full-text available
New therapeutic strategies are needed to overcome drawbacks in treatment of infections with intracellular bacteria. Chlamydiaceae are Gram-negative bacteria implicated in acute and chronic diseases such as abortion in animals and trachoma in humans. Water-filtered infrared A (wIRA) is short wavelength infrared radiation with a spectrum ranging from 780 to 1400 nm. In clinical settings, wIRA alone and in combination with visible light (VIS) has proven its efficacy in acute and chronic wound healing processes. This is the first study to demonstrate that wIRA irradiation combined with VIS (wIRA/VIS) diminishes recovery of infectious elementary bodies (EBs) of both intra- and extracellular Chlamydia (C.) in two different cell lines (Vero, HeLa) regardless of the chlamydial strain (C. pecorum, C. trachomatis serovar E) as shown by indirect immunofluorescence and titration by subpassage. Moreover, a single exposure to wIRA/VIS at 40 hours post infection (hpi) led to a significant reduction of C. pecorum inclusion frequency in Vero cells and C. trachomatis in HeLa cells, respectively. A triple dose of irradiation (24, 36, 40 hpi) during the course of C. trachomatis infection further reduced chlamydial inclusion frequency in HeLa cells without inducing the chlamydial persistence/stress response, as ascertained by electron microscopy. Irradiation of host cells (HeLa, Vero) neither affected cell viability nor induced any molecular markers of cytotoxicity as investigated by Alamar blue assay and Western blot analysis. Chlamydial infection, irradiation, and the combination of both showed a similar release pattern of a subset of pro-inflammatory cytokines (MIF/GIF, Serpin E1, RANTES, IL-6, IL-8) and chemokines (IL-16, IP-10, ENA-78, MIG, MIP-1α/β) from host cells. Initial investigation into the mechanism indicated possible thermal effects on Chlamydia due to irradiation. In summary, we demonstrate a non-chemical reduction of chlamydial infection using the combination of water-filtered infrared A and visible light.
Book
Full-text available
Article
Full-text available
Trachoma is the leading infectious cause of blindness and is endemic in 52 countries. There is a critical need to further our understanding of the host response during disease and infection, as millions of individuals are still at risk of developing blinding sequelae. Infection of the conjunctival epithelial cells by the causative bacterium, Chlamydia trachomatis, stimulates an acute host response. The main clinical feature is a follicular conjunctivitis that is incompletely defined at the tissue-specific gene expression and molecular levels. To explore the features of disease and the response to infection, we measured host gene expression in conjunctival samples from Gambian children with active trachoma and healthy controls. Genome-wide expression and transcription network analysis identified signatures characteristic of the expected infiltrating immune cell populations, such as neutrophils and T/B lymphocytes. The expression signatures were also significantly enriched for genes in pathways which regulate NK cell activation and cytotoxicity, antigen processing and presentation, chemokines, cytokines, and cytokine receptors. The data suggest that in addition to polymorph and adaptive cellular responses, NK cells may contribute to a significant component of the conjunctival inflammatory response to chlamydial infection.
Article
Chlamydiae replicate intracellularly within a unique vacuole termed the inclusion. The inclusion circumvents classical endosomal/lysosomal pathways but actively intercepts a subset of Golgi-derived exocytic vesicles containing sphingomyelin (SM) and cholesterol. To further examine this interaction, we developed a polarized epithelial cell model to study vectoral trafficking of lipids and proteins to the inclusion. We examined seven epithelial cell lines for their ability to form single monolayers of polarized cells and support chlamydial development. Of these cell lines, polarized colonic mucosal C2BBe1 cells were readily infected with Chlamydia trachomatis and remained polarized throughout infection. Trafficking of (6-((N-(7-nitrobenz-2-oxa-1, 3-diazol-4-yl) amino)hexanoyl)sphingosine) (NBD-C(6)-ceramide) and its metabolic derivatives, NBD-glucosylceramide (GlcCer) and NBD-SM, was analyzed. SM was retained within L2-infected cells relative to mock-infected cells, correlating with a disruption of basolateral SM trafficking. There was no net retention of GlcCer within L2-infected cells and purification of C. trachomatis elementary bodies from polarized C2BBe1 cells confirmed that bacteria retained only SM. The chlamydial inclusion thus appears to preferentially intercept basolaterally-directed SM-containing exocytic vesicles, suggesting a divergence in SM and GlcCer trafficking. The observed changes in lipid trafficking were a chlamydia-specific effect because Coxiella burnetii-infected cells revealed no changes in GlcCer or SM polarized trafficking.
Article
Transcript array analysis is a novel technique that examines the expression of thousands of genes simultaneously. Transcript array analyses are being used to clarify the diagnosis and prognosis of malignancies, and to understand the underlying pathogenesis of complex human disorders such as the rheumatic diseases. In this review, the authors will outline the use of transcript arrays to simultaneously assess gene activation of hundreds or thousands of genes, and their potential use in understanding and managing rheumatic disorders. The authors focus on the use of transcript arrays to confirm and refine disease diagnoses, to generate new hypotheses regarding pathophysiology of rheumatic diseases, and to the possible profiling of patients with respect to their likely response to therapies.
Article
Full-text available
The inflammatory response associated with Chlamydia trachomatis genital infections is thought to be initiated by the release of proinflammatory cytokines from infected epithelial cells. This study focuses on the interactions between C. trachomatis-infected HeLa cells and immune cells involved in the early stages of infection, i.e., neutrophils and macrophages. First, we showed that the expression of interleukin-11 (IL-11), an anti-inflammatory cytokine mainly active on macrophages, was upregulated at the mRNA level in the genital tracts of infected mice. Second, incubation of differentiated THP-1 (dTHP-1) cells or monocyte-derived macrophages (MdM) with basal culture supernatants from C. trachomatis serovar E- or serovar L2-infected HeLa cells resulted in macrophage activation with a differential release of tumor necrosis factor alpha (TNF-alpha) and upregulation of indoleamine 2,3-deoxygenase (IDO) but not of Toll-like receptor 2 and 4 mRNA expression. Third, coculture of infected HeLa cells with dTHP-1 cells resulted in a reduction in chlamydial growth, which was more dramatic for serovar E than for L2 and which was partially reversed by the addition of anti-TNF-alpha antibodies for serovar E or exogenous tryptophan for both serovars but was not reversed by the addition of superoxide dismutase or anti-IL-8 or anti-IL-1beta antibodies. A gamma interferon-independent IDO mRNA upregulation was also detected in dTHP-1 cells from infected cocultures. Lastly, with a two-stage coculture system, we found that (i) supernatants from neutrophils added to the apical side of infected HeLa cell cultures were chlamydicidal and induced MdM to express antichlamydial activity and (ii) although polymorphonuclear leukocytes released more proinflammatory cytokines in response to serovar E- than in response to L2-infected cells, MdM were strongly activated by serovar L2 infection, indicating that the early inflammatory response generated with a nondisseminating or a disseminating strain is different.
Article
Chlamydia trachomatis is one of the major causes of bacterial sexually transmitted disease worldwide. The initial infection of endocervical epithelium in females is asymptomatic and commonly ascends to fallopian tubes when left untreated. Immunity to Chlamydia develops after infection and appears to provide short-term protection. Consequently, a significant rate of reinfection occurs among sexually active individuals, which can result in reproductive disability. T helper type 1 responses are implicated in providing protective immunity but may also contribute to tubal infertility. The purpose of this chapter is to review the factors that regulate the induction and recruitment of protective cellular immune responses within the local genital mucosa. An understanding of these events is important for the design of a protective vaccine and control of immunopathologic reactions.
Article
Emerging evidence indicates that intracellular signaling cascades mediate entry of pathogenic adenoviruses into target host cells as well as some of the undesirable inflammatory responses to adenoviral gene vectors. We found that Ad19 infection of cultured human corneal fibroblasts induced IL-8 gene transcription independently of IL-1beta, TNF-alpha, and viral gene expression, suggesting that intracellular signaling events might mediate early inflammatory events in adenovirus keratitis. Heat but not UV light inactivation of the virus abrogated the effect of infection on IL-8 mRNA and protein levels, consistent with a viral binding-mediated mechanism. The tyrosine kinase inhibitor herbimycin blocked Ad19-induced IL-8 expression. Western blot analysis revealed tyrosine phosphorylation of the functionally related kinases c-Src and extracellular signal-regulated kinase (ERK) 1/2 in corneal fibroblasts within 15 min after infection. Respective inhibitors of these kinases, PP2 and PD98059, also blocked Ad19-induced IL-8 mRNA and protein expression. Application of inhibitors to Src and ERK kinase assays suggested an upstream relationship of c-Src to ERK. Finally, DNA microarray studies performed 1 h after Ad19 or mock infection of corneal fibroblasts in the presence or absence of the Src-specific inhibitor PP2 confirmed a relationship between c-Src and IL-8 expression in Ad19-infected corneal cells. c-Src may act as a global regulator of early proinflammatory host responses to Ad19 infection of the human cornea.
Article
Full-text available
To develop a model of pathogenesis by which Chlamydia trachomatis progresses from acute to chronic infection, and finally serious disease (salpingitis, tubal occlusion). Review of current literature located through web-based Medline searches using key words: Chlamydia trachomatis, immunology, cytokines, heat shock protein, infertility. Cell-mediated immune mechanisms appear to be critical in determining whether acute infection is resolved or progresses into chronicity with pathological outcome. What determines the particular immune pathway depends on a range of determinants-HLA subtype and human genetics, cytokine profile, infectious load, route of infection, and endocrinology. A clearer picture of the natural history of chlamydial pathology may assist in providing better predictors of those women who may go on to develop significant sequelae after infection. Predicting those who may develop serious disease, including infertility, may contribute to improved management of such persons during earlier stages of infection and assist in prevention.
Article
Full-text available
Chlamydia trachomatis infection is the most common cause of sexually transmitted disease, leading to female pelvic inflammatory disease and infertility. The disease process has been linked to cellular response to this bacterial pathogen. This obligate intracellular pathogen infects macrophages, fibroblast cells, and epithelial and endothelial cells. We show in this study that infection of cervical epithelial cells, the primary target of Chlamydia trachomatis, leads to up-regulation and activation of the JAK/STAT signal pathway. Specifically, Chlamydia trachomatis infection of HeLa 229 cells selectively induces STAT1, STAT2, and IFN-stimulated transcription factor 3gamma expression and promotes STAT1 activation. The up-regulation of STAT1 is dependent on bacterial replication, because treatment of infected cells with antibiotics prevents STAT1 up-regulation. By analysis of the gene transcriptional and cytokine expression profiles of host cells combined with the use of neutralizing Abs, we show that IFN-beta production is critical for STAT1 induction in epithelial cells. Finally, we demonstrate that the host up-regulates STAT1 to restrict bacterial infection, because Chlamydia propagates more efficiently in STAT1-null or STAT1 knockdown cells, whereas Chlamydia growth is inhibited in cells with up-regulated STAT1 expression. This study demonstrates that the infected cells up-regulate the host innate antimicrobial response to chlamydial infection. It also highlights the importance of cellular response by nonimmune cells in host clearance of chlamydial infection.
Article
Full-text available
Chlamydia trachomatis infection is the most common cause of bacterial sexually transmitted diseases. Infection of the urogenital tract by C. trachomatis causes chronic inflammation and related clinical complications. Unlike other invasive bacteria that induce a rapid cytokine/chemokine production, chlamydial infection induces delayed inflammatory response and proinflammatory chemokine production that is dependent on bacterial growth. We present data here to show that the lipid metabolism required for chlamydial growth contributes to Chlamydia-induced proinflammatory chemokine production. By gene microarray profiling, validated with biochemical studies, we found that C. trachomatis LGV2 selectively upregulated PTGS2 (COX2) and PTGER4 (EP4) in cervical epithelial HeLa 229 cells. COX2 is an enzyme that catalyzes the rate-limiting step of arachidonic acid conversion to prostaglandins, including prostaglandin E2 (PGE2) and other eicosanoids, whereas EP4 is a subtype of cell surface receptors for PGE2. We show that Chlamydia infection induced COX2 protein expression in both epithelial cells and peripheral blood mononuclear cells and promoted PGE2 release. Exogenous PGE2 was able to induce interleukin-8 release in HeLa 229 epithelial cells. Finally, we demonstrated that interleukin-8 induction by Chlamydia infection or PGE2 treatment was dependent on extracellular signal-regulated kinase/mitogen-activated protein activity. Together, these data demonstrate that the host lipid remodeling process required for chlamydial growth contributes to proinflammatory chemokine production. This study also highlights the importance of maintaining a balanced habitat for parasitic pathogens as obligate intracellular organisms.
Article
Chlamydia trachomatis is the most common bacterial sexually transmitted disease in the United States and a major cause of female infertility due to infection-induced Fallopian tube scarring. Epithelial cells are likely central to host defense and pathophysiology as they are the principal cell type productively infected by C. trachomatis. We generated cloned murine oviduct epithelial cell lines without viral or chemical transformation to investigate the role of the TLRs and cytosolic nucleotide binding site/leucine-rich repeat proteins Nod1 and Nod2 in epithelial responses to Chlamydia muridarum infection. RT-PCR assays detected mRNA for TLR2 (TLRs 1 and 6), TLR3, and TLR5. No mRNA was detected for TLRs 4, 7, 8, and 9. Messenger RNAs for Nod1 and Nod2 were present in the epithelial cell lines. Oviduct epithelial cell lines infected with C. muridarum or exposed to the TLR2 agonist peptidoglycan secreted representative acute phase cytokines IL-6 and GM-CSF in a MyD88-dependent fashion. Infected epithelial cell lines secreted the immunomodulatory cytokine IFN-beta, even though C. muridarum does not have a clear pathogen-associated molecular pattern (PAMP) for triggering IFN-beta transcription. The oviduct epithelial lines did not secrete IFN-beta in response to the TLR2 agonist peptidoglycan or to the TLR3 agonist poly(I:C). Our data identify TLR2 as the principal TLR responsible for secretion of acute phase cytokines by C. muridarum-infected oviduct epithelial cell lines. The pattern recognition molecule responsible for infection-induced IFN-beta secretion by oviduct epithelial cells remains to be determined.
Article
Several chlamydial antigens have been detected in the infected epithelial cell cytosol and on the host cell surface prior to their presumed natural release at the end of the 72-96 h developmental cycle. These extra-inclusion antigens are proposed to influence vital host cell functions, antigen trafficking and presentation and, ultimately, contribute to a prolonged inflammatory response. To begin to dissect the mechanisms for escape of these antigens from the chlamydial inclusion, which are enhanced on exposure to antibiotics, polarized endometrial epithelial cells (HEC-1B) were infected with Chlamydia trachomatis serovar E for 36 h or 48 h. Infected cells were then exposed to chemotactic human polymorphonuclear neutrophils not loaded or pre-loaded in vitro with the antibiotic azithromycin. Viewed by electron microscopy, the azithromycin-mediated killing of chlamydiae involved an increase in chlamydial outer membrane blebbing followed by the appearance of the blebs in larger vesicles (i) everting from but still associated with the inclusion as well as (ii) external to the inclusion. Evidence that the vesicles originated from the chlamydial inclusion membrane was shown by immuno-localization of inclusion membrane proteins A, F, and G on the vesicular membranes. Chlamydial heat shock protein 60 (chsp60) copies 2 and 3, but not copy 1, were released from RB and incorporated into the everted inclusion membrane vesicles and delivered to the infected cell surface. These data represent direct evidence for one mechanism of early antigen delivery, albeit membrane-bound, beyond the confines of the chlamydial inclusion.
Article
Chlamydia trachomatis is a major cause of sexually transmitted disease worldwide for which an effective vaccine is being actively pursued. Current vaccine efforts will be aided by elucidating the interaction between Chlamydia and dendritic cells (DCs). Protective immunity appears to develop slowly following natural infection in humans, and early vaccine trials using inactivated C. trachomatis resulted in partial, short-lived protection with possible enhanced inflammatory pathology during re-infection. Thus, immunity following natural infection with live chlamydia may differ fundamentally from immune responses induced by immunization with inactivated chlamydia. We explored this conjecture by studying the response of DCs exposed to either viable or inactivated [ultraviolet (UV) -irradiated] chlamydia elementary bodies (EBs; designated as Live-EB and UV-EB, respectively) using Affymetrix GeneChip microarrays. Thirty-one immunologically characterized genes were differentially expressed by DCs following exposure to Live-EB or UV-EB, including two glutamic acid-leucine-arginine cysteine-X-cysteine (ELR CXC) neutrophil chemoattractant chemokines, Cxcl1 (KC), and Cxcl2 (MIP-2). Up-regulation of these genes by Live-EB as compared to UV-EB was verified by quantitative reverse transcription-polymerase chain reaction and increased chemokine secretion was confirmed by enzyme-linked immunosorbent assay both in vitro and in vivo. Immunofluorescence and fluorescence-activated cell sorter analysis of chlamydia-infected lung tissue confirmed that Live-EB but not UV-EB induced significant DC and neutrophil infiltration during infection. These observations demonstrate that the development of an antichlamydial immune response is dramatically influenced by chlamydial viability. This has implications as to why early inactivated chlamydial vaccines were ineffective and suggests that new vaccine design efforts may benefit from in vitro DC screening for ELR chemokine expression profiles.
Article
A common model for studying Chlamydia trachomatis and growing chlamydial stocks uses Lymphogranuloma venereum serovar L2 and non-polarized HeLa cells. However, recent publications indicate that the growth rate and progeny yields can vary considerably for a particular strain depending on the cell line/type used, and seem to be partially related to cell tropism. In the present study, the growth of invasive serovar L2 was compared in endometrial HEC-1B and endocervical HeLa cells polarized on collagen-coated microcarrier beads, as well as in HeLa cells grown in tissue culture flasks. Microscopy analysis revealed no difference in chlamydial attachment/entry patterns or in inclusion development throughout the developmental cycle between cell lines. Very comparable growth curves in both cell lines were also found using real-time PCR analysis, with increases in chlamydial DNA content of 400-500-fold between 2 and 36 h post-inoculation. Similar progeny yields with comparable infectivity were recovered from HEC-1B and HeLa cell bead cultures, and no difference in chlamydial growth was found in polarized vs. non-polarized HeLa cells. In conclusion, unlike other C. trachomatis strains such as urogenital serovar E, invasive serovar L2 grows equally well in physiologically different endometrial and endocervical environments, regardless of the host cell polarization state.
Article
Full-text available
Inflammation is a hallmark of chlamydial infections, but how inflammatory cytokines are induced is not well understood. Pattern recognition receptors (PRR) of the host innate immune system recognize pathogen molecules and activate intracellular signaling pathways that modulate immune responses. The role of PRR such as Toll-like receptors (TLR) and nucleotide-binding oligomerization domain (NOD) proteins in the endogenous interleukin-8 (IL-8) response induced during Chlamydia trachomatis infection is not known. We hypothesized that a PRR is essential for the IL-8 response induced by C. trachomatis infection. RNA interference was used to knock down the TLR signaling partner MyD88 as well as NOD1 and its signaling molecule receptor-interacting protein 2 (RIP2). IL-8 induced at 30 h postinfection by C. trachomatis was dependent on NOD1 signaling through RIP2; however, the IL-8 response was independent of MyD88-dependent TLR signaling. Activation of the extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase cellular signaling pathway, which is essential for up-regulation of IL-8 in response to C. trachomatis infection, was independent of NOD1 or RIP2. We conclude that the endogenous IL-8 response induced by C. trachomatis infection is dependent upon NOD1 PRR signaling through RIP2 as part of a signal system requiring multiple inputs for optimal IL-8 induction. Since ERK is not activated through this pathway, a concomitant interaction between the host and bacteria is additionally required for full activation of the endogenous IL-8 response.
ResearchGate has not been able to resolve any references for this publication.