Skip to main content
mSystems logoLink to mSystems
. 2023 Oct 24;8(6):e00904-23. doi: 10.1128/msystems.00904-23

Chlamydial YAP activation in host endocervical epithelial cells mediates pro-fibrotic paracrine stimulation of fibroblasts

Liam Caven 1,2, Rey Carabeo 1,
Editor: Hiutung Chu3
PMCID: PMC10734534  PMID: 37874141

ABSTRACT

Infection of the female genital tract by Chlamydia trachomatis can produce severe fibrotic sequelae, including tubal factor infertility and ectopic pregnancy. While infection demonstrably mediates a pro-fibrotic response in host cells, it remains unclear if intrinsic properties of the upper genital tract exacerbate chlamydial fibrosis. The relatively sterile environment of the upper genital tract is primed for a pro-inflammatory response to infection, potentially enhancing fibrosis; however, subclinical C. trachomatis infections still develop fibrosis-related sequelae. Here, we compare infection-associated and steady-state gene expression of primary human cervical and vaginal epithelial cells. In the former, we observe enhanced baseline expression and infection-mediated induction of fibrosis-associated signal factors (e.g., TGFA, IL6, IL8, and IL20), implying predisposition to Chlamydia-associated pro-fibrotic signaling. Transcription factor enrichment analysis identified regulatory targets of YAP, a transcriptional cofactor induced by infection of cervical epithelial cells, but not vaginal epithelial cells. YAP target genes induced by infection include secreted fibroblast-activating signal factors; therefore, we developed an in vitro model involving coculture of infected endocervical epithelial cells with uninfected fibroblasts. Coculture enhanced fibroblast expression of type I collagen, as well as prompting reproducible (albeit statistically insignificant) induction of α-smooth muscle actin. Fibroblast collagen induction was sensitive to siRNA-mediated YAP knockdown in infected epithelial cells, implicating chlamydial YAP activation in this effect. Collectively, our results present a novel mechanism of fibrosis initiated by Chlamydia, wherein infection-mediated induction of host YAP facilitates pro-fibrotic intercellular communication. Chlamydial YAP activation in cervical epithelial cells is thus a determinant of this tissue’s susceptibility to fibrosis.

IMPORTANCE

Chronic or repeated infection of the female upper genital tract by C. trachomatis can lead to severe fibrotic sequelae, including tubal factor infertility and ectopic pregnancy. However, the molecular mechanisms underlying this effect are unclear. In this report, we define a transcriptional program specific to C. trachomatis infection of the upper genital tract, identifying tissue-specific induction of host YAP—a pro-fibrotic transcriptional cofactor—as a potential driver of infection-mediated fibrotic gene expression. Furthermore, we show that infected endocervical epithelial cells stimulate collagen production by fibroblasts and implicate chlamydial induction of YAP in this effect. Our results define a mechanism by which infection mediates tissue-level fibrotic pathology via paracrine signaling and identify YAP as a potential therapeutic target for the prevention of Chlamydia-associated scarring of the female genital tract.

KEYWORDS: Chlamydia trachomatis, host-pathogen interaction, fibrosis, YAP

INTRODUCTION

The Gram-negative, obligate intracellular pathogen Chlamydia trachomatis is responsible for the most common bacterial sexually transmitted infection worldwide. Critically, it is estimated that up to 80% of C. trachomatis infections of the female genital tract are asymptomatic (1). The relatively poor state of C. trachomatis surveillance is of particular concern to public health, given the severe complications associated with chronic or repeated infection. In females, dissemination of the infection into the upper genital tract can lead to pelvic inflammatory disease (PID) (2, 3). Additionally, chlamydial infection of the fallopian tubes is associated with progressive scarring that can block descension of the ovum, thereby resulting in ectopic pregnancy or tubal factor infertility (TIF) (4, 5). A retrospective study of reproductive complications following screening for C. trachomatis in Danish women found that repeat infections increased the risk of PID by 20% (6). Similarly, a study modeling the risk of specific infection-associated sequelae using epidemiological data from the United Kingdom estimated that up to 29% of all TIF cases are attributable to C. trachomatis infection (7). Given these data, it is unsurprising that the study of chlamydial pathogenesis and the mechanisms underlying Chlamydia-associated fibrotic sequelae comprise topics of ongoing investigation.

Recent work suggests that C. trachomatis induces pro-fibrotic signaling by host epithelial cells. Chlamydia infection has been shown to induce signal factors associated with myofibroblast activation and wound healing, such as IL-6, IL-11, EGF, and CTGF (812). The related species C. pneumoniae exhibits similar induction of pro-fibrotic signaling, including production of TGF-β1, a consistent stimulus of myofibroblast differentiation (1315). Additionally, infection-mediated induction of the epithelial-to-mesenchymal transition (EMT) in host epithelial cells has been consistently reported over the past decade, including upregulation of mesenchymal markers such as N-cadherin, MMP9, and fibronectin, as well as the pro-EMT regulators ZEB1/2, SNAIL/SLUG, and thrombospondin 1 (1618). Epithelial cells undergoing EMT transdifferentiate into a myofibroblast-like phenotype, subsequently remodeling the extracellular matrix (ECM) to drive scar formation (19). Accordingly, it has been observed that Chlamydia infection induces host expression of genes associated with ECM remodeling, including ECM components (collagens and laminins), and ECM-modifying matrix metalloproteases (20, 21). Taken together, these data lend credence to a model of chlamydial fibrosis enhanced by but not necessarily dependent upon immune cell recruitment to the site of infection.

Chlamydial induction of EMT has been associated with deposition of fibrillar collagen—a common hallmark of scar formation—however, the mechanisms underlying Chlamydia-induced fibrosis remain incompletely characterized. In particular, it is unclear to what extent the pro-fibrotic signaling of infected epithelial cells influences other cell types not directly accessible for infection by Chlamydia, such as tissue-resident fibroblasts. Importantly, signal factors produced by Chlamydia-infected epithelial cells have been shown in other contexts to promote fibroblast differentiation into contractile and collagen-producing myofibroblasts (2225). Given that aberrant fibroblast activation has been linked to idiopathic pulmonary fibrosis and other forms of scarring-associated disease (26), an assessment of the effect of Chlamydia-associated signaling on this cell type may provide key insight into the mechanisms underlying chlamydial fibrosis.

While the terminally differentiated epithelial cells of the stratum corneum are considered a poor host for C. trachomatis, given their lack of mitochondria and attendant metabolic inactivity (27, 28), a recent report from our laboratory indicated that undifferentiated epithelial cells from this tissue can support C. trachomatis serovars D and L2 in a three-dimensional organotypic model of infection (29). Accordingly, a study comparing C. trachomatis serovar E infection of squamous ectocervical epithelial cells and endometrial epithelial cells cultured in vitro reported viable infection of the former, albeit at reduced efficiency (30). In combination with more recent reports of early passage immortalized human vaginal epithelial cells supporting infection of genital (serovar D) and LGV (serovar L2) C. trachomatis biovars (27), these data collectively illustrate the pathogen’s capacity to infect transitional or undifferentiated vaginal epithelial cells. Critically, fibrotic pathology is rarely reported in Chlamydia-infected vaginal epithelium, suggesting that intrinsic differences between cervical and vaginal epithelial cells may contribute to Chlamydia-associated scarring of the upper genital tract.

To determine if infection of a specific cell type is associated with pro-fibrotic gene expression, we have examined the host response to C. trachomatis in primary human cervical epithelial cells (HCECs) and primary human vaginal epithelial cells (HVEs). We observe both steady-state and infection-associated induction of pro-inflammatory and pro-fibrotic signaling in the former, including increased expression of TGFA, IL6, IL8, and IL20. Transcription factor enrichment analysis of this gene set implicated a broad portfolio of known regulatory targets of YAP, a pro-fibrotic transcriptional cofactor. Infection promoted YAP activation in cervical epithelial cells, but not vaginal epithelial cells, suggesting that YAP may contribute to pro-fibrotic gene expression specific to the upper genital tract. To assess the downstream effects of pro-fibrotic/pro-inflammatory signaling by infected cells, we have developed an in vitro model of infection wherein Chlamydia-infected, immortalized endocervical epithelial cells (End1s) are cocultured with uninfected uterine fibroblasts (KCO2s). We show that fibroblasts cocultured with infected epithelial cells exhibit significantly increased expression of collagen I, a phenotype sensitive to siRNA-mediated knockdown of YAP in host epithelial cells. Taken together, our results define a novel means by which infection promotes fibrotic gene and protein expression, dependent upon host YAP activity and intercellular communication.

RESULTS

Cervical epithelial cells exhibit intrinsic pro-inflammatory and pro-EMT gene expression relative to vaginal epithelial cells

To identify gene expression potentially driving fibrotic pathologies in the epithelial tissues of the upper genital tract, we compared the transcriptomes of mock and Chlamydia-infected cervical epithelial cells to that of infected vaginal epithelial cells. Primary HCECs and HVEs were either mock-infected or infected with the anogenital C. trachomatis serovar L2, then harvested for polyadenylated RNA for bulk RNA sequencing at 24 hours post-infection (hpi). Comparison of the Chlamydia-infected HCEC and HVE transcriptomes identified 7,354 genes differentially expressed (false discovery rate P-value [FDRP] ≤ 0.05) in infected cervical cells relative to infected vaginal cells (Fig. 1A; Table S1). To avoid the introduction of selection bias by solely examining genes functionally associated with pro-fibrotic and pro-EMT signaling, further characterization of this gene set was performed using the ranked functional enrichment analysis functions of the STRING database (31, 32). The differentially expressed gene set exhibited disproportionately high representation of induced genes associated with pro-inflammatory signaling, with GO-BP (gene ontology, biological process) terms associated with a type-I interferon response, leukocyte chemotaxis, and complement activation showing significant enrichment (Table 1). Intriguingly, the infected HCEC transcriptome also exhibited enrichment of GO-BP terms associated with keratinocyte and epidermal differentiation (Table 1). Recent reports have also shown Chlamydia can induce an EMT in endocervical epithelial cells, a pro-fibrotic transdifferentiation event associated with loss of cytokeratin expression (16, 33, 34). Accordingly, we observe significantly increased expression of EMT-regulating transcription factors such as SNAIL/SNAI1 (FC, 7.35) and TWIST1 (FC, 11.37) in infected HCECs relative to the infected HVE transcriptome. These data collectively suggest that cervical epithelial cells may be more susceptible than vaginal epithelial cells to infection-associated EMT.

Fig 1.

Fig 1

Cervical epithelial cells exhibit intrinsic pro-inflammatory and pro-EMT gene expression relative to vaginal epithelial cells. (A and B) Volcano plot of differential gene expression of Chlamydia trachomatis serovar L2-infected (A) or mock-infected (B) primary HCECs relative to equivalent HVEs at 24 hpi (see also Table S1). n = 3, with a minimum of 3 × 107 unstranded single reads per replicate with a mean length of 150 bp. All fold changes are relative to the mock-infected control; red dots: FDRP ≤ 0.05, labels: top 20 genes with lowest FDRP values. (C) Heatmap of cytokine and EMT-associated gene expression in mock-infected (left columns) and C. trachomatis serovar L2-infected (right columns) HCECs. All fold changes are relative to an equivalent infection in HVEs; only genes differentially expressed (FDRP ≤ 0.05) in at least one comparison are shown.

TABLE 1.

GO biological process terms significantly enriched (FDR ≤ 0.05) in comparison of C. trachomatis serovar L2-infected primary HCECs to primary HVEs at 24 hours post-infection via bulk RNA sequencinga

GO-BP term description Genes mapped Enrichment score Modulation False discovery rate
Ribonucleoprotein complex biogenesis 166 0.81 Repressed 3.66E−23
Ribosome biogenesis 121 0.89 Repressed 2.02E−20
ncRNA metabolic process 171 0.70 Repressed 1.74E−19
ncRNA processing 136 0.80 Repressed 2.28E−18
rRNA processing 95 0.87 Repressed 1.05E−15
Type I interferon signaling pathway 48 2.17 Induced 1.55E−13
Response to type I interferon 52 2.06 Induced 1.55E−13
Epidermal cell differentiation 98 1.36 Repressed 5.09E−13
rRNA metabolic process 100 0.79 Repressed 5.09E−13
Epidermis development 137 1.11 Repressed 1.63E−12
Keratinization 66 1.71 Repressed 5.92E−12
Keratinocyte differentiation 87 1.36 Repressed 8.12E−11
Humoral immune response 84 1.35 Induced 1.02E−10
Response to interferon-gamma 95 1.45 Induced 1.14E−09
Cornification 59 1.60 Repressed 4.15E−09
a

Induction/repression of each term is relative to the HVE infection.

The differences we report between vaginal and cervical epithelial cell gene expression during infection could be attributed to HVEs being less permissive to chlamydial infection; should HVEs exhibit a reduction in bacterial burden at 24 hpi relative to HCECs, this discrepancy would account for the diminished host response observed in the former. To assess this possibility, we examined chlamydial replication in HCECs and HVEs via the quantification of chlamydial genomes over the course of a 36-hour infection. Surprisingly, pathogen replication was substantially increased in HVEs over HCECs (Fig. S1A), comparable to infection of immortalized endocervical epithelial cells (End1/E6E7) previously used by our laboratory and others to model chlamydial infection (12, 35). Consistent with this result, we observe significant differences in production of infectious chlamydial organisms after a 36-hour infection (Fig. S1B), with HCECs exhibiting a greater than 10-fold reduction in recovered inclusion-forming units (IFUs) relative to HVEs and End1s. Taken together, these results indicate that reduced chlamydial fitness in HVEs cannot account for their diminished pro-inflammatory and pro-EMT response to infection.

Alternatively, differences in pro-inflammatory and pro-EMT gene expression between infected HCECs and HVEs may be the product of cell type-intrinsic differences in basal gene expression. Thus, we next compared the transcriptome of mock-infected control HCECs to that of mock-infected HVEs, identifying 4,720 genes differentially expressed in an HCEC-intrinsic fashion (Fig. 1B). Functional characterization of this gene set via STRING revealed a similar pattern of enriched terms as in the comparison of Chlamydia-infected cells, with relative induction of innate immunity as well as repression of keratinization and epidermal differentiation (Table 2). Expression of specific interleukins and CXCL-family cytokines followed a similar trend, with infected HCECs exhibiting strikingly increased expression of CXCL10 (FC, 480.76), IL6 (FC, 1423.03), and IL8 (FC, 186.51), as well as modest induction of IL1B (FC, 3.73) and IL24 (FC, 6.52) (Table S1). However, expression of other interleukins, such as IL7, IL15, and IL20, differed only between Chlamydia-infected HCECs and HVEs (Fig. 1C). Importantly, both IL-15 and IL-20 have been implicated in fibrosis (36, 37), suggesting that Chlamydia-infected endocervical epithelial cells may produce a wider portfolio of fibromodulatory cytokines relative to infection of other cell types. In contrast cytokeratin repression in HCECs was far more consistent between uninfected and Chlamydia-infected transcriptomes, as was relative induction of SNAI1 and TWIST1 (Fig. 1C). These data suggest HCEC-specific expression of EMT-associated genes may be the product of an intrinsic predisposition toward EMT induction, rather than an altered response to infection.

TABLE 2.

GO biological process terms significantly enriched (FDR ≤ 0.05) in comparison of steady-state expression in primary HCECs and primary HVEs via bulk RNA sequencinga

GO-BP term description Genes mapped Enrichment score Modulation False discovery rate
Innate immune response 196 1.06082 Induced 1.04E−10
Humoral immune response 59 1.65343 Induced 1.07E−07
Epidermal cell differentiation 79 1.15647 Repressed 3.85E−07
Epidermis development 113 0.966933 Repressed 2.39E−06
Epithelial cell differentiation 191 0.58554 Repressed 1.42E−05
Keratinization 57 1.30416 Repressed 1.42E−05
Cornification 53 1.29258 Repressed 1.42E−05
Response to bacterium 168 0.767625 Induced 1.66E−05
Cytokine-mediated signaling pathway 200 0.872896 Induced 1.66E−05
Regulation of defense response 184 0.680786 Induced 1.66E−05
Inflammatory response 151 0.82777 Induced 2.73E−05
Keratinocyte differentiation 67 1.14483 Repressed 4.06E−05
a

Induction/repression of each term is relative to HVE expression.

Cervical epithelial cells do not exhibit collagen or EMT induction during midcycle infection by C. trachomatis

While the effect of C. trachomatis infection on gene expression of host cervical epithelial cells has been extensively reported (20, 21, 38), prior study has focused on human papillomavirus (HPV)-transformed cell lines (e.g., HeLa, HEp-2). Given the potential for HPV transformation to dysregulate gene expression (39, 40), we next defined a more physiologically relevant host transcriptome by assessing differential expression of Chlamydia-infected primary HCECs. Relative to mock-infected control HCECs, infection with C. trachomatis serovar L2 induced differential expression (FDRP ≤ 0.05) of 8,241 genes at 24 hpi (Table S1). In accordance with prior reports describing the extensive pro-inflammatory response to infection, functional characterization of this gene set via STRING exhibited enrichment of immune and inflammation-associated GO-BP terms, including both positive and negative regulation of type I/II interferon signaling (Table 3). Critically, this included infection-dependent induction of specific interleukins (e.g., IL17, IL15, and IL20) and other genes associated with a type-I IFN response previously noted to exhibit HCEC-specific expression (Fig. 1B, Fig. 2A), such as IFITM1 (lFC, 7.56), BST2 (logFC, 6.63), IFI44L (FC, 7.00), and CXCL10 (FC, 112.38).

TABLE 3.

GO biological process terms significantly enriched (FDR ≤ 0.05) in comparison of C. trachomatis serovar L2-infected HCECs to mock-infected control HCECs via bulk RNA sequencinga

GO-BP term description Genes mapped Enrichment score Modulation False discovery rate
Response to type I interferon 52 1.90 Induced 8.16E−17
Type I interferon signaling pathway 49 1.97 Induced 2.30E−16
Response to interferon-gamma 98 1.12 Induced 7.92E−15
Interferon-gamma-mediated signaling pathway 46 1.37 Induced 1.08E−11
ncRNA metabolic process 176 0.45 Repressed 1.37E−11
Cilium organization 153 0.68 Repressed 1.37E−11
Cilium assembly 146 0.69 Repressed 1.73E−11
Cellular response to interferon-gamma 84 1.09 Induced 4.37E−11
Positive regulation of cytokine production 196 0.77 Induced 5.52E−11
Regulation of immune effector process 177 0.81 Induced 8.84E−11
Negative regulation of viral process 63 1.38 Induced 3.19E−10
Regulation of inflammatory response 150 0.73 Induced 9.45E−10
Adaptive immune response 105 0.86 Induced 2.03E−09
Cellular response to interleukin-1 71 0.84 Induced 2.03E−09
Ribonucleoprotein complex biogenesis 143 0.43 Repressed 2.56E−09
a

Induction/repression of each term is relative to the mock infection.

Fig 2.

Fig 2

Cervical epithelial cells do not exhibit collagen or EMT induction during midcycle infection by C. trachomatis. (A and B) Heatmap of cytokine (A) and collagen/EMT-associated (B) gene expression in mock-infected HCECs relative to equivalent HVEs (left columns), Ct L2-infected HCECs relative to equivalent HVEs (center columns), and Ct L2-infected HCECs relative to mock-infected HCECs (right columns). Only genes differentially expressed (FDRP ≤ 0.05) in at least one comparison are shown. (C and D) Expression of epithelial/mesenchymal differentiation markers (C) and EMT-associated transcription factors (D) at 24 hpi in mock- and Ct L2-infected End1/E6E7 immortalized endocervical epithelial cells, as measured by reverse transcription-quantitative PCR. n = 3 biological replicates; fold changes are relative to mean expression of the mock-infected and untreated control. Whiskers: minimum to maximum; asterisks: P-values ≤ 0.05, using pairwise Student’s t-tests and Bonferroni’s correction for multiple comparisons.

Intriguingly, the infection-associated transcriptome exhibited repression of ECM components, with GO-CP (gene ontology, cellular component) terms associated with collagens and the ECM exhibiting modest enrichment among the downregulated gene set . Indeed, infected HCECs exhibited reduced expression of fibrillar (e.g., COL1A1 and COL5A1), fibril-associated (e.g., COL16A1 and COL21A1), and network-forming (COL4A1) collagens relative to mock-infected HCECs, despite collagen expression of infected HCECs generally exceeding that of infected HVEs (Fig. 2B). The mesenchymal marker vimentin/VIM was repressed by infection (FC, −2.22), whereas common markers of EMT (e.g., CDH1/2, SNAI1/2, and ZEB1/2) were not differentially expressed (Fig. 2B), suggesting that infection does not induce EMT in endocervical epithelial cells at 24 hpi. To confirm this hypothesis, we next examined expression of EMT-associated markers at this time point during C. trachomatis serovar L2 infection of immortalized endocervical epithelial cells (End1/E6E7), which have previously been demonstrated to undergo EMT in a chronic/persistent model of C. trachomatis infection (35). Critically, infected End1s did not exhibit induction of mesenchymal differentiation markers (e.g., CDH2 and VIM) or repression of epithelial differentiation markers (e.g., CDH1 and KRT13) (Fig. 2C). In similar fashion, transcription factors associated with EMT induction did not exhibit induction at 24 hpi, with the notable exception of SNAI1/SNAIL (Fig. 2D). Collectively, these data imply that chlamydial induction of EMT in cervical epithelial cells does not occur by 24 hpi, despite an apparent predisposition toward mesenchymal differentiation relative to vaginal epithelial cells. Thus, pro-fibrotic gene expression observed at this time point likely occurs via an EMT-independent mechanism.

Induction of the pro-fibrotic transcriptional cofactor YAP is specific to infection of cervical epithelial cells

To identify alternative mechanisms of pro-fibrotic gene expression potentially modulated by Chlamydia, we next defined a common signature of pro-fibrotic genes exhibiting distinct expression in the upper genital tract as well as differential expression during C. trachomatis infection. Importantly, only a subset of genes differentially expressed in an HCEC-intrinsic fashion (mock HCEC vs mock HVE expression) exhibited comparable induction/repression in an infection-mediated fashion (mock HCEC vs Ct L2-infected HCEC expression). Upon comparing the HCEC-intrinsic (4,720 genes) and infection-mediated (8,241 genes) differentially expressed gene sets, only 2,391 genes were common to both (Fig. 3A). Of these, 1,158 (49.4%) were equivalently induced or repressed in both comparisons, suggesting their likely relevance to infection-associated fibrosis of the upper genital tract (Fig. 3A). More than half (629 genes, or 54.3%) of this gene set mapped to the Comparative Toxicogenomics Database of pro-fibrotic genes (Fig. 3B), including numerous pro-fibrotic signal factors (e.g., IL6, IL24, and TGFA) and ECM-remodeling enzymes (e.g., ADAMTS9, MMP13, and TIMP2/3) (Table S2). Critically, expression of this gene set in infected HCECs significantly correlated with our previously reported bulk RNA sequencing of Ct serovar L2-infected End1/E6E7 immortalized endocervical epithelial cells (Fig. 3B). These data suggest that modulation of this putatively fibrotic gene set is conserved between in vitro models of chlamydial pathogenesis in the upper genital tract, additionally recommending use of End1/E6E7 cells for subsequent investigation of the mechanisms underlying infection-associated pro-fibrotic gene expression.

Fig 3.

Fig 3

Induction of the pro-fibrotic transcriptional cofactor YAP is specific to infection of cervical epithelial cells. (A) Venn diagrams of genes differentially expressed (top, FDRP ≤ 0.05), induced (middle, FDRP ≤ 0.05, log2FC > 0), or repressed (bottom, FDRP ≤ 0.05, log2FC < 0) in an HCEC-intrinsic (red, mock-infected HCECs vs mock-infected HVEs) or infection-mediated fashion (green, Ct L2-infected HCECs vs mock-infected HCECs). (B) Scatter plot of gene expression Ct L2 infection of HCECs (x-axis) and End1s (y-axis) at 24 hpi of the subset of genes equivalently modulated by steady-state HCEC expression relative to HVEs (see also Table S2). All fold changes are relative to each cell type’s respective mock-infected control; blue line: linear regression model of correlation; gray shading: 95% confidence interval. R2 and P-values calculated using Pearson’s correlation. (C) Representative micrographs of YAP (green) translocation into the nuclei (blue) of confluent mock- and Ct L2-infected HVEs, HCECs, and End1s at 24 hpi. Scale bar: 20 µm. (D) Quantification of YAP nuclear translocation in C as a ratio of nuclear to cytosolic YAP fluorescence normalized to each cell type’s respective mock-infected control. n = 3 biological replicates, 50 cells measured per sample. Black bars: group means; asterisks: P-values ≤ 0.05, using pairwise Wilcoxon rank sum tests and Bonferroni’s correction for multiple comparisons.

One potential means by which infection may facilitate host expression of pro-fibrotic gene expression is via modulation of host transcription factors. Indeed, past work by our laboratory and others have defined mechanisms by which C. trachomatis and related species manipulate host transcription factor activity (12, 41, 42). Thus, we next performed transcription factor enrichment analysis of this putatively fibrotic gene set via ChIP-X Enrichment Analysis 3 (ChEA3) (43), identifying 56 transcription factors exhibiting significant enrichment (Table S3). This included known mediators of pro-fibrotic and pro-inflammatory signaling, such as RELA, IRF1/8, SMAD3, and EGR1. Strikingly, nearly half (25) of the enriched transcription factors are known regulatory targets of YAP (Table 4), a pro-fibrotic transcriptional cofactor of which we recently reported Chlamydia-directed induction (12). Given the potential for YAP activation to drive expression of pro-fibrotic gene expression specific to the upper genital tract, we next assessed the degree to which C. trachomatis infection facilitates YAP nuclear translocation in vaginal epithelial cells. After infecting confluent monolayers of each cell type with C. trachomatis serovar L2, YAP nuclear translocation was quantified as a ratio of nuclear to cytosolic immunofluorescence. Critically, infected HVEs did not exhibit YAP nuclear translocation relative to mock-infected control monolayers, in stark contrast to the significant increase in nuclear YAP observed during infection of either primary (HCEC) or immortalized (End1) endocervical epithelial cells (Fig. 3C and D). Collectively, these data suggest that chlamydial YAP activation may mediate pro-fibrotic gene expression specific to infection of the upper genital tract.

TABLE 4.

YAP-associated transcription factors exhibiting significant enrichment (FDR ≤ 0.05) in the set of genes equivalently modulated both by Ct L2 infection of HCECs and steady-state HCEC expression relative to HVEs

Transcription factor Genes mapped Set length FDR YAP regulatory relationship
SOX2 236 2,308 8.68E−21 Expression induced by YAP/Oct4 (44)
ESR2 50 394 4.81E−08 Expression repressed by YAP/NuRD (45)
BACH1 114 1,298 1.96E−07 Coactivated by YAP (46)
STAT3 82 914 5.90E−06 Coactivated by YAP (47)
JUN 138 1,830 2.60E−05 Coactivated by YAP (47)
SMAD3 129 1,759 1.56E−04 Coactivated by YAP (48)
JUND 128 1,831 8.94E−04 Coactivated by YAP (47)
SALL4 62 775 0.00155 Expression induced by YAP/TEAD2 (49)
MYB 64 837 0.0036 Coactivated by YAP (50, 51)
EGR1 49 605 0.00438 Coactivated by YAP (52)
ATF3 130 1,971 0.00541 Expression induced by YAP/TAZ (53)
FOXA2 178 2,814 0.00541 Enhancer binding altered by YAP (49)
SOX9 89 1,270 0.00541 Induced/activated by YAP/TEAD (54)
NANOG 157 2,452 0.00582 Expression induced by YAP/TEAD2 (55)
WT1 99 1,450 0.00612 Represses CDH1 with YAP (56)
CEBPB 90 1,300 0.0064 Coactivated by YAP (47)
RXRA 106 1,584 0.00738 Coactivated by YAP (57)
TP53 69 981 0.0122 Expression induced by YAP (58)
CDX2 37 461 0.0141 Expression induced by YAP/TEAD4 (59)
MITF 300 5,140 0.0141 Expression induced by YAP/PAX3 (60)
SMAD4 31 371 0.0158 YAP-dependent nuclear retention (61)
RARG 29 342 0.0165 Coactivated by YAP (57)
KLF5 113 1,803 0.0271 Induced/stabilized by YAP (62)
FOXA1 113 1,842 0.0421 Bound by YAP/TFCP2 complex (63)
CEBPA 36 493 0.0461 Coactivated by YAP (47)

Coculture with infected endocervical epithelial cells alters the putative myofibroblast transcriptome

Given our data indicating that infected endocervical epithelial cells exhibit induction of pro-fibrotic signal factors, one potential mechanism through which C. trachomatis may induce fibrosis is via signaling to stromal fibroblasts. Indeed, differentiation of tissue-resident fibroblasts into ECM-remodeling myofibroblasts has been consistently associated with other forms of fibrotic disease (26); however, the contribution of fibroblasts to Chlamydia-associated scarring has not been assessed. Importantly, the lack of clinical evidence of Chlamydia-infected fibroblasts in vivo necessitates a model of infection wherein uninfected fibroblasts are cocultured with infected endocervical epithelial cells. Past study of the kinetics of C. trachomatis infection epithelial cells indicates that pathogen-directed uptake of the bacterium is rapid, with a majority of invading bacteria internalized as soon as 10 minutes post-infection (64, 65). Critically, this event requires interaction of Chlamydia with the host cell plasma membrane in two distinct stages: transient adhesion relying on electrostatic interactions with membrane glycosaminoglycans (66, 67), followed by irreversible attachment due to binding with host cell receptors and/or engagement of the pathogen’s type III secretion system (68). Thus, we infected immortalized End1s seeded at 35% of confluence with the genetically tractable C. trachomatis serovar L2, adding immortalized KCO2-44D uterine fibroblasts at 1 hpi. After 23 hours of incubation, cocultures prepared via this method exhibited a consistent morphology characterized by islands of epithelial cells interspersed with fibroblasts, of which the latter were readily distinguished by immunofluorescence staining of actin stress fibers (Fig. 4A). Importantly, fibroblasts did not exhibit visible chlamydial inclusions containing DAPI-positive organisms, in contrast to the readily apparent inclusions of infected epithelial cells (Fig. 4A).

Fig 4.

Fig 4

Coculture with infected endocervical epithelial cells alters the putative myofibroblast transcriptome. (A) Representative micrographs of mock- (left) or C. trachomatis serovar L2-infected (right) End1/E6E7 endocervical epithelial cells cocultured with KCO2 uterine fibroblasts for 23 hours (starting at 1 hpi); green: actin expression (phalloidin), blue: DAPI, arrowheads: chlamydial inclusions, scale bar: 20 µm. (B) Uniform manifold approximation and projection (UMAP) feature plot of KRT19 (top) and VIM (bottom) expression in same-well coculture of mock- or Ct serovar L2-infected End1 endocervical epithelial cells with KCO2 fibroblasts for 23 hours (starting at 1 hpi). n = 2 biological replicates, 1,000 cell libraries per sample, approximately 5–7 × 104 reads/cell. (C) Top, UMAP plot of fibroblast (VIM+) subclustering in same-well coculture of mock- or Ct serovar L2-infected End1 endocervical epithelial cells with KCO2 fibroblasts. Colors: cluster identity, all percentages relative to total fibroblast population of the respective mock-/Ct L2-infected coculture. Bottom, violin plot of fibroblast (VIM+) per-cluster expression of α-SMA/ACTA2 (left) and TIMP1 (right) in same-well coculture of mock- or Ct serovar L2-infected End1 endocervical epithelial cells with KCO2 fibroblasts. Colors: cluster identity, dots: per-cell expression values. (D) Per-cell expression heatmap of genes differentially expressed (P ≤ 0.05) by cluster F3 fibroblasts of Ct serovar L2-infected cocultures (right), relative to cluster F3 fibroblasts of mock-infected cocultures (left).

To both confirm the absence of Chlamydia-infected fibroblasts and examine the transcriptome of uninfected fibroblasts, we then harvested cocultures at 24 hpi via trypsinization for library preparation and single-cell RNA sequencing. End1/KCO2 cell populations were readily distinguished on the basis of epithelial and mesenchymal marker expression, with End1s exhibiting heightened expression of cytokeratin 19 (KRT19) and KCO2s exhibiting heightened expression of vimentin (VIM) (Fig. 4B). To identify Chlamydia-infected cells, sequencing results were aligned to the hg18 reference genome appended with C. trachomatis reference sequences for highly expressed, polyadenylated RNA transcripts most likely to be captured by library preparation and sequencing, including the chlamydial outer membrane protein ompA, the polymorphic membrane protein pmpC, the nucleoside phosphate kinase ndk, and the pgp8 antisense sRNA-2. Expression of chlamydial reference sequences identified an epithelial cell cluster distinct from the mesenchymal population—consistent with our previous findings in bulk RNA-sequencing of both HCECs and End1s, this cluster exhibited induction of genes associated with a type I interferon response (e.g., IFI6, IFITM2, IFITM3, and HLA-A/C) (Table S4). Importantly, chlamydial marker genes were not detectable in uninfected cocultures (Fig. S2)—taken together, these data demonstrate the viability of this approach as a means to render cocultured fibroblasts inaccessible to Chlamydia.

To determine if coculture with infected epithelial cells altered the transcriptome of “bystander” fibroblasts, we performed clustering analysis on the mesenchymal cell population via Seurat. We identified four mesenchymal subclusters distinguished by marker gene expression, designated clusters F1–F4 (Fig. 4C, top), with the mesenchymal population of Chlamydia-infected cocultures exhibiting a modest increase in the proportion of cluster F3 relative to a mock-infected control (24.3% and 17.4%, respectively). Given that cluster F3 also exhibited enhanced expression of the myofibroblast differentiation marker smooth muscle actin (α-SMA/ACTA2), as well as tissue inhibitor of matrix metallopeptidases 1 (TIMP1), this result suggests that coculture with infected-epithelial cells may enhance myofibroblast differentiation (Fig. 4C, bottom). Further comparison of cluster F3 gene expression in Chlamydia- and mock-infected cocultures revealed differential expression of a variety of genes associated with fibrosis (Fig. 4D). For example, cluster F3 fibroblasts from Chlamydia-infected cocultures exhibited significant repression of HES1, a bHLH-family transcription factor known to inhibit the regulator of myofibroblast differentiation MyoD (69, 70). Coculture with infected epithelial cells additionally inhibited expression of chromobox protein homolog 3 (CBX3), which was recently shown to inhibit proliferation and collagen production of vascular smooth muscle cells (71). “Bystander” cluster F3 fibroblasts also exhibited increased expression of pro-inflammatory genes, including C1QBP and ISG15, suggesting that cocultured fibroblasts may respond to infection-associated induction of pro-inflammatory signaling by host endocervical epithelial cells. Collectively, these data imply that infected epithelial cells may signal to cocultured fibroblasts, thereby stimulating pro-fibrotic gene expression.

Chlamydia-infected endocervical epithelial cells induce fibroblast collagen I expression in a YAP-dependent fashion

A key functional consequence of myofibroblast differentiation is the capacity to remodel the extracellular matrix via the production of collagens (72, 73). In healthy epithelial tissues, the ECM of the basement membrane is predominantly composed of type IV network-forming collagen, laminins, and the collagen/laminin crosslinking protein nidogen (74, 75). In contrast, fibrotic tissues exhibit heightened proportionality of fibrillar collagens (e.g., types I, II, III, and V) in the basement membrane, which has been attributed to deposition by myofibroblasts in idiopathic pulmonary fibrosis and other forms of scarring disease (72, 74, 76, 77). In similar fashion, it has been shown that Chlamydia-infected epithelial cells exhibit increased production of fibrillar collagen due to chlamydial induction of EMT and consequent host cell transdifferentiation into a myofibroblast-like phenotype (17).

In light of these results, one potential consequence of the altered fibroblast transcriptome in Chlamydia-infected cocultures may be increased production of fibrillar collagens. Thus, we prepared an alternative in vitro model of chlamydial infection, wherein End1 epithelial cells were seeded/infected on the semipermeable membrane of a transwell insert, washed with heparan sulfate at one hpi, then transferred to a plate containing seeded KCO2 fibroblasts (Fig. 5A). This approach provided a means to perform population-level measurement on cocultured fibroblasts, given that the 0.4-µm pore size of the transwell membrane would necessarily restrict passage of cells, while still allowing for cell-cell communication via the action of diffusible signal factors. Critically, KCO2s cocultured with Ct serovar L2-infected End1s in transwells for 23 hours (24 hpi) exhibited a significant increase in production of type I collagen relative to an uninfected control, as measured by immunofluorescence (Fig. 5B and C). Induction of collagen I expression occurred in a fashion consistent with mock-infected cocultures treated with TGF-β1 (a known inducer of myofibroblast differentiation and collagen deposition) (7880). Consistent with this result, we observed a significant increase in expression of COL1A1 in cocultured fibroblasts as measured by reverse transcription quantitative PCR (RT-qPCR); importantly, this phenotype was observed in both infections with C. trachomatis serovar L2 and serovar D (Fig. 5D), suggesting that infection-associated induction of fibroblast activity is conserved between anogenital and urogenital serovars of Chlamydia.

Fig 5.

Fig 5

Chlamydia-infected endocervical epithelial cells induce fibroblast expression of collagen I in a YAP-dependent fashion. (A) Schematic representation of the transwell-mediated approach of coculturing C. trachomatis-infected endocervical epithelial cells (top compartment) with uninfected uterine fibroblasts (bottom compartment). (B) Representative micrographs of collagen I (green) produced by KCO2 fibroblasts cocultured with mock-infected, Ct L2-infected, or TGF-β1-treated (20 ng/mL) End1 cells for 24 hours, starting at 1 hpi. Scale bar: 20 µm. (C) Quantification of mean fluorescence intensity of collagen I in B. n = 3 biological replicates, five fields measured per sample. Black bars: group means; asterisks: P-values ≤ 0.05, using pairwise Wilcoxon rank sum tests and Bonferroni’s correction for multiple comparisons. (D) COL1A1 expression as measured by RT-qPCR in KCO2 fibroblasts cocultured for 24 hours with mock-, Ct L2-, or Ct D-infected End1 cells (coculture starting at one hpi). n = 4 biological replicates; fold changes are relative to mean expression of the mock-infected control. Whiskers: minimum to maximum; asterisks: P-values ≤ 0.05, using Student’s t-tests and Bonferroni’s correction for multiple comparisons. (E) COL1A1 and ACTA2 expression as measured by RT-qPCR in KCO2 fibroblasts cocultured for 24 hours with siRNA-transfected (10 nM of YAP1-targeting or non-targeting siRNA, for 24 hours prior to infection), Ct L2-infected End1 cells (coculture starting at 1 hpi). n = 3 biological replicates; fold changes are relative to mean expression of the equivalent mock-infected control. Whiskers: minimum to maximum; asterisks: P-values ≤ 0.05, using Student’s t-tests and Bonferroni’s correction for multiple comparisons.

We have previously demonstrated that C. trachomatis infection of endocervical epithelial cells induces expression of the fibroblast-activating signal factors CTGF and INHBA, in a fashion dependent upon pathogen-directed induction of the host transcriptional cofactor YAP (12). Furthermore, here, we observe infection-mediated YAP activation specific to cervical epithelial cells, suggesting a role for chlamydial YAP activation in the fibrotic pathology associated with chlamydial infection of the upper genital tract. Thus, to assess the role of chlamydial YAP activation in stimulation of fibroblast collagen production, we next opted to knock down YAP expression in epithelial cells via siRNA at 24 hours prior to infection. After 23 hours of coculture (24 hpi), fibroblast coculture expression was assessed via RT-qPCR. Knockdown of YAP in Chlamydia-infected End1s was approximately 40% efficient, as measured by Western blotting of total YAP protein levels, in accordance with other reports of siRNA-mediated knockdown efficiency in End1s (12, 81) (Fig. S3). Fibroblast expression of collagen I was significantly increased in infected cocultures transfected with non-targeting control siRNA, consistent with our prior observations via immunofluorescence. However, transfection of infected End1s with YAP-targeting siRNA significantly attenuated this phenotype, with cocultured fibroblasts exhibiting collagen I (COL1A1) expression indistinguishable from mock-infected cocultures (Fig. 5D). Intriguingly, expression of the myofibroblast differentiation marker α-SMA (ACTA2) exhibited a similar, albeit statistically insignificant trend (Fig. 5E). Taken together, these data confirm that Chlamydia-infected epithelial cells stimulate fibroblast production of collagen, as well as implicate chlamydial induction of host cell YAP activity in this effect.

DISCUSSION

To elucidate the mechanisms underlying Chlamydia-mediated fibrosis of the upper genital tract, we have presented comparative analysis of both steady-state and infection-associated gene transcription in primary HCECs and primary HVEs. Both mock- and Chlamydia-infected HCECs exhibited enhanced expression of pro-inflammatory and pro-fibrotic cytokines (e.g., IL6 and IL8), induction of the pro-EMT transcription factors SNAI1/SNAIL and TWIST1, and relative repression of multiple cytokeratins. However, expression of specific fibromodulatory interleukins (e.g., IL7, IL15, and IL20) was enhanced only in Chlamydia-infected HCECs, suggesting a cell type-specific response to infection. Transcription factor enrichment analysis of genes equivalently modulated by infection and HCEC-intrinsic expression relative to HVEs revealed a broad portfolio of known YAP regulatory targets. YAP nuclear translocation of YAP was enhanced during infection of HCECs, but not HVEs, suggesting a role for this transcriptional cofactor in mediating the HCEC-specific induction of pro-fibrotic signaling. To investigate this possibility, we examined the response of uninfected fibroblasts to coculture with Chlamydia-infected endocervical epithelial cells, observing a modest expansion of α-SMA/TIMP1-expressing putative myofibroblasts. Further iteration on this coculture approach via use of transwells enabled population-level measurement of cocultured fibroblast gene expression. We find that cocultured fibroblasts exhibit heightened expression of type I collagen—critically, this phenotype was conserved between anogenital (L2) and urogenital (D) serovars of Chlamydia and sensitive to siRNA-mediated knockdown of YAP in infected endocervical epithelial cells. Taken together, our results present a novel mechanism by which Chlamydia mediates fibrotic tissue remodeling, wherein infection-mediated YAP activation in host endocervical epithelial cells promotes fibroblast collagen expression via the induction of pro-fibrotic signaling.

The apparent repression of collagen expression in infected HCECs is surprising, given past reports of chlamydial EMT leading to production of type I collagen in infected primary cells of the murine oviduct (17). Deposition of fibrillar collagen is a hallmark of pro-fibrotic remodeling of the ECM in other forms of scarring disease (7476); in light of this, the role of Chlamydia-infected epithelial cells in collagen synthesis is critically understudied. These data suggest two possibilities: either previous reports of fibrillar collagen production by infected cells are unique to infection of murine oviduct epithelial cells, or infection inhibits collagen expression in host cells prior to induction of EMT. Importantly, prior study of chlamydial EMT induction indicates this phenotype occurs in late-cycle or persistent models of infection, with murine oviduct epithelial cells and End1 cells expressing mesenchymal markers at 72 hours or 7 days post-infection, respectively. Consequently, infection-mediated repression at 24 hpi may be a transient effect, superseded by pathogen-directed host transdifferentiation into a myofibroblast-like and collagen-producing phenotype. Indeed, prior reports indicating collagen expression is not enhanced in HeLa cells at 24 hpi would seem to support this hypothesis (20). Further study is required to elucidate the impact of chlamydial EMT induction on basement membrane architecture and thereby determine the physiological relevance of this phenotype to the development of infection-associated fibrosis. Importantly, the infection-mediated induction of fibroblast collagen expression we observe may constitute a means by which Chlamydia may stimulate ECM deposition in an EMT-independent fashion.

Past study of C. trachomatis in murine models of infection has shown that the pathogen induces EMT in host epithelial cells of the murine oviduct, as well as host production of collagen I and TGF-β1 (a known inducer of myofibroblast differentiation) (13, 16, 17). However, fibrotic sequelae arising from C. trachomatis infection of the murine female genital tract has not been reported, suggesting that animal models of infection may not faithfully recapitulate the molecular events underlying Chlamydia-associated scarring in humans. A likely explanation for this discrepancy is the repeated observation that murine intravaginal infections with C. trachomatis fail to ascend into the upper genital tract and are readily cleared by the innate immune system (82, 83). That being said, recent work has demonstrated transcervical infections with C. trachomatis serovar D severely impact fertility in the mouse model (84, 85), emphasizing the importance of infection of cervical epithelial cells to the development of chlamydial sequelae. Intriguingly, the immune response to transcervical C. trachomatis infections includes robust recruitment of CD4+ T-cells and secretion of IL-17 (85, 86)—features consistent with the cell type-intrinsic induction of IL-17 expression and CD4+ T-cell survival and chemotactic factors (e.g., IL-6 and IL-8) we observe in both uninfected and infected HCECs. In light of this, use of immortalized human cell lines in an in vitro model of infection thus has the potential to provide greater insight into how Chlamydia-infected cervical epithelial cells drive tissue-wide fibrotic pathologies. Here, we report that infected endocervical epithelial cells alter gene expression and induce collagen production in cocultured, uninfected fibroblasts—demonstrating a means by which C. trachomatis may coordinate pro-fibrotic activity of cell types beyond the simple columnar cervical epithelium the pathogen demonstrably infects in vivo and complementary to the immune cell recruitment that has been repeatedly associated with clinical study of Chlamydia-associated infertility.

The observation that collagen I expression in fibroblasts was sensitive to siRNA-mediated knockdown of YAP in epithelial cells is in accordance with our prior report of infection- and YAP-dependent expression of pro-fibrotic signal factors (12). However, it is unclear to what extent specific signaling factors are relevant to or dispensable for induction of this phenotype. Given consistent role in induction of fibroblast differentiation and ECM remodeling (25, 8789), this signal factor constitutes a likely candidate for YAP- and infection-dependent induction of collagen synthesis in fibroblasts in cocultured fibroblasts. That being said, YAP has also been implicated in the expression of several other pro-fibrotic signaling factors. A recent report implicated YAP in inducing expression of the pro-fibrotic interleukin IL-6 in the context of endometrial cancer (90). YAP has also been shown to enhance TGF-β expression and signal transduction via binding to and enhancing the activity of SMAD2/3 (48). It has also been implicated in the expression of the myofibroblast-activating interleukin IL-33, with Cre/Lox-mediated YAP knockout significantly attenuating IL-33 expression and scarring in a murine model of post-infarction cardiac fibrosis (91). YAP has been additionally shown to modulate the Wnt/β-catenin signaling pathway, via enhancement of both β-catenin expression and transcriptional activity (92). Given the demonstrable role of this signaling pathway in fibrosis generally and regulation of TGF-β expression specifically (93, 94), these data collectively demonstrate multiple, complementary means by which chlamydial induction of YAP may facilitate pro-fibrotic signaling. Ultimately, further study of the role of YAP must begin with clearly defining the YAP regulon in Chlamydia-infected endocervical epithelial cells. This may necessitate the development of a YAP-knockout cell line, given the relative inefficiency of siRNA-mediated knockdown we and others observe in End1/E6E7 cells (81).

It is important to note that chlamydial fibrosis is clinically associated with chronic or repeated infections, with the latter demonstrably increasing the risk of scarring infertility and/or ectopic pregnancy (6). Critically, the lack of a viable and tractable animal model of C. trachomatis-associated scarring has heretofore precluded more longitudinal study of the pro-fibrotic effects of chronic or repeated C. trachomatis infection. Given our observation that Chlamydia-infected cells stimulate cocultured fibroblast expression of collagen, these data suggest intriguing possibilities: that infected epithelial cells sensitize affected tissues to other pro-fibrotic stimuli or that repeated infections at the same site have an aggregate effect in stimulating scar-associated tissue remodeling. In a similar fashion, collagen deposition facilitated by infected epithelial cells may itself act as a stimulus of myofibroblast activation. Ultimately, the capacity of coculture-stimulated fibroblasts to return to a naïve, collagen-repressed state must be assessed, as must the potential for successive rounds of coculture to enhance fibroblast activation and collagen synthesis. Further refinement of the in vitro model of chlamydial pathogenesis presented here may provide critical insight into how the pro-fibrotic stimulus of infected epithelial cells may effectively persist after host cell death and pathogen clearance, thereby facilitating the tissue-level scarring associated with chronic or persistent infection.

MATERIALS AND METHODS

Eukaryotic cell culture

Primary HCECs (ATCC PCS-0480–011, lot 80306190) were cultured at 37°C with 5% atmospheric CO2 in cervical epithelial cell basal medium (CECBM) (ATCC PCS-480-032) supplemented with all contents of a cervical epithelial growth kit (ATCC PCS-080-042), per manufacturer’s instructions. Primary HVEs (ATCC PCS-480-010, lot 80924222) were cultured at 37°C with 5% atmospheric CO2 in vaginal epithelial cell basal medium (VECBM) (ATCC PCS-480-030) supplemented with all contents of a vaginal epithelial cell growth kit (ATCC PCS-480-040), per manufacturer’s instructions. Human endocervical epithelial HPV-16 E6/E7-immortalized End1s (End1 E6/E7, ATCC CRL-2615) were cultured at 37°C with 5% atmospheric CO2 in keratinocyte serum-free medium (KSFM) (Thermo Fisher Scientific) supplemented with human recombinant epidermal growth factor, bovine pituitary extract, 5 µg/mL gentamicin, and 0.4 mM CaCl2 (unless otherwise indicated). Human endometrial hTERT-immortalized fibroblasts (KCO2-44D hTERT, ATCC SC-6000) and McCoy B mouse fibroblasts (McCoy, ATCC CRL-1696) were cultured in Dulbecco’s modification of Eagle’s medium (DMEM) (Thermo Fisher Scientific 11960069) supplemented with 10% fetal bovine serum (Fisher Scientific NC0327704), 2 mM L-glutamine (Thermo Fisher Scientific 25030081), and 5 µg/mL gentamicin. End1s, KCO2s, and McCoys were cultured at 37°C in 5% CO2, between passages 3 and 15. All cell lines were tested annually for mycoplasma contamination, using the ATCC PCR-based mycoplasma detection kit (ATCC 301-1012K), per manufacturer’s instructions.

Chlamydial infections

Chlamydia trachomatis serovars L2 (434/Bu) and D were originally obtained from Dr. Ted Hackstadt (Rocky Mountain National Laboratory, NIAID). Chlamydial EBs were isolated from infected, cycloheximide-treated McCoy cells at 36–40 hpi and purified by density gradient centrifugation as previously described (95). For infection of 6-well plates (Greiner Bio-One 657160 and 662160) or 12-well plates (Costar 3513), HCECs or HVEs seeded at 5,000 cells/cm2 and grown to confluence (6–7 days) or End1s seeded at 125% of confluence and incubated for 16 hours were washed with pre-warmed Hanks buffered saline solution (HBSS) prior to inoculation with Chlamydia-containing CECBM, VECBM, or KSFM (respectively) at the indicated multiplicity of infection (MOI). To synchronize infection of cultured monolayers, tissue culture plates were centrifuged at 4°C and 500 rcf (Eppendorf 5810 R tabletop centrifuge, A-4-81 rotor) for 15 minute to permit transient electrostatic adherence of C. trachomatis to the host cell surface. Inoculum was then aspirated, and cells were washed with chilled HBSS to remove any non-adherent EBs. After aspiration of the wash, chlamydial invasion was initiated via the addition of CECBM, VECBM, or KSFM (HCECs, HVEs, or End1s respectively) pre-warmed to 37°C. Infected cultures were then incubated for 24 hours at 37°C and 5% CO2 for subsequent harvesting of total RNA (see below).

For infection of End1/KCO2 same-well co-cultures, End1s were seeded at 35% of confluence on six-well tissue culture plates (Greiner Bio-One 657160), incubated for 16 hours, then infected at an MOI of 5 as described above. Infected End1 cultures were returned to a tissue culture incubator until 1 hpi, at which time cultures were washed three times with pre-warmed HBSS containing 100 µg/mL heparin (Sigma-Aldrich H3393) to remove any transiently adhered, non-internalized EBs (96). After aspiration of the final wash, KCO2s in complete DMEM were seeded in each well at 75% of confluence. Tissue culture plates were subsequently incubated until 24 hpi prior to harvest for single-cell RNA library preparation (see below).

For transwell coculture experiments, End1s seeded at 100% of confluence in six-well-sized transwell inserts (Costar 3450, Fisher Scientific) and incubated for 16 hours (or 40 hours, in the case of experiments involving siRNA transfection) were infected at an MOI of 5 as above. At 1 hpi, cells were washed with HBSS containing 100 µg/mL heparin (Millipore-Sigma H3393) to remove transiently adhered chlamydial organisms. After replacement of wash media with DMEM, transwells were subsequently transferred to six-well plates containing KCO2s seeded at 60% of confluence immediately after infection. Cocultures were then incubated for 24 hours prior to fixation or RNA harvest (see below).

Bulk RNA sequencing and analysis

HVEs infected at an MOI of 2 as described above were harvested for RNA at 24 hpi using TRIzol (Thermo Fisher Scientific 15596026) and the DNA-free DNA removal kit (Thermo Fisher Scientific AM1906), according to manufacturers’ protocols. Total RNA samples were subsequently assayed for fragmentation using an Agilent Bioanalyzer 2100. Polyadenylated transcript enrichment and cDNA library preparation of intact samples were performed using the NuGEN Universal mRNA-Seq Library Preparation kit, and sequencing of cDNA libraries was performed using the NextSeq 550 system (Illumina). HCEC and End1 libraries were prepared as described previously (12).

Read alignment and downstream analysis was performed using CLC Genomics Workbench (Qiagen); each treatment group was comprised of libraries from three biological replicates, each with a minimum of 30 million reads (unstranded single read, mean length 150 bp); and genes with an FDRP ≤ 0.05 were considered differentially expressed. Functional characterization of each differentially expressed gene set was performed using the STRING database of protein-protein association networks, using the ranked protein list functional enrichment analysis function (31, 32). GO/Wikipathway overrepresentation analysis was also performed in R, using the Bioconductor package clusterProfiler (97). Pearson’s correlation coefficients between HCEC, End1, and HeLa expressions of the fibrosis-associated gene set identified by clusterProfiler were subsequently calculated in R, using log2-transformed fold changes of all genes differentially expressed in at least one data set. Sequencing data for mock-infected and Ct L2-infected HVEs are available in the Gene Expression Omnibus, accession GSE228774; sequencing data for the equivalent HCEC and End1 infections are also available in GEO as part of our recent publication (accession no. GSE180784) (12).

Single-cell library preparation and sequencing

Infected cocultures were harvested as single-cell suspensions via the 10× Genomics Sample Preparation Demonstrated Protocol (Revision B). Briefly, cocultures were disassociated in 0.25% Trypsin-EDTA and then harvested in complete DMEM. After centrifugation and resuspension in complete DMEM to eliminate residual trypsin, cell suspensions were filtered over 30-µm cell strainers (Miltenyi Biotec, 130-098-458) to eliminate clustered cells, washed twice in 1× phosphate-buffered saline (PBS) containing 0.04% bovine serum albumin (BSA), then filtered again using Flowmi 40 µM cell strainers (Bel-Art H13680-0040). After staining, aliquots of each suspension with trypan blue were used to validate a minimum of 95% cell viability for each sample as well as to determine suspension density for controlling cell library size. Suspensions were stored on ice for library preparation according to the 10× single-cell protocol (10× Genomics, Chromium Single Cell 3’ v2 Chemistry). cDNA libraries were submitted to the UNMC Genomics Core facility for molarity quantification via qPCR, followed by sequencing using the NextSeq 550 platform and two 150-cycle High-Output flow cells. Sequencing data for this experiment are also available on GEO, accession no. GSE228647.

Single-cell RNA sequencing analysis

Sequencing data were demultiplexed and aligned to the hg38 reference genome via CellRanger (10× Genomics, version 6.0.1), using an expected cell count of 1,000 informed by hemacytometer counts performed on an aliquot of the single-cell suspension. After aggregating the mock- and Ct L2-infected alignment results for each replicate (n = 2), initial cell type determination was performed in Loupe Cell Browser (10× Genomics, version 3.1.1). Epithelial cell (End1) and fibroblast (KCO2) populations were identified on the basis of keratin 19 and vimentin expression, respectively. Subclustering analysis of the epithelial/fibroblast populations was performed in the Seurat R package (98). After associating per-cell expression data with their respective treatment (mock or Ct L2 infected) and cell type (epithelial cell or fibroblast) metadata, per-cell expression data were separated into separate End1/KCO2 Seurat objects.

Subsequent quality control and dimensionality reduction were performed analogously to the Seurat guided vignette on single-cell expression analysis. After trimming cells with extreme unique feature counts (200 < n < 8,000), outlier features were identified using the vst method (local polynomial regression of log-transformed mean/variance, selecting top 2,000 features). The dimensionality of each data set was determined via the Seurat JackStraw method; for both the End1 and KCO2 cell populations, the top 15 principal components encompassed the majority of variance in the data. Dimensionality reduction via uniform manifold approximation and projection was then performed on each data set, using their respective top 15 principal components, followed by cluster identification via Seurat’s shared nearest neighbor algorithm (using a clustering resolution of 0.5). Per-cluster differential expression for each cluster was then determined via the FindMarkers function, using a cutoff of genes expressed by at least 25% of cells in each compared population. Per-cell and per-cluster gene expression was visualized using the Seurat DimPlot, FeaturePlot, and VlnPlot functions, splitting plots across treatment groups or clusters where applicable.

Immunofluorescence microscopy

For assessment of chlamydial titer in infected HCECs, HVEs, and End1s, confluent monolayers of each cell type were infected with C. trachomatis serovar L2 in 12-well plates as described above, then harvested at 36 hpi via scraping. Host cells were then lysed via centrifugation (20,000 rcf for 30 min at 4°C); pelleted chlamydial organisms and host cell debris were resuspended in 100-µL SPG buffer (219 mM sucrose, 10 mM Na2HPO4, 3.8 mM KH2PO4, 5 mM glutamic acid) for storage at −80°C. Aliquots of each lysate were diluted serially 1:100 and 1:1,000 in HBSS for infection of McCoy cells seeded in triplicate on glass cover slips and then infected via centrifugation as described above.

For assessment of YAP nuclear translocation and fibroblast collagen expression, HCECs, HVEs, and End1s/KCO2s were seeded on glass cover slips (VWR) and infected as described above. At 24 hpi, McCoys, HCECs, HVEs, End1s, and/or KCO2s were fixed in 4% paraformaldehyde in PBS for 10 minutes at 37°C, washed in PBS, then blocked in 5% BSA in PBS for 1 hour at room temperature. Fixed and blocked cover slips were subsequently incubated overnight at 4°C with one of the following primary antibodies/sera: human anti-Chlamydia, rabbit anti-YAP (CST 4912, 1:100 dilution), and rabbit anti-collagen I (Abcam ab260043, 1:250 dilution). Cover slips were again washed in PBS and then incubated for 1 hour at room temperature with the following fluorophore-conjugated antibodies/dyes in 1% BSA-PBS: goat anti-human Alexa-488 conjugate (Thermo Fisher Scientific A-11013, 1:1,000 dilution), goat anti-rabbit Alexa-488 conjugate (Thermo Fisher Scientific A-11034, 1:1,000 dilution), phalloidin Alexa-594 conjugate (Thermo Fisher Scientific A-12381, 1:120 dilution), and DAPI (Sigma-Aldrich 10236276001, 1:1,000 dilution). Afterward, cover slips were washed in PBS and ultrapure water and then mounted on microscope slides using Shandon Immu-Mount (Thermo Fisher Scientific 9990402).

At least five fields of each cover slip were imaged using an Axio Observer inverted fluorescent microscope (Zeiss, chlamydial titers) or CSU-W1 Spinning-Disk Confocal Microscope (Nikon, all other experiments). For titration of chlamydial IFUs, the number of inclusions (defined here as nuclei-adjacent structures exhibiting DAPI and anti-Chlamydia staining) was counted per field and averaged across technical triplicates to determine a mean count of IFUs/field for each lysate, proportional to total IFUs/well by a ratio of areas (1:947.94). The total number of IFUs per lysate was then calculated by dividing the estimated total IFUs by volume of inoculum added to each well (500 µL), the multiplying this result by the dilution factor of the original sample lysate (1:100 or 1:1,000) and the total volume of each lysate (100 µL). Statistical analysis was performed in R, using pairwise Student’s t-tests and Bonferroni’s correction for multiple comparisons; P-values less than 0.05 were considered statistically significant.

For measurement of YAP nuclear translocation, 10 nuclei were selected at random per field using only the DAPI channel, manually masking the nuclear area, and recording the mean YAP fluorescence intensity per nucleus. Blinded image quantification was performed by assigning image filenames randomized number codes. To account for variation in total YAP between cells, staining efficiency between cover slips, and compression of the nuclear/cytosolic compartments by the chlamydial inclusion, 10 cytosolic regions not occluded by an inclusion body and adjacent to measured nuclei were selected per field, with the mean YAP fluorescence intensity of these regions averaged to produce a per-field measurement of mean cytosolic YAP fluorescence intensity; nuclear translocation of YAP was thereby expressed as a ratio of mean nuclear fluorescence to mean cytosolic fluorescence.

For measurement of KCO2 collagen I expression, a selection mask of all cell area per field was defined via fluorescence thresholding of the phalloidin counterstain in ImageJ; per-field measurements of mean collagen I fluorescence intensity were subsequently recorded using this selection mask. Blinded image quantification was performed by assigning image filenames randomized number codes. Statistical analysis of YAP nuclear translocation and fibroblast collagen expression was performed in R, using a Kruskal-Wallis test to first verify a statistically significant (P-value < 0.05) difference between treatment groups. Subsequent pairwise comparisons were performed using a Wilcoxon rank sum test and Bonferroni’s correction for multiple comparisons, with P-values less than 0.05 being considered statistically significant.

siRNA transfection

End1s were transfected with either an ON-TARGETplus non-targeting siRNA pool (Horizon Discovery D-001810-10-05) or an ON-TARGETplus YAP1-targeting siRNA SMARTpool (Horizon Discovery L-012200-00-0005) using Lipofectamine 3000 (Thermo Fisher Scientific L3000008), per manufacturer’s instructions, at an empirically determined optimal concentration of 10 nM. At 16 hours post-seeding of End1s at 125% of confluence on six-well transwell inserts as described above, siRNA was combined in Opti-MEM (Thermo Fisher Scientific 31985062) with the Lipofectamine 3000 reagent, incubated for 5 minutes at room temperature to allow for liposome formation, then added to wells dropwise with mixing. Transfected End1s were then incubated for 24 hours prior to infection with Chlamydia as described above.

SDS-PAGE and Western blotting

To minimize activity of the chlamydial protease CPAF, End1s seeded on six-well plates, transfected with siRNA, and then infected at an MOI of 5 as described above were subsequently lysed in 1% SDS buffer heated to 95°C, as previously described (99). After treatment with Pierce Universal Nuclease (Thermo Fisher Scientific, 1:1,000 dilution) for 5 minutes at room temperature, lysates were combined with 4× Laemmli Sample Buffer (Bio-Rad 1610747) for loading on a 10% acrylamide/bis-acrylamide gel for SDS-PAGE (1.5 hour, 100 V). Gels were then transferred to PVDF membranes (Bio-Rad 1620177) using a semi-dry transfer method (50 minutes, 20 V). After blocking in 5% BSA in PBST (PBS containing 0.1% Tween-20) for 1 hour at room temperature, membranes were incubated overnight at 4°C with primary antibodies in 5% BSA-PBST: rabbit anti-YAP (CST 4912, 1:1,000 dilution) and rabbit anti-GAPDH (CST 2118, 1:1,000 dilution). Membranes were subsequently washed in PBST and then incubated with a goat anti-rabbit HRP-conjugated secondary antibody (Dako P0448, 1:2,000 dilution in 5% BSA-PBST) for 2 hours at room temperature. After additional washing in PBST, membranes were imaged using Immobilon HRP Substrate (Millipore Sigma WBKLS0500) or an Azure Biosystems c600. Images were analyzed using the ImageJ gel analysis tool to quantify the fluorescence density of total YAP total protein relative to the GAPDH loading control.

Reverse transcription quantitative PCR

For assessment of host cell permissiveness to chlamydial infection, End1s, HCECs, and HVEs were seeded or grown to confluence on 12-well plates and infected with C. trachomatis serovar L2 at an MOI of 2, as described above. Wells were subsequently harvested for isolation of host and chlamydial genomic DNA (gDNA) at 0, 8, 16, 24, and 36 hpi, using the DNeasy Blood & Tissue kit (QIAGEN 69506), per manufacturer’s instructions for adherent cells. After confirming recovered gDNA concentration and purity using a NanoDrop One spectrophotometer (Thermo Fisher Scientific), the amount of chlamydial gDNA in each sample was determined via quantitative PCR. Briefly, 20-ng aliquots of each sample were tested in triplicate against a standard curve of 2 pg–20 ng C. trachomatis serovar L2 gDNA, prepared via 1:10 serial dilution (R2 ≥0.995). Quantitative PCR of the C. trachomatis serovar L2 gene euo was performed on a QuantStudio 3 (Thermo Fisher Scientific) using PowerUp SYBR Green (Thermo Fisher Scientific A25742) and the following primers: 5′-GCTGTTCCTGTTACTTCGCAAA-3′ (euo_F), 5′-AACATAGATAGCCTGACGAGTCACA-3′ (euo_R).

For measurement of EMT-associated gene expression, End1s were seeded on six-well plates (Corning 354402) at 100% of confluence and infected at an MOI of 5 as described above. At 24 hpi, RNA was harvested using TRIzol (Thermo Fisher Scientific 15596026) and the DNA-free DNA removal kit (Thermo Fisher Scientific AM1906), according to manufacturers’ protocols. For measurement of collagen I and α-SMA expression of cocultured fibroblasts, KCO2s seeded on six-well plates (Corning 354402) and cocultured as described above were harvested for RNA, again using TRIzol and the DNA-free DNA removal kit. cDNA libraries were subsequently prepared using SuperScript IV Reverse Transcriptase (Thermo Fisher Scientific 11766050) according to the manufacturer’s protocol. Quantitative PCR was performed on a QuantStudio 3 (Thermo Fisher Scientific) using TaqMan assay kits (Thermo Fisher Scientific) of the following genes: CDH1 (Hs01023895_m1), CDH2 (Hs00983056_m1), VIM (Hs00958111_m1), KRT13 (Hs02558881_s1), SNAI1 (Hs00195591_m1), SNAI2 (Hs00161904_m1), ZEB1 (Hs01566408_m1), ZEB2 (Hs00207691_m1), COL1A1, (Hs00164004_m1), ACTA2 (Hs00426835_g1), and the housekeeping gene HPRT (Hs02800695_m1). Statistical analysis was performed in R, using pairwise Student’s t-tests and Bonferroni’s correction for multiple comparisons; P-values less than 0.05 were considered statistically significant.

ACKNOWLEDGMENTS

The authors acknowledge the members of the Carabeo laboratory for their critical feedback in the development of this research. We also acknowledge the assistance of Maggie Sladek and Megan Otte in performing coculture experiments, Dr. Aileen Helsel and Dr. Nathan Law in single-cell RNA sequencing experimental design, Bianca Lopez-Biladeau for single-cell RNA library preparation, and the UNMC DNA Sequencing Core in bulk mRNA library preparation and bulk/single-cell RNA sequencing.

This work was supported by NIAID grant R01 AI065545 to R.C. The University of Nebraska DNA Sequencing Core receives partial support from the National Institute for General Medical Science (NIGMS) INBRE-P20GM103427-19 grant as well as The Fred & Pamela Buffett Cancer Center Support Grant P30 CA036727.

This publication’s contents are the sole responsibility of the authors and do not necessarily represent the official views of the NIH.

L.C., conceptualization, methodology, validation, formal analysis, investigation, data curation, writing – original draft, and visualization. R.C., conceptualization, methodology, resources, writing – review and editing, supervision, project administration, and funding acquisition.

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

Contributor Information

Rey Carabeo, Email: rey.carabeo@unmc.edu.

Hiutung Chu, University of California, San Diego, La Jolla, California, USA.

DATA AVAILABILITY

The bulk RNA sequencing data sets generated and analyzed for this study can be found in the Gene Expression Omnibus (100), GEO Series accession numbers GSE180784 (End1 and HCEC bulk RNA-sequencing), GSE228774 (HVE bulk RNA-sequencing), and GSE228647 (End1/KCO2 single-cell RNA-sequencing).

SUPPLEMENTAL MATERIAL

The following material is available online at https://doi.org/10.1128/msystems.00904-23.

Supplemental Data. msystems.00904-23-s0001.docx.

Captions for Tables S1 to S4; Fig. S1 to S3.

DOI: 10.1128/msystems.00904-23.SuF1
Table S1. msystems.00904-23-s0002.xlsx.

Summary of differential expression testing in bulk RNA-sequencing of HCECs/HVEs.

DOI: 10.1128/msystems.00904-23.SuF2
Table S2. msystems.00904-23-s0003.xlsx.

Summary of genes equivalently modulated by infection and steady-state expression in HCECs.

DOI: 10.1128/msystems.00904-23.SuF3
Table S3. msystems.00904-23-s0004.xlsx.

Summary of transcription factor enrichment analysis of equivalently modulated gene set.

DOI: 10.1128/msystems.00904-23.SuF4
Table S4. msystems.00904-23-s0005.xlsx.

List of differentially expressed genes.

DOI: 10.1128/msystems.00904-23.SuF5

ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

  • 1. Patel CG, Trivedi S, Tao G. 2018. The proportion of young women tested for chlamydia who had urogenital symptoms in physician offices. Sex Transm Dis 45:e72–e74. doi: 10.1097/OLQ.0000000000000858 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Haggerty CL, Gottlieb SL, Taylor BD, Low N, Xu F, Ness RB. 2010. Risk of sequelae after Chlamydia trachomatis genital infection in women. J Infect Dis 201 Suppl 2:S134–55. doi: 10.1086/652395 [DOI] [PubMed] [Google Scholar]
  • 3. Ness RB, Soper DE, Richter HE, Randall H, Peipert JF, Nelson DB, Schubeck D, McNeeley SG, Trout W, Bass DC, Hutchison K, Kip K, Brunham RC. 2008. Chlamydia antibodies, chlamydia heat shock protein, and adverse sequelae after pelvic inflammatory disease: the PID evaluation and clinical health (PEACH) study. Sex Transm Dis 35:129–135. doi: 10.1097/olq.0b013e3181557c25 [DOI] [PubMed] [Google Scholar]
  • 4. Chow JM, Yonekura ML, Richwald GA, Greenland S, Sweet RL, Schachter J. 1990. The association between Chlamydia trachomatis and ectopic pregnancy. A matched-pair, case-control study. JAMA 263:3164–3167. [PubMed] [Google Scholar]
  • 5. Passos LG, Terraciano P, Wolf N, Oliveira F de, Almeida I de, Passos EP. 2022. The correlation between Chlamydia trachomatis and female infertility: a systematic review. Rev Bras Ginecol Obstet 44:614–620. doi: 10.1055/s-0042-1748023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Davies B, Turner KME, Frølund M, Ward H, May MT, Rasmussen S, Benfield T, Westh H, Danish Chlamydia Study Group . 2016. Risk of reproductive complications following chlamydia testing: a population-based retrospective cohort study in Denmark. Lancet Infect Dis 16:1057–1064. doi: 10.1016/S1473-3099(16)30092-5 [DOI] [PubMed] [Google Scholar]
  • 7. Price MJ, Ades AE, Soldan K, Welton NJ, Macleod J, Simms I, DeAngelis D, Turner KM, Horner PJ. 2016. The natural history of Chlamydia trachomatis infection in women: a multi-parameter evidence synthesis. Health Technol Assess 20:1–250. doi: 10.3310/hta20220 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Rasmussen SJ, Eckmann L, Quayle AJ, Shen L, Zhang YX, Anderson DJ, Fierer J, Stephens RS, Kagnoff MF. 1997. 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 99:77–87. doi: 10.1172/JCI119136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Dessus-Babus S, Darville TL, Cuozzo FP, Ferguson K, Wyrick PB. 2002. Differences in innate immune responses (in vitro) to HeLa cells infected with nondisseminating serovar E and disseminating serovar L2 of Chlamydia trachomatis. Infect Immun 70:3234–3248. doi: 10.1128/IAI.70.6.3234-3248.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Coombes BK, Mahony JB. 2001. cDNA array analysis of altered gene expression in human endothelial cells in response to chlamydia pneumoniae infection. Infect Immun 69:1420–1427. doi: 10.1128/IAI.69.3.1420-1427.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Patel AL, Chen X, Wood ST, Stuart ES, Arcaro KF, Molina DP, Petrovic S, Furdui CM, Tsang AW. 2014. Activation of epidermal growth factor receptor is required for Chlamydia trachomatis development. BMC Microbiol 14:277. doi: 10.1186/s12866-014-0277-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Caven LT, Brinkworth AJ, Carabeo RA. 2023. Chlamydia trachomatis induces the transcriptional activity of host YAP in a Hippo-independent fashion. Front Cell Infect Microbiol 13:1098420. doi: 10.3389/fcimb.2023.1098420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Igietseme JU, Partin J, George Z, Omosun Y, Goldstein J, Joseph K, Ellerson D, Eko FO, Pohl J, Bandea C, Black CM. 2020. Epidermal growth factor receptor and transforming growth factor Β signaling pathways cooperate to mediate chlamydia pathogenesis. Infect Immun 88:e00819-19. doi: 10.1128/IAI.00819-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Carratelli CR, Paolillo R, Rizzo A. 2007. Chlamydia pneumoniae stimulates the proliferation of HUVEC through the induction of VEGF by THP-1. Int Immunopharmacol 7:287–294. doi: 10.1016/j.intimp.2006.10.007 [DOI] [PubMed] [Google Scholar]
  • 15. Paolillo R, Iovene MR, Romano Carratelli C, Rizzo A. 2012. Induction of VEGF and MMP-9 expression by toll-like receptor 2/4 in human endothelial cells infected with chlamydia pneumoniae. Int J Immunopathol Pharmacol 25:377–386. doi: 10.1177/039463201202500207 [DOI] [PubMed] [Google Scholar]
  • 16. Igietseme Joseph U., Omosun Y, Stuchlik O, Reed MS, Partin J, He Q, Joseph K, Ellerson D, Bollweg B, George Z, Eko FO, Bandea C, Liu H, Yang G, Shieh W-J, Pohl J, Karem K, Black CM, Seshu J. 2015. Role of epithelial-mesenchyme transition in chlamydia pathogenesis. PLOS ONE 10:e0145198. doi: 10.1371/journal.pone.0145198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Igietseme JU, Omosun Y, Nagy T, Stuchlik O, Reed MS, He Q, Partin J, Joseph K, Ellerson D, George Z, Goldstein J, Eko FO, Bandea C, Pohl J, Black CM. 2018. Molecular pathogenesis of chlamydia disease complications: epithelial-mesenchymal transition and fibrosis. Infect Immun 86:e00585-17. doi: 10.1128/IAI.00585-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Rajić J, Inic-Kanada A, Stein E, Dinić S, Schuerer N, Uskoković A, Ghasemian E, Mihailović M, Vidaković M, Grdović N, Barisani-Asenbauer T. 2017. Chlamydia trachomatis infection is associated with E-cadherin promoter methylation, downregulation of E-cadherin expression, and increased expression of fibronectin and α-SMA-implications for epithelial-mesenchymal transition. Front Cell Infect Microbiol 7:253. doi: 10.3389/fcimb.2017.00253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Marconi GD, Fonticoli L, Rajan TS, Pierdomenico SD, Trubiani O, Pizzicannella J, Diomede F. 2021. Epithelial-mesenchymal transition (EMT): the type-2 EMT in wound healing, tissue regeneration and organ fibrosis. Cells 10:1587. doi: 10.3390/cells10071587 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Humphrys MS, Creasy T, Sun Y, Shetty AC, Chibucos MC, Drabek EF, Fraser CM, Farooq U, Sengamalay N, Ott S, Shou H, Bavoil PM, Mahurkar A, Myers GSA. 2013. Simultaneous transcriptional profiling of bacteria and their host cells. PLOS ONE 8:e80597. doi: 10.1371/journal.pone.0080597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Porcella SF, Carlson JH, Sturdevant DE, Sturdevant GL, Kanakabandi K, Virtaneva K, Wilder H, Whitmire WM, Song L, Caldwell HD, Morrison RP. 2015. Transcriptional profiling of human epithelial cells infected with plasmid-bearing and plasmid-deficient Chlamydia trachomatis. Infect Immun 83:534–543. doi: 10.1128/IAI.02764-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Gallucci RM, Lee EG, Tomasek JJ. 2006. IL-6 modulates alpha-smooth muscle actin expression in dermal fibroblasts from IL-6-deficient mice. J Invest Dermatol 126:561–568. doi: 10.1038/sj.jid.5700109 [DOI] [PubMed] [Google Scholar]
  • 23. Yang L, Herrera J, Gilbertsen A, Xia H, Smith K, Benyumov A, Bitterman PB, Henke CA. 2018. IL-8 mediates idiopathic pulmonary fibrosis mesenchymal progenitor cell fibrogenicity. Am J Physiol Lung Cell Mol Physiol 314:L127–L136. doi: 10.1152/ajplung.00200.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Xu H, Liu L, Cong M, Liu T, Sun S, Ma H, You H, Jia J, Wang P. 2020. EGF neutralization antibodies attenuate liver fibrosis by inhibiting Myofibroblast proliferation in bile duct ligation mice. Histochem Cell Biol 154:107–116. doi: 10.1007/s00418-020-01867-9 [DOI] [PubMed] [Google Scholar]
  • 25. Lipson KE, Wong C, Teng Y, Spong S. 2012. CTGF is a central mediator of tissue remodeling and fibrosis and its inhibition can reverse the process of fibrosis. Fibrogenesis Tissue Repair 5. doi: 10.1186/1755-1536-5-S1-S24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Bochaton-Piallat M-L, Gabbiani G, Hinz B. 2016. The myofibroblast in wound healing and fibrosis: answered and unanswered questions. F1000Res 5:F1000 Faculty Rev-752. doi: 10.12688/f1000research.8190.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Faris R, Andersen SE, McCullough A, Gourronc F, Klingelhutz AJ, Weber MM. 2019. Chlamydia trachomatis serovars drive differential production of proinflammatory cytokines and chemokines depending on the type of cell infected. Front Cell Infect Microbiol 9:399. doi: 10.3389/fcimb.2019.00399 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Anderson DJ, Marathe J, Pudney J. 2014. The structure of the human vaginal stratum corneum and its role in immune defense. Am J Reprod Immunol 71:618–623. doi: 10.1111/aji.12230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Nogueira AT, Braun KM, Carabeo RA. 2017. Characterization of the growth of Chlamydia trachomatis in in vitro-generated stratified epithelium. Front Cell Infect Microbiol 7:438. doi: 10.3389/fcimb.2017.00438 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Moorman DR, Sixbey JW, Wyrick PBY. 1986. Interaction of Chlamydia trachomatis with human genital epithelium in culture. Microbiology 132:1055–1067. doi: 10.1099/00221287-132-4-1055 [DOI] [PubMed] [Google Scholar]
  • 31. Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, Doncheva NT, Legeay M, Fang T, Bork P, Jensen LJ, von Mering C. 2021. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res 49:D605–D612. doi: 10.1093/nar/gkaa1074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, Jensen LJ, Mering C von. 2019. STRING V11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47:D607–D613. doi: 10.1093/nar/gky1131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Lamouille S, Xu J, Derynck R. 2014. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 15:178–196. doi: 10.1038/nrm3758 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Huang RY-J, Guilford P, Thiery JP. 2012. Early events in cell adhesion and polarity during epithelial-mesenchymal transition. J Cell Sci 125:4417–4422. doi: 10.1242/jcs.099697 [DOI] [PubMed] [Google Scholar]
  • 35. Zadora PK, Chumduri C, Imami K, Berger H, Mi Y, Selbach M, Meyer TF, Gurumurthy RK. 2019. Integrated phosphoproteome and transcriptome analysis reveals chlamydia-induced epithelial-to-mesenchymal transition in host cells. Cell Rep 26:1286–1302. doi: 10.1016/j.celrep.2019.01.006 [DOI] [PubMed] [Google Scholar]
  • 36. Manohar M, Kandikattu HK, Verma AK, Mishra A. 2018. IL-15 regulates fibrosis and inflammation in a mouse model of chronic pancreatitis. Am J Physiol Gastrointest Liver Physiol 315:G954–G965. doi: 10.1152/ajpgi.00139.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Chiu Y-S, Wei C-C, Lin Y-J, Hsu Y-H, Chang M-S. 2014. IL-20 and IL-20R1 antibodies protect against liver fibrosis. Hepatology 60:1003–1014. doi: 10.1002/hep.27189 [DOI] [PubMed] [Google Scholar]
  • 38. Belland RJ, Nelson DE, Virok D, Crane DD, Hogan D, Sturdevant D, Beatty WL, Caldwell HD. 2003. Transcriptome analysis of chlamydial growth during IFN--mediated persistence and reactivation. Proc Natl Acad Sci U S A 100:15971–15976. doi: 10.1073/pnas.2535394100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Mittelman D, Wilson JH. 2013. The fractured genome of HeLa cells. Genome Biol 14:111. doi: 10.1186/gb-2013-14-4-111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Masters JR. 2002. Hela cells 50 years on: the good, the bad and the ugly. Nat Rev Cancer 2:315–319. doi: 10.1038/nrc775 [DOI] [PubMed] [Google Scholar]
  • 41. Le Negrate G, Krieg A, Faustin B, Loeffler M, Godzik A, Krajewski S, Reed JC. 2008. Chladub1 of Chlamydia trachomatis suppresses NF-κB activation and inhibits IκBα ubiquitination and degradation. Cell Microbiol 10:1879–1892. doi: 10.1111/j.1462-5822.2008.01178.x [DOI] [PubMed] [Google Scholar]
  • 42. Krämer S, Crauwels P, Bohn R, Radzimski C, Szaszák M, Klinger M, Rupp J, van Zandbergen G, Ehrt S. 2015. AP-1 transcription factor serves as a molecular switch between Chlamydia pneumoniae replication and persistence. Infect Immun 83:2651–2660. doi: 10.1128/IAI.03083-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Keenan AB, Torre D, Lachmann A, Leong AK, Wojciechowicz ML, Utti V, Jagodnik KM, Kropiwnicki E, Wang Z, Ma’ayan A. 2019. ChEA3: transcription factor enrichment analysis by orthogonal omics integration. Nucleic Acids Res 47:W212–W224. doi: 10.1093/nar/gkz446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Bora-Singhal N, Nguyen J, Schaal C, Perumal D, Singh S, Coppola D, Chellappan S. 2015. YAP1 regulates OCT4 activity and SOX2 expression to facilitate self-renewal and vascular mimicry of stem-like cells. Stem Cells 33:1705–1718. doi: 10.1002/stem.1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Zeng C, Wu P-L, Dong Z-T, Li X, Zhou Y-F, Xue Q. 2020. YAP1 inhibits ovarian endometriosis stromal cell invasion through ESR2. Reproduction 160:481–490. doi: 10.1530/REP-19-0565 [DOI] [PubMed] [Google Scholar]
  • 46. Jia M, Li Q, Guo J, Shi W, Zhu L, Huang Y, Li Y, Wang L, Ma S, Zhuang T, Wang X, Pan Q, Wei X, Qin Y, Li X, Jin J, Zhi X, Tang J, Jing Q, Li S, Jiang L, Qu L, Osto E, Zhang J, Wang X, Yu B, Meng D. 2022. Deletion of BACH1 attenuates atherosclerosis by reducing endothelial inflammation. Circ Res 130:1038–1055. doi: 10.1161/CIRCRESAHA.121.319540 [DOI] [PubMed] [Google Scholar]
  • 47. He L, Pratt H, Gao M, Wei F, Weng Z, Struhl K. 2021. YAP and TAZ are transcriptional co-activators of AP-1 proteins and STAT3 during breast cellular transformation. Elife 10:e67312. doi: 10.7554/eLife.67312 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Szeto SG, Narimatsu M, Lu M, He X, Sidiqi AM, Tolosa MF, Chan L, De Freitas K, Bialik JF, Majumder S, Boo S, Hinz B, Dan Q, Advani A, John R, Wrana JL, Kapus A, Yuen DA. 2016. YAP/TAZ are mechanoregulators of TGF-β-smad signaling and renal fibrogenesis. J Am Soc Nephrol 27:3117–3128. doi: 10.1681/ASN.2015050499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Alder O, Cullum R, Lee S, Kan AC, Wei W, Yi Y, Garside VC, Bilenky M, Griffith M, Morrissy AS, Robertson GA, Thiessen N, Zhao Y, Chen Q, Pan D, Jones SJM, Marra MA, Hoodless PA. 2014. Hippo signaling influences HNF4A and FOXA2 enhancer switching during hepatocyte differentiation. Cell Rep 9:261–271. doi: 10.1016/j.celrep.2014.08.046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Pattschull G, Walz S, Gründl M, Schwab M, Rühl E, Baluapuri A, Cindric-Vranesic A, Kneitz S, Wolf E, Ade CP, Rosenwald A, von Eyss B, Gaubatz S. 2019. The Myb-MuvB complex is required for YAP-dependent transcription of mitotic genes. Cell Rep 27:3533–3546. doi: 10.1016/j.celrep.2019.05.071 [DOI] [PubMed] [Google Scholar]
  • 51. Gründl M, Walz S, Hauf L, Schwab M, Werner KM, Spahr S, Schulte C, Maric HM, Ade CP, Gaubatz S. 2020. Interaction of YAP with the Myb-MuvB (MMB) complex defines a transcriptional program to promote the proliferation of cardiomyocytes. PLoS Genet 16:e1008818. doi: 10.1371/journal.pgen.1008818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Zagurovskaya M, Shareef MM, Das A, Reeves A, Gupta S, Sudol M, Bedford MT, Prichard J, Mohiuddin M, Ahmed MM. 2009. EGR-1 forms a complex with YAP-1 and upregulates BAX expression in irradiated prostate carcinoma cells. Oncogene 28:1121–1131. doi: 10.1038/onc.2008.461 [DOI] [PubMed] [Google Scholar]
  • 53. Plouffe SW, Lin KC, Moore JL, Tan FE, Ma S, Ye Z, Qiu Y, Ren B, Guan K-L. 2018. The Hippo pathway effector proteins YAP and TAZ have both distinct and overlapping functions in the cell. J Biol Chem 293:11230–11240. doi: 10.1074/jbc.RA118.002715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Liu Y, Zhuo S, Zhou Y, Ma L, Sun Z, Wu X, Wang XW, Gao B, Yang Y. 2022. Yap-Sox9 signaling determines hepatocyte plasticity and lineage-specific hepatocarcinogenesis. J Hepatol 76:652–664. doi: 10.1016/j.jhep.2021.11.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Tamm C, Böwer N, Annerén C. 2011. Regulation of mouse embryonic stem cell self-renewal by a Yes–YAP–TEAD2 signaling pathway downstream of LIF. J Cell Sci 124:1136–1144. doi: 10.1242/jcs.075796 [DOI] [PubMed] [Google Scholar]
  • 56. Park J, Kim D-H, Shah SR, Kim H-N, Kim P, Quiñones-Hinojosa A, Levchenko A. 2019. Switch-like enhancement of epithelial-mesenchymal transition by YAP through feedback regulation of Wt1 and Rho-family Gtpases. Nat Commun 10:2797. doi: 10.1038/s41467-019-10729-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Bauzone M, Souidi M, Dessein A-F, Wisztorski M, Vincent A, Gimeno J-P, Monté D, Van Seuningen I, Gespach C, Huet G. 2021. Cross-talk between YAP and RAR-RXR drives expression of stemness genes to promote 5-FU resistance and self-renewal in colorectal cancer cells. Mol Cancer Res 19:612–622. doi: 10.1158/1541-7786.MCR-20-0462 [DOI] [PubMed] [Google Scholar]
  • 58. Bai N, Zhang C, Liang N, Zhang Z, Chang A, Yin J, Li Z, Luo N, Tan X, Luo N, Luo Y, Xiang R, Li X, Reisfeld RA, Stupack D, Lv D, Liu C. 2013. Yes-associated protein (YAP) increases chemosensitivity of hepatocellular carcinoma cells by modulation of p53. Cancer Biol Ther 14:511–520. doi: 10.4161/cbt.24345 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Nishioka N, Inoue K, Adachi K, Kiyonari H, Ota M, Ralston A, Yabuta N, Hirahara S, Stephenson RO, Ogonuki N, Makita R, Kurihara H, Morin-Kensicki EM, Nojima H, Rossant J, Nakao K, Niwa H, Sasaki H. 2009. The Hippo signaling pathway components lats and Yap pattern Tead4 activity to distinguish Mouse trophectoderm from inner cell mass. Dev Cell 16:398–410. doi: 10.1016/j.devcel.2009.02.003 [DOI] [PubMed] [Google Scholar]
  • 60. Miskolczi Z, Smith MP, Rowling EJ, Ferguson J, Barriuso J, Wellbrock C. 2018. Collagen abundance controls melanoma phenotypes through lineage-specific microenvironment sensing. Oncogene 37:3166–3182. doi: 10.1038/s41388-018-0209-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Labibi B, Bashkurov M, Wrana JL, Attisano L. 2020. Modeling the control of TGF-β/smad nuclear accumulation by the Hippo pathway effectors, Taz/Yap. iScience 23:101416. doi: 10.1016/j.isci.2020.101416 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Zhi X, Zhao D, Zhou Z, Liu R, Chen C. 2012. YAP promotes breast cell proliferation and survival partially through stabilizing the KLF5 transcription factor. Am J Pathol 180:2452–2461. doi: 10.1016/j.ajpath.2012.02.025 [DOI] [PubMed] [Google Scholar]
  • 63. Zhang X, Sun F, Qiao Y, Zheng W, Liu Y, Chen Y, Wu Q, Liu X, Zhu G, Chen Y, Yu Y, Pan Q, Wang J. 2017. TFCP2 is required for YAP-dependent transcription to stimulate liver malignancy. Cell Rep 21:1227–1239. doi: 10.1016/j.celrep.2017.10.017 [DOI] [PubMed] [Google Scholar]
  • 64. Carabeo RA, Grieshaber SS, Fischer E, Hackstadt T. 2002. Chlamydia trachomatis induces remodeling of the actin cytoskeleton during attachment and entry into Hela cells. Infect Immun 70:3793–3803. doi: 10.1128/IAI.70.7.3793-3803.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Carabeo RA, Grieshaber SS, Hasenkrug A, Dooley C, Hackstadt T. 2004. Requirement for the RAC GTPase in Chlamydia trachomatis invasion of non-phagocytic cells. Traffic 5:418–425. doi: 10.1111/j.1398-9219.2004.00184.x [DOI] [PubMed] [Google Scholar]
  • 66. Moulder JW. 1991. Interaction of chlamydiae and host cells in vitro. Microbiol Rev 55:143–190. doi: 10.1128/mr.55.1.143-190.1991 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Tiwari V, Maus E, Sigar IM, Ramsey KH, Shukla D. 2012. Role of heparan sulfate in sexually transmitted infections. Glycobiology 22:1402–1412. doi: 10.1093/glycob/cws106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Romero MD, Mölleken K, Hegemann JH, Carabeo RA. 2020. Chlamydia adhesion and invasion, . In Chlamydia biology: From genome to disease. Caister Academic Press. doi: 10.21775/9781912530281 [DOI] [Google Scholar]
  • 69. Hecker L, Jagirdar R, Jin T, Thannickal VJ. 2011. Reversible differentiation of Myofibroblasts by MyoD. Exp Cell Res 317:1914–1921. doi: 10.1016/j.yexcr.2011.03.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Lahmann I, Bröhl D, Zyrianova T, Isomura A, Czajkowski MT, Kapoor V, Griger J, Ruffault P-L, Mademtzoglou D, Zammit PS, Wunderlich T, Spuler S, Kühn R, Preibisch S, Wolf J, Kageyama R, Birchmeier C. 2019. Oscillations of myoD and Hes1 proteins regulate the maintenance of activated muscle stem cells. Genes Dev 33:524–535. doi: 10.1101/gad.322818.118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Zhang C, Chen D, Maguire EM, He S, Chen J, An W, Yang M, Afzal TA, Luong LA, Zhang L, Lei H, Wu Q, Xiao Q. 2018. Cbx3 inhibits vascular smooth muscle cell proliferation, migration, and neointima formation. Cardiovasc Res 114:443–455. doi: 10.1093/cvr/cvx236 [DOI] [PubMed] [Google Scholar]
  • 72. Wynn TA. 2008. Cellular and molecular mechanisms of fibrosis. J Pathol 214:199–210. doi: 10.1002/path.2277 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. McAnulty RJ. 2007. Fibroblasts and myofibroblasts: their source, function and role in disease. Int J Biochem Cell Biol 39:666–671. doi: 10.1016/j.biocel.2006.11.005 [DOI] [PubMed] [Google Scholar]
  • 74. Karsdal MA, Nielsen SH, Leeming DJ, Langholm LL, Nielsen MJ, Manon-Jensen T, Siebuhr A, Gudmann NS, Rønnow S, Sand JM, Daniels SJ, Mortensen JH, Schuppan D. 2017. The good and the bad collagens of fibrosis – their role in signaling and organ function. Adv Drug Deliv Rev 121:43–56. doi: 10.1016/j.addr.2017.07.014 [DOI] [PubMed] [Google Scholar]
  • 75. Schuppan D. 1990. Structure of the extracellular matrix in normal and fibrotic liver: collagens and glycoproteins. Semin Liver Dis 10:1–10. doi: 10.1055/s-2008-1040452 [DOI] [PubMed] [Google Scholar]
  • 76. Kuhn C, McDonald JA. 1991. The roles of the myofibroblast in idiopathic pulmonary fibrosis. ultrastructural and immunohistochemical features of sites of active extracellular matrix synthesis. Am J Pathol 138:1257–1265. [PMC free article] [PubMed] [Google Scholar]
  • 77. Kuhn C, Boldt J, King TE, Crouch E, Vartio T, McDonald JA. 1989. An Immunohistochemical study of architectural remodeling and connective tissue synthesis in pulmonary fibrosis. Am Rev Respir Dis 140:1693–1703. doi: 10.1164/ajrccm/140.6.1693 [DOI] [PubMed] [Google Scholar]
  • 78. Desmoulière A, Geinoz A, Gabbiani F, Gabbiani G. 1993. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J Cell Biol 122:103–111. doi: 10.1083/jcb.122.1.103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Lijnen P, Petrov V. 2002. Transforming growth factor-beta 1-induced collagen production in cultures of cardiac fibroblasts is the result of the appearance of myofibroblasts. Methods Find Exp Clin Pharmacol 24:333–344. doi: 10.1358/mf.2002.24.6.693065 [DOI] [PubMed] [Google Scholar]
  • 80. Petrov VV, Fagard RH, Lijnen PJ. 2002. Stimulation of collagen production by transforming growth factor-β1 during differentiation of cardiac fibroblasts to myofibroblasts. Hypertension 39:258–263. doi: 10.1161/hy0202.103268 [DOI] [PubMed] [Google Scholar]
  • 81. Govender Y, Avenant C, Verhoog NJD, Ray RM, Grantham NJ, Africander D, Hapgood JP. 2014. The Injectable-only contraceptive medroxyprogesterone acetate, unlike norethisterone acetate and progesterone, regulates inflammatory genes in endocervical cells via the glucocorticoid receptor. PLoS One 9:e96497. doi: 10.1371/journal.pone.0096497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. De Clercq E, Kalmar I, Vanrompay D. 2013. Animal models for studying female genital tract infection with Chlamydia trachomatis. Infect Immun 81:3060–3067. doi: 10.1128/IAI.00357-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Sturdevant GL, Caldwell HD. 2014. Innate immunity is sufficient for the clearance of Chlamydia trachomatis from the female mouse genital tract. Pathog Dis 72:70–73. doi: 10.1111/2049-632X.12164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Rajeeve K, Sivadasan R. 2020. Transcervical Mouse infections with Chlamydia trachomatis and determination of bacterial burden. Bio Protoc 10:e3506. doi: 10.21769/BioProtoc.3506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Pal S, Tifrea DF, Zhong G, de la Maza LM. 2018. Transcervical inoculation with Chlamydia trachomatis induces infertility in HLA-Dr4 transgenic and wild-type mice. Infect Immun 86:e00722-17. doi: 10.1128/IAI.00722-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Gondek DC, Olive AJ, Stary G, Starnbach MN. 2012. CD4+ T cells are necessary and sufficient to confer protection against Chlamydia trachomatis infection in the murine upper genital tract. J Immunol 189:2441–2449. doi: 10.4049/jimmunol.1103032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Zhao B, Ye X, Yu J, Li L, Li W, Li S, Yu J, Lin JD, Wang C-Y, Chinnaiyan AM, Lai Z-C, Guan K-L. 2008. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev 22:1962–1971. doi: 10.1101/gad.1664408 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Garrett Q, Khaw PT, Blalock TD, Schultz GS, Grotendorst GR, Daniels JT. 2004. Involvement of CTGF in TGF-β1-stimulation of myofibroblast differentiation and collagen matrix contraction in the presence of mechanical stress. Invest Ophthalmol Vis Sci 45:1109–1116. doi: 10.1167/iovs.03-0660 [DOI] [PubMed] [Google Scholar]
  • 89. Tsai C-C, Wu S-B, Kau H-C, Wei Y-H. 2018. Essential role of connective tissue growth factor (CTGF) in transforming growth factor-β1 (TGF-β1)-Induced myofibroblast transdifferentiation from graves' orbital fibroblasts. Sci Rep 8:7276. doi: 10.1038/s41598-018-25370-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Wang J, Song T, Zhou S, Kong X. 2019. YAP promotes the malignancy of endometrial cancer cells via regulation of IL-6 and IL-11. Mol Med 25:32. doi: 10.1186/s10020-019-0103-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Mia MM, Cibi DM, Ghani S, Singh A, Tee N, Sivakumar V, Bogireddi H, Cook SA, Mao J, Singh MK. 2022. Loss of Yap/Taz in cardiac fibroblasts attenuates adverse remodelling and improves cardiac function. Cardiovasc Res 118:1785–1804. doi: 10.1093/cvr/cvab205 [DOI] [PubMed] [Google Scholar]
  • 92. Deng F, Peng L, Li Z, Tan G, Liang E, Chen S, Zhao X, Zhi F. 2018. YAP triggers the Wnt/β-catenin signalling pathway and promotes enterocyte self-renewal, regeneration and tumorigenesis after DSS-induced injury. Cell Death Dis 9:153. doi: 10.1038/s41419-017-0244-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Piersma B, Bank RA, Boersema M. 2015. Signaling in fibrosis: TGF-, WNT, and YAP/TAZ converge. Front Med (Lausanne) 2:59. doi: 10.3389/fmed.2015.00059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Lam AP, Gottardi CJ. 2011. β-catenin signaling: a novel mediator of fibrosis and potential therapeutic target. Curr Opin Rheumatol 23:562–567. doi: 10.1097/BOR.0b013e32834b3309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Caldwell HD, Kromhout J, Schachter J. 1981. Purification and partial characterization of the major outer membrane protein of Chlamydia trachomatis. Infect Immun 31:1161–1176. doi: 10.1128/iai.31.3.1161-1176.1981 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Matyszak MK, Young JL, Gaston JSH. 2002. Uptake and processing of Chlamydia trachomatis by human dendritic cells. Eur J Immunol 32:742–751. doi: [DOI] [PubMed] [Google Scholar]
  • 97. Yu G, Wang L-G, Han Y, He Q-Y. 2012. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16:284–287. doi: 10.1089/omi.2011.0118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, Lee MJ, Wilk AJ, Darby C, Zager M, Hoffman P, Stoeckius M, Papalexi E, Mimitou EP, Jain J, Srivastava A, Stuart T, Fleming LM, Yeung B, Rogers AJ, McElrath JM, Blish CA, Gottardo R, Smibert P, Satija R. 2021. Integrated analysis of multimodal single-cell data. Cell 184:3573–3587. doi: 10.1016/j.cell.2021.04.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Johnson KA, Lee JK, Chen AL, Tan M, Sütterlin C. 2015. Induction and inhibition of CPAF activity during analysis of Chlamydia-infected cells. Pathog Dis 73:1–8. doi: 10.1093/femspd/ftv007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Edgar R, Domrachev M, Lash AE. 2002. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res 30:207–210. doi: 10.1093/nar/30.1.207 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supplemental Data. msystems.00904-23-s0001.docx.

Captions for Tables S1 to S4; Fig. S1 to S3.

DOI: 10.1128/msystems.00904-23.SuF1
Table S1. msystems.00904-23-s0002.xlsx.

Summary of differential expression testing in bulk RNA-sequencing of HCECs/HVEs.

DOI: 10.1128/msystems.00904-23.SuF2
Table S2. msystems.00904-23-s0003.xlsx.

Summary of genes equivalently modulated by infection and steady-state expression in HCECs.

DOI: 10.1128/msystems.00904-23.SuF3
Table S3. msystems.00904-23-s0004.xlsx.

Summary of transcription factor enrichment analysis of equivalently modulated gene set.

DOI: 10.1128/msystems.00904-23.SuF4
Table S4. msystems.00904-23-s0005.xlsx.

List of differentially expressed genes.

DOI: 10.1128/msystems.00904-23.SuF5

Data Availability Statement

The bulk RNA sequencing data sets generated and analyzed for this study can be found in the Gene Expression Omnibus (100), GEO Series accession numbers GSE180784 (End1 and HCEC bulk RNA-sequencing), GSE228774 (HVE bulk RNA-sequencing), and GSE228647 (End1/KCO2 single-cell RNA-sequencing).


Articles from mSystems are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES