Skip to main content
iScience logoLink to iScience
. 2025 Aug 28;28(10):113453. doi: 10.1016/j.isci.2025.113453

Unveiling Chlamydia trachomatis immunity in urogenital secretions: A systematic review

Anne Van Caesbroeck 1,4,, Marijana Lipovac 1, Eef van den Borst 1,2, Paolo Palma 1,3, Laura Téblick 1, Alex Vorsters 1
PMCID: PMC12478110  PMID: 41031366

Summary

Chlamydia trachomatis (CT) is a highly prevalent bacterial sexually transmitted infection (STI), associated with severe disease complications. CT targets a unique immunological environment: the genital tract epithelium. Although sampling the genital tract is challenging, previous studies have shown that genital CT-specific antibodies exhibit enhanced neutralizing capacity compared with serum antibodies. Furthermore, tissue-resident memory T (TRM) cells provide superior protection compared with circulating T cells. However, further research is required to identify correlates of protection and explore correlations between local and systemic responses. This review provides an overview of the sampling methods suitable for identifying mucosal immune biomarkers associated with CT infection, as well as the immunoassays used. We identified the microbiome, presence of coinfections, hormonal influences, genetics, and CT infection state, load, and genotype as confounding factors to be considered in trial design. Finally, we discuss challenges related to the detection of mucosal immune biomarkers and offer recommendations for future research.

Subject areas: Sexual medicine, Mucosal Immunity, Sampling

Graphical abstract

graphic file with name fx1.jpg

Highlights

  • Genital samples are a source of DNA and humoral and cellular immune biomarkers

  • Harmonization of sampling methods is essential

  • No validated immunoassays for urogenital samples are available


Sexual medicine; Mucosal Immunity; Sampling

Introduction

Genital Chlamydia trachomatis (CT) is a highly prevalent sexually transmitted infection (STI), with a global prevalence in 2020 estimated at 4.0% in women and 2.5% in men aged 15 to 49 years, affecting 69.9 million women and 58.6 million men.1,2,3 This represents a substantial economic and social burden.4 Despite being curable5 and preventable through consistent and correct use of barrier contraception methods,6,7 CT incidence continues to rise annually.1,8 The true scale of this epidemic is presumably underestimated, as CT is asymptomatic in over 70% of female and 50% of male infections.9,10,11 Moreover, opportunistic testing is largely limited to high-income countries12 and depends on national guidelines.13 Urogenital symptoms and sequelae can be severe, ranging from cervicitis and aberrant vaginal discharge to salpingitis, pelvic inflammatory disease, tubal factor infertility, ectopic pregnancy, and chronic pelvic pain in women and urethritis and epididymitis in men.9,14 It is clear that current screening and treatment programs are insufficient to contain this epidemic, highlighting the need for an effective preventive vaccine.

In STI research, a wide variety of sample types have been evaluated for collecting genital secretions. For women, these include cervical cytobrushes, cervicovaginal swabs, cervicovaginal lavage (CVL), first-void urine (FVU), menstrual cups, sponges, filter paper strips, and aspirate samples.15,16,17,18,19,20,21,22,23 For men, commonly used sample types include total ejaculate, seminal plasma, urine, expressed prostatic secretions (EPSs), and urethral swabs.24,25,26,27 Studies monitoring the impact of future STI vaccines could benefit from incorporating standardized site-specific biological samples. Mucosal samples are versatile, as most can be used for both DNA testing and immunological endpoints,28 supporting their use as a universal sample in vaccine research. Sample types such as vaginal swabs and urine are particularly advantageous because they can be collected at home, a factor that has been shown to improve participant acceptance and compliance in clinical trials.29,30,31 Additionally, site-specific samples provide insights on local antibodies and immune cells, which show distinct differences compared with systemic immune responses in terms of both abundance and functionality.32,33,34,35,36,37 The ideal sample type for evaluating CT mucosal immune responses in genital secretions has yet to be determined. It is evident that not all samples are universally suitable for every biomarker or immunoassay, underscoring a significant research gap in this area.

Hence, the objective of this review is to give a comprehensive overview of local mucosal immune biomarkers associated with CT infection and explore their association with systemic immune responses. Identified prognostic biomarkers may support future vaccine monitoring studies, since a correlate of protection is unknown. This review will discuss methods used for sampling the local urogenital environment with regard to identification of those biomarkers and outline potential confounding factors influencing the detection and quantification of CT-associated immune responses. Finally, different immunoassays used for monitoring local immune responses will be discussed.

Results

Study selection and characteristics

A total of 5,968 records were identified through database searches conducted in October 2023 (Figure 1). Duplicates were removed using EndNote. Consequently, 3,686 articles remained for screening. An additional 47 articles were identified through screening of reference lists and evaluated based on title and abstract. Of those, 43 were retrieved and screened for eligibility. A total of 142 articles were included after full-text screening. An updated search in February 2025 identified an additional 383 records, of which three were included in the final analysis.

Figure 1.

Figure 1

PRISMA flow diagram of the literature search

Records were identified through three databases (Ovid MEDLINE, Web of Science, and Scopus). After duplicate removal, records were screened by title and abstract and excluded if they did not match the inclusion criteria. Articles were then retrieved for full-text screening and included if they matched inclusion criteria. Reference lists of included articles were manually screened for further records that were not yet included through database search and screened based on title and abstract. Full texts were included when appropriate.

Of the 145 included publications, 85 reported on the detection of CT immune responses in female urogenital secretions, 54 in male urogenital secretions, and six included both male and female participants. The selected reports included cross-sectional studies (n = 81), case-control studies (n = 39), and cohort studies (n = 19). Remaining publication types comprised randomized controlled trials (n = 2), case reports and case series (n = 3), and letters to the editor (n = 1). In most studies, concerns were raised about the risk of bias. Individual risk of bias assessment scores are presented in Table S2. Meta-analysis was not performed due to data heterogeneity; instead, a narrative synthesis was conducted.

Urogenital secretions used for monitoring local immune responses

An overview of specific characteristics and applications of the various sample types is provided in Table 1. Female urogenital secretions were collected using the following methods: CVL,36,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60 cervical cytobrush,37,39,41,42,43,44,45,50,51,54,60,61,62,63,64,65,66,67,68,69,70,71 cervical swab,57,69,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88 sponge,37,49,66,89,90,91,92,93,94,95,96,97,98,99,100,101,102 urethrogenital swabs,107,108,109,110,111,112 vaginal swabs,93,103,104,105,106 vaginal filter paper strips,113,114,115,116 aspirate,63,114,117,118,119 menstrual cup,120,121 urine,72,75,176,177,178 and cervical scraping54 (Figure 2). Notably, some sample collection methods were not clearly described, including the use of exfoliated cervical cells,179 endocervical secretions,180 or urine. Certain sample types were exclusively used for measuring soluble immune mediators (both cellular and humoral) including cervical swabs, sponges, filter paper strips, cervical aspirate, and menstrual cups. Urethrogenital swabs were used solely for antibody detection, whereas cervical scraping was used exclusively for phenotypical characterization of cells. Cervical cytobrushes, CVL, and vaginal swabs were used for both soluble and cell-based immunoassays.

Table 1.

Urogenital sample types used for detecting immune biomarkers

Sample type Sex N Possible additives Method Application Reference
CVL F 24 Protease inhibitors
  • 3–10 mL

  • Sterile saline or PBS

  • Repeatedly wash and aspirate fluid

Soluble & cell-based immune biomarkers
  • Antibodies (n = 8)

  • Cytokines (n = 17)

  • Cell phenotype (n = 3)

Ardizzone et al.36; Agrawal et al.38; Agrawal et al.39; Agrawal et al.40; Agrawal et al.41; Agrawal et al.42; Agrawal et al.43; Gupta et al.44; Jha et al.45; Mlisana et al.46; Ogendi et al.47; Spear et al.48; Sperling et al.49; Srivastava et al.50; Masson et al.51; Hwang et al.52; Richmond et al.53; Levine et al.54; Marconi et al.55; Jordan et al.56; Hedges et al.57; Barousse et al.58; Mott et al.59; Gupta et al.60
Cervical cytobrush F 22 Antibiotics, fungicide, glutamine, FBS, sodium azide
  • Collected in PBS, keratinocyte serum-free media, RPMI medium, or saline

  • Processed within 1–6 h

  • Cells can be dislodged using mechanical methods (vortex, rotate, or push through pipette point) or DTT treatment

  • Cell strainer (40–70 μm pore size) for homogenizationa

Soluble & cell-based immune biomarkers
  • Antibodies (n = 1)

  • Cytokines (n = 15)

  • Cell phenotype (n = 16)

Albritton et al.37; Agrawal et al.39; Agrawal et al.41; Agrawal et al.42; Agrawal et al.43; Gupta et al.44; Jha et al.45; Srivastava et al.50; Masson et al.51; Levine et al.54; Gupta et al.60; Agrawal et al.61; Agrawal et al.62; Cohen et al.63; Ficarra et al.64; Ibana et al.65; Kelly et al.66; Scott et al.67; Vats et al.68; Reddy et al.69; Schust et al.70; McClure et al.71
Cervical swab F 19 FBS (3%), 2X protease inhibitor cocktail, antibiotics, fungicide
  • Cotton, nylon or Dacron-tipped swabs

  • Collected in PBS, Amplicor buffer (Roche), 2SP buffer or sterile saline—buffer not always mentioned

  • Left in place for 5–10 s while applying pressure or scrapinga

Soluble immune biomarkers
  • Antibodies (n = 11)

  • Cytokines (n = 8)

Hedges et al.57; Reddy et al.69; Bua et al.72; Filardo et al.73; Fresse et al.74; Tsai et al.75; Fichorova et al.76; Fichorova et al.77; Witkin et al.78; Witkin et al.79; Zhang et al.80; Terho and Meurman81; Osser and Persson82; GrÖNroos et al.83; Honkonen et al.84; Kalimo et al.85; Puolakkainen et al.86; Witkin et al.87; Audu et al.88
Sponge F 17 IGEPAL detergent (0.25%–10%), protease inhibitors, BSA (0.5%), Tween 20 (0.05%)
  • Ophthalmic or nasal sponges

  • Elution method: immersing in medium or PBS, centrifugation using spin assembly apparatus, Spin-X microcentrifuge tube filters

  • Elution buffer: PBS, keratinocyte SFMa

  • Some researchers block spin filters or equilibrate sponges using PBS with NaCl, aprotinin (a trypsin inhibitor), and sodium azide before centrifugation

Soluble immune biomarkers
  • Antibodies (n = 9)

  • Cytokines (n = 8)

Albritton et al.37; Sperling et al.49; Kelly et al.66; Darville et al.89; Wang et al.90; Wang et al.91; Ziklo et al.92; Ziklo et al.93; Poston et al.94; Darougar et al.95; Schachter et al.96; Thejls et al.97; Treharne et al.98; Omer et al.99; Lewis et al.100; Mardh et al.101; Southgate et al.102
Vaginal swab F 5 IGEPAL detergent (10%), bacterial protease inhibitor (1%)
  • Clinician collected or collected by self-sampling

  • Collected in sterile saline, PBS, or RNAlater (Ambion)—buffer not always mentioned

Soluble & cell-based immune biomarkers
  • Antibodies (n = 2)

  • Cytokines (n = 3)

  • Cell phenotype (n = 2)

Ziklo et al.93; Cauci and Culhane103; Chen et al.104; van den Broek et al.105; Cai et al.106
Urethrogenital swab M/F 6 None Calcium alginate swabs (Calgiswab) Antibodies (n = 6) Hammerschlag et al.107; McCormack et al.108; Ng et al.109; Gump et al.110; McCormack et al.111; McComb et al.112
Filter paper F 4 None Let paper absorb secretions and elute in PBS Soluble immune biomarkers
  • Antibodies (n = 3)

  • Cytokines (n = 1)

Arno et al.113; Ruijs et al.114; Workowski et al.115; Brunham et al.116
Aspiration F 5 None Syringe, catheter, pipette, or Aspirette device Soluble immune biomarkers
  • Antibodies (n = 4)

  • Cytokines (n = 1)

Cohen et al.63; Ruijs et al.114; Mahmoud et al.117; Persson et al.118; Mahmoud et al.119
Menstrual cup F 2 None
  • Self-sampling

  • SoftCup (Instead)

  • Specified timinga

  • Diluted using PBSa

Soluble immune biomarkers
  • Antibodies (n = 1)

  • Cytokines (n = 1)

Abraham et al.120; Garrett et al.121
Cervical scraping F 1 None
  • Wooden spatula

  • Agitate spatula in saline

Cell phenotype (n = 1) Levine et al.54
Total ejaculate M 31 None
  • By masturbation

  • Period of sexual abstinence (2–24 days)

Soluble & cell-based immune biomarkers
  • Antibodies (n = 12)

  • Cytokines (n = 7)

  • Cell phenotype (n = 13)

Bua et al.72; Cai et al.122; Martínez-Prado and Camejo Bermúdez123; Mazzoli et al.124; Dehghan Marvast et al.125; Karaulov et al.126; Pérez-Soto et al.127; Mazzoli et al.128; Hakimi et al.129; Hakimi et al.130; Habermann and Krause131; Moazenchi et al.132; Markelova et al.133; Gdoura et al.134; Samra et al.135; Penna Videau et al.136; El Feky et al.137; Ruijs et al.138; Dieterle et al.139; Eggert-Kruse et al.140; EzzEl-Din et al.141; Kojima et al.142; Munoz and Witkin143; Suominen et al.144; Weidner et al.145; Eggert-Kruse et al.146; Eggert-Kruse et al.147; Eggert-Kruse et al.148; Cai et al.149; Mazzoli et al.150; Bjercke and Purvis151
Seminal plasma M 33 PureSperm (NidaCon Int.), PMSF protease inhibitor
  • By centrifugation of total ejaculate

  • Centrifugation speed: 300–10,000 ×ga

  • Centrifugation time: 5–10 mina

Soluble immune biomarkers
  • Antibodies (n = 27)

  • Cytokines (n = 11)

Martínez-Prado and Camejo Bermúdez123; Dehghan Marvast et al.125; Pérez-Soto et al.127; Habermann and Krause131; Moazenchi et al.132; Penna Videau et al.136; El Feky et al.137; Eggert-Kruse et al.140; EzzEl-Din et al.141; Munoz and Witkin143; Suominen et al.144; Weidner et al.145; Eggert-Kruse et al.146; Eggert-Kruse et al.147; Eggert-Kruse et al.148; Bjercke and Purvis151; Bollmann et al.152; Bollmann et al.153; Eggert-Kruse et al.154; Ochsendorf et al.155; Pérez-Soto et al.156; Kokab et al.157; Wolff et al.158; Wolff et al.159; Segnini et al.160; Motrich et al.161; Jungwirth et al.162; Nasr El-din et al.163; Munoz et al.164; Witkin et al.165; Yoshida et al.166; Gruschwitz et al.167
Urethral swab or smear M 7 FBS (3%), antibiotics, fungicide
  • Dacron-tipped stainless-steel shaft swabs, platinum wire loop swabs, cotton-tipped swabs, or calcium alginate urethrogenital swabs∗

  • Swab medium: PBS, 2SP buffera

Soluble & cell-based immune biomarkers
  • Antibodies (n = 5)

  • Cytokines (n = 1)

  • Cell phenotype (n = 2)

Fresse et al.74; Terho and Meurman81; Omer et al.99; Ng et al.109; Gdoura et al.134; Pate et al.168; Shahmanesh169
EPS M 6 None
  • By digital prostatic massage after voiding of urine

  • One author used a monofilament knitted polypropylene swab passed into the anterior urethra for collection of fluid after EPS expression

Antibodies (n = 6) Cai et al.122; Mazzoli et al.128; Cai et al.149; Ostaszewska-Puchalska et al.170; Mardh et al.171; Shortliffe et al.172
FVU M 3 Cytolyt, BSA (0.1%), DTT, Sputazol
  • Initial flush of urine, diluted in PBS or PBS with additivesa

  • 5–40 mL

  • Early morning or collected after not having urinated for 3 hoursa

Cell phenotype (n = 3) Ito et al.173; Wiggins et al.174; Shahmanesh et al.175
PPM urine M 3 None
  • After rectal digital massage

Antibodies (n = 3) Cai et al.122; Mazzoli et al.128; Cai et al.149
Other urine M/F 6 None
  • No further information

Soluble immune biomarkers
  • Cytokines (n = 2)

  • Antibodies (n = 2)

  • Cytokine mRNA (n = 2)

Bua et al.72; Tsai et al.75; Ochsendorf et al.155; Ohsawa et al.176; Ray et al.177; Ray et al.178

Sample type, sex, total number of studies (N) using a certain sample type, additives that can be added, collection method, application method used in provided references, and references (Refs.) are indicated in the table.

2SP, sucrose-phosphate buffer; F, female; FBS, fetal bovine serum; M, male; PPM urine, post-prostatic massage urine; SFM, serum-free media.

a

Method not specified in all articles.

Figure 2.

Figure 2

Sample types for collection of urogenital secretions in men and women

Overview of sample types used in men (left) and women (right), in order of appearance. The number of studies using a certain sample type is noted in brackets.

To collect male urogenital secretions, most studies used total ejaculate72,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151 or seminal plasma.123,125,127,131,132,136,137,140,141,143,144,145,146,147,148,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167 Less-common methods included urethral swab or smear,74,81,99,109,134,168,169 EPS,122,128,149,170,171,172 or urine samples,72,75,122,128,149,155,173,174,175 sometimes specified as FVU173,174,175 or post-prostatic massage urine.122,128,149 Total ejaculate was commonly used to assess both humoral and cellular immune responses, whereas cell-free seminal plasma was used exclusively to measure soluble immune mediators (antibodies and cytokines). EPS was used uniquely for measuring humoral immune responses. In men, urethral swabs and urine samples were used to evaluate both humoral and cellular immune responses.

Detection of antibodies in urogenital secretions

A variety of assays and antigens have been used for local antibody detection in urogenital secretions (Figure 3A). In men, 25 studies employed enzyme-linked immunosorbent assays (ELISA) to detect local CT-specific antibodies. Additionally, seven studies used microimmunofluorescence (MIF), 11 used whole-cell inclusion immunofluorescence (WIF), five used western blot, and two used radioimmunoassays (RIA). For antibody detection in female secretions, 18 studies used ELISA-based assays, 19 used MIF, four used WIF, four used western blot, and four used RIA. In total, 40 studies using immunofluorescence-based methods were identified in this review, including 24 using MIF and 16 using WIF.181,182,183,184 ELISA-based assays were used in 42 of the included studies, which are discussed in detail in Tables S3 and S4. One ELISA-based study included both female and male participants attending an STI clinic, but did not analyze them separately.74 Overall, western blot assays for detection of chlamydial antibodies were used in eight articles. Five of these studies used a commercial assay, the CT IgG + IgA western blot test AID (Autoimmun Diagnostika GmbH), including lipopolysaccharide (LPS), major outer membrane protein (MOMP), and chlamydial heat shock protein (cHsp) antigens.106,122,124,128,149 Three studies detected local antibodies against chlamydial inclusion membrane proteins (Incs) using an in-house immunoblot.44,60,75 RIAs were used in five articles for detection of CT-specific antibodies, all published in the 1980s. In these studies, L2 cell lysate coupled to polystyrene beads was used as antigen. Antibodies present in the sample were detected using radiolabeled anti-human antibodies.81,83,84,85,144

Figure 3.

Figure 3

Immunoassays for detecting immune biomarkers in urogenital secretions

Overview of different immunoassays for detecting immune biomarkers in urogenital secretions. Percentages indicate proportion of articles using the specified assay. Per immunoassay, different antigens that were used are given in the outer circle (proportional to the number of assays). CBA, cytokine bead array; Incs, inclusion proteins; LP assay, lymphoproliferation assay; L2, CT serovar L2 (lymphogranuloma venereum); Mip, macrophage infectivity potentiator; MOMP, major outer membrane protein; WB, western blot assay.

(A) Overview of immunoassays used for detection of antibodies in female (left) and male (right) genital secretions. Studies including both female and male participants were included in both proportions.

(B) Overview of different immunoassays used for measuring antigen-specific cellular immune responses in female urogenital secretions.

Functional antibody assays were performed in two studies, both focusing on female cervicovaginal secretions. The first study examined the inhibitory effects on inclusion formation using direct immunofluorescence-based staining of inclusion bodies. The study concluded that secretions of women with positive endocervical culture results exhibited higher anti-chlamydial activity compared with secretions of culture-negative women.117 More recently, a neutralization assay was performed by co-incubating serum- or CVL-derived antibodies with purified elementary bodies (EBs) on endocervical epithelial cell monolayers. Inclusion-forming units were then enumerated to assess neutralizing activity. The authors concluded that genital IgG, genital IgA, and serum IgA significantly inhibit the formation of inclusion bodies, whereas serum IgG did not. This finding may be explained by the higher affinity or broader reactivity of local mucosal antibodies. The study also investigated genogroup-specific neutralization and confirmed that the infecting strain can shape humoral immune responses.36

Association between local and serum antibody levels

In this review, we identified eight studies that investigated associations between local and serum antibody levels. An overview of these studies is provided in Table 2. Strong correlations between cervical and serum IgG antibodies were observed in two studies (Spearman’s ρ = 0.89, Pearson’s R = 0.57), whereas correlations for IgA were notably weaker (Spearman’s ρ = 0.18, Pearson’s R = 0.26, respectively).89,120 Supporting these findings, agreement (κ-values) between IgG positivity in serum and vaginal swab samples was modest, and even lower for IgA in a fertility clinic cohort. However, this trend was not observed in an STI clinic cohort.105 In male urogenital secretions, only two studies found a significant association between serum and seminal plasma Chlamydia IgA positivity.154,155

Table 2.

Relationship between local mucosal and serum antibody levels

Reference Total N Local secretion sample type Isotype Correlation between paired mucosal and serum samples Significance level
Abraham et al.120 35 Menstrual cup IgG ρ = 0.89 p ≤ 0.0001
IgA ρ = 0.18 p = 0.43
Darville et al.89 151 Cervical sponge IgG R = 0.57 p = 1.82 E−13
IgA R = 0.26 p = 0.002
van den Broek et al.,105 cohort 1 77 Vaginal swab IgG κ = 0.130 p = 0.2
IgA κ = 0.37 p < 0.001
van den Broek et al.,105 cohort 2 116 Vaginal swab IgG κ = 0.27 p = 0.015
IgA κ = 0.17 p = 0.055
Eggert-Kruse et al.154 173 Seminal plasma IgA Chi-square or Fisher’s exact test p < 0.001
Ochsendorf et al.155 125 Seminal plasma IgA Fisher’s exact test p = 0.0003
IgG Fisher’s exact test N.s.
Munoz et al.164 48 Seminal plasma, total ejaculate IgA Not reported No correlation found
Shortliffe et al.172 82 Expressed prostatic secretions IgA, IgG Linear regression analysis No correlation found
Weidner et al.145 131 Seminal plasma IgA Not reported No correlation found

Correlation coefficients (Spearman correlation coefficient, ρ, or Pearson correlation coefficient, R) or agreement (κ value for positivity in serum and positivity in local antibodies) and significance level (p value) are given.

N, number of participants in the study.

Detection of cellular immune responses in urogenital secretions

Overall, studies assessing antigen-specific immune responses in genitourinary secretions were limited (n = 9).39,44,45,50,60,61,62,68,69 An overview is provided in Table S5. Notably, one article focusing on cervical monocytes is included in this section.62 Although monocytes are not considered truly adaptive antigen-specific immune cells, they can play a major role in initiating both adaptive and innate immune responses against pathogens.185 Detection of cytokine-secreting cells following antigen stimulation by enzyme-linked immunosorbent spot (ELISpot) was performed in two articles.44,61 Another commonly used method for this endpoint is intracellular cytokine staining (ICS),186 which was applied in three studies; however, antigen-specific ICS was performed in only one of them.45,51,69 In total, 59 studies investigated cytokine and chemokine concentrations in urogenital secretions. Of these, 50 used ELISA-based techniques and 13 used cytokine magnetic bead assays. An overview of the number of studies measuring different cytokines and chemokines is presented in Figure S1. Seven studies measured cytokine concentrations following antigen stimulation using either cytokine ELISA or multiplex cytometric bead arrays.44,45,50,60,61,62,68 Three studies assessed lymphoproliferative responses following antigen stimulation. Two of these used radiolabeled 3H-thymidine incorporation into chromosomal DNA during mitosis for detection, whereas another employed a colorimetric MTT assay (3-(4,5-dimethyl thiazol-2-yl) 2, 5-diphenyl tetrazolium bromide) to determine stimulation indices.39,44,68 In this review, two studies were identified that involved cytotoxicity assays. The first study measured CT-specific cytotoxicity by quantifying lactate dehydrogenase release from damaged cells after antigen stimulation,44 whereas the second assessed non-antigen-specific cytotoxicity testing by measuring perforin expression of endocervical CD8+ T cells.65 Differential mRNA expression was reported in eight studies, three of which involved antigen stimulation.44,60,61,62,67,93,177,178 An overview of the antigens used for different antigen-specific T cell assays is presented in Figure 3B.

Seventeen studies used flow cytometry to determine immune cell phenotypes.39,41,42,43,45,47,51,54,61,63,64,65,66,68,69,70,71 Notably, this technique was exclusively applied to female urogenital secretions, with 16 studies analyzing cervical cytobrush samples and only one study using CVL.47 Only one study used flow cytometry to assess antigen-specific immune cell phenotypes.69 Overall, CD4+ T cells were analyzed in 14 studies, CD8+ lymphocytes in eight, and CD3 expression was explored in six studies. Some researchers also investigated additional surface markers.47,54,64,65,66 The CD4:CD8 T cell ratio was calculated in five articles. Dendritic cells (DCs) were analyzed in five articles. Some authors reported the percentage of lymphocytes among endocervical leukocytes (59%–86%, irrespective of infection or disease status),39,41,71 whereas others reported absolute cell counts. Multiple authors reported an increase in the mean number of CD4+ cells in CT-infected women, compared with those with fertility disorders or controls.41,54,61,63,69 This increase was further enhanced in women with mucopurulent cervicitis.42 Interestingly, only one study found a significant increase in the mean number of CD8+ mucosal T cells in CT-infected women compared with controls.69 B lymphocytes were present in low numbers, although they were only enumerated in two studies.39,71 Other methods for cell counting included microscopy-based techniques (n = 26) and the use of an automated urine particle analyzer (n = 1).178 In these studies, leukocytes or granulocytes were most commonly counted.

Association between local and systemic cellular immune responses

Peripheral blood mononuclear cell (PBMC) samples were collected in 16 articles to evaluate systemic cellular immune responses. Some studies additionally collected whole blood, serum, or plasma to determine cytokine concentrations.

Flow cytometry data have demonstrated distinct differences between local and systemic cellular immune phenotypes. As expected, lymphocyte counts are lower in cytobrush samples compared with cervical biopsies and peripheral blood.63 Data on CD4+/CD8+ ratios in blood and endocervical samples are inconsistent, with some authors reporting comparable ratios and others showing a higher percentage of systemic CD4+ cells.47,64 Notably, the endocervix is predominantly populated by CD45RO+ effector memory T cells (TEM), as indicated by a significantly lower CD45RA+/CD45RO+ T cell ratio compared with peripheral blood.64,65,66 This TEM dominance was also observed in cervical CD8+ cells, which were notably less perforin positive than systemic TEM cells.65 Regarding CD4+ helper T cell populations, a distinct Th1/Th2 polarization has been reported. PBMCs show higher expression of Th2-associated chemokine receptor CCR4, whereas mucosal lymphocytes express more Th1-associated CXCR3 and CCR5. This suggests that CD4+ cells may undergo changes in polarization upon arrival at the site of infection. Additionally, mucosal homing and retention markers including CD103 (αE) are upregulated in cervical T cells compared with peripheral blood.64,66 Although some studies suggest that α4 and β7 co-expression is lower in cervical compared with systemic compartments, others report a unique population of cervical memory T cells co-expressing both mucosal and peripheral homing receptors (α4β7/CLA).64,66 During CT infection, endocervical T cells become highly activated and upregulate HLA-DR and CD38.47,54,64,66 Furthermore, the absolute number of tolerogenic plasmacytoid DCs per milliliter of blood was significantly lower in Chlamydia-infected women compared with their cervical samples and with blood samples from controls. This difference was even more pronounced in women with fertility disorders or mucopurulent cervicitis.41,42 The percentage of mature myeloid DCs (mDCs) in paired cervical and blood samples of CT-infected women was comparable; however, the absolute number of mDCs per milliliter of blood was significantly lower in CT-infected women without mucopurulent cervicitis compared with controls.42 Moreover, the percentage of granulocytes in endocervical samples was lower compared with paired PBMCs (49%–55% vs 61%–63%).66

When comparing lymphoproliferative responses of cervical lymphocytes and PBMCs after stimulation with cHsp and MOMP peptides, no significant differences between stimulation indices were observed,39 suggesting a good correlation between the two. Similar trends were observed for Inc-specific responses.44 In contrast, another study reported significantly higher proliferative responses of cervical lymphocytes compared with PBMCs following stimulation with MOMP peptides in CT-positive women.68 Several studies compared antigen-specific cytokine secretion by cervical lymphocytes and PBMCs. Jha et al. reported interferon (IFN)-γ secretion was comparable, whereas levels of interleukin (IL)-17 and IL-22 were slightly higher in cervical cell supernatants compared with PBMC supernatants from CT-infected patients.45 Upon MOMP stimulation, IL-6 and IL-10 levels were significantly lower in PBMC supernatants compared with supernatants from cervical lymphocytes in healthy control subjects. Conversely, in CT-positive women, IL-6 and IL-8 levels were significantly higher in the systemic compartment.68 When measuring IFN-γ concentrations directly in the local sample, strong correlations with plasma levels were lacking.113,179

Only two studies used data from both mucosal and systemic assays to calculate correlations. Neither study found significant correlations between cervicovaginal and systemic cytokine concentrations.38,113

Factors influencing the detection of Chlamydia trachomatis immune biomarkers in urogenital secretions

Besides sample type and immunoassay, we identified several host-pathogen factors that can influence the immune response measured during chlamydial infection. An overview of the major contributing factors is shown in Figure 4. Table 3 indicates the number of studies that accounted for different confounding factors, while details regarding their effects are presented in Table S6.

Figure 4.

Figure 4

Different factors influencing mucosal immune responses to urogenital CT infections

Overview of different factors affecting measured mucosal immune responses to urogenital C. trachomatis infections. Infection status (persistence vs. spontaneous clearance, primary exposure vs. past exposure, frequency of reinfections), treatment status (before or after treatment, treated or untreated), host genetics, sample type (explained in detail in Table 1) and dilution, immunoassay choice (antigen specific or not, species vs. genus specific, assay performance characteristics), presence of coinfections, infecting genotype and CT load, microbiome composition, hormonal effects (time point during menstrual cycle, menstruating vs. premenarche or postmenopausal, hormonal contraceptives), urogenital symptoms (symptomatic vs. asymptomatic, mucopurulent infection or not, etc.) and fertility symptoms (infertile vs. fertile) can all affect whether, which, and how many immune biomarkers are found in the urogenital secretions of men and women.

Table 3.

Influencing factors taken into account in different papers

Influencing factor N° of papers (% of total)
Coinfections (tested or self-reported) 109 (75.2%a)
  • N = 41 excluded coinfected participants, 68 did not.

  • 28/68 investigated effect of other infections on outcome parameters.

  • Effects on immune biomarkers in coinfected participants compared with monoinfection were investigated in 9 papers.

Microbiome 11 (7.6%a)
  • 11 investigated effects of the microbiome; however, only 9 of them separately analyzed this for CT patients with and without microbiome parameters.

  • Two articles investigated effects of bacterial flora in men.

  • Seven articles investigated effects of bacterial vaginosis or female microbiome community state type.

Menstrual cycle 33 (37.1%b)
  • One reported days since last menstruation

  • 14 did not collect samples during menstruation.

  • 18 specified timing:
    • Midcycle, before ovulation: resembling late follicular phase (n = 11)
    • Early follicular phase (n = 1)
    • Pregnancy (n = 4)
    • Oocyte retrieval for IVF (n = 2)
Other hormonal effects 5 (5.6%a)
  • Two investigated effects of hormonal contraceptive use

  • Three investigated serum hormones (estradiol, progesterone)

Duration of infection (persistence, reinfections) 11 (7.6%a)
  • Eight investigated effect on outcome parameters compared with primary infection or patients who cleared infection during follow-up

Clinical outcomes 34 (23.4%a)
  • Fertility disorders (n = 22)

  • Urogenital tract symptoms including pain, discharge, inflammation (n = 12)

Number of papers out of total number of papers that accounted for coinfections, microbiome, menstrual cycle, hormonal effects, duration of infection, and clinical outcomes and details.

N° (%), number of papers (percentage); N, number, IVF, in vitro fertilization.

a

All 145 total articles.

b

89 articles including women.

Although several studies investigated the independent effects of CT and other STIs on immune biomarkers, only nine explored the combined effects of coinfections compared with participants infected with CT alone. Interestingly, not all studies reported a significant impact of coinfections for each individual pathogen.48,57,122,173 Multiple studies included in this review indicate that CT coinfections with human papillomavirus (HPV), Neisseria gonorrhea (NG), and genital herpes (herpes simplex virus-2) can alter mucosal cytokine levels.52,126,133,156 Moreover, an increased risk of anti-Chlamydia antibody presence has been observed in men with a history of NG infection.136 Furthermore, four studies independently demonstrated that cervicovaginal cytokine patterns are distinctly associated with microbiome community state types or bacterial vaginosis (BV).52,55,59,73 In men, colonization with Proteus spp. has been associated with the presence of anti-Chlamydia IgA in seminal plasma.154 Nonetheless, not all authors found microbiome-associated differences in mucosal immune parameters.56,64,103,172

When collecting female urogenital samples, hormonal influences can be considered by measuring serum hormone levels or taking into account the menstrual cycle. Serum hormones may affect local immune cells present and cytokine concentrations measured.38,41,177 Additionally, local cytokine measurements can be influenced by hormonal contraceptive use.77

Multiple studies independently demonstrate that patients with recurrent or persistent infections, as well as those with fertility disorders, exhibit distinct differences in mucosal cytokine concentrations or mRNA expression.38,39,40,41,43,56,90,93,94,104 Discrepancies arise when comparing these findings with antigen-specific cytokine responses, which may vary depending on the antigen used for stimulation.50,61,69 Furthermore, lower cervical CD4+ T cell counts and differences in DC phenotypes have also been reported in women with fertility disorders.41,43,61 Interestingly, lymphoproliferative responses of cervical lymphocytes in women with recurrent infection appear to depend on the antigen used for stimulation.39 Regarding humoral responses, increased mucosal anti-Chlamydia antibody levels have been observed in women with recurrent infections or fertility disorders, although this varies depending on the antigen used and the cohort sampled.39,40,43,50,78,79,82,83,105,144,154 Although not the focus of this review, higher Toll-like receptor 2 expression on cervical monocytes has been observed in seropositive women, indicating previous CT infection, who already experienced fertility disorders.62 However, the overall number of cervical monocytes was lower in this group.61

Host genetics can also influence immune responses, as shown by Wang et al., who identified two HLA variants and a single IL10 haplotype as independent markers of recurrent CT infection.91 Other key confounding factors include the infecting genotype and CT load. CT load can significantly impact immune cells present and local cytokine concentrations.42,43,94 Low levels of cervical (s)IgA and IgG have also been associated with high CT cervical load.89,116 It has long been known that antibody responses are serovar and genotype specific, but reinfection experiments have shown that antibody responses broaden over time until the infection is cleared. Surprisingly, secondary antibody responses are primarily directed against the serovar involved in the initial infection.187,188,189,190,191 One study included in this review found that local IgA is more broadly reactive against a range of CT ompA genotypes and may not only protect against the current infecting strain.36

Discussion

Comprehensively, there has been growing scientific interest in investigating local mucosal immune responses rather than systemic responses.

However, sampling the local environment presents several challenges, and differences between anatomical compartments and sample types should be considered in trial design. For example, proportional cell counts can differ between sample types.54,192,193 It should also be noted that superficial, apical immune cells might not be representative of cells residing in deeper layers of the tissue.193 In contrast, more invasive scraping or brushing techniques may collect circulating blood-derived cells rather than mucosal cells. The impact of blood contamination remains unclear, as even in visually contaminated samples most cells exhibit a mucosal phenotype. Lymphocytes in cervical samples are often outnumbered by other cell types including epithelial cells and neutrophils, which can hinder detection.192,193 Notably, although cells sampled through cervical cytobrushes are referred to as “cervical cells,” their exact anatomical origin is difficult to determine.192 In the context of cytokine quantification, variations in sample types can lead to inconsistent results. For instance, significantly higher tumor necrosis factor-α concentrations were observed in vaginal sponge samples before treatment compared with after treatment, whereas CVL samples showed no variations upon treatment.49 Discrepancies in cytokine concentrations are even more pronounced when comparing cytokine concentrations in the raw urogenital secretions with those in antigen-stimulated cervical cell supernatants (Figure 5). Sample-dependent differences have also been described for antibody detection.21,37,194 Table 4 provides an overview of different sample types and their suitability in different scenarios. This can help guide the selection of appropriate sample types depending on the study population, the immunological biomarker of interest, and whether self-collection or integrated DNA detection is desired. Combining DNA detection with both cellular and humoral immune biomarkers from a single sample offers complementary insights into mucosal immunity, enabling a more comprehensive understanding of local immune responses. In sample types that allow antibody detection, antibodies can be used to assess past exposure (or vaccination).214,215,216,217,218,219,220 Detecting pathogen-specific DNA alongside corresponding immunological endpoints is feasible in various sample types, including vaginal and cervical swabs, cervical cytobrushes, urethral or urethrogenital swabs, ejaculate, and FVU. Among these, vaginal swabs, ejaculate and (first-void) urine can be self-collected. Interestingly, FVU is a promising self-collectable sample type for evaluating HPV-related (functional) immune biomarkers and DNA in women.19,204,221

Figure 5.

Figure 5

Measurement of urogenital interferon-γ concentration depends on infection status, immunoassay choice, and sample type

Median IFN-γ concentrations (unless indicated as mean) in picograms per milliliter in different urogenital secretions retrieved from different studies (one data point indicates one population in a certain study). Infection status is indicated using the color and shape of data points. IFN-γ concentrations differ greatly when comparing sample types (cervicovaginal lavage, exfoliated cervical cells, menstrual cup, sponge, cervical swab, urethral swab, ejaculate) and CT infection status (CT [without further specifications], CT + Herpes coinfection, CT fertility disorders, CT mucopurulent cervicitis, CT mucopurulent cervicitis - treated, CT asymptomatic, CT asymptomatic - treated, CT clearers, CT fertile, CT persistent, CT primary infection, CT recurrent infection, CT symptomatic, CT after treatment, or negative uninfected controls) and depends on whether antigen-specific responses were considered (supernatant of antigen-stimulated cells) or not.

Table 4.

Adequacy of distinct urogenital sample types for measuring different mucosal biomarkers

Sample type Sex Self-collection NAAT Antibody detection Functional antibodies Cytokines Cellular Remarks Reference
CVL F + +++ ++ +++ +
  • Recommended sample for analysis of soluble immune factors.

  • Suitable for both soluble and cell-based assays.

  • Self-sampling devices available (e.g., Delphi Screener, Pantarhei Screener); however, not commonly used.

World Health Organization28; Ardizzone et al.36; Agrawal et al.38; Agrawal et al.39; Agrawal et al.40; Agrawal et al.41; Agrawal et al.42; Agrawal et al.43; Gupta et al.44; Jha et al.45; Mlisana et al.46; Ogendi et al.47; Spear et al.48; Sperling et al.49; Srivastava et al.50; Masson et al.51; Hwang et al.52; Richmond et al.53; Levine et al.54; Marconi et al.55; Jordan et al.56; Hedges et al.57; Barousse et al.58; Mott et al.59; Gupta et al.60; de Melo Kuil et al.195; Verhoef et al.196
Cervical cytobrush F +a ++ + ? ++ +++
  • Care should be taken to avoid bleeding.

  • Recommended sample for analysis of cells.

World Health Organization28; Albritton et al.37; Agrawal et al.39; Agrawal et al.41; Agrawal et al.42; Agrawal et al.43; Gupta et al.44; Jha et al.45; Srivastava et al.50; Masson et al.51; Levine et al.54; Gupta et al.60; Agrawal et al.61; Agrawal et al.62; Cohen et al.63; Ficarra et al.64; Ibana et al.65; Kelly et al.66; Scott et al.67; Vats et al.68; Reddy et al.69; Schust et al.70; McClure et al.71; McKinnon et al.192; Lund et al.193; Leinonen et al.197
Cervical swab F +a ++ ++ + ++ ?
  • Functional (neutralizing) antibodies demonstrated in other infections.

World Health Organization28; Hedges et al.57; Reddy et al.69; Bua et al.72; Filardo et al.73; Fresse et al.74; Tsai et al.75; Fichorova et al.76; Fichorova et al.77; Witkin et al.78; Witkin et al.79; Zhang et al.80; Terho and Meurman81; Osser and Persson82; GrÖNroos et al.83; Honkonen et al.84; Kalimo et al.85; Puolakkainen et al.86; Witkin et al.87; Audu et al.88; Leinonen et al.197; Draper et al.198
Sponge F ++ + ++ ?
  • Functional (neutralizing) antibodies demonstrated in other infections.

Kemp et al.21; World Health Organization28; Albritton et al.37; Sperling et al.49; Kelly et al.66; Darville et al.89; Wang et al.90; Wang et al.91; Ziklo et al.92; Ziklo et al.93; Poston et al.94; Darougar et al.95; Schachter et al.96; Thejls et al.97; Treharne et al.98; Omer et al.99; Lewis et al.100; Mardh et al.101; Southgate et al.102
Vaginal swab F ++ +++ + ? + +
  • Recommended sample for NAAT testing in women.

  • Can be self-collected or clinician collected.

World Health Organization28; Ziklo et al.93; Cauci and Culhane103; Chen et al.104; van den Broek et al.105; Cai et al.106
Filter paper F + ? + ? World Health Organization28; Arno et al.113; Ruijs et al.114; Workowski et al.115; Brunham et al.116
Aspiration F + ? + ? World Health Organization28; Cohen et al.63; Ruijs et al.114; Mahmoud et al.117; Persson et al.118; Mahmoud et al.119
Menstrual cup F ++ ++ + ++ +
  • Recently shown that it can be used for isolation of FGT immune cells.

  • Functional (neutralizing) antibodies. demonstrated in other infections.

World Health Organization28; Abraham et al.120; Garrett et al.121; Peters et al.199; Mkhize et al.200
Cervical scraping F ? ? ? +
  • Care should be taken to avoid bleeding.

World Health Organization28; Levine et al.54; McKinnon et al.192; Lund et al.193
Total ejaculate M ++ ++ ++ + + ++
  • Total ejaculate is expected to contain the same functional antibodies when compared with seminal plasma.

World Health Organization28; Bua et al.72; Cai et al.122; Martínez-Prado and Camejo Bermúdez123; Mazzoli et al.124; Dehghan Marvast et al.125; Karaulov et al.126; Pérez-Soto et al.127; Mazzoli et al.128; Hakimi et al.129; Hakimi et al.130; Habermann and Krause131; Moazenchi et al.132; Markelova et al.133; Gdoura et al.134; Samra et al.135; Penna Videau et al.136; El Feky et al.137; Ruijs et al.138; Dieterle et al.139; Eggert-Kruse et al.140; EzzEl-Din et al.141; Kojima et al.142; Munoz and Witkin143; Suominen et al.144; Weidner et al.145; Eggert-Kruse et al.146; Eggert-Kruse et al.147; Eggert-Kruse et al.148; Cai et al.149; Mazzoli et al.150; Bjercke and Purvis151
Seminal plasma M + + +
  • Centrifugation step necessary, making it less practical.

  • Inherently does not contain any cells.

  • Functional (ADCC-enhancing) antibodies demonstrated in other infections.

World Health Organization28; Martínez-Prado and Camejo Bermúdez123; Dehghan Marvast et al.125; Pérez-Soto et al.127; Habermann and Krause131; Moazenchi et al.132; Penna Videau et al.136; El Feky et al.137; Eggert-Kruse et al.140; EzzEl-Din et al.141; Munoz and Witkin143; Suominen et al.144; Eggert-Kruse et al.145; Eggert-Kruse et al.146; Eggert-Kruse et al.147; Eggert-Kruse et al.148; Bjercke and Purvis151; Bollmann et al.152; Bollmann et al.153; Eggert-Kruse et al.154; Ochsendorf et al.155; Pérez-Soto et al.156; Kokab et al.157; Wolff et al.158; Wolff et al.159; Segnini et al.160; Motrich et al.161; Jungwirth et al.162; Nasr El-din et al.163; Munoz et al.164; Witkin et al.165; Yoshida et al.166; Gruschwitz et al.167; Jecht and Poon201; Parsons et al.202
Urethral swab/smear M ++ + ? + + World Health Organization28; Fresse et al.74; Terho and Meurman81; Omer et al.99; Ng et al.109; Gdoura et al.134; Pate et al.168; Shahmanesh169
EPS M ++ ? + +
  • It has been demonstrated that immune cells and cytokines can be measured in EPS in other contexts (e.g., prostatitis).

World Health Organization28; Cai et al.122; Mazzoli et al.128; Cai et al.149; Ostaszewska-Puchalska et al.170; Mardh et al.171; Shortliffe et al.172; Nadler et al.203
FVU M/F ++ +++ ++ ++ ++ ++
  • In men: recommended sample for NAAT and for analysis of soluble and cell-based immune mediators (more practical compared with ejaculate).

  • In women: up to 10% less sensitive for NAAT compared with vaginal or endocervical swabs. In other genital infections, non-inferiority for NAAT and presence of functional (neutralizing) antibodies has been shown.

World Health Organization28; Ito et al.173; Wiggins et al.174; Shahmanesh et al.175; Teblick et al.204; Centers for Disease Control and Prevention205; Aaron et al.206; Van Keer et al.207
PPM urine M ++ ? + +
  • It has been demonstrated that immune cells, cytokines, and antibodies can be measured in PPM urine in other contexts (e.g., UTI, prostatitis)

World Health Organization28; Cai et al.122; Mazzoli et al.128; Cai et al.149; Ludwig et al.208; Fujita et al.209; Cao et al.210
Midstream urine M/F ++ ++ + + ++ +
  • It has been demonstrated that immune cells, cytokines and antibodies can be measured in midstream urine in other contexts (e.g., UTI).

World Health Organization28; Ludwig et al.208; Kellogg et al.211; Gibb and Edmond212; Drage et al.213
Urethrogenital swab M/F ++ + ? ? ? World Health Organization28; Hammerschlag et al.107; McCormack et al.108; Ng et al.109; Gump et al.110; McCormack et al.111; McComb et al.112

Sample type; sex it can be used for; self-collection potential; whether NAAT is available28; if antibody detection, cytokine measurement, and isolation of cells for cell-based assays is possible; and remarks are indicated.

+/++/+++, can be used for this endpoint; -, not suitable for this endpoint; ?, no indications whether this sample is suitable for the endpoint or not.

F, female; FGT, female genital tract; M, male; NAAT, nuclear acid amplification test; PPM urine, post-prostatic massage urine; UTI, urinary tract infection.

a

Cervicovaginal swabs and brushes can be self-collected, whereas endocervical swabs and brushes are always clinician collected.

Despite sampling challenges, researchers have demonstrated that CD69 and CD103 expressions are significantly higher in female reproductive tract tissues compared with blood, indicating the presence of cells with a TRM phenotype, even in a healthy population.193 After CT infection, noncanonical CD4γ13 TRM cells producing both IFN-γ and IL-13 are thought to expand. These memory T cells remain within lymphoid aggregates or memory lymphoid clusters in the genital tract after pathogen clearance by upregulating CD69 expression, alongside B cells and macrophages, which act as antigen-presenting cells. TRM cells are essential for a swift response to CT reinfection through local expansion, and provide superior protection compared with systemic CD4+ T cells.32,33,34,35 Although IFN-γ responses are commonly considered when selecting cytokine biomarkers, IL-13-related responses appear under-represented in the scientific literature, with only one study reporting significant findings.126 In men, CD103+ CD8+ TRM cells have been identified in the penile urethra and TRM cells have also been observed in the interstitial space of the testis, although evidence is sparse.222 No studies included in this review investigated CD69-expressing cells in genitourinary secretions. Most, instead, focused on cytokine concentrations. Interpreting these findings is challenging, as studies used different sample types and often compared heterogeneous populations in terms of infection status (e.g., duration of infection, primary infection vs. recurrent infection, before vs. after treatment, coinfections or not) (Figure 5). Moreover, most cytokines exhibit a protective optimum concentration range that facilitates pathogen clearance, whereas excessive levels can trigger inflammatory reactions and immunopathological tissue damage. Cytokine effects therefore depend on their concentration, immune microenvironment, and the stage of infection.223,224,225,226 Several studies also explored mucosal immune cells, most in a non-antigen-specific manner.

Nine studies investigating antigen-specific cellular responses were included in this review. Despite ELISpot being considered a highly sensitive, standardizable method for use with PBMCs, only two of the nine studies employed this technique.186 Given its adaptability to various (vaccine) antigens, ELISpot holds promise for measuring local immune responses in CT vaccine trials. T cell ELISpot has already been applied in human respiratory,227,228 gastrointestinal,229,230,231 and even cervicovaginal tissues,15,232 highlighting its suitability for mucosal applications. Notably, the two trials in this review that used ELISpot both relied on cervical cytobrush samples and excluded specimens with insufficient cervical cells or lymphocytes (fewer than 1–2 million cells/mL), suggesting that cell yield is a limiting factor.44,61 The low purity and yield, as well as contamination with either blood-derived cells or foreign pathogens and particles, have also been reported in other mucosal contexts, such as nasal T cells.233 Other techniques used to assess antigen-specific cellular immune responses included cytokine ELISA and mRNA expression in the supernatant of cultured cervical lymphocytes, lymphoproliferation assays, and cytotoxicity assays.39,44,45,50,60,61,62,68 Only one study employed flow cytometry or ICS.69 State-of-the-art techniques such as peptide-major histocompatibility complex multimers and T cell receptor sequencing have not yet been applied to assess antigen-specific cellular immune responses in the context of CT, although they have been performed in other diseases.234,235,236,237,238 Despite strong evidence that circulating T cells, particularly IFN-γ-producing CD4+ Th1 cells, play a key role in controlling CT infection,239,240,241,242,243,244,245,246,247,248 only two studies investigated correlations between local and systemic CT-specific cellular immune responses and neither reported significant associations.38,113

Several assays are available for detecting anti-CT serum antibodies.182 In this review, we investigated the use of these assays for detecting local antibodies in urogenital secretions. Multiple earlier studies used immunofluorescence-based assays, which were once considered the gold standard.182 However, these assays exhibit cross-reactivity with other Chlamydia spp., have low sensitivity, and require subjective microscopic interpretation, reducing diagnostic reproducibility.182 The RIA technique has largely been abandoned too due to cost, safety, and waste issues.249 More sensitive, specific, and safer ELISA-based techniques are now preferred. Notably, no studies included in this review employed more recent Luminex bead-based or luciferase immunosorbent assays.182 To quantify local antibody responses, various antigens were used, including whole EBs, recombinant cHsps, recombinant Incs, LPS, and MOMP proteins. However, none of these antigens are fully species specific to CT, which compromises specificity due to cross-reactivity.250,251,252,253 For cHsp in particular, cross-reactivity with human heat shock proteins has been suggested due to sequence homology.254,255 Despite this, immunoassays using whole EBs have demonstrated good sensitivity compared with MOMP-based ELISAs.256,257 One study used CTH522, a recombinant version of CT MOMP, to measure vaccine-induced antibodies.120,258,259 However, not all authors report the antigen used. For example, Ezz-El-Din and coworkers used a combination of antigens including MOMP, and two immunodominant species-specific antigens chlamydial protease-like activity factor and translocated actin-recruiting phosphoprotein, although this was not explicitly stated in the article.141,260,261 Notwithstanding its status as a superior antigen, as evidenced by its high immunogenicity and its lack of crossreaction with Chlamydia pneumoniae,182,262 no articles describing plasmid glycoprotein 3-based assays in urogenital secretions were retrieved in the present review.

The role of antibody-mediated immune responses in human Chlamydia infection is paradoxical. It has serum anti-CT IgG, and cervical anti-CT IgA and IgG have been shown to correlate with reduced cervical burden. However, the presence of both cervical and serum IgG has also been associated with an increased risk of incident infection. Antibodies alone are insufficient to prevent ascension.89 High antibody titers are often linked with enhanced disease severity, reflecting repeated or prolonged exposure rather than protective immunity.263,264 Proposed protective mechanisms include neutralization, antibody-dependent cellular cytotoxicity (ADCC), and antibody-mediated phagocytosis.262,265,266,267,268 In neutrophils, an additional role for complement receptor-mediated phagocytosis rather than FcγR receptor-mediated phagocytosis has also been suggested.266 Unfortunately, this literature search did not identify any studies investigating mucosal phagocytosis or ADCC enhancement by local antibodies. However, Ardizzone et al. demonstrated that mucosal antibodies in female genital secretions more efficiently neutralize EBs compared with serum-derived antibodies at equivalent concentrations, highlighting important functional differences between local and systemic antibody responses.36 Further research is warranted to elucidate the functional role of mucosal antibodies compared with serum antibodies. Only two studies investigated correlations between local and serum antibodies. Strong correlations for IgG support the notion that cervical IgG is a composite of both transudated serum IgG and locally produced IgG, whereas weaker correlations for IgA suggest that mucosal IgA is primarily produced locally by plasma cells.89,120 One study reported a difference in κ-agreement between an STI clinic vs. fertility clinic cohort and suggests that mucosal IgA is associated with tubal pathology in the latter.105 More research is needed to clarify the functionality of local antibodies and their correlation with serum antibodies in male genital secretions.

Several factors are known to influence immune responses to CT infection. One of the most critical is the microbiome. Urogenital CT strains can utilize a specific enzyme to convert indole (produced by, e.g., Prevotella spp., which are elevated in BV) back into tryptophan. This essential amino acid would otherwise be depleted under the influence of IFN-γ.225,269 However, the role of the microbiome in male CT infections remains poorly understood.270,271,272 Another major confounding factor is the presence of coinfections.264,273,274 Studies that did not exclude coinfected participants observed several pathogen-specific differences in mucosal cytokine responses, with varying combinations of coinfections producing distinct cytokine profiles. This underscores the importance of combined STI screening and the use of antigen-specific immunoassays. Additional confounding variables are host genetics,274,275,276,277,278 hormonal influences,279,280 the infecting genotype, and CT bacterial load.36,42,43,89,94,116,187,188,189,190 The characteristics of the study population, such as treatment status, clinical presentation, and fertility status, also significantly affect study outcomes. Persistent or recurrent infections can provoke an aberrant inflammatory response inducing fibrosis and tissue scarring, leading to reproductive sequelae. These pathological outcomes may also skew immune responses measured.32,274,281,282,283,284 Therefore, all these factors should be carefully considered during participant recruitment and when interpreting trial results.

Future directions

As urogenital sampling for detection of mucosal immune biomarkers has gained increasing interest, we propose several aspects that should be considered and optimized for future research in this field. First, we suggest that sample collection and processing be standardized across research groups to enable meaningful comparisons. Therefore, it is important to disclose detailed information regarding sample collection, processing, and dilution. Furthermore, protocols for isolation of cervical mononuclear cells should be harmonized, whereas protocols for isolation of male urogenital mononuclear cells still need to be developed. Uniform collection methods for measurement of local antibodies should also be agreed upon, with normalization for differences in sample volumes and dilution factors. Second, we believe that consensus regarding “gold-standard” assays and antigens for both humoral and cellular immune responses is essential. For validating assays that measure humoral immune responses, a reference standard using either antigen-specific monoclonal antibodies or a serum standard should be available.214 When measuring cellular immune responses, the focus should be on antigen-specific responses, as it seems impossible to account for all confounding factors affecting the local immune environment. Therefore, a reference antigen and a shared protocol for stimulating immune cells in antigen-specific testing would be highly beneficial. Third, differences between anatomical compartments and systemic immune responses should be further investigated and correlations should be evaluated. This can only be achieved when immunoassays are validated for the sample type used, ensuring equivalent assay performance between local and systemic compartments. This, combined with basic experimental research, will enhance our physiological understanding of how antibodies and immune cells reach the primary site of infection. Finally, long-term follow-up of both mucosal and systemic immunological endpoints, along with additional clinical endpoints, is needed to define a correlate of protection, which will be of paramount importance evaluating future vaccine efficacy.

Limitations of the study

Despite these interesting findings, it is important to acknowledge potential limitations in our study. This review did not consider clinical endpoints associated with proposed immunological endpoints, even though they are essential for defining a correlate of protection. However, we did highlight the effect of fertility disorders and urogenital symptoms as factors that can influence mucosal CT immunity. We did not investigate the effect of other confounding factors including demographic background or immune-related comorbidities. The data collected are highly variable regarding population characteristics, sample type and processing, specific biomarkers assessed, and detection methods. Therefore, a meta-analysis was unfortunately not feasible. Yet, the data are presented in a systematic way, and an overview is provided to allow comparison between the studies.

Resource availability

Lead contact

Requests for further information and resources should be directed to and will be fulfilled by the lead contact, Anne Van Caesbroeck (anne.vancaesbroeck@uantwerpen.be).

Materials availability

This study did not generate new unique reagents.

Data and code availability

  • Data reported in this paper will be shared by the lead contact upon request.

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

Acknowledgments

Our sincere gratitude goes to all authors willing to provide additional information or raw data related to studies included in this review. This work was supported by Universiteit Antwerpen (TT(ZAP)BOF 542300005) and the European Union (ERC, URISAMP, 101040588). E.v.d.B. is supported by a PhD fellowship of the Research Foundation Flanders (Belgium) (FWO; grant number 1SC9125N). Views and opinions expressed are, however, those of the authors only and do not necessarily reflect those of the European Union or the European Research Council Executive Agency. Neither the European Union nor the granting authority can be held responsible for them.

Author contributions

Conceptualization, A.V. and L.T.; methodology, A.V.C., A.V., L.T., M.L., and E.v.d.B.; investigation, A.V.C., M.L., and E.v.d.B.; writing—original draft, A.V.C.; writing—review & editing, A.V.C., A.V., L.T., E.v.d.B., M.L., and P.P.; funding acquisition, A.V.; resources, A.V.; supervision, A.V., L.T., and P.P.

Declaration of interests

The authors declare no competing interests.

Declaration of generative AI and AI-assisted technologies in the writing process

During the preparation of this work, the authors used systematic review management platform Ryyan285 to screen titles and abstracts retrieved in the database search based on inclusion and exclusion criteria. While using this tool, the authors reviewed and edited the content as needed and take full responsibility for the content of the publication.

STAR★Methods

Key resources table

REAGENT or RESOURCE SOURCE IDENTIFIER
Software and algorithms

Ryyan Ouzzani et al., 2016 https://doi.org/10.1186/s13643-016-0384-4
Microsoft Excel Microsoft RRID:SCR_016137
EndNote Clarivate Analytics RRID:SCR_014001
Biorender BioRender RRID:SCR_018361

Experimental model and study participant details

A literature search was conducted through Ovid MEDLINE, Web of Science, and Scopus databases on October 9th 2023, and updated on February 11th 2025. The main search concepts were urogenital secretions in men and women, Chlamydia trachomatis, and humoral and cellular immune responses. Animal studies were excluded using predefined filters for each database286,287,288 and studies were limited to English language. No limits regarding publication period were applied. The detailed search strategy is provided in Table S1. Duplicate records were removed using EndNote. Additional articles were manually searched through screening of reference lists of the articles identified via database search.

Method details

Selection criteria

Articles reporting on the detection of CT-specific antibodies or cellular immune responses in urogenital secretions were included. Further inclusion criteria were as follows: (1) English language, (2) Clinical trials, cohort studies, case-control studies, cross-sectional studies, letters to the editor, case reports and case series. Non-human studies, conference proceedings, reviews and meta-analyses were excluded. Articles were excluded if data were incomplete, and the author could not be reached to provide additional information regarding study protocol or outcome. Additionally, studies solely reporting on innate immune biomarkers were excluded. Studies reporting on cytokine concentrations were considered for inclusion, as changing cytokine environments can be mediated by both the innate and the adaptive immune system. Articles reporting on lymphocytes and CD45+ leukocytes (as the latter can include both innate and adaptive immune cells) were included as well. In addition, studies reporting on STIs overall or Chlamydia-associated diseases, without separate analysis of CT-infected or coinfected participants were excluded, unless the author was successfully contacted and could provide additional information.

A prospective protocol was registered on PROSPERO (CRD42023470875) on October 27th 2023, following PRISMA guidelines.289 Every record was screened independently by at least two authors and in case of disagreement, consensus was reached internally or by consulting a third reviewer. Risk of bias was assessed for every selected article. Appropriate JBI critical appraisal tools were used for cross-sectional studies, case reports, case series, randomized controlled trials and letters to the editor.290,291,292,293 For case-control and cohort studies, the Newcastle-Ottawa Scale was used.294

Data extraction, synthesis and presentation of the results

From each eligible article, data related to: (a) population characteristics, (b) CT antigen detection method used, (c) coinfections assessed, (d) sample type and collection method, (e) local immune parameters measured, and their quantification (f) laboratory techniques used for assessment of immune parameters, (g) systemic immune parameters measured and their correlation to local parameters, if available, (h) clinical signs and symptoms, and (i) relevant conclusions of the study were collected.

Quantification and statistical analysis

This study did not involve any statistical analyses.

Additional resources

PROSPERO protocol: https://www.crd.york.ac.uk/PROSPERO/view/CRD42023470875.

Published: August 28, 2025

Footnotes

Supplemental information can be found online at https://doi.org/10.1016/j.isci.2025.113453.

Supplemental information

Document S1. Figure S1 and Tables S1, S2, and S6
mmc1.pdf (489.4KB, pdf)
Table S3. Overview of studies on detection of antibodies in male urogenital secretions using ELISA-based assays
mmc2.xlsx (25.8KB, xlsx)
Table S4. Overview of studies on detection of antibodies in female urogenital secretions using ELISA-based assays
mmc3.xlsx (20.4KB, xlsx)
Table S5. Overview of studies on detection of antigen-specific adaptive cellular immune responses in urogenital secretions
mmc4.xlsx (20.3KB, xlsx)

References

  • 1.World Health Organization. Chlamydia https://www.who.int/news-room/fact-sheets/detail/chlamydia.
  • 2.European Centre for Disease Prevention and Control. (2024). A systematic review and meta-analysis of the prevalence of chlamydia, gonorrhoea, trichomoniasis and syphilis in Europe. 10.2900/0805127.
  • 3.World Health Organization . 2024. Implementing the global health sector strategies on HIV, viral hepatitis and sexually transmitted infections; pp. 2022–2030. report on progress and gaps. [Google Scholar]
  • 4.Chesson H., Spicknall I.H., Kreisel K.M., Gift T.L. Estimates of the Lifetime Productivity Costs of Chlamydia, Gonorrhea, and Syphilis in the United States. Sex. Transm. Dis. 2024;51:635–640. doi: 10.1097/OLQ.0000000000001973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.World Health Organization (2016). WHO guidelines for the treatment of Chlamydia trachomatis. ISBN 978 92 4 154971 4. [PubMed]
  • 6.Holmes K.K., Levine R., Weaver M. Effectiveness of condoms in preventing sexually transmitted infections. Bull. World Health Organ. 2004;82:454–461. [PMC free article] [PubMed] [Google Scholar]
  • 7.Shannon C.L., Klausner J.D. The growing epidemic of sexually transmitted infections in adolescents: a neglected population. Curr. Opin. Pediatr. 2018;30:137–143. doi: 10.1097/MOP.0000000000000578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.European Centre for Disease Prevention and Control. (2024). Annual Epidemiological Report for 2022: Chlamydia. [PubMed]
  • 9.Haggerty C.L., Gottlieb S.L., Taylor B.D., Low N., Xu F., Ness R.B. Risk of sequelae after Chlamydia trachomatis genital infection in women. J. Infect. Dis. 2010;201:S134–S155. doi: 10.1086/652395. [DOI] [PubMed] [Google Scholar]
  • 10.Korenromp E.L., Sudaryo M.K., de Vlas S.J., Gray R.H., Sewankambo N.K., Serwadda D., Wawer M.J., Habbema J.D.F. What proportion of episodes of gonorrhoea and chlamydia becomes symptomatic? Int. J. STD AIDS. 2002;13:91–101. doi: 10.1258/0956462021924712. [DOI] [PubMed] [Google Scholar]
  • 11.European Centre for Disease Prevention and Control Factsheet about Chlamydia. https://www.ecdc.europa.eu/en/chlamydia/facts#:∼:text=At%20least%2070%25%20of%20genital,asymptomatic%20infections%20is%20not%20uncommon
  • 12.Martin K., Wenlock R., Roper T., Butler C., Vera J.H. Facilitators and barriers to point-of-care testing for sexually transmitted infections in low- and middle-income countries: a scoping review. BMC Infect. Dis. 2022;22:561. doi: 10.1186/s12879-022-07534-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Geretti A.M., Mardh O., de Vries H.J.C., Winter A., McSorley J., Seguy N., Vuylsteke B., Gokengin D. Sex Transm Infect; 2022. Sexual Transmission of Infections across Europe: Appraising the Present, Scoping the Future. [DOI] [PubMed] [Google Scholar]
  • 14.Bébéar C., de Barbeyrac B. Genital Chlamydia trachomatis infections. Clin. Microbiol. Infection. 2009;15:4–10. doi: 10.1111/j.1469-0691.2008.02647.x. [DOI] [PubMed] [Google Scholar]
  • 15.Bere A., Denny L., Burgers W.A., Passmore J.A.S. Polyclonal expansion of cervical cytobrush-derived T cells to investigate HIV-specific responses in the female genital tract. Immunology. 2010;130:23–33. doi: 10.1111/j.1365-2567.2009.03172.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mbuya W., McHaro R., Mhizde J., Mnkai J., Mahenge A., Mwakatima M., Mwalongo W., Chiwerengo N., Hölscher M., Lennemann T., et al. Depletion and activation of mucosal CD4 T cells in HIV infected women with HPV-associated lesions of the cervix uteri. PLoS One. 2020;15 doi: 10.1371/journal.pone.0240154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mbuya W., Held K., McHaro R.D., Haule A., Mhizde J., Mnkai J., Mahenge A., Mwakatima M., Sembo M., Mwalongo W., et al. Depletion of Human Papilloma Virus E6- and E7-Oncoprotein-Specific T-Cell Responses in Women Living With HIV. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.742861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Jespers V., Francis S.C., van de Wijgert J., Crucitti T. Methodological issues in sampling the local immune system of the female genital tract in the context of HIV prevention trials. Am. J. Reprod. Immunol. 2011;65:368–376. doi: 10.1111/j.1600-0897.2010.00938.x. [DOI] [PubMed] [Google Scholar]
  • 19.Teblick L., Pattyn J., Van Keer S., De Smet A., De Coster I., Tjalma W.A.A., Rajbhandari I., Panicker G., Unger E.R., Vorsters A. Follow-up of humoral immune response after HPV vaccination using first-void urine: A longitudinal cohort study. J. Med. Virol. 2023;95 doi: 10.1002/jmv.29133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jaumdally S.Z., Jones H.E., Hoover D.R., Gamieldien H., Kriek J.M., Langwenya N., Myer L., Passmore J.A.S., Todd C.S. Comparison of sampling methods to measure HIV RNA viral load in female genital tract secretions. Am. J. Reprod. Immunol. 2017;77 doi: 10.1111/aji.12619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kemp T.J., Hildesheim A., Falk R.T., Schiller J.T., Lowy D.R., Rodriguez A.C., Pinto L.A. Evaluation of two types of sponges used to collect cervical secretions and assessment of antibody extraction protocols for recovery of neutralizing anti-human papillomavirus type 16 antibodies. Clin. Vaccine Immunol. 2008;15:60–64. doi: 10.1128/CVI.00118-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Vornhagen J., Quach P., Santana-Ufret V., Alishetti V., Brokaw A., Armistead B., Qing Tang H., MacDonald J.W., Bammler T.K., Adams Waldorf K.M., et al. Human Cervical Mucus Plugs Exhibit Insufficiencies in Antimicrobial Activity Towards Group B Streptococcus. J. Infect. Dis. 2018;217:1626–1636. doi: 10.1093/infdis/jiy076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sherlock C.H., Lott P.M., Money D.M., Merrick L., Arikan Y., Remple V.P., Craib K., Burdge D.R. Use of Sno Strip filter-paper wicks for collection of genital-tract samples allows reproducible determination of human immunodeficiency virus type I (HIV-1) RNA viral load with a commercial HIV-1 viral load assay. J. Clin. Microbiol. 2006;44:1115–1119. doi: 10.1128/JCM.44.3.1115-1119.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Peeling R.W., Embree J. Screening for sexually transmitted infection pathogens in semen samples. Can. J. Infect. Dis. Med. Microbiol. 2005;16:73–76. doi: 10.1155/2005/958374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Rieg G., Butler D.M., Smith D.M., Daar E.S. Seminal plasma HIV levels in men with asymptomatic sexually transmitted infections. Int. J. STD AIDS. 2010;21:207–208. doi: 10.1258/ijsa.2009.009331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kriesel J.D., Bhatia A.S., Barrus C., Vaughn M., Gardner J., Crisp R.J. Multiplex PCR testing for nine different sexually transmitted infections. Int. J. STD AIDS. 2016;27:1275–1282. doi: 10.1177/0956462415615775. [DOI] [PubMed] [Google Scholar]
  • 27.Smelov V., Eklund C., Bzhalava D., Novikov A., Dillner J. Expressed Prostate Secretions in the Study of Human Papillomavirus Epidemiology in the Male. PLoS One. 2013;8 doi: 10.1371/journal.pone.0066630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.World Health Organization . 2023. The diagnostics landscape for sexually transmitted infections. [Google Scholar]
  • 29.Hald S.R., Tatari C.R., Kirkegaard P., Tranberg M., Andersen B., Nielsen C.P. "Well, that Was Pretty Clever!"-Ethnic Minority Women's Views on HPV Self-Sampling Devices for Cervical Cancer Screening: Attitudes to Brushes, First-Void Urine, and Menstrual Blood Devices. J Racial Ethn Health. 2025;12:1304–1314. doi: 10.1007/s40615-024-01963-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Van Keer S., Tjalma W.A.A., Pattyn J., Biesmans S., Pieters Z., Van Ostade X., Ieven M., Van Damme P., Vorsters A. Human papillomavirus genotype and viral load agreement between paired first-void urine and clinician-collected cervical samples. Eur. J. Clin. Microbiol. Infect. Dis. 2018;37:859–869. doi: 10.1007/s10096-017-3179-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rochford H., Larsson L., Simms V., Mavodza C., Sigwadhi L., Dauya E., Machiha A., Mavhura M., Bipiti T., Mangena C., et al. Acceptability of self-collected vaginal swabs for sexually transmitted infection testing among youth in a community-based setting in Zimbabwe. Int. J. STD AIDS. 2023;34:574–580. doi: 10.1177/09564624231152804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Johnson R.M., Brunham R.C. Tissue-Resident T Cells as the Central Paradigm of Immunity. Infect. Immun. 2016;84:868–873. doi: 10.1128/Iai.01378-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stary G., Olive A., Radovic-Moreno A.F., Gondek D., Alvarez D., Basto P.A., Perro M., Vrbanac V.D., Tager A.M., Shi J., et al. VACCINES. A mucosal vaccine against Chlamydia trachomatis generates two waves of protective memory T cells. Science. 2015;348 doi: 10.1126/science.aaa8205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nguyen N.D.N.T., Guleed S., Olsen A.W., Follmann F., Christensen J.P., Dietrich J. Th1/Th17 T cell Tissue-Resident Immunity Increases Protection, But Is Not Required in a Vaccine Strategy Against Genital Infection With Chlamydia trachomatis. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.790463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Johnson R.M., Yu H., Strank N.O., Karunakaran K., Zhu Y., Brunham R.C. B Cell Presentation of Antigen Selects Out Protective CD4γ13 T Cells: Implications for Genital Tract Tissue-Resident Memory Lymphocyte Clusters. Infect. Immun. 2018;86 doi: 10.1128/IAI.00614-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ardizzone C.M., Albritton H.L., Lillis R.A., Bagnetto C.E.L., Shen L., Cavacini L.A., Kozlowski P.A., Quayle A.J. Human genital antibody-mediated inhibition of Chlamydia trachomatis infection and evidence for ompA genotype-specific neutralization. PLoS One. 2021;16 doi: 10.1371/journal.pone.0258759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Albritton H.L., Kozlowski P.A., Lillis R.A., McGowin C.L., Siren J.D., Taylor S.N., Ibana J.A., Buckner L.R., Shen L., Quayle A.J. A novel whole-bacterial enzyme linked-immunosorbant assay to quantify Chlamydia trachomatis specific antibodies reveals distinct differences between systemic and genital compartments. PLoS One. 2017;12 doi: 10.1371/journal.pone.0183101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Agrawal T., Vats V., Wallace P.K., Salhan S., Mittal A. Cervical cytokine responses in women with primary or recurrent chlamydial infection. J. Interferon Cytokine Res. 2007;27:221–226. doi: 10.1089/jir.2006.0132. [DOI] [PubMed] [Google Scholar]
  • 39.Agrawal T., Vats V., Salhan S., Mittal A. Mucosal and peripheral immune responses to chlamydial heat shock proteins in women infected with Chlamydia trachomatis. Clin. Exp. Immunol. 2007;148:461–468. doi: 10.1111/j.1365-2249.2007.03357.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Agrawal T., Vats V., Salhan S., Mittal A. Local markers for prediction of women at higher risk of developing sequelae to Chlamydia trachomatis infection. Am. J. Reprod. Immunol. 2007;57:153–159. doi: 10.1111/j.1600-0897.2006.00459.x. [DOI] [PubMed] [Google Scholar]
  • 41.Agrawal T., Vats V., Wallace P.K., Salhan S., Mittal A. Role of cervical dendritic cell subsets, co-stimulatory molecules, cytokine secretion profile and beta-estradiol in development of sequalae to Chlamydia trachomatis infection. Reprod. Biol. Endocrinol. 2008;6:46. doi: 10.1186/1477-7827-6-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Agrawal T., Vats V., Wallace P.K., Singh A., Salhan S., Mittal A. Recruitment of myeloid and plasmacytoid dendritic cells in cervical mucosa during Chlamydia trachomatis infection. Clin. Microbiol. Infect. 2009;15:50–59. doi: 10.1111/j.1469-0691.2008.02113.x. [DOI] [PubMed] [Google Scholar]
  • 43.Agrawal T., Vats V., Salhan S., Mittal A. Determination of chlamydial load and immune parameters in asymptomatic, symptomatic and infertile women. FEMS Immunol. Med. Microbiol. 2009;55:250–257. doi: 10.1111/j.1574-695X.2008.00530.x. [DOI] [PubMed] [Google Scholar]
  • 44.Gupta R., Srivastava P., Vardhan H., Salhan S., Mittal A. Host immune responses to chlamydial inclusion membrane proteins B and C in Chlamydia trachomatis infected women with or without fertility disorders. Reprod. Biol. Endocrinol. 2009;7:38. doi: 10.1186/1477-7827-7-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Jha R., Srivastava P., Salhan S., Finckh A., Gabay C., Mittal A., Bas S. Spontaneous secretion of interleukin-17 and -22 by human cervical cells in Chlamydia trachomatis infection. Microbes Infect. 2011;13:167–178. doi: 10.1016/j.micinf.2010.10.012. [DOI] [PubMed] [Google Scholar]
  • 46.Mlisana K., Naicker N., Werner L., Roberts L., Van Loggerenberg F., Baxter C., Passmore J.A.S., Grobler A.C., Sturm A.W., Williamson C., et al. Symptomatic vaginal discharge is a poor predictor of sexually transmitted infections and genital tract inflammation in high-risk women in South Africa. J. Infect. Dis. 2012;206:6–14. doi: 10.1093/infdis/jis298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ogendi B.M.O., Bakshi R.K., Sabbaj S., Brown L., Lee J.Y., Kapil R., Geisler W.M. Distinct peripheral vs mucosal T-cell phenotypes in chlamydia-infected women. Am. J. Reprod. Immunol. 2017;78 doi: 10.1111/aji.12768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Spear G.T., Kendrick S.R., Chen H.Y., Thomas T.T., Bahk M., Balderas R., Ghosh S., Weinberg A., Landay A.L. Multiplex immunoassay of lower genital tract mucosal fluid from women attending an urban STD clinic shows broadly increased IL1s and lactoferrin. PLoS One. 2011;6 doi: 10.1371/journal.pone.0019560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sperling R., Kraus T.A., Ding J., Veretennikova A., Lorde-Rollins E., Singh T., Lo Y., Quayle A.J., Chang T.L. Differential profiles of immune mediators and in vitro HIV infectivity between endocervical and vaginal secretions from women with Chlamydia trachomatis infection: A pilot study. J. Reprod. Immunol. 2013;99:80–87. doi: 10.1016/j.jri.2013.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Srivastava P., Jha R., Bas S., Salhan S., Mittal A. In infertile women, cells from Chlamydia trachomatis infected site release higher levels of interferon-gamma, interleukin-10 and tumor necrosis factor-alpha upon heat shock protein stimulation than fertile women. Reprod. Biol. Endocrinol. 2008;6:20. doi: 10.1186/1477-7827-6-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Masson L., Salkinder A.L., Olivier A.J., McKinnon L.R., Gamieldien H., Mlisana K., Scriba T.J., Lewis D.A., Little F., Jaspan H.B., et al. Relationship between female genital tract infections, mucosal interleukin-17 production and local T helper type 17 cells. Immunology. 2015;146:557–567. doi: 10.1111/imm.12527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hwang L.Y., Scott M.E., Ma Y., Moscicki A.-B. Diversity of Cervicovaginal Cytokine Response to Incident Chlamydia trachomatis Infection Among a Prospective Cohort of Young Women. Am. J. Reprod. Immunol. 2015;74:228–236. doi: 10.1111/aji.12401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Richmond S.J., Milne J.D., Hilton A.L., Caul E.O. Antibodies to chlamydia trachomatis in cervicovaginal secretions: Relation to serum antibodies and current chlamydial infection. Sex. Transm. Dis. 1980;7:11–15. doi: 10.1097/00007435-198001000-00003. [DOI] [PubMed] [Google Scholar]
  • 54.Levine W.C., Pope V., Bhoomkar A., Tambe P., Lewis J.S., Zaidi A.A., Farshy C.E., Mitchell S., Talkington D.F. Increase in endocervical CD4 lymphocytes among women with nonulcerative sexually transmitted diseases. J. Infect. Dis. 1998;177:167–174. doi: 10.1086/513820. [DOI] [PubMed] [Google Scholar]
  • 55.Marconi C., Santos-Greatti M.M.V., Parada C.M.G.L., Pontes A., Pontes A.G., Giraldo P.C., Donders G.G.G., da Silva M.G. Cervicovaginal Levels of Proinflammatory Cytokines Are Increased During Chlamydial Infection in Bacterial Vaginosis But Not in Lactobacilli-Dominated Flora. J. Low. Genit. Tract Dis. 2014;18:261–265. doi: 10.1097/LGT.0000000000000003. [DOI] [PubMed] [Google Scholar]
  • 56.Jordan S.J., Olson K.M., Barnes S., Wilson L.S., Berryhill T.F., Bakshi R., Brown L.T., Press C.G., Geisler W.M. Lower Levels of Cervicovaginal Tryptophan Are Associated With Natural Clearance of Chlamydia in Women. J. Infect. Dis. 2017;215:1888–1892. doi: 10.1093/infdis/jix240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hedges S.R., Sibley D.A., Mayo M.S., Hook E.W., 3rd, Russell M.W. Cytokine and antibody responses in women infected with Neisseria gonorrhoeae: effects of concomitant infections. J. Infect. Dis. 1998;178:742–751. doi: 10.1086/515372. [DOI] [PubMed] [Google Scholar]
  • 58.Barousse M.M., Theall K.P., Van Der Pol B., Fortenberry J.D., Orr D.P., Fidel P.L. Susceptibility of middle adolescent females to sexually transmitted infections: Impact of hormone contraception and sexual behaviors on vaginal immunity. Am. J. Reprod. Immunol. 2007;58:159–168. doi: 10.1111/j.1600-0897.2007.00504.x. [DOI] [PubMed] [Google Scholar]
  • 59.Mott P.D., Taylor C.M., Lillis R.A., Ardizzone C.M., Albritton H.L., Luo M., Calabresi K.G., Martin D.H., Myers L., Quayle A.J. Differences in the Genital Microbiota in Women Who Naturally Clear Chlamydia trachomatis Infection Compared to Women Who Do Not Clear; A Pilot Study. Front. Cell. Infect. Microbiol. 2021;11 doi: 10.3389/fcimb.2021.615770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Gupta R., Vardhan H., Srivastava P., Salhan S., Mittal A. Modulation of cytokines and transcription factors (T-Bet and GATA3) in CD4 enriched cervical cells of Chlamydia trachomatis infected fertile and infertile women upon stimulation with chlamydial inclusion membrane proteins B and C. Reprod. Biol. Endocrinol. 2009;7:84. doi: 10.1186/1477-7827-7-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Agrawal T., Gupta R., Dutta R., Srivastava P., Bhengraj A.R., Salhan S., Mittal A. Protective or pathogenic immune response to genital chlamydial infection in women-A possible role of cytokine secretion profile of cervical mucosal cells. Clin. Immunol. 2009;130:347–354. doi: 10.1016/j.clim.2008.10.004. [DOI] [PubMed] [Google Scholar]
  • 62.Agrawal T., Bhengraj A.R., Vats V., Salhan S., Mittal A. Expression of TLR 2, TLR 4 and iNOS in Cervical Monocytes of Chlamydia trachomatis-infected Women and Their Role in Host Immune Response. Am. J. Reprod. Immunol. 2011;66:534–543. doi: 10.1111/j.1600-0897.2011.01064.x. [DOI] [PubMed] [Google Scholar]
  • 63.Cohen C.R., Plummer F.A., Mugo N., Maclean I., Shen C., Bukusi E.A., Irungu E., Sinei S., Bwayo J., Brunham R.C. Increased interleukin-10 in the endocervical secretions of women with non-ulcerative sexually transmitted diseases: A mechanism for enhanced HIV-1 transmission? AIDS. 1999;13:327–332. doi: 10.1097/00002030-199902250-00004. [DOI] [PubMed] [Google Scholar]
  • 64.Ficarra M., Ibana J.S.A., Poretta C., Ma L., Myers L., Taylor S.N., Greene S., Smith B., Hagensee M., Martin D.H., Quayle A.J. A Distinct Cellular Profile Is Seen in The Human Endocervix During Chlamydia trachomatis Infection. Am. J. Reprod. Immunol. 2008;60:415–425. doi: 10.1111/j.1600-0897.2008.00639.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ibana J.A., Myers L., Porretta C., Lewis M., Taylor S.N., Martin D.H., Quayle A.J. The major CD8 T cell effector memory subset in the normal and Chlamydia trachomatis-infected human endocervix is low in perforin. BMC Immunol. 2012;13:66. doi: 10.1186/1471-2172-13-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kelly K.A., Wiley D., Wiesmeier E., Briskin M., Butch A., Darville T. 2009. The Combination of the Gastrointestinal Integrin (α4β7) and Selectin Ligand Enhances T-Cell Migration to the Reproductive Tract during Infection with chlamydia Trachomatis; pp. 446–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Scott M.E., Ma Y., Farhat S., Shiboski S., Moscicki A.-B. Covariates of cervical cytokine mRNA expression by real-time PCR in adolescents and young women: effects of Chlamydia trachomatis infection, hormonal contraception, and smoking. J. Clin. Immunol. 2006;26:222–232. doi: 10.1007/s10875-006-9010-x. [DOI] [PubMed] [Google Scholar]
  • 68.Vats V., Agrawal T., Salhan S., Mittal A. Primary and secondary immune responses of mucosal and peripheral lymphocytes during Chlamydia trachomatis infection. FEMS Immunol. Med. Microbiol. 2007;49:280–287. doi: 10.1111/j.1574-695X.2006.00196.x. [DOI] [PubMed] [Google Scholar]
  • 69.Reddy B.S., Rastogi S., Das B., Salhan S., Verma S., Mittal A. Cytokine expression pattern in the genital tract of Chlamydia trachomatis positive infertile women - Implication for T-cell responses. Clin. Exp. Immunol. 2004;137:552–558. doi: 10.1111/j.1365-2249.2004.02564.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Schust D.J., Ibana J.A., Buckner L.R., Ficarra M., Sugimoto J., Amedee A.M., Quayle A.J. Potential mechanisms for increased HIV-1 transmission across the endocervical epithelium during C. trachomatis infection. Curr. HIV Res. 2012;10:218–227. doi: 10.2174/157016212800618093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.McClure C.P., Bowman C.A., Geary I., Ryan C., Ball J.K., Eley A. HIV-1 co-receptor expression and epithelial immune cells of the cervix in asymptomatic women attending a genitourinary medicine clinic. HIV Med. 2013;14:108–114. doi: 10.1111/hiv.12002. [DOI] [PubMed] [Google Scholar]
  • 72.Bua A., Cannas S., Zanetti S., Molicotti P. Levels of different cytokines in women and men with asymptomatic genital infection caused by Chlamydia. J. Infect. Dev. Ctries. 2019;13:847–850. doi: 10.3855/jidc.9810. [DOI] [PubMed] [Google Scholar]
  • 73.Filardo S., Di Pietro M., Tranquilli G., Latino M.A., Recine N., Porpora M.G., Sessa R. Selected Immunological Mediators and Cervical Microbial Signatures in Women with Chlamydia trachomatis Infection. mSystems. 2019;4 doi: 10.1128/mSystems.00094-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Fresse A.S., Sueur J.M., Hamdad F. Diagnosis and follow-up of genital chlamydial infection by direct methods and by detection of serum IgG, IgA and secretory IgA. Indian J. Med. Microbiol. 2010;28:326–331. doi: 10.4103/0255-0857.71823. [DOI] [PubMed] [Google Scholar]
  • 75.Tsai P.Y., Hsu M.C., Huang C.T., Li S.Y. Human antibody and antigen response to IncA antibody of Chlamydia trachomatis. Int. J. Immunopathol. Pharmacol. 2007;20:155–161. doi: 10.1177/039463200702000118. [DOI] [PubMed] [Google Scholar]
  • 76.Fichorova R.N., Morrison C.S., Chen P.-L., Yamamoto H.S., Govender Y., Junaid D., Ryan S., Kwok C., Chipato T., Salata R.A., Doncel G.F. Aberrant cervical innate immunity predicts onset of dysbiosis and sexually transmitted infections in women of reproductive age. PLoS One. 2020;15 doi: 10.1371/journal.pone.0224359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fichorova R.N., Chen P.-L., Morrison C.S., Doncel G.F., Mendonca K., Kwok C., Chipato T., Salata R., Mauck C. The Contribution of Cervicovaginal Infections to the Immunomodulatory Effects of Hormonal Contraception. mBio. 2015;6 doi: 10.1128/mBio.00221-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Witkin S.S., Jeremias J., Neuer A., David S., Kligman I., Toth M., Willner E., Witkin K. Immune Recognition of the 60kD Heat Shock Protein: Implications for Subsequent Fertility. Infect. Dis. Obstet. Gynecol. 1996;4:152–158. doi: 10.1155/S1064744996000336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Witkin S.S., Sultan K.M., Neal G.S., Jeremias J., Grifo J.A., Rosenwaks Z. Unsuspected Chlamydia trachomatis infection and in vitro fertilization outcome. Am. J. Obstet. Gynecol. 1994;171:1208–1214. doi: 10.1016/0002-9378(94)90134-1. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang S., Funahashi Y., Tanaka S., Okubo T., Thapa J., Nakamura S., Higashi H., Yamaguchi H. Chlamydia trachomatis relies on the scavenger role of aryl hydrocarbon receptor with detyrosinated tubulin for its intracellular growth, but this is impaired by excess indole. Microbes Infect. 2023;25 doi: 10.1016/j.micinf.2022.105097. [DOI] [PubMed] [Google Scholar]
  • 81.Terho P., Meurman O. Chlamydial serum IgG, IgA and local IgA antibodies in patients with genital-tract infections measured by solid-phase radioimmunoassay. J. Med. Microbiol. 1981;14:77–87. doi: 10.1099/00222615-14-1-77. [DOI] [PubMed] [Google Scholar]
  • 82.Osser S., Persson K. Postabortal pelvic infection associated with Chlamydia trachomatis and the influence of humoral immunity. Am. J. Obstet. Gynecol. 1984;150:699–703. doi: 10.1016/0002-9378(84)90670-7. [DOI] [PubMed] [Google Scholar]
  • 83.GrÖNroos M., Honkonen E., Terho P., Punnonen R. Cervical and serum IgA and serum IgG antibodies to Chlamydia trachomatis and herpes simplex virus in threatened abortion: a prospective study. BJOG Int. J. Obstet. Gynaecol. 1983;90:167–170. doi: 10.1111/j.1471-0528.1983.tb08903.x. [DOI] [PubMed] [Google Scholar]
  • 84.Honkonen E., Punnonen R., Terho P. Chlamydia trachomatis in term pregnancy. Isolation and serological study with a case report. Int. J. Gynaecol. Obstet. 1983;21:473–476. doi: 10.1016/0020-7292(83)90037-1. [DOI] [PubMed] [Google Scholar]
  • 85.Kalimo K., Terho P., Honkonen E., Grönroos M., Halonen P. Chlamydia trachomatis and herpes simplex virus IgA antibodies in cervical secretions of patients with cervical atypia. Br. J. Obstet. Gynaecol. 1981;88:1130–1134. doi: 10.1111/j.1471-0528.1981.tb01767.x. [DOI] [PubMed] [Google Scholar]
  • 86.Puolakkainen M., Vesterinen E., Purola E., Saikku P., Paavonen J. Persistence of chlamydial antibodies after pelvic inflammatory disease. J. Clin. Microbiol. 1986;23:924–928. doi: 10.1128/jcm.23.5.924-928.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Witkin S.S., Bongiovanni A.M., Inglis S.R. Detection of endocervical anti-Chlamydia trachomatis immunoglobulin A in pregnant women by a rapid, 6-minute enzyme-linked immunosorbent assay: Comparison with PCR and chlamydial antigen detection methods. J. Clin. Microbiol. 1997;35:1781–1783. doi: 10.1128/JCM.35.7.1781-1783.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Audu O., Musa B.O.P., Usman A., Adekunle O.O., Opaluwa S.A., El-Fulaty A.A., Olayemi B., Okwubenata O.L., Ega B., Yaqub Y., Oraebosi M.I. Comparative assessment of Chlamydia trachomatis pathogen prevalence, and the determination of host gyneco-epidemiological and immunological associated risk factors in female infertility. Cytokine. 2025;185 doi: 10.1016/j.cyto.2024.156819. [DOI] [PubMed] [Google Scholar]
  • 89.Darville T., Albritton H.L., Zhong W., Dong L., O'Connell C.M., Poston T.B., Quayle A.J., Goonetilleke N., Wiesenfeld H.C., Hillier S.L., Zheng X. Anti-chlamydia IgG and IgA are insufficient to prevent endometrial chlamydia infection in women, and increased anti-chlamydia IgG is associated with enhanced risk for incident infection. Am. J. Reprod. Immunol. 2019;81 doi: 10.1111/aji.13103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wang C., Tang J., Crowley-Nowick P.A., Wilson C.M., Kaslow R.A., Geisler W.M. Interleukin (IL)-2 and IL-12 responses to Chlamydia trachomatis infection in adolescents. Clin. Exp. Immunol. 2005;142:548–554. doi: 10.1111/j.1365-2249.2005.02946.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wang C., Tang J., Geisler W.M., Crowley-Nowick P.A., Wilson C.M., Kaslow R.A. Human leukocyte antigen and cytokine gene variants as predictors of recurrent Chlamydia trachomatis infection in high-risk adolescents. J. Infect. Dis. 2005;191:1084–1092. doi: 10.1086/428592. [DOI] [PubMed] [Google Scholar]
  • 92.Ziklo N., Vidgen M.E., Taing K., Huston W.M., Timms P. Dysbiosis of the vaginal microbiota and higher vaginal kynurenine/tryptophan ratio reveals an association with Chlamydia trachomatis genital infections. Front. Cell. Infect. Microbiol. 2018;8:1. doi: 10.3389/fcimb.2018.00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ziklo N., Huston W.M., Taing K., Timms P. High expression of IDO1 and TGF-beta1 during recurrence and post infection clearance with Chlamydia trachomatis, are independent of host IFN-gamma response. BMC Infect. Dis. 2019;19:218. doi: 10.1186/s12879-019-3843-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Poston T.B., Lee D.E., Darville T., Zhong W., Dong L., O'Connell C.M., Wiesenfeld H.C., Hillier S.L., Sempowski G.D., Zheng X. Cervical Cytokines Associated With Chlamydia trachomatis Susceptibility and Protection. J. Infect. Dis. 2019;220:330–339. doi: 10.1093/infdis/jiz087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Darougar S., Forsey T., Wood J.J., Bolton J.P., Allan A. Chlamydia and the Curtis-Fitz-Hugh syndrome. Br. J. Vener. Dis. 1981;57:391–394. doi: 10.1136/sti.57.6.391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Schachter J., Cles L., Ray R., Hines P.A. Failure of serology in diagnosing chlamydial infections of the female genital tract. J. Clin. Microbiol. 1979;10:647–649. doi: 10.1128/jcm.10.5.647-649.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Thejls H., Rahm V.A., Gnarpe J., Gnarpe H. Diagnostic efficacy of chlamydial antibodies in cervical secretions from pregnant women and adolescent girls. Genitourin. Med. 1995;71:370–374. doi: 10.1136/sti.71.6.370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Treharne J.D., Darougar S., Simmons P.D., Thin R.N. Rapid diagnosis of chlamydial infection of the cervix. Br. J. Vener. Dis. 1978;54:403–408. doi: 10.1136/sti.54.6.403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Omer E.E., Forsey T., Darougar S., Ali M.H., el-Naeem H.A. Seroepidemiological survey of chlamydial genital infections in Khartoum, Sudan. Genitourin. Med. 1985;61:261–263. doi: 10.1136/sti.61.4.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lewis M.E., Belland R.J., AbdelRahman Y.M., Beatty W.L., Aiyar A.A., Zea A.H., Greene S.J., Marrero L., Buckner L.R., Tate D.J., et al. Morphologic and molecular evaluation of Chlamydia trachomatis growth in human endocervix reveals distinct growth patterns. Front. Cell. Infect. Microbiol. 2014;4:71. doi: 10.3389/fcimb.2014.00071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Mardh P.A., Moller B.R., Ingerselv H.J. Endometritis caused by Chlamydia trachomatis. Br. J. Vener. Dis. 1981;57:191–195. doi: 10.1136/sti.57.3.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Southgate L.J., Treharne J.D., Forsey T. Chlamydia trachomatis and Neisseria gonorrhoeae infections in women attending inner city general practices. Br. Med. J. 1983;287:879–881. doi: 10.1136/bmj.287.6396.879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Cauci S., Culhane J.F. Modulation of vaginal immune response among pregnant women with bacterial vaginosis by Trichomonas vaginalis, Chlamydia trachomatis, Neisseria gonorrhoeae, and yeast. Am. J. Obstet. Gynecol. 2007;196:133.e1–133.e7. doi: 10.1016/j.ajog.2006.08.033. [DOI] [PubMed] [Google Scholar]
  • 104.Chen H., Wang L., Zhao L., Luo L., Min S., Wen Y., Lei W., Shu M., Li Z. Alterations of Vaginal Microbiota in Women With Infertility and Chlamydia trachomatis Infection. Front. Cell. Infect. Microbiol. 2021;11 doi: 10.3389/fcimb.2021.698840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.van den Broek I.V.F., Land J.A., van Bergen J.E.A.M., Morré S.A., van der Sande M.A.B. Chlamydia trachomatis Antibody Testing in Vaginal Mucosal Material versus Blood Samples of Women Attending a Fertility Clinic and an STI Clinic. Obstet. Gynecol. Int. 2014;2014 doi: 10.1155/2014/601932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Cai T., Mondaini N., Migno S., Meacci F., Boddi V., Gontero P., Malossini G., Geppetti P., Mazzoli S., Bartoletti R. Genital Chlamydia trachomatis Infection is Related to Poor Sexual Quality of Life in Young Sexually Active Women. J. Sex. Med. 2011;8:1131–1137. doi: 10.1111/j.1743-6109.2010.02194.x. [DOI] [PubMed] [Google Scholar]
  • 107.Hammerschlag M.R., Alpert S., Rosner I., Thurston P., Semine D., McComb D., McCormack W.M. Microbiology of the vagina in children: Normal and potentially pathogenic organisms. Pediatrics. 1978;62:57–62. [PubMed] [Google Scholar]
  • 108.McCormack W.M., Alpert S., McComb D.E., Nichols R.L., Semine D.Z., Zinner S.H. Fifteen-month follow-up study of women infected with Chlamydia trachomatis. N. Engl. J. Med. 1979;300:123–125. doi: 10.1056/NEJM197901183000305. [DOI] [PubMed] [Google Scholar]
  • 109.Ng K.M., Graham D.M., Forsyth J.R., Brennan K.F. Antichlamydial antibody in genital exudates of men and women with non-gonococcal genital infections. Lancet (London, England) 1978;1:507. doi: 10.1016/s0140-6736(78)90177-0. [DOI] [PubMed] [Google Scholar]
  • 110.Gump D.W., Dickstein S., Gibson M. Endometritis related to Chlamydia trachomatis infection. Ann. Intern. Med. 1981;95:61–63. doi: 10.7326/0003-4819-95-1-61. [DOI] [PubMed] [Google Scholar]
  • 111.McCormack W.M., Rosner B., McComb D.E., Evrard J.R., Zinner S.H. Infection with Chlamydia trachomatis in female college students. Am. J. Epidemiol. 1985;121:107–115. doi: 10.1093/oxfordjournals.aje.a113971. [DOI] [PubMed] [Google Scholar]
  • 112.McComb D.E., Nichols R.L., Semine D.Z., Evrard J.R., Alpert S., Crockett V.A., Rosner B., Zinner S.H., McCormack W.M. Chlamydia trachomatis in women: Antibody in cervical secretions as a possible indicator of genital infection. J. Infect. Dis. 1979;139:628–633. doi: 10.1093/infdis/139.6.628. [DOI] [PubMed] [Google Scholar]
  • 113.Arno J.N., Ricker V.A., Batteiger B.E., Katz B.P., Caine V.A., Jones R.B. Interferon-gamma in endocervical secretions of women infected with Chlamydia trachomatis. J. Infect. Dis. 1990;162:1385–1389. doi: 10.1093/infdis/162.6.1385. [DOI] [PubMed] [Google Scholar]
  • 114.Ruijs G.J., Kauer F.M., Jager S., Schröder F.P., Schirm J., Kremer J. Further details on sequelae at the cervical and tubal level of Chlamydia trachomatis infection in infertile women. Fertil. Steril. 1991;56:20–26. doi: 10.1016/s0015-0282(16)54410-0. [DOI] [PubMed] [Google Scholar]
  • 115.Workowski K.A., Lampe M.F., Wong K.G., Watts M.B., Stamm W.E. Long-term Eradication of Chlamydia trachomatis Genital Infection After Antimicrobial Therapy: Evidence Against Persistent Infection. JAMA, J. Am. Med. Assoc. 1993;270:2071–2075. doi: 10.1001/jama.1993.03510170061031. [DOI] [PubMed] [Google Scholar]
  • 116.Brunham R.C., Kuo C.C., Cles L., Holmes K.K. Correlation of host immune response with quantitative recovery of Chlamydia trachomatis from the human endocervix. Infect. Immun. 1983;39:1491–1494. doi: 10.1128/iai.39.3.1491-1494.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Mahmoud E.A., Hamad E.E., Bassiri M., Mardh P.A. Antichlamydial activity of cervical secretion. Journal of Obstetrics and Gynaecology. 1994;14:166–171. doi: 10.3109/01443619409004068. [DOI] [Google Scholar]
  • 118.Persson E., Eneroth P., Grillner L. Immunoglobulin contents in cervical secretions of women with chlamydial cervicitis. Gynecol. Obstet. Invest. 1990;30:109–113. doi: 10.1159/000293229. [DOI] [PubMed] [Google Scholar]
  • 119.Mahmoud E.A., Svensson L.O., Olsson S.E., Mårdh P.A. Antichlamydial activity of vaginal secretion. Am. J. Obstet. Gynecol. 1995;172:1268–1272. doi: 10.1016/0002-9378(95)91491-9. [DOI] [PubMed] [Google Scholar]
  • 120.Abraham S., Juel H.B., Bang P., Cheeseman H.M., Dohn R.B., Cole T., Kristiansen M.P., Korsholm K.S., Lewis D., Olsen A.W., et al. Safety and immunogenicity of the chlamydia vaccine candidate CTH522 adjuvanted with CAF01 liposomes or aluminium hydroxide: a first-in-human, randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Infect. Dis. 2019;19:1091–1100. doi: 10.1016/S1473-3099(19)30279-8. [DOI] [PubMed] [Google Scholar]
  • 121.Garrett N., Mtshali A., Osman F., Masson L., McKinnon L.R., Singh R., Mitchev N., Ngobese H., Kharsany A.B.M., Abdool Karim S., et al. Impact of point-of-care testing and treatment of sexually transmitted infections and bacterial vaginosis on genital tract inflammatory cytokines in a cohort of young South African women. Sex. Transm. Infect. 2021;97:555–565. doi: 10.1136/sextrans-2020-054740. [DOI] [PubMed] [Google Scholar]
  • 122.Cai T., Wagenlehner F.M.E., Mondaini N., D'Elia C., Meacci F., Migno S., Malossini G., Mazzoli S., Bartoletti R. Effect of human papillomavirus and Chlamydia trachomatis co-infection on sperm quality in young heterosexual men with chronic prostatitis-related symptoms. BJU Int. 2014;113:281–287. doi: 10.1111/bju.12244. [DOI] [PubMed] [Google Scholar]
  • 123.Martínez-Prado E., Camejo Bermúdez M.I. Expression of IL-6, IL-8, TNF-α, IL-10, HSP-60, Anti-HSP-60 Antibodies, and Anti-sperm Antibodies, in Semen of Men with Leukocytes and/or Bacteria. Am. J. Reprod. Immunol. 2010;63:233–243. doi: 10.1111/j.1600-0897.2009.00786.x. [DOI] [PubMed] [Google Scholar]
  • 124.Mazzoli S., Cai T., Rupealta V., Gavazzi A., Castricchi Pagliai R., Mondaini N., Bartoletti R. Interleukin 8 and Anti-Chlamydia trachomatis Mucosal IgA as Urogenital Immunologic Markers in Patients with C. trachomatis Prostatic Infection. Eur. Urol. 2007;51:1385–1393. doi: 10.1016/j.eururo.2006.10.059. [DOI] [PubMed] [Google Scholar]
  • 125.Dehghan Marvast L., Aflatoonian A., Talebi A.R., Ghasemzadeh J., Pacey A.A. Semen inflammatory markers and Chlamydia trachomatis infection in male partners of infertile couples. Andrologia. 2016;48:729–736. doi: 10.1111/and.12501. [DOI] [PubMed] [Google Scholar]
  • 126.Karaulov A.V., Markelova E.V., Chepurnova N.S., Tulupova M.S., Nevezhkina T.A., Hamoshina M.B., Zhdanova O.L., Mizanova V.Z., Ivanjuk N.A. Cytokine profile and intracellular matrix component degradation indicators in male ejaculate with chronic urethritis before and following the treatment. Asian J. Pharm. 2017;11:S865–S872. [Google Scholar]
  • 127.Pérez-Soto E., Oros-Pantoja R., Fernández-Martínez E., Carbonell-Campos J.M., Sánchez Monroy V. Seminal pro-inflammatory cytokines and pH are affected by Chlamydia infection in asymptomatic patients with teratozoospermia. Cent. Eur. J. Immunol. 2021;46:76–81. doi: 10.5114/ceji.2021.105247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Mazzoli S., Cai T., Addonisio P., Bechi A., Mondaini N., Bartoletti R. Chlamydia trachomatis Infection Is Related to Poor Semen Quality in Young Prostatitis Patients. Eur. Urol. 2010;57:708–714. doi: 10.1016/j.eururo.2009.05.015. [DOI] [PubMed] [Google Scholar]
  • 129.Hakimi H., Akhondi M.M., Sadeghi M.R., Chamani L., Arababadi M.K., Ahmadabadi B.N., Hassanshahi G., Fathollahi M.S. Seminal Levels of IL-10, IL-12, and IL-17 in Men with Asymptomatic Chlamydia Infection. Inflammation. 2014;37:122–126. doi: 10.1007/s10753-013-9719-7. [DOI] [PubMed] [Google Scholar]
  • 130.Hakimi H., Zainodini N., Khorramdelazad H., Kazemi Arababadi M., Hassanshahi G. Seminal levels of pro-inflammatory (CXCL1, CXCL9, CXCL10) and homeostatic (CXCL12) chemokines in men with asymptomatic Chlamydia trachomatis infection. Jundishapur J. Microbiol. 2014;7 doi: 10.5812/jjm.11152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Habermann B., Krause W. Altered sperm function or sperm antibodies are not associated with chlamydial antibodies in infertile men with leucocytospermia. J. Eur. Acad. Dermatol. Venereol. 1999;12:25–29. doi: 10.1111/j.1468-3083.1999.tb00803.x. [DOI] [PubMed] [Google Scholar]
  • 132.Moazenchi M., Totonchi M., Salman Yazdi R., Hratian K., Mohseni Meybodi M.A., Ahmadi Panah M., Chehrazi M., Mohseni Meybodi A. The impact of Chlamydia trachomatis infection on sperm parameters and male fertility: A comprehensive study. Int. J. STD AIDS. 2018;29:466–473. doi: 10.1177/0956462417735245. [DOI] [PubMed] [Google Scholar]
  • 133.Markelova E.V., Chepurnova N.S., Tulupova M.S., Karaulov A.V., Baibarina E.V., Mel'nikova Y.A., Shadrina M.N., Yakovleva Y., Rabota N.S., Ziangirova Y.V. Study of the system of cytokines in case of herpetic and chlamydia-herpetic infection. Journal of Global Pharma Technology. 2016;8:10–14. [Google Scholar]
  • 134.Gdoura R., Daoudi F., Bouzid F., Ben Salah F., Chaigneau C., Sueur J.M., Eb F., Rekik S., Hammami A., Orfila J. Detection of Chlamydia trachomatis in semen and urethral specimens from male members of infertile couples in Tunisia. Eur. J. Contracept. Reprod. Health Care. 2001;6:14–20. [PubMed] [Google Scholar]
  • 135.Samra Z., Soffer Y., Pansky M. Prevalence of genital chlamydia and mycoplasma infection in couples attending a male infertility clinic. Eur. J. Epidemiol. 1994;10:69–73. doi: 10.1007/BF01717455. [DOI] [PubMed] [Google Scholar]
  • 136.Penna Videau S., Cermeno Vivas J., Salazar N. IgA antibodies to Chlamydia trachomatis and seminal parameters in asymptomatic infertile males. Arch. Androl. 2001;46:189–195. doi: 10.1080/01485010151096469. [DOI] [PubMed] [Google Scholar]
  • 137.El Feky M.A., Hassan E.A.S., El Din A.M.E., Hofny E.R.M., Afifi N.A.H., Eldin S.S.S., Baker M.O. Chlamydia trachomatis: methods of identification and impact on semen quality. Egypt. J. Immunol. 2009;16:49–59. [PubMed] [Google Scholar]
  • 138.Ruijs G.J., Kauer F.M., Jager S., Schröder P.F., Schirm J., Kremer J. Is serology of any use when searching for correlations between Chlamydia trachomatis infection and male infertility? Fertil. Steril. 1990;53:131–136. doi: 10.1016/s0015-0282(16)53228-2. [DOI] [PubMed] [Google Scholar]
  • 139.Dieterle S., Mahony J.B., Luinstra K.E., Stibbe W. Immunology: Chlamydial immunoglobulin IgG and IgA antibodies in serum and semen are not associated with the presence of chlamydia trachomatis DNA or rRNA in semen from male partners of infertile couples. Hum. Reprod. 1995;10:315–319. doi: 10.1093/oxfordjournals.humrep.a135934. [DOI] [PubMed] [Google Scholar]
  • 140.Eggert-Kruse W., Buhlinger-Göpfarth N., Rohr G., Probst S., Aufenanger J., Näher H., Runnebaum B. Antibodies to Chlamydia trachomatis in semen and relationship with parameters of male fertility. Hum. Reprod. 1996;11:1408–1417. doi: 10.1093/oxfordjournals.humrep.a019410. [DOI] [PubMed] [Google Scholar]
  • 141.EzzEl-Din A.M., Gaber H.D., Kamal D.T. Chlamydia trachomatis Infection: its relation to semen parameters and sperm DNA integrity. Egypt. J. Immunol. 2021;28:290–298. [PubMed] [Google Scholar]
  • 142.Kojima H., Wang S.P., Kuo C.C., Grayston J.T. Local antibody in semen for rapid diagnosis of Chlamydia trachomatis epididymitis. J. Urol. 1988;140:528–531. doi: 10.1016/S0022-5347(17)41710-1. [DOI] [PubMed] [Google Scholar]
  • 143.Munoz M.G., Witkin S.S. Immunology: Autoimmunity to spermatozoa, asymptomatic chlamydia trachomatis genital tract infection and γδ t lymphocytes in seminal fluid from the male partners of couples with unexplained infertility. Hum. Reprod. 1995;10:1070–1074. doi: 10.1093/oxfordjournals.humrep.a136096. [DOI] [PubMed] [Google Scholar]
  • 144.Suominen J., Grönroos M., Terho P., Wichmann L. Chronic prostatitis, chlamydia trachomatis and infertility. Int. J. Androl. 1983;6:405–413. doi: 10.1111/j.1365-2605.1983.tb00555.x. [DOI] [PubMed] [Google Scholar]
  • 145.Weidner W., Floren E., Zimmermann O., Thiele D., Ludwig M. Chlamydial antibodies in semen: Search for ''silent'' chlamydial infections in asymptomatic andrological patients. Infection. 1996;24:309–313. doi: 10.1007/BF01743366. [DOI] [PubMed] [Google Scholar]
  • 146.Eggert-Kruse W., Boit R., Rohr G., Aufenanger J., Hund M., Strowitzki T. Relationship of seminal plasma interleukin (IL)-8 and IL-6 with semen quality. Hum. Reprod. 2001;16:517–528. doi: 10.1093/humrep/16.3.517. [DOI] [PubMed] [Google Scholar]
  • 147.Eggert-Kruse W., Kiefer I., Beck C., Demirakca T., Strowitzki T. Role for tumor necrosis factor alpha (TNF-α) and interleukin 1-beta (IL-1β) determination in seminal plasma during infertility investigation. Fertil. Steril. 2007;87:810–823. doi: 10.1016/j.fertnstert.2006.08.103. [DOI] [PubMed] [Google Scholar]
  • 148.Eggert-Kruse W., Neuer A., Clussmann C., Boit R., Geissler W., Rohr G., Strowitzki T. Seminal antibodies to human 60kd heat shock protein (HSP 60) in male partners of subfertile couples. Hum. Reprod. 2002;17:726–735. doi: 10.1093/humrep/17.3.726. [DOI] [PubMed] [Google Scholar]
  • 149.Cai T., Wagenlehner F.M.E., Mazzoli S., Meacci F., Mondaini N., Nesi G., Tiscione D., Malossini G., Bartoletti R. Semen quality in patients with Chlamydia trachomatis genital infection treated concurrently with prulifloxacin and a phytotherapeutic agent. J. Androl. 2012;33:615–623. doi: 10.2164/jandrol.111.013961. [DOI] [PubMed] [Google Scholar]
  • 150.Mazzoli S., Meacci F., Cosco E., Poggiali C. Clinical consequences of immune responses to Chlamydia in men. Infect. Dis. Obstet. Gynecol. 1996;4:136–142. doi: 10.1155/S1064744996000300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Bjercke S., Purvis K. Chlamydial serology in the investigation of infertility. Hum. Reprod. 1992;7:621–624. doi: 10.1093/oxfordjournals.humrep.a137707. [DOI] [PubMed] [Google Scholar]
  • 152.Bollmann R., Engel S., Sagert D., Göbel U.B. Investigations on the detection of Chlamydia trachomatis infections in infertile male outpatients. Andrologia. 1998;30:23–27. doi: 10.1111/j.1439-0272.1998.tb02822.x. [DOI] [PubMed] [Google Scholar]
  • 153.Bollmann R., Engel S., Petzoldt R., Göbel U.B. Chlamydia trachomatis in andrologic patients - Direct and indirect detection. Infection. 2001;29:113–118. doi: 10.1007/s15010-001-0088-z. [DOI] [PubMed] [Google Scholar]
  • 154.Eggert-Kruse W., Weltin M., Strowitzki T. Are Chlamydial Lipopolysaccharide-directed Antibodies in Seminal Plasma or Serum Clinically Significant During Investigation of Male Infertility? Urology. 2011;77:1101–1106. doi: 10.1016/j.urology.2010.11.014. [DOI] [PubMed] [Google Scholar]
  • 155.Ochsendorf F., Özdemir K., Rabenau H., Fenner T., Oremek R., Milbradt R., Doerr H.W. Chlamydia trachomatis and male infertility: Chlamydia-IgA antibodies in seminal plasma are C. trachomatis specific and associated with an inflammatory response. J. Eur. Acad. Dermatol. Venereol. 1999;12:143–152. doi: 10.1016/S0926-9959(98)00150-0. [DOI] [PubMed] [Google Scholar]
  • 156.Pérez-Soto E., Fernández-Martínez E., Oros-Pantoja R., Medel-Flores O., Mira-Covarrubias J.C., Sánchez-Monroy V. Proinflammatory and oxidative stress states induced by human papillomavirus and chlamydia trachomatis coinfection affect sperm quality in asymptomatic infertile men. Medicina (Lithuania) 2021;57 doi: 10.3390/medicina57090862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Kokab A., Akhondi M.M., Sadeghi M.R., Modarresi M.H., Aarabi M., Jennings R., Pacey A.A., Eley A. Raised Inflammatory Markers in Semen From Men With Asymptomatic Chlamydial Infection. J. Androl. 2010;31:114–120. doi: 10.2164/jandrol.109.008300. [DOI] [PubMed] [Google Scholar]
  • 158.Wolff H., Neubert U., Zebhauser M., Bezold G., Korting H.C., Meurer M. Chlamydia trachomatis induces an inflammatory response in the male genital tract and is associated with altered semen quality. Fertil. Steril. 1991;55:1017–1019. doi: 10.1016/S0015-0282(16)54318-0. [DOI] [PubMed] [Google Scholar]
  • 159.Wolff H., Neubert U., Volkenandt M., Zöchling N., Schlüpen E.M., Bezold G., Meurer M. Detection of Chlamydia trachomatis in semen by antibody-enzyme immunoassay compared with polymerase chain reaction, antigen-enzyme immunoassay, and urethral cell culture. Fertil. Steril. 1994;62:1250–1254. doi: 10.1016/s0015-0282(16)57194-5. [DOI] [PubMed] [Google Scholar]
  • 160.Segnini A., Camejo M.I., Proverbio F. Chlamydia trachomatis and sperm lipid peroxidation in infertile men. Asian J. Androl. 2003;5:47–49. [PubMed] [Google Scholar]
  • 161.Motrich R.D., Cuffini C., Oberti J.P.M., Maccioni M., Rivero V.E. Chlamydia trachomatis occurrence and its impact on sperm quality in chronic prostatitis patients. J. Infect. 2006;53:175–183. doi: 10.1016/j.jinf.2005.11.007. [DOI] [PubMed] [Google Scholar]
  • 162.Jungwirth A., Straberger A., Esterbauer B., Fink K., Schmeller N. 2003. Acrosome Reaction in Chlamydia-Positive and Negative Patients; pp. 314–316. [PubMed] [Google Scholar]
  • 163.Nasr El-din A., Sorour H., Fattouh M., Abu El-Hamd M. Evaluation of the role of Chlamydia trachomatis in primary male infertility. Int. J. Clin. Pract. 2021;75 doi: 10.1111/ijcp.14702. [DOI] [PubMed] [Google Scholar]
  • 164.Munoz M.G., Jeremias J., Witkin S.S. The 60 kDa heat shock protein in human semen: Relationship with antibodies to spermatozoa and Chlamydia trachomatis. Hum. Reprod. 1996;11:2600–2603. doi: 10.1093/oxfordjournals.humrep.a019177. [DOI] [PubMed] [Google Scholar]
  • 165.Witkin S.S., Kligman I., Bongiovanni A.M. Relationship between an asymptomatic male genital tract exposure to Chlamydia trachomatis and an autoimmune response to spermatozoa. Hum. Reprod. 1995;10:2952–2955. doi: 10.1093/oxfordjournals.humrep.a135827. [DOI] [PubMed] [Google Scholar]
  • 166.Yoshida K., Kobayashi N., Negishi T. Chlamydia trachomatis infection in the semen of asymptomatic infertile men: detection of the antigen by in situ hybridization. Urol. Int. 1994;53:217–221. doi: 10.1159/000282675. [DOI] [PubMed] [Google Scholar]
  • 167.Gruschwitz M.S., Brezinschek R., Brezinschek H.P. Cytokine levels in the seminal plasma of infertile males. J. Androl. 1996;17:158–163. [PubMed] [Google Scholar]
  • 168.Pate M.S., Hedges S.R., Sibley D.A., Russell M.W., Hook E.W., Mestecky J. Urethral cytokine and immune responses in Chlamydia trachomatis-infected males. Infect. Immun. 2001;69:7178–7181. doi: 10.1128/IAI.69.11.7178-7181.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Shahmanesh M. Characteristics of inflammatory cells in urethral smears from men with non-gonococcal urethritis. Genitourin. Med. 1989;65:18–21. doi: 10.1136/sti.65.1.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Ostaszewska-Puchalska I., Zdrodowska-Stefanow B., Badyda J., Bułhak-Kozioł V., Puciło K., Darewicz B. Antichlamydial antibodies in the serum and expressed prostatic secretion in prostatitis. Arch. Immunol. Ther. Exp. 2004;52:277–283. [PubMed] [Google Scholar]
  • 171.Mardh P.A., Ripa K.T., Colleen S., Treharne J.D., Darougar S. Role of Chlamydia trachomatis in non-acute prostatitis. Br. J. Vener. Dis. 1978;54:330–334. doi: 10.1136/sti.54.5.330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Shortliffe L.M., Sellers R.G., Schachter J. THE CHARACTERIZATION OF NONBACTERIAL PROSTATITIS - SEARCH FOR AN ETIOLOGY. J. Urol. 1992;148:1461–1466. doi: 10.1016/s0022-5347(17)36940-9. [DOI] [PubMed] [Google Scholar]
  • 173.Ito S., Horie K., Seike K., Yasuda M., Tsuchiya T., Yokoi S., Nakano M., Deguchi T. Usefulness of quantifying leukocytes in first-voided urine to predict positivity for Chlamydia trachomatis in asymptomatic men at high risk for chlamydial infection. J. Infect. Chemother. 2014;20:748–751. doi: 10.1016/j.jiac.2014.08.002. [DOI] [PubMed] [Google Scholar]
  • 174.Wiggins R.C., Holmes C.H., Andersson M., Ibrahim F., Low N., Horner P.J. Quantifying leukocytes in first catch urine provides new insights into our understanding of symptomatic and asymptomatic urethritis. Int. J. STD AIDS. 2006;17:289–295. doi: 10.1258/095646206776790268. [DOI] [PubMed] [Google Scholar]
  • 175.Shahmanesh M., Pandit P.G., Round R. Urethral lymphocyte isolation in non-gonococcal urethritis. Genitourin. Med. 1996;72:362–364. doi: 10.1136/sti.72.5.362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Ohsawa I., Ohi H., Endo M., Fujita T., Hidaka M., Satomura A., Yamaguchi Y. A case of renal involvement in persistent immune activation caused by chlamydial salpingitis. Virchows Arch. 2001;438:306–311. doi: 10.1007/s004280000314. [DOI] [PubMed] [Google Scholar]
  • 177.Ray A., Bhati T., Arora R., Rastogi S. Progesterone-mediated immunoregulation of cytokine signaling by miRNA-133a and 101-3p in Chlamydia trachomatis-associated recurrent spontaneous abortion. Mol. Immunol. 2023;164:47–57. doi: 10.1016/j.molimm.2023.10.012. [DOI] [PubMed] [Google Scholar]
  • 178.Ray A., Pradhan D., Siraj F., Arora R., Rastogi S. MicroRNA mediated regulation of oxidative stress and cytokines in Chlamydia trachomatis-infected recurrent spontaneous abortion: A case-control study. Am. J. Reprod. Immunol. 2024;91 doi: 10.1111/aji.13821. [DOI] [PubMed] [Google Scholar]
  • 179.Lugo L.Z.A., Puga M.A.M., Jacob C.M.B., Padovani C.T.J., Nocetti M.C., Tupiná M.S., Pina A.F.S., de Freitas J.N.M., Ferreira A.M.T., Fernandes C.E.D.S., et al. Cytokine profiling of samples positive for Chlamydia trachomatis and Human papillomavirus. PLoS One. 2023;18 doi: 10.1371/journal.pone.0279390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Hammerschlag M.R., Anderka M., Semine D.Z., McComb D., McCormack W.M. Prospective study of maternal and infantile infection with Chlamydia trachomatis. Pediatrics. 1979;64:142–148. [PubMed] [Google Scholar]
  • 181.Wang S.P., Grayston J.T. Human serology in Chlamydia trachomatis infection with microimmunofluorescence. J. Infect. Dis. 1974;130:388–397. doi: 10.1093/infdis/130.4.388. [DOI] [PubMed] [Google Scholar]
  • 182.Waters M.B., Hybiske K., Ikeda R., Kaltenboeck B., Manhart L.E., Kreisel K.M., Khosropour C.M. Chlamydia trachomatis Seroassays Used in Epidemiologic Research: A Narrative Review and Practical Considerations. J. Infect. Dis. 2024;230:250–262. doi: 10.1093/infdis/jiae199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Treharne J.D., Darougar S., Jones B.R. Modification of Microimmunofluorescence Test to Provide a Routine Serodiagnostic Test for Chlamydial Infection. J. Clin. Pathol. 1977;30:510–517. doi: 10.1136/jcp.30.6.510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Richmond S.J., Caul E.O. Fluorescent antibody studies in Chlamydial infections. J. Clin. Microbiol. 1975;1:345–352. doi: 10.1128/jcm.1.4.345-352.1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Lauvau G., Loke P., Hohl T.M. Monocyte-mediated defense against bacteria, fungi, and parasites. Semin. Immunol. 2015;27:397–409. doi: 10.1016/j.smim.2016.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Coughlan L., Lambe T. Measuring Cellular Immunity to Influenza: Methods of Detection, Applications and Challenges. Vaccines (Basel) 2015;3:293–319. doi: 10.3390/vaccines3020293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Kari L., Bakios L.E., Goheen M.M., Bess L.N., Watkins H.S., Southern T.R., Song L., Whitmire W.M., Olivares-Zavaleta N., Caldwell H.D. Antibody signature of spontaneous clearance of Chlamydia trachomatis ocular infection and partial resistance against re-challenge in a nonhuman primate trachoma model. PLoS Negl. Trop. Dis. 2013;7 doi: 10.1371/journal.pntd.0002248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Hanna L., Jawetz E., Dawson C.R., Thygeson P. Long-Term Clinical, Microbiological, and Immunological Observations of a Volunteer Repeatedly Infected with Chlamydia-Trachomatis. J. Clin. Microbiol. 1982;16:895–900. doi: 10.1128/Jcm.16.5.895-900.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Kumar A., Mittal A. Production and characterization of monoclonal antibodies to Chlamydia trachomatis. Hybridoma. 2006;25:293–299. doi: 10.1089/hyb.2006.25.293. [DOI] [PubMed] [Google Scholar]
  • 190.Zhao Q., Lammel C.J., Lindquist E.A., Stephens R.S. Recall of original serologic response after challenge with homologous and heterologous Chlamydia trachomatis serovars. J. Infect. Dis. 1996;173:609–618. doi: 10.1093/infdis/173.3.609. [DOI] [PubMed] [Google Scholar]
  • 191.de Vries H.J.C., Pannekoek Y., Dean D., Bavoil P.M., Borel N., Greub G., Morré S.A., ICSP Subcommittee on the Taxonomy of Chlamydiae Call for consensus in Chlamydia trachomatis nomenclature: moving from biovars, serovars, and serotypes to genovariants and genotypes. Clin. Microbiol. Infect. 2022;28:761–763. doi: 10.1016/j.cmi.2022.02.013. [DOI] [PubMed] [Google Scholar]
  • 192.McKinnon L.R., Hughes S.M., De Rosa S.C., Martinson J.A., Plants J., Brady K.E., Gumbi P.P., Adams D.J., Vojtech L., Galloway C.G., et al. Optimizing Viable Leukocyte Sampling from the Female Genital Tract for Clinical Trials: An International Multi-Site Study. PLoS One. 2014;9 doi: 10.1371/journal.pone.0085675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Lund J.M., Hladik F., Prlic M. Advances and challenges in studying the tissue-resident T cell compartment in the human female reproductive tract. Immunol. Rev. 2023;316:52–62. doi: 10.1111/imr.13212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Quesnel A., CuUvin S., Murphy D., Ashley R.L., Flanigan T., Neutra M.R. Comparative analysis of methods for collection and measurement of immunoglobulins in cervical and vaginal secretions of women. J. Immunol. Methods. 1997;202:153–161. doi: 10.1016/S0022-1759(97)00003-3. [DOI] [PubMed] [Google Scholar]
  • 195.de Melo Kuil L., Lorenzi A.T., Stein M.D., Resende J.C.P., Antoniazzi M., Longatto-Filho A., Scapulatempo-Neto C., Fregnani J.H.T.G. The Role of Self-Collection by Vaginal Lavage for the Detection of HPV and High-Grade Intraepithelial Neoplasia. Acta Cytol. 2017;61:425–433. doi: 10.1159/000477331. [DOI] [PubMed] [Google Scholar]
  • 196.Verhoef V.M.J., Dijkstra M.G., Bosgraaf R.P., Hesselink A.T., Melchers W.J.G., Bekkers R.L.M., Berkhof J., van Kemenade F.J. A second generation cervico-vaginal lavage device shows similar performance as its preceding version with respect to DNA yield and HPV DNA results. BMC Womens Health. 2013;13 doi: 10.1186/1472-6874-13-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Leinonen M.K., Schee K., Jonassen C.M., Lie A.K., Nystrand C.F., Rangberg A., Furre I.E., Johansson M.J., Tropé A., Sjøborg K.D., et al. Safety and acceptability of human papillomavirus testing of self-collected specimens: A methodologic study of the impact of collection devices and HPV assays on sensitivity for cervical cancer and high-grade lesions. J. Clin. Virol. 2018;99–100:22–30. doi: 10.1016/j.jcv.2017.12.008. [DOI] [PubMed] [Google Scholar]
  • 198.Draper E., Bissett S.L., Howell-Jones R., Waight P., Soldan K., Jit M., Andrews N., Miller E., Beddows S. A Randomized, Observer-Blinded Immunogenicity Trial of Cervarix® and Gardasil® Human Papillomavirus Vaccines in 12-15 Year Old Girls. PLoS One. 2013;8 doi: 10.1371/journal.pone.0061825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Peters M.Q., Domenjo-Vila E., Carlson M., Armistead B., Edlefsen P.T., Gasper M., Dabee S., Whidbey C., Jaspan H.B., Prlic M., Harrington W.E. A Noninvasive Method to Sample Immune Cells in the Lower Female Genital Tract Using Menstrual Discs. Immunohorizons. 2024;8:182–192. doi: 10.4049/immunohorizons.2300105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Mkhize N.N., Durgiah R., Ashley V., Archary D., Garrett N.J., Karim Q.A., Karim S.S.A., Moore P.L., Yates N., Passmore J.A.S., et al. Broadly neutralizing antibody specificities detected in the genital tract of HIV-1 infected women. AIDS. 2016;30:1005–1014. doi: 10.1097/QAD.0000000000001038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Jecht E.W., Poon C.H. Preparation of Sperm-Free Seminal Plasma from Human Semen. Fertil. Steril. 1975;26:1–5. doi: 10.1016/s0015-0282(16)40868-x. [DOI] [PubMed] [Google Scholar]
  • 202.Parsons M.S., Madhavi V., Ana-Sosa-Batiz F., Center R.J., Wilson K.M., Bunupuradah T., Ruxrungtham K., Kent S.J. Brief Report: Seminal Plasma Anti-HIV Antibodies Trigger Antibody-dependent Cellular Cytotoxicity: Implications for HIV Transmission. J. Acquir. Immune Defic. Syndr. 2016;71:17–23. doi: 10.1097/QAI.0000000000000804. [DOI] [PubMed] [Google Scholar]
  • 203.Nadler R.B., Koch A.E., Calhoun E.A., Campbell P.L., Pruden D.L., Bennett C.L., Yarnold P.R., Schaeffer A.J. IL-1beta and TNF-alpha in prostatic secretions are indicators in the evaluation of men with chronic prostatitis. J. Urol. 2000;164:214–218. [PubMed] [Google Scholar]
  • 204.Teblick L., Lipovac M., Molenberghs F., Delputte P., De Vos W.H., Vorsters A. HPV-specific antibodies in female genital tract secretions captured via first-void urine retain their neutralizing capacity. Hum Vacc Immunother. 2024;20 doi: 10.1080/21645515.2024.2330168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Centers for Disease Control and Prevention Recommendations for the Laboratory-Based Detection of Chlamydia trachomatis and Neisseria gonorrhoeae. MMWR (Morb. Mortal. Wkly. Rep. 2014;63:1–19. [PMC free article] [PubMed] [Google Scholar]
  • 206.Aaron K.J., Griner S., Footman A., Boutwell A., van der Pol B. Vaginal Swab vs Urine for Detection of Neisseria gonorrhoeae, and Trichomonas vaginalis: A Meta-Analysis. Ann. Fam. Med. 2023;21:172–179. doi: 10.1370/afm.2942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Van Keer S., Peeters E., Vanden Broeck D., De Sutter P., Donders G., Doyen J., Tjalma W.A.A., Weyers S., Vorsters A., Arbyn M. Clinical and analytical evaluation of the RealTime High Risk HPV assay in Colli-Pee collected first-void urine using the VALHUDES protocol. Gynecol. Oncol. 2021;162:575–583. doi: 10.1016/j.ygyno.2021.06.010. [DOI] [PubMed] [Google Scholar]
  • 208.Ludwig M., Steltz C., Huwe P., Schäffer R., Altmannsberger M., Weidner W. Immunocytological analysis of leukocyte subpopulations in urine specimens before and after prostatic massage. Eur. Urol. 2001;39:277–282. doi: 10.1159/000052453. [DOI] [PubMed] [Google Scholar]
  • 209.Fujita K., Ewing C.M., Isaacs W.B., Pavlovich C.P. Immunomodulatory IL-18 binding protein is produced by prostate cancer cells and its levels in urine and serum correlate with tumor status. Int. J. Cancer. 2011;129:424–432. doi: 10.1002/ijc.25705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Cao Y.Z., Friedmankien A.E., Mirabile M., Li X.L., Alam M., Dieterich D., Ho D.D. Hiv-1 Neutralizing Antibodies in Urine from Seropositive Individuals. J. Acquir. Immune Defic. Syndr. 1990;3:195–199. [PubMed] [Google Scholar]
  • 211.Kellogg J.A., Manzella J.P., Seiple J.W., Fortna S.J., Cook J.W., Levisky J.S. Efficacy of an enzyme-linked immunosorbent assay for detection of urinary tract immunoglobulins for diagnosis of urinary tract infections. J. Clin. Microbiol. 1992;30:1711–1715. doi: 10.1128/jcm.30.7.1711-1715.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Gibb A.P., Edmond D.M. URINARY IGG ANTIBODY AGAINST MIXED HEAT-KILLED COLIFORM ANTIGEN AND LIPOPOLYSACCHARIDE CORE ANTIGEN. J. Clin. Pathol. 1992;45:161–164. doi: 10.1136/jcp.45.2.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Drage L.K.L., Robson W., Mowbray C., Ali A., Perry J.D., Walton K.E., Harding C., Pickard R., Hall J., Aldridge P.D. Elevated urine IL-10 concentrations associate with Escherichia coli persistence in older patients susceptible to recurrent urinary tract infections. Immun. Ageing. 2019;16:16. doi: 10.1186/s12979-019-0156-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Woodhall S.C., Gorwitz R.J., Migchelsen S.J., Gottlieb S.L., Horner P.J., Geisler W.M., Winstanley C., Hufnagel K., Waterboer T., Martin D.L., et al. Advancing the public health applications of Chlamydia trachomatis serology. Lancet Infect. Dis. 2018;18:e399–e407. doi: 10.1016/S1473-3099(18)30159-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Woodhall S.C., Wills G.S., Horner P.J., Craig R., Mindell J.S., Murphy G., McClure M.O., Soldan K., Nardone A., Johnson A.M. Chlamydia trachomatis Pgp3 Antibody Population Seroprevalence before and during an Era of Widespread Opportunistic Chlamydia Screening in England (1994-2012) PLoS One. 2017;12 doi: 10.1371/journal.pone.0152810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Horner P.J., Wills G.S., Righarts A., Vieira S., Kounali D., Samuel D., Winston A., Muir D., Dickson N.P., McClure M.O. Chlamydia trachomatis Pgp3 Antibody Persists and Correlates with Self-Reported Infection and Behavioural Risks in a Blinded Cohort Study. PLoS One. 2016;11 doi: 10.1371/journal.pone.0151497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Mazraani R., Timms P., Hill P.C., Suaalii-Sauni T., Niupulusu T., Temese S.V.A., Iosefa-Siitia L., Auvaa L., Tapelu S.A., Motu M.F., et al. Evaluation of a PGP3 ELISA for surveillance of the burden of Chlamydia infection in women from Australia and Samoa. Pathog. Dis. 2019;77 doi: 10.1093/femspd/ftz031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Anyalechi G.E., Hong J., Danavall D.C., Martin D.L., Gwyn S.E., Horner P.J., Raphael B.H., Kirkcaldy R.D., Kersh E.N., Bernstein K.T. High Plasmid Gene Protein 3 (Pgp3) Chlamydia trachomatis Seropositivity, Pelvic Inflammatory Disease, and Infertility Among Women, National Health and Nutrition Examination Survey, United States, 2013-2016. Clin. Infect. Dis. 2021;73:1507–1516. doi: 10.1093/cid/ciab506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.van Aar F., de Moraes M., Morré S.A., van Bergen J.E.A.M., van der Klis F.R.M., Land J.A., van der Sande M.A.B., van den Broek I.V.F. Chlamydia trachomatis IgG seroprevalence in the general population of the Netherlands in 1996 and in 2007: differential changes by gender and age. Sex. Transm. Infect. 2014;90:434–440. doi: 10.1136/sextrans-2013-051074. [DOI] [PubMed] [Google Scholar]
  • 220.Alexiou Z.W., van Aar F., Hoenderboom B.M., Morre S.A., Heijne J.C.M. Trends in Chlamydia trachomatis IgG seroprevalence in the general population of the Netherlands over 20 years. Sex. Transm. Infect. 2024;100:31–38. doi: 10.1136/sextrans-2023-055888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Poljak M., Cuschieri K., Alemany L., Vorsters A. Testing for Human Papillomaviruses in Urine, Blood, and Oral Specimens: an Update for the Laboratory. J. Clin. Microbiol. 2023;61:e0140322–e0141422. doi: 10.1128/jcm.01403-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Yuzen D., Arck P.C., Thiele K. Tissue-resident immunity in the female and male reproductive tract. Semin. Immunopathol. 2022;44:785–799. doi: 10.1007/s00281-022-00934-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Xiang W.J., Yu N.Y., Lei A.H., Li X.F., Tan S., Huang L.J., Zhou Z. Insights Into Host Cell Cytokines in Infection. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.639834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Murray S.M., McKay P.F. Chlamydia trachomatis: Cell biology, immunology and vaccination. Vaccine. 2021;39:2965–2975. doi: 10.1016/j.vaccine.2021.03.043. [DOI] [PubMed] [Google Scholar]
  • 225.Aiyar A., Quayle A.J., Buckner L.R., Sherchand S.P., Chang T.L., Zea A.H., Martin D.H., Belland R.J. Influence of the tryptophan-indole-IFNgamma axis on human genital Chlamydia trachomatis infection: role of vaginal co-infections. Front. Cell. Infect. Microbiol. 2014;4:72. doi: 10.3389/fcimb.2014.00072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.McClarty G., Caldwell H.D., Nelson D.E. Chlamydial interferon gamma immune evasion influences infection tropism. Curr. Opin. Microbiol. 2007;10:47–51. doi: 10.1016/j.mib.2006.12.003. [DOI] [PubMed] [Google Scholar]
  • 227.Lim J.M.E., Tan A.T., Le Bert N., Hang S.K., Low J.G.H., Bertoletti A. SARS-CoV-2 breakthrough infection in vaccinees induces virus-specific nasal-resident CD8+ and CD4+ T cells of broad specificity. J. Exp. Med. 2022;219 doi: 10.1084/jem.20220780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Kalsdorf B., Scriba T.J., Wood K., Day C.L., Dheda K., Dawson R., Hanekom W.A., Lange C., Wilkinson R.J. HIV-1 Infection Impairs the Bronchoalveolar T-Cell Response to Mycobacteria. Am. J. Respir. Crit. Care Med. 2009;180:1262–1270. doi: 10.1164/rccm.200907-1011OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Shacklett B.L., Yang O., Hausner M.A., Elliott J., Hultin L., Price C., Fuerst M., Matud J., Hultin P., Cox C., et al. Optimization of methods to assess human mucosal T-cell responses to HIV infection. J. Immunol. Methods. 2003;279:17–31. doi: 10.1016/S0022-1759(03)00255-2. [DOI] [PubMed] [Google Scholar]
  • 230.Bontems P., Robert F., Van Gossum A., Cadranel S., Mascart F. modulation of gastric and duodenal mucosal T cell cytokine secretions in children compared with adults. Helicobacter. 2003;8:216–226. doi: 10.1046/j.1523-5378.2003.00147.x. [DOI] [PubMed] [Google Scholar]
  • 231.Salvati V.M., Mazzarella G., Gianfrani C., Levings M.K., Stefanile R., De Giulio B., Iaquinto G., Giardullo N., Auricchio S., Roncarolo M.G., Troncone R. Recombinant human interleukin 10 suppresses gliadin dependent T cell activation in ex vivo cultured coeliac intestinal mucosa. Gut. 2005;54:46–53. doi: 10.1136/gut.2003.023150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Shacklett B.L., Cu-Uvin S., Beadle T.J., Pace C.A., Fast N.M., Donahue S.M., Caliendo A.M., Flanigan T.P., Carpenter C.C., Nixon D.F. Quantification of HIV-1-specific T-cell responses at the mucosal cervicovaginal surface. AIDS. 2000;14:1911–1915. doi: 10.1097/00002030-200009080-00005. [DOI] [PubMed] [Google Scholar]
  • 233.Lim J.M.E., Tan A.T., Bertoletti A. Protocol to detect antigen-specific nasal-resident T cells in humans. STAR Protoc. 2023;4 doi: 10.1016/j.xpro.2022.101995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Rha M.-S., Kim G., Lee S., Kim J., Jeong Y., Jung C.M., Noh H.E., Noh J.Y., Kim Y.M., Cho H.-J., et al. SARS-CoV-2 spike-specific nasal-resident CD49a+CD8+ memory T cells exert immediate effector functions with enhanced IFN-γ production. Nat. Commun. 2024;15:8355. doi: 10.1038/s41467-024-52689-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Vyas V., Sandhar B., Keane J.M., Wood E.G., Blythe H., Jones A., Shahaj E., Fanti S., Williams J., Metic N., et al. Tissue-resident memory T cells in epicardial adipose tissue comprise transcriptionally distinct subsets that are modulated in atrial fibrillation. Nat. Cardiovasc. Res. 2024;3:1067–1082. doi: 10.1038/s44161-024-00532-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Binayke A., Zaheer A., Vishwakarma S., Singh S., Sharma P., Chandwaskar R., Gosain M., Raghavan S., Murugesan D.R., Kshetrapal P., et al. A quest for universal anti-SARS-CoV-2 T cell assay: systematic review, meta-analysis, and experimental validation. NPJ Vaccines. 2024;9:3. doi: 10.1038/s41541-023-00794-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Tippalagama R., Chihab L.Y., Kearns K., Lewis S., Panda S., Willemsen L., Burel J.G., Lindestam Arlehamn C.S. Antigen-specificity measurements are the key to understanding T cell responses. Front. Immunol. 2023;14 doi: 10.3389/fimmu.2023.1127470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Bacher P., Scheffold A. Flow-Cytometric Analysis of Rare Antigen-Specific T Cells. Cytometry. A. 2013;83:692–701. doi: 10.1002/cyto.22317. [DOI] [PubMed] [Google Scholar]
  • 239.Helble J.D., Starnbach M.N. T cell responses to Chlamydia. Pathog. Dis. 2021;79 doi: 10.1093/femspd/ftab014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Jordan S.J., Bakshi R.K., Brown L.T., Chi X., Geisler W.M. Stimulated peripheral blood mononuclear cells from chlamydia-infected women release predominantly Th1-polarizing cytokines. Cytokine. 2019;113:458–461. doi: 10.1016/j.cyto.2018.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Ogendi B.M.O., Bakshi R.K., Gupta K., Kapil R., Brown L.T., Jordan S.J., Sabbaj S., Press C.G., Lee J.Y., Geisler W.M. T cell phenotypes in women with Chlamydia trachomatis infection and influence of treatment on phenotype distributions. Microbes Infect. 2018;20:176–184. doi: 10.1016/j.micinf.2017.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Min S.L., He P., Zhou Q.T., Chen H.L. The dual role of cytokine responses to infection in host pathogen crosstalk. Microb. Pathog. 2022;173 doi: 10.1016/j.micpath.2022.105812. [DOI] [PubMed] [Google Scholar]
  • 243.Loomis W.P., Starnbach M.N. Chlamydia trachomatis infection alters the development of memory CD8+ T cells. J. Immunol. 2006;177:4021–4027. doi: 10.4049/jimmunol.177.6.4021. [DOI] [PubMed] [Google Scholar]
  • 244.Fankhauser S.C., Starnbach M.N. PD-L1 Limits the Mucosal CD8 T Cell Response to. J. Immunol. 2014;192:1079–1090. doi: 10.4049/jimmunol.1301657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Fling S.P., Sutherland R.A., Steele L.N., Hess B., D'Orazio S.E., Maisonneuve J., Lampe M.F., Probst P., Starnbach M.N. CD8 T cells recognize an inclusion membrane-associated protein from the vacuolar pathogen. Proc. Natl. Acad. Sci. USA. 2001;98:1160–1165. doi: 10.1073/pnas.98.3.1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Starnbach M.N., Loomis W.P., Ovendale P., Regan D., Hess B., Alderson M.R., Fling S.P. An inclusion membrane protein from Chlamydia trachomatis enters the MHC class I pathway and stimulates a CD8+ T cell response. J. Immunol. 2003;171:4742–4749. doi: 10.4049/jimmunol.171.9.4742. [DOI] [PubMed] [Google Scholar]
  • 247.Russell A.N., Zheng X., O'Connell C.M., Wiesenfeld H.C., Hillier S.L., Taylor B.D., Picard M.D., Flechtner J.B., Zhong W., Frazer L.C., Darville T. Identification of Chlamydia trachomatis Antigens Recognized by T Cells From Highly Exposed Women Who Limit or Resist Genital Tract Infection. J. Infect. Dis. 2016;214:1884–1892. doi: 10.1093/infdis/jiw485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Poston T.B. Advances in vaccine development for Chlamydia trachomatis. Pathog. Dis. 2024;82 doi: 10.1093/femspd/ftae017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Cerda-Kipper A.S., Montiel B.E., Hosseini S. In: Encyclopedia of Analytical Science. Third Edition. Worsfold P., Poole C., Townshend A., Miró M., editors. Academic Press; 2019. Immunoassays | Radioimmunoassays and Enzyme-Linked Immunosorbent Assay; pp. 55–75. [DOI] [Google Scholar]
  • 250.Baud D., Regan L., Greub G. Comparison of five commercial serological tests for the detection of anti-Chlamydia trachomatis antibodies. Eur. J. Clin. Microbiol. Infect. Dis. 2010;29:669–675. doi: 10.1007/s10096-010-0912-4. [DOI] [PubMed] [Google Scholar]
  • 251.Wang X.S., Rockey D.D., Dolan B.P. Lipooligosaccharide Has Varied Direct and Indirect Roles in Evading both Innate and Adaptive Host Immune Responses. Infect. Immun. 2020;88 doi: 10.1128/IAI.00198-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.den Hartog J.E., Land J.A., Stassen F.R.M., Slobbe-van Drunen M.E.P., Kessels A.G.H., Bruggeman C.A. The role of chlamydia genus-specific and species-specific IgG antibody testing in predicting tubal disease in subfertile women. Hum. Reprod. 2004;19:1380–1384. doi: 10.1093/humrep/deh267. [DOI] [PubMed] [Google Scholar]
  • 253.Andersen, S.E., Bulman, L.M., Steiert, B., Faris, R., and Weber, M.M. Got mutants? How advances in chlamydial genetics have furthered the study of effector proteins. LID - 10.1093/femspd/ftaa078 [doi] LID - ftaa078. [DOI] [PMC free article] [PubMed]
  • 254.Cappello F., de Macario E.C., Di Felice V., Zummo G., Macario A.J.L. Infection and Anti-Hsp60 Immunity: The Two Sides of the Coin. PLoS Pathog. 2009;5 doi: 10.1371/journal.ppat.1000552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Jonsson S., Oda H., Lundin E., Olsson J., Idahl A. Chlamydial Heat Shock Protein 60 and Anti-Chlamydial Antibodies in Women with Epithelial Ovarian Tumors. Transl. Oncol. 2018;11:546–551. doi: 10.1016/j.tranon.2018.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Horner P.J., Anyalechi G.E., Geisler W.M. What Can Serology Tell Us About the Burden of Infertility in Women Caused by Chlamydia? J. Infect. Dis. 2021;224:S80–S85. doi: 10.1093/infdis/jiab047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Rahman K.S., Darville T., Wiesenfeld H.C., Hillier S.L., Kaltenboeck B. Mixed Chlamydia trachomatis Peptide Antigens Provide a Specific and Sensitive Single-Well Colorimetric Enzyme-Linked Immunosorbent Assay for Detection of Human Anti-C. trachomatis Antibodies. mSphere. 2018;3 doi: 10.1128/mSphere.00484-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Olsen A.W., Rosenkrands I., Jacobsen C.S., Cheeseman H.M., Kristiansen M.P., Dietrich J., Shattock R.J., Follmann F. Immune signature of Chlamydia vaccine CTH522/CAF(R)01 translates from mouse-to-human and induces durable protection in mice. Nat. Commun. 2024;15:1665. doi: 10.1038/s41467-024-45526-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Rosenkrands I., Olsen A.W., Knudsen S., Dehari N., Juel H.B., Cheeseman H.M., Andersen P., Shattock R.J., Follmann F. Human antibody signatures towards the major outer membrane protein after natural infection and vaccination. EBioMedicine. 2024;104 doi: 10.1016/j.ebiom.2024.105140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.van Ess, E.A.-O., Eck-Hauer, A., Land, J.A., Morré, S.A.-O., and Ouburg, S.A.-O. Combining individual Chlamydia trachomatis IgG antibodies MOMP, TARP, CPAF, OMP2, and HSP60 for Tubal Factor Infertility Prediction. [DOI] [PMC free article] [PubMed]
  • 261.Orgentec Diagnostika GmbH. Product catalogue. https://www.orgentec.com/articles/ORGENTEC_Catalogue_QM164166_2019_12_09.pdf.
  • 262.Yu H., Geisler W.M., Dai C., Gupta K., Cutter G., Brunham R.C. Antibody responses to Chlamydia trachomatis vaccine candidate antigens in Chlamydia-infected women and correlation with antibody-mediated phagocytosis of elementary bodies. Front. Cell. Infect. Microbiol. 2024;14 doi: 10.3389/fcimb.2024.1342621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Batteiger B.E., Xu F., Johnson R.E., Rekart M.L. Protective immunity to Chlamydia trachomatis genital infection: Evidence from human studies. J. Infect. Dis. 2010;201 Suppl 2:S178–S189. doi: 10.1086/652400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Darville T. Pelvic Inflammatory Disease Due to Neisseria gonorrhoeae and Chlamydia trachomatis: Immune Evasion Mechanisms and Pathogenic Disease Pathways. J. Infect. Dis. 2021;224:S39–S46. doi: 10.1093/infdis/jiab031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Naglak E.K., Morrison S.G., Morrison R.P. Neutrophils Are Central to Antibody-Mediated Protection against Genital Chlamydia. Infect. Immun. 2017;85 doi: 10.1128/IAI.00409-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Hybiske K., Paktinat S., Newman K., Patton D., Khosropour C., Roxby A.C., Mugo N.R., Oluoch L., Ngure K., Suchland R., et al. Antibodies from chlamydia-infected individuals facilitate phagocytosis via Fc receptors. Infect. Immun. 2024;92 doi: 10.1128/iai.00503-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Grasse M., Rosenkrands I., Olsen A., Follmann F., Dietrich J. A flow cytometry-based assay to determine the phagocytic activity of both clinical and nonclinical antibody samples against Chlamydia trachomatis. Cytometry. A. 2018;93:525–532. doi: 10.1002/cyto.a.23353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Yu H., Geisler W.M., Dai C., Gupta K., Cutter G., Brunham R.C. Neutralizing Antibody Responses to Chlamydia trachomatis in Women and Associations with Chlamydia Outcomes. J. Infect. Dis. 2025;231:e531–e535. doi: 10.1093/infdis/jiae519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Ziklo N., Huston W.M., Hocking J.S., Timms P. Chlamydia trachomatis Genital Tract Infections: When Host Immune Response and the Microbiome Collide. Trends Microbiol. 2016;24:750–765. doi: 10.1016/j.tim.2016.05.007. [DOI] [PubMed] [Google Scholar]
  • 270.Onywera H., Williamson A.L., Ponomarenko J., Meiring T.L. The Penile Microbiota in Uncircumcised and Circumcised Men: Relationships With HIV and Human Papillomavirus Infections and Cervicovaginal Microbiota. Front. Med. 2020;7:383. doi: 10.3389/fmed.2020.00383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Mofolorunsho K.C., Mabaso N.G., Nundlall N., Ojo A.O., Cason E.D., Abbai N.S. Comparison of the urinary microbiome in men who have sex with men with and without Chlamydia trachomatis infection. Eur. J. Clin. Microbiol. Infect. Dis. 2024;43:2159–2170. doi: 10.1007/s10096-024-04930-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Galiwango R.M., Okech B., Park D.E., Buchanan L., Shao Z., Bagaya B., Mpendo J., Joag V., Yegorov S., Nanvubya A., et al. Impact of antimicrobials on penile HIV susceptibility and immunology in uncircumcised men: A randomized phase 1/2 clinical trial. Cell Rep. Med. 2024;5 doi: 10.1016/j.xcrm.2024.101705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Ghasemian E., Harding-Esch E., Mabey D., Holland M.J. When Bacteria and Viruses Collide: A Tale of Chlamydia trachomatis and Sexually Transmitted Viruses. Viruses. 2023;15 doi: 10.3390/v15091954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Menon S., Timms P., Allan J.A., Alexander K., Rombauts L., Horner P., Keltz M., Hocking J., Huston W.M. Human and Pathogen Factors Associated with Chlamydia trachomatis-Related Infertility in Women. Clin. Microbiol. Rev. 2015;28:969–985. doi: 10.1128/CMR.00035-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Kinnunen A.H., Surcel H.M., Lehtinen M., Karhukorpi J., Tiitinen A., Halttunen M., Bloigu A., Morrison R.P., Karttunen R., Paavonen J. HLA DQ alleles and interleukin-10 polymorphism associated with Chlamydia trachomatis-related tubal factor infertility: a case-control study. Hum. Reprod. 2002;17:2073–2078. doi: 10.1093/humrep/17.8.2073. [DOI] [PubMed] [Google Scholar]
  • 276.Abdelsamed H., Peters J., Byrne G.I. Genetic variation in Chlamydia trachomatis and their hosts: impact on disease severity and tissue tropism. Future Microbiol. 2013;8:1129–1146. doi: 10.2217/fmb.13.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Murillo L.S., Land J.A., Pleijster J., Bruggeman C.A., Peña A.S., Morré S.A. Interleukin-1B (IL-1B) and interleukin-1 receptor antagonist (IL-1RN) gene polymorphisms are not associated with tubal pathology and Chlamydia trachomatis-related tubal factor subfertility. Hum. Reprod. 2003;18:2309–2314. doi: 10.1093/humrep/deg436. [DOI] [PubMed] [Google Scholar]
  • 278.Ohman H., Tiitinen A., Halttunen M., Birkelund S., Christiansen G., Koskela P., Lehtinen M., Paavonen J., Surcel H.M. IL-10 polymorphism and cell-mediated immune response to Chlamydia trachomatis. Genes Immun. 2006;7:243–249. doi: 10.1038/sj.gene.6364293. [DOI] [PubMed] [Google Scholar]
  • 279.Berry A., Hall J.V. The Complexity of Interactions Between Female Sex Hormones and Chlamydia trachomatis Infections. Curr. Clin. Microbiol. Rep. 2019;6:67–75. doi: 10.1007/s40588-019-00116-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Hickey D.K., Mulvey P., Bryan E.R., Trim L., Beagley K.W. Mucosal Vaccines: Innovation for Preventing Infectious Diseases. 2019. Regulation of mucosal immunity in the genital tract: Balancing reproduction and protective immunity; pp. 255–297. [DOI] [Google Scholar]
  • 281.Rey-Ladino J., Ross A.G.P., Cripps A.W. Immunity, immunopathology, and human vaccine development against sexually transmitted. Hum Vacc Immunother. 2014;10:2664–2673. doi: 10.4161/hv.29683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Stephens R.S. The cellular paradigm of chlamydial pathogenesis. Trends Microbiol. 2003;11:44–51. doi: 10.1016/S0966-842X(02)00011-2. [DOI] [PubMed] [Google Scholar]
  • 283.Horner P.J., Flanagan H., Horne A.W. Is There a Hidden Burden of Disease as a Result of Epigenetic Epithelial-to-Mesenchymal Transition Following Chlamydia trachomatis Genital Tract Infection? J. Infect. Dis. 2021;224:S128–S136. doi: 10.1093/infdis/jiab088. [DOI] [PubMed] [Google Scholar]
  • 284.Brunham R.C. Problems With Understanding Chlamydia trachomatis Immunology. J. Infect. Dis. 2022;225:2043–2049. doi: 10.1093/infdis/jiab610. [DOI] [PubMed] [Google Scholar]
  • 285.Ouzzani M., Hammady H., Fedorowicz Z., Elmagarmid A. Rayyan-a web and mobile app for systematic reviews. Syst. Rev. 2016;5:210. doi: 10.1186/s13643-016-0384-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Lefebvre C., Glanville J., Briscoe S., Featherstone R., Littlewood A., Metzendorf M.-I., Noel-Storr A., Paynter R., Rader T., Thomas J., Wieland L.S. In: Cochrane Handbook for Systematic Reviews of Interventions version 6.4. Higgins J.P.T., Chandler J., Cumpston M., Li T., Page M.J., Welch V.A., editors. 2024. Technical Supplement to Chapter 4: Searching for and selecting studies. [Google Scholar]
  • 287.Mierden, S.v., & Leenaars, C. (2023). Animal Search Filter. osf.io/q6uxs.
  • 288.University of Tasmania. Systematic Resviews for Health: 8. Search Limits. https://utas.libguides.com/SystematicReviews/SearchLimits.
  • 289.Page M.J., Moher D., Bossuyt P.M., Boutron I., Hoffmann T.C., Mulrow C.D., Shamseer L., Tetzlaff J.M., Akl E.A., Brennan S.E., et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ Br. Med. J. (Clin. Res. Ed.) 2021;372 doi: 10.1136/bmj.n160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Moola S, M.Z., Tufanaru C, Aromataris E, Sears K, Sfetcu R, Currie M, Qureshi R, Mattis P, Lisy K, Mu P-F. (2020). Chapter 7: Systematic reviews of etiology and risk. In JBI Manual for Evidence Synthesis, M.Z. Aromataris E, ed.
  • 291.McArthur A., Klugárová J., Yan H., Florescu S. Innovations in the systematic review of text and opinion. Int. J. Evid. Based. Healthc. 2015;13:188–195. doi: 10.1097/XEB.0000000000000060. [DOI] [PubMed] [Google Scholar]
  • 292.Munn Z, B.T., Moola S, Tufanaru C, Stern C, McArthur A, Stephenson M, Aromataris, and E. Methodological quality of case series studies. JBI Evidence Synthesis. doi: 10.11124/JBISRIR-D-19-00099. [DOI] [PubMed]
  • 293.Tufanaru C, M.Z., Aromataris E, Campbell J, Hopp L. (2020). Chapter 3: Systematic reviews of effectiveness. In JBI Manual for Evidence Synthesis. , M.Z. Aromataris E, ed.
  • 294.Wells G.A., Wells G., Shea B., Shea B., O'Connell D., Peterson J., Welch V., Losos M., Tugwell P., Ga S.W., et al. 2014. The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. [Google Scholar]

Associated Data

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

Supplementary Materials

Document S1. Figure S1 and Tables S1, S2, and S6
mmc1.pdf (489.4KB, pdf)
Table S3. Overview of studies on detection of antibodies in male urogenital secretions using ELISA-based assays
mmc2.xlsx (25.8KB, xlsx)
Table S4. Overview of studies on detection of antibodies in female urogenital secretions using ELISA-based assays
mmc3.xlsx (20.4KB, xlsx)
Table S5. Overview of studies on detection of antigen-specific adaptive cellular immune responses in urogenital secretions
mmc4.xlsx (20.3KB, xlsx)

Data Availability Statement

  • Data reported in this paper will be shared by the lead contact upon request.

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.


Articles from iScience are provided here courtesy of Elsevier

RESOURCES