Skip to main content
Microbial Cell logoLink to Microbial Cell
. 2016 Sep 5;3(9):390–403. doi: 10.15698/mic2016.09.525

Chlamydia trachomatis Genital Infections

Catherine M O’Connell 1,*, Morgan E Ferone 1
PMCID: PMC5354567  PMID: 28357377

Abstract

Etiology, transmission and protection: Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infection (STI) globally. However, C. trachomatis also causes trachoma in endemic areas, mostly Africa and the Middle East, and is a leading cause of preventable blindness worldwide. Epidemiology, incidence and prevalence: The World Health Organization estimates 131 million new cases of C. trachomatis genital infection occur annually. Globally, infection is most prevalent in young women and men (14-25 years), likely driven by asymptomatic infection, inadequate partner treatment and delayed development of protective immunity. Pathology/Symptomatology: C. trachomatis infects susceptible squamocolumnar or transitional epithelial cells, leading to cervicitis in women and urethritis in men. Symptoms are often mild or absent but ascending infection in some women may lead to Pelvic Inflammatory Disease (PID), resulting in reproductive sequelae such as ectopic pregnancy, infertility and chronic pelvic pain. Complications of infection in men include epididymitis and reactive arthritis. Molecular mechanisms of infection: Chlamydiae manipulate an array of host processes to support their obligate intracellular developmental cycle. This leads to activation of signaling pathways resulting in disproportionate influx of innate cells and the release of tissue damaging proteins and pro-inflammatory cytokines. Treatment and curability: Uncomplicated urogenital infection is treated with azithromycin (1 g, single dose) or doxycycline (100 mg twice daily x 7 days). However, antimicrobial treatment does not ameliorate established disease. Drug resistance is rare but treatment failures have been described. Development of an effective vaccine that protects against upper tract disease or that limits transmission remains an important goal.

Keywords: Chlamydia, urogenital, infection, epidemiology, reproductive morbidity, treatment

INTRODUCTION

Chlamydia trachomatis infections are the most commonly reported sexually transmitted bacterial infections in the US and globally. Ascending infection may result in infertility, ectopic pregnancy and chronic pelvic pain in some women. Despite widespread screening and treatment programs, the Chlamydia epidemic continues unabated with yearly increases in the number of reported cases. C. trachomatis is a gram-negative obligate intracellular pathogen with a unique developmental cycle that infects ocular, genital and respiratory tissues. Intriguingly, chlamydial serovars display specific tropisms for different mucosal sites but the molecular mechanisms controlling these processes are not fully understood. C. trachomatis can be classified into 15 serovars (genovars) 1 based on antigenic variation in the major outer membrane protein (MOMP) encoded by ompA 2. Serovars A-C are associated with trachoma, serovars D-K are most commonly with urogenital infection and serovars L1-L3 represent strains causing invasive lymphoma granuloma venereum (LGV). Although we have developed insights into how this bacterium infects and establishes a protected niche within epithelial cells using cell culture models, and have established animal models of in vivo infection, we lack information regarding the mechanisms that promote ascension and elicit damaging immunopathology in humans. Consequently, we are challenged to define chlamydial markers of virulence or biomarkers of host disease that could predict risk for severe reproductive sequelae and improve targeted screening and treatment. Chlamydial research is entering a period of rapid expansion with the advent of molecular epidemiology techniques, abundant genome sequences, and new approaches for effective genetic manipulation. With new tools to investigate the pathogenic mechanisms driving chlamydial disease in humans we hope to see accelerated progress towards an effective vaccine. In this review we provide an overview of current knowledge regarding epidemiology, disease outcomes and effective treatment of chlamydial genital tract infection. We also explore potential mechanisms facilitating C. trachomatis infection of genital mucosa identified via bioinformatics and other molecular approaches.

EPIDEMIOLOGY

C. trachomatis is the leading cause of bacterial sexually transmitted infection (STI) in the world. However, in endemic areas, mostly in Africa and the Middle East, C. trachomatis also causes trachoma, a leading cause of preventable blindness worldwide. The World Health Organization estimated a global prevalence of chlamydia at 4.2% (95% uncertainty interval: 3.7-4.7) among women aged 15-49 years for 2012 3. These figures correspond to an estimated 131 million new cases of chlamydia (100-166 million) 3. The majority of infections are observed within the Western Pacific Region and the Region of the Americas. Within the USA, 1,441,789 chlamydial infections were reported to CDC in 2014 4. Most infected men and women are either asymptomatic or minimally symptomatic and diagnosis occurs after screening or because a contact is symptomatic.

Rates of reported cases of chlamydia are highest among adolescents and young adults aged 15-24 years. In 2014, the rate among 15-19 year olds was 1,804.0 cases per 100,000 and the rate among 20-24 year olds was 2,484.6 cases per 100,000 4. Prevalence is relatively high when compared with other bacterial STIs because asymptomatically infected individuals may not seek treatment and repeat infection after single dose therapy is common. Infection is more frequently reported in young women rather than young men 4,5. Additional predictors of incident chlamydial infection in young women include single marital status, having a new sex partner or concurrent partnerships, smoking and associated signifiers of socioeconomic status, having gonorrhea or bacterial vaginosis, and presence of carcinogenic human papillomavirus 5,6,7,8,9.

GENITOURINARY TRACT INFECITON, DISEASE and REPRODUCTIVE SEQUELAE

Symptoms of genital C. trachomatis infection in women, when present, include changes in vaginal discharge, intermittent, intermenstrual and/or post-coital bleeding. C. trachomatis can also infect the urethra and some patients may present with symptoms of urinary tract infection (frequency and dysuria) 10. Mucopurulent endocervical discharge, easily induced endocervical bleeding, or edematous ectopy are clinical signs that may be observed upon exam 11. Untreated, infection may persist for up to 4 years 12 although spontaneous clearance of infection after diagnosis has been described 13, suggesting development of some degree of protective immunity.

Infection may ascend from the cervix, resulting in endometritis and salpingitis. Chlamydial PID can present as pelvic or lower abdominal pain with cervical motion tenderness or uterine or adnexal tenderness at exam 14 but even upper genital tract infection may be asymptomatic 15. In a high-risk population, 2% to 5% of untreated women developed PID within a ~2-week elapse between testing positive for C. trachomatis and returning for treatment 16,17. Repeated chlamydial infection has been associated with PID and other reproductive sequelae 16,18. A direct assessment of the risk for infertility after untreated C. trachomatis infection has not been performed but it has been determined that up to 18% of women may develop infertility after symptomatic PID of any cause 19.

C. trachomatis genital tract infection can also negatively impact pregnancy. Prior chlamydial infection is associated with elevated risk for ectopic pregnancy 20,21. C. trachomatis infection has been associated with spontaneous abortion, stillbirth and preterm delivery 22,23,24. C. trachomatis can also be transmitted to a neonate during delivery via contact with infected cervix tissue and secretions leading to infection of mucous membranes of the eye, oropharynx, urogenital tract, and rectum. Infection may be asymptomatic in these locations. C. trachomatis conjunctivitis that develops 5-12 days after birth is the most common presentation 25 but C. trachomatis also can cause a subacute, afebrile pneumonia with onset at ages 1-3 months 26. These outcomes are best avoided by screening and treatment prior to delivery.

In addition to serious reproductive consequences such as infertility, ectopic pregnancy and chronic pelvic pain C. trachomatis has also been proposed as a possible risk factor for cervical cancer. Human Papillomavirus (HPV) is a known cause of cervical cancer, but exposure to HPV does not necessarily result in the development of HPV-related cervical cancer. Persistent, high-risk HPV infections are more likely to progress to squamous cell carcinoma (SCC) or invasive cervical cancer (ICC) in the presence of identified cofactors including smoking, behavioral factors, age, genetic background and individual immune variation 27. Chronic cervical infection by C. trachomatis has been proposed as a cofactor based on detection of chlamydial DNA in HPV-associated lesions 28 and studies correlating the presence of anti-CT antibodies with risk for ICC or SCC 29. A recent meta analysis of 22 studies (19 retrospective, 3 prospective) determined that C. trachomatis was significantly linked to increased cervical cancer risk prospectively (OR = 2.21, 95% CI: 1.88-2.61, P < 0.001), and retrospectively (OR = 2.19, 95% CI: 1.74-2.74, P < 0.001) 30. The overlap in factors that contribute to HPV and chlamydial infection such as age 31 and number of sex partners 32 makes it challenging to determine if this association reflects concurrent infection or if chlamydial infection acts indirectly to facilitate HPV infection and/or promote HPV persistence. However, C. trachomatis infection was identified as an independent predictor of cervical cancer in 11 of these studies (OR = 1.76, 95% CI: 1.03-3.01, P = 0.04) with a multivariate logistic regression analysis adjusted for HPV and age 30.

Recent studies indicate that the origin of high-grade serous carcinomas is the fallopian tube 33. Precursor lesions that contain evidence of DNA damage and p53 mutations 34 have been detected in the fimbriated portion of the tubes of women with BRCA mutations 35. This association has prompted some gynecologic oncologists to advocate prophylactic bilateral salpingectomy for low risk women, coincident with benign gynaecologic surgery, e.g. hysterectomy or tubal ligation, as a primary preventive for ovarian cancer 36. Primary fallopian tube carcinomas have described in patients with chronic PID 37 and infertility is a known risk factor for epithelial ovarian cancer (reviewed in 38). A pilot study initially supported an association with anti-chlamydial antibody and ovarian cancer 39 but a subsequent study in a larger cohort could not confirm this finding 40. Anti-chlamydial IgG has been associated with type II ovarian cancer (P=0.002) in women with plasma samples obtained >1 year prior to diagnosis (n=7) 41. Positive 42 and negative 43 reports of the detection of chlamydial DNA in tumor tissue specimens have been published. Should future studies validate these still equivocal findings, women with a history of ascended chlamydial infection may be at increased risk for neoplasia.

C. trachomatis is the most common cause of nongonococcal urethritis in men. As in women, infections are often asymptomatic (40% to 96%) 44,45. The incubation period is variable but is typically 5 to 10 days after exposure. When men have symptoms, they may present with a mucoid or watery urethral discharge, and complain of dysuria. Their discharge may be scanty, clear or only observed after milking the urethra 46. Chlamydial infection may result in inflammation of the epididymides and testes. C. trachomatis is one of the most frequent pathogens in epididymitis among sexually active men <35 years of age. Symptoms of acute epididymitis include testicular pain and tenderness, hydrocele and epididymal swelling 47. Approximately 1% of men with nongonococcal urethritis develop reactive arthritis, and about one-third of these patients display the complete reactive arthritis triad previously termed Reiter syndrome (arthritis, uveitis, and urethritis) 48,49. Chlamydial nucleic acids have been detected in synovial tissues from patients with sexually transmitted reactive arthritis 50,51. The potential for C. trachomatis to cause chronic prostatitis and it’s potential to negatively impact male infertility is controversial (reviewed by 52).

Chlamydial proctitis, inflammation of the distal rectal mucosa, occurs primarily in men who have sex with men (MSM) who engage in receptive anal intercourse. In this group, infection is not uncommon and can be caused by D-K and L serovars. The presentation and severity of disease depends on the infecting chlamydial serovars. L1, L2 and L3 serovars of C. trachomatis cause LGV. In tropical and sub tropical regions, LGV infections are associated with urogenital ulceration and invasion of the lymphatic system in both men and women, which can result in bubo formation, fistulae, fibrosis and rectal stenosis. Outbreaks of anorectal disease caused by the L1-3 serovars have been reported amongst European and North American MSM, particularly those who are HIV-infected 53. In contrast to infection with serovars associated with genital tract infection, these infections are frequently symptomatic. Symptoms include anorectal pain, discharge, tenesmus, rectal bleeding and constipation, and are often accompanied by fever 54. Lack of treatment may result in strictures and severe scarring.

CHLAMYDIA TRACHOMATIS - GENITAL TRACT PATHOGEN

C. trachomatis is a strict intracellular pathogen with a unique biphasic lifecycle. Upon attachment, infectious elementary bodies (EB) stimulate uptake into epithelial cells where they differentiate into vegetative reticulate bodies (RB) to grow and divide within a membrane bound, host-derived parasitophorous vacuole called an inclusion. Within 8-12 divisions 55, differentiation to EB is initiated and the cycle is complete when the cell releases the contents of the inclusion to attach to adjacent cells and reinitiate the cycle 56. Chlamydial appropriation and exploitation of host cell machinery during invasion, inclusion formation and development is fundamental to replicative success. However, this "hijack" of cellular processes and intermediates triggers pro-inflammatory signaling pathways that drive innate cell influx with cytokine and chemokine release. For a subset of women with ascending infection the ultimate outcome is severe immunopathology and fibrosis leading to tubal occlusion (reviewed by Haftner 57). Similarly, processes that support chlamydial multiplication intracellularly can predispose the host cell towards transformation. Much of our current understanding of the roles that specific chlamydial effectors and their interactive host partners play in these processes is derived from cell systems 58,59,60,61 and animal models 62,63. Candidate receptors that promote chlamydial attachment to susceptible cells have been identified 64,65,66. Not unsurprisingly for a microorganism that interacts with its host cell across the plasma membrane at attachment and entry and with the inclusion membrane during the remainder of the developmental cycle, secretion systems represent a significant portion of the chlamydial genome. Type III secretion is important to effective cellular entry with a key role for the secreted effector TARP as well as potential accessory proteins (reviewed in 67). Furthermore, chlamydiae secrete a strikingly large number of proteins (Inc) to the inclusion membrane that play critical roles in membrane fusion, promote nutrient acquisition, avoidance of autophagy, engagement of innate signaling etc. 68. Global mapping of the Inc-Human interactome via affinity purification-mass spectroscopy (AP-MS) has identified associations or engagement of host proteins during all aspects of the chlamydial developmental cycle and revealed a previously unappreciated role for IncE in disruption of retromer trafficking via sequestration of SNX5/6 69. A recent review of the role(s) of these proteins and their cellular targets has been published which discusses these interactions in great detail, revealing the extent to which chlamydiae re-engineer the cell to support their multiplication 61.

Defining a spectrum of virulence for C. trachomatis strains in the context of genital tract infection has been challenging. Identifying clinical features that predict ascending infection and disease development in susceptible individuals is problematic because even PID may be asymptomatic 15. Disease outcome is also influenced by the genetic predisposition of an infected individual (~40%) 70. Genome studies and innovative efforts to better characterize infection kinetics and host response may prove useful, while genetic manipulation of strains will provide a direct route to examining the contribution of candidate virulence loci to infectivity, transmission, immunopathology or cancer.

The sequence of C. trachomatis D/UW-3/Cx was published in 1988 71 and since then many more strains have been sequenced (137 complete or partial genomes in Genbank, July 2016). Overall, C. trachomatis strains are strikingly similar with respect to size, GC content, similarity and synteny with near overlap between their core and pan genomes 72,73,74. This is preserved at the level of the resident plasmid with concordance of chromosome and plasmid phylogenies 75. Plasmid host range is highly restricted because shuttle vectors constructed from plasmids obtained from C. muridarum, C. trachomatis LGV or trachoma plasmids could not be stably transformed outside their lineage 76. The C. trachomatis phylogenetic tree parallels tissue-tropic groupings, LGV strains splitting first from urogenital and trachoma strains that subsequently diverged 75. Strains causing genital infections form two clades 72,73, one encompassing the most commonly isolated serovars E and F 77, with a second group comprised of serovars D, G-K. Within the clades, genetic exchange or recombination within the ompA locus has been detected where the major proportion of the genome remains consistent with the clade even when ompA type is discordant 72,78. Sequencing of trachoma strains endemic in Australian aboriginal communities determined that these isolates are more closely related to urogenital strains than to the classic trachoma lineage with the exception of their ompA and pmpEFGH loci 79 indicating that trachoma lineages have arisen from urogenital strains more than once.

Interestingly, these trachoma strains retain a functional TrpA 79. Previously, truncations of trpA were considered an important feature of ocular strains, contrasting with urogenital strains that express functional TrpA 80 and synthesize tryptophan if provided with indole 81. The potential that urogenital strains might be able to avoid IFN-γ -mediated chlamydial killing, which acts via IDO-induced tryptophan degradation 82, via cross-feeding from indole-producing commensals of the genital tract was thought to reflect niche expansion. The observation that trachoma strains may have arisen more than once, suggests that mutation of trpA could be pathoadaptive with respect to overall metabolism 83. More recently, the potential that TARP, a type III effector critically important in chlamydial entry and the highly polymorphic surface proteins (Pmps) play important roles in tissue tropism has been described 64,79,84. Cell culture-based studies of invasive, lypmphotropic LGV strains suggested that they were capable of surviving within macrophages 85,86, and were less susceptible to IFN-γ 87. However, L2 strains are no better at resisting perforin-2 mediated killing by activated human macrophages than the urogenital serovars B or D 88. Bioinformatic approaches have been employed to identify highly polymorphic loci (pmp, inc, TARP), recombination hotspots, and loci under positive or purifying selection with the goal of identifying individual genes that contribute to tissue tropism and virulence in LGV and urogenital strains 74,78,89,90,91. Although this approach is unbiased and the sequences analyzed represent the range of natural variation compatible with successful occupation of this ecologic niche, teasing out the individual contributions of candidate virulence factors still requires functional and mechanistic studies.

Another approach to investigate virulence differences between urogenital strains is to identify in vivo phenotypes predicting superior pathogenic potential. The conserved plasmid that is present in nearly all strains of C. trachomatis and in C. muridarum plays an important, highly pleiotropic role in virulence. Plasmid-deficient C. muridarum are attenuated in the murine model of genital tract infection because their ability to elicit damaging upper tract inflammation is reduced 92. Plasmid-deficient C. muridarum compete poorly with their plasmid-containing parent in vivo 93 and are less successful establishing oviduct infection 92,93. A plasmid-cured derivative of trachoma-causing C. trachomatis is attenuated in a non-human primate model of ocular infection 94. Cynomolgus macaques inoculated with strain A/2497P- displayed reduced inflammation and infection was cleared rapidly. In contrast, infection parameters did not differ significantly between C. trachomatis CTD153, a plasmid-cured derivative of the urogenital strain, D/UW-3/Cx and its parent when inoculated intravaginally in rhesus macaques 95. Similarly, accelerated clearance of infection by plasmid-deficient chlamydia is not observed in mice 92. It is possible that this phenotype reflects a plasmid-associated difference that contributes to tissue tropism. The chlamydial plasmid is also required for accumulation of glycogen within inclusions 96,97,98. Pgp4 is the plasmid-borne transcriptional regulator of the adjacent pgp3 99 and a conserved group of chromosomal loci, including glgA, which are differentially expressed in plasmid-deficient strains 98,100. C. muridarum pgp3 mutants are attenuated in vivo 101, indicating that this protein likely plays an important role in chlamydial virulence. The mechanism(s) by which Pgp3 contributes to chlamydial pathogenesis remains unclear, although roles in TLR2 activation 102 and immune avoidance via binding of the antimicrobial peptide LL-37 have been proposed 103. These studies demonstrate that chlamydial virulence is not intrinsically linked to fitness and that chlamydiae coordinate expression of genes or pathways important for pathogenesis. Conditions that stress chlamydiae 104,105,106 result in distinctive and profound transcriptional changes, superimposed on the complex transcriptional program that regulates the chlamydial developmental cycle 107. TLR2 activation and PRCL transcription by C. trachomatis is reduced in cell culture when glucose is limited 100, indicating that regulatory networks could potentially modulate virulence effector expression during infection in response to environmental stressors.

Association of signs, symptoms, and serovar with chlamydial load in diagnostic samples has been investigated as a way to assess infection severity (reviewed by 108). Increased cervical burden correlated with ascending infection using endometrial biopsies to monitor upper tract infection 109. However, a recent attempt to associate the presence or absence of the plasmid with reproductive morbidities in women presenting with gynecologic complications or subfertility was unsuccessful because the prevalence of plasmid-deficient strains in the study population was too low 110. The feasibility for extensive evaluation of cervical infection, coordinating immunofluorescent, ultrastructural genomic or flow cytometric analysis of infected cervical cells recovered via cytobrush, has been demonstrated 111,112. Transcriptional profiling using blood obtained from women with PID or asymptomatic cervical infection suggests that a blood borne inflammatory signature could enable the identification of specific biomarkers of damaging host responses 113,114. Correlation of infecting strain with engagement of such biomarkers may also be a way to identify virulent strains. However, the requirement for large numbers of infected patients combined with the expense associated with molecular and/or immunologic assays may render studies of sufficient statistical power cost prohibitive.

Determining the mechanisms by which chlamydial infection contributes to cellular transformation is key to understanding its potential role in reproductive cancers. HPV can induce genetic instability via dysregulation of centrosome duplication and p53 suppression 115. Multiple phenotypes related to genetic instability have been observed in chlamydial infection, such as supernumerary centrosomes, abnormal spindle poles, multinucleation, and chromosomal segregation defects 116,117,118. Dysregulation of host centrosome duplication during chlamydial infection occurs at procentriole formation, requires host kinases Cdk2 and Plk4 and progression through S-phase 117. Chlamydial disruption of this host pathway does not impact generation of infectious progeny 117. However, chlamydiae also usurp host microtubule networks as they establish their intracellular niche. Initial trafficking to the centrosome along microtubules involves the recruitment of Src kinases to the inclusion membrane, where their interaction with inclusion membrane (Inc) proteins facilitates access to the microtubule network at the centrosome 116,119,120. Chlamydia then orchestrate reorganization of the host microtubule network via Inc protein IPAM (inclusion protein acting on microtubules) and host-encoded CEP170 into a scaffold to support and maintain the inclusion within the cell 121, at the apparent cost of further centrosomal abnormality. There is no direct evidence to indicate that such abnormalities directly mediate tumor initiation but centrosomal abnormalities are observed in early, pre-cancerous lesions, hinting of a contribution to tumor progression 122. Regardless, cytokinesis failure and/or centrosome overduplication normally activates the tumor supressor p53 pathway 123.

Chlamydial infection also inhibits cellular DNA damage repair pathways directly, leading to heritable defects 124. Chlamydial infection triggers formation of reactive oxidative species, which promotes double stranded DNA breaks (DSBs). Downstream DNA repair responses and DSB relevant cell-cycle checkpoints are overridden 124 because intracellular chlamydiae activate a host pathway that culminates in proteasomal degradation of p53 125,126. These events grant chlamydiae access to vital energy intermediates because p53 down-regulates the pentose phosphate pathway within it damage surveillance program 126. Infected cells continue to proliferate despite the damage they sustain 124. Thus, in the context of acute infection, chlamydiae successfully meet their metabolic requirements and preserve their cellular niche. Current understanding of the developmental cycle suggests that the damaged cell will be destroyed rather than transformed after inclusion lysis and bacterial release. However, infected cells are able to divide and pass genetic defects onto daughter cells in culture 116,127 and in mice 127. Furthermore, 3T3 cells infected and cured of chlamydia exhibit anchorage-independent growth and increased rates of colony formation compared to mock-infected 3T3s 127, suggesting that mutagenized cells could escape infection and initiate neoplasm. Cervical dysplasia has been observed in both wild type and HPV transgenic mice infected with C. muridarum. Cervical dysplasia scored as CIN II was detected in both infected groups (WT, 3.3 ± 0.3; K14-HPV-E7, 3.5 ± 0.3) but cervical tissues from the respective uninfected control groups were normal (WT, 1.3 ± 0.3; K14-HPV-E7, 1.8 ± 0.5) 127. While similar studies cannot be undertaken in humans, it is possible that future studies using human-derived cervical 128,129 or fallopian 125,126 epithelial cell or organ models in conjunction with low passage clinical isolates of known virulence or mutagenic potential will provide future insights.

TREATMENT AND PREVENTION

Antibiotics effective against chlamydial infections cross host membranes and are active intracellularly. These target protein biosynthesis, primarily by interactions with the 50S or 30S ribosomal subunits. Antibiotics that target cell wall biosynthesis are also effective. The current recommendation of the CDC for treatment for uncomplicated genital infections in nonpregnant adolescents and adults is doxycycline for 7 days or azithromycin in a single dose 14. Azithromycin is the recommended first choice for treatment of pregnant women, with amoxicillin as alternative 14. Doxycycline and ofloxacin are contraindicated in pregnant women. Treatment for chlamydial infection in the context of PID is similar with the addition of a second-generation (cefoxitin) and all third-generation (ceftriaxone) cephalosporins for treatment of possible co-infection by other STI pathogens e.g. Neisseria gonorrheae 14. Treatment of LGV is more protracted (doxycycline 100 mg orally twice a day for 21 days) and may require aspiration/drainage to prevent ulcer formation. Sex partners should be evaluated, tested and treated. A test of cure is not recommended after completing treatment unless symptoms persist or if reinfection is suspected. However, testing sooner than 3-4 weeks post therapy completion may not be valid because of persisting, residual pathogen-derived nucleic acids 130,131. Treatment usually resolves infection but does not ameliorate preexisting inflammatory-mediated tissue damage.

Although in vivo development of homotypic drug resistance has never been documented for C. trachomatis, spontaneous drug resistant mutants have been selected in cell culture or after passage with sub-inhibitory concentrations of drug (reviewed by 132). However, their reduced ability to infect in cell culture or in animal models of infection suggests that these mutations are associated with such significant metabolic compromise that they will be lost from a population in the absence of selection 133,134. Nevertheless, treatment failure has been described for individuals who completed a course of therapy and who were reportedly not at risk for reinfection 7,135. Mechanisms that could contribute to these clinical observations include the potential for ongoing infection in a drug-protected reservoir that facilitates autoinoculation after therapy 136 and/or changes in the metabolic or physiologic state of chlamydiae that alter sensitivity to antimicrobial treatment. Recent studies have revealed that long lasting C. muridarum colonization of the murine gastrointestinal (GI) tract can be established with very low inocula administered orally 137. Intravenous inoculation with a bioluminescent derivative of C. muridarum also resulted in GI colonization 138, revealing a systemic route to this mucosal site. Treatment with doxycycline cleared GI infection but azithromycin treatment was ineffective 139. Intriguingly, a very recent study revealed that GI-colonized female mice failed to auto-inoculate their genital tract 140. However, the extent to which colonization elicited or modulated a protective adaptive response was not reported. It is possible that protracted colonization may have induced an adaptive response that protected their reproductive tracts from infection. Analogies with rectal infection/carriage of C. trachomatis in women abound and have been extensively reviewed by Borel and colleagues 141. Protective immunity in humans is slow to develop and thus, women may be more vulnerable to reinfection after transmission to a treated partner via unprotected rectal intercourse or via auto-inoculation.

C. trachomatis development and replication in vivo may be subject to stresses imposed by nutritional requirements 142, innate and adaptive immune responses 143, host physiology via hormones 144,145,146 and even competition with commensals or co-pathogens 147,148. Conditions that delay bacterial multiplication impair effectiveness of antibiotics in many microorganisms 149,150,151. Asymptomatic cervical infection lasting up to four years has been documented 12 but it is not known if infection could have been detected throughout or if infection waxed and waned entering periods of persistence or dormancy. Conditions that arrest chlamydiae mid-cycle or promote aberrant forms influence antimicrobial sensitivity in cell culture 152,153. Prospective observational studies in women are unethical and the establishment of animal models of persistent infection has been challenging. Nevertheless, azithromycin failure is more frequent in the murine model in the context of amoxicillin-induced persistence 154. Failure was more frequent when azithromycin was administered as a single dose rather than distributed over a period of days, suggesting that it might be prevented by improved absorption or extended exposure to the drug. A study performed with 85 patients (men and women) with uncomplicated dual infection with C. trachomatis and Mycoplasma genitalium receiving an extended treatment regimen achieved an eradication rate of 98.8% 155, suggesting that this approach may be sufficient to limit therapy failures. Practical aspects related to patient compliance with prolonged therapy must also be balanced with the impact on potential co-pathogens such as N. gonorrheae and M. genitalium. STI treatment guidelines now advise the use of single dose azithromycin in combination with ceftriaxone for treatment of uncomplicated N. gonorrheae infection in an effort to preserve this antibiotic in the face of increasing resistance 14,156,157. Resistance to tetracyclines is increasingly prevalent in this STI, limiting the usefulness of doxycycline in this context. M. genitalium, an etiology for nongonococcal urethritis in men 158, is associated with cervicitis and PID in women 159,160. Doxycycline is ineffective against this pathogen and homotypic resistance to azithromycin is well recognized 161,162,163. Thus, anti-chlamydial therapy in co-infected patients could potentially select or fix resistant M. genitalium strains within the population 164. There is an increasing need to consider the development of new regimens or novel antimicrobials to treat polymicrobial STI.

Women with any of the following risk factors should be tested routinely for Chlamydia: mucopurulent cervicitis, sexually active and <20 years of age, >1 sex partner during the last 3 months, or inconsistent use of barrier contraception while in a nonmonogamous relationship 14. Public health measures have encouraged screening, treatment and barrier contraception for more than 20 years. Although minimal rates of screening coverage have yet to be achieved in vulnerable populations, reductions in PID incidence have been observed 165,166. However, simply expanding screening risks becoming cost ineffective 167 and early treatment may blunt the development of protective immunity 168.

Evidence for natural immunity in humans includes decreased prevalence with increasing age 169 and decreased infection concordance with increased age of sexual partnerships 7,170. IFN-γ-producing Chlamydia-responsive CD4 T cells are key mediators of protection 171,172 in mice and the relative ability of several candidate vaccine preparations to protect murine oviducts from disease correlated directly with their induction of CD4 T cell IFN-γ 173,174,175. Chlamydial proteins that induce CD4 and CD8 T cell production of IFN-γ in humans have been identified 176,177. Longitudinal analysis of PBMC responses to cHSP60 and EB conducted in sex workers revealed IFN-γ responses to cHSP60, but not to EB, were associated with protection from incident infection 178. Anti-chlamydial antibody contributes to resistance to reinfection 179. These may act indirectly by promoting T-helper 1 activation and cellular effector responses 180 because epidemiological studies associate high antibody titers with infertility 181 and do not correlate with infection resolution or control of ascending infection 109.

Nevertheless, there is no evidence that natural immunity provides complete, long-term protection sufficient to prevent damaging immune pathology. Consequently, developing an effective vaccine is a highly desired, ambitious goal (reviewed by 182,183). Candidate vaccines against C. trachomatis have languished in preclinical testing but Phase I trials of chlamydial vaccine candidates are anticipated. Furthermore, advances in adjuvant development hold promise for additional candidates to enter clinical evaluation. MOMP is a highly abundant surface antigen that has long been considered a promising candidate. Novel formulations delivering this protein via cationic liposomes induced antibody, type-1 immunity and partial protection from infection in minipigs 184 and significant protection against upper tract disease in mice 185,186. Intranasal immunization using MOMP in combination with Nanostat, oil-in-water nanoemulsion in mice after challenge 187. A polyvalent vaccine comprised of MOMP with PMPs formulated with DDA/MPL adjuvants reduces chlamydial shedding when tested in a transcervical C. trachomatis mouse model 188. Route of delivery has proven particularly important. Uterine vaccination with inactivated C. trachomatis complexed with charge switching synthetic adjuvant particles (cSAPs) linked with a TLR7-agonist, resiquimod, induced superior chlamydial clearance when compared to intranasal or intramuscular delivery because it elicited resident memory T cells in murine genital mucosa 189. This study highlighted the importance of investigating immunologic responses specific to the genital tract to determine optimal strategies for developing vaccines that elicit broad, long lasting protection against urogenital infection.

Funding Statement

We would like to thank Taylor Poston for helpful discussions while we prepared this review. We would like to acknowledge the support of NIH/NIAID via U19 AI084024 and AI098660 (Darville PI) and the University of North Carolina at Chapel Hill via SOM/TraCS TTSA Phase I Award (Randell and Darville PIs).

References

  • 1.Wang SP, Grayston JT. Immunologic relationship between genital TRIC, lymphogranuloma venereum, and related organisms in a new microtiter indirect immunofluorescence test. Am J Ophthalmol. 1970;70(3):67–374. doi: 10.1016/0002-9394(70)90096-6. [DOI] [PubMed] [Google Scholar]
  • 2.Stephens RS, Sanchez-Pescador R, Wagar EA, Inouye C, Urdea MS. Diversity of Chlamydia trachomatis major outer membrane protein genes. J Bacteriol. 1987;169(9):3879–3885. doi: 10.1128/jb.169.9.3879-3885.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Newman L, Rowley J, Vander Hoorn S, Wijesooriya NS, Unemo M, Low N, Stevens G, Gottlieb S, Kiarie J, Temmerman M. Global estimates of the prevalence and incidence of four curable sexually transmitted infections in 2012 based on systematic review and global reporting. PLoS One. 2015;10(12):e0143304. doi: 10.1371/journal.pone.0143304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Centers for Disease Control and Prevention (2015) Sexually Transmitted Disease Surveillance 2014. Atlanta: U.S. Department of Health and Human Services. . Available at: http://www.cdc.gov/std/stats14/surv-2014-print.pdf. [Accessed: 16.07.2016] [Google Scholar]
  • 5.Crichton J, Hickman M, Campbell R, Batista-Ferrer H, Macleod J. Socioeconomic factors and other sources of variation in the prevalence of genital chlamydia infections: A systematic review and meta-analysis. BMC Public Health. 2015;15:729. doi: 10.1186/s12889-015-2069-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Aghaizu A, Reid F, Kerry S, Hay PE, Mallinson H, Jensen JS, Kerry S, Kerry S, Oakeshott P. Frequency and risk factors for incident and redetected Chlamydia trachomatis infection in sexually active, young, multi-ethnic women: a community based cohort study. Sex Transm Infect. 2014;90(7):524–528. doi: 10.1136/sextrans-2014-051607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Batteiger BE, Tu W, Ofner S, Van Der Pol B, Stothard DR, Orr DP, Katz BP, Fortenberry JD. Repeated Chlamydia trachomatis genital infections in adolescent women. J Infect Dis. 2010;201(1):42–51. doi: 10.1086/648734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hwang LY, Ma Y, Moscicki AB. Biological and behavioral risks for incident Chlamydia trachomatis infection in a prospective cohort. Obstet Gynecol. 2014;124(5):954–960. doi: 10.1097/AOG.0000000000000429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jorgensen MJ, Maindal HT, Larsen MB, Christensen KS, Olesen F, Andersen B. Chlamydia trachomatis infection in young adults - association with concurrent partnerships and short gap length between partners. Infect Dis. 2015;47(12):838–845. doi: 10.3109/23744235.2015.1071916. [DOI] [PubMed] [Google Scholar]
  • 10.Stamm WE, Wagner KF, Amsel R, Alexander ER, Turck M, Counts GW, Holmes KK. Causes of the acute urethral syndrome in women. N Eng J Med. 1980;303(8):409–415. doi: 10.1056/NEJM198008213030801. [DOI] [PubMed] [Google Scholar]
  • 11.Marrazzo JM, Handsfield HH, Whittington WL. Predicting chlamydial and gonococcal cervical infection: implications for management of cervicitis. Obstet Gynecol. 2002;100(3):579–584. doi: 10.1016/S0029-7844(02)02140-3. [DOI] [PubMed] [Google Scholar]
  • 12.Molano M, Meijer CJ, Weiderpass E, Arslan A, Posso H, Franceschi S, Ronderos M, Munoz N, van den Brule AJ. The natural course of Chlamydia trachomatis infection in asymptomatic Colombian women: a 5-year follow-up study. J Infect Dis. 2005;191(6):907–916. doi: 10.1086/428287. [DOI] [PubMed] [Google Scholar]
  • 13.Geisler WM, Lensing SY, Press CG, Hook 3rd EW. Spontaneous resolution of genital Chlamydia trachomatis infection in women and protection from reinfection. J Infect Dis. 2013;207(12):1850–1856. doi: 10.1093/infdis/jit094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Workowski KA, Bolan GA, Centers for Disease Control and Prevention Sexually transmitted diseases treatment guidelines, 2015. MMWR Recomm Rep. 2015;64(RR-03):1–137. Available at: https://www.cdc.gov/std/tg2015/tg-2015-print.pdf. [Accessed: 16.07.2016] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wiesenfeld HC, Hillier SL, Meyn LA, Amortegui AJ, Sweet RL. Subclinical pelvic inflammatory disease and infertility. Obstet Gynecol. 2012;120(1):37–43. doi: 10.1097/AOG.0b013e31825a6bc9. [DOI] [PubMed] [Google Scholar]
  • 16.Bachmann LH, Richey CM, Waites K, Schwebke JR, Hook 3rd EW. Patterns of Chlamydia trachomatis testing and follow-up at a university hospital medical center. Sex Transm Dis. 1999;26(9):496–499. doi: 10.1097/00007435-199910000-00002. [DOI] [PubMed] [Google Scholar]
  • 17.Geisler WM, Wang C, Morrison SG, Black CM, Bandea CI, Hook 3rd EW. The natural history of untreated Chlamydia trachomatis infection in the interval between screening and returning for treatment. Sex Transm Dis. 2008;35(2):119–123. doi: 10.1097/OLQ.0b013e318151497d. [DOI] [PubMed] [Google Scholar]
  • 18.Kimani J, Maclean IW, Bwayo JJ, MacDonald K, Oyugi J, Maitha GM, Peeling RW, Cheang M, Nagelkerke NJ, Plummer FA, Brunham RC. Risk factors for Chlamydia trachomatis pelvic inflammatory disease among sex workers in Nairobi, Kenya. J Infect Dis. 1996;173(6):1437–1444. doi: 10.1093/infdis/173.6.1437. [DOI] [PubMed] [Google Scholar]
  • 19.Ness RB, Smith KJ, Chang CC, Schisterman EF, Bass DC. Prediction of pelvic inflammatory disease among young, single, sexually active women. Sex Transm Dis. 2006;33(3):137–142. doi: 10.1097/01.olq.0000187205.67390.d1. [DOI] [PubMed] [Google Scholar]
  • 20.Bakken IJ, Skjeldestad FE, Nordbo SA. Chlamydia trachomatis infections increase the risk for ectopic pregnancy: a population-based, nested case-control study. Sex Transm Dis. 2007;34(3):166–169. doi: 10.1097/01.olq.0000230428.06837.f7. [DOI] [PubMed] [Google Scholar]
  • 21.Egger M, Low N, Smith GD, Lindblom B, Herrmann B. Screening for chlamydial infections and the risk of ectopic pregnancy in a county in Sweden: ecological analysis. BMJ. 1998;316(7147):1776–1780. doi: 10.1136/bmj.316.7147.1776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Liu B, Roberts CL, Clarke M, Jorm L, Hunt J, Ward J. Chlamydia and gonorrhoea infections and the risk of adverse obstetric outcomes: a retrospective cohort study. Sex Transm Infect. 2013;89(8):672–678. doi: 10.1136/sextrans-2013-051118. [DOI] [PubMed] [Google Scholar]
  • 23.Hollegaard S, Vogel I, Thorsen P, Jensen IP, Mordhorst CH, Jeune B. Chlamydia trachomatis C-complex serovars are a risk factor for preterm birth. In Vivo. 2007;21(1):107–112. [PubMed] [Google Scholar]
  • 24.Andrews WW, Goldenberg RL, Mercer B, Iams J, Meis P, Moawad A, Das A, Vandorsten JP, Caritis SN, Thurnau G, Miodovnik M, Roberts J, McNellis D. The Preterm Prediction Study: association of second-trimester genitourinary chlamydia infection with subsequent spontaneous preterm birth. Am J Obstet Gynecol. 2000;183(3):662–668. doi: 10.1067/mob.2000.106556. [DOI] [PubMed] [Google Scholar]
  • 25.Rours IG, Hammerschlag MR, Ott A, De Faber TJ, Verbrugh HA, de Groot R, Verkooyen RP. Chlamydia trachomatis as a cause of neonatal conjunctivitis in Dutch infants. Pediatrics. 2008;121(2):e321–e326. doi: 10.1542/peds.2007-0153. [DOI] [PubMed] [Google Scholar]
  • 26.Rours GI, Hammerschlag MR, Van Doornum GJ, Hop WC, de Groot R, Willemse HF, Verbrugh HA, Verkooyen RP. Chlamydia trachomatis respiratory infection in Dutch infants. Arch Dis Child. 2009;94(9):705–707. doi: 10.1136/adc.2008.152066. [DOI] [PubMed] [Google Scholar]
  • 27.Silva J, Cerqueira F, Medeiros R. Chlamydia trachomatis infection: implications for HPV status and cervical cancer. Arch Gynecol Obstet. 2014;289(4):715–723. doi: 10.1007/s00404-013-3122-3. [DOI] [PubMed] [Google Scholar]
  • 28.Paba P, Bonifacio D, Di Bonito L, Ombres D, Favalli C, Syrjanen K, Ciotti M. Co-expression of HSV2 and Chlamydia trachomatis in HPV-positive cervical cancer and cervical intraepithelial neoplasia lesions is associated with aberrations in key intracellular pathways. Intervirology. 2008;51(4):230–234. doi: 10.1159/000156481. [DOI] [PubMed] [Google Scholar]
  • 29.Paavonen J, Karunakaran KP, Noguchi Y, Anttila T, Bloigu A, Dillner J, Hallmans G, Hakulinen T, Jellum E, Koskela P, Lehtinen M, Thoresen S, Lam H, Shen C, Brunham RC. Serum antibody response to the heat shock protein 60 of Chlamydia trachomatis in women with developing cervical cancer. Am J Obstet Gynecol. 2003;189(5):1287–1292. doi: 10.1067/s0002-9378(03)00755-5. [DOI] [PubMed] [Google Scholar]
  • 30.Zhu H, Shen Z, Luo H, Zhang W, Zhu X. Chlamydia Trachomatis Infection-Associated Risk of Cervical Cancer: A Meta-Analysis. Medicine (Baltimore) 2016;95(13):e3077. doi: 10.1097/MD.0000000000003077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Nonato DR, Alves RR, Ribeiro AA, Saddi VA, Segati KD, Almeida KP, de Lima YA, D'Alessandro WB, Rabelo-Santos SH. Prevalence and factors associated with co-infection of human papillomavirus and Chlamydia trachomatis in adolescents and young women. Am J Obstet Gynecol. 2016;S0002-9378 (16):30436–7. doi: 10.1016/j.ajog.2016.07.003. [DOI] [PubMed] [Google Scholar]
  • 32.Quinonez-Calvache EM, Rios-Chaparro DI, Ramirez JD, Soto-De Leon SC, Camargo M, Del Rio-Ospina L, Sanchez R, Patarroyo ME, Patarroyo MA. Chlamydia trachomatis frequency in a cohort of HPV-Infected Colombian women. PLoS One. 2016;11(1):e0147504. doi: 10.1371/journal.pone.0147504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Crum CP, Drapkin R, Miron A, Ince TA, Muto M, Kindelberger DW, Lee Y. The distal fallopian tube: a new model for pelvic serous carcinogenesis. Curr Opin Obstet Gynecol. 2007;19(1):3–9. doi: 10.1097/GCO.0b013e328011a21f. [DOI] [PubMed] [Google Scholar]
  • 34.Lee Y, Miron A, Drapkin R, Nucci MR, Medeiros F, Saleemuddin A, Garber J, Birch C, Mou H, Gordon RW, Cramer DW, McKeon FD, Crum CP. A candidate precursor to serous carcinoma that originates in the distal fallopian tube. J Pathol. 2007;211(1):26–35. doi: 10.1002/path.2091. [DOI] [PubMed] [Google Scholar]
  • 35.Medeiros F, Muto MG, Lee Y, Elvin JA, Callahan MJ, Feltmate C, Garber JE, Cramer DW, Crum CP. The tubal fimbria is a preferred site for early adenocarcinoma in women with familial ovarian cancer syndrome. Am J Surg Pathol. 2006;30(2):230–236. doi: 10.1097/01.pas.0000180854.28831.77. [DOI] [PubMed] [Google Scholar]
  • 36.Oliver Perez MR, Magrina J, Garcia AT, Jimenez Lopez JS. Prophylactic salpingectomy and prophylactic salpingoophorectomy for adnexal high-grade serous epithelial carcinoma: A reappraisal. Surg Oncol. 2015;24(4):335–344. doi: 10.1016/j.suronc.2015.09.008. [DOI] [PubMed] [Google Scholar]
  • 37.Zardawi IM. Primary fallopian tube carcinoma arising in the setting of chronic pelvic inflammatory disease. Case Rep Med. 2014;2014:645045. doi: 10.1155/2014/645045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Salvador S, Gilks B, Kobel M, Huntsman D, Rosen B, Miller D. The fallopian tube: primary site of most pelvic high-grade serous carcinomas. Int J Gynecol Cancer. 2009;19(1):58–64. doi: 10.1111/IGC.0b013e318199009c. [DOI] [PubMed] [Google Scholar]
  • 39.Ness RB, Goodman MT, Shen C, Brunham RC. Serologic evidence of past infection with Chlamydia trachomatis, in relation to ovarian cancer. J Infect Dis. 2003;187(7):1147–1152. doi: 10.1086/368380. [DOI] [PubMed] [Google Scholar]
  • 40.Ness RB, Soper DE, Richter HE, Randall H, Peipert JF, Nelson DB, Schubeck D, McNeeley SG, Trout W, Bass DC, Hutchison K, Kip K, Brunham RC. Chlamydia Antibodies, Chlamydia Heat Shock Protein, and Adverse Sequelae After Pelvic Inflammatory Disease: The PID Evaluation and Clinical Health (PEACH) Study. Sex Transm Dis. 2008;35(2):129–135. doi: 10.1097/OLQ.0b013e3181557c25. [DOI] [PubMed] [Google Scholar]
  • 41.Idahl A, Lundin E, Jurstrand M, Kumlin U, Elgh F, Ohlson N, Ottander U. Chlamydia trachomatis and Mycoplasma genitalium plasma antibodies in relation to epithelial ovarian tumors. Infect Dis Obstet Gynecol. 2011;2011(824627) doi: 10.1155/2011/824627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Shanmughapriya S, Senthilkumar G, Vinodhini K, Das BC, Vasanthi N, Natarajaseenivasan K. Viral and bacterial aetiologies of epithelial ovarian cancer. Eur J Clin Microbiol Infect Dis. 2012;31(9):2311–2317. doi: 10.1007/s10096-012-1570-5. [DOI] [PubMed] [Google Scholar]
  • 43.Idahl A, Lundin E, Elgh F, Jurstrand M, Moller JK, Marklund I, Lindgren P, Ottander U. Chlamydia trachomatis, Mycoplasma genitalium, Neisseria gonorrhoeae, Human PapillomaVirus, and Polyomavirus are not detectable in human tissue with epithelial ovarian cancer, borderline tumor, or benign conditions. Am J Obstet Gynecol. 2010;202(1):e71–e76. doi: 10.1016/j.ajog.2009.07.042. [DOI] [PubMed] [Google Scholar]
  • 44.Stamm WE. Chlamydia trachomatis infections: progress and problems. J Infect Dis . 1999;179(Suppl 2):S380–S383. doi: 10.1086/513844. [DOI] [PubMed] [Google Scholar]
  • 45.Gonzales GF, Munoz G, Sanchez R, Henkel R, Gallegos-Avila G, Diaz-Gutierrez O, Vigil P, Vasquez F, Kortebani G, Mazzolli A, Bustos-Obregon E. Update on the impact of Chlamydia trachomatis infection on male fertility. Andrologia. 2004;36(1):1–23. doi: 10.1046/j.0303-4569.2003.00594.x. [DOI] [PubMed] [Google Scholar]
  • 46.Stamm WE, Koutsky LA, Benedetti JK, Jourden JL, Brunham RC, Holmes KK. Chlamydia trachomatis urethral infections in men. Prevalence, risk factors, and clinical manifestations. Ann Intern Med. 1984;100(1):47–51. doi: 10.7326/0003-4819-100-1-47. [DOI] [PubMed] [Google Scholar]
  • 47.Hedger MP. Immunophysiology and pathology of inflammation in the testis and epididymis. J Androl. 2011;32(6):625–640. doi: 10.2164/jandrol.111.012989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Keat A, Thomas BJ, Taylor Robinson D. Chlamydial infection in the aetiology of arthritis. Br Med Bull. 1983;39(2):168–174. doi: 10.1093/oxfordjournals.bmb.a071811. [DOI] [PubMed] [Google Scholar]
  • 49.Hannu T. Reactive arthritis. Best Pract Res Clin Rheumatol. 2011;25(3):347–357. doi: 10.1016/j.berh.2011.01.018. [DOI] [PubMed] [Google Scholar]
  • 50.Rahman MU, Cheema MA, Schumacher HR, Hudson AP. Molecular evidence for the presence of chlamydia in the synovium of patients with Reiter's syndrome. Arthritis Rheum. 1992;35(5):521–529. doi: 10.1002/art.1780350506. [DOI] [PubMed] [Google Scholar]
  • 51.Taylor-Robinson D, Gilroy CB, Thomas BJ, Keat ACS. Detection of Chlamydia trachomatis DNA in joints of reactive arthritis patients by polymerase chain reaction. Lancet. 1992;340(8811):81–82. doi: 10.1016/0140-6736(92)90399-N. [DOI] [PubMed] [Google Scholar]
  • 52.Redgrove KA, McLaughlin EA. The role of the immune response in Chlamydia trachomatis infection of the male genital tract: A double-edged sword. Front Immunol. 2014;5:534. doi: 10.3389/fimmu.2014.00534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.de Vrieze NH, de Vries HJ. Lymphogranuloma venereum among men who have sex with men. An epidemiological and clinical review. Expert Rev Anti Infect Ther. 2014;12(6):697–704. doi: 10.1586/14787210.2014.901169. [DOI] [PubMed] [Google Scholar]
  • 54.de Vries HJ, Zingoni A, White JA, Ross JD, Kreuter A. 2013 European guideline on the management of proctitis, proctocolitis and enteritis caused by sexually transmissible pathogens. Int J STD AIDS. 2014;25(7):465–474. doi: 10.1177/0956462413516100. [DOI] [PubMed] [Google Scholar]
  • 55.Lambden PR, Pickett MA, Clarke IN. The effect of penicillin on Chlamydia trachomatis DNA replication. Microbiol. 2006;152(Pt 9):2573–2578. doi: 10.1099/mic.0.29032-0. [DOI] [PubMed] [Google Scholar]
  • 56.Moulder JW. Interaction of chlamydiae and host cells in vitro. Microbiol Rev. 1991;55(1):143–190. doi: 10.1128/mr.55.1.143-190.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hafner LM. Pathogenesis of fallopian tube damage caused by Chlamydia trachomatis infections. Contraception. 2015;92(2):108–115. doi: 10.1016/j.contraception.2015.01.004. [DOI] [PubMed] [Google Scholar]
  • 58.Mueller KE, Plano GV, Fields KA. New frontiers in type III secretion biology: the Chlamydia perspective. Infect Immun. 2014;82(1):2–9. doi: 10.1128/IAI.00917-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Omsland A, Sixt BS, Horn M, Hackstadt T. Chlamydial metabolism revisited: interspecies metabolic variability and developmental stage-specific physiologic activities. FEMS Microbiol Rev. 2014;38(4):779–801. doi: 10.1111/1574-6976.12059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bastidas RJ, Elwell CA, Engel JN, Valdivia RH. Chlamydial intracellular survival strategies. Cold Spring Harb Perspect Med. 2013;3(5):a010256. doi: 10.1101/cshperspect.a010256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Elwell C, Mirrashidi K, Engel J. Chlamydia cell biology and pathogenesis. Nat Rev Microbiol. 2016;14(6):385–400. doi: 10.1038/nrmicro.2016.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Rank RG, Whittum-Hudson JA. Protective immunity to chlamydial genital infection: evidence from animal studies. J Infect Dis. 2010;201(Suppl 2):S168–177. doi: 10.1086/652399. [DOI] [PubMed] [Google Scholar]
  • 63.Rank RG. Animal models for urogenital infections. Method Enzymol. 1994;235(83-93) doi: 10.1016/0076-6879(94)35133-3. [DOI] [PubMed] [Google Scholar]
  • 64.Kari L, Southern TR, Downey CJ, Watkins HS, Randall LB, Taylor LD, Sturdevant GL, Whitmire WM, Caldwell HD. Chlamydia trachomatis polymorphic membrane protein D is a virulence factor involved in early host cell interactions. Infect. 2014;Immun. doi: 10.1128/IAI.01686-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Moelleken K, Hegemann JH. The Chlamydia outer membrane protein OmcB is required for adhesion and exhibits biovar-specific differences in glycosaminoglycan binding. Mol Microbiol. 2008;67(2):403–419. doi: 10.1111/j.1365-2958.2007.06050.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Stallmann S, Hegemann JH. The Chlamydia trachomatis Ctad1 invasin exploits the human integrin beta1 receptor for host cell entry. Cell Microbiol. 2016;18(5):761–775. doi: 10.1111/cmi.12549. [DOI] [PubMed] [Google Scholar]
  • 67.Ferrell JC, Fields KA. A working model for the type III secretion mechanism in Chlamydia. Microbes Infect. 2016;18(2):84–92. doi: 10.1016/j.micinf.2015.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Moore ER, Ouellette SP. Reconceptualizing the chlamydial inclusion as a pathogen-specified parasitic organelle: an expanded role for Inc proteins. Front Cell Infect Microbiol. 2014;4(157) doi: 10.3389/fcimb.2014.00157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Mirrashidi KM, Elwell CA, Verschueren E, Johnson JR, Frando A, Von Dollen J, Rosenberg O, Gulbahce N, Jang G, Johnson T, Jager S, Gopalakrishnan AM, Sherry J, Dunn JD, Olive A, Penn B, Shales M, Cox JS, Starnbach MN, Derre I, Valdivia R, Krogan NJ, Engel J. Global mapping of the Inc-human interactome reveals that retromer restricts chlamydia infection. Cell Host Microbe. 2015;18(1):109–121. doi: 10.1016/j.chom.2015.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bailey RL, Natividad-Sancho A, Fowler A, Peeling RW, Mabey DC, Whittle HC, Jepson AP. ). Host genetic contribution to the cellular immune response to Chlamydia trachomatis: Heritability estimate from a Gambian twin study. Front Cell Infect Microbiol. 2009;45(Suppl B):45–50. [PubMed] [Google Scholar]
  • 71.Stephens RS, Kalman S, Lammel C, Fan J, Marathe R, Aravind L, Mitchell W, Olinger L, Tatusov RL, Zhao Q, Koonin EV, Davis RW. Genome sequence of an obligate intracellular pathogen of humans: Chlamydia trachomatis. Science. 1998;282(5389):754–759. doi: 10.1126/science.282.5389.754. [DOI] [PubMed] [Google Scholar]
  • 72.Harris SR, Clarke IN, Seth-Smith HM, Solomon AW, Cutcliffe LT, Marsh P, Skilton RJ, Holland MJ, Mabey D, Peeling RW, Lewis DA, Spratt BG, Unemo M, Persson K, Bjartling C, Brunham R, de Vries HJ, Morre SA, Speksnijder A, Bebear CM, Clerc M, de Barbeyrac B, Parkhill J, Thomson NR. Whole-genome analysis of diverse Chlamydia trachomatis strains identifies phylogenetic relationships masked by current clinical typing. Nat Genet. 2012;44(4):413–419. doi: 10.1038/ng.2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Joseph SJ, Didelot X, Rothschild J, de Vries HJ, Morre SA, Read TD, Dean D. Population genomics of Chlamydia trachomatis: insights on drift, selection, recombination, and population structure. Mol Biol Evol. 2012;29(12):3933–3946. doi: 10.1093/molbev/mss198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Joseph SJ, Didelot X, Gandhi K, Dean D, Read TD. Interplay of recombination and selection in the genomes of Chlamydia trachomatis. Biol Direct. 2011;6(1):28. doi: 10.1186/1745-6150-6-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Seth-Smith HMB, Harris SR, Persson K, Marsh P, Barron A, Bignell A, Bjartling C, Clark L, Cutcliffe LT, Lambden PR, Lennard N, Lockey SJ, Quail MA, Salim O, Skilton RJ, Wang YB, Holland MJ, Parkhill J, Thomson NR, Clarke IN. Co-evolution of genomes and plasmids within Chlamydia trachomatis and the emergence in Sweden of a new variant strain. Bmc Genomics. 2009;10(1):239. doi: 10.1186/1471-2164-10-239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Song L, Carlson JH, Zhou B, Virtaneva K, Whitmire WM, Sturdevant GL, Porcella SF, McClarty G, Caldwell HD. Plasmid-mediated transformation tropism of chlamydial biovars. Pathog Dis. 2014;70(2):189–193. doi: 10.1111/2049-632X.12104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Nunes A, Nogueira PJ, Borrego MJ, Gomes JP. Adaptive evolution of the Chlamydia trachomatis dominant antigen reveals distinct evolutionary scenarios for B- and T-cell epitopes: worldwide survey. PLoS One. 2010;5(10) doi: 10.1371/journal.pone.0013171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ferreira R, Antelo M, Nunes A, Borges V, Damiao V, Borrego MJ, Gomes JP. In silico scrutiny of genes revealing phylogenetic congruence with clinical prevalence or tropism properties of Chlamydia trachomatis strains. G3 (Bethesda) 2014;5(1):9–19. doi: 10.1534/g3.114.015354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Andersson P, Harris SR, Seth Smith HM, Hadfield J, O'Neill C, Cutcliffe LT, Douglas FP, Asche LV, Mathews JD, Hutton SI, Sarovich DS, Tong SY, Clarke IN, Thomson NR, Giffard PM. Chlamydia trachomatis from Australian Aboriginal people with trachoma are polyphyletic composed of multiple distinctive lineages. 2016;(7):10688. doi: 10.1038/ncomms10688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Caldwell HD, Wood H, Crane D, Bailey R, Jones RB, Mabey D, Maclean I, Mohammed Z, Peeling R, Roshick C, Schachter J, Solomon AW, Stamm WE, Suchland RJ, Taylor L, West SK, Quinn TC, Belland RJ, McClarty G. Polymorphisms in Chlamydia trachomatis tryptophan synthase genes differentiate between genital and ocular isolates. J Clin Invest. 2003;111(11):1757–1769. doi: 10.1172/JC17993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Fehlner-Gardiner C, Roshick C, Carlson JH, Hughes S, Belland RJ, Caldwell HD, McClarty G. Molecular basis defining human Chlamydia trachomatis tissue tropism. A possible role for tryptophan synthase. . J Biol Chem. 2002;277(30):26893–26903. doi: 10.1074/jbc.M203937200. [DOI] [PubMed] [Google Scholar]
  • 82.Beatty WL, Belanger TA, Desai AA, Morrison RP, Byrne GI. Tryptophan depletion as a mechanism of gamma interferon-mediated chlamydial persistence. Infect Immun. 1994;62(9):3705–3711. doi: 10.1128/iai.62.9.3705-3711.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sokurenko EV, Hasty DL, Dykhuizen DE. Pathoadaptive mutations: gene loss and variation in bacterial pathogens. Trends Microbiol. 1999;7(5):191–195. doi: 10.1016/s0966-842x(99)01493-6. [DOI] [PubMed] [Google Scholar]
  • 84.Carlson J, Porcella S, McClarty G, Caldwell H. Comparative genomic analysis of Chlamydia trachomatis oculotropic and genitotropic strains. Infect Immun. 2005;73:6407–6418. doi: 10.1128/IAI.73.10.6407-6418.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sarov I, Geron E, Shemer-Avni Y, Manor E, Zvillich M, Wallach D, Schmitz E, Holtman H. Implications for persistent chlamydial infections of phagocyte- microorganism interplay. Eur J Clin Microbiol Infect Dis. 1991;10:119–123. doi: 10.1007/bf01964423. [DOI] [PubMed] [Google Scholar]
  • 86.Manor E, Sarov I. Fate of Chlamydia trachomatis in human monocytes and monocyte-derived macrophages. Infect Immun. 1986;54(1):90–95. doi: 10.1128/iai.54.1.90-95.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Morrison RP. Differential sensitivities of Chlamydia trachomatis strains to inhibitory effects of gamma interferon. Infect Immun. 2000;68(10):6038–6040. doi: 10.1128/iai.68.10.6938-6040.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Fields KA, McCormack R, de Armas LR, Podack ER. Perforin-2 restricts growth of Chlamydia trachomatis in macrophages. Infect Immun. 2013;81(8):3045–3054. doi: 10.1128/IAI.00497-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Borges V, Gomes JP. Deep comparative genomics among Chlamydia trachomatis lymphogranuloma venereum isolates highlights genes potentially involved in pathoadaptation. Infect Genet Evol. 2015;32:74–88. doi: 10.1016/j.meegid.2015.02.026. [DOI] [PubMed] [Google Scholar]
  • 90.Gomes JP, Bruno WJ, Nunes A, Santos N, Florindo C, Borrego MJ, Dean D. Evolution of Chlamydia trachomatis diversity occurs by widespread interstrain recombination involving hotspots. Genome. 2007;Res17(1):50–60. doi: 10.1101/gr.5674706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Nunes A, Borrego MJ, Gomes JP. Genomic features beyond Chlamydia trachomatis phenotypes: what do we think we know? Infect Genet Evol. 2013;16:392–400. doi: 10.1016/j.meegid.2013.03.018. [DOI] [PubMed] [Google Scholar]
  • 92.O'Connell CM, Ingalls RR, Andrews Jr CW, Skurlock AM, Darville T. Plasmid-deficient Chlamydia muridarum fail to induce immune pathology and protect against oviduct disease. J Immunol. 2007;179(6):4027–4034. doi: 10.4049/jimmunol.179.6.4027. [DOI] [PubMed] [Google Scholar]
  • 93.Russell M, Darville T, Chandra-Kuntal K, Smith B, Andrews Jr CW, O'Connell CM. Infectivity acts as in vivo selection for maintenance of the chlamydial cryptic plasmid. Infect Immun. 2011;79(1):98–107. doi: 10.1128/IAI.01105-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kari L, Whitmire WM, Olivares-Zavaleta N, Goheen MM, Taylor LD, Carlson JH, Sturdevant GL, Lu C, Bakios LE, Randall LB, Parnell MJ, Zhong G, Caldwell HD. A live-attenuated chlamydial vaccine protects against trachoma in nonhuman primates. J Exp Med. 2011;208(11):2217–2223. doi: 10.1084/jem.20111266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Qu Y, Frazer LC, O'Connell CM, Tarantal AF, Andrews Jr CW, O'Connor SL, Russell AN, Sullivan JE, Poston TB, Vallejo AN, Darville T. Comparable genital tract infection, pathology, and immunity in rhesus macaques inoculated with wild-type or plasmid-deficient Chlamydia trachomatis serovar D. Infect Immun. 2015;83(10):4056–4067. doi: 10.1128/IAI.00841-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Matsumoto A, Izutsu H, Miyashita N, Ohuchi M. Plaque formation by and plaque cloning of Chlamydia trachomatis biovar trachoma. J Clinical Microbiol. 1998;36(10):3013–3019. doi: 10.1128/jcm.36.10.3013-3019.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.O'Connell CM, Nicks KM. A plasmid-cured Chlamydia muridarum strain displays altered plaque morphology and reduced infectivity in cell culture. Microbiol. 2006;152(6):1601–1607. doi: 10.1099/mic.0.28658-0. [DOI] [PubMed] [Google Scholar]
  • 98.Carlson JH, Whitmire WM, Crane DD, Wicke L, Virtaneva K, Sturdevant DE, Kupko JJ III, Porcella SF, Martinez-Orengo N, Heinzen RA, Kari L, Caldwell HD. The Chlamydia trachomatis plasmid is a transcriptional regulator of chromosomal genes and a virulence vactor. Infect Immun. 2008;76(6):2273–2283. doi: 10.1128/iai.00102-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Song L, Carlson JH, Whitmire WM, Kari L, Virtaneva K, Sturdevant DE, Watkins H, Zhou B, Sturdevant GL, Porcella SF, McClarty G, Caldwell HD. Chlamydia trachomatis plasmid-encoded Pgp4 is a transcriptional regulator of virulence-associated genes. Infect Immun. 2013;81(3):636–644. doi: 10.1128/IAI.01305-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.O'Connell CM, AbdelRahman YM, Green E, Darville HK, Saira K, Smith B, Darville T, Scurlock AM, Meyer CR, Belland RJ. Toll-like receptor 2 activation by Chlamydia trachomatis is plasmid dependent, and plasmid-responsive chromosomal loci are coordinately regulated in response to glucose limitation by C. trachomatis but not by C. muridarum. . Infect Immun. 2011;79(3):1044–1056. doi: 10.1128/IAI.01118-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Liu Y, Huang Y, Yang Z, Sun Y, Gong S, Hou S, Chen C, Li Z, Liu Q, Wu Y, Baseman J, Zhong G. Plasmid-encoded Pgp3 is a major virulence factor for Chlamydia muridarum to induce hydrosalpinx in mice. Infect Immun. 2014;82(12):5327–5335. doi: 10.1128/IAI.02576-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Li Z, Chen D, Zhong Y, Wang S, Zhong G. The chlamydial plasmid-encoded protein pgp3 is secreted into the cytosol of Chlamydia-infected cells. Infect Immun. 2008;76(8):3415–3428. doi: 10.1128/IAI.01377-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hou S, Dong X, Yang Z, Li Z, Liu Q, Zhong G. Chlamydial plasmid-encoded virulence factor Pgp3 neutralizes the antichlamydial activity of human cathelicidin LL-37. Infect Immun. 2015;83(12):4701–4709. doi: 10.1128/IAI.00746-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Belland RJ, Nelson DE, Virok D, Crane DD, Hogan D, Sturdevant D, Beatty WL, Caldwell HD. Transcriptome analysis of chlamydial growth during IFN-gamma-mediated persistence and reactivation. Proc Natl Acad Sci USA. 2003;100(26):15971–15976. doi: 10.1073/pnas.2535394100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Nicholson T, Stephens RS. Proceedings of Tenth Meeting of the International Society of Human Chlamydial Infections. San Francisco, CA USA: > Schachter, J, Christiansen, G et. al. (Eds), International Chlamydia Symposium ; 2002. Chlamydial genomic transcriptional profile for penicillin-induced persistence. pp. 611–614. [Google Scholar]
  • 106.Carrasco JA, Tan C, Rank RG, Hsia RC, Bavoil PM. Altered developmental expression of polymorphic membrane proteins in penicillin-stressed Chlamydia trachomatis. Cell Microbiol. 2011;13(7):1014–1025. doi: 10.1111/j.1462-5822.2011.01598.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Belland RJ, Zhong G, Crane DD, Hogan D, Sturdevant D, Sharma J, Beatty WL, Caldwell HD. Genomic transcriptional profiling of the developmental cycle of Chlamydia trachomatis. Proc Natl Acad Sci USA. 2003;100(14):8478–8483. doi: 10.1073/pnas.1331135100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Vodstrcil LA, McIver R, Huston WM, Tabrizi SN, Timms P, Hocking JS. The Epidemiology of Chlamydia trachomatis organism load during genital infection: a systematic review. J Infect Dis. 2015;211(10):1628–1645. doi: 10.1093/infdis/jiu670. [DOI] [PubMed] [Google Scholar]
  • 109.Russell AN, Zheng X, O'Connell CM, Taylor BD, Wiesenfeld HC, Hillier SL, Zhong W, Darville T. Analysis of factors driving incident and ascending infection and the role of serum antibody in Chlamydia trachomatis genital tract infection. 2016;(4):523–531. doi: 10.1093/infdis/jiv438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Yeow TC, Wong WF, Sabet NS, Sulaiman S, Shahhosseini F, Tan GM, Movahed E, Looi CY, Shankar EM, Gupta R, Arulanandam BP, Hassan J, Abu Bakar S. Prevalence of plasmid-bearing and plasmid-free Chlamydia trachomatis infection among women who visited obstetrics and gynecology clinics in Malaysia. BMC Microbiol. 2016;16(1):45. doi: 10.1186/s12866-016-0671-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Lewis ME, Belland RJ, AbdelRahman YM, Beatty WL, Aiyar AA, Zea AH, Greene SJ, Marrero L, Buckner LR, Tate DJ, McGowin CL, Kozlowski PA, O'Brien M, Lillis RA, Martin DH, Quayle AJ. 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]
  • 112.Kawana K, Quayle AJ, Ficarra M, Ibana JA, Shen L, Kawana Y, Yang H, Marrero L, Yavagal S, Greene SJ, Zhang YX, Pyles RB, Blumberg RS, Schust DJ. CD1d degradation in Chlamydia trachomatis-infected epithelial cells is the result of both cellular and chlamydial proteasomal activity. J Biol Chem. 2007;282(10):7368–7375. doi: 10.1074/jbc.M610754200. [DOI] [PubMed] [Google Scholar]
  • 113.Zheng XOC CM, Nagarajan U, Wiesenfeld H, Hillier S, Darville T. Proceedings of Thirteenth Meeting of the International Society of Human Chlamydial Infections. San Francisco, CA USA: Schachter, J, Christiansen, G et. al. (Eds), International Chlamydia Symposium ; 2014. Identification of a blood transcriptional signature for chlamydial PID. [Google Scholar]
  • 114.Balamuth F, Zhang Z, Rappaport E, Hayes K, Mollen C, Sullivan KE. RNA biosignatures in adolescent patients in a pediatric emergency department with pelvic inflammatory disease. Pediatr Emerg Care. 2015;31(7):465–472. doi: 10.1097/PEC.0000000000000483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wallace NA, Robinson K, Galloway DA. Beta human papillomavirus E6 expression inhibits stabilization of p53 and increases tolerance of genomic instability. J Virol. 2014;88(11):6112–6127. doi: 10.1128/JVI.03808-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Grieshaber SS, Grieshaber NA, Miller N, Hackstadt T. Chlamydia trachomatis causes centrosomal defects resulting in chromosomal segregation abnormalities. Traffic. 2006;7(8):940–949. doi: 10.1111/j.1600-0854.2006.00439.x. [DOI] [PubMed] [Google Scholar]
  • 117.Johnson KA, Tan M, Sutterlin C. Centrosome abnormalities during a Chlamydia trachomatis infection are caused by dysregulation of the normal duplication pathway. Cell Microbiol. 2009;11(7):1064–1073. doi: 10.1111/j.1462-5822.2009.01307.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Knowlton AE, Brown HM, Richards TS, Andreolas LA, Patel RK, Grieshaber SS. Chlamydia trachomatis infection causes mitotic spindle pole defects independently from its effects on centrosome amplification. Traffic. 2011;12(7):854–866. doi: 10.1111/j.1600-0854.2011.01204.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Mital J, Miller NJ, Fischer ER, Hackstadt T. Specific chlamydial inclusion membrane proteins associate with active Src family kinases in microdomains that interact with the host microtubule network. Cell Microbiol. 2010;12(9):1235–1249. doi: 10.1111/j.1462-5822.2010.01465.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Richards TS, Knowlton AE, Grieshaber SS. Chlamydia trachomatis homotypic inclusion fusion is promoted by host microtubule trafficking. BMC microbiology. 2013;13(185) doi: 10.1186/1471-2180-13-185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Dumoux M, Menny A, Delacour D, Hayward RD. A Chlamydia effector recruits CEP170 to reprogram host microtubule organization. J Cell Sci. 2015;128(18):3420–3434. doi: 10.1242/jcs.169318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nigg EA. Centrosome aberrations: cause or consequence of cancer progression? Nat Rev Cancer. 2002;2(11):815–825. doi: 10.1038/nrc924. [DOI] [PubMed] [Google Scholar]
  • 123.Bolgioni AF, Ganem NJ. The interplay between centrosomes and the Hippo tumor suppressor pathway. Chromosome Res. 2016;24(1):93–104. doi: 10.1007/s10577-015-9502-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Chumduri C, Gurumurthy RK, Zadora PK, Mi Y, Meyer TF. Chlamydia infection promotes host DNA damage and proliferation but impairs the DNA damage response. Cell Host Microbe. 2013;13(6):746–758. doi: 10.1016/j.chom.2013.05.010. [DOI] [PubMed] [Google Scholar]
  • 125.Gonzalez E, Rother M, Kerr MC, Al-Zeer MA, Abu-Lubad M, Kessler M, Brinkmann V, Loewer A, Meyer TF. Chlamydia infection depends on a functional MDM2-p53 axis. Nat Commun. 2014;5:5201. doi: 10.1038/ncomms6201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Siegl C, Prusty BK, Karunakaran K, Wischhusen J, Rudel T. Tumor suppressor p53 alters host cell metabolism to limit Chlamydia trachomatis infection. Cell Rep. 2014;9(3):918–929. doi: 10.1016/j.celrep.2014.10.004. [DOI] [PubMed] [Google Scholar]
  • 127.Knowlton AE, Fowler LJ, Patel RK, Wallet SM, Grieshaber SS. Chlamydia induces anchorage independence in 3T3 cells and detrimental cytological defects in an infection model. PLoS One. 2013;8(1):e54022. doi: 10.1371/journal.pone.0054022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Merbah M, Introini A, Fitzgerald W, Grivel JC, Lisco A, Vanpouille C, Margolis L. Cervico-vaginal tissue ex vivo as a model to study early events in HIV-1 infection. A Reprod Immunol. 2011;65(3):268–278. doi: 10.1111/j.1600-0897.2010.00967.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Buckner LR, Amedee AM, Albritton HL, Kozlowski PA, Lacour N, McGowin CL, Schust DJ, Quayle AJ. Chlamydia trachomatis infection of endocervical epithelial cells enhances early HIV transmission events. PLoS One. 2016;11(1):e0146663. doi: 10.1371/journal.pone.0146663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Gaydos CA, Crotchfelt KA, Howell MR, Kralian S, Hauptman P, Quinn TC. Molecular amplification assays to detect chlamydial infections in urine specimens from high school female students and to monitor the persistence of chlamydial DNA after therapy. J Infect Dis. 1998;177(2):417–424. doi: 10.1086/514207. [DOI] [PubMed] [Google Scholar]
  • 131.Geisler WM. Diagnosis and management of uncomplicated Chlamydia trachomatis infections in adolescents and adults: summary of evidence reviewed for the 2015 Centers for Disease Control and Prevention sexually transmitted diseases treatment guidelines. Clin Infect Dis 61 Suppl. 2015;8:S774–S784. doi: 10.1093/cid/civ694. [DOI] [PubMed] [Google Scholar]
  • 132.Sandoz KM, Rockey DD. Antibiotic resistance in Chlamydiae. Future Microbiol. 2010;5(9):1427–1442. doi: 10.2217/fmb.10.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Binet R, Maurelli AT. Frequency of development and associated physiological cost of azithromycin resistance in Chlamydia psittaci 6BC and C. trachomatis L2. Antimicrob Agents Chemother. 2007;51(12):4267–4275. doi: 10.1128/AAC.00962-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Binet R, Maurelli AT. Fitness cost due to mutations in the 16S rRNA associated with spectinomycin resistance in Chlamydia psittaci 6BC. Antimicrob Agents Chemother. 2005;49(11):4455–4464. doi: 10.1128/AAC.49.11.4455-4464.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Golden MR, Whittington WL, Handsfield HH, Hughes JP, Stamm WE, Hogben M, Clark A, Malinski C, Helmers JR, Thomas KK, Holmes KK. Effect of expedited treatment of sex partners on recurrent or persistent gonorrhea or chlamydial infection. N Engl J Med. 2005;352(7):676–685. doi: 10.1056/NEJMoa041681. [DOI] [PubMed] [Google Scholar]
  • 136.Rank RG, Yeruva L. Hidden in plain sight: chlamydial gastrointestinal infection and its relevance to persistence in human genital infection. Infect Immun. 2014;82(4):1362–1371. doi: 10.1128/IAI.01244-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Yeruva L, Spencer N, Bowlin AK, Wang Y, Rank RG. Chlamydial infection of the gastrointestinal tract: a reservoir for persistent infection. Pathog Dis. 2013;68(3):88–95. doi: 10.1111/2049-632X.12052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Dai J, Zhang T, Wang L, Shao L, Zhu C, Zhang Y, Failor C, Schenken R, Baseman J, He C, Zhong G. Intravenous inoculation of Chlamydia muridarum leads to a long-lasting infection restricted to the gastrointestinal tract. Infect Immun. 2016;84(8):2382–2388. doi: 10.1128/IAI.00432-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Yeruva L, Melnyk S, Spencer N, Bowlin A, Rank RG. Differential susceptibilities to azithromycin treatment of chlamydial infection in the gastrointestinal tract and cervix. Antimicrob Agents Chemother. 2013;57(12):6290–6294. doi: 10.1128/AAC.01405-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Wang L, Zhang Q, Zhang T, Zhang Y, Zhu C, Sun X, Zhang N, Xue M, Zhong G. The Chlamydia muridarum organisms fail to auto-inoculate the mouse genital tract after colonization in the gastrointestinal tract for 70 days. PLoS One. 2016;11(5):e0155880. doi: 10.1371/journal.pone.0155880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Borel N, Leonard C, Slade J, Schoborg RV. Chlamydial antibiotic resistance and treatment failure in veterinary and human medicine. Curr Clin Microbiol Rep. 2016;3:10–18. doi: 10.1007/s40588-016-0028-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Leonhardt RM, Lee SJ, Kavathas PB, Cresswell P. Severe tryptophan starvation blocks onset of conventional persistence and reduces reactivation of Chlamydia trachomatis. Infect Immun. 2007;75(11):5105–5117. doi: 10.1128/IAI.00668-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Leonard CA, Schoborg RV, Borel N. Damage/danger Associated Molecular Patterns (DAMPs) modulate Chlamydia pecorum and C. trachomatis serovar E inclusion development In vitro. . PLoS One. 2015;10(8):e0134943. doi: 10.1371/journal.pone.0134943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Guseva NV, Knight ST, Whittimore JD, Wyrick PB. Primary cultures of female swine genital epithelial cells in vitro: a new approach for the study of hormonal modulation of Chlamydia infection. Infect Immun. 2003;71(8):4700–4710. doi: 10.1128/IAI.71.8.4700-4710.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kintner J, Schoborg RV, Wyrick PB, Hall JV. Progesterone antagonizes the positive influence of estrogen on Chlamydia trachomatis serovar E in an Ishikawa/SHT-290 co-culture model. Pathog Dis. 2015;73(4) doi: 10.1093/femspd/ftv015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hall JV, Schell M, Dessus-Babus S, Moore CG, Whittimore JD, Sal M, Dill BD, Wyrick PB. The multifaceted role of oestrogen in enhancing Chlamydia trachomatis infection in polarized human endometrial epithelial cells. Cell Microbiol. 2011;13(8):1183–1199. doi: 10.1111/j.1462-5822.2011.01608.x. [DOI] [PubMed] [Google Scholar]
  • 147.Deka S, Vanover J, Dessus-Babus S, Whittimore J, Howett MK, Wyrick PB, Schoborg RV. Chlamydia trachomatis enters a viable but non-cultivable (persistent) state within Serpes Simplex Virus Type 2 (HSV-2) co-infected host cells. Cell Microbiol. 2006;8(1):149–162. doi: 10.1111/j.1462-5822.2005.00608.x. [DOI] [PubMed] [Google Scholar]
  • 148.Romano JD, de Beaumont C, Carrasco JA, Ehrenman K, Bavoil PM, Coppens I. A novel co-infection model with Toxoplasma and Chlamydia trachomatis highlights the importance of host cell manipulation for nutrient scavenging. Cell Microbiol. 2013;15(4):619–646. doi: 10.1111/cmi.12060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kell D, Potgieter M, Pretorius E. Individuality, phenotypic differentiation, dormancy and 'persistence' in culturable bacterial systems: commonalities shared by environmental, laboratory, and clinical microbiology. F1000Res. 2015;4(179) doi: 10.12688/f1000research.6709.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Kint CI, Verstraeten N, Fauvart M, Michiels J. New-found fundamentals of bacterial persistence. Trends Microbiol. 2012;20(12):577–585. doi: 10.1016/j.tim.2012.08.009. [DOI] [PubMed] [Google Scholar]
  • 151.Fauvart M, De Groote VN, Michiels J. Role of persister cells in chronic infections: clinical relevance and perspectives on anti-persister therapies. J Med Microbiol. 2011;60(Pt 6):699–709. doi: 10.1099/jmm.0.030932-0. [DOI] [PubMed] [Google Scholar]
  • 152.Wyrick PB, Knight ST. Pre-exposure of infected human endometrial epithelial cells to penicillin in vitro renders Chlamydia trachomatis refractory to azithromycin. J Antimicrob Chemother. 2004;54(1):79–85. doi: 10.1093/jac/dkh283. [DOI] [PubMed] [Google Scholar]
  • 153.Reveneau N, Crane DD, Fischer E, Caldwell HD. Bactericidal activity of first-choice antibiotics against gamma interferon-induced persistent infection of human epithelial cells by Chlamydia trachomatis. Antimicrob Agents Chemother. 2005;49(5):1787–1793. doi: 10.1128/AAC.49.5.1787-1793.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Phillips-Campbell R, Kintner J, Schoborg RV. Induction of the Chlamydia muridarum stress/persistence response increases azithromycin treatment failure in a murine model of infection. Antimicrob Agents Chemother. 2014;58(3):1782–1784. doi: 10.1128/AAC.02097-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Unemo M, Endre KM, Moi H. Five-day azithromycin treatment regimen for Mycoplasma genitalium infection also effectively eradicates Chlamydia trachomatis. Acta Derm Venereol. 2015;95(6):730–732. doi: 10.2340/00015555-2108. [DOI] [PubMed] [Google Scholar]
  • 156.Lanjouw E, Ouburg S, de Vries HJ, Stary A, Radcliffe K, Unemo M. 2015 European guideline on the management of Chlamydia trachomatis infections. Int J STD AIDS. 2016;27(5):333–348. doi: 10.1177/0956462415618837. [DOI] [PubMed] [Google Scholar]
  • 157.Lanjouw E, Ouburg S, de Vries HJ, Stary A, Radcliffe K, Unemo M. Background review for the '2015 European guideline on the management of Chlamydia trachomatis infections'. Int J STD AIDS. 2015; pii: 0956462415618838 doi: 10.1177/0956462415618838. [DOI] [PubMed] [Google Scholar]
  • 158.Schwebke JR, Rompalo A, Taylor S, Sena AC, Martin DH, Lopez LM, Lensing S, Lee JY. Re-evaluating the treatment of nongonococcal urethritis: emphasizing emerging pathogens--a randomized clinical trial. Clin Infect Dis. 2011;52(2):163–170. doi: 10.1093/cid/ciq074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Bjartling C, Osser S, Persson K. Mycoplasma genitalium in cervicitis and pelvic inflammatory disease among women at a gynecologic outpatient service. Am J Obstet Gynecol. 2012;206(6):e471–e478. doi: 10.1016/j.ajog.2012.02.036. [DOI] [PubMed] [Google Scholar]
  • 160.McGowin CL, Anderson-Smits C. Mycoplasma genitalium: an emerging cause of sexually transmitted disease in women. PLoS Pathog. 2011;7(5):e1001324. doi: 10.1371/journal.ppat.1001324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Bissessor M, Tabrizi SN, Twin J, Abdo H, Fairley CK, Chen MY, Vodstrcil LA, Jensen JS, Hocking JS, Garland SM, Bradshaw CS. Macrolide resistance and azithromycin failure in a Mycoplasma genitalium-infected cohort and response of azithromycin failures to alternative antibiotic regimens. Clin Infect Dis. 2015;60(8):1228–1236. doi: 10.1093/cid/ciu1162. [DOI] [PubMed] [Google Scholar]
  • 162.Pond MJ, Nori AV, Witney AA, Lopeman RC, Butcher PD, Sadiq ST. High prevalence of antibiotic-resistant Mycoplasma genitalium in nongonococcal urethritis: the need for routine testing and the inadequacy of current treatment options. Clin Infect Dis. 2014;58(5):631–637. doi: 10.1093/cid/cit752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Tagg KA, Jeoffreys NJ, Couldwell DL, Donald JA, Gilbert GL. Fluoroquinolone and macrolide resistance-associated mutations in Mycoplasma genitalium. J Clin Microbiol. 2013;51(7):2245–2249. doi: 10.1128/JCM.00495-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Horner P, Blee K, Adams E. Time to manage Mycoplasma genitalium as an STI: but not with azithromycin 1 g! Curr Opin Infect Dis. 2014;27(1):68–74. doi: 10.1097/QCO.0000000000000030. [DOI] [PubMed] [Google Scholar]
  • 165.Oakeshott P, Kerry S, Aghaizu A, Atherton H, Hay S, Taylor-Robinson D, Simms I, Hay P. Randomised controlled trial of screening for Chlamydia trachomatis to prevent pelvic inflammatory disease: the POPI (prevention of pelvic infection) trial. BMJ. 2010;340:c1642. doi: 10.1136/bmj.c1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Gottlieb SL, Xu F, Brunham RC. Screening and treating Chlamydia trachomatis genital infection to prevent pelvic inflammatory disease: interpretation of findings from randomized controlled trials. Sex Transm Dis. 2013;40(2):97–102. doi: 10.1097/OLQ.0b013e31827bd637. [DOI] [PubMed] [Google Scholar]
  • 167.Aghaizu A, Adams EJ, Turner K, Kerry S, Hay P, Simms I, Oakeshott P. What is the cost of pelvic inflammatory disease and how much could be prevented by screening for Chlamydia trachomatis? Cost analysis of the Prevention of Pelvic Infection (POPI) trial. . Sex Transm Infect. 2011;87(4):312–317. doi: 10.1136/sti.2010.048694. [DOI] [PubMed] [Google Scholar]
  • 168.Brunham RC, Rekart ML. The arrested immunity hypothesis and the epidemiology of chlamydia control. Sex Transm Dis. 2008;35(1):53–54. doi: 10.1097/OLQ.0b013e31815e41a3. [DOI] [PubMed] [Google Scholar]
  • 169.Arno JN, Katz BP, McBride R, Carty GA, Batteiger BE, Caine VA, Jones RB. Age and clinical immunity to infections with Chlamydia trachomatis. Sex Transm Dis. 1994;21(1):47–52. doi: 10.1097/00007435-199401000-00010. [DOI] [PubMed] [Google Scholar]
  • 170.Batteiger BE, Xu F, Johnson RE, Rekart ML. Protective immunity to Chlamydia trachomatis genital infection: evidence from human studies. J Infect Dis. 2010;2(S178-189) doi: 10.1086/652400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Ramsey KH, Rank RG. Resolution of chlamydial genital infection with antigen-specific T- lymphocyte lines. Infect Immun. 1991;59(3):925–931. doi: 10.1128/iai.59.3.925-931.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Riley MM, Zurenski MA, Frazer LC, O'Connell CM, Andrews Jr CW, Mintus M, Darville T. The recall response induced by genital challenge with Chlamydia muridarum protects the oviduct from pathology but not from reinfection. Infect Immun. 2012;80(6):2194–2203. doi: 10.1128/IAI.00169-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Murthy AK, Chambers JP, Meier PA, Zhong G, Arulanandam BP. Intranasal vaccination with a secreted chlamydial protein rnhances resolution of genital Chlamydia muridarum infection, protects against oviduct pathology, and is gighly dependent upon endogenous gamma interferon production. Infect Immun. 2007;75(2):666–676. doi: 10.1128/IAI.01280-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Cong Y, Jupelli M, Guentzel MN, Zhong G, Murthy AK, Arulanandam BP. Intranasal immunization with chlamydial protease-like activity factor and CpG deoxynucleotides enhances protective immunity against genital Chlamydia muridarum infection. Vaccine. 2007;25(19):3773–3780. doi: 10.1016/j.vaccine.2007.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Yu H, Jiang X, Shen C, Karunakaran KP, Jiang J, Rosin NL, Brunham RC. Chlamydia muridarum T-cell antigens formulated with the adjuvant DDA/TDB induce immunity against infection that correlates with a high frequency of gamma interferon (IFN-gamma)/tumor necrosis factor alpha and IFN-gamma/interleukin-17 double-positive CD4+ T cells. Infect Immun. 2010;78(5):2272–2282. doi: 10.1128/IAI.01374-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Olsen AW, Follmann F, Jensen K, Hojrup P, Leah R, Sorensen H, Hoffmann S, Andersen P, Theisen M. Identification of CT521 as a Frequent Target of Th1 Cells in patients with urogenital Chlamydia trachomatis infection. J Infect Dis. 2006;194(9):1258–1266. doi: 10.1086/508203. [DOI] [PubMed] [Google Scholar]
  • 177.Gervassi AL, Grabstein KH, Probst P, Hess B, Alderson MR, Fling SP. Human CD8+ T cells recognize the 60-kDa cysteine-rich outer membrane protein from Chlamydia trachomatis. J Immunol. 2004;173(11):6905–6913. doi: 10.4049/jimmunol.173.11.6905. [DOI] [PubMed] [Google Scholar]
  • 178.Cohen CR, Koochesfahani KM, Meier AS, Shen C, Karunakaran K, Ondondo B, Kinyari T, Mugo NR, Nguti R, Brunham RC. Immunoepidemiologic profile of Chlamydia trachomatis infection: importance of heat-shock protein 60 and interferon- gamma. J InfectDis. 2005;192(4):591–599. doi: 10.1086/432070. [DOI] [PubMed] [Google Scholar]
  • 179.Morrison SG, Morrison RP. A predominant role for antibody in acquired immunity to chlamydial genital tract reinfection. J Immunol. 2005;175(11):7536–7542. doi: 10.4049/jimmunol.175.11.7536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Brady LJ. Antibody-mediated immunomodulation: a strategy to improve host responses against microbial antigens. Infect Immun. 2005;73(2):671–678. doi: 10.1128/IAI.73.2.671-678.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Punnonen R, Terho P, Nikkanen V, Meurman O. Chlamydial serology in infertile women by immunofluorescence. Fert Steril. 1979;31(656-659) doi: 10.1016/S0015-0282(16)44056-2. [DOI] [PubMed] [Google Scholar]
  • 182.Brunham RC. Immunity to Chlamydia trachomatis. J Infect Dis. 2013;207(12):1796–1797. doi: 10.1093/infdis/jit095. [DOI] [PubMed] [Google Scholar]
  • 183.Poston TB, Darville T. Chlamydia trachomatis: protective adaptive responses and prospects for a vaccine. Curr Top Microbiol Immunol. 2016 doi: 10.1007/82_2016_6. [DOI] [PubMed] [Google Scholar]
  • 184.Lorenzen E, Follmann F, Boje S, Erneholm K, Olsen AW, Agerholm JS, Jungersen G, Andersen P. Intramuscular priming and intranasal boosting induce strong genital immunity through secretory IgA in minipigs infected with Chlamydia trachomatis. Front Immunol. 2015;6:628. doi: 10.3389/fimmu.2015.00628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Olsen AW, Follmann F, Erneholm K, Rosenkrands I, Andersen P. Protection Against Chlamydia trachomatis infection and upper genital tract pathological changes by vaccine-promoted neutralizing antibodies directed to the VD4 of the major outer membrane protein. J Infect Dis. 2015;212(6):978–989. doi: 10.1093/infdis/jiv137. [DOI] [PubMed] [Google Scholar]
  • 186.Boje S, Olsen AW, Erneholm K, Agerholm JS, Jungersen G, Andersen P, Follmann F. A multi-subunit Chlamydia vaccine inducing neutralizing antibodies and strong IFN-gamma(+) CMI responses protects against a genital infection in minipigs. Immunol Cell Biol. 2016;94(2):185–195. doi: 10.1038/icb.2015.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.NanoBio Corporation (2015) NanoBio’s chlamydia vaccine improves clearance of bacteria and prevents Pelvic Inflammatory Disease in mice. Press release available at: http://www.nanobio.com/chlamydia-vaccine-update. [Accessed: 26.08.2016] [Google Scholar]
  • 188.Karunakaran KP, Yu H, Jiang X, Chan Q, Moon KM, Foster LJ, Brunham RC. Outer membrane proteins preferentially load MHC class II peptides: implications for a Chlamydia trachomatis T cell vaccine. Vaccine. 2015;33(18):2159–2166. doi: 10.1016/j.vaccine.2015.02.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Stary G, Olive A, Radovic-Moreno AF, Gondek D, Alvarez D, Basto PA, Perro M, Vrbanac VD, Tager AM, Shi J, Yethon JA, Farokhzad OC, Langer R, Starnbach MN, von Andrian UH. VACCINES. A mucosal vaccine against Chlamydia trachomatis generates two waves of protective memory T cells. . Science. 2015;348(6241):aaa8205. doi: 10.1126/science.aaa8205. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Microbial Cell are provided here courtesy of Shared Science Publishers

RESOURCES