Reviews:

Microbial Cell, Vol. 3, No. 9, pp. 390 - 403; doi: 10.15698/mic2016.09.525

Chlamydia trachomatis Genital Infections

Catherine M. O’Connell and Morgan E. Ferone

Download PDF download pdf
Show/hide additional information

    Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.

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

Abbreviations:

EB – elementary body,

GI – gastrointestinal,

HPV – human papillomavirus

LGV – lymphoma granuloma venereum,

MOMP – major outer membrane protein,

PID – pelvic inflammatory disease,

PMP – polymorphic surface protein,

STI – sexually transmitted infection.
Received originally: 25/07/2016 Received in revised form: 30/08/2016
Accepted: 30/08/2016 Published: 05/09/2016

Correspondence:
Catherine M. O’Connell, University of North Carolina at Chapel Hill, 8301B MBRB, CB# 7509, 111 Mason Farm Road, Chapel Hill, NC 27599-7509, USA catherine.oconnell@unc.edu

Conflict of interest statement: The authors have no conflict of interest to declare.
Please cite this article as: Catherine M. O’Connell and Morgan E. Ferone (2016). Chlamydia trachomatis Genital Infections. Microbial Cell 3(9): 390-403.

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.

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, elicited high levels of serum and vaginal antibody with chlamydia specific IL-17/IFN-γ responses and reduced rates of oviduct pathology 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.

References

  1. S.P. Wang, and J.T. Grayston, "Immunologic relationship between genital TRIC, lymphogranuloma venereum, and related organisms in a new microtiter indirect immunofluorescence test.", American journal of ophthalmology, 1970. http://www.ncbi.nlm.nih.gov/pubmed/4915925
  2. R.S. Stephens, R. Sanchez-Pescador, E.A. Wagar, C. Inouye, and M.S. Urdea, "Diversity of Chlamydia trachomatis major outer membrane protein genes.", Journal of bacteriology, 1987. http://www.ncbi.nlm.nih.gov/pubmed/3040664
  3. L. Newman, J. Rowley, S. Vander Hoorn, N.S. Wijesooriya, M. Unemo, N. Low, G. Stevens, S. Gottlieb, J. Kiarie, and M. Temmerman, "Global Estimates of the Prevalence and Incidence of Four Curable Sexually Transmitted Infections in 2012 Based on Systematic Review and Global Reporting", PLOS ONE, vol. 10, pp. e0143304, 2015. http://dx.doi.org/10.1371/journal.pone.0143304
  4. . Centers for Disease Control and Prevention, "Sexually Transmitted Disease Surveillance 2014.", Available at : http://www.cdc.gov/std/stats14/surv-2014-print.pdf. Accessed: 16.07.2016., 2015.
  5. J. Crichton, M. Hickman, R. Campbell, H. Batista-Ferrer, and J. Macleod, "Socioeconomic factors and other sources of variation in the prevalence of genital chlamydia infections: A systematic review and meta-analysis", BMC Public Health, vol. 15, 2015. http://dx.doi.org/10.1186/s12889-015-2069-7
  6. A. Aghaizu, F. Reid, S. Kerry, P.E. Hay, H. Mallinson, J.S. Jensen, S. Kerry, S. Kerry, and P. Oakeshott, "Frequency and risk factors for incident and redetectedChlamydia trachomatisinfection in sexually active, young, multi-ethnic women: a community based cohort study", Sexually Transmitted Infections, vol. 90, pp. 524-528, 2014. http://dx.doi.org/10.1136/sextrans-2014-051607
  7. B. Batteiger, W. Tu, S. Ofner, B. Van Der Pol, D. Stothard, D. Orr, B. Katz, and J. Fortenberry, "RepeatedChlamydia trachomatisGenital Infections in Adolescent Women", The Journal of Infectious Diseases, vol. 201, pp. 42-51, 2010. http://dx.doi.org/10.1086/648734
  8. L.Y. Hwang, Y. Ma, and A. Moscicki, "Biological and Behavioral Risks for Incident Chlamydia trachomatis Infection in a Prospective Cohort", Obstetrics & Gynecology, vol. 124, pp. 954-960, 2014. http://dx.doi.org/10.1097/AOG.0000000000000429
  9. M.J. Jørgensen, H.T. Maindal, M.B. Larsen, K.S. Christensen, F. Olesen, and B. Andersen, "Chlamydia trachomatis infection in young adults — association with concurrent partnerships and short gap length between partners", Infectious Diseases, vol. 47, pp. 838-845, 2015. http://dx.doi.org/10.3109/23744235.2015.1071916
  10. W.E. Stamm, K.F. Wagner, R. Amsel, E.R. Alexander, M. Turck, G.W. Counts, and K.K. Holmes, "Causes of the Acute Urethral Syndrome in Women", New England Journal of Medicine, vol. 303, pp. 409-415, 1980. http://dx.doi.org/10.1056/NEJM198008213030801
  11. J. Marrazzo, "Predicting chlamydial and gonococcal cervical infection: implications for management of cervicitis", Obstetrics & Gynecology, vol. 100, pp. 579-584, 2002. http://dx.doi.org/10.1016/S0029-7844(02)02140-3
  12. M. Molano, C. Meijer, E. Weiderpass, A. Arslan, H. Posso, S. Franceschi, M. Ronderos, N. Muñoz, and A. van den Brule, "The Natural Course ofChlamydia trachomatisInfection in Asymptomatic Colombian Women: A 5‐Year Follow‐Up Study", The Journal of Infectious Diseases, vol. 191, pp. 907-916, 2005. http://dx.doi.org/10.1086/428287
  13. W.M. Geisler, S.Y. Lensing, C.G. Press, and E.W. Hook, "Spontaneous Resolution of Genital Chlamydia trachomatis Infection in Women and Protection from Reinfection", The Journal of Infectious Diseases, vol. 207, pp. 1850-1856, 2013. http://dx.doi.org/10.1093/infdis/jit094
  14. K. Workoswki, G. Bolan, and . Centers for Disease Control and Prevention, "Sexually transmitted diseases treatment guidelines, 2015.", MMWR Recomm Rep 64(RR-03): 1-137., 2015.
  15. H.C. Wiesenfeld, S.L. Hillier, L.A. Meyn, A.J. Amortegui, and R.L. Sweet, "Subclinical Pelvic Inflammatory Disease and Infertility", Obstetrics & Gynecology, vol. 120, pp. 37-43, 2012. http://dx.doi.org/10.1097/AOG.0b013e31825a6bc9
  16. L.H. Bachmann, C.M. Richey, K. Waites, J.R. Schwebke, and E.W. Hook, "Patterns of Chlamydia trachomatis Testing and Follow-Up at a University Hospital Medical Center", Sexually Transmitted Diseases, vol. 26, pp. 496-499, 1999. http://dx.doi.org/10.1097/00007435-199910000-00002
  17. W.M. Geisler, C. Wang, S.G. Morrison, C.M. Black, C.I. Bandea, and E.W. Hook, "The Natural History of Untreated Chlamydia trachomatis Infection in the Interval Between Screening and Returning for Treatment", Sexually Transmitted Diseases, vol. 35, pp. 119-123, 2008. http://dx.doi.org/10.1097/OLQ.0b013e318151497d
  18. J. Kimani, I.W. Maclean, J.J. Bwayo, K. MacDonald, J. Oyugi, G.M. Maitha, R.W. Peeling, M. Cheang, N.J.D. Nagelkerke, F.A. Plummer, and R.C. Brunham, "Risk Factors for Chlamydia trachomatis Pelvic Inflammatory Disease among Sex Workers in Nairobi, Kenya", Journal of Infectious Diseases, vol. 173, pp. 1437-1444, 1996. http://dx.doi.org/10.1093/infdis/173.6.1437
  19. R.B. Ness, K.J. Smith, C.H. Chang, E.F. Schisterman, and D.C. Bass, "Prediction of Pelvic Inflammatory Disease Among Young, Single, Sexually Active Women", Sexually Transmitted Diseases, vol. 33, pp. 137-142, 2006. http://dx.doi.org/10.1097/01.olq.0000187205.67390.d1
  20. I.J. Bakken, F.E. Skjeldestad, and S.A. Nordbø, "Chlamydia Trachomatis Infections Increase the Risk for Ectopic Pregnancy: A Population-Based, Nested Case–Control Study", Sexually Transmitted Diseases, vol. 34, pp. 166-169, 2007. http://dx.doi.org/10.1097/01.olq.0000230428.06837.f7
  21. M. Egger, N. Low, G.D. Smith, B. Lindblom, and B. Herrmann, "Screening for chlamydial infections and the risk of ectopic pregnancy in a county in Sweden: ecological analysis", BMJ, vol. 316, pp. 1776-1780, 1998. http://dx.doi.org/10.1136/bmj.316.7147.1776
  22. B. Liu, C.L. Roberts, M. Clarke, L. Jorm, J. Hunt, and J. Ward, "Chlamydia and gonorrhoea infections and the risk of adverse obstetric outcomes: a retrospective cohort study", Sexually Transmitted Infections, vol. 89, pp. 672-678, 2013. http://dx.doi.org/10.1136/sextrans-2013-051118
  23. S. Hollegaard, I. Vogel, P. Thorsen, I.P. Jensen, C. Mordhorst, and B. Jeune, "Chlamydia trachomatis C-complex serovars are a risk factor for preterm birth.", In vivo (Athens, Greece), 2007. http://www.ncbi.nlm.nih.gov/pubmed/17354622
  24. W.W. Andrews, R.L. Goldenberg, B. Mercer, J. Iams, P. Meis, A. Moawad, A. Das, J. VanDorsten, S.N. Caritis, G. Thurnau, M. Miodovnik, J. Roberts, and D. McNellis, "The Preterm Prediction Study: Association of second-trimester genitourinary chlamydia infection with subsequent spontaneous preterm birth", American Journal of Obstetrics and Gynecology, vol. 183, pp. 662-668, 2000. http://dx.doi.org/10.1067/mob.2000.106556
  25. I.G. Rours, M.R. Hammerschlag, A. Ott, T.J. De Faber, H.A. Verbrugh, R. de Groot, and R.P. Verkooyen, "Chlamydia trachomatis as a Cause of Neonatal Conjunctivitis in Dutch Infants", Pediatrics, vol. 121, pp. e321-e326, 2008. http://dx.doi.org/10.1542/peds.2007-0153
  26. G.I.J.G. Rours, M.R. Hammerschlag, G.J.J. Van Doornum, W.C.J. Hop, R. de Groot, H.F.M. Willemse, H.A. Verbrugh, and R.P. Verkooyen, "Chlamydia trachomatis respiratory infection in Dutch infants", Archives of Disease in Childhood, vol. 94, pp. 705-707, 2009. http://dx.doi.org/10.1136/adc.2008.152066
  27. J. Silva, F. Cerqueira, and R. Medeiros, "Chlamydia trachomatis infection: implications for HPV status and cervical cancer", Archives of Gynecology and Obstetrics, vol. 289, pp. 715-723, 2013. http://dx.doi.org/10.1007/s00404-013-3122-3
  28. P. Paba, D. Bonifacio, L. Di Bonito, D. Ombres, C. Favalli, K. Syrjänen, and M. Ciotti, "Co-Expression of HSV2 and <i>Chlamydia trachomatis</i> in HPV-Positive Cervical Cancer and Cervical Intraepithelial Neoplasia Lesions Is Associated with Aberrations in Key Intracellular Pathways", Intervirology, vol. 51, pp. 230-234, 2008. http://dx.doi.org/10.1159/000156481
  29. J. Paavonen, K.P. Karunakaran, Y. Noguchi, T. Anttila, A. Bloigu, J. Dillner, G. Hallmans, T. Hakulinen, E. Jellum, P. Koskela, M. Lehtinen, S. Thoresen, H. Lam, C. Shen, and R.C. Brunham, "Serum antibody response to the heat shock protein 60 of Chlamydia trachomatis in women with developing cervical cancer", American Journal of Obstetrics and Gynecology, vol. 189, pp. 1287-1292, 2003. http://dx.doi.org/10.1067/s0002-9378(03)00755-5
  30. H. Zhu, Z. Shen, H. Luo, W. Zhang, and X. Zhu, "Chlamydia Trachomatis Infection-Associated Risk of Cervical Cancer", Medicine, vol. 95, pp. e3077, 2016. http://dx.doi.org/10.1097/MD.0000000000003077
  31. D.R. Nonato, R.R. Alves, A.A. Ribeiro, V.A. Saddi, K.D. Segati, K.P. Almeida, Y.A. de Lima, W.B. D’Alessandro, and S.H. Rabelo-Santos, "Prevalence and factors associated with coinfection of human papillomavirus and Chlamydia trachomatis in adolescents and young women", American Journal of Obstetrics and Gynecology, vol. 215, pp. 753.e1-753.e9, 2016. http://dx.doi.org/10.1016/j.ajog.2016.07.003
  32. E.M. Quinónez-Calvache, D.I. Ríos-Chaparro, J.D. Ramírez, S.C. Soto-De León, M. Camargo, L. Del Río-Ospina, R. Sánchez, M.E. Patarroyo, and M.A. Patarroyo, "Chlamydia trachomatis Frequency in a Cohort of HPV-Infected Colombian Women", PLOS ONE, vol. 11, pp. e0147504, 2016. http://dx.doi.org/10.1371/journal.pone.0147504
  33. C.P. Crum, R. Drapkin, A. Miron, T.A. Ince, M. Muto, D.W. Kindelberger, and Y. Lee, "The distal fallopian tube: a new model for pelvic serous carcinogenesis", Current Opinion in Obstetrics & Gynecology, vol. 19, pp. 3-9, 2007. http://dx.doi.org/10.1097/GCO.0b013e328011a21f
  34. Y. Lee, A. Miron, R. Drapkin, M. Nucci, F. Medeiros, A. Saleemuddin, J. Garber, C. Birch, H. Mou, R. Gordon, D. Cramer, F. McKeon, and C. Crum, "A candidate precursor to serous carcinoma that originates in the distal fallopian tube", The Journal of Pathology, vol. 211, pp. 26-35, 2006. http://dx.doi.org/10.1002/path.2091
  35. F. Medeiros, M.G. Muto, Y. Lee, J.A. Elvin, M.J. Callahan, C. Feltmate, J.E. Garber, D.W. Cramer, and C.P. Crum, "The Tubal Fimbria Is a Preferred Site for Early Adenocarcinoma in Women With Familial Ovarian Cancer Syndrome", American Journal of Surgical Pathology, vol. 30, pp. 230-236, 2006. http://dx.doi.org/10.1097/01.pas.0000180854.28831.77
  36. M.R. Oliver Perez, J. Magriñá, A.T. García, and J.S. Jiménez Lopez, "Prophylactic salpingectomy and prophylactic salpingoophorectomy for adnexal high-grade serous epithelial carcinoma: A reappraisal", Surgical Oncology, vol. 24, pp. 335-344, 2015. http://dx.doi.org/10.1016/j.suronc.2015.09.008
  37. I.M. Zardawi, "Primary Fallopian Tube Carcinoma Arising in the Setting of Chronic Pelvic Inflammatory Disease", Case Reports in Medicine, vol. 2014, pp. 1-7, 2014. http://dx.doi.org/10.1155/2014/645045
  38. S. Salvador, B. Gilks, M. Köbel, D. Huntsman, B. Rosen, and D. Miller, "The Fallopian Tube: Primary Site of Most Pelvic High-grade Serous Carcinomas", International Journal of Gynecologic Cancer, vol. 19, pp. 58-64, 2009. http://dx.doi.org/10.1111/IGC.0b013e318199009c
  39. R. Ness, M. Goodman, C. Shen, and R. Brunham, "Serologic Evidence of Past Infection withChlamydia trachomatis,in Relation to Ovarian Cancer", The Journal of Infectious Diseases, vol. 187, pp. 1147-1152, 2003. http://dx.doi.org/10.1086/368380
  40. R.B. Ness, D.E. Soper, H.E. Richter, H. Randall, J.F. Peipert, D.B. Nelson, D. Schubeck, S.G. McNeeley, W. Trout, D.C. Bass, K. Hutchison, K. Kip, and R.C. Brunham, "Chlamydia Antibodies, Chlamydia Heat Shock Protein, and Adverse Sequelae After Pelvic Inflammatory Disease: The PID Evaluation and Clinical Health (PEACH) Study", Sexually Transmitted Diseases, vol. 35, pp. 129-135, 2008. http://dx.doi.org/10.1097/OLQ.0b013e3181557c25
  41. A. Idahl, E. Lundin, M. Jurstrand, U. Kumlin, F. Elgh, N. Ohlson, and U. Ottander, "Chlamydia trachomatisandMycoplasma genitaliumPlasma Antibodies in Relation to Epithelial Ovarian Tumors", Infectious Diseases in Obstetrics and Gynecology, vol. 2011, pp. 1-10, 2011. http://dx.doi.org/10.1155/2011/824627
  42. S. Shanmughapriya, G. SenthilKumar, K. Vinodhini, B.C. Das, N. Vasanthi, and K. Natarajaseenivasan, "Viral and bacterial aetiologies of epithelial ovarian cancer", European Journal of Clinical Microbiology & Infectious Diseases, vol. 31, pp. 2311-2317, 2012. http://dx.doi.org/10.1007/s10096-012-1570-5
  43. A. Idahl, E. Lundin, F. Elgh, M. Jurstrand, J.K. Møller, I. Marklund, P. Lindgren, and U. Ottander, "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", American Journal of Obstetrics and Gynecology, vol. 202, pp. 71.e1-71.e6, 2010. http://dx.doi.org/10.1016/j.ajog.2009.07.042
  44. W. Stamm, "Chlamydia trachomatisInfections: Progress and Problems", The Journal of Infectious Diseases, vol. 179, pp. S380-S383, 1999. http://dx.doi.org/10.1086/513844
  45. G.F. Gonzales, G. Munoz, R. Sanchez, R. Henkel, G. Gallegos-Avila, O. Diaz-Gutierrez, P. Vigil, F. Vasquez, G. Kortebani, A. Mazzolli, and E. Bustos-Obregon, "Update on the impact of Chlamydia trachomatis infection on male fertility", Andrologia, vol. 36, pp. 1-23, 2004. http://dx.doi.org/10.1046/j.0303-4569.2003.00594.x
  46. W.E. STAMM, "Chlamydia trachomatisUrethral Infections in Men", Annals of Internal Medicine, vol. 100, pp. 47, 1984. http://dx.doi.org/10.7326/0003-4819-100-1-47
  47. M.P. Hedger, "Immunophysiology and Pathology of Inflammation in the Testis and Epididymis", Journal of Andrology, vol. 32, pp. 625-640, 2011. http://dx.doi.org/10.2164/jandrol.111.012989
  48. A. Keat, B.J. Thomas, and D. Taylor-Robinson, "Chlamydial infection in the aetiology of arthritis.", British medical bulletin, 1983. http://www.ncbi.nlm.nih.gov/pubmed/6347328
  49. T. Hannu, "Reactive arthritis", Best Practice & Research Clinical Rheumatology, vol. 25, pp. 347-357, 2011. http://dx.doi.org/10.1016/j.berh.2011.01.018
  50. M.U. Rahman, M.A. Cheema, H.R. Schumacher, and A.P. Hudson, "Molecular evidence for the presence of chlamydia in the synovium of patients with reiter's syndrome", Arthritis & Rheumatism, vol. 35, pp. 521-529, 1992. http://dx.doi.org/10.1002/art.1780350506
  51. D. Taylor-Robinson, C. Gilroy, B. Thomas, and A. Keat, "Detection of Chlamydia trachomatis DNA in joints of reactive arthritis patients by polymerase chain reaction", The Lancet, vol. 340, pp. 81-82, 1992. http://dx.doi.org/10.1016/0140-6736(92)90399-N
  52. K.A. Redgrove, and E.A. McLaughlin, "The Role of the Immune Response in Chlamydia trachomatis Infection of the Male Genital Tract: A Double-Edged Sword", Frontiers in Immunology, vol. 5, 2014. http://dx.doi.org/10.3389/fimmu.2014.00534
  53. N.H.N. de Vrieze, and H.J.C. de Vries, "Lymphogranuloma venereum among men who have sex with men. An epidemiological and clinical review", Expert Review of Anti-infective Therapy, vol. 12, pp. 697-704, 2014. http://dx.doi.org/10.1586/14787210.2014.901169
  54. H.J. de Vries, A. Zingoni, J.A. White, J.D. Ross, and A. Kreuter, "2013 European Guideline on the management of proctitis, proctocolitis and enteritis caused by sexually transmissible pathogens", International Journal of STD & AIDS, vol. 25, pp. 465-474, 2013. http://dx.doi.org/10.1177/0956462413516100
  55. P.R. Lambden, M.A. Pickett, and I.N. Clarke, "The effect of penicillin on Chlamydia trachomatis DNA replication", Microbiology, vol. 152, pp. 2573-2578, 2006. http://dx.doi.org/10.1099/mic.0.29032-0
  56. J.W. Moulder, "Interaction of chlamydiae and host cells in vitro.", Microbiological reviews, 1991. http://www.ncbi.nlm.nih.gov/pubmed/2030670
  57. L.M. Hafner, "Pathogenesis of fallopian tube damage caused by Chlamydia trachomatis infections", Contraception, vol. 92, pp. 108-115, 2015. http://dx.doi.org/10.1016/j.contraception.2015.01.004
  58. K.E. Mueller, G.V. Plano, and K.A. Fields, "New Frontiers in Type III Secretion Biology: the Chlamydia Perspective", Infection and Immunity, vol. 82, pp. 2-9, 2014. http://dx.doi.org/10.1128/IAI.00917-13
  59. A. Omsland, B.S. Sixt, M. Horn, and T. Hackstadt, "Chlamydial metabolism revisited: interspecies metabolic variability and developmental stage-specific physiologic activities", FEMS Microbiology Reviews, vol. 38, pp. 779-801, 2014. http://dx.doi.org/10.1111/1574-6976.12059
  60. R.J. Bastidas, C.A. Elwell, J.N. Engel, and R.H. Valdivia, "Chlamydial Intracellular Survival Strategies", Cold Spring Harbor Perspectives in Medicine, vol. 3, pp. a010256-a010256, 2013. http://dx.doi.org/10.1101/cshperspect.a010256
  61. C. Elwell, K. Mirrashidi, and J. Engel, "Chlamydia cell biology and pathogenesis", Nature Reviews Microbiology, vol. 14, pp. 385-400, 2016. http://dx.doi.org/10.1038/nrmicro.2016.30
  62. R. Rank, and J. Whittum‐Hudson, "Protective Immunity to Chlamydial Genital Infection: Evidence from Animal Studies", The Journal of Infectious Diseases, vol. 201, pp. 168-177, 2010. http://dx.doi.org/10.1086/652399
  63. R.G. Rank, "[7] Animal models for urogenital infections", Methods in Enzymology, pp. 83-93, 1994. http://dx.doi.org/10.1016/0076-6879(94)35133-3
  64. L. Kari, T.R. Southern, C.J. Downey, H.S. Watkins, L.B. Randall, L.D. Taylor, G.L. Sturdevant, W.M. Whitmire, and H.D. Caldwell, "Chlamydia trachomatis Polymorphic Membrane Protein D Is a Virulence Factor Involved in Early Host-Cell Interactions", Infection and Immunity, vol. 82, pp. 2756-2762, 2014. http://dx.doi.org/10.1128/IAI.01686-14
  65. K. Moelleken, and J.H. Hegemann, "The Chlamydia outer membrane protein OmcB is required for adhesion and exhibits biovar‐specific differences in glycosaminoglycan binding", Molecular Microbiology, vol. 67, pp. 403-419, 2007. http://dx.doi.org/10.1111/j.1365-2958.2007.06050.x
  66. S. Stallmann, and J.H. Hegemann, "TheChlamydia trachomatisCtad1 invasin exploits the human integrin β1 receptor for host cell entry", Cellular Microbiology, vol. 18, pp. 761-775, 2016. http://dx.doi.org/10.1111/cmi.12549
  67. J.C. Ferrell, and K.A. Fields, "A working model for the type III secretion mechanism in Chlamydia", Microbes and Infection, vol. 18, pp. 84-92, 2016. http://dx.doi.org/10.1016/j.micinf.2015.10.006
  68. E.R. Moore, and S.P. Ouellette, "Reconceptualizing the chlamydial inclusion as a pathogen-specified parasitic organelle: an expanded role for Inc proteins", Frontiers in Cellular and Infection Microbiology, vol. 4, 2014. http://dx.doi.org/10.3389/fcimb.2014.00157
  69. K. Mirrashidi, C. Elwell, E. Verschueren, J. Johnson, A. Frando, J. Von Dollen, O. Rosenberg, N. Gulbahce, G. Jang, T. Johnson, S. Jäger, A. Gopalakrishnan, J. Sherry, J. Dunn, A. Olive, B. Penn, M. Shales, J. Cox, M. Starnbach, I. Derre, R. Valdivia, N. Krogan, and J. Engel, "Global Mapping of the Inc-Human Interactome Reveals that Retromer Restricts Chlamydia Infection", Cell Host & Microbe, vol. 18, pp. 109-121, 2015. http://dx.doi.org/10.1016/j.chom.2015.06.004
  70. R.L. Bailey, A. Natividad-Sancho, A. Fowler, R.W.W. Peeling, D.C.W. Mabey, H.C. Whittle, and A.P. Jepson, "Host genetic contribution to the cellular immune response to Chlamydia trachomatis: Heritability estimate from a Gambian twin study.", Drugs of today (Barcelona, Spain : 1998), 2009. http://www.ncbi.nlm.nih.gov/pubmed/20011694
  71. R.S. Stephens, S. Kalman, C. Lammel, J. Fan, R. Marathe, L. Aravind, W. Mitchell, L. Olinger, R.L. Tatusov, Q. Zhao, E.V. Koonin, and R.W. Davis, "Genome Sequence of an Obligate Intracellular Pathogen of Humans: Chlamydia trachomatis", Science, vol. 282, pp. 754-759, 1998. http://dx.doi.org/10.1126/science.282.5389.754
  72. S.R. Harris, I.N. Clarke, H.M.B. Seth-Smith, A.W. Solomon, L.T. Cutcliffe, P. Marsh, R.J. Skilton, M.J. Holland, D. Mabey, R.W. Peeling, D.A. Lewis, B.G. Spratt, M. Unemo, K. Persson, C. Bjartling, R. Brunham, H.J.C. de Vries, S.A. Morré, A. Speksnijder, C.M. Bébéar, M. Clerc, B. de Barbeyrac, J. Parkhill, and N.R. Thomson, "Whole-genome analysis of diverse Chlamydia trachomatis strains identifies phylogenetic relationships masked by current clinical typing", Nature Genetics, vol. 44, pp. 413-419, 2012. http://dx.doi.org/10.1038/ng.2214
  73. S.J. Joseph, X. Didelot, J. Rothschild, H.J. de Vries, S.A. Morré, T.D. Read, and D. Dean, "Population Genomics of Chlamydia trachomatis: Insights on Drift, Selection, Recombination, and Population Structure", Molecular Biology and Evolution, vol. 29, pp. 3933-3946, 2012. http://dx.doi.org/10.1093/molbev/mss198
  74. S.J. Joseph, X. Didelot, K. Gandhi, D. Dean, and T.D. Read, "Interplay of recombination and selection in the genomes of Chlamydia trachomatis", Biology Direct, vol. 6, 2011. http://dx.doi.org/10.1186/1745-6150-6-28
  75. H.M. Seth-Smith, S.R. Harris, K. Persson, P. Marsh, A. Barron, A. Bignell, C. Bjartling, L. Clark, L.T. Cutcliffe, P.R. Lambden, N. Lennard, S.J. Lockey, M.A. Quail, O. Salim, R.J. Skilton, Y. Wang, M.J. Holland, J. Parkhill, N.R. Thomson, and I.N. Clarke, "Co-evolution of genomes and plasmids within Chlamydia trachomatis and the emergence in Sweden of a new variant strain", BMC Genomics, vol. 10, 2009. http://dx.doi.org/10.1186/1471-2164-10-239
  76. L. Song, J.H. Carlson, B. Zhou, K. Virtaneva, W.M. Whitmire, G.L. Sturdevant, S.F. Porcella, G. McClarty, and H.D. Caldwell, "Plasmid-mediated transformation tropism of chlamydial biovars", Pathogens and Disease, vol. 70, pp. 189-193, 2013. http://dx.doi.org/10.1111/2049-632X.12104
  77. A. Nunes, P.J. Nogueira, M.J. Borrego, and J.P. Gomes, "Adaptive Evolution of the Chlamydia trachomatis Dominant Antigen Reveals Distinct Evolutionary Scenarios for B- and T-cell Epitopes: Worldwide Survey", PLoS ONE, vol. 5, pp. e13171, 2010. http://dx.doi.org/10.1371/journal.pone.0013171
  78. R. Ferreira, M. Antelo, A. Nunes, V. Borges, V. Damiao, M. Borrego, and J. Gomes, "In silico scrutiny of genes revealing phylogenetic congruence with clinical prevalence or tropism properties of Chlamydia trachomatis strains.", G3 (Bethesda) 5(1): 9-19., 2014.
  79. P. Andersson, S.R. Harris, H.M.B.S. Smith, J. Hadfield, C. O’Neill, L.T. Cutcliffe, F.P. Douglas, L.V. Asche, J.D. Mathews, S.I. Hutton, D.S. Sarovich, S.Y.C. Tong, I.N. Clarke, N.R. Thomson, and P.M. Giffard, "Chlamydia trachomatis from Australian Aboriginal people with trachoma are polyphyletic composed of multiple distinctive lineages", Nature Communications, vol. 7, 2016. http://dx.doi.org/10.1038/ncomms10688
  80. H.D. Caldwell, H. Wood, D. Crane, R. Bailey, R.B. Jones, D. Mabey, I. Maclean, Z. Mohammed, R. Peeling, C. Roshick, J. Schachter, A.W. Solomon, W.E. Stamm, R.J. Suchland, L. Taylor, S.K. West, T.C. Quinn, R.J. Belland, and G. McClarty, "Polymorphisms in Chlamydia trachomatis tryptophan synthase genes differentiate between genital and ocular isolates.", The Journal of clinical investigation, 2003. http://www.ncbi.nlm.nih.gov/pubmed/12782678
  81. C. Fehlner-Gardiner, C. Roshick, J.H. Carlson, S. Hughes, R.J. Belland, H.D. Caldwell, and G. McClarty, "Molecular Basis Defining Human Chlamydia trachomatis Tissue Tropism", Journal of Biological Chemistry, vol. 277, pp. 26893-26903, 2002. http://dx.doi.org/10.1074/jbc.M203937200
  82. W.L. Beatty, T.A. Belanger, A.A. Desai, R.P. Morrison, and G.I. Byrne, "Tryptophan depletion as a mechanism of gamma interferon-mediated chlamydial persistence.", Infection and immunity, 1994. http://www.ncbi.nlm.nih.gov/pubmed/8063385
  83. E.V. Sokurenko, D.L. Hasty, and D.E. Dykhuizen, "Pathoadaptive mutations: gene loss and variation in bacterial pathogens", Trends in Microbiology, vol. 7, pp. 191-195, 1999. http://dx.doi.org/10.1016/s0966-842x(99)01493-6
  84. J.H. Carlson, S.F. Porcella, G. McClarty, and H.D. Caldwell, "Comparative Genomic Analysis ofChlamydia trachomatisOculotropic and Genitotropic Strains", Infection and Immunity, vol. 73, pp. 6407-6418, 2005. http://dx.doi.org/10.1128/IAI.73.10.6407-6418.2005
  85. I. Sarov, E. Geron, Y. Shemer-Avni, E. Manor, M. Zvillich, D. Wallach, E. Schmitz, and H. Holtman, "Implications for persistent chlamydial infections of phagocyte-microorganism interplay", European Journal of Clinical Microbiology & Infectious Diseases, vol. 10, pp. 119-123, 1991. http://dx.doi.org/10.1007/bf01964423
  86. E. Manor, and I. Sarov, "Fate of Chlamydia trachomatis in human monocytes and monocyte-derived macrophages.", Infection and immunity, 1986. http://www.ncbi.nlm.nih.gov/pubmed/3759241
  87. R.P. Morrison, "Differential sensitivities of Chlamydia trachomatis strains to inhibitory effects of gamma interferon.", Infection and immunity, 2000. http://www.ncbi.nlm.nih.gov/pubmed/10992517
  88. K.A. Fields, R. McCormack, L.R. de Armas, and E.R. Podack, "Perforin-2 Restricts Growth of Chlamydia trachomatis in Macrophages", Infection and Immunity, vol. 81, pp. 3045-3054, 2013. http://dx.doi.org/10.1128/IAI.00497-13
  89. V. Borges, and J.P. Gomes, "Deep comparative genomics among Chlamydia trachomatis lymphogranuloma venereum isolates highlights genes potentially involved in pathoadaptation", Infection, Genetics and Evolution, vol. 32, pp. 74-88, 2015. http://dx.doi.org/10.1016/j.meegid.2015.02.026
  90. J.P. Gomes, W.J. Bruno, A. Nunes, N. Santos, C. Florindo, M.J. Borrego, and D. Dean, "Evolution ofChlamydia trachomatisdiversity occurs by widespread interstrain recombination involving hotspots", Genome Research, vol. 17, pp. 50-60, 2006. http://dx.doi.org/10.1101/gr.5674706
  91. A. Nunes, M.J. Borrego, and J.P. Gomes, "Genomic features beyond Chlamydia trachomatis phenotypes: What do we think we know?", Infection, Genetics and Evolution, vol. 16, pp. 392-400, 2013. http://dx.doi.org/10.1016/j.meegid.2013.03.018
  92. C.M. O’Connell, R.R. Ingalls, C.W. Andrews, A.M. Scurlock, and T. Darville, "Plasmid-Deficient Chlamydia muridarum Fail to Induce Immune Pathology and Protect against Oviduct Disease", The Journal of Immunology, vol. 179, pp. 4027-4034, 2007. http://dx.doi.org/10.4049/jimmunol.179.6.4027
  93. M. Russell, T. Darville, K. Chandra-Kuntal, B. Smith, C.W. Andrews, and C.M. O'Connell, "Infectivity Acts as In Vivo Selection for Maintenance of the Chlamydial Cryptic Plasmid", Infection and Immunity, vol. 79, pp. 98-107, 2011. http://dx.doi.org/10.1128/IAI.01105-10
  94. L. Kari, W.M. Whitmire, N. Olivares-Zavaleta, M.M. Goheen, L.D. Taylor, J.H. Carlson, G.L. Sturdevant, C. Lu, L.E. Bakios, L.B. Randall, M.J. Parnell, G. Zhong, and H.D. Caldwell, "A live-attenuated chlamydial vaccine protects against trachoma in nonhuman primates", Journal of Experimental Medicine, vol. 208, pp. 2217-2223, 2011. http://dx.doi.org/10.1084/jem.20111266
  95. Y. Qu, L.C. Frazer, C.M. O'Connell, A.F. Tarantal, C.W. Andrews, S.L. O'Connor, A.N. Russell, J.E. Sullivan, T.B. Poston, A.N. Vallejo, and T. Darville, "Comparable Genital Tract Infection, Pathology, and Immunity in Rhesus Macaques Inoculated with Wild-Type or Plasmid-Deficient Chlamydia trachomatis Serovar D", Infection and Immunity, vol. 83, pp. 4056-4067, 2015. http://dx.doi.org/10.1128/IAI.00841-15
  96. A. Matsumoto, H. Izutsu, N. Miyashita, and M. Ohuchi, "Plaque formation by and plaque cloning of Chlamydia trachomatis biovar trachoma.", Journal of clinical microbiology, 1998. http://www.ncbi.nlm.nih.gov/pubmed/9738059
  97. C.M. O'Connell, and K.M. Nicks, "A plasmid-cured Chlamydia muridarum strain displays altered plaque morphology and reduced infectivity in cell culture", Microbiology, vol. 152, pp. 1601-1607, 2006. http://dx.doi.org/10.1099/mic.0.28658-0
  98. J.H. Carlson, W.M. Whitmire, D.D. Crane, L. Wicke, K. Virtaneva, D.E. Sturdevant, J.J. Kupko, S.F. Porcella, N. Martinez-Orengo, R.A. Heinzen, L. Kari, and H.D. Caldwell, "TheChlamydia trachomatisPlasmid Is a Transcriptional Regulator of Chromosomal Genes and a Virulence Factor", Infection and Immunity, vol. 76, pp. 2273-2283, 2008. http://dx.doi.org/10.1128/iai.00102-08
  99. L. Song, J.H. Carlson, W.M. Whitmire, L. Kari, K. Virtaneva, D.E. Sturdevant, H. Watkins, B. Zhou, G.L. Sturdevant, S.F. Porcella, G. McClarty, and H.D. Caldwell, "Chlamydia trachomatis Plasmid-Encoded Pgp4 Is a Transcriptional Regulator of Virulence-Associated Genes", Infection and Immunity, vol. 81, pp. 636-644, 2013. http://dx.doi.org/10.1128/IAI.01305-12
  100. C.M. O'Connell, Y.M. AbdelRahman, E. Green, H.K. Darville, K. Saira, B. Smith, T. Darville, A.M. Scurlock, C.R. Meyer, and R.J. Belland, "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", Infection and Immunity, vol. 79, pp. 1044-1056, 2011. http://dx.doi.org/10.1128/IAI.01118-10
  101. Y. Liu, Y. Huang, Z. Yang, Y. Sun, S. Gong, S. Hou, C. Chen, Z. Li, Q. Liu, Y. Wu, J. Baseman, and G. Zhong, "Plasmid-Encoded Pgp3 Is a Major Virulence Factor for Chlamydia muridarum To Induce Hydrosalpinx in Mice", Infection and Immunity, vol. 82, pp. 5327-5335, 2014. http://dx.doi.org/10.1128/IAI.02576-14
  102. Z. Li, D. Chen, Y. Zhong, S. Wang, and G. Zhong, "The Chlamydial Plasmid-Encoded Protein pgp3 Is Secreted into the Cytosol ofChlamydia-Infected Cells", Infection and Immunity, vol. 76, pp. 3415-3428, 2008. http://dx.doi.org/10.1128/IAI.01377-07
  103. S. Hou, X. Dong, Z. Yang, Z. Li, Q. Liu, and G. Zhong, "Chlamydial Plasmid-Encoded Virulence Factor Pgp3 Neutralizes the Antichlamydial Activity of Human Cathelicidin LL-37", Infection and Immunity, vol. 83, pp. 4701-4709, 2015. http://dx.doi.org/10.1128/IAI.00746-15
  104. R.J. Belland, D.E. Nelson, D. Virok, D.D. Crane, D. Hogan, D. Sturdevant, W.L. Beatty, and H.D. Caldwell, "Transcriptome analysis of chlamydial growth during IFN-γ-mediated persistence and reactivation", Proceedings of the National Academy of Sciences, vol. 100, pp. 15971-15976, 2003. http://dx.doi.org/10.1073/pnas.2535394100
  105. T. Nicholson, and R. Stephens, "Chlamydial genomic transcriptional profile for penicillin-induced persistence. Proceedings of Tenth Meeting of the International Society of Human Chlamydial Infections. ", Schachter, J, Christiansen, G et. al. (Eds), International Chlamydia Symposium, San Francisco, CA USA p 611-614., 2002.
  106. J.A. Carrasco, C. Tan, R.G. Rank, R. Hsia, and P.M. Bavoil, "Altered developmental expression of polymorphic membrane proteins in penicillin-stressed Chlamydia trachomatis", Cellular Microbiology, vol. 13, pp. 1014-1025, 2011. http://dx.doi.org/10.1111/j.1462-5822.2011.01598.x
  107. R.J. Belland, G. Zhong, D.D. Crane, D. Hogan, D. Sturdevant, J. Sharma, W.L. Beatty, and H.D. Caldwell, "Genomic transcriptional profiling of the developmental cycle of Chlamydia trachomatis", Proceedings of the National Academy of Sciences, vol. 100, pp. 8478-8483, 2003. http://dx.doi.org/10.1073/pnas.1331135100
  108. L.A. Vodstrcil, R. McIver, W.M. Huston, S.N. Tabrizi, P. Timms, and J.S. Hocking, "The Epidemiology of Chlamydia trachomatis Organism Load During Genital Infection: A Systematic Review", Journal of Infectious Diseases, vol. 211, pp. 1628-1645, 2014. http://dx.doi.org/10.1093/infdis/jiu670
  109. A.N. Russell, X. Zheng, C.M. O'Connell, B.D. Taylor, H.C. Wiesenfeld, S.L. Hillier, W. Zhong, and T. Darville, "Analysis of Factors Driving Incident and Ascending Infection and the Role of Serum Antibody inChlamydia trachomatisGenital Tract Infection", Journal of Infectious Diseases, vol. 213, pp. 523-531, 2015. http://dx.doi.org/10.1093/infdis/jiv438
  110. T.C. Yeow, W.F. Wong, N.S. Sabet, S. Sulaiman, F. Shahhosseini, G.M.Y. Tan, E. Movahed, C.Y. Looi, E.M. Shankar, R. Gupta, B.P. Arulanandam, J. Hassan, and S. Abu Bakar, "Prevalence of plasmid-bearing and plasmid-free Chlamydia trachomatis infection among women who visited obstetrics and gynecology clinics in Malaysia", BMC Microbiology, vol. 16, 2016. http://dx.doi.org/10.1186/s12866-016-0671-1
  111. M.E. Lewis, R.J. Belland, Y.M. AbdelRahman, W.L. Beatty, A.A. Aiyar, A.H. Zea, S.J. Greene, L. Marrero, L.R. Buckner, D.J. Tate, C.L. McGowin, P.A. Kozlowski, M. O'Brien, R.A. Lillis, D.H. Martin, and A.J. Quayle, "Morphologic and molecular evaluation of Chlamydia trachomatis growth in human endocervix reveals distinct growth patterns", Frontiers in Cellular and Infection Microbiology, vol. 4, 2014. http://dx.doi.org/10.3389/fcimb.2014.00071
  112. K. Kawana, A.J. Quayle, M. Ficarra, J.A. Ibana, L. Shen, Y. Kawana, H. Yang, L. Marrero, S. Yavagal, S.J. Greene, Y. Zhang, R.B. Pyles, R.S. Blumberg, and D.J. Schust, "CD1d Degradation in Chlamydia trachomatis-infected Epithelial Cells Is the Result of Both Cellular and Chlamydial Proteasomal Activity", Journal of Biological Chemistry, vol. 282, pp. 7368-7375, 2007. http://dx.doi.org/10.1074/jbc.M610754200
  113. C. Zheng XOC, U. Nagarajan, H. Wiesenfeld, S. Hillier, and T. Darville, "Identification of a blood transcriptional signature for chlamydial PID. Proceedings of Thirteenth Meeting of the International Society of Human Chlamydial Infections.", Schachter, J, Christiansen, G et. al. (Eds), International Chlamydia Symposium, San Francisco, CA USA., 2014.
  114. F. Balamuth, Z. Zhang, E. Rappaport, K. Hayes, C. Mollen, and K.E. Sullivan, "RNA Biosignatures in Adolescent Patients in a Pediatric Emergency Department With Pelvic Inflammatory Disease", Pediatric Emergency Care, vol. 31, pp. 465-472, 2015. http://dx.doi.org/10.1097/PEC.0000000000000483
  115. N.A. Wallace, K. Robinson, and D.A. Galloway, "Beta Human Papillomavirus E6 Expression Inhibits Stabilization of p53 and Increases Tolerance of Genomic Instability", Journal of Virology, vol. 88, pp. 6112-6127, 2014. http://dx.doi.org/10.1128/JVI.03808-13
  116. S.S. Grieshaber, N.A. Grieshaber, N. Miller, and T. Hackstadt, "Chlamydia trachomatis Causes Centrosomal Defects Resulting in Chromosomal Segregation Abnormalities", Traffic, vol. 7, pp. 940-949, 2006. http://dx.doi.org/10.1111/j.1600-0854.2006.00439.x
  117. K.A. Johnson, M. Tan, and C. Sütterlin, "Centrosome abnormalities during aChlamydia trachomatisinfection are caused by dysregulation of the normal duplication pathway", Cellular Microbiology, vol. 11, pp. 1064-1073, 2009. http://dx.doi.org/10.1111/j.1462-5822.2009.01307.x
  118. A.E. Knowlton, H.M. Brown, T.S. Richards, L.A. Andreolas, R.K. Patel, and S.S. Grieshaber, "Chlamydia trachomatis Infection Causes Mitotic Spindle Pole Defects Independently from its Effects on Centrosome Amplification", Traffic, vol. 12, pp. 854-866, 2011. http://dx.doi.org/10.1111/j.1600-0854.2011.01204.x
  119. J. Mital, N.J. Miller, E.R. Fischer, and T. Hackstadt, "Specific chlamydial inclusion membrane proteins associate with active Src family kinases in microdomains that interact with the host microtubule network", Cellular Microbiology, vol. 12, pp. 1235-1249, 2010. http://dx.doi.org/10.1111/j.1462-5822.2010.01465.x
  120. T.S. Richards, A.E. Knowlton, and S.S. Grieshaber, "Chlamydia trachomatis homotypic inclusion fusion is promoted by host microtubule trafficking", BMC Microbiology, vol. 13, 2013. http://dx.doi.org/10.1186/1471-2180-13-185
  121. M. Dumoux, A. Menny, D. Delacour, and R.D. Hayward, "A Chlamydia effector recruits CEP170 to reprogram host microtubule organization", Journal of Cell Science, 2015. http://dx.doi.org/10.1242/jcs.169318
  122. E.A. Nigg, "Centrosome aberrations: cause or consequence of cancer progression?", Nature Reviews Cancer, vol. 2, pp. 815-825, 2002. http://dx.doi.org/10.1038/nrc924
  123. A.F. Bolgioni, and N.J. Ganem, "The interplay between centrosomes and the Hippo tumor suppressor pathway", Chromosome Research, vol. 24, pp. 93-104, 2015. http://dx.doi.org/10.1007/s10577-015-9502-8
  124. C. Chumduri, R. Gurumurthy, P. Zadora, Y. Mi, and T. Meyer, "Chlamydia Infection Promotes Host DNA Damage and Proliferation but Impairs the DNA Damage Response", Cell Host & Microbe, vol. 13, pp. 746-758, 2013. http://dx.doi.org/10.1016/j.chom.2013.05.010
  125. E. González, M. Rother, M.C. Kerr, M.A. Al-Zeer, M. Abu-Lubad, M. Kessler, V. Brinkmann, A. Loewer, and T.F. Meyer, "Chlamydia infection depends on a functional MDM2-p53 axis", Nature Communications, vol. 5, 2014. http://dx.doi.org/10.1038/ncomms6201
  126. C. Siegl, B. Prusty, K. Karunakaran, J. Wischhusen, and T. Rudel, "Tumor Suppressor p53 Alters Host Cell Metabolism to Limit Chlamydia trachomatis Infection", Cell Reports, vol. 9, pp. 918-929, 2014. http://dx.doi.org/10.1016/j.celrep.2014.10.004
  127. A.E. Knowlton, L.J. Fowler, R.K. Patel, S.M. Wallet, and S.S. Grieshaber, "Chlamydia Induces Anchorage Independence in 3T3 Cells and Detrimental Cytological Defects in an Infection Model", PLoS ONE, vol. 8, pp. e54022, 2013. http://dx.doi.org/10.1371/journal.pone.0054022
  128. M. Merbah, A. Introini, W. Fitzgerald, J. Grivel, A. Lisco, C. Vanpouille, and L. Margolis, "Cervico‐Vaginal Tissue Ex Vivo as a Model to Study Early Events in HIV‐1 Infection", American Journal of Reproductive Immunology, vol. 65, pp. 268-278, 2011. http://dx.doi.org/10.1111/j.1600-0897.2010.00967.x
  129. L.R. Buckner, A.M. Amedee, H.L. Albritton, P.A. Kozlowski, N. Lacour, C.L. McGowin, D.J. Schust, and A.J. Quayle, "Chlamydia trachomatis Infection of Endocervical Epithelial Cells Enhances Early HIV Transmission Events", PLOS ONE, vol. 11, pp. e0146663, 2016. http://dx.doi.org/10.1371/journal.pone.0146663
  130. C. Gaydos, K. Crotchfelt, M. Howell, S. Kralian, P. Hauptman, and T. Quinn, "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", The Journal of Infectious Diseases, vol. 177, pp. 417-424, 1998. http://dx.doi.org/10.1086/514207
  131. W.M. Geisler, "Diagnosis and Management of UncomplicatedChlamydia trachomatisInfections in Adolescents and Adults: Summary of Evidence Reviewed for the 2015 Centers for Disease Control and Prevention Sexually Transmitted Diseases Treatment Guidelines", Clinical Infectious Diseases, vol. 61, pp. S774-S784, 2015. http://dx.doi.org/10.1093/cid/civ694
  132. K.M. Sandoz, and D.D. Rockey, "Antibiotic resistance in Chlamydiae", Future Microbiology, vol. 5, pp. 1427-1442, 2010. http://dx.doi.org/10.2217/fmb.10.96
  133. R. Binet, and A.T. Maurelli, "Frequency of Development and Associated Physiological Cost of Azithromycin Resistance in Chlamydia psittaci 6BC and C. trachomatis L2", Antimicrobial Agents and Chemotherapy, vol. 51, pp. 4267-4275, 2007. http://dx.doi.org/10.1128/AAC.00962-07
  134. R. Binet, and A.T. Maurelli, "Fitness Cost Due to Mutations in the 16S rRNA Associated with Spectinomycin Resistance in Chlamydia psittaci 6BC", Antimicrobial Agents and Chemotherapy, vol. 49, pp. 4455-4464, 2005. http://dx.doi.org/10.1128/AAC.49.11.4455-4464.2005
  135. M.R. Golden, W.L. Whittington, H.H. Handsfield, J.P. Hughes, W.E. Stamm, M. Hogben, A. Clark, C. Malinski, J.R. Helmers, K.K. Thomas, and K.K. Holmes, "Effect of Expedited Treatment of Sex Partners on Recurrent or Persistent Gonorrhea or Chlamydial Infection", New England Journal of Medicine, vol. 352, pp. 676-685, 2005. http://dx.doi.org/10.1056/NEJMoa041681
  136. R.G. Rank, and L. Yeruva, "Hidden in Plain Sight: Chlamydial Gastrointestinal Infection and Its Relevance to Persistence in Human Genital Infection", Infection and Immunity, vol. 82, pp. 1362-1371, 2014. http://dx.doi.org/10.1128/IAI.01244-13
  137. L. Yeruva, N. Spencer, A.K. Bowlin, Y. Wang, and R.G. Rank, "Chlamydial infection of the gastrointestinal tract: a reservoir for persistent infection", Pathogens and Disease, vol. 68, pp. 88-95, 2013. http://dx.doi.org/10.1111/2049-632X.12052
  138. J. Dai, T. Zhang, L. Wang, L. Shao, C. Zhu, Y. Zhang, C. Failor, R. Schenken, J. Baseman, C. He, and G. Zhong, "Intravenous Inoculation with Chlamydia muridarum Leads to a Long-Lasting Infection Restricted to the Gastrointestinal Tract", Infection and Immunity, vol. 84, pp. 2382-2388, 2016. http://dx.doi.org/10.1128/IAI.00432-16
  139. L. Yeruva, S. Melnyk, N. Spencer, A. Bowlin, and R.G. Rank, "Differential Susceptibilities to Azithromycin Treatment of Chlamydial Infection in the Gastrointestinal Tract and Cervix", Antimicrobial Agents and Chemotherapy, vol. 57, pp. 6290-6294, 2013. http://dx.doi.org/10.1128/AAC.01405-13
  140. L. Wang, Q. Zhang, T. Zhang, Y. Zhang, C. Zhu, X. Sun, N. Zhang, M. Xue, and G. Zhong, "The Chlamydia muridarum Organisms Fail to Auto-Inoculate the Mouse Genital Tract after Colonization in the Gastrointestinal Tract for 70 days", PLOS ONE, vol. 11, pp. e0155880, 2016. http://dx.doi.org/10.1371/journal.pone.0155880
  141. N. Borel, C. Leonard, J. Slade, and R.V. Schoborg, "Chlamydial Antibiotic Resistance and Treatment Failure in Veterinary and Human Medicine", Current Clinical Microbiology Reports, vol. 3, pp. 10-18, 2016. http://dx.doi.org/10.1007/s40588-016-0028-4
  142. R.M. Leonhardt, S. Lee, P.B. Kavathas, and P. Cresswell, "Severe Tryptophan Starvation Blocks Onset of Conventional Persistence and Reduces Reactivation of Chlamydia trachomatis", Infection and Immunity, vol. 75, pp. 5105-5117, 2007. http://dx.doi.org/10.1128/IAI.00668-07
  143. C.A. Leonard, R.V. Schoborg, and N. Borel, "Damage/Danger Associated Molecular Patterns (DAMPs) Modulate Chlamydia pecorum and C. trachomatis Serovar E Inclusion Development In Vitro", PLOS ONE, vol. 10, pp. e0134943, 2015. http://dx.doi.org/10.1371/journal.pone.0134943
  144. N.V. Guseva, S.T. Knight, J.D. Whittimore, and P.B. Wyrick, "Primary Cultures of Female Swine Genital Epithelial Cells In Vitro: a New Approach for the Study of Hormonal Modulation of Chlamydia Infection", Infection and Immunity, vol. 71, pp. 4700-4710, 2003. http://dx.doi.org/10.1128/IAI.71.8.4700-4710.2003
  145. J. Kintner, R.V. Schoborg, P.B. Wyrick, and J.V. Hall, "Progesterone antagonizes the positive influence of estrogen on Chlamydia trachomatis serovar E in an Ishikawa/SHT-290 co-culture model", Pathogens and Disease, vol. 73, 2015. http://dx.doi.org/10.1093/femspd/ftv015
  146. J.V. Hall, M. Schell, S. Dessus-Babus, C.G. Moore, J.D. Whittimore, M. Sal, B.D. Dill, and P.B. Wyrick, "The multifaceted role of oestrogen in enhancing Chlamydia trachomatis infection in polarized human endometrial epithelial cells", Cellular Microbiology, vol. 13, pp. 1183-1199, 2011. http://dx.doi.org/10.1111/j.1462-5822.2011.01608.x
  147. S. Deka, J. Vanover, S. Dessus-Babus, J. Whittimore, M.K. Howett, P.B. Wyrick, and R.V. Schoborg, "Chlamydia trachomatis enters a viable but non-cultivable (persistent) state within herpes simplex virus type 2 (HSV-2) co-infected host cells", Cellular Microbiology, vol. 8, pp. 149-162, 2006. http://dx.doi.org/10.1111/j.1462-5822.2005.00608.x
  148. J.D. Romano, C. de Beaumont, J.A. Carrasco, K. Ehrenman, P.M. Bavoil, and I. Coppens, "A novel co-infection model withToxoplasmaandChlamydia trachomatishighlights the importance of host cell manipulation for nutrient scavenging", Cellular Microbiology, vol. 15, pp. 619-646, 2012. http://dx.doi.org/10.1111/cmi.12060
  149. D. Kell, M. Potgieter, and E. Pretorius, "Individuality, phenotypic differentiation, dormancy and ‘persistence’ in culturable bacterial systems: commonalities shared by environmental, laboratory, and clinical microbiology", F1000Research, vol. 4, pp. 179, 2015. http://dx.doi.org/10.12688/f1000research.6709.2
  150. C.I. Kint, N. Verstraeten, M. Fauvart, and J. Michiels, "New-found fundamentals of bacterial persistence", Trends in Microbiology, vol. 20, pp. 577-585, 2012. http://dx.doi.org/10.1016/j.tim.2012.08.009
  151. M. Fauvart, V.N. De Groote, and J. Michiels, "Role of persister cells in chronic infections: clinical relevance and perspectives on anti-persister therapies", Journal of Medical Microbiology, vol. 60, pp. 699-709, 2011. http://dx.doi.org/10.1099/jmm.0.030932-0
  152. P.B. Wyrick, "Pre-exposure of infected human endometrial epithelial cells to penicillin in vitro renders Chlamydia trachomatis refractory to azithromycin", Journal of Antimicrobial Chemotherapy, vol. 54, pp. 79-85, 2004. http://dx.doi.org/10.1093/jac/dkh283
  153. N. Reveneau, D.D. Crane, E. Fischer, and H.D. Caldwell, "Bactericidal Activity of First-Choice Antibiotics against Gamma Interferon-Induced Persistent Infection of Human Epithelial Cells by Chlamydia trachomatis", Antimicrobial Agents and Chemotherapy, vol. 49, pp. 1787-1793, 2005. http://dx.doi.org/10.1128/AAC.49.5.1787-1793.2005
  154. R. Phillips-Campbell, J. Kintner, and R.V. Schoborg, "Induction of the Chlamydia muridarum Stress/Persistence Response Increases Azithromycin Treatment Failure in a Murine Model of Infection", Antimicrobial Agents and Chemotherapy, vol. 58, pp. 1782-1784, 2014. http://dx.doi.org/10.1128/AAC.02097-13
  155. M. Unemo, K. Endre, and H. Moi, "Five-day Azithromycin Treatment Regimen for Mycoplasma genitalium Infection Also Effectively Eradicates Chlamydia trachomatis", Acta Dermato Venereologica, vol. 95, pp. 730-732, 2015. http://dx.doi.org/10.2340/00015555-2108
  156. E. Lanjouw, S. Ouburg, H. de Vries, A. Stary, K. Radcliffe, and M. Unemo, "2015 European guideline on the management of Chlamydia trachomatis infections", International Journal of STD & AIDS, vol. 27, pp. 333-348, 2015. http://dx.doi.org/10.1177/0956462415618837
  157. E. Lanjouw, S. Ouburg, H. de Vries, A. Stary, K. Radcliffe, and M. Unemo, "Background review for the ‘2015 European guideline on the management ofChlamydia trachomatisinfections’", International Journal of STD & AIDS, pp. 095646241561883, 2015. http://dx.doi.org/10.1177/0956462415618838
  158. J.R. Schwebke, A. Rompalo, S. Taylor, A.C. Sena, D.H. Martin, L.M. Lopez, S. Lensing, and J.Y. Lee, "Re-Evaluating the Treatment of Nongonococcal Urethritis: Emphasizing Emerging Pathogens-A Randomized Clinical Trial", Clinical Infectious Diseases, vol. 52, pp. 163-170, 2010. http://dx.doi.org/10.1093/cid/ciq074
  159. C. Bjartling, S. Osser, and K. Persson, "Mycoplasma genitalium in cervicitis and pelvic inflammatory disease among women at a gynecologic outpatient service", American Journal of Obstetrics and Gynecology, vol. 206, pp. 476.e1-476.e8, 2012. http://dx.doi.org/10.1016/j.ajog.2012.02.036
  160. C.L. McGowin, and C. Anderson-Smits, "Mycoplasma genitalium: An Emerging Cause of Sexually Transmitted Disease in Women", PLoS Pathogens, vol. 7, pp. e1001324, 2011. http://dx.doi.org/10.1371/journal.ppat.1001324
  161. M. Bissessor, S.N. Tabrizi, J. Twin, H. Abdo, C.K. Fairley, M.Y. Chen, L.A. Vodstrcil, J.S. Jensen, J.S. Hocking, S.M. Garland, and C.S. Bradshaw, "Macrolide Resistance and Azithromycin Failure in a Mycoplasma genitalium-Infected Cohort and Response of Azithromycin Failures to Alternative Antibiotic Regimens", Clinical Infectious Diseases, vol. 60, pp. 1228-1236, 2014. http://dx.doi.org/10.1093/cid/ciu1162
  162. M.J. Pond, A.V. Nori, A.A. Witney, R.C. Lopeman, P.D. Butcher, and S.T. Sadiq, "High Prevalence of Antibiotic-Resistant Mycoplasma genitalium in Nongonococcal Urethritis: The Need for Routine Testing and the Inadequacy of Current Treatment Options", Clinical Infectious Diseases, vol. 58, pp. 631-637, 2013. http://dx.doi.org/10.1093/cid/cit752
  163. K.A. Tagg, N.J. Jeoffreys, D.L. Couldwell, J.A. Donald, and G.L. Gilbert, "Fluoroquinolone and Macrolide Resistance-Associated Mutations in Mycoplasma genitalium", Journal of Clinical Microbiology, vol. 51, pp. 2245-2249, 2013. http://dx.doi.org/10.1128/JCM.00495-13
  164. P. Horner, K. Blee, and E. Adams, "Time to manage Mycoplasma genitalium as an STI", Current Opinion in Infectious Diseases, vol. 27, pp. 68-74, 2014. http://dx.doi.org/10.1097/QCO.0000000000000030
  165. P. Oakeshott, S. Kerry, A. Aghaizu, H. Atherton, S. Hay, D. Taylor-Robinson, I. Simms, and P. Hay, "Randomised controlled trial of screening for Chlamydia trachomatis to prevent pelvic inflammatory disease: the POPI (prevention of pelvic infection) trial", BMJ, vol. 340, pp. c1642-c1642, 2010. http://dx.doi.org/10.1136/bmj.c1642
  166. S.L. Gottlieb, F. Xu, and R.C. Brunham, "Screening and Treating Chlamydia trachomatis Genital Infection to Prevent Pelvic Inflammatory Disease", Sexually Transmitted Diseases, vol. 40, pp. 97-102, 2013. http://dx.doi.org/10.1097/OLQ.0b013e31827bd637
  167. A. Aghaizu, E.J. Adams, K. Turner, S. Kerry, P. Hay, I. Simms, and P. Oakeshott, "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", Sexually Transmitted Infections, vol. 87, pp. 312-317, 2011. http://dx.doi.org/10.1136/sti.2010.048694
  168. R.C. Brunham, and M.L. Rekart, "The Arrested Immunity Hypothesis and the Epidemiology of Chlamydia Control", Sexually Transmitted Diseases, vol. 35, pp. 53-54, 2008. http://dx.doi.org/10.1097/OLQ.0b013e31815e41a3
  169. J.N. Arno, B.P. Katz, R. McBride, G.A. Carty, B.E. Batteiger, V.A. Caine, and R.B. Jones, "Age and clinical immunity to infections with Chlamydia trachomatis.", Sexually transmitted diseases, 1994. http://www.ncbi.nlm.nih.gov/pubmed/8140489
  170. B. Batteiger, F. Xu, R. Johnson, and M. Rekart, "Protective Immunity toChlamydia trachomatisGenital Infection: Evidence from Human Studies", The Journal of Infectious Diseases, vol. 201, pp. 178-189, 2010. http://dx.doi.org/10.1086/652400
  171. K.H. Ramsey, and R.G. Rank, "Resolution of chlamydial genital infection with antigen-specific T-lymphocyte lines.", Infection and immunity, 1991. http://www.ncbi.nlm.nih.gov/pubmed/1705244
  172. M.M. Riley, M.A. Zurenski, L.C. Frazer, C.M. O'Connell, C.W. Andrews, M. Mintus, and T. Darville, "The Recall Response Induced by Genital Challenge with Chlamydia muridarum Protects the Oviduct from Pathology but Not from Reinfection", Infection and Immunity, vol. 80, pp. 2194-2203, 2012. http://dx.doi.org/10.1128/IAI.00169-12
  173. A.K. Murthy, J.P. Chambers, P.A. Meier, G. Zhong, and B.P. Arulanandam, "Intranasal Vaccination with a Secreted Chlamydial Protein Enhances Resolution of GenitalChlamydia muridarumInfection, Protects against Oviduct Pathology, and Is Highly Dependent upon Endogenous Gamma Interferon Production", Infection and Immunity, vol. 75, pp. 666-676, 2007. http://dx.doi.org/10.1128/IAI.01280-06
  174. Y. Cong, M. Jupelli, M.N. Guentzel, G. Zhong, A.K. Murthy, and B.P. Arulanandam, "Intranasal immunization with chlamydial protease-like activity factor and CpG deoxynucleotides enhances protective immunity against genital Chlamydia muridarum infection", Vaccine, vol. 25, pp. 3773-3780, 2007. http://dx.doi.org/10.1016/j.vaccine.2007.02.010
  175. H. Yu, X. Jiang, C. Shen, K.P. Karunakaran, J. Jiang, N.L. Rosin, and R.C. Brunham, "Chlamydia muridarumT-Cell Antigens Formulated with the Adjuvant DDA/TDB Induce Immunity against Infection That Correlates with a High Frequency of Gamma Interferon (IFN-γ)/Tumor Necrosis Factor Alpha and IFN-γ/Interleukin-17 Double-Positive CD4+T Cells", Infection and Immunity, vol. 78, pp. 2272-2282, 2010. http://dx.doi.org/10.1128/IAI.01374-09
  176. A. Olsen, F. Follmann, K. Jensen, P. Højrup, R. Leah, H. Sørensen, S. Hoffmann, P. Andersen, and M. Theisen, "Identification of CT521 as a Frequent Target of Th1 Cells in Patients with UrogenitalChlamydia trachomatisInfection", The Journal of Infectious Diseases, vol. 194, pp. 1258-1266, 2006. http://dx.doi.org/10.1086/508203
  177. A.L. Gervassi, K.H. Grabstein, P. Probst, B. Hess, M.R. Alderson, and S.P. Fling, "Human CD8+ T Cells Recognize the 60-kDa Cysteine-Rich Outer Membrane Protein fromChlamydia trachomatis", The Journal of Immunology, vol. 173, pp. 6905-6913, 2004. http://dx.doi.org/10.4049/jimmunol.173.11.6905
  178. C. Cohen, K. Koochesfahani, A. Meier, C. Shen, K. Karunakaran, B. Ondondo, T. Kinyari, N. Mugo, R. Nguti, and R. Brunham, "Immunoepidemiologic Profile ofChlamydia trachomatisInfection: Importance of Heat‐Shock Protein 60 and Interferon‐γ", The Journal of Infectious Diseases, vol. 192, pp. 591-599, 2005. http://dx.doi.org/10.1086/432070
  179. S.G. Morrison, and R.P. Morrison, "A Predominant Role for Antibody in Acquired Immunity to Chlamydial Genital Tract Reinfection", The Journal of Immunology, vol. 175, pp. 7536-7542, 2005. http://dx.doi.org/10.4049/jimmunol.175.11.7536
  180. L.J. Brady, "Antibody-Mediated Immunomodulation: a Strategy To Improve Host Responses against Microbial Antigens", Infection and Immunity, vol. 73, pp. 671-678, 2005. http://dx.doi.org/10.1128/IAI.73.2.671-678.2005
  181. R. Punnonen, P. Terho, V. Nikkanen, and O. Meurman, "Chlamydial Serology in Infertile women by Immunofluorescence", Fertility and Sterility, vol. 31, pp. 656-659, 1979. http://dx.doi.org/10.1016/S0015-0282(16)44056-2
  182. R.C. Brunham, "Immunity to Chlamydia trachomatis", Journal of Infectious Diseases, vol. 207, pp. 1796-1797, 2013. http://dx.doi.org/10.1093/infdis/jit095
  183. T.B. Poston, and T. Darville, "Chlamydia trachomatis: Protective Adaptive Responses and Prospects for a Vaccine", Biology of Chlamydia, pp. 217-237, 2016. http://dx.doi.org/10.1007/82_2016_6
  184. E. Lorenzen, F. Follmann, S. Bøje, K. Erneholm, A.W. Olsen, J.S. Agerholm, G. Jungersen, and P. Andersen, "Intramuscular Priming and Intranasal Boosting Induce Strong Genital Immunity Through Secretory IgA in Minipigs Infected with Chlamydia trachomatis", Frontiers in Immunology, vol. 6, 2015. http://dx.doi.org/10.3389/fimmu.2015.00628
  185. A.W. Olsen, F. Follmann, K. Erneholm, I. Rosenkrands, and P. Andersen, "Protection AgainstChlamydia trachomatisInfection and Upper Genital Tract Pathological Changes by Vaccine-Promoted Neutralizing Antibodies Directed to the VD4 of the Major Outer Membrane Protein", Journal of Infectious Diseases, vol. 212, pp. 978-989, 2015. http://dx.doi.org/10.1093/infdis/jiv137
  186. S. Bøje, A.W. Olsen, K. Erneholm, J.S. Agerholm, G. Jungersen, P. Andersen, and F. Follmann, "A multi‐subunit Chlamydia vaccine inducing neutralizing antibodies and strong IFN‐γ+ CMI responses protects against a genital infection in minipigs", Immunology & Cell Biology, vol. 94, pp. 185-195, 2015. http://dx.doi.org/10.1038/icb.2015.79
  187. . NanoBio Corporation, "NanoBio’s chlamydia vaccine improves clearance of bacteria and prevents Pelvic Inflammatory Disease in mice.", Available at: http://www.nanobio.com/chlamydia-vaccine-update/. Accessed: 26.08.2016, 2015.
  188. K.P. Karunakaran, H. Yu, X. Jiang, Q. Chan, K. Moon, L.J. Foster, and R.C. Brunham, "Outer membrane proteins preferentially load MHC class II peptides: Implications for a Chlamydia trachomatis T cell vaccine", Vaccine, vol. 33, pp. 2159-2166, 2015. http://dx.doi.org/10.1016/j.vaccine.2015.02.055
  189. G. Stary, A. Olive, A.F. Radovic-Moreno, D. Gondek, D. Alvarez, P.A. Basto, M. Perro, V.D. Vrbanac, A.M. Tager, J. Shi, J.A. Yethon, O.C. Farokhzad, R. Langer, M.N. Starnbach, and U.H. von Andrian, "A mucosal vaccine against Chlamydia trachomatis generates two waves of protective memory T cells", Science, vol. 348, 2015. http://dx.doi.org/10.1126/science.aaa8205

ACKNOWLEDGMENTS

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).

COPYRIGHT

© 2016

Creative Commons License
Chlamydia trachomatis Genital Infections by O'Connell and Ferone is licensed under a Creative Commons Attribution 4.0 International License.

By continuing to use the site, you agree to the use of cookies. more information

The cookie settings on this website are set to "allow cookies" to give you the best browsing experience possible. If you continue to use this website without changing your cookie settings or you click "Accept" below then you are consenting to this. Please refer to our "privacy statement" and our "terms of use" for further information.

Close