Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Infection

DOI 10.1007/s15010-015-0860-0

REVIEW

Trichomonas vaginalis: pathogenicity and potential role in human


reproductive failure
Ewelina Mielczarek1 · Joanna Blaszkowska2 

Received: 27 September 2015 / Accepted: 27 October 2015


© Springer-Verlag Berlin Heidelberg 2015

Abstract  Keywords  Trichomoniasis · Trichomonas vaginalis ·


Purpose  Trichomonas vaginalis, which colonizes the Infertility · Pathogenesis
genitourinary tract of men and women, is a sexually trans-
mitted parasite causing symptomatic or asymptomatic Abbreviations
trichomoniasis. The host–parasite relationship is very com- AP Adhesion proteins
plex, and clinical symptoms cannot likely be attributed to a CDC Centers for Disease Control and Prevention
single pathogenic effect. Among the many factors respon- CDF Cell-detaching factor
sible for interactions between T. vaginalis and host tissues, CN Cervical neoplasia
contact-dependent and contact-independent mechanisms CP Cysteine peptidases
are important in pathogenicity, as is the immune response. FDA Food and Drug Administration
Methods  This review focuses on the potential virulence prop- GFB Transforming growth factor beta
erties of T. vaginalis and its role in female and male infertility. HIV Human immunodeficiency virus
Results  It highlights the association between T. vaginalis HPV Human papillomavirus
infection and serious adverse health consequences experi- HSV Herpes simplex virus
enced by women, including infertility, preterm birth and IgA Immunoglobulin A
low-birth-weight infants. Long-term clinical observations IgG Immunoglobulin G
and results of in vitro experimental studies indicate that in IL Interleukin
men, trichomoniasis has been also associated with infertil- ITS Internal transcribed spacer
ity through inflammatory damage to the genitourinary tract NAATs Nucleic acid amplification tests
or interference with sperm function. LPG Lipophosphoglycan
Conclusion  These results contribute significantly to PID Pelvic inflammatory disease
improving our knowledge of the role of parasitic viru- PCR Polymerase chain reaction
lence factors in the development of infection and its role in STD Sexually transmitted disease
human infertility. STI Sexually transmitted infection
TVVs  Trichomonas vaginalis viruses
* Joanna Blaszkowska VECs Vaginal epithelial cells
joanna.blaszkowska@umed.lodz.pl
Ewelina Mielczarek
ewelina_mielczarek@outlook.com Introduction
1
Department of Obstetrics and Gynaecology, The Ludwig
The flagellated protozoan parasite Trichomonas vaginalis,
Rydygier Medical Center in Lodz, 13 Sterlinga Street,
90‑217 Lodz, Poland occurring in the human urogenital tract, is the etiological
2 agent of trichomoniasis, the most common worldwide non-
Department of Diagnostics and Treatment of Parasitic
Diseases and Mycoses, Medical University of Lodz, Pl. viral sexually transmitted infection (STD) [1]. Trichomonal
Hallera 1, 90‑647 Lodz, Poland infection has been noted in every continent and climate [1,

13
E. Mielczarek, J. Blaszkowska

2] and with no seasonal variability. The outcome of infec- environments [17, 18]. Outside the host, the parasite can
tion with T. vaginalis varies depending on many factors, survive for 6–24 h in urine, semen and swimming pool
such as the genetic variability of the isolates and the host water [19], but only up to 30 min when exposed to air. T.
immune response [3, 4]. The incidence of trichomoniasis vaginalis can grow over a wide range of pH values with an
rate depends on many factors including age, sexual activ- optimum level between pH 6 and 6.3 [20].
ity, number of sexual partners, the presence of other STDs, Trichomonas vaginalis exists as a trophozoite with-
sexual customs, phase of the menstrual cycle, techniques of out the true cystic stages. The two described forms of
examination, specimen collection and laboratory technique. trophozoite are characterized by different morphologies:
Together with Chagas disease, cysticercosis, toxocariasis free-swimming, flagellated or amoeboid cells. It has been
and toxoplasmosis, trichomoniasis belongs to the group of recently shown that T. vaginalis occurs in another cellular
neglected parasitic infections (NPIs), which has been tar- form: a pseudocyst described as a non-motile stage, which
geted by the CDC as a priority for public health action [5]. is rounded without a true cyst wall, and an external fla-
The public health importance of the problem is under- gella [21]. It has been suggested that this form is created
lined by the fact that T. vaginalis infection is prevalent in in response to unfavorable culture conditions [20]. Addi-
women of reproductive age, and is associated with serious tionally, T. vaginalis pseudocysts have been isolated from
adverse reproductive outcomes [6, 7]. This infection may cervical neoplasia (CN) patients. The study conducted
also increase the risk of human immunodeficiency virus by Afzan and Suresh [11] confirms that the pseudocystic
(HIV) transmission and other STD infections which are stage from CN has unique biochemical, surface, and ultra-
observed significantly more frequently in women [8, 9]. structural properties compared to the other forms. Moreo-
Infection may cause chronic inflammation of the genitouri- ver, Hussein and Atwa [22] report that the pseudocysts
nary tract, which may even lead to infertility. Several stud- were able to cause infection in mice, with trophozoites of
ies report that patients with T. vaginalis infection display T. vaginalis being detected in the urine of experimentally
deleterious outcomes in reproduction. Additionally, the infected animals on day 4 after inoculation. They postulate
incidence and severity of cervical dysplasia and prostate that pseudocysts play a significant role in the life cycle of
cancer is also associated with trichomoniasis [10, 11]. T. vaginalis and the pathophysiology of the infection.
Very probably, the true prevalence of T. vaginalis is
much higher due to its frequent asymptomatic course. This
parasite is not routinely screened in asymptomatic patients Epidemiology and modes of transmission
and the infection can persist for 3–12 months in the geni-
tal tract [4]. Moreover, the detectability of trichomonads in The available WHO estimates suggest that the global inci-
examined biological material substantially depends on the dence of T. vaginalis infection increased between 1995 and
choice of applied diagnostic method [12, 13]. 2008 (Table 1) [1, 2]. Its prevalence has increased from
152.9 million (22.2 %) since 2005 to 187.1 million concur-
rent infections at any point in 2008. However, these esti-
Brief characteristics of T. vaginalis mates may be low, since they are based on wet mounts that
are not as sensitive as new molecular diagnostic techniques
Trichomonas vaginalis is an amitochondrial, microaero- [13].
tolerant flagellate which has various shapes and sizes, but Trichomonas vaginalis is a sexually transmitted patho-
on average measures about 9 × 7 µm. It has five flagella; gen with the highest annual incidence of all curable and
four are present anteriorly and the other flagellum is incor- non-viral STIs. At the 2008, the prevalence of T. vaginalis
porated within the undulating membrane. It has unique was higher than Chlamydia trachomatis (100.4 million),
energy-producing double membrane organelles known as Neisseria gonorrhoeae (36.4) and Treponema pallidum
hydrogenosomes. It reproduces every 8–12 h by longitu- (36.4) infections combined [2]. In addition, trichomoniasis
dinal binary fission. T. vaginalis is a obligate parasite that is the most common parasitic infection in the US, affect-
phagocytoses bacteria, vaginal epithelial cells (VECs), ing an estimated 3.7 million persons nationwide, including
spermatozoids and erythrocytes [3, 14–16]. This unicellular 2.3 million women and 1.4 million men [5]. There are large
parasite (strain G3) has a genome of over 176 megabases differences in the prevalence of Trichomonas infections
with 60,000 genes, the largest number of genes ever identi- between different WHO regions populations (Table 2).
fied in protozoans. It also has a massive proteome, express- Additionally, epidemiological studies of this protozoan in
ing ∼4000 genes. Until now, 411 proteins have been identi- local communities have revealed a wide range of preva-
fied, many of which are known to be membrane proteins lence. In the US, the prevalence among women ranges from
with possible roles in the pathogenesis of T. vaginalis or 2.8 % in adolescents nationwide to up 50 % among Afri-
which may help parasite to adapt and survive in diverse can–American women in urban communities [23].

13
Trichomonas vaginalis: pathogenicity and potential role in human reproductive failure

Table 1  Estimates of the global Year Incidencea [millions] Prevalenceb [millions]


incidence and prevalence of
T. vaginalis infection among Female Male Total Female Male Total
adults aged 15–49-year
1995 84.57 82.55 167.12 Not reported
1999 87.68 85.78 173.46 Not reported
2005 105.63 142.85 248.48 135.52 17.38 152.90
2008 132.20 144.20 276.40 169.50 17.60 187.10

Estimates from the World Health Organization [1, 2]


a
  The number of new cases of T. vaginalis infection occurring per year
b
  The number of cases of T. vaginalis infection at any point in time of year

Table 2  Regional prevalence estimates for T. vaginalis infection for and respiratory tracts have been reported in newborns of
2008 infected mothers [28].
WHO region Prevalence [millions] Non-sexual transmission is rare, but has been observed
Female Male Total
in cases involving contaminated showers, moist wash-
clothes, toilet seats, or specula [29–31]. Crucitti et al. [32]
African Region 38.9 3.9 42.8 found a high prevalence of trichomoniasis in adolescent
Region of the Americas 52.7 5.2 57.9 girls attending school in Ndola, Zambia following non-sex-
South-East Asia Region 25.7 3.0 28.7 ual transmission of trichomoniasis via shared bathing water
European Region 13.0 1.3 14.3 and inconsistent use of soap. Adu-Sarkodie [30] reports
Eastern Mediterranean Region 12.0 1.3 13.3 the transmission of T. vaginalis from mother to children
Western Pacific Region 27.2 2.9 30.1 through the sharing of bathing implements in Ghana. Addi-
Global total 169.5 17.6 187.1 tionally, Charles [29] describes an epidemic of trichomo-
niasis in young girls in a rural area in India spread through
Estimates from the World Health Organization [1]
contaminated water.

Biological differences between the sexes contribute to


these striking differences in the incidence and prevalence Virulence factors of T. vaginalis
of T. vaginalis infection between men and women [4]. For
men it was calculated as one tenth of the prevalence in The host–parasite relationship is very complex [33, 34]. A
women [1, 2]. An individual with no symptoms of tricho- successful invasion of T. vaginalis requires the existence of
moniasis is classified as a chronic asymptomatic carrier, several virulence mechanisms such as the transformation
and these constitute 70–100 % of male population and in of the trophozoite into an ameboid form, cell-to-cell adhe-
35–85 % of the female population with confirmed T. vagi- sions [20, 35–37] hemolytic activity [20], trichomonad pro-
nalis infection [3]. teinase activity [38], contact-independent mechanisms and
Trichomonas vaginalis is commonly spread through cell-detaching factor [36, 39] phagocytosis [3, 36] and host
sexual contact with the vaginal or urethral discharges of immune system evasion [3, 20, 36, 40].
infected persons [20]. Urethral infection by T. vaginalis is
present in at least 80 % of infected women and over 73 % The morphological change of the trophozoite
of male partners of women diagnosed with vaginal tricho-
moniasis [24]. Additionally, transmission of protozoa is One of the determinants of pathogenicity of T. vaginalis
also possible via artificial insemination of infected cry- is the ability of the trophozoite from a flagellated free-
obanked semen [25, 26]. However, in some rare cases, the swimming cell to an amoeboid-adherent stage. Scanning
presence of T. vaginalis has also been documented outside electron microscope studies by Figueroa-Angulo et al. [36]
the genitourinary tract: in the pharynx and lower respira- suggest that the amoeboid form is the key player of patho-
tory tract of adults. The occurrence of this protozoan in the genesis, as this morphological transition occurs when the
respiratory tract has been linked to orogenital sexual activ- parasite attaches itself to cervical and prostatic cells. Mor-
ity [27]. Although T. vaginalis commonly affects adults, phological transformations are associated with cytoskel-
documented cases of T. vaginalis infections have also been eton protein rearrangement and an increased expression of
seen in neonates who acquired the infection vertically genes encoding cytoskeletal proteins. Upon contact with
from the maternal genitourinary tract. Some rare cases of vaginal epithelial cells, the diffuse localization of actin and
infections with T. vaginalis of the vaginal, urinary, nasal fimbrin 1 (Tv Fim1) changes dramatically [36]. Live cell

13
E. Mielczarek, J. Blaszkowska

imaging demonstrates that trichomonal amoeboid stages do also shown to modulate host cell cytokine/interleukins 6
not just adhere to host tissue, but actively migrate across (IL-6) and 8 (IL-8)/production [42].
human epithelial cells in a concerted manner, with an aver-
age speed of 20 µm per minute [37]. Hydrolases and cytotoxic molecules

Cytoadherence A wide range of hydrolases (20–110 kDa) have been iso-


lated from T. vaginalis, including cysteine proteinases
Several proteomic analyses have also provided an insight which appear to be strongly correlated with virulence [38].
into the surface, soluble and secreted proteins that may be Currently, this parasite is estimated to contain 156 cysteine
involved in T. vaginalis pathogenesis [17, 33, 34]. The con- peptidases (CPs) [18], most of which are necessary for effi-
tact-dependent mechanisms described above play an impor- cient AP-mediated adhesion of T. vaginalis to target cells.
tant role in cell destruction of host. Adhesion of T. vagi- These hydrolases have trypsin-like activity and function as
nalis to red blood cells or the urogenital epithelium triggers cell-detaching factors by degrading host cell proteins such
the degradation of the skeleton of the host cell membrane, as laminin, fibronectin, and other components.
which can lead to cytolysis [34]. All 26 strains of T. vagi- Trichomonas vaginalis produces several cytotoxic mole-
nalis examined by Lustig et al. [35], with various degrees cules and mediates cytotoxicity by damaging the target cell
of virulence and isolated from various geographic loca- plasma membrane. Since T. vaginalis lacks the ability to
tions, required contact with host cells to induce cytolysis. synthesize lipids, the parasite obtains essential fatty acids
The lipophosphoglycan (LPG) is a major adherence by hemolytic activity [36]. The identification of parasite
factor in T. vaginalis, although molecular studies have 60-kDa CP, which is capable of degrading hemoglobin into
revealed that the expression of several other genes coding heme and globin, supports the supposition that this parasite
adhesion proteins (AP), fibronectin (FN)-binding protein, may use hemoglobin as a source of iron [43]. The lysis of
laminin-binding protein, actinin, enolase, phosphogluco- erythrocytes appears to be mediated by protein receptors on
mutase, and conserved GTP-binding protein (GTP-BP) are the surfaces of erythrocytes and parasites. Some molecules
up-regulated [36]. The human cell receptor of T. vaginalis, have a perforin-like activity and create pores in erythrocyte
galectin-1, has been identified on cervical epithelial cells membranes [36]. Twelve genes (TVSaplip1 to TVSaplip
and has been shown to bind to Trichomonas LPG [36]. 12) containing pore-forming domains have been identified.
The adhesion of the parasite to the epithelial cells is T. vaginalis also secretes various lytic factors with phos-
mediated by numerous adhesins, including AP120, AP65, pholipase A2 activity to destroy nucleated cells, erythro-
AP51, AP33, AP23, GAPDH, and several hypothetical cytes and specifically degrade phosphatidylcholine [44].
proteins [18, 36] which act in a specific receptor-ligand Pindak et al. [39] showed that apoptosis of the host cells
fashion, dependent on time, temperature, and pH. Gene may occur without the contact-dependent mechanisms of T.
expression of these APs is coordinately upregulated at the vaginalis, but also through the production of acidic metab-
transcriptional level by iron. The parasite AP65-1 gene olites and other factors by the living parasite. One of these
encodes a 65-kDa protein, which is believed to be one of factors is known as cell-detaching factor (CDF), which is
the surface adhesins upon iron repletion. Iron-inducible released by the parasite and is known to have trypsin-like
transcription of the AP65-1 gene in T. vaginalis involves activity. CDF, a 200-kDa glycoprotein that causes detach-
at least three Myb-like transcriptional factors (tvMyb1, ment of monolayer cells in vitro, analogous to the vaginal
tvMyb2 and tvMyb3) that differentially bind to two epithelial cell sloughing seen in trichomoniasis [20]. CDF
closely spaced promoter sites, MRE-1/MRE-2r and MRE- activity is pH dependent. Further, the production of CDF
2f [41]. has been shown to decrease in the presence of estrogen.
Twu et al. [42] report that T. vaginalis secretes small
vesicles called exosomes that are capable of fusing with, Phagocytosis
and delivering their contents to, host cells. A total of 215
proteins including surface proteins (Tvag_240680, BspA The cytopathogenic action of T. vaginalis may be divided
family) and proteases (Tvag_224980, metallopeptidase) into four stages: adhesion, cytolysis following contact,
have been detected in exosomes isolated from T. vaginalis phagocytosis and intracellular digestion [14, 15]. The
strain B7RC2. Hence, proteins that may have a role in T. phagocytosis of various cell types by T. vaginalis has
vaginalis pathogenesis are also present in the exosome been described: this parasite is able to efficiently ingest
proteome. These parasite-secreted vesicles were found to and degrade lactobacilli, yeast cells, vaginal and cervical
induce changes in the host cell and to mediate the interac- epithelial cells, leukocytes, erythrocytes, prostatic cells
tion of T. vaginalis with host by increasing the adherence and spermatozoids [14–16]. T. vaginalis phagocytosis is
of the parasite to host cells. T. vaginalis exosomes were thought to be both an efficient means of obtaining nutrients

13
Trichomonas vaginalis: pathogenicity and potential role in human reproductive failure

for the parasite and an important factor in the pathogenesis reaction to the parasite exceeds the boundaries of the repro-
of trichomonal infections of the human genitourinary tract. ductive tract mucosa [7].
Two types of yeast cell phagocytosis have been observed
by virulent strains of T. vaginalis [15]. The first involves Immune evasion strategy of T. vaginalis
a ‘sinking’ process, without any apparent participation of
plasma membrane extensions, while the second resem- Trichomonas vaginalis has numerous ways of evading the
bles the classical phagocytosis described for professional immune system, which include evasion of complement-
phagocytes, where pseudopodia are extended toward the mediated destruction, molecular mimicry, and the ability to
target cell, which is subsequently engulfed. Although these coat itself with host plasma proteins [33, 36, 40, 47].
two phagocytic processes were detected in both strains of Provenzano and Alderete [48] report that numerous CPs
T. vaginalis investigated by Pereira-Neves and Benchimol secreted by T. vaginalis degrade different types of immu-
[15], the first mechanism was found to occur more com- noglobulins which allows the parasite to survive the anti-
monly with the more virulent T016 strain. body response. Degradation of IgG and IgA was observed
The in vitro adherence and phagocytosis of sperm cells following incubation with lysates and culture supernatants
by T. vaginalis was demonstrated by Benchimol et al. [16] of T. vaginalis. Among the CPs, TVCP39 is one of the
using transmission and scanning electron microscopy. The papain-like proteinases that degrades both several extracel-
adhesion between trichomonads to the sperm cell occurred lular matrix proteins, including fibronectin, various types
either by the flagella or sperm head. The parasites were of collagen, IgG, IgA and hemoglobin. In addition, the T.
able to attach to the sperm cells by their cell surface, before vaginalis cysteine proteases, including CP30, induce apop-
commencing phagocytosis. This process began with a tight tosis in vaginal epithelial cells and in multiple mucosal
membrane–membrane adhesion and the incorporation immune cell types. T. vaginalis produces immuno-sup-
of the sperm cell within an intracellular vacuole. After- pressive cytokines (IL-10, TGFB) and causes caspase-
wards, the sperm cell was gradually digested in lysosomes. mediated apoptosis in T-cells, macrophages and dendritic
Another study conducted by Midlej and Benchimol [14] cells [40].
reports that the cytopathic effect of T. vaginalis occurs due A recent study by Ibáñez-Escribano et al. [47] describes
to mechanical stress and subsequent phagocytosis of the a novel strategy by T. vaginalis to evade the lytic action of
necrotic cells. the complement based on its procurement of host protein.
Trichomonads with surface-bound host-CD59 were pro-
Host response to T. vaginalis infections tected from lysis.
Upon T. vaginalis infection, cervical and vaginal epithe-
LPG plays a major pathogenic and immunoregulatory role lial cells release galectin-1 and galectin-3. These modulate
in adherence with VECs [36]. LPG triggers leukocytes to the inflammatory responses, with galectin-1 suppressing
secrete IL-8, parasite-specific immunoglobulin G (IgG) and galectin-3 enhancing the leucocyte response to inflam-
and A (IgA), Th1 cytokines, leukotrienes, reactive nitro- mation [40]. In addition, galectin-1 acts as a soluble adhe-
gen intermediates and macrophage inflammatory protein- sion molecule by facilitating the attachment of HIV-1 to the
3a [40, 45]. Reighard et al. [46] report increased numbers cell surface, thereby augmenting the efficiency of the viral
of polymorphonuclear leukocytes (PMNs), T cells, B cells, infection process [8].
and plasma cells in the endometrial tissue of the lower gen-
ital tract of women with T. vaginalis infection. Although The presence of dsRNA viral infection of T. vaginalis
the endometrial mucosa is home to immune responses
capable of eradicating pathogens, the increased inflamma- Trichomonas vaginalis can be infected with four dsRNA
tory infiltrate associated with this infection may increase viruses (TVVs), which are members of the Totiviridae. The
the likelihood for immunopathological upper genital tract genome of each TVVs is a single dsRNA molecule, approx-
damage. imately 4300–5500 bp in length [49]. It is suggested that
The mechanisms of pregnancy complications linked to the presence of TVV in T. vaginalis may have an impact
T. vaginalis infection remain unexplained. Asymptomatic on its virulence. This infection causes the up-regulation of
trichomoniasis in pregnancy is accompanied by a state of synthesis and surface expression of highly immunogenic
neutrophil activation. Additionally, women with asympto- proteins, especially P270, and significant differences in
matic trichomoniasis have been reported to have detectable the total protein composition of the parasite. The presence
vaginal fluid neutrophil defensins and cervical IL-8 concen- of TVV is also associated with differential qualitative and
trations [40]. The presence of increased C-reactive protein quantitative expression of cysteine proteinases, which are
levels in the sera of pregnant women infected with T. vagi- linked with cytoadherence, cytotoxicity, and degradation of
nalis suggests that the impact of the immunoinflammatory basement membrane components.

13
E. Mielczarek, J. Blaszkowska

A study investigating the genetic diversity and popula- cancer. Sutcliffe et al. [55] describe two possible molecular
tion structure of T. vaginalis identified two major genotypes mechanisms for T. vaginalis-mediated prostate carcinogen-
for the parasite (type 1 and type 2) with distinct properties esis: the first occurring via the inflammation process, and
[3, 50]. T. vaginalis isolates type 1 are more sensitive to the second proceeding through initiation of the cell-sign-
metronidazole, and are also significantly more likely to be aling cascade using proto-oncogenes PIM1, c-MYC, and
infected with TVV than T. vaginalis type 2, in a ratio ∼3:1. HMGA1.
Type 2 isolates are notable for having a higher prevalence
of metronidazole resistance [51]. Role of T. vaginalis in male infertility

Several studies report that men with T. vaginalis infection


Consequence of T. vaginalis infections in man display deleterious outcomes in reproduction [24, 52, 56,
57]. This has been associated with urethritis, prostatitis,
Trichomonas vaginalis has been detected in up to 77 % of epididymitis, and infertility through inflammatory damage
the male partners of infected women, and of those men, or interference with sperm function.
about 70 % were asymptomatic [24]. This high prevalence Sexual transmission of T. vaginalis is well established,
of T. vaginalis among sexual partners confirms its easy either via urethral discharge of men with urethritis or
transmission. Male trichomoniasis is most often associated through the semen. In men, trichomoniasis has been most
with urethritis, but infection may occur in other areas of frequently related to infertility by deficit of sperm cell
the urogenital system [20]. In rare cases, T. vaginalis also quality and function due to physical damage. Significantly
infects the epididymis and prostate gland and occasionally, higher levels of seminal contamination with the parasite
the testis [3, 4, 52]. have been found in infertile men compared to fertile con-
Although T. vaginalis has been identified in urethral trols [56]. The semen characteristics of males affected by
discharge, urine, semen and prostatic fluid, its presence in T. vaginalis have been described in several studies [24, 58].
these fluids does not confirm infection of either the prostate Gopalkrishnan et al. [58] note differences to the semen
or seminal vesicles, since fluid can be contaminated when from asymptomatic men, and showed that spermatozoan
it passes through the urethra. Although some authors regard motility, viability and morphologically normal forms were
the prostate as an ecological niche for T. vaginalis, others decreased significantly during infection. Additionally,
consider it to be an etiologic agent of chronic prostatitis. seminal fluid viscosity has been reported to be increased in
Gardner et al. [53] reported 5 well-documented cases of asymptomatic men with trichomoniasis, without significant
T. vaginalis detection in prostates obtained through autop- change in pH.
sies, with clinical evidence of the parasite in the urine of In vitro studies have shown that proteins secreted by T.
patients. Microscopic examination of all specimens dem- vaginalis rapidly immobilize or kill sperm. Sperm agglu-
onstrated multiple foci of nonspecific acute and chronic tination has also been described after exposure of human
prostatitis characterized by intraluminal macrophages and semen to high concentrations of live trophozoites ranging
a mixed chronic inflammatory infiltrate in the adjacent from 104 to 107 parasites per ml [59]. Benchimol et al. [16]
stroma. Other studies have shown the presence of T. vagi- report that, after 1 h of interaction between trichomonads
nalis in the prostate gland: trophozoites have been identi- and sperm cells under incubation conditions, 75 % of the
fied in the prostatic urethra, glandular lumina, submucosa, sperm cells were immotile or dead. Under scanning elec-
and stroma, in benign hyperplastic prostatic tissue [42, 54]. tron microscopy, the interaction of T. vaginalis and sperm
Mitteregger et al. [54] detected T. vaginalis DNA in 34 % cells results in high agglutination, membrane protrusions,
of investigated samples from 86 patients suffering from or the formation of channels between the sperm plasma
benign prostatic hyperplasia (BPH), with chronic inflam- membrane and parasite surface. Additionally, Mali et al.
matory tissue infiltration of unknown etiology. They also [57] reported a case of phagocytic interaction and agglu-
observed foci with nonspecific acute and chronic inflam- tinated human spermatozoa by T. vaginalis, observed after
mation, as well as intraepithelial vacuolization near trich- a Papanicolaou-stained cervical smear under light micros-
omonads which may lead to prostate carcinogenesis. Twu copy. Recently, experimental studies found that the secre-
et al. [42] demonstrated that a secreted T. vaginalis protein tory products of trichomonads decrease fertilizing capacity
(TvMIF—T. vaginalis macrophage migration inhibitory of mice sperm [60]. The incubation of sperm with extra-
factor) homologous to human macrophage migration inhib- cellular polymeric substances (EPS) from T. vaginalis, sig-
itory factor (HuMIF) elicits antibodies in infected individu- nificantly decreased sperm motility, viability and functional
als, increases prostate cell proliferation and invasiveness, integrity in a concentration and time-dependent manner.
and induces cellular pathways linked to inflammation that These effects on sperm quality also resulted in a decreased
contribute to the promotion and progression of prostate fertilization rate in vitro.

13
Trichomonas vaginalis: pathogenicity and potential role in human reproductive failure

A strong association between inflammation of the male trichomoniasis may promote HPV infection and further
reproductive system and infertility has been reported [52, increase the risk of CN [67, 68]. A study of rural Tanza-
61, 62]. The role of chronic infection by T. vaginalis on nian patients found that those with T. vaginalis infection
male infertility via the inflammatory process is not com- were 6.5 times more likely to have HPV type 16 than those
pletely understood. However, rare cases of sterile men with without [69]. The proposed mechanism for increased risk
trichomoniasis suggest the significant role played by these of cervical dysplasia in the case of Trichomonas invasion
protozoa in fertility disorder. One such difficult diagnosis is that the parasite produces mediators of inflammatory
of T. vaginalis orchitis was detected in men with severe response which cause cervical inflammation. The cervical
oligoasthenoteratospermia [63]. Semen analysis showed epithelium, as a protective barrier, is disrupted by inflam-
a sperm concentration of less than 100,000/ml with 0 % mation, allowing for penetrance of HPV to the basement
motility and 0 % morphological normal forms. Pathologi- membrane [69, 70]. As co-infections of T. vaginalis with
cal analysis confirmed the diagnosis of right orchitis caused viral and bacterial pathogens are known to occur, Chigbu
by T. vaginalis. Janssenswillen et al. [64] report a case et al. [71] suggest that the detection of T. vaginalis from the
of non-obstructive azoospermia in a 34-year-old patient lower genital tract can serve as an indicator for other more
resulting from maturation arrest associated with Tricho- serious STIs caused by N. gonorrhoeae, Gardnerella vagi-
monas infection at the level of the epididymis. The flag- nalis and C. trachomatis, as well as HIV and HPV.
ellated T. vaginalis cells were observed in the epididymal Trichomonas vaginalis infections have been associated
aspiration, while spermatozoa were not present and a wet with 1.5–2.7 times greater risks of HIV acquisition and
preparation of multiple testicular biopsies failed to demon- transmission [9, 72, 73]. According to Coleman et al. [72]
strate any spermatozoon. T. vaginalis has emerged as a cofactor for HIV transmis-
In HIV-positive men, adverse effects on sperm qual- sion and potential mechanisms for increased susceptibility
ity have been noted and semen quality has been found to include various complex processes: (1) recruitment of HIV
deteriorate with the progression of immunodeficiency [65]. target cells (CD4+ T cells) to the genital tract as a result
Hobbs et al. [66] report that the HIV RNA concentration of the host immune response to T. vaginalis, (2) degrada-
observed in the seminal plasma of HIV-positive Malawian tion of HIV-protective factors such as secretory leukocyte
men with symptomatic urethritis was significantly higher in protease inhibitor, and (3) the direct and indirect cyto-
those also infected with T. vaginalis. The role of co-infec- toxic effects of the parasite itself. Additionally, this pro-
tions of T. vaginalis with HIV in the impairment of sperm tozoan secretes a CDF that releases epithelial cells from
quality and motility still needs further investigation. the tissue, thus thinning the protective barrier against HIV
invasion. T. vaginalis also elicits minor extravasation in
mucosal genital tract tissue, which may facilitate transmis-
Consequence of T. vaginalis infections in women sion for HIV [8].
The factors that influence the host inflammatory
In females, 50 % of cases of T. vaginalis infections are response to T. vaginalis remain not fully understood, how-
asymptomatic and parasites can persist for long periods ever, hormonal levels, iron resource and the coexisting
of time in the urogenital tract of women. Between 25 and vaginal flora in the vagina have an important influence on
50 % are asymptomatic for the first 6 months of infec- the severity of trichomoniasis in female patients [36]. The
tion, and organisms can survive indefinitely in the lower rise of vaginal pH during infection may therefore be cru-
urogenital tract if left untreated [4, 20]. The parasite is cial in the pathogenesis of the disease. This phenomenon
normally isolated from the vagina, cervix, bladder, Bartho- is accompanied by a concomitant reduction of protective
lin’s, Skene’s, and periurethral glands [4]. Clinical forms lactobacilli that facilitates colonization of the reproductive
of trichomoniasis in women ranges from an asymptomatic tract with opportunistic pathogens [4]. The roles played by
carrier state to symptomatic vaginitis [3, 4]. Rarer compli- pH and hormones in trichomoniasis may explain the obser-
cations may result in pyosalpinx endometritis, adnexitis, vation that the symptoms of the disease are often worse
and can trigger inflammatory responses in the mucosal gen- during menstruation [20].
ital tract, increasing the risk of pelvic inflammatory disease
(PID) by microhemorrhages [4, 20]. Role of T. vaginalis in female infertility
Some authors suggested that in women, trichomonia-
sis increases the predisposition to cervical neoplasia (CN) The presence of pathogens in the reproductive tract can
[11]. The epidemiological data suggest that the aetiological result in fertility disorders, and similarly, the presence
agents of CN are genital pathogens, mainly human papil- of T. vaginalis in semen samples affects the quality of
lomavirus (HPV), herpes simplex virus (HSV) type-2, and sperm cells and impedes their ability to achieve oocyte
C. trachomatis. Moreover, concomitant STDs, including fertilization.

13
E. Mielczarek, J. Blaszkowska

The clinical data show that the prevalence of T. vagi- neonates [80–83]. For recent decades, the prevalence of
nalis is significantly higher among infertile women than newborn infection with T. vaginalis is unknown, but liter-
fertile controls [74]. El-Sharkawy et al. [75] showed that ature data indicate rare cases of vertical transmission [28,
infertile women with T. vaginalis have decreased C3 and 84–89]. However, 40 years ago, al-Salihi et al. [90] esti-
C4, increased IgA level in vaginal discharge and increased mated that transmission to newborns occurred in 2–17 %
serum prolactin. of female neonates of infected women. The mode of trans-
More than 20 % of women with trichomoniasis have a mission is proposed to occur through direct contact with
chronic inflammatory process in the reproductive tract. The the maternal genital tract during birth or contact between
protozoan attaches to vaginal epithelial cells and secretes the neonate and infected maternal fluids after delivery. It
various proteases and a CDF, leading to an intense host has been hypothesized that the premature rupture of fetal
inflammatory response, inducing local cytotoxic effects, membranes promotes the transmission of T. vaginalis,
genital tract damage, and reproductive effects [20, 39, 72]. because prolonged exposure to the vaginal flora enhances
This parasite has been isolated from fallopian tubes, peri- the chance of neonatal colonization and infection by these
toneal fluid, and the pouch Douglas, suggesting that motile organisms [91]. A few reports have shown the presence of
trophozoites may be able to invade the whole genital tract T. vaginalis in the respiratory tract of neonates [87–89].
[6, 71]. By virtue of their mobility, T. vaginalis not only Most documented cases of neonatal trichomoniasis include
infect the cervical canal, but simultaneous endometrial vaginal or urinary tract infections and parasitosis were
infection is also present, affecting the development of the diagnosed in the newborn from 5 to 28 days after birth [28,
uterine lining [46]. 81–85]. A high risk of developing urinary tract or vaginal
While roles for C. trachomatis and N. gonorrhoeae T. vaginalis infections exists in newborns due to the influ-
infections in pelvic inflammatory disease (PID) pathogen- ence of maternal estrogen, which remains at a high level in
esis are well delineated [76], the etiologic contributions the neonate for four to 6 weeks after birth [90]. Under the
of T. vaginalis have been not clarified. Some authors sug- influence of high maternal estrogen, the vaginal milieu of
gest that in rare cases, untreated trichomoniasis in HIV- newborn females has more glycogen deposits and a thicker
positive patients or during co-infection of T. vaginalis with epithelium than found in older children, which increases
other genital pathogens can lead even to PID [4, 77, 78]. the susceptibility to infection with T. vaginalis during the
Cherpes [78] notes that T. vaginalis infection and positive neonatal period [89]. According to Jenny et al. [92], the
HSV-2 serology are associated with endometritis. Accord- identification of sexually transmissible agents, including T.
ing to Moodley [77], T. vaginalis infection of the lower vaginalis in children beyond the neonatal period strongly
genital tract is associated with a clinical diagnosis of PID suggests sexual abuse, and every such case requires spe-
in HIV-1–infected women. Unfortunately, no data exist cial attention. It is also important to note that perinatally
regarding the mechanism for such an association. Theo- acquired rectal or vaginal C. trachomatis infection may
retically, the proteinases produced by T. vaginalis have persist for 2–3 years after birth while T. vaginalis neonatal
the potential to disrupt the protective cervical mucus plug, infections may continue for several months [93].
allowing the protozoan to pass from the lower to the upper Genitourinary tract infections are one cause of preterm
genital tract [38]. Reighard et al. [46] demonstrated an delivery. Approximately 40 % of spontaneous premature
increase in endometrial leukocyte subpopulations among births are thought to be caused by infection. Many authors
women with lower genital tract T. vaginalis infections, have confirmed the relationship of T. vaginalis with preterm
which suggests that the protozoan may play a role in upper labor. An analysis of the 178 articles published before May
genital tract inflammatory processes. Additionally, motile 2013 concerning perinatal outcomes in women infected
trichomonads have been isolated from samples of perito- with T. vaginalis provides strong evidence that trichomo-
neal fluid from women with vaginal trichomoniasis [79]. niasis in pregnancy is associated with an increased risk of
Although some evidence links T. vaginalis infection to preterm birth [6]. Uterine contractions may be induced by
upper genital tract infection, no conclusive proof of causal- cytokines (IL-1β), IL-6), and prostaglandins (PGE2 and
ity exists. Further studies will be needed to determine the PGE2a), which are released following T. vaginalis infec-
potential roles this parasite may play in PID pathogenesis. tion [82].

T. vaginalis infection and its effects on pregnancy


outcomes Diagnostic tests

During pregnancy, T. vaginalis infection has been asso- Microscopic examination of a wet mount preparation of
ciated with such adverse outcomes as preterm rupture genital secretions is still the most common method for
of membranes, preterm delivery, and low-birth-weight T. vaginalis detection because of convenience and low

13
Trichomonas vaginalis: pathogenicity and potential role in human reproductive failure

cost. However, the sensitivity of this traditional method is pregnancy complications, infertility and an increased risk
60–70 %, and decreases by as much as 35 % within half of HIV acquisition, as well as cervical and prostate cancer,
hour after collection [13, 94, 95]. Culture has a sensitiv- have been linked to T. vaginalis infection. This infection
ity of 75–96 % and a specificity of up to 100 % [96], but has a cosmopolitan distribution and has been identified in
is rarely used in routine laboratory tests [20]. Culture was all racial groups and socioeconomic strata. For this reason,
considered the gold standard method for diagnosing T. vag- trichomoniasis is one of CDC’s neglected parasitic infec-
inalis infection before molecular detection methods became tions, which are a group of five parasitic diseases that have
available [97]. The newly designed NAATs (e.g., APTIMA been targeted as priorities for public health action.
T. vaginalis FDA-approved assay) are the most sensitive Thanks to its wide clinical spectrum, control of this
tool for identifying Trichomonas asymptomatic infection infection must be based on the screening of symptomatic
in both women and men [12, 13], but their high cost and women and their partners and the appropriate treatment
required infrastructure prevent their widespread use in lab- of infected individuals to prevent the continued spread of
oratories. Additionally, point-of-care tests that detect pro- the disease. In addition, screening might be advisable for
tozoan antigens (e.g., OSOM® Trichomonas Rapid Test) or asymptomatic women at high risk of infection (e.g., with a
unamplified RNA (e.g., Affirm VP III) has also been devel- history of any STD or multiple sex partners).
oped [13, 97]. Unfortunately, these commercially kits have
limited availability. Additionally none of these tests has
Compliance with ethical standards 
been evaluated for screening asymptomatic women or for
diagnosis of trichomoniasis in men. Current recommenda- Conflict of interest  The authors declare that they have no conflict of
tions for T. vaginalis testing and screening, can be found interest to disclose. On behalf of all authors, the corresponding author
in CDC’s STD Treatment Guidelines, available online at: states that there is no conflict of interest.
http://www.cdc.gov/std/treatment [97].

References
Treatment
1. World Health Organization. Prevalence and incidence of selected
sexually transmitted infections, Chlamydia trachomatis, Neisse-
Most strains of T. vaginalis are sensitive to nitroimidazoles. ria gonorrhoeae, syphilis, and Trichomonas vaginalis: methods
Currently FDA-cleared metronidazole and tinidazole are and results used by the WHO to generate 2005 estimates. Geneva,
the standard drugs for therapy of trichomoniasis [97, 98]. Switzerland: World Health Organization, 2011. http://www.who.
int/reproductivehealth/publications/rtis/9789241502450/en/index.
According to CDC guidelines, a single 2 g dose of metro- htlm.
nidazole or 2 g dose of tinidazole is sufficient to cure more 2. World Health Organization. Global incidence and prevalence of
than 90 % of women with trichomoniasis, while metroni- selected curable sexually transmitted infections—2008. Geneva,
dazole 500 mg orally twice a day for 7 days can be used as Switzerland: WHO; 2012. http://www.who.int/reproductive-
health/publications/rtis/stisestimates/en/index.htlm.
an alternative [97]. Both drugs are recommended treatment 3. Harp DF, Chowdhury I. Trichomoniasis: evaluation to execution.
regimens in infected men. Metronidazole (FDA pregnancy Eur J Obstet Gynecol Reprod Biol. 2011;157:3–9.
category B) is recommended for pregnant women which 4. Lewis DA. Trichomoniasis. Medicine. 2010;38:291–3.
can be treated with 2 g metronidazole in a single dose at 5. Secor WE, Meites E, Starr MC, Workowski KA. Neglected para-
sitic infections in the United States: trichomoniasis. Am J Trop
any stage of pregnancy [98]. Because tinidazole is qualified Med Hyg. 2014;90:800–4.
to pregnancy category C the manufacturers advise against 6. Silver BJ, Guy RJ, Kaldor JM, Jamil MS, Rumbold AR. Tricho-
its use during the first trimester. Detailed guidelines regard- monas vaginalis as a cause of perinatal morbidity: a systematic
ing therapy of trichomoniasis, including cases of nitroimi- review and meta-analysis. Sex Transm Dis. 2014;41:369–76.
7. Simhan HN, Anderson BL, Krohn MA, Heine RP, Martinez de
dazole-resistant infections, are available in the 2015 Sexu- Tejada B, Landers DV, Hillier SL. Host immune consequences of
ally Transmitted Disease Treatment Guidelines published asymptomatic Trichomonas vaginalis infection in pregnancy. Am
by the Centers for Disease Control and Prevention [97]. J Obstet Gynecol. 2007;196:59.e1-5.
8. Kissinger P, Adamsku A. Trichomoniasis and HIV interactions: a
review. Sex Transm Infect. 2013;89:426–33.
9. McClelland RS, Sangare L, Hassan WM, Lavreys L, Mandaliya
Conclusions K, Kiarie J, Ndinya-Achola J, Jaoko W, Baeten JM. Infection
with Trichomonas vaginalis increases the risk of HIV-1 acquisi-
Despite the fact that trichomoniasis is the most common tion. J Infect Dis. 2007;195:698–702.
10. Stark JR, Judson G, Alderete JF, Mundodi V, Kucknoor AS,
curable sexually transmitted infection worldwide, the medi- Giovannucci EL, Platz EA, Sutcliffe S, Fall K, Kurth T, Ma J,
cal community still considers T. vaginalis a harmless inhab- Stampfer MJ, Mucci LA. Prospective study of Trichomonas
itant of the human reproductive tract. In the meantime, vaginalis infection and prostate cancer incidence and mortality:
serious adverse reproductive health outcomes, including physicians’ health study. J Natl Cancer Inst. 2009;101:1406–11.

13
E. Mielczarek, J. Blaszkowska

11. Afzan MY, Suresh K. Pseudocyst forms of Trichomonas vagi- 33. Ryan CM, de Migue N, Johnson PJ. Trichomonas vaginalis:
nalis from cervical neoplasia. Parasitol Res. 2012;111:371–81. current understanding of host–parasite interactions. Essays Bio-
12. Chapin K, Sndrea S. APTIMA Trichomonas vaginalis, a tran- chem. 2011;51:161–75.
scription-mediated amplification assay for detection of Tricho- 34. Lin WC, Chang WT, Chang TY, Shin JW. The pathogenesis of
monas vaginalis in urogenital specimens. Expert Rev Mol Diagn. human cervical epithelium cells induced by interacting with
2011;11:679–88. Trichomonas vaginalis. PLoS ONE. 2015;10:e0124087.
13. Hobbs MM, Seña AC. Modern diagnosis of Trichomonas vagi- 35. Lustig G, Ryan CM, Secor WE, Johnson PJ. Trichomonas

nalis infection. Sex Transm Infect. 2013;89:434–8. vaginalis contact-dependent cytolysis of epithelial cells. Infect
14. Midlej V, Benchimol M. Trichomonas vaginalis kills and eats- Immun. 2013;81:1411–9.
evidence for phagocytic activity as a cytopathic effect. Parasitol- 36. Figueroa-Angulo EE, Rendón-Gandarilla FJ, Puente-Rivera J,
ogy. 2010;137:65–76. Calla-Choque JS, Cárdenas-Guerra RE, Ortega-López J, Quin-
15. Pereira-Neves A, Benchimol M. Phagocytosis by Trichomonas tas-Granados LI, Alvarez-Sánchez ME, Arroyo R. The effects of
vaginalis: new insights. Biol Cell. 2007;99:87–101. environmental factors on the virulence of Trichomonas vaginalis.
16. Benchimol M, de Andrade Rosa I, da Silva Fontes R, Burla Dias Microbes Infect. 2012;14:1411–27.
AJ. Trichomonas adhere and phagocytose sperm cells: adhesion 37. Kusdian G, Woehle C, Martin WF, Gould SB. The actin-based
seems to be a prominent stage during interaction. Parasitol Res. machinery of Trichomonas vaginalis mediates flagellate-amoe-
2008;102:597–604. boid transition and migration across host tissue. Cell Microbiol.
17. de Miguel N, Lustig G, Twu O, Chattopadhyay A, Wohlschlegel 2013;15:1707–21.
JA, Johnson PJ. Proteome analysis of the surface of Trichomonas 38. Hernandez HM, Marcet R, Sarracent J. Biological roles of

vaginalis reveals novel proteins and strain-dependent differential cysteine proteinases in the pathogenesis of Trichomonas vagi-
expression. Mol Cell Proteomics. 2010;9:1554–66. nalis. Parasite. 2014;21:54. doi:10.1051/parasite/2014054.
18. Hirt RP, de Miguel N, Nakjang S, Dessi D, Liu YC, Diaz N, Rap- 39. Pindak FF, Mora de Pindak M, Jr Gardner WA. Contact-inde-
pelli P, Acosta-Serrano A, Fiori PL, Mottram JC. Trichomonas pendent cytotoxicity of Trichomonas vaginalis. Genitourin Med.
vaginalis pathobiology new insights from the genome sequence. 1993;69:35–40.
Adv Parasitol. 2011;77:87–140. 40. Fichorova RN. Impact of T. vaginalis infection on innate

19. Pereira-Neves A, Benchimol M. Trichomonas vaginalis: in vitro immune responses and reproductive outcome. J Reprod Immu-
survival in swimming pool water samples. Exp Parasitol. nol. 2009;83:185–9.
2008;118:438–41. 41. Jiang I, Tsai CK, Chen SC, Wang SH, Amiraslanov I, Chang CF,
20. Petrin D, Delgaty K, Bhatt R, Garber G. Clinical and microbio- Wu WJ, Tai JH, Liaw YC, Huang TH. Molecular basis of the rec-
logical aspects of Trichomonas vaginalis. Clin Microbiol Rev. ognition of the ap65-1 gene transcription promoter elements by a
1998;11:300–17. Myb protein from the protozoan parasite Trichomonas vaginalis.
21. Pereira-Neves A, Riberio KC, Benchimol M. Pseudocysts in
Nucl Acids Res. 2011;39:8992–9008.
trichomonads—new insights. Protist. 2003;154:313–29. 42. Twu O, de Miguel N, Lustig G, Stevens GC, Vashisht AA,

22. Hussein EM, Atwa MM. Infectivity of Trichomonas vaginalis Wohlschlegel JA, Johnson PJ. Trichomonas vaginalis exosomes
pseudocysts inoculated intra-vaginally in mice. J Egypt Soc Par- deliver cargo to host cells and mediate host:parasite interactions.
asitol. 2008;38:749–62. PLoS Pathog. 2013;9:e1003482.
23. Shafir SC, Sorvillo FJ, Smith L. Current issues and considera- 43. Min DY, Hyun KH, Ryu JS, Ahn MH, Cho MH. Degrada-

tions regarding trichomoniasis and human immunodeficiency tions of human immunoglobulins and hemoglobin by a 60 kDa
virus in African-Americans. Clin Micobiol Rev. 2009;22:37–45. cysteine proteinase of Trichomonas vaginalis. Korean J Parasi-
24. Seña AC, Miller WC, Hobbs MM, Schwebke JR, Leone PA, tol. 1998;36:261–8.
Swygard H, Atashili J, Cohen MS. Trichomonas vaginalis infec- 44. Lubick KJ, Burgess DE. Purification and analysis of a phos-
tion in male sexual partners: implications for diagnosis, treat- pholipase A2-like lytic factor of Trichomonas vaginalis. Infect
ment, and prevention. Clin Infect Dis. 2007;44:13–22. Immun. 2004;72:1284–90.
25. Sherman JK, Hostetle TL, McHenry K, Daly JJ. Cryosurvival of 45. Yadav M, Gupta I, Malla N. Kinetics of immunoglobulin G, M,
Trichomonas vaginalis during cryopreservation of human semen. A and IgG subclass responses in experimental intravaginal trich-
Cryobiology. 1991;28:246–50. omoniasis: prominence of IgG1 response. Parasite Immunol.
26. Habib FS, Metwally DM, Habib KS. Cryopreservation of Trich- 2005;27:461–7.
omonas vaginalis: a trial of using four different cryoprotectants. 46. Reighard SD, Sweet RL, Vicetti MC, Vicetti MRD, Chivukula
J Egypt Soc Parasitol. 2004;34:931–40. M, Krishnamurti U, Cherpes TL. Endometrial leukocyte sub-
27. Rebhun J. Pulmonary Trichomoniasis associated with a fever of populations associated with Chlamydia trachomatis, Neisseria
unknown origin. Calif Med. 1964;100:443–4. gonorrhoeae, and Trichomonas vaginalis genital tract infection.
28. Schwandt A, William C, Beig RH. Perinatal transmis-
Am J Obstet Gynecol. 2011;205:324.
sion of Trichomonas vaginalis: a case report. J Reprod Med. 47. Ibáñez-Escribano A, Nogal-Ruiz JJ, Pérez-Serrano J, Gómez-
2008;53:59–61. Barrio A, Escario JA, Alderete JF. Sequestration of host-CD59
29. Charles SX. Epidemiology of Trichomonas vaginalis (TV)
as potential immune evasion strategy of Trichomonas vaginalis.
in rural adolescent and juvenile children. J Trop Pediatr. Acta Trop. 2015;149:1–7.
1991;37:90. 48. Provenzano D, Alderete JF. Analysis of human immunoglobulin-
30. Adu-Sarkodie Y. Trichomonas vaginalis transmission in a family. degrading cysteine proteinases of Trichomonas vaginalis. Infect
Genitourin Med. 1995;71:199–200. Immun. 1995;63:3388–95.
31. Peterson K, Drame D. Iatrogenic transmission of Tricho-
49. Parent KN, Takagi Y, Cardone G, Olson NH, Ericsson M, Yang
monas vaginalis by a traditional healer. Sex Transm Infect. M, Lee Y, Asara JM, Fichorova RN, Baker TS, Nibert ML. Struc-
2010;86:353–4. ture of a protozoan virus from the human genitourinary parasite
32. Crucitti T, Jespers V, Mulenga C, Khondowe S, Vandepitte J, Trichomonas vaginalis. MBio. 2013;4:e00056-13.
Buvé A. Non-sexual transmission of Trichomonas vaginalis in 50. Conrad MD, Gorman AW, Schillinger JA, Fiori PL, Arroyo R,
adolescent girls attending school in Ndola, Zambia. PLoS ONE. Malla N, Dubey ML, Gonzalez J, Blank S, Secor WE, Carlton
2011;6:e16310. JM. Extensive genetic diversity, unique population structure and

13
Trichomonas vaginalis: pathogenicity and potential role in human reproductive failure

evidence of genetic exchange in the sexually transmitted parasite 69. Lazenby GB, Taylor PT, Badman BS, McHaki E, Korte JE,

Trichomonas vaginalis. PLoS Negl Trop Dis. 2012;6(3):e1573. Soper DE, Young Pierce J. An association between Trichomonas
51. Malla N, Kaul P, Sehgal R, Gupta I. The presence of dsRNA vaginalis and high-risk human papillomavirus in rural Tanza-
virus in Trichomonas vaginalis isolates from symptomatic nian women undergoing cervical cancer screening. Clin Ther.
and asymptomatic Indian women and its correlation with 2014;36:38–45.
in vitro metronidazole sensitivity. Indian J Med Microbiol. 70. Pustan L, Ailiesei O, Dunca S. Trichomonas vaginalis—a risk
2011;29:152–7. factor for cervical cancer. Gen Mol Biol. 2010;11:107–12.
52. La Vignera S, Vicari E, Condorelli RA, D’Agata R, Calogero 71. Chigbu LN, Aluka C, Eke RA. Trichomoniasis as an indicator
AE. Male accessory gland infection and sperm parameters for existing sexually transmitted infections in women in Aba,
(review). Int J Androl. 2011;34:e330–47. Nigeria. Ann Afr Med. 2006;5:1–5.
53. Gardner WA Jr, Culberson DE, Bennett BD. Trichomonas
72. Coleman JS, Gaydos CA, Witter F. Trichomonas vaginalis vagi-
vaginalis in the prostate gland. Arch Pathol Lab Med. nitis in obstetrics and gynecology practice: new concepts and
1986;110:430–2. controversies. Obstet Gynecol Surv. 2013;68:43–50.
54. Mitteregger D, Aberle SW, Makristathis A, Walochnik J, Brozek 73. Guenthner PC, Secor WE, Dezzutti CS. Trichomonas vagi-

W, Marberger M, Kramer G. High detection rate of Trichomonas nalis—induced epithelial monolayer disruption and human
vaginalis in benign hyperplastic prostatic tissue. Med Microbiol immunodeficiency virus type 1 (HIV-1) replication: implica-
Immunol. 2012;201:113–6. tions for the sexual transmission of HIV-1. Infect Immun.
55. Sutcliffe S, Neace C, Magnuson NS, Reeves R, Alderete JF. 2005;73:4155–60.
Trichomonosis, a common curable STI, and prostate carcino- 74. El-Shazly AM, El-Naggar HM, Soliman M, El-Negeri M, El-
genesis—a proposed molecular mechanism. PLoS Pathog. Nemr HE, Handousa AE, Morsy TA. A study on trichomo-
2012;8:e1002801. niasis vaginalis and female infertility. J Egypt Soc Parasitol.
56. Martinez-Garcia F, Regadera J, Mayer R, Sanchez S, Nistal
2001;31:545–53.
M. Protozoan infections in the male genital tract. J Urol. 75. El-Sharkawy IM, Hamza SM, El-Sayed MK, El-Negeri M, El-
1996;156:340–9. Nemr HE, Handousa AE, Morsy TA. Correlation between tricho-
57. Mali NM, Hazari KT, Meherji PK. Interaction between T. vagi- moniasis vaginalis and female infertility. J Egypt Soc Parasitol.
nalis and human spermatozoa in the genital tract: Papanicolaou- 2000;30:287–94.
stained cervical smear findings. Acta Cytol. 2006;50:357–8. 76. Ness RB, Kip KE, Hillier SL, Soper DE, Stamm CA, Sweet RL,
58. Gopalkrishnan K, Hinduja IN, Kumar TC. Semen characteristics Rice P, Richter HE. A cluster analysis of bacterial vaginosis-
of asymptomatic males affected by Trichomonas vaginalis. J In associated microflora and pelvic inflammatory disease. Am J
Vitro Fert Embryo Transf. 1990;7:165–7. Epidemiol. 2005;162:585–90.
59. Tuttle JP, Holbrook TW, Derrick FC. Interference of human
77. Moodley P, Wilkinson D, Connolly C, Moodley J, Sturm AW.
spermatozoal motility by Trichomonas vaginalis. J Urol. Trichomonas vaginalis is associated with pelvic inflammatory
1977;118:1024–5. disease in women infected with human immunodeficiency virus.
60. Roh J, Lim YS, Seo MY, Choi Y, Ryu JS. The secretory products Clin Infect Dis. 2002;34:519–22.
of Trichomonas vaginalis decrease fertilizing capacity of mice 78. Cherpes TL, Wiesenfeld HC, Melan MA, Kant JA, Cosentino
sperm in vitro. Asian J Androl. 2015;17:319–23. LA, Meyn LA, Hillier SL. The associations between pelvic
61. Gimenes F, Souza RP, Bento JC, Teixeira JJV, Maria-Engler SS, inflammatory disease, Trichomonas vaginalis infection, and
Bonini MG, Consolaro MEL. Male infertility: a public health positive herpes simplex virus type 2 serology. Sex Transm Dis.
issue caused by sexually transmitted pathogen. Nat Rev Urol. 2006;33:747–52.
2014;11:672–7. 79. Hammond TL, Hankins GD, Snyder RR. Transvaginal-perito-
62. Lucena E, Moreno-Ortiz H, Coral L, Lombana O, Moran A, neal migration of Trichomonas vaginalis as a cause of ascites: a
Esteban-Pérez CI. Unexplained infertility caused by a latent report of two cases. J Reprod Med. 1990;35:179–81.
but serious intruder: Trichomonas vaginalis? JFIV Reprod Med 80. Simhan HN, Caritis SN, Krohn MA, Hillier SL. The vaginal
Genet. 2015;3:139. doi:10.4172/2375-4508.1000139. inflammatory milieu and the risk of early premature preterm rup-
63. Lloyd G, Case JR, De Frias D, Brannigan RE. Trichomonas vag- ture of membranes. Am J Obstet Gynecol. 2005;192:213–8.
inalis orchitis with associated severe oligoasthenoteratospermia 81. Smith LM, Wang M, Zangwill K, Yeh S. Trichomonas vaginalis
and hypogonadism. J Urol. 2003;170:924–5. infection in a premature newborn. J Perinatol. 2002;22:502–3.
64. Janssenswillen C, Tournaye H, Pierard D, Devroey P, Van
82. Cram LF, Zapata MI, Toy EC, Baker B. Genitourinary infections
Steirteghem A. Microsurgical epididymal sperm aspiration with and their association with preterm labor. Am Fam Physician.
motile trophozoite cells but no spermatozoa. Hum Reprod. 2002;65:241–8.
1997;12:2217–9. 83. Cotch MF, Pastorek JG II, Nugent RP, Hillier SL, Gibbs RS,
65. Le Tortorec A, Dejucq-Rainsford N. HIV infection of
Martin DH, Eschenbach DA, Edelman R, Carey JC, Regan JA,
the male genital tract–consequences for sexual transmis- Krohn MA, Klebanoff MA, Rao AV, Rhoads GG. Trichomonas
sion and reproduction. Int J Androl. 2010;33:e98–108. vaginalis associated with low birth weight and preterm delivery.
doi:10.1111/j.1365-2605.2009.00973.x. The vaginal infections and prematurity study group. Sex Transm
66. Hobbs MM, Kazembe P, Reed AW, Miller WC, Nkata E, Zimba Dis. 1997;24:353–60.
D, Daly CC, Chakraborty H, Cohen MS, Hoffman I. Tricho- 84. Trintis J, Epie N, Boss R, Riedel S. Neonatal Trichomonas vagi-
monas vaginalis as a cause of urethritis in Malawian men. Sex nalis infection: a case report and review of literature. Int J STD
Transm Dis. 1999;26:381–7. AIDS. 2010;21:606–7.
67. Yap EH, Ho TH, Chan YC, Thong TW, Ng GC, Ho LC, Singh 85. Rasti S, Behrashi M, Mousavi G, Moniri R. Complications of
M. Serum antibodies to Trichomonas vaginalis in invasive cervi- trichomoniasis on the pregnant women. Jundishapur J Microbiol.
cal cancer patients. Genitourin Med. 1995;71:402–4. 2011;4:61–3.
68. Viikki M, Pukkala E, Nieminen P, Hakama M. Gynaecological 86. Mann JR, McDermott S, Zhou L, Barnes TL, Hardin J. Treat-
infections as risk determinants of subsequent cervical neoplasia. ment of trichomoniasis in pregnancy and preterm birth: an obser-
Acta Oncol. 2000;39:71–5. vational study. J Womens Health (Larchmt). 2009;18:493–7.

13
E. Mielczarek, J. Blaszkowska

87. Bruins MJ, van Straaten IL, Ruijs GJ. Respiratory disease and 93. Hammerschlag MR. Sexual assault and abuse of children. Clin
Trichomonas vaginalis in premature newborn twins. Pediatr Infect Dis. 2011;53:S103–9. doi:10.1093/cid/cir700.
Infect Dis J. 2013;32:1029–30. 94. Kingston MA, Carlin EM, Bansal D. ‘Shelf life’ of Trichomonas
88. Temesvari P, Kerekes A, Tege A, Szarka K. Demonstra-
vaginalis. Int J STD AIDS. 2003;14:28–9.
tion of Trichomonas vaginalis in tracheal aspirates in infants 95. Patil MJ, Nagamoti JM, Metgud SC. Diagnosis of Tricho-

with early respiratory failure. J Matern Fetal Neonatal Med. monas vaginalis from vaginal specimens by wet mount micros-
2002;11:347–9. copy, In Pouch TV culture system, and PCR. J Glob Infect Dis.
89. Carter JE, Whithaus KC. Neonatal respiratory tract involvement 2012;4:22–5.
by Trichomonas vaginalis: a case report and review of the litera- 96. Nye MB, Schwebke JR, Body BA. Comparison of APTIMA
ture. Am J Trop Med Hyg. 2008;78:17–9. Trichomonas vaginalis transcription-mediated amplification to
90. al-Salihi F L, Curran J P, Wang J. Neonatal Trichomonas vagi- wet mount microscopy, culture, and polymerase chain reaction
nalis: report of three cases and review of the literature. Pediatrics for diagnosis of trichomoniasis in men and women. Am J Obstet
1974;53:196–200. Gynecol. 2009;200:188.e1-7. doi:10.1016/j.ajog.2008.10.005.
91. Azargoon A, Darvishzadeh S. Association of bacterial vagino- 97. Workowski KA, Bolan GA, Centers for Disease Control and
sis, Trichomonas vaginalis, and vaginal acidity with outcome of Prevention. Sexually transmitted diseases treatment guidelines,
pregnancy. Arch Iran Med. 2006;9:213–7. 2015. MMWR Recomm Rep. 2015;5:1–137.
92. Jenny C, Crawford-Jakubiak JE. Committee on child abuse and 98. Sherrard J, Ison C, Moody J, Wainwright E, Wilson J, Sullivan
neglect, American Academy of Pediatrics. The evaluation of chil- A. United Kingdom National Guideline on the Management of
dren in the primary care setting when sexual abuse is suspected. Trichomonas vaginalis 2014. Int J STD AIDS. 2014;25:541–9.
Pediatrics. 2013;132:e558–67. doi:10.1542/peds.2013-1741.

13

You might also like