Trichomonas 3

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

TREPAR 1764 No.

of Pages 11

Review

Trichomonas vaginalis: Pathogenesis,


Symbiont Interactions, and Host Cell
Immune Responses
Frances Mercer1 and Patricia J. Johnson2,*

The parasite Trichomonas vaginalis (Tv) causes a highly prevalent sexually Highlights
transmitted infection. As an extracellular pathogen, the parasite mediates New players in the multifactorial pro-
cess of host–parasite interactions have
adherence to epithelial cells to colonize the human host. In addition, the
been identified, including parasite-
parasite interfaces with the host immune system and the vaginal microbiota. secreted vesicles (exosomes) and var-
Modes of Tv pathogenesis include damage to host tissue mediated by parasite ious surface and secreted proteases
and effector proteins.
killing of host cells, disruption of steady-state vaginal microbial ecology, and
eliciting inflammation by activating the host immune response. Recent Tv Tv is associated with disruption of the
research has uncovered new players that contribute to multifactorial mecha- vaginal microbiota; specific interac-
tions with vaginal microbes represen-
nisms of host–parasite adherence and killing, and has examined the relation- tative of the healthy and disrupted-
ship between Tv and vaginal bacteria. Mechanisms that may lead to parasite state are being examined.
recognition and killing, or the evasion of host immune cells, have also been
Neutrophils were found to kill Tv using a
revealed. novel antimicrobial mechanism, trogo-
cytosis. Identification of the mechan-
isms immune cells use to kill Tv allows
Trichomonas vaginalis (Tv) Is the Causative Agent of Trichomoniasis analysis of modes that the parasite
Tv is a unicellular, flagellated, microaerophilic (see Glossary) protist that causes trichomonia- might use to evade immune elimination.
sis, a highly prevalent sexually transmitted infection (STI) [1–3]. With a global prevalence of
250 million, trichomoniasis is the most common nonviral sexually transmitted infection [4]. A holistic view of how Tv modulates
host immunity is emerging. New roles
While >95% of Tv infections can be cleared using the 5-nitro-imidazole drug metronidazole, for three microbial symbionts, a para-
drug-resistant strains are on the rise [1]. Common symptoms in infected women are vaginitis, site-derived cytokine mimic (TvMIF),
and vaginal discharge; however, serious adverse consequences include pelvic inflammatory exosomes, a surface glycoconjugate,
disease, increased risk of HIV transmission and acquisition, and increased risk of malignant and the selective targeting of lympho-
cyte populations for killing have been
cervical cancers [5,6–8]. Tv is also associated with female and male infertility, and with preterm/ revealed.
low-weight infant births [5,6,9,10]. Notably, 50% of women are asymptomatic [11]. Greater
than 75% of men are asymptomatic [2]; however, men are carriers, and Tv can cause
inflammation in the prostate.
1
Department of Biological Sciences,
To establish infection, the parasite must mediate contact with host epithelial cells, evade the California State Polytechnic University,
onslaught of the host immune response, and contend with the vaginal microbiota (Figure 1, 3801 West Temple Avenue, Pomona,
Key Figure). The parasite also has a microbiota of its own, harboring two Mycoplasma species CA 91768, USA
2
Department of Microbiology,
and a virus, which contribute to its pathogenesis [12]. In the wake of the Tv genome sequence Immunology & Molecular Genetics,
publication, the adaptation of omics, in vitro adherence and cytotoxicity assays, and imaging University of California, 1602
technologies to interrogate Tv host–pathogen interactions, recent years have seen much Molecular Sciences Building, 609
Charles E. Young Drive East, Los
progress in our understanding of how this important parasite causes disease. Angeles, CA 90095-1489, USA

Cellular Pathogenesis: Tv Adherence to Host Cells and Host-Cell Killing


*Correspondence:
Tv is an extracellular pathogen; it adheres to host cells as its mode of infection and pathogen- frances.mercer@gmail.com (F. Mercer)
esis [13], likely relying on adherence to genital epithelial cells to survive elimination through and johnsonp@ucla.edu (P.J. Johnson).

Trends in Parasitology, Month Year, Vol. xx, No. yy https://doi.org/10.1016/j.pt.2018.05.006 1


© 2018 Elsevier Ltd. All rights reserved.
TREPAR 1764 No. of Pages 11

Key Figure Glossary


Bacterial vaginosis (BV): an
Pathogenic Behaviors of Trichomonas vaginalis overgrowth of anaerobic vaginal
bacteria, especially Gardnerella
vaginalis.
Host immunity Candida albicans: a fungus
normally found in humans. It can
overgrow and cause ‘yeast
infections’ in the vagina.
Cytokine: a secreted protein used
for communication among immune
Subvert Incite inflammaƟon cells.
ECM (extracellular matrix):
Trichomonas secreted proteins and
polysaccharides that bind cells
vaginalis
together.
Ectosomes: extracellular vesicles of
Disrupt? moderate size (100–350 nm) which
Damage bud directly from the plasma
membrane.
Exosomes: small extracellular
vesicles (30–100 nm), secreted from
cells, that transfer information to
Host epithelia Vaginal microbiota neighboring cells.
Extracellular degranulation: the
process by which neutrophils excrete
vesicles containing toxic effector
molecules onto a pathogen to elicit
Figure 1. The parasite T. vaginalis (blue) is a sexually transmitted parasite that primarily produces symptoms and
pathogen death.
pathology in the cervical and vaginal mucosa. The parasite can adhere to, and lyse, host epithelial cells (pink), causing
Humoral immunity: the arm of the
tissue damage. Immune responses (gray) during T. vaginalis infection cause inflammation, which can also contribute to
immune response that generates
pathology, and the parasite has mechanisms to evade clearance by host immunity. Finally, the parasite lives alongside the
antibodies and implements antibody-
vaginal microbiota, and is associated with pathogenic disruptions of vaginal bacteria.
mediated pathogen attack.
Imaging flow cytometry: a
technology that combines the speed
and quantitation of flow cytometry
gravity and secretions. Tv also derives nutrients from lysing and feeding off host cells [13]. Tv with high-resolution microscopy,
allowing localization and quantitation
can adhere to a broad range of hosts [14], and key cytoskeletal changes appear to facilitate
of fluorescent signals in single cells.
host-cell binding. However, specific molecular moieties on the surface of parasite and host Inflammasome: an intracellular
have also been shown to play a role in adherence and host-cell killing, including the parasite multiprotein complex in immune cells
surface lipoglycan (LG), which binds to host cell galectin-1 [15], and Tv membrane proteins that can recognize pathogens or
stress, and activate some
TVAG_244130 (TvBAP1) and TVAG_188850 (TvBAP2) [16]. Cellular pathogenesis is almost
inflammatory cytokines, including IL-
certainly multifactorial, as loss-of-function experiments with individual candidate players never 1b.
completely inhibit adherence or host cell killing, and several surface molecules have been Inflammation: a state of immune
shown to play a role in adherence (Table 1). activation which can mediate
pathogen clearance, but which can
also become pathogenic.
Clinical isolates of the parasite vary considerably in their ability to adhere to and kill host cells. Lactobacillus: the most abundant
Work in our laboratory comparing adherence and host-cell killing among clinical isolates bacteria in healthy vaginas; it
revealed that although the two behaviors do not share a strictly linear correlation, a threshold produces lactic acid.
Metronidazole: an antibiotic
of sufficient adherence must be reached for host-cell killing to occur [17]. Also, a few strains that commonly used to treat Trichomonas
are highly adherent do not kill host cells [17], indicating that adherence per se is not sufficient for vaginalis and other anaerobic
host-cell killing, and that the latter is mediated by specific effectors that vary between strains. organisms.
Microaerophilic: refers to growth in
Recently, some new players have been shown to contribute to both processes (Table 1).
the presence of limited oxygen.
Microbiota: all the microbes in a
New Players in Adherence particular environment.
Recent experiments assaying the ability of parasites to adhere to host cells in vitro have identified MIF (macrophage migration
inhibitory factor): a highly
new players. Knock-down of TvPP1 g, a protein phosphatase, reduced host cell adherence by

2 Trends in Parasitology, Month Year, Vol. xx, No. yy


TREPAR 1764 No. of Pages 11

Table 1. Factors That Promote Adherence of Trichomonas vaginalis to Host Cells conserved secreted protein that
functions as a pleiotropic
Category Parasite effector Mechanism Refs
inflammatory cytokine.
Surface glycoconjugate Lipoglygan Bind host Galectin-1 [15] Mycoplasma: a genus of bacteria
which lack cell walls; they are the
Surface proteins TvBAP-1 Unknown [16]
smallest known living cells.
Tv BAP-2 Unknown [16] NETosis (neutrophil extracellular
trap): the process by which
Enzymes TvPP1g Unknown [18]
neutrophils expel their nuclear DNA
TvLegu 1 Unknown [20] to form extracellular webs of nucleic
acid that can ensnare, and lead to
TvROM1 Processes surface proteins [21]
the death of pathogens.
Vesicles Exosomes Deliver protein and RNA to host cell [32] Neutrophils: the most abundant
immune cells in mammals, and
Modulating vaginal microbiota Unknown Inhibit growth of lactobacilli [39]
usually the first to arrive at the site of
infection. Neutrophils have multiple
50% [18]. Additionally, it has been reported that TvTim, a Tv glycolytic enzyme, binds laminin and mechanisms to kill pathogens.
Opsonization: the process by which
fibronectin, suggesting that TvTim could play a role in ECM adherence during infection [19]. serum factors coat a pathogen
surface, providing cross-linking to
Tv proteases have also been shown to be important mediators of adherence. The cysteine receptors on phagocytic or
protease TvLegu1 appears to play a role, as treatment of host cells with antisera raised against trogocytic cells, thus facilitating
engulfment.
TvLegu1 showed a 45% decrease in adherence compared to preimmune serum control. Phagocytosis: the process by
Furthermore, cysteine protease inhibitors reduce adherence by around 66%, indicating that which one cell engulfs another cell
TvLegu1 and other cysteine proteases play an important role in adherence [20]. Finally, we whole. Neutrophils can phagocytose
and subsequently degrade many
found that a serine protease inhibitor reduced adherence by 60%, and rhomboid serine
pathogens.
protease TvROM1 was shown to increase adherence by 1.5-fold. A biochemical assay to PRRs (pattern-recognition
identify TvROM1 substrates identified TvBAP-1, which is cleaved by TvROM1 and also receptors): molecules on immune
mediates adherence [16,21]. Interestingly, however, mutation in the TvROM1 cleavage site cells that recognize specific microbial
molecular signatures and relay a
of TvBAP-1 further increased parasite adherence to host cells, indicating that Tv adherence is
signal to the cytoplasm upon
most effective when a dynamic on–off approach is utilized, possibly in the context of parasite engagement, resulting in initiation of
aggregates, as we discuss below [21]. an immune response.
SLPI (secretory leukocyte
protease inhibitor): a protein
Protein palmitoylation also likely plays a role in cytoadherence, as treatment with a palmitoy-
secreted by host cells which is
lation inhibitor decreased host-cell adherence by 60% [22]. Players in adherence were also thought to protect host cells from
found to be regulated epigenetically, as treatment with a histone acetylation inhibitor increased proteases secreted by immune
adherence and also activated transcription at the promoters of genes shown to play a role in effector cells.
TLR3 (Toll-like receptor 3): a
adherence [23]. Consistent with these results, a genome-wide approach found histone H3 member of the TLR family of PRR
modification associated with epigenetic upregulation of gene expression [24]. signaling proteins found on the
surface of immune cells; it
Effectors of Host-Cell Killing recognizes dsRNA.
Trogocytosis: (trogo = to nibble); a
Few studies have identified specific effectors in host-cell destruction. We showed that host-cell process by which one cell uptakes
cytosol is released after coculture with parasites [17], indicating that host-cell killing proceeds fragments of an adjacent cell.
through necrosis, rather than induction of apoptosis. Spillage of cytosol was also observed in
the killing of host lymphocytes [25]. Most recently, two proteases have been shown to play a
role in host-cell killing. We found that overexpression of TvROM1 increased host-cell lysis by 4-
fold, while only increasing adherence 1.5-fold, indicating that the protease may affect players of
a lytic mechanism. Others have found that killing of epithelial cells was reduced 20% in the
presence of polyclonal antibody to metalloprotease TvMP50, and that recombinant TvMP50
showed moderate lytic activity to host-cell monolayers [26].

High-resolution imaging has also provided insight into host-cell killing mechanisms. Interest-
ingly, very tight interdigitations between Tv and host-cell membranes have been observed in
electron microscopy (EM) pictures [27], suggesting that Tv may kill host epithelial cells by a

Trends in Parasitology, Month Year, Vol. xx, No. yy 3


TREPAR 1764 No. of Pages 11

mechanism – trogocytosis (trogo = to nibble) – similar to that recently reported in Entamoeba


histolytica [27,28]. In contrast, whole-cell phagocytosis of smaller lymphoid cells and other
microbes has been observed [29,30].

Cellularity and Cellular Morphology during Adherence


It was recently observed that treatments with acetylation or palmitoylation inhibitors, which
increase host-adherent behavior, also increase parasite clumping (parasite–parasite adher-
ence) [22,23]. Parasite clumping may facilitate social behavior for group-mediated adherence
and host-cell killing. A scenario in which several parasites ‘hold on’ to host cells to avoid being
swept away, while others in the clump lyse host cells for nutrients or invasion, can be
envisioned. In this regard, it is interesting that Tv overexpressing the tetraspanin protein
Tsp8 increases clumping 8-fold [31]. We have also observed that exosomes with Tsp1
on their surface increase parasite adherence to host cells [32]. Together, these findings indicate
that tetraspanins contribute to Tv virulence.

EM analyses have revealed striking images of amoeboid-shaped Tv adhered to host cells [33],
suggesting that cytoskeletal rearrangement is involved in host-cell adherence. Investigators have
therefore sought to determine players in this amoeboid transition, and have identified a fimbrin
protein TvFim1 associated with actin bundles during amoeboid transition [34], consistent with
TvFim1 playing a role in mediating host-cell adherence. It will be interesting to determine the
relative contribution of cytoskeletal versus surface receptor factors in adherence to host cells. It is
likely that the parasite employs different strategies in different in vivo circumstances or that different
strategies are more effective on some cellular targets compared to others.

Effect of Tv Extracellular Vesicles on Adherence


We showed that Tv secretes exosomes, small extracellular vesicles that can fuse with host cells
[32]. Remarkably, these exosomes were found to ‘prime’ host cells for adherence: parasites
adhered better to host cells that were pretreated with exosomes, suggesting that Tv can
‘prepare the environment’ for optimal colonization. Since exosomes deliver cargo into the host-
cell cytosol, Tv exosomes may affect host cell expression of adherence ligands. Moreover,
host-cell adherence was more enhanced when both host cells and parasites were preincu-
bated with exosomes before the adherence assay. Combined with the observation that more
adherent strains of parasites clump together, this suggests that parasites could use the same
molecular moieties to bind to host cells as they do to bind each other, and exosomes serve as a
way for the parasite to deposit a prefabricated ligand for its receptor onto the host cell. Further
supporting this hypothesis is the striking phenotype that exosomes from more adherent strains
of Tv can transfer their more-adherent phenotype to less-adherent strains, through exosomes
[32]. However, it remains to be determined whether exosomes from more-adherent strains
facilitate enhanced adherence through their surface-associated moieties, or through activity of
the cytosolic cargo. Furthermore, ectosomes, larger extracellular vesicles that bud off the
plasma membrane, are also produced by Tv and are upregulated upon host cell encounter,
suggesting that they may also modulate host-cell pathogenesis [35].

Tv Interactions with the Vaginal Microbiota


The vaginal microbiota likely plays a key role in Tv pathogenesis. In 2011, five main types of vaginal
microbiota were established, ranging in their relative abundance of lactobacillus and overall taxa
diversity, with type IV being the most diverse, and most commonly associated with pathogenic
dysbioses, including bacterial vaginosis (BV), which is characterized by a major shift from
lactobacillus dominance to mostly anaerobic species [36]. Interestingly, Tv is associated with a
type IV microbiome. One report found that 72% of Tv+ women have a type IV vaginal microbiome

4 Trends in Parasitology, Month Year, Vol. xx, No. yy


TREPAR 1764 No. of Pages 11

[37]. Another study showed that Tv is present in 30% of cases of BV [38]. While it is not clear
whether anaerobic microbiomes predispose to the acquisition of Tv, or whether Tv disrupts the
vaginal microbiome causing dysbioses, some recent studies have examined outcomes of Tv
interactions with common vaginal residents. Notably, in a vaginal ectocervical cell -microbiome-Tv
culture model, Tv was shown to inhibit growth of lactobacillus, but not BV-associated bacteria [39],
suggesting Tv-driven modification of vaginal microbes. On the other hand, lactobacillus was found
to decrease Tv adherence to host epithelial cells by 60% [40], suggesting that lactobacillus-
dominated vaginas are less hospitable to the parasite. Also, in the presence of Candida albicans,
Tv was found to be more cytopathic towards fibroblast cultures [41], suggesting that Tv patho-
genesis is increased in Tv–C. albicans concomitant infections.

Another factor in Tv niche establishment is likely to be the human vaginal pH, which is typically
4.5 [39]. One study reported elevated vaginal pH as a key correlate of Tv infection [42]. Similarly,
the type IV microbiome, which is most commonly associated with Tv, has an elevated pH
compared to other vaginal microbiome types [37]. In vitro, the optimal pH for axenic Tv growth
is 6.2, consistent with Tv either preferring a higher than normal vaginal pH, or with the parasite
contributing to elevating vaginal pH. Future work to better discern vaginal environments that
favor or discourage Tv growth and pathogenesis may inform design of probiotic or other
suppository therapies to treat infection.

Tv Symbionts Contribute to Pathogenesis


Tv is known to commonly harbor Trichomonas vaginalis virus (TVV), a double-stranded RNA
totivirus, and two different mycoplasma species, both of which likely contribute to Tv patho-
genesis [12].

TVV
TVV can be classified into types I–IV; the parasite may harbor none, one, or multiple types. TVV
and the percentage of global isolates harboring it has been recently reviewed [12]. Analysis of
TVV+ versus TVV clinical isolates is consistent with adherence to host cells being increased in
TVV+ strains [12]; however, caution is required in interpreting these results as adherence of
isogenic strains differing only in the presence of TVV has not been examined. Antivirals to clear
strains of TVV, and which could determine whether these correlations are strictly due to the
virus, are also lacking. However, one report suggests a twofold increase in symptoms in TVV+
compared to TVV cases, consistent with the virus possibly contributing to enhanced patho-
genesis [43]. Interestingly, TVV+ isolates were also found to elicit greater inflammatory cytokine
production from host cells, in a TLR3-dependent manner [44], suggesting that host recognition
of TVV could enhance inflammation. As an antiviral program of immunity is not likely effective in
clearing Tv, this inflammation may rather contribute to infection-associated pathologies.
Consistent with this hypothesis, structural analysis of TVV revealed a capsid with unusually
large channels posited to facilitate release of the viral genome [45], which could enhance
immunogenicity, since dsRNA is the ligand for TLR-3. Furthermore, metronidazole treatment
was shown to increase inflammatory cytokine induction by TVV+ strains, likely due to Tv lysis
releasing virus and allowing enhanced access to TLR3 [44].

Tv-Associated Mycoplasmas
Mycoplasma hominis (Mh) is another common symbiont of Tv that is highly prevalent in clinical
isolates, as recently reviewed [12]. Mh can increase Tv fitness, as a recent study reports that Mh
is beneficial to the parasite by increasing its growth rate and ATP production by around 50%
[46]. Furthermore, Mh has also been shown to contribute to inflammation [47]. Mh alone, or in
combination with Tv, was shown to upregulate inflammatory cytokine secretion from a

Trends in Parasitology, Month Year, Vol. xx, No. yy 5


TREPAR 1764 No. of Pages 11

monocytic cell line [47]. Similarly, we found that isogenic strains harboring, or cleared of, Mh
show dramatically different induction of key inflammatory cytokines from primary human
monocytes [25]. It is not clear whether enhanced immune responses caused by Mh presence
in Tv promote parasite clearance or merely contribute to inflammation-associated pathologies.

Recently, a new mycoplasma species, Mycoplasma girerdii (Mg) was identified in association
with Tv [48–50]. One study reported this species to be present in 44% of Tv clinical isolates [48],
and notably, 96% of women carrying Mg were positive for Tv [48]. It will be interesting to
determine whether Mg similarly lives within Tv, similarly benefits Tv growth and metabolism, and
whether it can also induce inflammatory cytokine secretion in the context of Tv symbiosis.

Tv Recognition by Host Immunity and Cytokine Induction


It has long been known that the pleiotropic cytokine IL-8 is released in response to Tv infection
[51–54]. Galectin 3 has recently been shown to modulate IL-8 secretion through engagement
of the LG predominant on the parasite surface [55]. Since host-cell galectins are becoming
appreciated as pattern-recognition receptors (PRRs) [56], the LG–Galectin interaction is an
exciting area of future research into parasite-derived immune signaling. Recent reports have
also shown IL-1b, CCL2, IL-17, IL-22, IFNb, IL-6, RANTES, MIP-3a, TNF, and IL-23 to be
associated with Tv infection either in vivo or in vitro [25,47,55,57–59]. Furthermore, parasite-
produced exosomes were shown to induce IL-8 and IL-6 from host cells [32]. However, as
mentioned above, a striking percentage of the cytokine abundance observed in vitro appears to
be due to Tv symbionts. In comparing six strains harboring TVV to three strains without TVV,
Fichorova et al. observed that TVV+ strains were greatly enhanced in their ability to induce
cytokines [44], with strains lacking the virus showing very little, if any, induction. Similarly, Fiori
et al. [47] reported that Mh induced several cytokines from THP-1 macrophages, while Tv alone
only stimulated small amounts of IL-1B. However, the Tv–Mh consortium was able to induce
greater amounts of cytokine than Mh alone, suggesting a synergism between the two microbes
in inducing cytokines. We also observed an Mh-dependent induction of cytokines from primary
human monocytes [25]. Interestingly, each of these studies indicate that no, or very little,
cytokine was induced from the parasite alone; however, these studies may not be sensitive
enough to detect parasite-alone induced cytokine, or may not capture damage-associated
cytokine induction that may occur in vivo. Furthermore, none of these studies considered both
symbionts at that same time. Future work should determine cytokine induction from strains that
are cleared of virus and mycoplasma, as well as having both symbionts present, to determine
the relative contribution of each and the immunogenicity of the parasite alone.

Mature IL-1b secretion is promoted by the inflammasome, a protein complex in the cytosol
downstream of PRRs. Recently, it was suggested that Tv can activate the inflammasome, as
silencing of Capsase-1, and NLRP3, two players in inflammasome signaling, decreased the
amount of IL-1b observed in response to Tv at commensurate levels to their knockdowns [57].
Interestingly, the recently discovered murine gut trichomonad, Tritrichomonas muris, was
observed to induce an inflammasome-dependent program of immunity in the mouse gut
[60]. However, it remains to be determined whether inflammasome activation associated with
trichomonads is dependent on their symbionts.

Parasite-Derived Cytokine Mimic


We also reported that Tv secretes TvMIF, a homolog of human MIF (macrophage migration
inhibitory factor), which can bind the human MIF receptor, and increase host inflammatory
cytokine production [61]. In addition, TvMIF could be involved in promotion of some metastatic
cancers, as we found it to promote cell growth and invasion in a tissue culture model [61].

6 Trends in Parasitology, Month Year, Vol. xx, No. yy


TREPAR 1764 No. of Pages 11

Immune Recognition of Tv Could Increase Spread of HIV


In vitro work observing immune activation by Tv has also shed light on how the parasite might
elicit increased HIV susceptibility. SLPI, an antimicrobial and immunoregulatory peptide in the
vaginal mucosa, was shown to be decreased by 50% in Tv+ compared to Tv controls.
Elevated SLPI levels have been associated with greater control of HIV [62,63], indicating that
reduced SLPI could be a contributing factor in the increased transmission of HIV among Tv+
individuals. Tv-induced inflammatory cytokines, especially RANTES and MIP3a, could con-
tribute to the recruitment of HIV targets. Interestingly, these two cytokines were found to be
released in response to strains harboring TVV only [44], indicating that TVV could act as a
cofactor in increasing Tv-associated HIV spread.

Immune Clearance of Tv, and Tv Evasion of Host Immunity


Tv infection is known to activate the immune system through cytokine induction and inducing
antibody production [64–67]. However, it is not clear why many infections persist, and why
partner reinfection appears to occur in some cases [68]. It remains to be seen how symbionts
contribute to antibody responses to the parasite. However, recent adaptations of flow cyto-
metric analyses, including imaging flow cytometry and of live imaging conditions for super-
high-resolution videos of Tv interactions with immune cells, have allowed better characteriza-
tion of effective antitrichomonal responses, and how parasites may evade these. Potential
evasion strategies are noted below and are outlined in Table 2.

Neutrophil Killing of Tv
Neutrophils have long been known to be a critical cell-type recruited to the site of Tv infection
[42,69]. Additionally, IL-8, which predominantly functions in neutrophil recruitment, is consis-
tently the most abundant cytokine induced by Tv [51–54]. New experiments also show that
supernatants from Tv–epithelial cell cultures induce neutrophil migration over a filter [58]. We
therefore recently tested and confirmed that human neutrophils are able to kill Tv in vitro, and
found this to occur rapidly, within 20 min [70]. To investigate the mechanisms that neutrophils
use to kill Tv, we individually blocked each of the known neutrophil killing pathways at a time:
phagocytosis, extracellular degranulation, and NETosis [71]. We found that neither extra-
cellular degranulation nor NETosis was involved in rapid Tv killing, but that killing was inhibited in
the presence of engulfment inhibitors. Interestingly however, when we imaged this engulfment
process, we found that, in contrast to neutrophils phagocytosing Tv whole, they were found to
be ‘nibbling’ on the parasite, taking up to 25 bites before the parasite died [70] (Figure 2). This is
consistent with killing using trogocytosis [72,73], and is the first demonstration of neutrophils
using trogocytosis to kill a pathogen. We found trogocytosis and Tv killing to be almost
completely inhibited when neutrophils were preincubated with a serine protease inhibitor,

Table 2. Trichomonas vaginalis Immune-Evasive Behaviors


Behavior Function or effector Predicted outcome Refs

Immune cell lysis Kill T cells Dampen adaptive immunity [25]

Kill B cells Dampen humoral immunity [25]

DNase II production Degrade NETs (putative) Evade neutrophil NETosis [74]

Cysteine protease production Degrade antibody Evade neutrophil trogocytosis [70,75]

Clumping Tetraspanin 8, palmitoylated Evade neutrophil trogocytosis [21–23,31,70]


proteins, others unknown Enhance adherence to host cells

Exosome secretion Increase IL-10, decrease Promote immunological tolerance [32,59,78]


IL-8 and IL-17

Trends in Parasitology, Month Year, Vol. xx, No. yy 7


TREPAR 1764 No. of Pages 11

Figure 2. Human Neutrophils Kill Tri-


5 μm chomonas vaginalis Using Trogocy-
tosis. Primary human neutrophils were
labeled with Cell Tracker Deep Red
(magenta), and the T. vaginalis surface
was biotinylated and labelled with strep-
tavidin-488 (green). Neutrophils and para-
sites were allowed to interact at a ratio of
10 neutrophils:1 parasite in the presence
of 10% human AB serum at 37 C. HyVo-
lution, super-resolution live confocal ima-
ging (Leica Microsystems) was used to
take 60 cross-sections of the cells inter-
acting in real time. A cross-section of a
three-dimensional image is shown,
revealing neutrophil trogocytosis (nib-
bling) of the parasite. Location of ‘nib-
bling’ in action is noted with white
arrows. Complete data and methods
are published [70].

indicating that one of four known neutrophil serine proteases may contribute to degradation of
the parasite surface into ‘bites’. We hypothesize that Tv size and motility preclude whole
parasite engulfment; the parasite is rather killed by nibbling. It is not clear why Tv is not
susceptible to killing by extracellular degranulation, or whether the parasite is also eventually
killed using NETosis, a later-stage killing process. However, it is interesting that a DNase was
recently discovered to be part of the Tv constitutive secretome [74].

We also showed that neutrophil trogocytosis and killing of Tv is dependent on human serum,
and that 50% of killing is inhibited in the presence of an Fc-receptor blocking agent [70],
suggesting a role for adaptive immunity in enhancing Tv killing by neutrophils. Interestingly, old
reports identified cysteine proteases of the parasites that showed ability to degrade immuno-
globulin in vitro [75], suggesting that the parasite could evade trogocytosis by degrading
antibody. It is also interesting that the Fc-receptor inhibitor blocked only 50% of killing, and we
found that complement factors also coat the Tv surface, indicating that opsonization by
complement factors could also mediate Tv killing by neutrophils [70]. This is consistent with old
reports of complement-mediated parasite clearance [76].

We found that an average of five neutrophils were needed to achieve killing of a trichomonad,
surrounding and sequestering the parasite before and during nibbling [70]. In this regard, it is
highly likely that Tv clumping discussed above [22,23,31] could serve as a neutrophil-evasive
behavior. It remains to be seen how symbionts contribute to neutrophil killing. Also, Tv has been
reported to induce apoptosis of neutrophils in vitro [77], indicating that killing of neutrophils
could be a strategy that some strains use to evade killing. However, we observed that
neutrophil killing of Tv was completed within 20 min, while induction of neutrophil apoptosis
took hours. However, as neutrophil killing of Tv is contact-dependent, it is possible that large
clumps of Tv may induce apoptosis of neutrophils at a distance.

8 Trends in Parasitology, Month Year, Vol. xx, No. yy


TREPAR 1764 No. of Pages 11

Tv Has Lymphotoxic Activity Outstanding Questions


In a broad-survey of challenge experiments between Tv and different immune-cell subsets, Is there a correlation between Tv
we found that Tv was able to kill T cells and B cells, but not monocytes [25]. We also found strains that display high levels of path-
ogenic behavior in vitro and increased
that, in mixed lymphocyte cultures, B cells were preferentially targeted for killing, and that symptoms and complications in
this killing could be mediated by Tv soluble factors [25]. It is interesting that Tv shows a patients?
preference for killing B cells, the producers of antibody, particularly in light of the fact that
antibody enhances neutrophil killing of the parasite. It therefore appears that the parasite Does genomic variance and differential
gene expression contribute to the
employs a program of humoral immunity evasion by killing B cells, and degrading
broad range of pathogenic properties
antibody [75]. of Tv strains?

Immunomodulation by Tv Exosomes What are the major host cell receptors


Collectively, these data support a model by which inflammatory cytokine induced during Tv for Tv? Do specific protein receptors
and proteoglycans play a role in
infection, induces neutrophil recruitment and attack of Tv. In this regard, it is interesting that Tv
adherence?
exosomes were also found to be immunomodulatory. We demonstrated that pretreatment with
exosomes reduced subsequent IL-8 responses [32]. Moreover, another group found that 2- What induces cytoskeletal rearrange-
day pretreatment of mice with exosomes reduced IL-17 secretion and reduced gross inflam- ment when parasites bind to host
mation score, indicating that exosomes could be a way in which parasites dampen the immune cells? How critical is cytoskeletal reor-
ganization to adherence and
response [78]. Interestingly, IL-17 responses have been reported in Tv+ women [59], and a
cytotoxicity?
major function of IL-17 adaptive responses is neutrophil recruitment. Moreover, the same
report found that exosome pretreatment increased levels of the anti-inflammatory cytokine IL- How is host cell biology modified upon
10 in response to subsequent infection. This is consistent with another report that secretory uptake of Tv exosomes? What mech-
products from Tv induce IL-10 from dendritic cells [79], and with reports of IL-10 induction from anisms mediate host cell–exosome
interaction?
bovine cells with the bovine trichomonad Tritrichomonas foetus [80]. Tv exosome generation
and secretion could therefore be a mechanism that the parasite uses to subvert host immunity, What is the function of the human cyto-
allowing it to gain a foothold. It is not clear whether these anti-inflammatory properties of kine mimic, TvMIF, in the parasite?
exosomes also have beneficial anti-inflammatory roles in the host, or whether they merely
stymie immune clearance of the parasite, allowing more host damage and pathology. Do lactobacilli protect against acquisi-
tion of Tv, or favor displacement of the
parasite from the vagina?
Concluding Remarks and Future Directions
The publication of the Tv genome in 2007 paved the way to identify molecular players in Tv Do lactobacilli show utility as a probi-
pathogenesis [33]. Our recent adaptation of the CRISPR/Cas9 gene editing system in Tv will otic in Tv-infected women?
potentiate more powerful interrogation of molecules that the parasite uses to establish
infection and subvert host immunity [81]. In addition, datasets of the membrane proteome Is the co-occurrence of Tv and type IV
vaginal microbiome due to specific
[16], exosome proteome [32], phosphoproteome [82], secretome [74], palmitoylated prote-
metabolic conditions of type IV micro-
ome [22], and genes regulated epigenetically [23,24], are now all available for researchers to biome that favor Tv growth?
mine. However, a key limitation remains incomplete annotation of the genome, abundant
repetition, predominant transposable elements [83], and absent genome sequences for more Do Tv strains cleared of all symbionts,
virulent parasite strains. Lack of robust genomic DNA models continue to hinder the use of and lacking any host-lytic behavior,
display any immunogenicity?
RNAseq to identify and interrogate genes that are regulated upon host contact. Flow
cytometric analyses of trichomonads has also allowed better quantitation for in vitro patho-
Do TVV and Mycoplasma symbionts
genicity assays [25,70,84]. Implementation of the oxygen-independent fluorophore, iLOV, increase adherence and cytolytic
has improved fluorescent tagging in the microaerophilic organism [85], and should facilitate capacity of Tv in vitro and in vivo?
improved in vivo imaging and tracking. However, a major limitation to studying pathogenesis
has been lack of a robust mouse model that sustains adequate Tv titers. The variation in Does the recently identified Tv symbi-
ont, Mycoplasma girerdii, contribute to
microbiota and pH between human and mouse vaginas likely contributes to this. Future
Tv fitness, and does it affect the path-
efforts to ‘humanize’ the mouse vaginal environment using hormonal and microbial supple- ogenesis or immunogenicity of Tv?
ments may help to overcome this considerable challenge We outline some important open
questions (see Outstanding Questions) that need to be addressed to better characterize Tv What is the effectiveness of neutrophil
pathogenesis and inform therapeutic and management strategies for this highly prevalent trogocytosis in killing Tv in vivo, and

infection.

Trends in Parasitology, Month Year, Vol. xx, No. yy 9


TREPAR 1764 No. of Pages 11

what trogocytic-evasive behaviors


References may the parasite employ?
1. Leitsch, D. (2016) Recent advances in the Trichomonas vaginalis Published online February 2018. http://dx.doi.org/10.1074/
field. F1000Research 5, 162 mcp.RA117.000018
2. Hirt, R.P. and Sherrard, J. (2015) Trichomonas vaginalis origins, 23. Pachano, T. et al. (2017) Epigenetics regulates transcription and Is trogocytic killing of Tv an adaptive
molecular pathobiology and clinical considerations. Curr. Opin. pathogenesis in the parasite Trichomonas vaginalis. Cell. Micro- immune mechanism or can it be
Infect. Dis. 28, 72–79 biol. Published online June 2017. http://dx.doi.org/10.1111/ employed as an innate immune
3. Secor, W.E. et al. (2014) Neglected parasitic infections in the cmi.12716
mechanism?
United States: trichomoniasis. Am. J. Trop. Med. Hyg. 90, 800– 24. Song, M.J. et al. (2017) Epigenome mapping highlights chroma-
804 tin-mediated gene regulation in the protozoan parasite Tricho-
4. World Health Organization (2011) Prevalence and Incidence of monas vaginalis. Sci. Rep. 7, 45365 What are the downstream immunolog-
Selected Sexually Transmitted Infections, Chlamydia trachoma- 25. Mercer, F. et al. (2016) Leukocyte lysis and cytokine induction by ical consequences of neutrophil killing
tis, Neisseria gonorrhoeae, Syphilis, and Trichomonas vaginalis: the human sexually transmitted parasite Trichomonas vaginalis. of Tv using trogocytosis; does it pro-
Methods and Results Used by the WHO to Generate 2005 PLoS Negl. Trop. Dis. 10, e0004913 mote or decrease inflammation?
Estimates, WHO 26. Puente-Rivera, J. et al. (2017) The 50 kDa metalloproteinase
5. Fichorova, R.N. (2009) Impact of T. vaginalis infection on innate TvMP50 is a zinc-mediated Trichomonas vaginalis virulence fac-
immune responses and reproductive outcome. J. Reprod. Immu- tor. Mol. Biochem. Parasitol. 217, 32–41 What effector proteins does Tv use to
nol. 83, 185–189 27. Midlej, V. and Benchimol, M. (2010) Trichomonas vaginalis kills
kill T cells and B cells?
6. Mielczarek, E. and Blaszkowska, J. (2016) Trichomonas vaginalis: and eats – evidence for phagocytic activity as a cytopathic effect.
pathogenicity and potential role in human reproductive failure. Parasitology 137, 65–76 What are the conditions needed to
Infection 44, 447–458 28. Fiori, P.L. et al. (1999) The flagellated parasite Trichomonas establish a mouse model that will sus-
7. Tao, L. et al. (2014) Prevalence and risk factors for cervical vaginalis: new insights into cytopathogenicity mechanisms.
tain adequate titers of Tv to study viru-
neoplasia: a cervical cancer screening program in Beijing. Microbes Infect. 1, 149–156
BMC Public Health 14, 1185
lence and cellular immune responses
29. Rendon-Maldonado, J.G. et al. (1998) Trichomonas vaginalis: in
in vivo?
8. Ghosh, I. et al. (2017) Association between high risk human vitro phagocytosis of lactobacilli, vaginal epithelial cells, leuko-
papillomavirus infection and co-infection with Candida spp. cytes, and erythrocytes. Exp. Parasitol. 89, 241–250
and Trichomonas vaginalis in women with cervical premalignant 30. Pereira-Neves, A. and Benchimol, M. (2007) Phagocytosis by
and malignant lesions. J. Clin. Virol. 87, 43–48 Trichomonas vaginalis: new insights. Biol. Cell 99, 87–101
9. Nakubulwa, S. et al. (2015) Genital infections and risk of prema- 31. Coceres, V.M. et al. (2015) The C-terminal tail of tetraspanin
ture rupture of membranes in Mulago Hospital, Uganda: a case proteins regulates their intracellular distribution in the parasite
control study. BMC Res. Notes 8, 573 Trichomonas vaginalis. Cell. Microbiol. 17, 1217–1229
10. Hamilton, H. et al. (2018) Trichomonas vaginalis brain abscess in 32. Twu, O. et al. (2013) Trichomonas vaginalis exosomes deliver
a neonate. Clin. Infect. Dis. 66, 604–607 cargo to host cells and mediate hostratioparasite interactions.
11. Sutton, M. et al. (2007) The prevalence of Trichomonas vaginalis PLoS Pathog. 9, e1003482
infection among reproductive-age women in the United States, 33. Carlton, J.M. et al. (2007) Draft genome sequence of the sexually
2001–2004. Clin. Infect. Dis. 45, 1319–1326 transmitted pathogen Trichomonas vaginalis. Science 315, 207–
12. Fichorova, R. et al. (2017) Trichomonas vaginalis infection in 212
symbiosis with Trichomonasvirus and Mycoplasma. Res. Micro- 34. Kusdian, G. et al. (2013) The actin-based machinery of Tricho-
biol. 168, 882–891 monas vaginalis mediates flagellate-amoeboid transition and
13. Ryan, C.M. et al. (2011) Trichomonas vaginalis: current under- migration across host tissue. Cell. Microbiol. 15, 1707–1721
standing of host–parasite interactions. Essays Biochem. 51, 161– 35. Nievas, Y.R. et al. (2017) Membrane-shed vesicles from the
175 parasite Trichomonas vaginalis: characterization and their asso-
14. Addis, M.F. et al. (2000) Host and tissue specificity of Trichomo- ciation with cell interaction. Cell. Mol. Life Sci. 75, 2211–2226
nas vaginalis is not mediated by its known adhesion proteins. 36. Ravel, J. et al. (2011) Vaginal microbiome of reproductive-age
Infect. Immun. 68, 4358–4360 women. Proc. Natl. Acad. Sci. U. S. A. 108, 4680–4687
15. Okumura, C.Y. et al. (2008) Galectin-1 on cervical epithelial cells is 37. Brotman, R.M. et al. (2012) Association between Trichomonas
a receptor for the sexually transmitted human parasite Trichomo- vaginalis and vaginal bacterial community composition among
nas vaginalis. Cell. Microbiol. 10, 2078–2090 reproductive-age women. Sex. Transm. Dis. 39, 807–812
16. de Miguel, N. et al. (2010) Proteome analysis of the surface of 38. El Sayed Zaki, M. et al. (2010) Correlation of Trichomonas vag-
Trichomonas vaginalis reveals novel proteins and strain-depen- inalis to bacterial vaginosis: a laboratory-based study. J. Infect.
dent differential expression. Mol. Cell. Proteom. 9, 1554–1566 Dev. Ctries. 4, 156–163
17. Lustig, G. et al. (2013) Trichomonas vaginalis contact-dependent 39. Fichorova, R.N. et al. (2013) The villain team-up or how Tricho-
cytolysis of epithelial cells. Infect. Immun. 81, 1411–1419 monas vaginalis and bacterial vaginosis alter innate immunity in
18. Munoz, C. et al. (2012) A protein phosphatase 1 gamma concert. Sex. Transm. Infect. 89, 460–466
(PP1gamma) of the human protozoan parasite Trichomonas vag- 40. Phukan, N. et al. (2013) The adherence of Trichomonas vaginalis
inalis is involved in proliferation and cell attachment to the host to host ectocervical cells is influenced by lactobacilli. Sex.
cell. Int. J. Parasitol. 42, 715–727 Transm. Infect. 89, 455–459
19. Miranda-Ozuna, J.F. et al. (2016) The glycolytic enzyme triose- 41. Ozcelik, S. et al. (2012) The cytopathic effects of Trichomonas
phosphate isomerase of Trichomonas vaginalis is a surface-asso- vaginalis on fibroblast cell culture alone and with C. albicans and
ciated protein induced by glucose that functions as a laminin- and E. coli. Turkiye Parazitol. Derg. 36, 193–197
fibronectin-binding protein. Infect. Immun. 84, 2878–2894
42. Lazenby, G.B. et al. (2013) Correlation of leukorrhea and Tricho-
20. Rendon-Gandarilla, F.J. et al. (2013) The TvLEGU-1, a legumain- monas vaginalis infection. J. Clin. Microbiol. 51, 2323–2327
like cysteine proteinase, plays a key role in Trichomonas vaginalis
43. Jehee, I. et al. (2017) Direct detection of Trichomonas vaginalis
cytoadherence. BioMed Res. Int. 2013, 561979
virus in Trichomonas vaginalis positive clinical samples from the
21. Riestra, A.M. et al. (2015) A Trichomonas vaginalis rhomboid Netherlands. J. Virol. Methods 250, 1–5
protease and its substrate modulate parasite attachment and
44. Fichorova, R.N. et al. (2012) Endobiont viruses sensed by the
cytolysis of host cells. PLoS Pathog. 11, e1005294
human host – beyond conventional antiparasitic therapy. PLoS
22. Nievas, Y.R. et al. (2018) Protein palmitoylation plays an important One 7, e48418
role in Trichomonas vaginalis adherence. Mol. Cell. Proteom.

10 Trends in Parasitology, Month Year, Vol. xx, No. yy


TREPAR 1764 No. of Pages 11

45. Parent, K.N. et al. (2013) Structure of a protozoan virus from the 65. Kaur, S. et al. (2008) Antitrichomonas IgG, IgM, IgA, and IgG
human genitourinary parasite Trichomonas vaginalis. mBio 4, subclass responses in human intravaginal trichomoniasis. Para-
e00056-13 sitol. Res. 103, 305–312
46. Margarita, V. et al. (2016) Symbiotic association with Myco- 66. Bastida-Corcuera, F.D. et al. (2013) Antibodies to Trichomonas
plasma hominis can influence growth rate, ATP production, cytol- vaginalis surface glycolipid. Sex. Transm. Infect. 89, 467–472
ysis and inflammatory response of Trichomonas vaginalis. Front. 67. Ton Nu, P.A. et al. (2015) Kinetics of circulating antibody
Microbiol. 7, 953 response to Trichomonas vaginalis: clinical and diagnostic impli-
47. Fiori, P.L. et al. (2013) Association of Trichomonas vaginalis with cations. Sex. Transm. Infect. 91, 561–563
its symbiont Mycoplasma hominis synergistically upregulates the 68. Forna, F. and Gulmezoglu, A.M. (2003) Interventions for treating
in vitro proinflammatory response of human monocytes. Sex. trichomoniasis in women. Cochrane Database Syst. Rev.
Transm. Infect. 89, 449–454 CD000218
48. Fettweis, J.M. et al. (2014) An emerging mycoplasma associated 69. Rein, M.F. et al. (1980) Trichomonacidal activity of human poly-
with trichomoniasis, vaginal infection and disease. PLoS One 9, morphonuclear neutrophils: killing by disruption and fragmenta-
e110943 tion. J. Infect. Dis. 142, 575–585
49. Martin, D.H. et al. (2013) Unique vaginal microbiota that includes 70. Mercer, F. et al. (2018) Neutrophils kill the parasite Trichomonas
an unknown Mycoplasma-like organism is associated with Trich- vaginalis using trogocytosis. PLoS Biol. 16, e2003885
omonas vaginalis infection. J. Infect. Dis. 207, 1922–1931
71. Kolaczkowska, E. and Kubes, P. (2013) Neutrophil recruitment
50. Costello, E.K. et al. (2017) Candidatus Mycoplasma girerdii rep- and function in health and inflammation. Nat. Rev. Immunol. 13,
licates, diversifies, and co-occurs with Trichomonas vaginalis in 159–175
the oral cavity of a premature infant. Sci. Rep. 7, 3764
72. Ralston, K.S. et al. (2014) Trogocytosis by Entamoeba histolytica
51. Ryu, J.S. et al. (2004) Production of interleukin-8 by human contributes to cell killing and tissue invasion. Nature 508, 526–
neutrophils stimulated with Trichomonas vaginalis. Infect. Immun. 530
72, 1326–1332
73. Gilmartin, A.A. et al. (2017) Inhibition of amebic lysosomal acidifi-
52. Shaio, M.F. et al. (1995) Generation of interleukin-8 from human cation blocks amebic trogocytosis and cell killing. mBio 8,
monocytes in response to Trichomonas vaginalis stimulation. e01187-17
Infect. Immun. 63, 3864–3870
74. Stafkova, J. et al. (2018) Dynamic secretome of Trichomonas
53. Shaio, M.F. et al. (1994) Monocyte-derived interleukin-8 involved vaginalis: case study of beta-amylases. Mol. Cell. Proteom. 17,
in the recruitment of neutrophils induced by Trichomonas vagi- 304–320
nalis infection. J. Infect. Dis. 170, 1638–1640
75. Provenzano, D. and Alderete, J.F. (1995) Analysis of human
54. Jarrett, O.D. et al. (2015) T. vaginalis infection is associated with immunoglobulin-degrading cysteine proteinases of Trichomonas
increased IL-8 and TNFr1 levels but with the absence of CD38 vaginalis. Infect. Immun. 63, 3388–3395
and HLADR activation in the cervix of ESN. PLoS One 10,
76. Gillin, F.D. and Sher, A. (1981) Activation of the alternative com-
e0130146
plement pathway by Trichomonas vaginalis. Infect. Immun. 34,
55. Fichorova, R.N. et al. (2016) Trichomonas vaginalis lipophospho- 268–273
glycan exploits binding to Galectin-1 and -3 to modulate epithelial
77. Song, H.O. et al. (2008) Trichomonas vaginalis: reactive oxygen
immunity. J. Biol. Chem. 291, 998–1013
species mediates caspase-3 dependent apoptosis of human
56. Vasta, G.R. (2012) Galectins as pattern recognition receptors: neutrophils. Exp. Parasitol. 118, 59–65
structure, function, and evolution. Adv. Exp. Med. Biol. 946, 21–
78. Olmos-Ortiz, L.M. et al. (2017) Trichomonas vaginalis exosome-
36
like vesicles modify the cytokine profile and reduce inflammation
57. Gu, N.Y. et al. (2016) Trichomonas vaginalis induces IL-1beta in parasite-infected mice. Parasite Immunol. 39, e12426
production in a human prostate epithelial cell line by activating the
79. Song, M.J. et al. (2015) Modulation of dendritic cell function by
NLRP3 inflammasome via reactive oxygen species and potas-
Trichomonas vaginalis-derived secretory products. BMB Rep.
sium ion efflux. Prostate 76, 885–896
48, 103–108
58. Seo, M.Y. et al. (2014) Inflammatory response of prostate epithe-
80. Vilela, R.C. and Benchimol, M. (2013) IL-10 release by bovine
lial cells to stimulation by Trichomonas vaginalis. Prostate 74,
epithelial cells cultured with Trichomonas vaginalis and Tritricho-
441–449
monas foetus. Mem. Inst. Oswaldo Cruz 108, 110–112
59. Makinde, H.M. et al. (2013) IL-22 levels are associated with
81. Janssen, B.D. et al. (2018) CRISPR/Cas9-mediated gene modi-
Trichomonas vaginalis infection in the lower genital tract. Am.
fication and gene knock out in the human-infective parasite
J. Reprod. Immunol. 70, 38–44
Trichomonas vaginalis. Sci. Rep. 8, 270
60. Chudnovskiy, A. et al. (2016) Host–protozoan interactions protect
82. Yeh, Y.M. et al. (2013) Phosphoproteome profiling of the sexually
from mucosal infections through activation of the inflammasome.
transmitted pathogen Trichomonas vaginalis. J. Microbiol. Immu-
Cell 167, 444–456 e14
nol. Infect. 46, 366–373
61. Twu, O. et al. (2014) Trichomonas vaginalis homolog of macro-
83. Bradic, M. et al. (2014) The Tc1/mariner transposable element
phage migration inhibitory factor induces prostate cell growth,
family shapes genetic variation and gene expression in the protist
invasiveness, and inflammatory responses. Proc. Natl. Acad. Sci.
Trichomonas vaginalis. Mob. DNA 5, 12
U. S. A. 111, 8179–8184
84. Brooks, A.E. et al. (2013) An improved quantitative method to
62. Taborda, N.A. et al. (2012) Higher SLPI expression, lower immune
assess adhesive properties of Trichomonas vaginalis to host
activation, and increased frequency of immune cells in a cohort of
vaginal ectocervical cells using flow cytometry. J. Microbiol.
Colombian HIV-1 controllers. J. Acquir. Immune Defic. Syndr. 60,
Methods 92, 73–78
12–19
85. Wang, S.E. et al. (2017) The fluorescent protein iLOV outperforms
63. Pillay, K. et al. (2001) Secretory leukocyte protease inhibitor in
eGFP as a reporter gene in the microaerophilic protozoan Trich-
vaginal fluids and perinatal human immunodeficiency virus type 1
omonas vaginalis. Mol. Biochem. Parasitol. 216, 1–4
transmission. J. Infect. Dis. 183, 653–656
64. Paintlia, M.K. et al. (2002) Specific IgA response, T-cell subtype
and cytokine profile in experimental intravaginal trichomoniasis.
Parasitol. Res. 88, 338–343

Trends in Parasitology, Month Year, Vol. xx, No. yy 11

You might also like