Skip to main content
Faculty Reviews logoLink to Faculty Reviews
. 2021 Mar 4;10:26. doi: 10.12703/r/10-26

Recent advances in the molecular biology of the protist parasite Trichomonas vaginalis

David Leitsch 1,*
PMCID: PMC7946386  PMID: 33718943

Abstract

Trichomonas vaginalis is an anaerobic/microaerophilic protist parasite which causes trichomoniasis, one of the most prevalent sexually transmitted diseases worldwide. T. vaginalis not only is important as a human pathogen but also is of great biological interest because of its peculiar cell biology and metabolism, in earlier times fostering the erroneous notion that this microorganism is at the root of eukaryotic evolution. This review summarizes the major advances in the last five years in the T. vaginalis field with regard to genetics, molecular biology, ecology, and pathogenicity of the parasite.

Keywords: Trichomonas vaginalis, Sexually Transmitted Diseases, Microbiome

Introduction

The protist parasite Trichomonas vaginalis is the causative agent of trichomoniasis, one of the most prevalent sexually transmitted diseases worldwide. In the most recent estimate by the World Health Organization1, 110 million infections worldwide were ascribed to T. vaginalis in 2016; that is somewhat less than ascribed to chlamydia but much more than to gonorrhea and syphilis. Symptoms of trichomoniasis are usually more severe in women than in men and affect the vagina, the cervix, or the urethra or a combination of these. Vaginal pruritus and odorous vaginal discharge are the most common sequelae, but severe inflammation of the cervix can also occur. In men, T. vaginalis can cause urethritis and prostatitis, albeit with lower frequency. Trichomoniasis can persist for years and sour the life of those infected, but it also increases the risk for adverse pregnancy outcomes and the risk of acquiring HIV2. It is now firmly established that underlying trichomoniasis increases the risk to acquire HIV by 1.5- to 2-fold3. This poses serious problems in countries where both pathogens are highly endemic (for example, in the southern part of Africa). T. vaginalis has also been suggested to predispose for cervical cancer and prostate cancer, although there is far less support for this supposed link46 than with HIV.

However, T. vaginalis not only is of medical importance but also is a fascinating microorganism in its own right. Once considered a primordial eukaryote because of its anaerobic metabolism and the absence of mitochondria, it is now known to host hydrogenosomes7, hydrogen gas–producing organelles derived from mitochondria. Furthermore, T. vaginalis, like other trichomonads, has a huge haploid genome of 160 Mb which contains a very large number of transposable elements (TEs)8. T. vaginalis is commonly believed to be asexual and to exist only as a trophozoite-stage flagellate, but the recent discovery of meiosis-specific genes9, amongst other features typical of sexual organisms, and of a cyst-like stage with a cell wall10 might lead to a revaluation of these tenets.

The present overview focuses on the major scientific advances in the T. vaginalis field in the last five years and covers genetics, biochemistry, cell biology, ecology, and infection biology of the parasite. Drug testing and resistance will also be discussed. However, more clinical aspects of trichomoniasis, such as epidemiology, diagnosis, and management, were surveyed recently in another review published at F1000 Research11 and are not included here.

Genome and gene expression

As compared with other protists, T. vaginalis has an enormously large genome of 160 Mb in length12, which encodes about 60,000 protein-coding genes. A surprisingly large portion of the genome consists of TEs13,14, and as many as 40,000 TEs come from several families. This has slowed down efforts to assemble the genome because of the immensely high number of sequence repeats. Still, the availability of the genome sequence, if not fully assembled, has allowed substantial scientific advances on T. vaginalis evolution on the one hand and on its genetic machinery on the other.

Strikingly, T. vaginalis has a predicted number of only 62 introns. A detailed study on introns in T. vaginalis confirmed the existence of 32 genes with introns, whereas 18 candidates were found to be non-functional as they were not removed from the respective transcripts15. Eleven new introns which group into two different types with regard to sequence and splice motifs were identified in this study. Transcription of protein-encoding genes is commonly initiated at an initiator element16 or two alternative promoter elements, M3 and M5, respectively17. Interestingly, the M3 element resembles a Myb recoginition element and was found to be bound by a novel transcription factor with a Myb-like DNA-binding domain17. Surprisingly, however, a TATA box was found to be missing from promoters of protein-encoding genes. T. vaginalis nonetheless does encode two TATA box-binding proteins (TBPs) which bind to initiator-binding protein 39 (IBP39)16 and to DNA as determined in electrophoretic mobility shift assays18. However, it seems that binding to DNA by these T. vaginalis TBPs is unspecific19. Instead, they might have a role in the transcription of spliceosomal U6 snRNA. Also of interest is the mRNA capping apparatus in T. vaginalis which is structurally similar to its counterparts in metazoans and plants rather than to those in other protists20.

Transcription rates not only are determined by sequences proximal to the transcription start site but also are dependent on epigenetic regulation. Consequently, epigenetic regulation in T. vaginalis was addressed in a number of studies. N6-methyladenine (6mA) was found to be the major DNA methylation mark in T. vaginalis21. It occurs frequently in intergenic regions (in 94% of all sequenced methylated DNA stretches) and probably localizes to chromatin loops, indicating a role of 6mA in the regulation of gene expression. Also, histone acetylation was identified as an important modulator of gene expression in two independent studies22,23. Indeed, T. vaginalis has a large arsenal of histone deacetylases (HDACs) of the Sir2 type and of histone acetyl transferases22. Methylation and acetylation of histone 3 (H3K27Ac and H3K4Me3, respectively) near transcription initiation sites were found to be positively correlated with transcription rate, and the HDAC inhibitor trichostatin A was found to strongly affect gene expression23. Independently of the transcription rate, mRNA levels can also be regulated, a process termed RNA interference (RNAi). For RNAi, the presence of small RNAs (sRNAs), which are complementary to their target mRNA and recruit the so-called RNA-induced silencing complex, is a prerequisite. Importantly, two Argonaute proteins (T. vaginalis AGO1 and 2), which constitute central components of this complex, have been found in the genome14. Furthermore, a large number of sRNAs of the PIWI domain–interacting type (piRNA) were found. Arguably, T. vaginalis AGO1 and 2 interact with sequence-specific piRNA to degrade transposase mRNAs as encoded by myriads of TEs in the T. vaginalis genome14.

The last step of expression of protein-coding genes (that is, translation) has also been studied in recent years. Translation efficiency of transcripts was found to be tuned by stretches of low sequence complexity at the 5′ end of the mRNA, followed by a highly structured region more downstream24. The newly introduced iLOV fluorescent protein25 was used as a marker to gauge the effect of the prospective regulatory sequences on protein expression.

In addition to the fundamental research on T. vaginalis genetics as stated above, the repertoire of genetic tools in this parasite was refined and enlarged. In a comprehensive study, reference genes for quantitative polymerase chain reaction studies were evaluated26. Importantly, the frequently chosen glyceraldehyde 3-phosphate dehydrogenase (GAPDH) gene was identified as highly unreliable when exposing T. vaginalis to various conditions due to the strongly variable expression levels observed. In contrast, the genes for DNA topisomerase 2, α-tubulin, and actin were found to be much more suitable; the best dual combination was DNA topisomerase 2 and α-tubulin. Hopefully, these results will be taken into account in future studies on gene expression in T. vaginalis. Furthermore, RNAi was applied successfully for the first time in T. vaginalis27, although downregulation of the respective transcripts was rather low (33–72% at the most), which might be insufficient for many scientific questions. As a word of caution, the feasibility of the method still awaits confirmation from other researchers in the field. In the meantime, gene knockouts using CRISPR/Cas-9 technology might prove highly instrumental. In a pilot study28, CRISPR/Cas-9 was successfully applied to knock out the non-essential genes for ferredoxin-1 and migration inhibitory factor. Importantly, the efficiency of the necessary transfection procedures was highly improved when a newly developed nucleofection protocol was applied.

Cell biology and biochemistry

T. vaginalis diverges strongly from most other protists by having an anaerobic metabolism and by hosting an unusual organelle, the hydrogenosome. These issues have attracted considerable interest throughout the last 50 years, and the last five years have yielded further important insights into the physiology of this parasite.

The hydrogenosome was originally believed to have evolved from a bacterial endosymbiont in a trichomonad progenitor but is now known to derive from mitochondria29. Many biological processes relating to hydrogenosomal function, however, remained incompletely understood in the past. Protein import into hydrogenosomes, for example, is of pivotal importance as no protein translation takes place within the organelle. Some hydrogenosomal proteins have an N-terminal targeting sequence (NTS) but others do not30,31. Moreover, in many cases, the NTS seems to be dispensable for protein import, and even ectopic import into yeast mitochondria without NTS is possible31,32. This indicates that predominantly internal signals earmark certain proteins for import into hydrogenosomes. In the case of tail-anchored (TA) proteins which localize to the T. vaginalis hydrogenosome33, the responsible amino acid sequences have been studied in more detail: in the C-terminal region next to the trans-membrane domain of TA proteins, the net charge must be positive to ensure reliable import. The translocase of the outer membrane (TOM) complex is mainly responsible for conducting protein import into the hydrogenosome34. The translocation channel is mainly formed by TvTOM40-2 which is divergent from TOM40 proteins from other eukaryotes but which nevertheless can partially complement for TOM40 mutations in yeast34. Despite its apparent simplicity, the protein import machinery of the hydrogenosome is highly effective. In recent decades, there was an intense discussion about whether certain highly expressed hydrogenosomal proteins such as pyruvate:ferredoxin oxidoreductase (PFOR), malic enzyme, and succinyl-coA synthetase (SCS) are also trafficked to the cell surface to act there as adhesin proteins, facilitating adhesion to host epithelium. This issue has now been conclusively settled by a recent study which showed that these enzymes are exclusively trafficked into the hydrogenosomes35. However, trafficking of proteins to membrane compartments in general (that is, to destinations including the hydrogenosome but not being restricted to it) seems to have a different underlying mechanism as demonstrated for the cyclophilins TvCyP1 and TvCyP236.

The major hydrogenosomal pathway deploys PFOR, SCS, malic enzyme, and hydrogenase for the breakdown of pyruvate and malate to carbon dioxide and hydrogen gas. But also pathways for amino acid catabolism are hosted in the organelle. These become more important under glucose restriction, especially the arginine dihydrolase pathway37. The arginine dehydrolase pathway also indirectly leads to the formation of nitric oxide (NO) levels and this has a stabilizing effect on hydrogenosomal membranes under glucose restriction38. Several hydrogenosomal enzymes (for example, PFOR and hydrogenase) are sensitive to oxygen and its derivatives such as hydrogen peroxide and superoxide radical anion and need to be protected by appropriate antioxidant enzymes (reviewed in 39). The most recently discovered antioxidant enzyme of the hydrogenosome is the osmotically inducible protein C (OsmC), which detoxifies peroxides after receiving electrons from lipoate via the glycine decarboxylase L and H proteins40.

Pyruvate and malate are broken down in the hydrogenosome but derive from glycolysis taking place in the cytosol. Glucose, in turn, is obtained mostly from the breakdown of intracellular glycogen41 or of glycogen from the vaginal environment. T. vaginalis secretes several glycosidases42 (including most notably β-amylase43,44), which break down glycogen to maltose. Maltose is further broken down to glucose by another glucosidase45, followed by glucose import into the cell. Of course, carbohydrate uptake is not the only form of nutrient uptake T. vaginalis is capable of. The hydrolysis of nucleotides from the host by T. vaginalis ectonucleoside triphosphate diphosphohydrolase (E-NTPDase) and the subsequent uptake of the resulting nucleosides have been studied in a suite of studies in recent years46,47.

Trichomonas vaginalis as a member of the vaginal microbiome

The main habitat of T. vaginalis, the human vagina, accommodates a highly complex microbiome48 which strongly influences the chances of T. vaginalis to successfully colonize its host. The microbiota of the vagina are roughly grouped into five community state types (CSTs) of which four (CST-I, -II, -III, and -V) are dominated by lactobacilli, whereas CST-IV is dominated by anaerobic bacteria such as Gardnerella vaginalis and mollicutes such as Mycoplasma spp.49. Lactobacilli preserve an acidic environment (pH ~4.5), whereas CST-IV leads to a higher vaginal pH. T. vaginalis is predominantly associated with CST-IV50. The presence of certain anaerobic bacteria (that is, Prevotella amnii and Sneathia sanguinegens) is positively correlated with the acquisition of T. vaginalis51. Interestingly, representatives of the CST-IV microbiome enhance adherence of T. vaginalis to epithelial cells through formation of a biofilm52 and both can cooperatively compromise the integrity of the vaginal epithelium by disrupting intercellular tight junctions53. In contrast, lactobacilli clearly antagonize T. vaginalis. In a revelatory study, Lactobacillus gasseri strain ATCC 985754 was shown to strongly inhibit adhesion of T. vaginalis to host cells in a contact-dependent manner. In fact, L. gasseri ATCC 9857 could even displace already-adhering trichomonads from vaginal epithelial cells. Arguably, aggregation-promoting factor 2 (APF-2) as encoded by the lactobacilli is responsible for this remarkable capability. T. vaginalis, however, is by no means defenseless. It expresses nine peptidoglycan hydrolases of the NlpC/P60 family which can kill bacteria55. To date, these enzymes have been tested only on Escherichia coli and it will be interesting to learn more about their activity against vaginal bacteria.

Even if most details of the interplay of T. vaginalis with the microbiota of the vagina remain to be discovered, the association of T. vaginalis with mollicutes, predominantly with Mycoplasma spp., has been studied in considerable detail. The proportion of T. vaginalis isolates harboring Mycoplasma hominis varies strongly depending on geographic origin but usually is substantial and can be even higher than 80%56. Thus, coinfections with T. vaginalis and M. hominis are very common and therefore should be taken into account by all means when speaking of trichomoniasis. The interactions of T. vaginalis and M. hominis are mutually beneficial. Most importantly, the growth rate of T. vaginalis is greatly enhanced (that is, by 20%) if intracellular M. hominis is present57. This is probably due to the arginine dihydrolase (ADH) pathway which is shared by the two microorganisms and which leads to the production of more ATP when L-arginine is present as a substrate. Indeed, intracellular M. hominis was shown to enhance ATP production substantially after supplementation with L-arginine whereas in symbiont-free T. vaginalis supplementation of arginine had only a minimal effect. This also has implications for the host defense because NO production by immune cells is greatly diminished if L-arginine is scavenged by the T. vaginalis/M. hominis consortium. There are probably many more interactions, and there is some evidence that M. hominis can alter gene expression in T. vaginalis to a certain extent58. Like T. vaginalis, M. hominis has been linked to adverse pregnancy outcomes59. There is an indication that the cohabitation with T. vaginalis promotes this, especially when T. vaginalis is eliminated by metronidazole treatment and liberated M. hominis infects host tissue60,61. In addition to M. hominis, another Mycoplasma species is closely associated with T. vaginalis: Mycoplasma girerdii. This species was discovered only recently62, possibly because it might exist merely as an intracellular symbiont of T. vaginalis. A recent study, however, casts doubt on this notion51. In any case, M. girerdii is at least as common as M. hominis63 but its pathogenic potential remains to be elucidated.

In addition to hosting bacterial symbionts, T. vaginalis hosts T. vaginalis virus (TVV), which is grouped into four strains (TVV1–4) and belongs to the family of Totiviridae64. Little is known about the life cycle of TVV but it has been suggested to exacerbate trichomoniasis by enhancing the immune response65, and a recent study showed that the presence of TVV mitigates the response of vaginal epithelial cells to trichomonads66. However, another recent study of 355 T. vaginalis isolates, of which 40% hosted TVV, did not find any association of TVV and clinical symptoms67.

Pathogenesis of trichomoniasis

After a long period of scientific neglect, the pathogenesis of trichomoniasis is now finally receiving the attention it deserves. In recent years, pertinent studies have been forthcoming in increasing numbers, covering numerous aspects ranging from host response to parasite virulence factors (reviewed in 68).

Arguably the most significant response of the host to T. vaginalis is the production of cytokines by immune cells at the site of infection. Mainly interleukin 1 (IL-1), IL-6, IL-8, and IL-176971 are induced and this is characteristic of a pro-inflammatory response. Interestingly, the cytokine profile was influenced by the presence of the M. hominis as IL-1 and IL-6 levels were several-fold higher after exposure to T. vaginalis G3 with the endosymbiont as compared with the same strain without69. Notably, IL-6 was reported to induce polarization of THP-1–derived macrophages into M2-type macrophages72 and IL-1 production is linked to activation of the NLRP3 inflammasome in macrophages resulting in processing of precursor IL-1β to bioactive IL-173. IL-1, in turn, can induce pyroptotic cell death in macrophages73. The host cell response to T. vaginalis further centers on Toll-like receptor 2 (TLR2), whose expression is also triggered by T. vaginalis71,74. Consequently, in TLR2−/− mouse macrophages, immune mediators such as p38, ERK, and p65 NF-κB were found to be phosphorylated to a lesser extent after stimulation with T. vaginalis75. In contrast to these observations, intraepithelial dendritic cells and regulatory T cells were shown to react to exposure with T. vaginalis or one of its major antigens, actinin-2, with the expression of IL-10, which is an anti-inflammatory cytokine75. Cytokine production, however, is not restricted to the human host as T. vaginalis also produces cytokines to ensure its survival. The parasite secretes a homologue of human macrophage migration inhibition factor (TvMIF), which increases survivability under serum starvation several-fold76. Serum contains several essential nutrients for T. vaginalis, such as amino acids, lipids, and precursors of nucleotides. T. vaginalis also secretes leukotriene B4 (LTB4), which induces exocytotic degranulation in mast cells, thereby promoting tissue inflammation77.

T. vaginalis interacts with the host tissue by shedding extracellular vesicles (EVs) carrying cargo which promotes the infection process (thoroughly reviewed in 78). Two types of T. vaginalis EVs have been identified: exosomes (50–150 nm)79 and microvesicles80, which are considerably larger (100–1000 nm). The former derive from intracellular multivesicular bodies whereas the latter are shed from the cell membrane. The vesicles contain protein factors involved in mediating adherence, such as tetraspanins or BspA-domain proteins, and arguably in tuning the host’s response. Notably, the content of the small vesicles was found to be altered in the presence of TVV66. In addition to proteins, EVs contain RNA81; that is, mainly tRNA fragments or tRNA halves (tsRNA), respectively, which have also been described to be part of the cargo in EVs of trypanosomatid parasites82,83. The precise role of tsRNA in host–parasite interactions, however, remains to be elucidated. Finally, the uptake of exosomes by host cells is mediated by 4-α-glucanotransferase (Tv4AGT) on the EV surface which binds to heparan sulfate of host cell surface proteoglycans84.

Adherence of T. vaginalis to host epithelium is indeed a key event in trichomoniasis. In this process, the host’s surface proteins galectin-185 and -386 have an essential role by binding to lipoglycan (LG) on the parasite’s surface85. Host galectins are also instrumental in dimming the host response to T. vaginalis, and TvLG binding can further tune this86. However, numerous other cell surface–associated factors, such as a novel cadherin-like protein87, actinin-288, and triosephosphate isomerase89, promote adherence of T. vaginalis to host epithelium. The last of these is a glycolytic protein which can also be associated with the parasite’s cell surface and bind to fibronectin and laminin. Two other groups of proteins, the BspA and Pmp domain-containing proteins, seem to enhance adherence90, and the former have been found to be transported to the host in EVs79. Finally, palmitoylation of proteins was found to positively affect adherence of T. vaginalis to host cells91.

Eventually, adherence is followed by cell lysis. Damage to host cells can be inflicted by parasite proteases such as metalloprotease TvMP5092 or cysteine proteinase 2 (CP2)93,94, and antibody treatment against either of these factors greatly diminished cytotoxicity as exerted by T. vaginalis. Interestingly, T. vaginalis cysteine proteases do also degrade the anion channel CFTR on the host cell surface, leading to elevated intracellular Cl concentrations and induction of NF-κB signaling95. Another protease, the rhomboid protease TvROM1, exerts an indirect effect by cleaving T. vaginalis substrate proteins, resulting in enhanced attachment and damage to host cells96. Just recently, saponin-like pore-forming proteins (TvSaplips) were identified in the T. vaginalis genome97 and one of these, TvSaplip12, was expressed and characterized. TvSaplip12 is strongly upregulated upon contact with host cells and has a strong lytic activity against bacteria and HeLa cells. Consequently, it has been proposed to act as a Trichopore, in accordance with Amoebapore in Entamoeba histolytica98. To summarize, the picture of pathogenesis of trichomoniasis is still incomplete but the gaps are being filled at an increasing pace.

Anti-trichomonadal drugs: established and experimental

Throughout the last six decades, the 5-nitroimidazole drug metronidazole has remained the mainstay of anti-trichomonadal chemotherapy99, although resistance has become an increasingly worrying issue. Clinical metronidazole resistance is a complex phenomenon affecting numerous enzymatic pathways in T. vaginalis99. In a large-scale study on gene expression in metronidazole-sensitive and -resistant strains13, several genes were found to be differentially expressed in three metronidazole-resistant strains assayed. Flavin reductase 1 (FR1), an oxygen-scavenging enzyme which produces hydrogen peroxide, had been previously identified as a mediator of metronidazole resistance100 and was found to be downregulated in all three isolates. Various nitroreductases, likewise identified previously101103, were also downregulated. In contrast, multidrug resistance pump and metal ABC transporter genes were upregulated in all three resistant strains. The relative contribution of all of these factors to metronidazole resistance remains to be determined.

Importantly, treatment failures with metronidazole are not always caused by resistance as such. Treatment regimens can also strongly affect treatment outcome. It was demonstrated recently that a seven-day course with 500 mg metronidazole twice per day is clearly more effective than a single dose with 2 g104. In addition, it is necessary to test alternative treatments within the drug class of 5-nitroimidazoles, such as secnidazole105 or novel derivatives106,107.

Cross resistance, however, is often a problem with 5-nitroimidazole drugs, so completely different drugs have also been explored for anti-trichomonadal activity. Arguably, the most promising candidate is auranofin, a repurposed anti-rheumatic drug which was shown to be effective against a larger number of parasites108. Indeed, auranofin is also effective against T. vaginalis and can cure experimentally infected mice109. It was also successfully administered topically in mice on nanoparticles suspended in a hydrogel110. Thioredoxin reductase has been proposed to be the main target of auranofin, but this needs further confirmation as observations in another parasitic anaerobic flagellate, Giardia lamblia, contradict this notion111. Numerous other candidate drugs, including proteasome inhibitors such as carmaphycin-17112, zinc sulfate and zinc complexes113,114, bisbenzimidazole analogues115, boric acid116, and tetracycline117, were also evaluated. For a more complete overview of recent anti-trichomonadal drug research, a comprehensive review is available118.

As a concluding note, it is important to emphasize that assay conditions can be of very high importance when evaluating efficacies of established and novel anti-trichomonadal drugs. For example, cysteine, which is routinely used in growth media for T. vaginalis, has a highly protective effect against metronidazole and auranofin119.

Concluding remarks

During the writing of this review, it became quickly apparent that the number of high-quality research articles in the T. vaginalis field has increased in the last five years as compared with the preceding quinquennial period120, a development which is also reflected in a larger number of references cited in this review. This is highly encouraging and indicates that T. vaginalis now receives more attention than before. There were substantial advances in our understanding of how T. vaginalis interacts with its host and, equally important, with the microbiome of which it is a part. As to the latter, the formation of consortia of protistologists, bacteriologists, and mycologists might even accelerate the pace of insightful discoveries in the future.

The peer reviewers who approve this article are:

  • Augusto Simoes-Barbosa, School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand

  • Natalia de Miguel, Laboratorio de Parásitos Anaerobios, Instituto Tecnológico Chascomús, National Scientific and Technical Research Council, National University of San Martin (CONICET-UNSAM), Chascomús, Argentina

  • Paola Rappelli, Department of Biomedical Sciences, University of Sassari, Sassari, Italy

Funding Statement

The author declares that no grants were involved in supporting this work.

References

  • 1. Rowley J, Vander Hoorn S, Korenromp E, et al. : Chlamydia, gonorrhoea, trichomoniasis and syphilis: Global prevalence and incidence estimates, 2016. Bull World Health Organ. 2019; 97(8): 548–562P. 10.2471/BLT.18.228486 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 2. Kissinger P: Trichomonas vaginalis: A review of epidemiologic, clinical and treatment issues. BMC Infect Dis. 2015; 15: 307. 10.1186/s12879-015-1055-0 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 3. Masha SC, Cools P, Sanders EJ, et al. : Trichomonas vaginalis and HIV infection acquisition: A systematic review and meta-analysis. Sex Transm Infect. 2019; 95(1): 36–42. 10.1136/sextrans-2018-053713 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 4. Ghosh I, Muwonge R, Mittal S, et al. : Association between high risk human papillomavirus infection and co-infection with Candida spp. and Trichomonas vaginalis in women with cervical premalignant and malignant lesions. J Clin Virol. 2017; 87: 43–8. 10.1016/j.jcv.2016.12.007 [DOI] [PubMed] [Google Scholar]
  • 5. Shui IM, Kolb S, Hanson C, et al. : Trichomonas vaginalis infection and risk of advanced prostate cancer. Prostate. 2016; 76(7): 620–3. 10.1002/pros.23153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Tsang SH, Peisch SF, Rowan B, et al. : Association between Trichomonas vaginalis and prostate cancer mortality. Int J Cancer. 2019; 144(10): 2377–80. 10.1002/ijc.31885 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Lindmark DG, Müller M: Hydrogenosome, a Cytoplasmic Organelle of the Anaerobic Flagellate Tritrichomonas foetus, and Its Role in Pyruvate Metabolism. J Biol Chem. 1973; 248(22): 7724–8. 10.1016/S0021-9258(19)43249-3 [DOI] [PubMed] [Google Scholar]
  • 8. Conrad MD, Bradic M, Warring SD, et al. : Getting trichy: Tools and approaches to interrogating Trichomonas vaginalis in a post-genome world. Trends Parasitol. 2013; 29(1): 17–25. 10.1016/j.pt.2012.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Bradic M, Carlton JM: Does the common sexually transmitted parasite Trichomonas vaginalis have sex? PLoS Pathog. 2018; 14(3): e1006831. 10.1371/journal.ppat.1006831 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 10. Beri D, Yadav P, Devi HRN, et al. : Demonstration and Characterization of Cyst-Like Structures in the Life Cycle of Trichomonas vaginalis. Front Cell Infect Microbiol. 2019; 9: 430. 10.3389/fcimb.2019.00430 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 11. van Gerwen OT, Muzny CA: Recent advances in the epidemiology, diagnosis, and management of Trichomonas vaginalis infection [version 1; peer review: 2 approved]. F1000Res. 2019; 8. 10.12688/f1000research.19972.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Carlton JM, Hirt RP, Silva JC: Draft genome sequence of the sexually transmitted pathogen Trichomonas vaginalis. Science. 2007; 315(5809): 207–12. 10.1126/science.1132894 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 13. Bradic M, Warring SD, Tooley GE, et al. : Genetic Indicators of Drug Resistance in the Highly Repetitive Genome of Trichomonas vaginalis. Genome Biol Evol. 2017; 9(6): 1658–72. 10.1093/gbe/evx110 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 14. Warring SD, Blow F, Avecilla G, et al. : Small RNAs Are Implicated in Regulation of Gene and Transposable Element Expression in the Protist Trichomonas vaginalis. mSphere. 2021; 6(1): e01061-20. 10.1128/mSphere.01061-20 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 15. Wang SE, Amir AS, Nguyen T, et al. : Spliceosomal introns in Trichomonas vaginalis revisited. Parasit Vectors. 2018; 11(1): 607. 10.1186/s13071-018-3196-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 16. Liston DR, Lau AO, Ortiz D, et al. : Initiator recognition in a primitive eukaryote: IBP39, an initiator-binding protein from Trichomonas vaginalis. Mol Cell Biol. 2001; 21(22): 7872–82. 10.1128/MCB.21.22.7872-7882.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 17. Smith AJ, Chudnovsky L, Simoes-Barbosa A, et al. : Novel core promoter elements and a cognate transcription factor in the divergent unicellular eukaryote Trichomonas vaginalis. Mol Cell Biol. 2011; 31(7): 1444–58. 10.1128/MCB.00745-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Parra-Marín O, Rosas-Hernández L, López-Pacheco K, et al. : An in vitro characterisation of the Trichomonas vaginalis TATA box-binding proteins (TBPs). Parasitol Res. 2019; 118(10): 3019–31. 10.1007/s00436-019-06438-z [DOI] [PubMed] [Google Scholar]
  • 19. Simoes-Barbosa A, Hirt RP, Johnson PJ: A metazoan/plant-like capping enzyme and cap modified nucleotides in the unicellular eukaryote Trichomonas vaginalis. PLoS Pathog. 2010; 6(7): e1000999. 10.1371/journal.ppat.1000999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Simoes-Barbosa A, Meloni D, Wohlschlegel JA, et al. : Spliceosomal snRNAs in the unicellular eukaryote Trichomonas vaginalis are structurally conserved but lack a 5'-cap structure. RNA. 2008; 14(8): 1617–31. 10.1261/rna.1045408 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Lizarraga A, O'Brown ZK, Boulias K, et al. : Adenine DNA methylation, 3D genome organization, and gene expression in the parasite Trichomonas vaginalis. Proc Natl Acad Sci U S A. 2020; 117(23): 13033–43. 10.1073/pnas.1917286117 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 22. Song MJ, Kim M, Choi Y, et al. : Epigenome mapping highlights chromatin-mediated gene regulation in the protozoan parasite Trichomonas vaginalis. Sci Rep. 2017; 7: 45365. 10.1038/srep45365 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 23. Pachano T, Nievas YR, Lizarraga A, et al. : Epigenetics regulates transcription and pathogenesis in the parasite Trichomonas vaginalis. Cell Microbiol. 2017; 19(6): e12716. 10.1111/cmi.12716 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 24. Wang SE, Brooks AES, Poole AM, et al. : Determinants of translation efficiency in the evolutionarily-divergent protist Trichomonas vaginalis. BMC Mol Cell Biol. 2020; 21(1): 54. 10.1186/s12860-020-00297-8 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 25. Wang SE, Brooks AES, Cann B, et al. : The fluorescent protein iLOV outperforms eGFP as a reporter gene in the microaerophilic protozoan Trichomonas vaginalis. Mol Biochem Parasitol. 2017; 216: 1–4. 10.1016/j.molbiopara.2017.06.003 [DOI] [PubMed] [Google Scholar]
  • 26. dos Santos O, de Vargas Rigo G, Frasson AP, et al. : Optimal Reference Genes for Gene Expression Normalization in Trichomonas vaginalis. PLoS One. 2015; 10(9): e0138331. 10.1371/journal.pone.0138331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Ravaee R, Ebadi P, Hatam G, et al. : Synthetic siRNAs effectively target cystein protease 12 and α-actinin transcripts in Trichomonas vaginalis. Exp Parasitol. 2015; 157: 30–4. 10.1016/j.exppara.2015.06.012 [DOI] [PubMed] [Google Scholar]
  • 28. Janssen BD, Chen YP, Molgora BM, et al. : CRISPR/Cas9-mediated gene modification and gene knock out in the human-infective parasite Trichomonas vaginalis. Sci Rep. 2018; 8(1): 270. 10.1038/s41598-017-18442-3 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 29. Hrdy I, Hirt RP, Dolezal P, et al. : Trichomonas hydrogenosomes contain the NADH dehydrogenase module of mitochondrial complex I. Nature. 2004; 432(7017): 618–22. 10.1038/nature03149 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 30. Mentel M, Zimorski V, Haferkamp P, et al. : Protein import into hydrogenosomes of Trichomonas vaginalis involves both N-terminal and internal targeting signals: A case study of thioredoxin reductases. Eukaryotic Cell. 2008; 7(10): 1750–7. 10.1128/EC.00206-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Rada P, Makki AR, Zimorski V, et al. : N-Terminal Presequence-Independent Import of Phosphofructokinase into Hydrogenosomes of Trichomonas vaginalis. Eukaryotic Cell. 2015; 14(12): 1264–75. 10.1128/EC.00104-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Garg S, Stölting J, Zimorski V, et al. : Conservation of Transit Peptide-Independent Protein Import into the Mitochondrial and Hydrogenosomal Matrix. Genome Biol Evol. 2015; 7(9): 2716–26. 10.1093/gbe/evv175 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Rada P, Makki A, Žárský V, et al. : Targeting of tail-anchored proteins to Trichomonas vaginalis hydrogenosomes. Mol Microbiol. 2019; 111(3): 588–603. 10.1111/mmi.14175 [DOI] [PubMed] [Google Scholar]
  • 34. Makki A, Rada P, Žárský V, et al. : Triplet-pore structure of a highly divergent TOM complex of hydrogenosomes in Trichomonas vaginalis. PLoS Biol. 2019; 17(1): e3000098. 10.1371/journal.pbio.3000098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Rada P, Kellerová P, Verner Z, et al. : Investigation of the Secretory Pathway in Trichomonas vaginalis Argues against a Moonlighting Function of Hydrogenosomal Enzymes. J Eukaryot Microbiol. 2019; 66(6): 899–910. 10.1111/jeu.12741 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 36. Hsu HM, Huang YH, Aryal S, et al. : Endomembrane Protein Trafficking Regulated by a TvCyP2 Cyclophilin in the Protozoan Parasite, Trichomonas vaginalis. Sci Rep. 2020; 10(1): 1275. 10.1038/s41598-020-58270-6 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 37. Huang KY, Ong SC, Wu CC, et al. : Metabolic reprogramming of hydrogenosomal amino acids in Trichomonas vaginalis under glucose restriction. J Microbiol Immunol Infect. 2019; 52(4): 630–7. 10.1016/j.jmii.2017.10.005 [DOI] [PubMed] [Google Scholar]
  • 38. Cheng WH, Huang KY, Huang PJ, et al. : Nitric oxide maintains cell survival of Trichomonas vaginalis upon iron depletion. Parasit Vectors. 2015; 8: 393. 10.1186/s13071-015-1000-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Leitsch D, Williams CF, Hrdý I: Redox Pathways as Drug Targets in Microaerophilic Parasites. Trends Parasitol. 2018; 34(7): 576–89. 10.1016/j.pt.2018.04.007 [DOI] [PubMed] [Google Scholar]
  • 40. Nývltová E, Smutná T, Tachezy J, et al. : OsmC and incomplete glycine decarboxylase complex mediate reductive detoxification of peroxides in hydrogenosomes of Trichomonas vaginalis. Mol Biochem Parasitol. 2016; 206(1–2): 29–38. 10.1016/j.molbiopara.2016.01.006 [DOI] [PubMed] [Google Scholar]
  • 41. Wilson WA, Pradhan P, Madhan N, et al. : Glycogen synthase from the parabasalian parasite Trichomonas vaginalis: An unusual member of the starch/glycogen synthase family. Biochimie. 2017; 138: 90–101. 10.1016/j.biochi.2017.04.016 [DOI] [PubMed] [Google Scholar]
  • 42. Huffman RD, Nawrocki LD, Wilson WA, et al. : Digestion of glycogen by a glucosidase released by Trichomonas vaginalis. Exp Parasitol. 2015; 159: 151–9. 10.1016/j.exppara.2015.09.011 [DOI] [PubMed] [Google Scholar]
  • 43. Smith RW, Brittingham A, Wilson WA: Purification and identification of amylases released by the human pathogen Trichomonas vaginalis that are active towards glycogen. Mol Biochem Parasitol. 2016; 210(1–2): 22–31. 10.1016/j.molbiopara.2016.08.002 [DOI] [PubMed] [Google Scholar]
  • 44. Štáfková J, Rada P, Meloni D, et al. : Dynamic secretome of Trichomonas vaginalis: Case study of β-amylases. Mol Cell Proteomics. 2018; 17(2): 304–20. 10.1074/mcp.RA117.000434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. ter Kuile BH, Müller M: Maltose utilization by extracellular hydrolysis followed by glucose transport in Trichomonas vaginalis. Parasitology. 1995; 110(Pt 1): 37–44. 10.1017/s0031182000081026 [DOI] [PubMed] [Google Scholar]
  • 46. Frasson AP, dos Santos O, Meirelles LC, et al. : Five putative nucleoside triphosphate diphosphohydrolase genes are expressed in Trichomonas vaginalis. FEMS Microbiol Lett. 2016; 363(2): fnv221. 10.1093/femsle/fnv221 [DOI] [PubMed] [Google Scholar]
  • 47. Menezes CB, Durgante J, de Oliveira RR, et al. : Trichomonas vaginalis NTPDase and ecto-5'-nucleotidase hydrolyze guanine nucleotides and increase extracellular guanosine levels under serum restriction. Mol Biochem Parasitol. 2016; 207(1): 10–8. 10.1016/j.molbiopara.2016.04.003 [DOI] [PubMed] [Google Scholar]
  • 48. Pekmezovic M, Mogavero S, Naglik JR, et al. : Host-Pathogen Interactions during Female Genital Tract Infections. Trends Microbiol. 2019; 27(12): 982–96. 10.1016/j.tim.2019.07.006 [DOI] [PubMed] [Google Scholar]
  • 49. Ravel J, Gajer P, Abdo Z, et al. : Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci U S A. 2011; 108 Suppl 1(Suppl 1): 4680–7. 10.1073/pnas.1002611107 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 50. Brotman RM, Bradford LL, Conrad M, et al. : Association between Trichomonas vaginalis and vaginal bacterial community composition among reproductive-age women. Sex Transm Dis. 2012; 39(10): 807–12. 10.1097/OLQ.0b013e3182631c79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Jarrett OD, Srinivasan S, Richardson BA, et al. : Specific Vaginal Bacteria Are Associated With an Increased Risk of Trichomonas vaginalis Acquisition in Women. J Infect Dis. 2019; 220(9): 1503–10. 10.1093/infdis/jiz354 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 52. Hinderfeld AS, Simoes-Barbosa A: Vaginal dysbiotic bacteria act as pathobionts of the protozoal pathogen Trichomonas vaginalis. Microb Pathog. 2020; 138: 103820. 10.1016/j.micpath.2019.103820 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 53. Hinderfeld AS, Phukan N, Bär AK, et al. : Cooperative Interactions between Trichomonas vaginalis and Associated Bacteria Enhance Paracellular Permeability of the Cervicovaginal Epithelium by Dysregulating Tight Junctions. Infect Immun. 2019; 87(5): e00141–19. 10.1128/IAI.00141-19 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 54. Phukan N, Brooks AES, Simoes-Barbosa A: A Cell Surface Aggregation-Promoting Factor from Lactobacillus gasseri Contributes to Inhibition of Trichomonas vaginalis Adhesion to Human Vaginal Ectocervical Cells. Infect Immun. 2018; 86(8): e00907-17. 10.1128/IAI.00907-17 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 55. Pinheiro J, Biboy J, Vollmer W, et al. : The Protozoan Trichomonas vaginalis Targets Bacteria with Laterally Acquired NlpC/P60 Peptidoglycan Hydrolases. MBio. 2018; 9(6): e01784–18. 10.1128/mBio.01784-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Fichorova R, Fraga J, Rappelli P, et al. : Trichomonas vaginalis infection in symbiosis with Trichomonasvirus and Mycoplasma. Res Microbiol. 2017; 168(9–10): 882–91. 10.1016/j.resmic.2017.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Margarita V, Rappelli P, Dessì D, et al. : Symbiotic Association with Mycoplasma hominis Can Influence Growth Rate, ATP Production, Cytolysis and Inflammatory Response of Trichomonas vaginalis. Front Microbiol. 2016; 7: 953. 10.3389/fmicb.2016.00953 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 58. Fürnkranz U, Henrich B, Walochnik J: Mycoplasma hominis impacts gene expression in Trichomonas vaginalis. Parasitol Res. 2018; 117(3): 841–7. 10.1007/s00436-018-5761-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Larsen B, Hwang J: Mycoplasma, Ureaplasma, and adverse pregnancy outcomes: A fresh look. Infect Dis Obstet Gynecol. 2010; 2010: 521921. 10.1155/2010/521921 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Margarita V, Fiori PL, Rappelli P: Impact of Symbiosis Between Trichomonas vaginalis and Mycoplasma hominis on Vaginal Dysbiosis: A Mini Review. Front Cell Infect Microbiol. 2020; 10: 179. 10.3389/fcimb.2020.00179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Thi Trung Thu T, Margarita V, Cocco AR, et al. : Trichomonas vaginalis Transports Virulent Mycoplasma hominis and Transmits the Infection to Human Cells after Metronidazole Treatment: A Potential Role in Bacterial Invasion of Fetal Membranes and Amniotic Fluid. J Pregnancy. 2018; 2018: 5037181. 10.1155/2018/5037181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Fettweis JM, Serrano MG, Huang B, et al. : An emerging mycoplasma associated with trichomoniasis, vaginal infection and disease. PLoS ONE. 2014; 9(10): e110943. 10.1371/journal.pone.0110943 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Ioannidis A, Papaioannou P, Magiorkinis E, et al. : Detecting the Diversity of Mycoplasma and Ureaplasma Endosymbionts Hosted by Trichomonas vaginalis Isolates. Front Microbiol. 2017; 8: 1188. 10.3389/fmicb.2017.01188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Graves KJ, Ghosh AP, Kissinger PJ, et al. : Trichomonas vaginalis virus: A review of the literature. Int J STD AIDS. 2019; 30: 496–504. 10.1177/0956462418809767 [DOI] [PubMed] [Google Scholar]
  • 65. Fichorova RN, Buck OR, Yamamoto HS, et al. : The villain team-up or how Trichomonas vaginalis and bacterial vaginosis alter innate immunity in concert. Sex Transm Infect. 2013; 89(6): 460–6. 10.1136/sextrans-2013-051052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Govender Y, Chan T, Yamamoto HS, et al. : The Role of Small Extracellular Vesicles in Viral-Protozoan Symbiosis: Lessons From Trichomonasvirus in an Isogenic Host Parasite Model. Front Cell Infect Microbiol. 2020; 10: 591172. 10.3389/fcimb.2020.591172 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 67. Graves KJ, Ghosh AP, Schmidt N, et al. : Trichomonas vaginalis Virus Among Women With Trichomoniasis and Associations With Demographics, Clinical Outcomes, and Metronidazole Resistance. Clin Infect Dis. 2019; 69(12): 2170–6. 10.1093/cid/ciz146 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 68. Mercer F, Johnson PJ: Trichomonas vaginalis: Pathogenesis, Symbiont Interactions, and Host Cell Immune Responses. Trends Parasitol. 2018; 34(8): 683–93. 10.1016/j.pt.2018.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Mercer F, Diala FGI, Chen YP, et al. : Leukocyte Lysis and Cytokine Induction by the Human Sexually Transmitted Parasite Trichomonas vaginalis. PLoS Negl Trop Dis. 2016; 10(8): e0004913. 10.1371/journal.pntd.0004913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Olmos-Ortiz LM, Barajas-Mendiola MA, Barrios-Rodiles M, et al. : Trichomonas vaginalis exosome-like vesicles modify the cytokine profile and reduce inflammation in parasite-infected mice. Parasite Immunol. 2017; 39(6): e12426. 10.1111/pim.12426 [DOI] [PubMed] [Google Scholar]
  • 71. Han IH, Kim JH, Kim SS, et al. : Signalling pathways associated with IL-6 production and epithelial-mesenchymal transition induction in prostate epithelial cells stimulated with Trichomonas vaginalis. Parasite Immunol. 2016; 38(11): 678–87. 10.1111/pim.12357 [DOI] [PubMed] [Google Scholar]
  • 72. Han IH, Song HO, Ryu JS: IL-6 produced by prostate epithelial cells stimulated with Trichomonas vaginalis promotes proliferation of prostate cancer cells by inducing M2 polarization of THP-1-derived macrophages. PLoS Negl Trop Dis. 2020; 14(3): e0008126. 10.1371/journal.pntd.0008126 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 73. Riestra AM, Valderrama JA, Patras KA, et al. : Trichomonas vaginalis Induces NLRP3 Inflammasome Activation and Pyroptotic Cell Death in Human Macrophages. J Innate Immun. 2019; 11(1): 86–98. 10.1159/000493585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Li L, Li X, Gong P, et al. : Trichomonas vaginalis Induces Production of Proinflammatory Cytokines in Mouse Macrophages Through Activation of MAPK and NF-κB Pathways Partially Mediated by TLR2. Front Microbiol. 2018; 9: 712. 10.3389/fmicb.2018.00712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Lee HY, Kim J, Ryu JS, et al. : Trichomonas vaginalis α-Actinin 2 Modulates Host Immune Responses by Inducing Tolerogenic Dendritic Cells via IL-10 Production from Regulatory T Cells. Korean J Parasitol. 2017; 55: 375–84. 10.3347/kjp.2017.55.4.375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Chen YP, Twu O, Johnson PJ: Trichomonas vaginalis Macrophage Migration Inhibitory Factor Mediates Parasite Survival during Nutrient Stress. MBio. 2018; 9(3): e00910–18. 10.1128/mBio.00910-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Min A, Lee YA, Kim KA, et al. : SNAP23-Dependent Surface Translocation of Leukotriene B4 (LTB4) Receptor 1 Is Essential for NOX2-Mediated Exocytotic Degranulation in Human Mast Cells Induced by Trichomonas vaginalis-Secreted LTB4. Infect Immun. 2017; 85(1): e00526–16. 10.1128/IAI.00526-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Nievas YR, Lizarraga A, Salas N, et al. : Extracellular vesicles released by anaerobic protozoan parasites: Current situation. Cell Microbiol. 2020; 22(11): e13257. 10.1111/cmi.13257 [DOI] [PubMed] [Google Scholar]
  • 79. Twu O, de Miguel N, Lustig G, et al. : Trichomonas vaginalis exosomes deliver cargo to host cells and mediate host:parasite interactions. PLoS Pathog. 2013; 9: e1003482. 10.1371/journal.ppat.1003482 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 80. Nievas YR, Coceres VM, Midlej V, et al. : Membrane-shed vesicles from the parasite Trichomonas vaginalis: Characterization and their association with cell interaction. Cell Mol Life Sci. 2018; 75(12): 2211–26. 10.1007/s00018-017-2726-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Artuyants A, Campos TL, Rai AK, et al. : Extracellular vesicles produced by the protozoan parasite Trichomonas vaginalis contain a preferential cargo of tRNA-derived small RNAs. Int J Parasitol. 2020; 50(14): 1145–55. 10.1016/j.ijpara.2020.07.003 [DOI] [PubMed] [Google Scholar]
  • 82. Garcia-Silva MR, Sanguinetti J, Cabrera-Cabrera F, et al. : A particular set of small non-coding RNAs is bound to the distinctive Argonaute protein of Trypanosoma cruzi: Insights from RNA-interference deficient organisms. Gene. 2014; 538(2): 379–84. 10.1016/j.gene.2014.01.023 [DOI] [PubMed] [Google Scholar]
  • 83. Lambertz U, Oviedo Ovando ME, Vasconcelos EJR, et al. : Small RNAs derived from tRNAs and rRNAs are highly enriched in exosomes from both old and new world Leishmania providing evidence for conserved exosomal RNA Packaging. BMC Genomics. 2015; 16(1): 151. 10.1186/s12864-015-1260-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Rai AK, Johnson PJ: Trichomonas vaginalis extracellular vesicles are internalized by host cells using proteoglycans and caveolin-dependent endocytosis. Proc Natl Acad Sci U S A. 2019; 116(43): 21354–60. 10.1073/pnas.1912356116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Okumura CYM, Baum LG, Johnson PJ: Galectin-1 on cervical epithelial cells is a receptor for the sexually transmitted human parasite Trichomonas vaginalis. Cell Microbiol. 2008; 10(10): 2078–90. 10.1111/j.1462-5822.2008.01190.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Fichorova RN, Yamamoto HS, Fashemi T, et al. : Trichomonas vaginalis Lipophosphoglycan Exploits Binding to Galectin-1 and -3 to Modulate Epithelial Immunity. J Biol Chem. 2016; 291(2): 998–1013. 10.1074/jbc.M115.651497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Chen YP, Riestra AM, Rai AK, et al. : A Novel Cadherin-like Protein Mediates Adherence to and Killing of Host Cells by the Parasite Trichomonas vaginalis. mBio. 2019; 10(3): e00720–19. 10.1128/mBio.00720-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Lee HY, Kim J, Park SJ: Role of α-Actinin 2 in Cytoadherence and Cytotoxicity of Trichomonas vaginalis. J Microbiol Biotechnol. 2017; 27(10): 1844–54. 10.4014/jmb.1706.06050 [DOI] [PubMed] [Google Scholar]
  • 89. Miranda-Ozuna JFT, Hernández-García MS, Brieba LG, et al. : The Glycolytic Enzyme Triosephosphate Isomerase of Trichomonas vaginalis Is a Surface-Associated Protein Induced by Glucose That Functions as a Laminin- and Fibronectin-Binding Protein. Infect Immun. 2016; 84(10): 2878–94. 10.1128/IAI.00538-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Handrich MR, Garg SG, Sommerville EW, et al. : Characterization of the BspA and Pmp protein family of trichomonads. Parasit Vectors. 2019; 12(1): 406. 10.1186/s13071-019-3660-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Nievas YR, Vashisht AA, Corvi MM, et al. : Protein Palmitoylation Plays an Important Role in Trichomonas vaginalis Adherence. Mol Cell Proteomics. 2018; 17(11): 2229–41. 10.1074/mcp.RA117.000018 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 92. Puente-Rivera J, Villalpando JL, Villalobos-Osnaya A, et al. : The 50kDa metalloproteinase TvMP50 is a zinc-mediated Trichomonas vaginalis virulence factor. Mol Biochem Parasitol. 2017; 217: 32–41. 10.1016/j.molbiopara.2017.09.001 [DOI] [PubMed] [Google Scholar]
  • 93. Miranda-Ozuna JFT, Rivera-Rivas LA, Cárdenas-Guerra RE, et al. : Glucose-restriction increases Trichomonas vaginalis cellular damage towards HeLa cells and proteolytic activity of cysteine proteinases (CPs), such as TvCP2. Parasitology. 2019; 146(9): 1156–1166. 10.1017/S0031182019000209 [DOI] [PubMed] [Google Scholar]
  • 94. Rivera-Rivas LA, Lorenzo-Benito S, Sánchez-Rodríguez DB, et al. : The effect of iron on Trichomonas vaginalis TvCP2: A cysteine proteinase found in vaginal secretions of trichomoniasis patients. Parasitology. 2020; 147(7): 760–74. 10.1017/S0031182020000438 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Xu JB, Zhang YL, Huang J, et al. : Increased intracellular Cl- concentration mediates Trichomonas vaginalis-induced inflammation in the human vaginal epithelium. Int J Parasitol. 2019; 49(9): 697–704. 10.1016/j.ijpara.2019.04.005 [DOI] [PubMed] [Google Scholar]
  • 96. Riestra AM, Gandhi S, Sweredoski MJ, et al. : A Trichomonas vaginalis Rhomboid Protease and Its Substrate Modulate Parasite Attachment and Cytolysis of Host Cells. PLoS Pathog. 2015; 11(12): e1005294. 10.1371/journal.ppat.1005294 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 97. Diaz N, Lico C, Capodicasa C, et al. : Production and Functional Characterization of a Recombinant Predicted Pore-Forming Protein (TVSAPLIP12) of Trichomonas vaginalis in Nicotiana benthamiana Plants. Front Cell Infect Microbiol. 2020; 10: 581066. 10.3389/fcimb.2020.581066 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 98. Leippe M: Pore-forming toxins from pathogenic amoebae. Appl Microbiol Biotechnol. 2014; 98(10): 4347–53. 10.1007/s00253-014-5673-z [DOI] [PubMed] [Google Scholar]
  • 99. Leitsch D: A review on metronidazole: An old warhorse in antimicrobial chemotherapy. Parasitology. 2019; 146(9): 1167–78. 10.1017/S0031182017002025 [DOI] [PubMed] [Google Scholar]
  • 100. Leitsch D, Janssen BD, Kolarich D, et al. : Trichomonas vaginalis flavin reductase 1 and its role in metronidazole resistance. Mol Microbiol. 2014; 91(1): 198–208. 10.1111/mmi.12455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Pal D, Banerjee S, Cui J, et al. : Giardia, Entamoeba, and Trichomonas enzymes activate metronidazole (nitroreductases) and inactivate metronidazole (nitroimidazole reductases). Antimicrob Agents Chemother. 2009; 53(2): 458–64. 10.1128/AAC.00909-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Paulish-Miller TE, Augostini P, Schuyler JA, et al. : Trichomonas vaginalis metronidazole resistance is associated with single nucleotide polymorphisms in the nitroreductase genes ntr4Tv and ntr6Tv. Antimicrob Agents Chemother. 2014; 58(5): 2938–43. 10.1128/AAC.02370-13 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 103. Beltrán NC, Horváthová L, Jedelský PL, et al. : Iron-induced changes in the proteome of Trichomonas vaginalis hydrogenosomes. PLoS One. 2013; 8(5): e65148. 10.1371/journal.pone.0065148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Kissinger P, Muzny CA, Mena LA, et al. : Single-dose versus 7-day-dose metronidazole for the treatment of trichomoniasis in women: An open-label, randomised controlled trial. Lancet Infect Dis. 2018; 18(11): 1251–1259. 10.1016/S1473-3099(18)30423-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Ghosh AP, Aycock C, Schwebke JR: In Vitro Study of the Susceptibility of Clinical Isolates of Trichomonas vaginalis to Metronidazole and Secnidazole. Antimicrob Agents Chemother. 2018; 62: e02329–17. 10.1128/AAC.02329-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Mandalapu D, Kushwaha B, Gupta S, et al. : 2-Methyl-4/5-nitroimidazole derivatives potentiated against sexually transmitted Trichomonas: Design, synthesis, biology and 3D-QSAR study. Eur J Med Chem. 2016; 124: 820–39. 10.1016/j.ejmech.2016.09.006 [DOI] [PubMed] [Google Scholar]
  • 107. Jarrad AM, Debnath A, Miyamoto Y, et al. : Nitroimidazole carboxamides as antiparasitic agents targeting Giardia lamblia, Entamoeba histolytica and Trichomonas vaginalis. Eur J Med Chem. 2016; 120: 353–62. 10.1016/j.ejmech.2016.04.064 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 108. May HC, Yu JJ, Guentzel MN, et al. : Repurposing Auranofin, Ebselen, and PX-12 as Antimicrobial Agents Targeting the Thioredoxin System. Front Microbiol. 2018; 9: 336. 10.3389/fmicb.2018.00336 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
  • 109. Hopper M, Yun JF, Zhou B, et al. : Auranofin inactivates Trichomonas vaginalis thioredoxin reductase and is effective against trichomonads in vitro and in vivo. Int J Antimicrob Agents. 2016; 48(6): 690–4. 10.1016/j.ijantimicag.2016.09.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Zhang Y, Miyamoto Y, Ihara S, et al. : Composite thermoresponsive hydrogel with auranofin-loaded nanoparticles for topical treatment of vaginal trichomonad infection. Adv Ther (Weinh). 2019; 2(12): 1900157. 10.1002/adtp.201900157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Leitsch D, Müller J, Müller N: Evaluation of Giardia lamblia thioredoxin reductase as drug activating enzyme and as drug target. Int J Parasitol Drugs Drug Resist. 2016; 6(3): 148–53. 10.1016/j.ijpddr.2016.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. O'Donoghue AJ, Bibo-Verdugo B, Miyamoto Y, et al. : 20S Proteasome as a Drug Target in Trichomonas vaginalis. Antimicrob Agents Chemother. 2019; 63(11): e00448–19. 10.1128/AAC.00448-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Byun JM, Jeong DH, Kim YN, et al. : Experience of successful treatment of patients with metronidazole-resistant Trichomonas vaginalis with zinc sulfate: A case series. Taiwan J Obstet Gynecol. 2015; 54(5): 617–20. 10.1016/j.tjog.2015.08.018 [DOI] [PubMed] [Google Scholar]
  • 114. Gilboa-Garber N: Pseudomonas aeruginosa Lectins as a Model for Lectin Production, Properties, Applications and Functions. Zentralbl Bakteriol Mikrobiol Hyg A. 1988; 270(1–2): 3–15. 10.1016/s0176-6724(88)80135-4 [DOI] [PubMed] [Google Scholar]
  • 115. Korosh T, Bujans E, Morada M, et al. : Potential of bisbenzimidazole-analogs toward metronidazole-resistant Trichomonas vaginalis isolates. Chem Biol Drug Des. 2017; 90(4): 489–95. 10.1111/cbdd.12972 [DOI] [PubMed] [Google Scholar]
  • 116. Backus KV, Muzny CA, Beauchamps LS: Trichomonas vaginalis Treated With Boric Acid in a Metronidazole Allergic Female. Sex Transm Dis. 2017; 44(2): 120. 10.1097/OLQ.0000000000000559 [DOI] [PubMed] [Google Scholar]
  • 117. Huang KY, Ku FM, Cheng WH, et al. : Novel insights into the molecular events linking to cell death induced by tetracycline in the amitochondriate protozoan Trichomonas vaginalis. Antimicrob Agents Chemother. 2015; 59(11): 6891–903. 10.1128/AAC.01779-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Küng E, Fürnkranz U, Walochnik J: Chemotherapeutic options for the treatment of human trichomoniasis. Int J Antimicrob Agents. 2019; 53(2): 116–27. 10.1016/j.ijantimicag.2018.10.016 [DOI] [PubMed] [Google Scholar]
  • 119. Leitsch D: Drug susceptibility testing in microaerophilic parasites: Cysteine strongly affects the effectivities of metronidazole and auranofin, a novel and promising antimicrobial. Int J Parasitol Drugs Drug Resist. 2017; 7(3): 321–327. 10.1016/j.ijpddr.2017.09.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Leitsch D: Recent Advances in the Trichomonas vaginalis Field [version 1; peer review: 2 approved]. F1000Res. 2016; 5: F1000 Faculty Rev-162. 10.12688/f1000research.7594.1 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Faculty Reviews are provided here courtesy of Faculty Opinions Ltd.

RESOURCES