1
|
de Jesus Ramires M, Hummel K, Hatfaludi T, Hess M, Bilic I. Host-specific targets of Histomonas meleagridis antigens revealed by immunoprecipitation. Sci Rep 2025; 15:5800. [PMID: 39962091 PMCID: PMC11832935 DOI: 10.1038/s41598-025-88855-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025] Open
Abstract
Histomonas meleagridis, a protozoan parasite responsible for histomonosis (syn. Blackhead disease, histomoniasis), presents an increasing challenge for poultry health, particularly with the ban of licensed prophylactic and treatment options. Recent studies have explored H. meleagridis proteome, exoproteome, and surfaceome, linking molecular data to virulence and in vitro attenuation. Nevertheless, proteins involved in interactions with hosts remain largely unknown. In this study, we conducted immunoproteome analyses to identify key antigens involved in the humoral immune response of the parasite's main hosts, turkeys and chickens. Immunogenic proteins were isolated via immunoprecipitation using sera from chickens and turkeys that were vaccinated with a single attenuated strain and challenged with virulent strains of the protozoan, respectively. Mass spectrometry identified 155 putative H. meleagridis immunogenic proteins, of which 43 were recognized by sera from both hosts. In silico antigenicity screening (VaxElan) identified 33 pan-reactive antigens, with VaxiDL further highlighting 10 as potential vaccine candidates. Comparative analysis revealed host-specific immune responses, with 16 differential immunogenic proteins in chickens (6 specific to virulent and 10 to attenuated preparations) and 19 unique proteins in turkeys, all associated with virulent strains. These results enhance our understanding of H. meleagridis immunogenic protein dynamics and host-pathogen specificities, supporting the development of improved diagnostic tools and potential protective measures against the infection.
Collapse
Affiliation(s)
- Marcelo de Jesus Ramires
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria
| | - Karin Hummel
- VetCore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria
| | - Tamas Hatfaludi
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria
| | - Michael Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria
| | - Ivana Bilic
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Veterinärplatz 1, Vienna, A-1210, Austria.
| |
Collapse
|
2
|
Artuyants A, Hong J, Dauros-Singorenko P, Phillips A, Simoes-Barbosa A. Lactobacillus gasseri and Gardnerella vaginalis produce extracellular vesicles that contribute to the function of the vaginal microbiome and modulate host-Trichomonas vaginalis interactions. Mol Microbiol 2024; 122:357-371. [PMID: 37485746 DOI: 10.1111/mmi.15130] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/22/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023]
Abstract
Trichomonas vaginalis is an extracellular protozoan parasite of the human urogenital tract, responsible for a prevalent sexually transmitted infection. Trichomoniasis is accompanied by a dysbiotic microbiome that is characterised by the depletion of host-protective commensals such as Lactobacillus gasseri, and the flourishing of a bacterial consortium that is comparable to the one seen for bacterial vaginosis, including the founder species Gardnerella vaginalis. These two vaginal bacteria are known to have opposite effects on T. vaginalis pathogenicity. Studies on extracellular vesicles (EVs) have been focused on the direction of a microbial producer (commensal or pathogen) to a host recipient, and largely in the context of the gut microbiome. Here, taking advantage of the simplicity of the human cervicovaginal microbiome, we determined the molecular cargo of EVs produced by L. gasseri and G. vaginalis and examined how these vesicles modulate the interaction of T. vaginalis and host cells. We show that these EVs carry a specific cargo of proteins, which functions can be attributed to the opposite roles that these bacteria play in the vaginal biome. Furthermore, these bacterial EVs are delivered to host and protozoan cells, modulating host-pathogen interactions in a way that mimics the opposite effects that these bacteria have on T. vaginalis pathogenicity. This is the first study to describe side-by-side the protein composition of EVs produced by two bacteria belonging to the opposite spectrum of a microbiome and to demonstrate that these vesicles modulate the pathogenicity of a protozoan parasite. Such as in trichomoniasis, infections and dysbiosis co-occur frequently resulting in significant co-morbidities. Therefore, studies like this provide the knowledge for the development of antimicrobial therapies that aim to clear the infection while restoring a healthy microbiome.
Collapse
Affiliation(s)
| | - Jiwon Hong
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, University of Auckland, Auckland, New Zealand
| | | | - Anthony Phillips
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
3
|
Rivero MB, Alonso AM, Abdala ME, Luque ME, Carranza PG, Coceres VM, Rivero FD. Comparative membrane proteomic analysis of Tritrichomonas foetus isolates. Sci Rep 2024; 14:17033. [PMID: 39043862 PMCID: PMC11266394 DOI: 10.1038/s41598-024-67827-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Tritrichomonas foetus is a flagellated and anaerobic parasite able to infect cattle and felines. Despite its prevalence, there is no effective standardized or legal treatment for T. foetus-infected cattle; the vaccination still has limited success in mitigating infections and reducing abortion risk; and nowadays, the diagnosis of T. foetus presents important limitations in terms of sensitivity and specificity in bovines. Here, we characterize the plasma membrane proteome of T. foetus and identify proteins that are represented in different isolates of this protozoan. Additionally, we performed a bioinformatic analysis that revealed the antigenicity potential of some of those proteins. This analysis is the first study to identify common proteins at the plasma membrane of different T. foetus isolates that could be targets for alternative diagnostic or vaccine techniques in the future.
Collapse
Affiliation(s)
- Maria B Rivero
- Laboratorio de Biología Molecular, Inmunología y Microbiología (LaBIM), Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero, Argentina
- Facultad de Ciencias Médicas (FCM-UNSE), Santiago del Estero, Argentina
| | - Andrés M Alonso
- Laboratorio de Parásitos Anaerobios, Instituto Tecnológico Chascomús (INTECH), CONICET-UNSAM, B7130IWA, Chascomús, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), Buenos Aires, Argentina
| | - Maria E Abdala
- Laboratorio de Biología Molecular, Inmunología y Microbiología (LaBIM), Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero, Argentina
- Facultad de Ciencias Médicas (FCM-UNSE), Santiago del Estero, Argentina
- Facultad de Agronomía y Agroindustrias (FAyA-UNSE), Santiago del Estero, Argentina
| | - Melchor E Luque
- Laboratorio de Biología Molecular, Inmunología y Microbiología (LaBIM), Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero, Argentina
- Facultad de Ciencias Médicas (FCM-UNSE), Santiago del Estero, Argentina
- Facultad de Agronomía y Agroindustrias (FAyA-UNSE), Santiago del Estero, Argentina
| | - Pedro G Carranza
- Laboratorio de Biología Molecular, Inmunología y Microbiología (LaBIM), Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero, Argentina
- Facultad de Ciencias Médicas (FCM-UNSE), Santiago del Estero, Argentina
- Facultad de Agronomía y Agroindustrias (FAyA-UNSE), Santiago del Estero, Argentina
| | - Veronica M Coceres
- Laboratorio de Parásitos Anaerobios, Instituto Tecnológico Chascomús (INTECH), CONICET-UNSAM, B7130IWA, Chascomús, Argentina.
- Escuela de Bio y Nanotecnologías (UNSAM), Buenos Aires, Argentina.
| | - Fernando D Rivero
- Laboratorio de Biología Molecular, Inmunología y Microbiología (LaBIM), Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero, Argentina.
- Facultad de Ciencias Médicas (FCM-UNSE), Santiago del Estero, Argentina.
- Facultad de Agronomía y Agroindustrias (FAyA-UNSE), Santiago del Estero, Argentina.
| |
Collapse
|
4
|
Bongiorni Galego G, Tasca T. Infinity war: Trichomonas vaginalis and interactions with host immune response. MICROBIAL CELL (GRAZ, AUSTRIA) 2023; 10:103-116. [PMID: 37125086 PMCID: PMC10140678 DOI: 10.15698/mic2023.05.796] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/07/2023] [Accepted: 03/21/2023] [Indexed: 05/02/2023]
Abstract
Trichomonas vaginalis is the pathological agent of human trichomoniasis. The incidence is 156 million cases worldwide. Due to the increasing resistance of isolates to approved drugs and clinical complications that include increased risk in the acquisition and transmission of HIV, cervical and prostate cancer, and adverse outcomes during pregnancy, increasing our understanding of the pathogen's interaction with the host immune response is essential. Production of cytokines and cells of innate immunity: Neutrophils and macrophages are the main cells involved in the fight against the parasite, while IL-8, IL-6 and TNF-α are the most produced cytokines in response to this infection. Clinical complications: T. vaginalis increases the acquisition of HIV, stimulates the invasiveness and growth of prostate cells, and generates an inflammatory environment that may lead to preterm birth. Endosymbiosis: Mycoplasma hominis increased cytotoxicity, growth, and survival rate of the parasite. Purinergic signaling: NTPD-ases and ecto-5'-nucleotidase helps in parasite survival by modulating the nucleotides levels in the microenvironment. Antibodies: IgG was detected in serum samples of rodents infected with isolates from symptomatic patients as well as patients with symptoms. However, antibody production does not protect against a reinfection. Vaccine candidate targets: The transient receptor potential- like channel of T. vaginalis (TvTRPV), cysteine peptidase, and α-actinin are currently cited as candidate targets for vaccine development. In this context, the understanding of mechanisms involved in the host-T. vaginalis interaction that elicit the immune response may contribute to the development of new targets to combat trichomoniasis.
Collapse
Affiliation(s)
- Giulia Bongiorni Galego
- Grupo de Pesquisa em Tricomonas, Faculdade de Farmácia e Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre, 90610-000, Rio Grande do Sul, Brazil
| | - Tiana Tasca
- Grupo de Pesquisa em Tricomonas, Faculdade de Farmácia e Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Ipiranga, 2752, Porto Alegre, 90610-000, Rio Grande do Sul, Brazil
- * Corresponding Author: Tiana Tasca, Avenida Ipiranga, 2752. 90610-000. Porto Alegre, Rio Grande do Sul, Brazil; Tel: +555133085325;
| |
Collapse
|
5
|
Multiple Regulations of Parasitic Protozoan Viruses: A Double-Edged Sword for Protozoa. mBio 2023; 14:e0264222. [PMID: 36633419 PMCID: PMC9973342 DOI: 10.1128/mbio.02642-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Parasite infections affect human and animal health significantly and contribute to a major burden on the global economy. Parasitic protozoan viruses (PPVs) affect the protozoan parasites' morphology, phenotypes, pathogenicity, and growth rates. This discovery provides an opportunity to develop a novel preventive and therapeutic strategy for parasitic protozoan diseases (PPDs). Currently, there is greater awareness regarding PPVs; however, knowledge of viruses and their associations with host diseases remains limited. Parasite-host interactions become more complex owing to PPVs; however, few studies have investigated underlying viral regulatory mechanisms in parasites. In this study, we reviewed relevant studies to identify studies that investigated PPV development and life cycles, the triangular association between viruses, parasites, and hosts, and the effects of viruses on protozoan pathogenicity. This study highlights that viruses can alter parasite biology, and viral infection of parasites may exacerbate the adverse effects of virus-containing parasites on hosts or reduce parasite virulence. PPVs should be considered in the prevention of parasitic epidemics and outbreaks, although their effects on the host and the complexity of the triangular association between PPVs, protozoans, and hosts remain unclear. IMPORTANCE PPVs-based regulation of parasitic protozoa can provide a theoretical basis and direction for PPD prevention and control, although PPVs and PPV regulatory mechanisms remain unclear. In this review, we investigated the differences between PPVs and the unique properties of each virus regarding virus discovery, structures, and life cycles, focused on the Trichomonas vaginalis virus, Giardia lamblia virus, Leishmania RNA virus, and the Cryptosporidium parvum virus 1. The triangular association between PPVs, parasitic protozoa, and hosts reveals the "double-edged sword" property of PPVs, which maintains a balance between parasitic protozoa and hosts in both positive and negative respects. These studies discuss the complexity of parasitic protozoa and their co-existence with hosts and suggest novel pathways for using PPVs as tools to gain a deeper understanding of protozoal infection and treatment.
Collapse
|
6
|
Prevalence and Genotype of Trichomonas vaginalis among Men in Xinxiang City, Henan Province, China. J Trop Med 2023; 2023:4119956. [PMID: 36895425 PMCID: PMC9991476 DOI: 10.1155/2023/4119956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/28/2023] [Accepted: 02/09/2023] [Indexed: 03/05/2023] Open
Abstract
Trichomonas vaginalis (T. vaginalis) could cause trichomoniasis through sexual transmission, which was globally distributed. In this study, the prevalence and phylogenetic analyses of T. vaginalis among men in Xinxiang were conducted. From October 2018 to December 2019, a total of 634 male clinical samples were collected, including 254 samples of semen, 43 samples of prostate fluid, and 337 samples of urine. These samples were examined by nested PCR and a total of 32 (5.05%) T. vaginalis-positive samples were detected. Among these samples, the positive rates of T. vaginalis in semen, prostate fluid, and urine were 7.87% (20/254), 4.65% (2/43), and 2.97% (10/337), respectively. Three actin genes were successfully isolated and sequenced from the 32 positive DNA samples, and the analysis of the sequence and phylogenetic tree showed that the three actin gene sequences exhibited 99.7%-100% homology to the published actin gene sequence (EU076580) in NCBI, and the T. vaginalis strains in the three positive samples were identified as genotype E. Our results demonstrate a notable genotype of T. vaginalis in the male population and provide insight into the performance of these genetic markers in the molecular epidemiology of trichomoniasis. However, further studies are needed to research the association between the genotype and the pathogenicity of T. vaginalis.
Collapse
|
7
|
The L-Rhamnose Biosynthetic Pathway in Trichomonas vaginalis: Identification and Characterization of UDP-D-Glucose 4,6-dehydratase. Int J Mol Sci 2022; 23:ijms232314587. [PMID: 36498914 PMCID: PMC9741107 DOI: 10.3390/ijms232314587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Trichomonas vaginalis is the causative agent of one of the most widespread sexually transmitted diseases in the world. The adhesion of the parasite to the vaginal epithelial cells is mediated by specific proteins and by a complex glycan structure, the lipoglycan (TvLG), which covers the pathogen surface. L-rhamnose is an important component of TvLG, comprising up to 40% of the monosaccharides. Thus, the inhibition of its production could lead to a severe alteration in the TvLG structure, making the L-rhamnose biosynthetic pathway an attractive pharmacologic target. We report the identification and characterization of the first committed and limiting step of the L-rhamnose biosynthetic pathway, UDP-D-glucose 4,6-dehydratase (UGD, EC 4.2.1.76). The enzyme shows a strong preference for UDP-D-glucose compared to dTDP-D-glucose; we propose that the mechanism underlying the higher affinity for the UDP-bound substrate is mediated by the differential recognition of ribose versus the deoxyribose of the nucleotide moiety. The identification of the enzymes responsible for the following steps of the L-rhamnose pathway (epimerization and reduction) was more elusive. However, sequence analyses suggest that in T. vaginalis L-rhamnose synthesis proceeds through a mechanism different from the typical eukaryotic pathways, displaying intermediate features between the eukaryotic and prokaryotic pathways and involving separate enzymes for the epimerase and reductase activities, as observed in bacteria. Altogether, these results form the basis for a better understanding of the formation of the complex glycan structures on TvLG and the possible use of L-rhamnose biosynthetic enzymes for the development of selective inhibitors.
Collapse
|
8
|
Local cytokine/chemokine profiles in BALB/c and C57BL/6 mice in response to T. vaginalis infection. Exp Parasitol 2022; 239:108287. [PMID: 35660531 DOI: 10.1016/j.exppara.2022.108287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 02/27/2022] [Accepted: 05/30/2022] [Indexed: 11/19/2022]
Abstract
Trichomonas vaginalis is the causative agent of Trichomoniasis (a sexually transmitted infection). Recent reports have shown that stimulation of cellular immunity can reduce trichomoniasis infection. Animal studies are essential to understanding the pathogenesis of infection and developing new potential drugs and vaccines to treat the infection. Therefore, we have tried to understand the pathogenesis of T. vaginalis infection by investigating the differences in the expression of chemokine/cytokine levels in vaginal and cervical tissues of BALB/c and C57BL/6 mice. Different pathological symptoms, like desquamation, neutrophil infiltration, and hemorrhage, were recorded in BALB/c and C57BL/6 in response to T. vaginalis infection. Vaginal and cervical tissues of BALB/c showed these symptoms on 2nd dpi, which became severe on 7th dpi and turned to mild or normal till 14th dpi compared to C57BL/6 strain. Immunohistochemistry in the vagina and cervical tissues of BALB/c and C57BL/6 mice was done to assess cytokines at different time intervals post-infection. Significant expression of Interleukin-1β (IL-1β) (a pro-inflammatory cytokine) was found in BALB/c compared to the C57BL/6 mice, on 7th dpi and 2nd dpi in vaginal and cervical tissues, respectively. Higher expression of MIP-2 (neutrophil chemoattractant) was observed in the vaginal tissues of BALB/c mice on 7th dpi compared to the C57BL/6 group. In addition, higher expression of TGF-β (immune-suppressor) was observed on 7th dpi in the vaginal tissue of BALB/c mice. The present study demonstrates that more pathological signs of T. vaginalis infection developed in BALB/c mice than C57BL/6 mice. Also, significant levels of IL-1β and MIP-2 were measured in BALB/c mice in response to T. vaginalis compared to C57BL/6.
Collapse
|
9
|
Trichomonas vaginalis triggers neutrophil extracellular traps reducing parasite integrity and growth. Parasitol Res 2022; 121:1355-1367. [PMID: 35258690 DOI: 10.1007/s00436-022-07475-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/21/2022] [Indexed: 10/18/2022]
Abstract
Trichomoniasis-caused by the parasite Trichomonas vaginalis-is associated with a high inflammatory process that may contribute to the risk of suffering from other medical complications. Our study focused on the in vitro interaction of T. vaginalis with human neutrophils because these are the most abundant cells implicated in the characteristic inflammatory process of trichomoniasis. This study showed that T. vaginalis and its surface glycoconjugates (lipophosphoglycan and/or lipoglycan) induced the formation of human neutrophil extracellular traps (NETs). After the trichomonad-neutrophil interaction, parasite integrity was at 32.9%, and the subsequent parasite growth was at 35.2% compared to those of control trophozoites (100%) incubated under the same conditions without neutrophils. In the presence of an antibody against the TLR-4 receptor, DNase I or micrococcal nuclease (MNase), neutrophils reduced the DNA fibres of the NETs and the amount of extracellular DNA, allowing a higher subsequent growth of T. vaginalis, at 52% with the anti-TLR-4 antibody and 62.6% with the enzymes. These results indicated that T. vaginalis induced the formation of extracellular traps by human neutrophils and, because of the interaction with neutrophils and NETs, parasite integrity and growth decreased.
Collapse
|
10
|
Fichorova RN, DeLong AK, Cu-Uvin S, King CC, Jamieson DJ, Klein RS, Sobel JD, Vlahov D, Yamamoto HS, Mayer KH. Protozoan-Viral-Bacterial Co-Infections Alter Galectin Levels and Associated Immunity Mediators in the Female Genital Tract. Front Cell Infect Microbiol 2021; 11:649940. [PMID: 34422675 PMCID: PMC8375472 DOI: 10.3389/fcimb.2021.649940] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/29/2021] [Indexed: 01/05/2023] Open
Abstract
Co-infections with sexually transmittable pathogens are common and more likely in women with disturbed vaginal bacteriome. Among those pathogens, the protozoan parasite Trichomonas vaginalis (TV) is most common after accounting for the highly persistent DNA viruses human papillomavirus (HPV) and genital herpes. The parasitic infection often concurs with the dysbiotic syndrome diagnosed as bacterial vaginosis (BV) and both are associated with risks of superimposed viral infections. Yet, the mechanisms of microbial synergisms in evading host immunity remain elusive. We present clinical and experimental evidence for a new role of galectins, glycan-sensing family of proteins, in mixed infections. We assessed participants of the HIV Epidemiology Research Study (HERS) at each of their incident TV visits (223 case visits) matched to controls who remained TV-negative throughout the study. Matching criteria included age, race, BV (by Nugent score), HIV status, hysterectomy, and contraceptive use. Non-matched variables included BV status at 6 months before the matched visit, and variables examined at baseline, within 6 months of and/or at the matched visit e.g. HSV-2, HPV, and relevant laboratory and socio-demographic parameters. Conditional logistic regression models using generalized estimating equations calculated odds ratios (OR) for incident TV occurrence with each log10 unit higher cervicovaginal concentration of galectins and cytokines. Incident TV was associated with higher levels of galectin-1, galectin-9, IL-1β and chemokines (ORs 1.53 to 2.91, p <0.001). Galectin-9, IL-1β and chemokines were up and galectin-3 down in TV cases with BV or intermediate Nugent versus normal Nugent scores (p <0.001). Galectin-9, IL-1β and chemokines were up in TV-HIV and down in TV-HPV co-infections. In-vitro, TV synergized with its endosymbiont Trichomonasvirus (TVV) and BV bacteria to upregulate galectin-1, galectin-9, and inflammatory cytokines. The BV-bacterium Prevotella bivia alone and together with TV downregulated galectin-3 and synergistically upregulated galectin-1, galectin-9 and IL-1β, mirroring the clinical findings of mixed TV–BV infections. P. bivia also downregulated TVV+TV-induced anti-viral response e.g. IP-10 and RANTES, providing a mechanism for conducing viral persistence in TV-BV co-infections. Collectively, the experimental and clinical data suggest that galectin-mediated immunity may be dysregulated and exploited by viral–protozoan–bacterial synergisms exacerbating inflammatory complications from dysbiosis and sexually transmitted infections.
Collapse
Affiliation(s)
- Raina N Fichorova
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Allison K DeLong
- Center for Statistical Sciences, School of Public Health, Brown University, Providence, RI, United States
| | - Susan Cu-Uvin
- Department of Obstetrics and Gynecology, Brown University, The Miriam Hospital, Providence, RI, United States
| | - Caroline C King
- National Center for Chronic Disease Prevention and Health Promotion/Division of Reproductive Health, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Denise J Jamieson
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Robert S Klein
- Hudson Infectious Diseases Associates, Briarcliff Manor, NY, United States
| | - Jack D Sobel
- Division of Infectious Diseases, School of Medicine, Wayne State University, Detroit, MI, United States
| | - David Vlahov
- Department of Community Health Systems, School of Nursing, University of California at San Francisco, San Francisco, CA, United States
| | - Hidemi S Yamamoto
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Kenneth H Mayer
- The Fenway Institute, Fenway Health, Boston, MA, United States.,Department of Medicine, Beth Israel Deaconess Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Han Y, Liu Z, Chen T. Role of Vaginal Microbiota Dysbiosis in Gynecological Diseases and the Potential Interventions. Front Microbiol 2021; 12:643422. [PMID: 34220737 PMCID: PMC8249587 DOI: 10.3389/fmicb.2021.643422] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
Vaginal microbiota dysbiosis, characterized by the loss of Lactobacillus dominance and increase of microbial diversity, is closely related to gynecological diseases; thus, intervention on microbiota composition is significant and promising in the treatment of gynecological diseases. Currently, antibiotics and/or probiotics are the mainstay of treatment, which show favorable therapeutic effects but also bring problems such as drug resistance and high recurrence. In this review, we discuss the role of vaginal microbiota dysbiosis in various gynecological infectious and non-infectious diseases, as well as the current and potential interventions.
Collapse
Affiliation(s)
- Yiwen Han
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
12
|
HIV susceptibility in women: The roles of genital inflammation, sexually transmitted infections and the genital microbiome. J Reprod Immunol 2021; 145:103291. [PMID: 33647576 DOI: 10.1016/j.jri.2021.103291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/29/2021] [Accepted: 02/12/2021] [Indexed: 12/24/2022]
Abstract
Given that heterosexual transmission of HIV across the genital mucosa is the most common route of infection in women, an in-depth understanding of the biological mechanisms associated with HIV risk in the female genital tract (FGT) is essential for effective control of the epidemic. Genital pro-inflammatory cytokines are well-described biological co-factors to HIV risk. Increased levels of pro-inflammatory cytokines in the FGT have been associated with a 3-fold higher-risk of acquiring HIV, presumably through involvement in barrier compromise and the recruitment of highly activated HIV target cells to the site of initial viral infection and replication. Sexually transmitted infections (STIs) and bacterial vaginosis (BV) are suggested possible contributors to genital inflammation in the FGT, and this, coupled with the relationship between genital inflammation and HIV risk, underscores the importance of effective treatment of STI and BV in the promotion of women's health. In most low- and middle-income countries, STIs are treated syndromically, a practice providing rapid treatment without identifying the infection source. However, this approach has been associated with over-diagnosis and the overuse of drugs. Further, because many women with STIs are asymptomatic, syndromic management also fails to treat a vast proportion of infected women. Although several studies have explored the role of STIs and the vaginal microbiome on genital inflammation and HIV risk, the impact of STI and BV management on genital inflammation remains poorly understood. This review aimed to collate the evidence on how BV and STI management efforts affect genital inflammation and the genital microbiome in women.
Collapse
|
13
|
Bhakta SB, Moran JA, Mercer F. Neutrophil interactions with the sexually transmitted parasite Trichomonas vaginalis: implications for immunity and pathogenesis. Open Biol 2020; 10:200192. [PMID: 32873151 PMCID: PMC7536067 DOI: 10.1098/rsob.200192] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Trichomoniasis is the third most common sexually transmitted infection in humans and is caused by the protozoan parasite, Trichomonas vaginalis (Tv). Pathogenic outcomes are more common in women and generally include mild vaginitis or cervicitis. However, more serious effects associated with trichomoniasis include adverse reproductive outcomes. Like other infectious agents, pathogenesis from Tv infection is predicted to be the result of both parasite and host factors. At the site of infection, neutrophils are the most abundant immune cells present and probably play key roles in both parasite clearance and inflammatory pathology. Here, we discuss the evidence that neutrophils home to the site of Tv infection, kill the parasite, and that in some circumstances, parasites possibly evade neutrophil-directed killing. In vitro, the parasite is killed by neutrophils using a novel antimicrobial mechanism called trogocytosis, which probably involves both innate and adaptive immunity. While mechanisms of evasion are mostly conjecture at present, the persistence of Tv infections in patients argues strongly for their existence. Additionally, many strains of Tv harbour microbial symbionts Mycoplasma hominis or Trichomonasvirus, which are both predicted to impact neutrophil responses against the parasite. Novel research tools, especially animal models, will help to reveal the true outcomes of many factors involved in neutrophil-Tv interactions during trichomoniasis.
Collapse
Affiliation(s)
| | | | - Frances Mercer
- Department of Biological Sciences, California State Polytechnic University, Pomona, CA, USA
| |
Collapse
|
14
|
Yadav S, Verma V, Singh Dhanda R, Yadav M. Insights into the toll-like receptors in sexually transmitted infections. Scand J Immunol 2020; 93:e12954. [PMID: 32762084 DOI: 10.1111/sji.12954] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 07/10/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are like soldiers of an innate immune system, which protects vital biological processes against invading pathogens. TLR signalling pathways help in the removal of pathogens and mediate well-established inflammatory processes. However, these processes may also aid in the development or augmentation of an infection or an autoimmune disease. Recent studies have delineated TLR polymorphism's role in the loss of function, making hosts more resistant or vulnerable to the development of an infection. In this review, we have discussed the association of TLRs with sexually transmitted infections (STIs), especially to the pathogen-specific ligands. We have also assessed the impact on TLR downstream signalling and the maintenance of cellular homeostasis during immune responses. Besides, we have discussed the role of TLRs single nucleotide polymorphisms in various STIs. Since TLRs are known to play a part in defence mechanisms and in aiding infections therefore, a thorough understanding of TLRs structure and molecular mechanisms is required to explain how they can influence the outcome of an STI. Such a strategy may lead to the development of novel and useful immunotherapeutic approaches to control pathogen progression and prevent transmission.
Collapse
Affiliation(s)
- Sonal Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | - Vivek Verma
- Dr. B. R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | | | - Manisha Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, Delhi, India.,Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
15
|
Morales-Luna L, Hernández-Ochoa B, Ramírez-Nava EJ, Martínez-Rosas V, Ortiz-Ramírez P, Fernández-Rosario F, González-Valdez A, Cárdenas-Rodríguez N, Serrano-Posada H, Centeno-Leija S, Arreguin-Espinosa R, Cuevas-Cruz M, Ortega-Cuellar D, Pérez de la Cruz V, Rocha-Ramírez LM, Sierra-Palacios E, Castillo-Rodríguez RA, Vega-García V, Rufino-González Y, Marcial-Quino J, Gómez-Manzo S. Characterizing the Fused TvG6PD::6PGL Protein from the Protozoan Trichomonas vaginalis, and Effects of the NADP + Molecule on Enzyme Stability. Int J Mol Sci 2020; 21:E4831. [PMID: 32650494 PMCID: PMC7402283 DOI: 10.3390/ijms21144831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/30/2022] Open
Abstract
This report describes a functional and structural analysis of fused glucose-6-phosphate dehydrogenase dehydrogenase-phosphogluconolactonase protein from the protozoan Trichomonas vaginalis (T. vaginalis). The glucose-6-phosphate dehydrogenase (g6pd) gene from T. vaginalis was isolated by PCR and the sequence of the product showed that is fused with 6pgl gene. The fused Tvg6pd::6pgl gene was cloned and overexpressed in a heterologous system. The recombinant protein was purified by affinity chromatography, and the oligomeric state of the TvG6PD::6PGL protein was found as tetramer, with an optimal pH of 8.0. The kinetic parameters for the G6PD domain were determined using glucose-6-phosphate (G6P) and nicotinamide adenine dinucleotide phosphate (NADP+) as substrates. Biochemical assays as the effects of temperature, susceptibility to trypsin digestion, and analysis of hydrochloride of guanidine on protein stability in the presence or absence of NADP+ were performed. These results revealed that the protein becomes more stable in the presence of the NADP+. In addition, we determined the dissociation constant for the binding (Kd) of NADP+ in the protein and suggests the possible structural site in the fused TvG6PD::6PGL protein. Finally, computational modeling studies were performed to obtain an approximation of the structure of TvG6PD::6PGL. The generated model showed differences with the GlG6PD::6PGL protein (even more so with human G6PD) despite both being fused.
Collapse
Affiliation(s)
- Laura Morales-Luna
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico; (L.M.-L.); (E.J.R.-N.); (V.M.-R.); (P.O.-R.); (F.F.-R.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, 06720 Ciudad de México, Mexico;
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, 11340 Ciudad de México, Mexico
| | - Edson Jiovany Ramírez-Nava
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico; (L.M.-L.); (E.J.R.-N.); (V.M.-R.); (P.O.-R.); (F.F.-R.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico
| | - Víctor Martínez-Rosas
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico; (L.M.-L.); (E.J.R.-N.); (V.M.-R.); (P.O.-R.); (F.F.-R.)
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, 11340 Ciudad de México, Mexico
| | - Paulina Ortiz-Ramírez
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico; (L.M.-L.); (E.J.R.-N.); (V.M.-R.); (P.O.-R.); (F.F.-R.)
| | - Fabiola Fernández-Rosario
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico; (L.M.-L.); (E.J.R.-N.); (V.M.-R.); (P.O.-R.); (F.F.-R.)
| | - Abigail González-Valdez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico;
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico;
| | - Hugo Serrano-Posada
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Laboratorio de Agrobiotecnología, Tecnoparque CLQ, Universidad de Colima, Carretera los Limones-Loma de Juárez, 28629 Colima, Mexico; (H.S.-P.); (S.C.-L.)
| | - Sara Centeno-Leija
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Laboratorio de Agrobiotecnología, Tecnoparque CLQ, Universidad de Colima, Carretera los Limones-Loma de Juárez, 28629 Colima, Mexico; (H.S.-P.); (S.C.-L.)
| | - Roberto Arreguin-Espinosa
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico; (R.A.-E.); (M.C.-C.)
| | - Miguel Cuevas-Cruz
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico; (R.A.-E.); (M.C.-C.)
| | - Daniel Ortega-Cuellar
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, 04530 Secretaría de Salud, Mexico;
| | - Verónica Pérez de la Cruz
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Secretaria de Salud, 14269 Ciudad de México, Mexico;
| | - Luz María Rocha-Ramírez
- Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Dr. Márquez No. 162, Col Doctores, 06720 Delegación Cuauhtémoc, Mexico;
| | - Edgar Sierra-Palacios
- Colegio de Ciencias y Humanidades, Plantel Casa Libertad, Universidad Autónoma de la Ciudad de México, 09620 Ciudad de México, Mexico;
| | - Rosa Angélica Castillo-Rodríguez
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico;
| | - Vanesa Vega-García
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico;
| | - Yadira Rufino-González
- Laboratorio de Parasitología Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico;
| | - Jaime Marcial-Quino
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, 04530 Ciudad de México, Mexico; (L.M.-L.); (E.J.R.-N.); (V.M.-R.); (P.O.-R.); (F.F.-R.)
| |
Collapse
|
16
|
Blois SM, Verlohren S, Wu G, Clark G, Dell A, Haslam SM, Barrientos G. Role of galectin-glycan circuits in reproduction: from healthy pregnancy to preterm birth (PTB). Semin Immunopathol 2020; 42:469-486. [PMID: 32601855 PMCID: PMC7508936 DOI: 10.1007/s00281-020-00801-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
Abstract
Growing evidence suggests that galectins, an evolutionarily conserved family of glycan-binding proteins, fulfill key roles in pregnancy including blastocyst implantation, maternal-fetal immune tolerance, placental development, and maternal vascular expansion, thereby establishing a healthy environment for the growing fetus. In this review, we comprehensively present the function of galectins in shaping cellular circuits that characterize a healthy pregnancy. We describe the current understanding of galectins in term and preterm labor and discuss how the galectin-glycan circuits contribute to key immunological pathways sustaining maternal tolerance and preventing microbial infections. A deeper understanding of the glycoimmune pathways regulating early events in preterm birth could offer the broader translational potential for the treatment of this devastating syndrome.
Collapse
Affiliation(s)
- Sandra M Blois
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, AG GlycoImmunology, Berlin, Germany. .,Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Stefan Verlohren
- Department of Obstetrics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gang Wu
- Department of Life Sciences, Imperial College London, London, UK
| | - Gary Clark
- Department of Obstetrics, Gynaecology and Women's Health, University of Missouri, Columbia, Missouri, USA
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London, UK
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London, UK
| | - Gabriela Barrientos
- Laboratory of Experimental Medicine, Hospital Alemán, School of Medicine, University of Buenos Aires, CONICET, Buenos Aires, Argentina
| |
Collapse
|
17
|
Pérot P, Falguieres M, Arowas L, Laude H, Foy JP, Goudot P, Corre-Catelin N, Ungeheuer MN, Caro V, Heard I, Eloit M, Gessain A, Bertolus C, Berthet N. Investigation of viral etiology in potentially malignant disorders and oral squamous cell carcinomas in non-smoking, non-drinking patients. PLoS One 2020; 15:e0232138. [PMID: 32348362 PMCID: PMC7190135 DOI: 10.1371/journal.pone.0232138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are the seventh most frequent cancers. Among HNSCCs, oral squamous cell carcinomas (OSCCs) include several anatomical locations of the oral cavity, but exclude the oropharynx. The known risk factors for OSCCs are mainly alcohol consumption and tobacco use for at least 75-80% of cases. In addition to these risk factors, Human papillomavirus (HPV) types 16 and 18, classified as high-risk (HR) HPV genotypes, are considered as risk factors for oropharyngeal cancers, but their role in the development of OSCC remains unclear. We tested the hypothesis of viral etiology in a series of 68 well-characterized OSCCs and 14 potentially malignant disorders (PMD) in non-smoking, non-drinking (NSND) patients using broad-range, sensitive molecular methodologies. Deep-sequencing of the transcriptome did not reveal any vertebrate virus sequences other than HPV transcripts, detected in only one case. In contrast, HPV DNA was detected in 41.2% (28/68) and 35.7% (5/14) of OSCC and PMD cases, respectively. Importantly, 90.9% (30/33) of these belonged to the Betapapillomavirus genus, but no viral transcripts were detected. Finally, high-throughput sequencing revealed reads corresponding to transcripts of the Trichomonas vaginalis virus (TVV), which were confirmed by RT-PCR in two OSCCs. Our results strongly suggest that Alphapapillomavirus genotypes classified as HR are not involved in the development of OSCCs in NSND patients and that known oncogenic infectious agents are absent in these specific OSCCs. Any possible direct or indirect role of Betapapillomavirus genus members and TVV in OSCCs remains speculative and requires further investigation.
Collapse
Affiliation(s)
- Philippe Pérot
- Pathogen Discovery Laboratory, Institut Pasteur, Biology of Infection Unit, Paris, France
| | - Michaël Falguieres
- Institut Pasteur, Centre national de référence (CNR) des papillomavirus humains (HPV), Paris, France
| | - Laurence Arowas
- Institut Pasteur, Centre national de référence (CNR) des papillomavirus humains (HPV), Paris, France
| | - Hélène Laude
- Institut Pasteur, Centre national de référence (CNR) des papillomavirus humains (HPV), Paris, France
| | - Jean-Philippe Foy
- Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Patrick Goudot
- Department of Oral and Maxillo-Facial Surgery, Pitié-Salpêtrière Hospital, Paris, France
| | - Nicole Corre-Catelin
- Institut Pasteur, Clinical Investigation and Acces to Bioresources Department, Paris, France
| | - Marie-Noëlle Ungeheuer
- Institut Pasteur, Clinical Investigation and Acces to Bioresources Department, Paris, France
| | - Valérie Caro
- Institut Pasteur, Unité Environnement et risques infectieux, Cellule d’Intervention Biologique d’Urgence, Paris, France
| | - Isabelle Heard
- Institut Pasteur, Centre national de référence (CNR) des papillomavirus humains (HPV), Paris, France
| | - Marc Eloit
- Pathogen Discovery Laboratory, Institut Pasteur, Biology of Infection Unit, Paris, France
- National Veterinary School of Alfort, Paris-Est University, Maisons-Alfort, France
| | - Antoine Gessain
- Institut Pasteur, Unité d'Epidémiologie et Physiopathologie des Virus Oncogènes, CNRS UMR3569, Paris, France
| | - Chloé Bertolus
- Department of Oral and Maxillo-Facial Surgery, Pitié-Salpêtrière Hospital, Paris, France
- Sorbonne University, Paris, France
| | - Nicolas Berthet
- Institut Pasteur, Unité Environnement et risques infectieux, Cellule d’Intervention Biologique d’Urgence, Paris, France
- Institut Pasteur, Unité d'Epidémiologie et Physiopathologie des Virus Oncogènes, CNRS UMR3569, Paris, France
| |
Collapse
|
18
|
Kalia N, Singh J, Kaur M. Microbiota in vaginal health and pathogenesis of recurrent vulvovaginal infections: a critical review. Ann Clin Microbiol Antimicrob 2020; 19:5. [PMID: 31992328 PMCID: PMC6986042 DOI: 10.1186/s12941-020-0347-4] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
Recurrent vulvovaginal infections (RVVI) has not only become an epidemiological and clinical problem but also include large social and psychological consequences. Understanding the mechanisms of both commensalism and pathogenesis are necessary for the development of efficient diagnosis and treatment strategies for these enigmatic vaginal infections. Through this review, an attempt has been made to analyze vaginal microbiota (VMB) from scratch and to provide an update on its current understanding in relation to health and common RVVI i.e. bacterial vaginosis, vulvovaginal candidiaisis and Trichomoniasis, making the present review first of its kind. For this, potentially relevant studies were retrieved from data sources and critical analysis of the literature was made. Though, culture-independent methods have greatly unfolded the mystery regarding vaginal bacterial microbiome, there are only a few studies regarding the composition and diversity of vaginal mycobiome and different Trichomonas vaginalis strains. This scenario suggests a need of further studies based on comparative genomics of RVVI pathogens to improve our perceptive of RVVI pathogenesis that is still not clear (Fig. 5). Besides this, the review details the rationale for Lactobacilli dominance and changes that occur in healthy VMB throughout a women's life. Moreover, the list of possible agents continues to expand and new species recognised in both health and VVI are updated in this review. The review concludes with the controversies challenging the widely accepted dogma i.e. "VMB dominated with Lactobacilli is healthier than a diverse VMB". These controversies, over the past decade, have complicated the definition of vaginal health and vaginal infections with no definite conclusion. Thus, further studies on newly recognised microbial agents may reveal answers to these controversies. Conversely, VMB of women could be an answer but it is not enough to just look at the microbiology. We have to look at the woman itself, as VMB which is fine for one woman may be troublesome for others. These differences in women's response to the same VMB may be determined by a permutation of behavioural, cultural, genetic and various other anonymous factors, exploration of which may lead to proper definition of vaginal health and disease.
Collapse
Affiliation(s)
- Namarta Kalia
- Department of Molecular Biology & Biochemistry, Guru Nanak Dev University, Amritsar, 143005 India
| | - Jatinder Singh
- Department of Molecular Biology & Biochemistry, Guru Nanak Dev University, Amritsar, 143005 India
| | - Manpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, 143005 India
| |
Collapse
|
19
|
Malli S, Loiseau PM, Bouchemal K. Trichomonas vaginalis Motility Is Blocked by Drug-Free Thermosensitive Hydrogel. ACS Infect Dis 2020; 6:114-123. [PMID: 31713413 DOI: 10.1021/acsinfecdis.9b00243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Trichomonas vaginalis motility in biological fluids plays a prominent, but understudied, role in parasite infectivity. In this study, the ability of a thermosensitive hydrogel (pluronic F127) to physically immobilize T. vaginalis was investigated. Blocking parasite motility could prevent its attachment to the mucosa, thus reducing the acquisition of the infection. The trajectory of individual parasites was monitored by multiple particle tracking. Mean square displacement, diffusivity, and velocity were calculated from x, y coordinates during time. Major results are that T. vaginalis exhibited different types of trajectories in a diluted solution composed of lactate buffer similar to "run-and-tumble" motion reported for flagellated bacteria. The fastest T. vaginalis specimen moves with a velocity of 19 μm/s. Observation of T. vaginalis movements showed that the cell body remains rigid during swimming and that the propulsive forces necessary to generate the movement are the result of flagellar beating. Parasite motility was partially slowed down using hydroxyethylcellulose hydrogel, used as a reference for the development of vaginal microbicides, while 100% of T. vaginalis were immobile in F127 hydrogel. Once completed by biological investigations on mice, this report suggests using drug-free formulation composed of F127 as a new strategy to prevent T. vaginalis attachment to the mucosa. The concept will be extended to other flagellated organisms where the motility is driven by cilia and flagella.
Collapse
Affiliation(s)
- Sophia Malli
- Institut Galien Paris Sud, UMR CNRS 8612, Université Paris-Sud, Faculté de Pharmacie, Université Paris-Saclay, 5, rue J-B. Clément, 92296, Châtenay-Malabry, France
- Institut Galien Paris Sud, Junior member of the Institut Universitaire de France, UMR CNRS 8612, Université Paris-Sud, Faculté de Pharmacie, Université Paris-Saclay, 5, rue J-B. Clément, 92296 Châtenay-Malabry, France
| | - Philippe M. Loiseau
- Antiparasite Chemotherapy PARACHEM, Université Paris-Sud, CNRS, 5, rue J-B. Clément, 92290 Châtenay-Malabry, France
| | - Kawthar Bouchemal
- Institut Galien Paris Sud, Junior member of the Institut Universitaire de France, UMR CNRS 8612, Université Paris-Sud, Faculté de Pharmacie, Université Paris-Saclay, 5, rue J-B. Clément, 92296 Châtenay-Malabry, France
| |
Collapse
|
20
|
Kalia N, Singh J, Kaur M. Immunopathology of Recurrent Vulvovaginal Infections: New Aspects and Research Directions. Front Immunol 2019; 10:2034. [PMID: 31555269 PMCID: PMC6722227 DOI: 10.3389/fimmu.2019.02034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/12/2019] [Indexed: 12/25/2022] Open
Abstract
Recurrent vulvovaginal infections (RVVI), a devastating group of mucosal infection, are severely affecting women's quality of life. Our understanding of the vaginal defense mechanisms have broadened recently with studies uncovering the inflammatory nature of bacterial vaginosis, inflammatory responses against novel virulence factors, innate Type 17 cells/IL-17 axis, neutrophils mediated killing of pathogens by a novel mechanism, and oxidative stress during vaginal infections. However, the pathogens have fine mechanisms to subvert or manipulate the host immune responses, hijack them and use them for their own advantage. The odds of hijacking increases, due to impaired immune responses, the net magnitude of which is the result of numerous genetic variations, present in multiple host genes, detailed in this review. Thus, by underlining the role of the host immune responses in disease etiology, modern research has clarified a major hypothesis shift in the pathophilosophy of RVVI. This knowledge can further be used to develop efficient immune-based diagnosis and treatment strategies for this enigmatic disease conditions. As for instance, plasma-derived MBL replacement, adoptive T-cell, and antibody-based therapies have been reported to be safe and efficacious in infectious diseases. Therefore, these emerging immune-therapies could possibly be the future therapeutic options for RVVI.
Collapse
Affiliation(s)
- Namarta Kalia
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, India
| | - Jatinder Singh
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, India
| | - Manpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
21
|
Mwatelah R, McKinnon LR, Baxter C, Abdool Karim Q, Abdool Karim SS. Mechanisms of sexually transmitted infection-induced inflammation in women: implications for HIV risk. J Int AIDS Soc 2019; 22 Suppl 6:e25346. [PMID: 31468677 PMCID: PMC6715949 DOI: 10.1002/jia2.25346] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Globally, sexually transmitted infections (STI) affect >300 million people annually, and are a major cause of sexual and reproductive health complications in women. In this commentary, we describe how STIs interact with the immune and non-immune cells, both within and below the cervicovaginal mucosal barrier, to cause inflammation, which in turn has been associated with increased HIV acquisition risk. DISCUSSION STIs have a major impact on the female genital mucosa, which is an important biological and physical barrier that forms the first line of defence against invading microorganisms such as HIV. Pattern recognition of STI pathogens, by receptors expressed either on the cell surface or inside the cell, typically triggers inflammation at the mucosal barrier. The types of mucosal responses vary by STI, and can be asymptomatic or culminate in the formation of discharge, ulcers and/or warts. While the aim of this response is to clear the invading microbes, in many cases these responses are either evaded or cause pathology that impairs barrier integrity and increases HIV access to target cells in the sub-mucosa. In addition, innate responses to STIs can result in an increased number of immune cells, including those that are the primary targets of HIV, and may contribute to the association between STIs and increased susceptibility to HIV acquisition. Many of these cells are mediators of adaptive immunity, including tissue-resident cells that may also display innate-like functions. Bacterial vaginosis (BV) is another common cause of inflammation, and evidence for multiple interactions between BV, STIs and HIV suggest that susceptibility to these conditions should be considered in concert. CONCLUSIONS STIs and other microbes can induce inflammation in the genital tract, perturbing the normal robust function of the mucosal barrier against HIV. While the impact of STIs on the mucosal immune system and HIV acquisition is often under-appreciated, understanding their interactions of the infections with the immune responses play an important role in improving treatment and reducing the risk of HIV acquisition. The frequent sub-clinical inflammation associated with STIs underscores the need for better STI diagnostics to reverse the immunological consequences of infection.
Collapse
Affiliation(s)
- Ruth Mwatelah
- Department of Medical Microbiology and Infectious DiseasesUniversity of ManitobaWinnipegCanada
| | - Lyle R McKinnon
- Department of Medical Microbiology and Infectious DiseasesUniversity of ManitobaWinnipegCanada
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurbanSouth Africa
| | - Cheryl Baxter
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurbanSouth Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurbanSouth Africa
- Department of EpidemiologyColumbia UniversityNew YorkNYUSA
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurbanSouth Africa
- Department of EpidemiologyColumbia UniversityNew YorkNYUSA
| |
Collapse
|
22
|
Pandit H, Kale K, Yamamoto H, Thakur G, Rokade S, Chakraborty P, Vasudevan M, Kishore U, Madan T, Fichorova RN. Surfactant Protein D Reverses the Gene Signature of Transepithelial HIV-1 Passage and Restricts the Viral Transfer Across the Vaginal Barrier. Front Immunol 2019; 10:264. [PMID: 30984160 PMCID: PMC6447669 DOI: 10.3389/fimmu.2019.00264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/31/2019] [Indexed: 01/02/2023] Open
Abstract
Effective prophylactic strategy against the current epidemic of sexually transmitted HIV-1 infection requires understanding of the innate gatekeeping mechanisms at the genital mucosa. Surfactant protein D (SP-D), a member of the collectin family of proteins naturally present in the vaginal tract, is a potential HIV-1 entry inhibitor at the cellular level. Human EpiVaginal tissues compartmentalized in culture inserts were apically exposed to HIV-1 and/or a recombinant fragment of human SP-D (rfhSP-D) and viral passage was assessed in the basal chamber containing mononuclear leukocytes. To map the gene signature facilitating or resisting the transepithelial viral transfer, microarray analysis of the HIV-1 challenged EpiVaginal tissues was performed in the absence or presence of rfhSP-D. Mucosal biocompatibility of rfhSP-D was assessed ex vivo and in the standard rabbit vaginal irritation model. The passage of virus through the EpiVaginal tissues toward the underlying target cells was associated with a global epithelial gene signature including differential regulation of genes primarily involved in inflammation, tight junctions and cytoskeletal framework. RfhSP-D significantly inhibited HIV-1 transfer across the vaginal tissues and was associated with a significant reversal of virus induced epithelial gene signature. Pro-inflammatory NF-κB and mTOR transcripts were significantly downregulated, while expression of the tight junctions and cytoskeletal genes was upheld. In the absence of virus, rfhSP-D directly interacted with the EpiVaginal tissues and upregulated expression of genes related to structural stability of the cell and epithelial integrity. There was no increment in the viral acquisition by the PBMCs present in basal chambers wherein, the EpiVaginal tissues in apical chambers were treated with rfhSP-D. The effective concentrations of rfhSP-D had no effect on lactobacilli, epithelial barrier integrity and were safe on repeated applications onto the rabbit vaginal mucosa. This pre-clinical safety data, coupled with its efficacy of restricting viral passage via reversal of virus-induced gene expression of the vaginal barrier, make a strong argument for clinical trials of rfhSP-D as a topical anti-HIV microbicide.
Collapse
Affiliation(s)
- Hrishikesh Pandit
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India.,Laboratory of Genital Tract Biology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Kavita Kale
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India
| | - Hidemi Yamamoto
- Laboratory of Genital Tract Biology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Gargi Thakur
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India
| | - Sushama Rokade
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India
| | - Payal Chakraborty
- Genome Informatics Research Group, Bionivid Technology Pvt. Ltd., Bengaluru, India
| | - Madavan Vasudevan
- Genome Informatics Research Group, Bionivid Technology Pvt. Ltd., Bengaluru, India
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Taruna Madan
- Department of Innate Immunity, ICMR National Institute for Research in Reproductive Health, Mumbai, India
| | - Raina Nakova Fichorova
- Laboratory of Genital Tract Biology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
23
|
Riestra AM, Valderrama JA, Patras KA, Booth SD, Quek XY, Tsai CM, Nizet V. Trichomonas vaginalis Induces NLRP3 Inflammasome Activation and Pyroptotic Cell Death in Human Macrophages. J Innate Immun 2018; 11:86-98. [PMID: 30391945 DOI: 10.1159/000493585] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/09/2018] [Indexed: 12/16/2022] Open
Abstract
Trichomonas vaginalis is a sexually transmitted, eukaryotic parasite that causes trichomoniasis, the most common nonviral, sexually transmitted disease in the USA and worldwide. Little is known about the molecular mechanisms involved in the host immune response to this widespread parasite. Here we report that T. vaginalis induces NLRP3 inflammasome activation in human macrophages, leading to caspase-1 activation and the processing of pro-IL-1β to the mature and bioactive form of the cytokine. Using inhibitor-based approaches, we show that NLRP3 activation by T. vaginalis involves host cell detection of extracellular ATP via P2X7 receptors and potassium efflux. In addition, our data reveal that T. vaginalis inflammasome activation induces macrophage inflammatory cell death by pyroptosis, known to occur via caspase-1 cleavage of the gasdermin D protein, which assembles to form pores in the host cell membrane. We found that T. vaginalis-induced cytolysis of macrophages is attenuated in gasdermin D knockout cells. Lastly, in a murine challenge model, we detected IL-1β production in vaginal fluids in response to T. vaginalis infection in vivo. Together, our findings mechanistically dissect how T. vaginalis contributes to the production of the proinflammatory IL-1β cytokine and uncover pyroptosis as a mechanism by which the parasite can trigger host macrophage cell death.
Collapse
Affiliation(s)
- Angelica Montenegro Riestra
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - J Andrés Valderrama
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Kathryn A Patras
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Sharon D Booth
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Xing Yen Quek
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Chih-Ming Tsai
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA, .,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA,
| |
Collapse
|
24
|
Min A, Lee YA, Kim KA, Shin MH. BLT1-mediated O-GlcNAcylation is required for NOX2-dependent migration, exocytotic degranulation and IL-8 release of human mast cell induced by Trichomonas vaginalis-secreted LTB 4. Microbes Infect 2018; 20:376-384. [PMID: 29859938 DOI: 10.1016/j.micinf.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/10/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Abstract
Trichomonas vaginalis is a sexually-transmitted protozoan parasite that causes vaginitis and cervicitis. Although mast cell activation is important for provoking tissue inflammation during infection with parasites, information regarding the signaling mechanisms in mast cell activation and T. vaginalis infection is limited. O-linked N-acetylglucosamine (O-GlcNAc) is a post-translational modification of serine and threonine residues that functions as a critical regulator of intracellular signaling, regulated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). We investigated if O-GlcNAcylation was associated with mast cell activation induced by T. vaginalis-derived secretory products (TvSP). Modified TvSP collected from live trichomonads treated with the 5-lipooxygenase inhibitor AA861 inhibited migration of mast cells. This result suggested that mast cell migration was caused by stimulation of T. vaginalis-secreted leukotrienes. Using the BLT1 antagonist U75302 or BLT1 siRNA, we found that migration of mast cells was evoked via LTB4 receptor (BLT1). Furthermore, TvSP induced protein O-GlcNAcylation and OGT expression in HMC-1 cells, which was prevented by transfection with BLT1 siRNA. TvSP-induced migration, ROS generation, CD63 expression and IL-8 release were significantly suppressed by pretreatment with OGT inhibitor ST045849 or OGT siRNA. These results suggested that BLT1-mediated OGlcNAcylation was important for mast cell activation during trichomoniasis.
Collapse
Affiliation(s)
- Arim Min
- Department of Environmental Medical Biology, Institute of Tropical Medicine, South Korea; Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - Young Ah Lee
- Department of Environmental Medical Biology, Institute of Tropical Medicine, South Korea; Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - Kyeong Ah Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, South Korea
| | - Myeong Heon Shin
- Department of Environmental Medical Biology, Institute of Tropical Medicine, South Korea; Yonsei University College of Medicine, Seoul 120-752, South Korea.
| |
Collapse
|
25
|
Holm M, Morken TS, Fichorova RN, VanderVeen DK, Allred EN, Dammann O, Leviton A. Systemic Inflammation-Associated Proteins and Retinopathy of Prematurity in Infants Born Before the 28th Week of Gestation. Invest Ophthalmol Vis Sci 2017; 58:6419-6428. [PMID: 29260199 PMCID: PMC5736326 DOI: 10.1167/iovs.17-21931] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To assess the association between systemic levels of inflammation-associated proteins and severe retinopathy of prematurity (ROP) in extremely preterm infants. Methods We collected whole blood on filter paper on postnatal days 1, 7, 14, 21, and 28 from 1205 infants born before the 28th week of gestation, and measured the concentrations of 27 inflammation-associated, angiogenic, and neurotrophic proteins. We calculated odds ratios with 95% confidence intervals for the association between top quartile concentrations of each protein and prethreshold ROP. Results During the first three weeks after birth, high concentrations of VEGF-R1, myeloperoxidase (MPO), IL-8, intercellular adhesion molecule (ICAM)-1, matrix metalloproteinase 9, erythropoietin, TNF-α, and basic fibroblast growth factor were associated with an increased risk for prethreshold ROP. On day 28, high levels of serum amyloid A, MPO, IL-6, TNF-α, TNF-R1/-R2, IL-8, and ICAM-1 were associated with an increased risk. Top quartile concentrations of the proinflammatory cytokines TNF-α and IL-6 were associated with increased risks of ROP when levels of neuroprotective proteins and growth factors, including BDNF, insulin-like growth factor 1, IGFBP-1, VEGFR-1 and -2, ANG-1 and PlGF, were not in the top quartile. In contrast, high concentrations of NT-4 and BDNF appeared protective only in infants without elevated inflammatory mediators. Conclusions Systemic inflammation during the first postnatal month was associated with an increased risk of prethreshold ROP. Elevated concentrations of growth factors, angiogenic proteins, and neurotrophins appeared to modulate this risk, and were capable of reducing the risk even in the absence of systemic inflammation.
Collapse
Affiliation(s)
- Mari Holm
- Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Pediatrics, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tora S Morken
- Department of Neuromedicine and Movement Science (INB), Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Ophthalmology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Raina N Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Deborah K VanderVeen
- Department of Ophthalmology, Children's Hospital Boston, Boston, Massachusetts, United States.,Department of Ophthalmology, Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Elizabeth N Allred
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, United States.,Department of Neurology, Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Olaf Dammann
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, United States.,Perinatal Epidemiology Unit, Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Alan Leviton
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, United States.,Department of Neurology, Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | | |
Collapse
|
26
|
Application of standard cell cultures and 3D in vitro tissue models as an effective tool in drug design and development. Pharmacol Rep 2017. [DOI: 10.1016/j.pharep.2017.03.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
27
|
Lee HY, Kim J, Ryu JS, Park SJ. Trichomonas vaginalis α-Actinin 2 Modulates Host Immune Responses by Inducing Tolerogenic Dendritic Cells via IL-10 Production from Regulatory T Cells. THE KOREAN JOURNAL OF PARASITOLOGY 2017; 55:375-384. [PMID: 28877568 PMCID: PMC5594722 DOI: 10.3347/kjp.2017.55.4.375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 12/25/2022]
Abstract
Trichomonas vaginalis is a pathogen that triggers severe immune responses in hosts. T. vaginalis α-actinin 2, Tvα-actinin 2, has been used to diagnose trichomoniasis. This study was undertaken to examine the role of Tvα-actinin 2 as an antigenic molecule to induce immune responses from humans. Western blot analysis using anti-Tvα-actinin 2 antibodies indicated its presence in the secreted proteins of T. vaginalis. ELISA was employed to measure cytokine production by vaginal epithelial cells, prostate cells, mouse dendritic cells (DCs), or T cells stimulated with T. vaginalis or Tvα-actinin 2 protein. Both T. vaginalis and rTvα-actinin 2 induced cytokine production from epithelial cell lines, including IL-10. Moreover, CD4+CD25− regulatory T cells (Treg cells) incubated with rTvα-actinin 2-treated DCs produced high levels of IL-10. These data indicate that Tvα-actinin 2 modulates immune responses via IL-10 production by Treg cells.
Collapse
Affiliation(s)
- Hye-Yeon Lee
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Juri Kim
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jae-Sook Ryu
- Department of Environmental Biology and Medical Parasitology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Soon-Jung Park
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
28
|
Adenosine reduces reactive oxygen species and interleukin-8 production by Trichomonas vaginalis-stimulated neutrophils. Purinergic Signal 2017; 13:569-577. [PMID: 28879644 DOI: 10.1007/s11302-017-9584-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 08/23/2017] [Indexed: 02/08/2023] Open
Abstract
Trichomonas vaginalis is a flagellated protozoan that affects the human urogenital tract causing 276.4 million new infections a year. The parasite elicits a vaginal mucosal infiltration of immune cells, especially neutrophils which are considered to be primarily responsible for cytological change observed at the infection site as well as the major contributor in the inflammatory response against the parasite. Extracellular nucleotides and their nucleosides are signaling compounds involved in several biological processes, including inflammation and immune responses. Once in the extracellular space, the nucleotides and nucleosides can directly activate the purinergic receptors. Herein, we investigated the involvement of purinergic signaling on the production of reactive oxygen species (ROS) and cytokines by T. vaginalis-stimulated neutrophils. Parasites were able to induce an increase in ROS and IL-8 levels while they did not promote IL-6 secretion or neutrophil elastase activity. Adenine and guanine nucleotides or nucleosides were not able to modulate ROS and cytokine production; however, when T. vaginalis-stimulated neutrophils were incubated with adenosine and adenosine deaminase inhibitor, the levels of ROS and IL-8 were significantly reduced. These immunosuppressive effects were probably a response to the higher bioavailability of adenosine found in the supernatant as result of inhibition of enzyme activity. The involvement of P1 receptors was investigated by immunofluorescence and A1 receptor was the most abundant. Our data show that the influence of purinergic signaling, specifically those effects associated with adenosine accumulation, on the modulation of production of proinflammatory mediators by T. vaginalis-stimulated neutrophils contribute to the understanding of immunological aspects of trichomoniasis.
Collapse
|
29
|
Basnet P, Jøraholmen MW, Acharya G, Skalko-Basnet N. P 25 Vesicle-based resveratrol formulation for the topical treatment of Trichomonas vaginalis caused vaginal infection and inflammation. Pregnancy Hypertens 2017. [DOI: 10.1016/j.preghy.2017.07.103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
30
|
Human infectious diseases and risk of preeclampsia: an updated review of the literature. Infection 2017; 45:589-600. [PMID: 28577241 DOI: 10.1007/s15010-017-1031-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/25/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Preeclampsia (PE) is one of the major causes of maternal and perinatal morbidity and mortality, especially in low- and middle-income countries. In recent years, a growing body of literatures suggests that infections by bacteria, viruses, and parasites and their related inflammations play an important role in the pathogenesis of PE. METHODS We searched PubMed, Google scholar, and Cochrane databases using the following search words: "infection and preeclampsia," "bacterial infection and preeclampsia," "viral infection and preeclampsia" and "parasitic infection and preeclampsia." RESULTS The literature review revealed that many bacteria including Helicobacter pylori, Chlamydia pneumonia, and those are involved in periodontal disease or urinary tract infections (UTIs) and some viral agents such as Cytomegalovirus, herpes simplex virus type-2, human immunodeficiency virus, and some parasites especially Plasmodium spp. and Toxoplasma gondii can be effective in development of PE. Inflammation responses against infections has major role in the inducement of PE. The shift of immunological cytokine profile of Th2 toward Th1 and high levels of pro-inflammatory cytokines (TNF-ɑ, IL-12, IFN-γ, etc.), increase of oxidative stress, increase of anti-angiogenic proteins, increase of vascular endothelial growth factor receptor 1 (sVEGFR1), and complement C5a are the main potential mechanisms related to infections and enhanced development of PE. CONCLUSION Thus, early diagnosis and treatment of bacterial, viral, and parasitic infections could be an effective strategy to reduce the incidence of PE.
Collapse
|
31
|
Menezes CB, Rigo GV, Bridi H, Trentin DDS, Macedo AJ, von Poser GL, Tasca T. The anti-Trichomonas vaginalis phloroglucinol derivative isoaustrobrasilol B modulates extracellular nucleotide hydrolysis. Chem Biol Drug Des 2017; 90:811-819. [PMID: 28390095 DOI: 10.1111/cbdd.13002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/30/2017] [Accepted: 04/03/2017] [Indexed: 12/20/2022]
Abstract
Trichomonas vaginalis causes trichomoniasis, a neglected sexually transmitted disease. Due to severe health consequences and treatment failure, new therapeutic alternatives are crucial. Phloroglucinols from southern Brazilian Hypericum species demonstrated anti-T. vaginalis and anti-Leishmania amazonensis activities. The modulation of biochemical pathways involved in the control of inflammatory response by ectonucleotidases, NTPDase, and ecto-5'-nucleotidase represents new targets for combating protozoa. This study investigated the activity of phloroglucinol derivatives of Hypericum species from southern Brazil against T. vaginalis as well as its ability on modulating parasite ectonucleotidases and, consequently, immune parameters through ATP and adenosine effects. Phloroglucinol derivatives screening revealed activity for isoaustrobrasilol B (IC50 38 μm) with no hemolytic activity. Although the most active compound induced cytotoxicity against a mammalian cell lineage, the in vivo model evidenced absence of toxicity. Isoaustrobrasilol B significantly inhibited NTPDase and ecto-5'-nucleotidase activities, and the immune modulation attributed to extracellular nucleotide accumulation was evaluated. The production of ROS and IL-6 by T. vaginalis-stimulated neutrophils was not affected by the treatment. Conversely, IL-8 levels were significantly enhanced. The associative mechanism of trophozoites death and ectonucleotidases modulation by isoaustrobrasilol B may increase the susceptibility of T. vaginalis to host innate immune cell like neutrophils consequently, contributing to parasite clearance.
Collapse
Affiliation(s)
- Camila Braz Menezes
- Laboratório de Pesquisa em Parasitologia, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Graziela Vargas Rigo
- Laboratório de Pesquisa em Parasitologia, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Henrique Bridi
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Danielle da Silva Trentin
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Alexandre José Macedo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Faculdade de Farmácia e Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gilsane Lino von Poser
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tiana Tasca
- Laboratório de Pesquisa em Parasitologia, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
32
|
Olmos-Ortiz LM, Barajas-Mendiola MA, Barrios-Rodiles M, Castellano LE, Arias-Negrete S, Avila EE, Cuéllar-Mata P. Trichomonas vaginalisexosome-like vesicles modify the cytokine profile and reduce inflammation in parasite-infected mice. Parasite Immunol 2017; 39. [DOI: 10.1111/pim.12426] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/17/2017] [Indexed: 12/27/2022]
Affiliation(s)
- L. M. Olmos-Ortiz
- Departamento de Biología; Universidad de Guanajuato; Guanajuato Mexico
| | | | - M. Barrios-Rodiles
- Center for Systems Biology; Lunenfeld-Tanenbaum Research Institute; Mount Sinai Hospital; Toronto ON Canada
| | - L. E. Castellano
- Departamento de Ingenierías Química, Electrónica y Biomédica; Universidad de Guanajuato; León México
| | - S. Arias-Negrete
- Departamento de Biología; Universidad de Guanajuato; Guanajuato Mexico
| | - E. E. Avila
- Departamento de Biología; Universidad de Guanajuato; Guanajuato Mexico
| | - P. Cuéllar-Mata
- Departamento de Biología; Universidad de Guanajuato; Guanajuato Mexico
| |
Collapse
|
33
|
Trichomonas vaginalis infection in symbiosis with Trichomonasvirus and Mycoplasma. Res Microbiol 2017; 168:882-891. [PMID: 28366838 DOI: 10.1016/j.resmic.2017.03.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/19/2017] [Accepted: 03/20/2017] [Indexed: 02/04/2023]
Abstract
Trichomonas vaginalis is a protozoan with an extracellular obligatory parasitic lifestyle exclusively adapted to the human urogenital tract and responsible for nearly a quarter billion sexually transmitted infections worldwide each year. This review focuses on symbiotic Trichomonasvirus and mycoplasmas carried by the protozoan, their molecular features and their role in altering the human vaginal microbiome and the immunopathogenicity of the parasite. Improved diagnostics and larger clinical interventional studies are needed to confirm the causative role of protozoan symbionts in the variable clinical presentation of trichomoniasis and its morbid sequelae, including adverse reproductive outcome, susceptibility to viral infections and cancer.
Collapse
|
34
|
Menezes CB, Tasca T. Trichomoniasis immunity and the involvement of the purinergic signaling. Biomed J 2016; 39:234-243. [PMID: 27793265 PMCID: PMC6138788 DOI: 10.1016/j.bj.2016.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/27/2016] [Accepted: 06/30/2016] [Indexed: 12/31/2022] Open
Abstract
Innate and adaptive immunity play a significant role in trichomoniasis, the most common non-viral sexually transmitted disease worldwide. In the urogenital tract, innate immunity is accomplished by a defense physical barrier constituted by epithelial cells, mucus, and acidic pH. During infection, immune cells, antimicrobial peptides, cytokines, chemokines, and adaptive immunity evolve in the reproductive tract, and a proinflammatory response is generated to eliminate the invading extracellular pathogen Trichomonas vaginalis. However, the parasite has developed complex evolutionary mechanisms to evade the host immune response through cysteine proteases, phenotypic variation, and molecular mimicry. The purinergic system constitutes a signaling cellular net where nucleotides and nucleosides, enzymes, purinoceptors and transporters are involved in almost all cells and tissues signaling pathways, especially in central and autonomic nervous systems, endocrine, respiratory, cardiac, reproductive, and immune systems, during physiological as well as pathological processes. The involvement of the purinergic system in T. vaginalis biology and infection has been demonstrated and this review highlights the participation of this signaling pathway in the parasite immune evasion strategies.
Collapse
Affiliation(s)
- Camila Braz Menezes
- Parasitology Research Laboratory, Pharmacy Faculty, Federal University of Rio Grande do Sul, Brazil
| | - Tiana Tasca
- Parasitology Research Laboratory, Pharmacy Faculty, Federal University of Rio Grande do Sul, Brazil.
| |
Collapse
|
35
|
Cytokine response of human THP-1 macrophages to Trichomonas tenax. Exp Parasitol 2016; 169:77-80. [PMID: 27497807 DOI: 10.1016/j.exppara.2016.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 07/13/2016] [Accepted: 07/24/2016] [Indexed: 01/09/2023]
Abstract
Trichomonas tenax is a protozoan that inhabits the oral cavity of humans, most often those with poor oral hygiene. Although T. tenax is widely considered a commensal, recent studies have suggested a pathogenic role for the protozoan in persons with periodontitis. Here we investigated the capacity of T. tenax to induce pro-inflammatory cytokine secretion in human macrophages, with the idea that elicitation of inflammation may be one mechanism by which T. tenax contributes to oral pathology. Human THP-1 cells differentiated to the macrophage phenotype (dTHP-1) were incubated with live or sonicated T. tenax at trophozoite:dTHP-1 ratios of 1:5, 1:10, and 1:20. Culture media removed from the wells after 4, 8, and 16 h of stimulation were assayed by ELISA for tumor necrosis factor alpha, interleukin-1 beta, interleukin-8, and the immunoregulatory cytokine interleukin-10. Live T. tenax trophozoites failed to induce production of any of the cytokines tested, regardless of trophozoite:dTHP-1 cell ratio or length of co-incubation. T. tenax lysates stimulated interleukin-8 synthesis, but only after 16 h of incubation at the 1:5 trophozoite:dTHP-1 cell ratio. These results suggest that pro-inflammatory cytokine synthesis by human macrophages in direct response to T. tenax contributes little to oral pathology.
Collapse
|
36
|
Roditi I. The languages of parasite communication. Mol Biochem Parasitol 2016; 208:16-22. [DOI: 10.1016/j.molbiopara.2016.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 05/16/2016] [Indexed: 10/21/2022]
|
37
|
Menezes CB, Frasson AP, Tasca T. Trichomoniasis - are we giving the deserved attention to the most common non-viral sexually transmitted disease worldwide? MICROBIAL CELL (GRAZ, AUSTRIA) 2016; 3:404-419. [PMID: 28357378 PMCID: PMC5354568 DOI: 10.15698/mic2016.09.526] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/18/2016] [Indexed: 02/03/2023]
Abstract
ETIOLOGY Trichomonas vaginalis is the etiologic agent of trichomoniasis, the most common non-viral sexually transmitted disease (STD) in the world. Transmission: Trichomoniasis is transmitted by sexual intercourse and transmission via fomites is rare. Epidemiology, incidence and prevalence: The WHO estimates an incidence of 276 million new cases each year and prevalence of 187 million of infected individuals. However, the infection is not notifiable. Pathology/Symptomatology: The T. vaginalis infection results in a variety of clinical manifestations - in most cases the patients are asymptomatic, but some may develop signs typically associated to the disease. Importantly, the main issue concerning trichomoniasis is its relationship with serious health consequences such as cancer, adverse pregnancy outcomes, infertility, and HIV acquisition. Molecular mechanisms of infection: To achieve success in parasitism trichomonads develop a complex process against the host cells that includes dependent- and independent-contact mechanisms. This multifactorial pathogenesis includes molecules such as soluble factors, secreted proteinases, adhesins, lipophosphoglycan that culminate in cytoadherence and cytotoxicity against the host cells. Treatment and curability: The treatment with metronidazole or tinidazole is recommended; however, cure failures remain problematic due to noncompliance, reinfection and/or lack of treatment of sexual partners, inaccurate diagnosis, or drug resistance. Therefore, new therapeutic alternatives are urgently needed. Protection: Strategies for protection including sexual behavior, condom usage, and therapy have not contributed to the decrease on disease prevalence, pointing to the need for innovative approaches. Vaccine development has been hampered by the lack of long-lasting humoral immunity associated to the absence of good animal models.
Collapse
Affiliation(s)
- Camila Braz Menezes
- Laboratório de Pesquisa em Parasitologia, Faculdade de Farmácia,
Universidade Federal do Rio Grande do Sul. Porto Alegre, Rio Grande do Sul, Brazil
| | - Amanda Piccoli Frasson
- Laboratório de Pesquisa em Parasitologia, Faculdade de Farmácia,
Universidade Federal do Rio Grande do Sul. Porto Alegre, Rio Grande do Sul, Brazil
| | - Tiana Tasca
- Laboratório de Pesquisa em Parasitologia, Faculdade de Farmácia,
Universidade Federal do Rio Grande do Sul. Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
38
|
Tolbert MK, Gookin JL. Mechanisms of Tritrichomonas foetus Pathogenicity in Cats with Insights from Venereal Trichomonosis. J Vet Intern Med 2016; 30:516-26. [PMID: 26946069 PMCID: PMC4913604 DOI: 10.1111/jvim.13920] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/26/2015] [Accepted: 02/09/2016] [Indexed: 12/18/2022] Open
Abstract
Almost 20 years has passed since trichomonosis was first recognized as a potential cause of diarrhea in domestic cats. Despite progress in confirming disease causation, developing means for diagnosis, and identifying approaches to treatment of the infection, we still know very little about how this parasite causes diarrhea. With increasing recognition of resistance of trichomonosis to treatment with 5‐nitroimidazole drugs, new treatment strategies based on an understanding of disease pathogenesis are needed. In this review, lessons learned from the pathogenesis of venereal trichomonosis in people and cattle are applied to clinical observations of trichomonosis in cats in effort to generate insight into areas where further research may be beneficial.
Collapse
Affiliation(s)
- M K Tolbert
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN
| | - J L Gookin
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| |
Collapse
|
39
|
Fichorova RN, Yamamoto HS, Fashemi T, Foley E, Ryan S, Beatty N, Dawood H, Hayes GR, St-Pierre G, Sato S, Singh BN. Trichomonas vaginalis Lipophosphoglycan Exploits Binding to Galectin-1 and -3 to Modulate Epithelial Immunity. J Biol Chem 2016; 291:998-1013. [PMID: 26589797 PMCID: PMC4705417 DOI: 10.1074/jbc.m115.651497] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 10/29/2015] [Indexed: 11/06/2022] Open
Abstract
Trichomoniasis is the most common non-viral sexually transmitted infection caused by the vaginotropic extracellular protozoan parasite Trichomonas vaginalis. The infection is recurrent, with no lasting immunity, often asymptomatic, and linked to pregnancy complications and risk of viral infection. The molecular mechanisms of immune evasion by the parasite are poorly understood. We demonstrate that galectin-1 and -3 are expressed by the human cervical and vaginal epithelial cells and act as pathogen-recognition receptors for the ceramide phosphoinositol glycan core (CPI-GC) of the dominant surface protozoan lipophosphoglycan (LPG). We used an in vitro model with siRNA galectin knockdown epithelial clones, recombinant galectins, clinical Trichomonas isolates, and mutant protozoan derivatives to dissect the function of galectin-1 and -3 in the context of Trichomonas infection. Galectin-1 suppressed chemokines that facilitate recruitment of phagocytes, which can eliminate extracellular protozoa (IL-8) or bridge innate to adaptive immunity (MIP-3α and RANTES (regulated on activation normal T cell expressed and secreted)). Silencing galectin-1 increased and adding exogenous galectin-1 suppressed chemokine responses to Trichomonas or CPI-GC/LPG. In contrast, silencing galectin-3 reduced IL-8 response to LPG. Live Trichomonas depleted the extracellular levels of galectin-3. Clinical isolates and mutant Trichomonas CPI-GC that had reduced affinity to galectin-3 but maintained affinity to galectin-1 suppressed chemokine expression. Thus via CPI-GC binding, Trichomonas is capable of regulating galectin bioavailability and function to the benefit of its parasitic survival. These findings suggest novel approaches to control trichomoniasis and warrant further studies of galectin-binding diversity among clinical isolates as a possible source for symptom disparity in parasitic infections.
Collapse
Affiliation(s)
- Raina N Fichorova
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115,
| | - Hidemi S Yamamoto
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Titilayo Fashemi
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Evan Foley
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Stanthia Ryan
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Noah Beatty
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Hassan Dawood
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Gary R Hayes
- the Departments of Biochemistry and Molecular Biology and Obstetrics and Gynecology, State University of New York Upstate Medical University, Syracuse, New York 13210, and
| | - Guillaume St-Pierre
- the Laboratory of Glycobiology and Bioimaging, Research Centre for Infectious Diseases, Faculty of Medicine, Laval University, Quebec, Quebec G1V 4G2, Canada
| | - Sachiko Sato
- the Laboratory of Glycobiology and Bioimaging, Research Centre for Infectious Diseases, Faculty of Medicine, Laval University, Quebec, Quebec G1V 4G2, Canada
| | - Bibhuti N Singh
- the Departments of Biochemistry and Molecular Biology and Obstetrics and Gynecology, State University of New York Upstate Medical University, Syracuse, New York 13210, and
| |
Collapse
|
40
|
Oliveira AS, Ferrão AR, Pereira FM, Martinez-de-Oliveira J, Palmeira-de-Oliveira A. Trichomonas vaginalis: An Updated Overview Towards Diagnostic Improvement. Acta Parasitol 2016; 61:10-21. [PMID: 26751868 DOI: 10.1515/ap-2016-0002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 09/15/2015] [Indexed: 11/15/2022]
Abstract
The protozoan Trichomonas vaginalis (TV) is responsible for trichomonosis, a sexually transmitted disease (STD) with a significant incidence worldwide. This infection is one of the most common non-viral STDs, representing almost 50% of all curable STDs. Trichomonosis has an incidence of 180 million new cases worldwide. Nowadays, the 'gold standard' for TV diagnosis remains the use of in vitro cultures combined with daily visual microscopic evaluations, which is a time-consuming and low sensitive method. Recent diagnostic methodologies include imunocromatographic assays and molecular biology techniques. The use of the latter has improved enormously the sensitivity and specificity of TV diagnosis, despite, however, none being unable to identify the presence of live parasites. By understanding the biology, the pathogenesis, the proteomic profile and its relation with the parasite's virulence mechanisms, new possibilities towards diagnostic techniques can arise. This review covers various important aspects of vaginal trichomonosis from the parasite's biology and virulence to recent improvements in diagnostic techniques and also metabolic and protein discoveries.
Collapse
|
41
|
Heiss C, Wang Z, Black I, Azadi P, Fichorova RN, Singh BN. Novel structural features of the immunocompetent ceramide phospho-inositol glycan core from Trichomonas vaginalis. Carbohydr Res 2015; 419:51-9. [PMID: 26671321 DOI: 10.1016/j.carres.2015.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/28/2015] [Accepted: 11/02/2015] [Indexed: 12/23/2022]
Abstract
The ceramide phosphoinositol glycan core (CPI-GC) of the lipophosphoglycan of Trichomonas vaginalis is a major virulent factor of this common genitourinary parasite. While its carbohydrate composition has been reported before, its structure has remained largely unknown. We isolated the glycan portions of CPI-GC by nitrous acid deamination and hydrofluoric acid treatment and investigated their structures by methylation analysis and 1- and 2-D NMR. We found that the α-anomer of galactose is a major constituent of CPI-GC. The β-anomer was found exclusively at the non-reducing end of CPI-GC side chains. Furthermore the data showed that the rhamnan backbone is more complex than previously thought and that the inositol residue at the reducing end is linked to a 4-linked α-glucuronic acid (GlcA) residue. This appears to be the most striking and novel feature of this GPI-anchor type molecule.
Collapse
Affiliation(s)
- Christian Heiss
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA.
| | - Zhirui Wang
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Ian Black
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Raina N Fichorova
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA
| | - Bibhuti N Singh
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, 750 E Adams St, Syracuse, NY 13210, USA.
| |
Collapse
|
42
|
Fichorova RN, Chen PL, Morrison CS, Doncel GF, Mendonca K, Kwok C, Chipato T, Salata R, Mauck C. The Contribution of Cervicovaginal Infections to the Immunomodulatory Effects of Hormonal Contraception. mBio 2015; 6:e00221-15. [PMID: 26330510 PMCID: PMC4556810 DOI: 10.1128/mbio.00221-15] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/23/2015] [Indexed: 01/15/2023] Open
Abstract
UNLABELLED Particular types of hormonal contraceptives (HCs) and genital tract infections have been independently associated with risk of HIV-1 acquisition. We examined whether immunity in women using injectable depot medroxyprogesterone acetate (DMPA), combined oral contraceptives (COC), or no HCs differs by the presence of cervicovaginal infections. Immune mediators were quantified in cervical swabs from 832 HIV-uninfected reproductive-age Ugandans and Zimbabweans. Bacterial infections and HIV were diagnosed by PCR, genital herpes serostatus by enzyme-linked immunosorbent assay (ELISA), altered microflora by Nugent score, and Trichomonas vaginalis and Candida albicans infection by wet mount. Generalized linear models utilizing Box-Cox-Power transformation examined associations between levels of mediators, infection status, and HCs. In no-HC users, T. vaginalis was associated with broadest spectrum of aberrant immunity (higher interleukin 1β [IL-1β], IL-8, macrophage inflammatory protein 3α [MIP-3α], β-defensin 2 [BD2], and IL-1 receptor antigen [IL-1RA]). In women with a normal Nugent score and no genital infection, compared to the no-HC group, COC users showed higher levels of IL-1β, IL-6, IL-8, and IL-1RA, while DMPA users showed higher levels of RANTES and lower levels of BD2, both associated with HIV seroconversion. These effects of COC were blunted in the presence of gonorrhea, chlamydia, trichomoniasis, candidiasis, and an abnormal Nugent score; however, RANTES was increased among COC users with herpes, chlamydia, and abnormal Nugent scores. The effect of DMPA was exacerbated by lower levels of IL-1RA in gonorrhea, chlamydia, or herpes, SLPI in gonorrhea, and IL-1β, MIP-3α, and IL-1RA/IL1β ratio in trichomoniasis. Thus, the effects of HC on cervical immunity depend on the genital tract microenvironment, and a weakened mucosal barrier against HIV may be a combined resultant of genital tract infections and HC use. IMPORTANCE In this article, we show that in young reproductive-age women most vulnerable to HIV, hormonal contraceptives are associated with altered cervical immunity in a manner dependent on the presence of genital tract infections. Through altered immunity, hormones may predispose women to bacterial and viral pathogens; conversely, a preexisting specific infection or disturbed vaginal microbiota may suppress the immune activation by levonorgestrel or exacerbate the suppressed immunity by DMPA, thus increasing HIV risk by their cumulative action. Clinical studies assessing the effects of contraception on HIV susceptibility and mucosal immunity may generate disparate results in populations that differ by microbiota background or prevalence of undiagnosed genital tract infections. A high prevalence of asymptomatic infections among HC users that remain undiagnosed and untreated raises even more concerns in light of their combined effects on biomarkers of HIV risk. The molecular mechanisms of the vaginal microbiome's simultaneous interactions with hormones and HIV remain to be elucidated.
Collapse
Affiliation(s)
- Raina N Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | - Kevin Mendonca
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Robert Salata
- Case Western Reserve University, Cleveland, Ohio, USA
| | | |
Collapse
|
43
|
Quintero CA, Tudela JG, Damiani MT. Rho GTPases as pathogen targets: Focus on curable sexually transmitted infections. Small GTPases 2015; 6:108-18. [PMID: 26023809 DOI: 10.4161/21541248.2014.991233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pathogens have evolved highly specialized mechanisms to infect hosts. Several microorganisms modulate the eukaryotic cell surface to facilitate their engulfment. Once internalized, they hijack the molecular machinery of the infected cell for their own benefit. At different stages of phagocytosis, particularly during invasion, certain pathogens manipulate pathways governed by small GTPases. In this review, we focus on the role of Rho proteins on curable, sexually transmitted infections caused by Chlamydia trachomatis, Neisseria gonorrhoeae, Trichomonas vaginalis and Treponema pallidum. Despite the high, worldwide frequencies of these sexually-transmitted diseases, very little is known about the strategies developed by these microorganisms to usurp key eukaryotic proteins that control intracellular signaling and actin dynamics. Improved knowledge of these molecular mechanisms will contribute to the elucidation of how these clinically important pathogens manipulate intracellular processes and parasitize their hosts.
Collapse
Affiliation(s)
- Cristián A Quintero
- a Laboratory of Phagocytosis and Intracellular Trafficking; IHEM-CONICET; School of Medicine; University of Cuyo ; Mendoza , Argentina
| | | | | |
Collapse
|
44
|
Lin WC, Chang WT, Chang TY, Shin JW. The Pathogenesis of Human Cervical Epithelium Cells Induced by Interacting with Trichomonas vaginalis. PLoS One 2015; 10:e0124087. [PMID: 25901354 PMCID: PMC4406492 DOI: 10.1371/journal.pone.0124087] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 02/25/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Trichomonas vaginalis is a protozoan parasite that occurs in the urogenital-vaginal tract and is the primary causative agent of trichomoniasis, a common sexually transmitted disease in humans. The aggregation of this protozoan tends to destroy epithelial cells and induce pathogenesis. PRINCIPAL FINDINGS This study cultured T. vaginalis and human cervical epithelial cells (Z172) under the same conditions in the experiments. Following co-culturing for ten hours, the protozoans became attached to Z172, such that the cells presented a round shape and underwent shrinkage. Time-lapse recording and flow cytometry on interacted Z172 revealed that 70% had been disrupted, 18% presented a necrosis-like morphology and 8% showed signs of apoptosis. Gene expression profiling revealed in the seven inflammatory Z172 genes as well as in T. vaginalis genes that code for adhesion proteins 65 and 65-1. SIGNIFICANCE These results suggest that cytopathogenic effects progress while Z172 is in contact with T. vaginalis, and the resulting morphological changes can be categorized as disruption.
Collapse
Affiliation(s)
- Wei-Chen Lin
- Department of Parasitology, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Wei-Ting Chang
- Department of Parasitology, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Tsuey-Yu Chang
- Department of Parasitology, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Jyh-Wei Shin
- Department of Parasitology, National Cheng Kung University, Tainan, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
45
|
Elevated endogenous erythropoietin concentrations are associated with increased risk of brain damage in extremely preterm neonates. PLoS One 2015; 10:e0115083. [PMID: 25793991 PMCID: PMC4368546 DOI: 10.1371/journal.pone.0115083] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/18/2014] [Indexed: 12/17/2022] Open
Abstract
Background We sought to determine, in very preterm infants, whether elevated perinatal erythropoietin (EPO) concentrations are associated with increased risks of indicators of brain damage, and whether this risk differs by the co-occurrence or absence of intermittent or sustained systemic inflammation (ISSI). Methods Protein concentrations were measured in blood collected from 786 infants born before the 28th week of gestation. EPO was measured on postnatal day 14, and 25 inflammation-related proteins were measured weekly during the first 2 postnatal weeks. We defined ISSI as a concentration in the top quartile of each of 25 inflammation-related proteins on two separate days a week apart. Hypererythropoietinemia (hyperEPO) was defined as the highest quartile for gestational age on postnatal day 14. Using logistic regression and multinomial logistic regression models, we compared risks of brain damage among neonates with hyperEPO only, ISSI only, and hyperEPO+ISSI, to those who had neither hyperEPO nor ISSI, adjusting for gestational age. Results Newborns with hyperEPO, regardless of ISSI, were more than twice as likely as those without to have very low (< 55) Mental (OR 2.3; 95% CI 1.5-3.5) and/or Psychomotor (OR 2.4; 95% CI 1.6-3.7) Development Indices (MDI, PDI), and microcephaly at age two years (OR 2.4; 95%CI 1.5-3.8). Newborns with both hyperEPO and ISSI had significantly increased risks of ventriculomegaly, hemiparetic cerebral palsy, microcephaly, and MDI and PDI < 55 (ORs ranged from 2.2-6.3), but not hypoechoic lesions or other forms of cerebral palsy, relative to newborns with neither hyperEPO nor ISSI. Conclusion hyperEPO, regardless of ISSI, is associated with elevated risks of very low MDI and PDI, and microcephaly, but not with any form of cerebral palsy. Children with both hyperEPO and ISSI are at higher risk than others of very low MDI and PDI, ventriculomegaly, hemiparetic cerebral palsy, and microcephaly.
Collapse
|
46
|
Soto-Rivera CL, Fichorova RN, Allred EN, Van Marter LJ, Shah B, Martin CR, Agus MSD, Leviton A. The relationship between TSH and systemic inflammation in extremely preterm newborns. Endocrine 2015; 48:595-602. [PMID: 24996532 PMCID: PMC4285685 DOI: 10.1007/s12020-014-0329-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 06/05/2014] [Indexed: 01/06/2023]
Abstract
Elevated thyrotropin (TSH) levels in critically ill extremely premature infants have been attributed to transient hypothyroidism of prematurity or non-thyroidal illness syndrome. We evaluated the hypothesis that relatively high TSH levels in the first 2 postnatal weeks follow recovery from systemic inflammation, similar to non-thyroidal illness syndrome. The study was conducted in 14 Neonatal Intensive Care Units and approved by each individual Institutional Review Board. We measured the concentrations of TSH and 25 inflammation-related proteins in blood spots obtained on postnatal days 1, 7, and 14. We then evaluated the temporal relationships between hyperthyrotropinemia (HTT), defined as a TSH concentration in the highest quartile for gestational age and postnatal day, and elevated levels of inflammation-related proteins. 880 newborns less than 28 weeks of gestation were included. Elevated concentrations of inflammation-related proteins during the first or second week did not precede day-14 HTT. Systemic inflammation on day 7 was associated with day-14 HTT only if inflammation persisted through the end of the 2 week period. HTT frequently accompanied elevated concentrations of inflammation-related proteins on the same day. The hypothesis that HTT follows recovery from severe illness, defined as preceding systemic inflammation, is weakly supported by our study. Our findings more prominently support the hypothesis that TSH conveys information about concomitant inflammation in the extremely premature newborn.
Collapse
Affiliation(s)
- Carmen L Soto-Rivera
- Department of Medicine, Divisions of Endocrinology and Medicine Critical Care, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA,
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Kusdian G, Gould SB. The biology of Trichomonas vaginalis in the light of urogenital tract infection. Mol Biochem Parasitol 2015; 198:92-9. [PMID: 25677793 DOI: 10.1016/j.molbiopara.2015.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/29/2015] [Accepted: 01/30/2015] [Indexed: 12/20/2022]
Abstract
The human pathogen Trichomonas vaginalis is a parasitic protist. It is a representative of the eukaryotic supergroup Excavata that includes a few other protist parasites such as Leishmania, Trypanosoma and Giardia. T. vaginalis is the agent of trichomoniasis and in the US alone, one in 30 women tests positive for this parasite. The disease is easily treated with metronidazole in most cases, but resistant strains are on the rise. The biology of Trichomonas is remarkable: it includes for example the biggest protist genome currently sequenced, the expression of about 30,000 protein-encoding genes (and thousands of lncRNAs and pseudogenes), anaerobic hydrogenosomes, rapid morphogenesis during infection, the secretion of exosomes, the manipulation of the vaginal microbiota through phagocytosis and a rich strain-dependent diversity. Here we provide an overview of Trichomonas biology with a focus on its relevance for pathogenicity and summarise the most recent advances. With some respect this parasite offers the opportunity to serve as a model system to study certain aspects of cell and genome biology, but tackling the complex biology of T. vaginalis is also important to better understand the effects that accompany infection and direct symptoms.
Collapse
Affiliation(s)
- Gary Kusdian
- Institute for Molecular Evolution, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Sven B Gould
- Institute for Molecular Evolution, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
48
|
Russell MW, Whittum-Hudson J, Fidel PL, Hook EW, Mestecky J. Immunity to Sexually Transmitted Infections. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00112-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
Malla N, Goyal K, Dhanda RS, Yadav M. Immunity in urogenital protozoa. Parasite Immunol 2014; 36:400-8. [PMID: 25201404 DOI: 10.1111/pim.12114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/20/2014] [Indexed: 01/15/2023]
Abstract
Innate and adaptive immunity play a significant role in urogenital infections. Innate immunity is provided by the epithelial cells and mucus lining along with acidic pH, which forms a strong physical barrier against the pathogens in female reproductive tract. Cells of innate immune system, antimicrobial peptides, cytokines, chemokines and adaptive immunity in the reproductive tract are evolved during infection, and a pro-inflammatory response is generated to fight against the invading pathogen Trichomonas vaginalis, a primary urogenital protozoa, the etiological agent of human trichomoniasis, a curable sexually transmitted infection. The involvement of the urogenital tract by other protozoal infections such as P. falciparum, Trypanosoma, Leishmania, Toxoplasma, Entamoeba histolytica and Acanthamoeba infection is rarely reported. Trichomonas induce pro-inflammatory and immunosuppressive responses in infected subjects. Multifactorial pathogenic mechanisms including parasite adherence, cysteine proteases, lipophosphoglycan, free radical, cytokine generation and Toll-like receptors appear to interplay with the induction of local and systemic immune responses that ultimately determine the outcome of the infection. However, the involvement of urogenital pathogen-specific immune mechanisms and effect of normal local resident flora on the outcome (symptomatic vs. asymptomatic) of infection are poorly understood. Moreover, immune interactions in trichomoniasis subjects co-infected with bacterial and viral pathogens need to be elucidated.
Collapse
Affiliation(s)
- N Malla
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | |
Collapse
|
50
|
Edwards T, Burke P, Smalley H, Hobbs G. Trichomonas vaginalis: Clinical relevance, pathogenicity and diagnosis. Crit Rev Microbiol 2014; 42:406-17. [PMID: 25383648 DOI: 10.3109/1040841x.2014.958050] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Trichomonas vaginalis is the etiological agent of trichomoniasis, the most prevalent non-viral sexually transmitted disease worldwide. Trichomoniasis is a widespread, global health concern and occurring at an increasing rate. Infections of the female genital tract can cause a range of symptoms, including vaginitis and cervicitis, while infections in males are generally asymptomatic. The relatively mild symptoms, and lack of evidence for any serious sequelae, have historically led to this disease being under diagnosed, and under researched. However, growing evidence that T. vaginalis infection is associated with other disease states with high morbidity in both men and women has increased the efforts to diagnose and treat patients harboring this parasite. The pathology of trichomoniasis results from damage to the host epithelia, caused by a variety of processes during infection and recent work has highlighted the complex interactions between the parasite and host, commensal microbiome and accompanying symbionts. The commercial release of a number of nucleic acid amplification tests (NAATs) has added to the available diagnostic options. Immunoassay based Point of Care testing is currently available, and a recent initial evaluation of a NAAT Point of Care system has given promising results, which would enable testing and treatment in a single visit.
Collapse
Affiliation(s)
- Thomas Edwards
- a Liverpool John Moores University, School of Pharmacy and Biomolecular Sciences , Byrom Street , Liverpool , UK
| | - Patricia Burke
- a Liverpool John Moores University, School of Pharmacy and Biomolecular Sciences , Byrom Street , Liverpool , UK
| | - Helen Smalley
- a Liverpool John Moores University, School of Pharmacy and Biomolecular Sciences , Byrom Street , Liverpool , UK
| | - Glyn Hobbs
- a Liverpool John Moores University, School of Pharmacy and Biomolecular Sciences , Byrom Street , Liverpool , UK
| |
Collapse
|