Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
Parasit Vectors ; 17(1): 271, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926778

ABSTRACT

BACKGROUND: Trichomonosis is a common infection in small animals, mostly manifesting in gastrointestinal symptoms such as diarrhea. Although oral trichomonads are also known, the species found colonizing the large intestine are more frequently detected protozoa. METHODS: In the present study, four wildcats, 94 domestic cats, and 25 dogs, originating from 18 different locations in Hungary, were investigated for the presence of oral and large intestinal trichomonads based on the 18S rRNA gene and ITS2. RESULTS: All oral swabs were negative by polymerase chain reaction (PCR). However, Tritrichomonas foetus was detected in a high proportion among tested domestic cats (13.8%) and dogs (16%), and Pentatrichomonas hominis only in two domestic cats. In addition, a novel Tritrichomonas genotype was identified in one cat, probably representing a new species that was shown to be phylogenetically most closely related to Tritrichomonas casperi described recently from mice. All positive dogs and half of the positive cats showed symptoms, and among cats, the most frequent breed was the Ragdoll. CONCLUSIONS: With molecular methods, this study evaluated the prevalence of oral and intestinal trichomonads in clinical samples of dogs and cats from Hungary, providing the first evidence of T. foetus in dogs of this region. In contrast to literature data, P. hominis was more prevalent in cats than in dogs. Finally, a hitherto unknown large intestinal Tritrichomonas species (closely related to T. casperi) was shown to be present in a cat, raising two possibilities. First, this novel genotype might have been a rodent-associated pseudoparasite in the relevant cat. Otherwise, the cat was actually infected, thus suggesting the role of a predator-prey link in the evolution of this trichomonad.


Subject(s)
Cat Diseases , Dog Diseases , Phylogeny , Protozoan Infections, Animal , RNA, Ribosomal, 18S , Animals , Cats , Dogs , Cat Diseases/parasitology , Cat Diseases/epidemiology , Dog Diseases/parasitology , Dog Diseases/epidemiology , Protozoan Infections, Animal/parasitology , Protozoan Infections, Animal/epidemiology , Hungary/epidemiology , RNA, Ribosomal, 18S/genetics , Tritrichomonas/genetics , DNA, Protozoan/genetics , Female , Male , Genotype , Prevalence , Polymerase Chain Reaction , Tritrichomonas foetus/genetics , Tritrichomonas foetus/isolation & purification , Tritrichomonas foetus/classification
2.
ISME J ; 18(1)2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38366179

ABSTRACT

Commensal protists and gut bacterial communities exhibit complex relationships, mediated at least in part through host immunity. To improve our understanding of this tripartite interplay, we investigated community and functional dynamics between the murine protist Tritrichomonas musculus and intestinal bacteria in healthy and B-cell-deficient mice. We identified dramatic, protist-driven remodeling of resident microbiome growth and activities, in parallel with Tritrichomonas musculus functional changes, which were accelerated in the absence of B cells. Metatranscriptomic data revealed nutrient-based competition between bacteria and the protist. Single-cell transcriptomics identified distinct Tritrichomonas musculus life stages, providing new evidence for trichomonad sexual replication and the formation of pseudocysts. Unique cell states were validated in situ through microscopy and flow cytometry. Our results reveal complex microbial dynamics during the establishment of a commensal protist in the gut, and provide valuable data sets to drive future mechanistic studies.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Tritrichomonas , Animals , Mice , Eukaryota , Bacteria
3.
J Eukaryot Microbiol ; 70(6): e12989, 2023.
Article in English | MEDLINE | ID: mdl-37300520

ABSTRACT

Tritrichomonas muris is a common flagellated protist isolated from the cecum of wild rodents. This commensal protist has been shown previously to alter immune phenotypes in laboratory mice. Other trichomonads, referred to as Tritrichomonas musculis and Tritrichomonas rainier, also naturally colonize laboratory mice and cause immune alterations. This report formally describes two new trichomonads, Tritrichomonas musculus n. sp., and Tritrichomonas casperi n. sp., at the ultrastructural and molecular level. These two protists were isolated from laboratory mice and were differentiated by their size and the structure of their undulating membrane and posterior flagellum. Analysis at the 18S rRNA and trans-ITS genetic loci supported their designation as distinct species, related to T. muris. To assess the true extent of parabasalid diversity infecting laboratory mice, 135 mice bred at the National Institutes of Health (NIH) were screened using pan-parabasalid primers that amplify the trans-ITS region. Forty-four percent of mice were positive for parabasalids, encompassing a total of eight distinct sequence types. Tritrichomonas casperi and Trichomitus-like protists were dominant. T. musculus and T. rainier were also detected, but T. muris was not. Our work establishes a previously underappreciated diversity of commensal trichomonad flagellates that naturally colonize the enteric cavity of laboratory mice.


Subject(s)
Parabasalidea , Trichomonadida , Tritrichomonas , Animals , Mice , Tritrichomonas/ultrastructure , Trichomonadida/genetics , Eukaryota , Flagella/ultrastructure
4.
J Immunol ; 208(7): 1782-1789, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35256512

ABSTRACT

Commensal intestinal protozoa, unlike their pathogenic relatives, are neglected members of the mammalian microbiome. These microbes have a significant impact on the host's intestinal immune homeostasis, typically by elevating anti-microbial host defense. Tritrichomonas musculis, a protozoan gut commensal, strengthens the intestinal host defense against enteric Salmonella infections through Asc- and Il1r1-dependent Th1 and Th17 cell activation. However, the underlying inflammasomes mediating this effect remain unknown. In this study, we report that colonization with T. musculis results in an increase in luminal extracellular ATP that is followed by increased caspase activity, higher cell death, elevated levels of IL-1ß, and increased numbers of IL-18 receptor-expressing Th1 and Th17 cells in the colon. Mice deficient in either Nlrp1b or Nlrp3 failed to display these protozoan-driven immune changes and lost resistance to enteric Salmonella infections even in the presence of T. musculis These findings demonstrate that T. musculis-mediated host protection requires sensors of extracellular and intracellular ATP to confer resistance to enteric Salmonella infections.


Subject(s)
Apoptosis Regulatory Proteins , Microbiota , NLR Family, Pyrin Domain-Containing 3 Protein , Tritrichomonas , Animals , Apoptosis Regulatory Proteins/immunology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Mammals/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Symbiosis , Tritrichomonas/metabolism
5.
Int J Mol Sci ; 22(19)2021 Oct 02.
Article in English | MEDLINE | ID: mdl-34639039

ABSTRACT

The mucus layer in the intestine plays a critical role in regulation of host-microbe interactions and maintaining homeostasis. Disruptions of the mucus layer due to genetic, environmental, or immune factors may lead to inflammatory bowel diseases (IBD). IBD frequently are accompanied with infections, and therefore are treated with antibiotics. Hence, it is important to evaluate risks of antibiotic treatment in individuals with vulnerable gut barrier and chronic inflammation. Mice with a knockout of the Muc2 gene, encoding the main glycoprotein component of the mucus, demonstrate a close contact of the microbes with the gut epithelium which leads to chronic inflammation resembling IBD. Here we demonstrate that the Muc2-/- mice harboring a gut protozoan infection Tritrichomonas sp. are susceptible to an antibiotic-induced depletion of the bacterial microbiota. Suppression of the protozoan infection with efficient metronidazole dosage or L-fucose administration resulted in amelioration of an illness observed in antibiotic-treated Muc2-/- mice. Fucose is a monosaccharide presented abundantly in gut glycoproteins, including Mucin2, and is known to be involved in host-microbe interactions, in particular in microbe adhesion. We suppose that further investigation of the role of fucose in protozoan adhesion to host cells may be of great value.


Subject(s)
Fucose/metabolism , Mucin-2/deficiency , Protozoan Infections/etiology , Protozoan Infections/metabolism , Tritrichomonas/physiology , Animals , Anti-Bacterial Agents/pharmacology , Disease Susceptibility , Female , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Mortality , Protozoan Infections/drug therapy , Protozoan Infections/mortality , Tritrichomonas/classification
6.
Nat Commun ; 12(1): 3371, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099671

ABSTRACT

The role of p53 in tumor suppression has been extensively studied and well-established. However, the role of p53 in parasitic infections and the intestinal type 2 immunity is unclear. Here, we report that p53 is crucial for intestinal type 2 immunity in response to the infection of parasites, such as Tritrichomonas muris and Nippostrongylus brasiliensis. Mechanistically, p53 plays a critical role in the activation of the tuft cell-IL-25-type 2 innate lymphoid cell circuit, partly via transcriptional regulation of Lrmp in tuft cells. Lrmp modulates Ca2+ influx and IL-25 release, which are critical triggers of type 2 innate lymphoid cell response. Our results thus reveal a previously unrecognized function of p53 in regulating intestinal type 2 immunity to protect against parasitic infections, highlighting the role of p53 as a guardian of immune integrity.


Subject(s)
Immunity, Innate/immunology , Intestines/immunology , Nippostrongylus/immunology , Parasitic Diseases/immunology , Tritrichomonas/immunology , Tumor Suppressor Protein p53/immunology , Animals , Cell Line, Tumor , Eosinophils/immunology , Eosinophils/parasitology , Gene Expression Regulation , Goblet Cells/immunology , Goblet Cells/parasitology , Host-Parasite Interactions/immunology , Humans , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/parasitology , Intestines/parasitology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/physiology , Parasitic Diseases/metabolism , Parasitic Diseases/parasitology , Tritrichomonas/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Curr Protoc ; 1(3): e77, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33740294

ABSTRACT

Recent findings position tuft cells as key mediators of intestinal immunity through their production of the cytokine interleukin (IL)-25 and activation of group 2 innate lymphoid cells (ILC2s). Though tuft cells are found in numerous epithelial tissues, their phenotype and function have been best characterized in the small intestine, where robust in vivo techniques have enabled the dissection of their cellular function, ontogeny, and key signaling pathways. We describe methods for the identification, quantification, and manipulation of tuft cells, focusing on analysis of ILC2s as a readout of tuft cell function. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Ex vivo analysis of small intestinal tuft cells and ILC2 by flow cytometry Alternate Protocol: Ex vivo analysis of small intestinal tuft cells and ILC2 by flow cytometry in the context of type 2 inflammation Basic Protocol 2: Ex vivo analysis of small intestinal tuft cells by imaging of intestinal Swiss roll Basic Protocol 3: Tuft-ILC2 circuit activation by oral gavage of adult Nippostrongylus brasiliensis worms Basic Protocol 4: Circuit activation by colonization with Tritrichomonas spp. Basic Protocol 5: Circuit activation by treatment with succinate in drinking water Basic Protocol 6: Circuit activation by treatment with recombinant IL-25.


Subject(s)
Immunity, Innate , Tritrichomonas , Animals , Intestine, Small , Lymphocytes , Nippostrongylus
8.
Appl Environ Microbiol ; 86(11)2020 05 19.
Article in English | MEDLINE | ID: mdl-32198171

ABSTRACT

Our current understanding of the host-microbiota interaction in the gut is dominated by studies focused primarily on prokaryotic bacterial communities. However, there is an underappreciated symbiotic eukaryotic protistic community that is an integral part of mammalian microbiota. How commensal protozoan bacteria might interact to form a stable microbial community remains poorly understood. Here, we describe a murine protistic commensal, phylogenetically assigned as Tritrichomonas musculis, whose colonization in the gut resulted in a reduction of gut bacterial abundance and diversity in wild-type C57BL/6 mice. Meanwhile, dietary nutrient and commensal bacteria also influenced the protozoan's intestinal colonization and stability. While mice fed a normal chow diet had abundant T. musculis organisms, switching to a Western-type high-fat diet led to the diminishment of the protozoan from the gut. Supplementation of inulin as a dietary fiber to the high-fat diet partially restored the protozoan's colonization. In addition, a cocktail of broad-spectrum antibiotics rendered permissive engraftment of T. musculis even under a high-fat, low-fiber diet. Furthermore, oral administration of Bifidobacterium spp. together with dietary supplementation of inulin in the high-fat diet impacted the protozoan's intestinal engraftment in a bifidobacterial species-dependent manner. Overall, our study described an example of dietary-nutrient-dependent murine commensal protozoan-bacterium cross talk as an important modulator of the host intestinal microbiome.IMPORTANCE Like commensal bacteria, commensal protozoa are an integral part of the vertebrate intestinal microbiome. How protozoa integrate into a commensal bacterium-enriched ecosystem remains poorly studied. Here, using the murine commensal Tritrichomonas musculis as a proof of concept, we studied potential factors involved in shaping the intestinal protozoal-bacterial community. Understanding the rules by which microbes form a multispecies community is crucial to prevent or correct microbial community dysfunctions in order to promote the host's health or to treat diseases.


Subject(s)
Bacterial Physiological Phenomena , Diet, High-Fat , Gastrointestinal Microbiome/physiology , Host Microbial Interactions , Tritrichomonas/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Nutrients/physiology
9.
Immunohorizons ; 4(1): 23-32, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980480

ABSTRACT

Tuft cells are an epithelial cell type critical for initiating type 2 immune responses to parasites and protozoa in the small intestine. To respond to these stimuli, intestinal tuft cells use taste chemosensory signaling pathways, but the role of taste receptors in type 2 immunity is poorly understood. In this study, we show that the taste receptor TAS1R3, which detects sweet and umami in the tongue, also regulates tuft cell responses in the distal small intestine. BALB/c mice, which have an inactive form of TAS1R3, as well as Tas1r3-deficient C57BL6/J mice both have severely impaired responses to tuft cell-inducing signals in the ileum, including the protozoa Tritrichomonas muris and succinate. In contrast, TAS1R3 is not required to mount an immune response to the helminth Heligmosomoides polygyrus, which infects the proximal small intestine. Examination of uninfected Tas1r3-/- mice revealed a modest reduction in the number of tuft cells in the proximal small intestine but a severe decrease in the distal small intestine at homeostasis. Together, these results suggest that TAS1R3 influences intestinal immunity by shaping the epithelial cell landscape at steady-state.


Subject(s)
Epithelial Cells/immunology , Intestinal Mucosa/immunology , Intestine, Small/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Animals , Epithelial Cells/metabolism , Gastrointestinal Microbiome , Homeostasis , Ileum/immunology , Ileum/parasitology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Intestine, Small/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nematospiroides dubius/immunology , Receptors, G-Protein-Coupled/deficiency , Strongylida Infections/immunology , Strongylida Infections/parasitology , Taste/physiology , Tritrichomonas/immunology
10.
Immunity ; 49(1): 33-41.e7, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30021144

ABSTRACT

In the small intestine, type 2 responses are regulated by a signaling circuit that involves tuft cells and group 2 innate lymphoid cells (ILC2s). Here, we identified the microbial metabolite succinate as an activating ligand for small intestinal (SI) tuft cells. Sequencing analyses of tuft cells isolated from the small intestine, gall bladder, colon, thymus, and trachea revealed that expression of tuft cell chemosensory receptors is tissue specific. SI tuft cells expressed the succinate receptor (SUCNR1), and providing succinate in drinking water was sufficient to induce a multifaceted type 2 immune response via the tuft-ILC2 circuit. The helminth Nippostrongylus brasiliensis and a tritrichomonad protist both secreted succinate as a metabolite. In vivo sensing of the tritrichomonad required SUCNR1, whereas N. brasiliensis was SUCNR1 independent. These findings define a paradigm wherein tuft cells monitor microbial metabolites to initiate type 2 immunity and suggest the existence of other sensing pathways triggering the response to helminths.


Subject(s)
Immunity, Mucosal/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Succinic Acid/pharmacology , Animals , Cell Line , Female , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Intestine, Small/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/drug effects , Nippostrongylus/immunology , Nippostrongylus/metabolism , Organ Specificity , Protozoan Infections/immunology , Receptors, G-Protein-Coupled/immunology , Signal Transduction/immunology , Species Specificity , Strongylida Infections/immunology , TRPM Cation Channels/metabolism , Th2 Cells/immunology , Tritrichomonas/drug effects , Tritrichomonas/immunology , Tritrichomonas/metabolism
11.
Cell ; 174(2): 271-284.e14, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887373

ABSTRACT

The small intestinal tuft cell-ILC2 circuit mediates epithelial responses to intestinal helminths and protists by tuft cell chemosensory-like sensing and IL-25-mediated activation of lamina propria ILC2s. Small intestine ILC2s constitutively express the IL-25 receptor, which is negatively regulated by A20 (Tnfaip3). A20 deficiency in ILC2s spontaneously triggers the circuit and, unexpectedly, promotes adaptive small-intestinal lengthening and remodeling. Circuit activation occurs upon weaning and is enabled by dietary polysaccharides that render mice permissive for Tritrichomonas colonization, resulting in luminal accumulation of acetate and succinate, metabolites of the protist hydrogenosome. Tuft cells express GPR91, the succinate receptor, and dietary succinate, but not acetate, activates ILC2s via a tuft-, TRPM5-, and IL-25-dependent pathway. Also induced by parasitic helminths, circuit activation and small intestinal remodeling impairs infestation by new helminths, consistent with the phenomenon of concomitant immunity. We describe a metabolic sensing circuit that may have evolved to facilitate mutualistic responses to luminal pathosymbionts.


Subject(s)
Intestine, Small/physiology , Tritrichomonas/metabolism , Acetates/metabolism , Animals , Dietary Fiber/metabolism , Energy Metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Interleukins/genetics , Interleukins/metabolism , Intestinal Mucosa/cytology , Intestine, Small/microbiology , Intestine, Small/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbiota , Plasmids/genetics , Plasmids/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Succinic Acid/metabolism , TRPM Cation Channels/metabolism , Tritrichomonas/growth & development , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
12.
BMC Vet Res ; 13(1): 348, 2017 Nov 23.
Article in English | MEDLINE | ID: mdl-29169366

ABSTRACT

BACKGROUND: Tritrichomonads like porcine Tritrichomonas foetus (previously named Tritrichomonas suis), can commensally live in nasal cavity of pigs, but it is rare to cause pulmonary tritrichomonosis. CASE PRESENTATION: A 40-day-old piglet was presented for persistent labor breathing and diagnosed with parasite infections in the lung by analysis of bronchoalveolar lavage (BAL) under microscope. By taking advantage of next-generation sequencing approach, we found 9611 homologous tags belonging to 50 annotated genes of tritrichomonads by analysis of mRNA of the bronchoalveolar lavage with the parasite infection. Furthermore, RT-PCR and DNA sequencing analysis confirmed the presence of the tritrichomonad. FINDINGS: Here, we report a case of pulmonary tritrichomonosis in a pig. By taking advantage of next-generation sequencing approach, we found 9611 homologous tags belonging to 50 annotated genes of tritrichomonads by analysis of mRNA of the bronchoalveolar lavage with the parasite infections. Furthermore, RT-PCR and DNA sequencing analysis confirmed the presence of the tritrichomonad. CONCLUSION: Our results demonstrate that tritrichomonads like porcine Tritrichomonas foetus can cause lung infections of pigs and reveal that next-generation sequencing is potential to identify rare diseases like pulmonary tritrichomonosis in clinical.


Subject(s)
Lung Diseases/veterinary , Protozoan Infections, Animal/diagnosis , Swine Diseases/diagnosis , Tritrichomonas , Animals , Bronchoalveolar Lavage Fluid/parasitology , Genes, Protozoan/genetics , High-Throughput Nucleotide Sequencing/veterinary , Lung Diseases/diagnosis , Lung Diseases/parasitology , Microscopy/veterinary , Protozoan Infections, Animal/parasitology , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Swine , Swine Diseases/parasitology , Tritrichomonas/genetics
13.
Sci Rep ; 7(1): 5580, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28717211

ABSTRACT

Intestinal tuft cells are one of 4 secretory cell linages in the small intestine and the source of IL-25, a critical initiator of the type 2 immune response to parasite infection. When Raptor, a critical scaffold protein for mammalian target of rapamycin complex 1 (mTORC1), was acutely deleted in intestinal epithelium via Tamoxifen injection in Tritrichomonas muris (Tm) infected mice, tuft cells, IL-25 in epithelium and IL-13 in the mesenchyme were significantly reduced, but Tm burden was not affected. When Tm infected mice were treated with rapamycin, DCLK1 and IL-25 expression in enterocytes and IL-13 expression in mesenchyme were diminished. After massive small bowel resection, tuft cells and Tm were diminished due to the diet used postoperatively. The elimination of Tm and subsequent re-infection of mice with Tm led to type 2 immune response only in WT, but Tm colonization in both WT and Raptor deficient mice. When intestinal organoids were stimulated with IL-4, tuft cells and IL-25 were induced in both WT and Raptor deficient organoids. In summary, our study reveals that enterocyte specific Raptor is required for initiating a type 2 immune response which appears to function through the regulation of mTORC1 activity.


Subject(s)
Enterocytes/cytology , Intestine, Small/cytology , Protozoan Infections, Animal/immunology , Regulatory-Associated Protein of mTOR/deficiency , Sirolimus/administration & dosage , Tritrichomonas/immunology , Animals , Doublecortin-Like Kinases , Down-Regulation , Enterocytes/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Immunity, Mucosal/drug effects , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukins/genetics , Interleukins/metabolism , Intestine, Small/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protozoan Infections, Animal/drug therapy , Sirolimus/pharmacology , Tamoxifen/administration & dosage , Tamoxifen/pharmacology
14.
J Eukaryot Microbiol ; 64(1): 4-17, 2017 01.
Article in English | MEDLINE | ID: mdl-27222431

ABSTRACT

We developed nested PCR protocols and performed a multiyear survey on the prevalence of several protozoan parasites in wild northern bobwhite (Colinus virginianus) and scaled quail (Callipepla squamata) in the Rolling Plains ecoregion of Texas and Oklahoma (i.e. fecal pellets, bird intestines and blood smears collected between 2010 and 2013). Coccidia, cryptosporidia, and microsporidia were detected in 46.2%, 11.7%, and 44.0% of the samples (n = 687), whereas histomona and hematozoa were undetected. Coccidia consisted of one major and two minor Eimeria species. Cryptosporidia were represented by a major unknown Cryptosporidium species and Cryptosporidium baileyi. Detected microsporidia species were highly diverse, in which only 11% were native avian parasites including Encephalitozoon hellem and Encephalitozoon cuniculi, whereas 33% were closely related to species from insects (e.g. Antonospora, Liebermannia, and Sporanauta). This survey suggests that coccidia infections are a significant risk factor in the health of wild quail while cryptosporidia and microsporidia may be much less significant than coccidiosis. In addition, the presence of E. hellem and E. cuniculi (known to cause opportunistic infections in humans) suggests that wild quail could serve as a reservoir for human microsporidian pathogens, and individuals with compromised or weakened immunity should probably take precautions while directly handling wild quail.


Subject(s)
Bird Diseases/parasitology , Coccidia/isolation & purification , Cryptosporidium/isolation & purification , Microsporidia/isolation & purification , Microsporidiosis/veterinary , Protozoan Infections, Animal/parasitology , Quail/parasitology , Trichomonadida/isolation & purification , Tritrichomonas/isolation & purification , Animals , Bird Diseases/epidemiology , Coccidia/genetics , Colinus/parasitology , Cryptosporidiosis/epidemiology , Cryptosporidiosis/parasitology , Cryptosporidium/genetics , DNA, Protozoan/analysis , DNA, Protozoan/genetics , Feces/parasitology , Female , Male , Microsporidia/genetics , Microsporidiosis/epidemiology , Microsporidiosis/parasitology , Oklahoma/epidemiology , Polymerase Chain Reaction/methods , Protozoan Infections, Animal/diagnosis , Protozoan Infections, Animal/epidemiology , Quail/blood , Risk Factors , Surveys and Questionnaires , Texas/epidemiology , Trichomonadida/genetics , Tritrichomonas/genetics
15.
J Exp Med ; 213(13): 2841-2850, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27836928

ABSTRACT

The mammalian gastrointestinal tract hosts a diverse community of microbes including bacteria, fungi, protozoa, helminths, and viruses. Through coevolution, mammals and these microbes have developed a symbiosis that is sustained through the host's continuous sensing of microbial factors and the generation of a tolerant or pro-inflammatory response. While analyzing T cell-driven colitis in nonlittermate mouse strains, we serendipitously identified that a nongenetic transmissible factor dramatically increased disease susceptibility. We identified the protozoan Tritrichomonas muris as the disease-exacerbating element. Furthermore, experimental colonization with T. muris induced an elevated Th1 response in the cecum of naive wild-type mice and accelerated colitis in Rag1-/- mice after T cell transfer. Overall, we describe a novel cross-kingdom interaction within the murine gut that alters immune cell homeostasis and disease susceptibility. This example of unpredicted microbial priming of the immune response highlights the importance of studying trans-kingdom interactions and serves as a stark reminder of the importance of using littermate controls in all mouse research.


Subject(s)
Colitis/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Tritrichomonas/immunology , Animals , Colitis/genetics , Colitis/parasitology , Colitis/pathology , Disease Susceptibility/immunology , Disease Susceptibility/parasitology , Disease Susceptibility/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Knockout
16.
J Am Assoc Lab Anim Sci ; 55(4): 480-3, 2016.
Article in English | MEDLINE | ID: mdl-27423157

ABSTRACT

Tritrichomonas muris is occasionally identified during routine fecal screening of laboratory mice. Frequently, entire racks are affected, and because no effective treatment is available, culling of affected mice and rederivation by embryo transfer have been suggested. The current study evaluated whether treatment with ronidazole, a nitroimidazole efficacious against T. fetus infections in cats, combined with limited culling was effective against T. muris in laboratory mice (Mus musculus). A subset (n = 39) of mice were treated with ronidazole (400 mg/L in drinking water) for 15 d, after which 6 of the mice still shed T. muris. Consequently all mice in the affected rack received ronidazole (500 mg /L in drinking water) for 25 d. All mice were retested by using pooled samples, and those positive for T. muris (except for a valuable breeding pair) were culled. The remaining mice continued to receive ronidazole for another 17 d. At the end of the treatment period, all mice were tested (days 60 and 81) and were shown to be negative for T. muris. Over the following year, sentinel mice from the rack were tested every 3 mo and remained negative for tritrichomonads by fecal smear. Thus, a combination of limited culling and treatment with ronidazole in the drinking water successfully cleared research mice of infection with T. muris.


Subject(s)
Antiprotozoal Agents/administration & dosage , Protozoan Infections, Animal/drug therapy , Protozoan Infections, Animal/prevention & control , Rodent Diseases/drug therapy , Rodent Diseases/prevention & control , Ronidazole/administration & dosage , Tritrichomonas/drug effects , Animals , Disease Eradication/methods , Feces/parasitology , Mice , Protozoan Infections, Animal/parasitology , Rodent Diseases/physiopathology , Treatment Outcome
17.
Science ; 351(6279): 1329-33, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26847546

ABSTRACT

The intestinal epithelium forms an essential barrier between a host and its microbiota. Protozoa and helminths are members of the gut microbiota of mammals, including humans, yet the many ways that gut epithelial cells orchestrate responses to these eukaryotes remain unclear. Here we show that tuft cells, which are taste-chemosensory epithelial cells, accumulate during parasite colonization and infection. Disruption of chemosensory signaling through the loss of TRMP5 abrogates the expansion of tuft cells, goblet cells, eosinophils, and type 2 innate lymphoid cells during parasite colonization. Tuft cells are the primary source of the parasite-induced cytokine interleukin-25, which indirectly induces tuft cell expansion by promoting interleukin-13 production by innate lymphoid cells. Our results identify intestinal tuft cells as critical sentinels in the gut epithelium that promote type 2 immunity in response to intestinal parasites.


Subject(s)
Chemoreceptor Cells/immunology , Intestinal Diseases, Parasitic/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Microbiota/immunology , TRPM Cation Channels/immunology , Animals , Doublecortin-Like Kinases , Eosinophils/immunology , Goblet Cells/immunology , Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Immunity, Mucosal , Interleukin-13/immunology , Interleukin-17/immunology , Intestinal Diseases, Parasitic/parasitology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Serine-Threonine Kinases/immunology , Protozoan Infections/immunology , Protozoan Infections/parasitology , Signal Transduction , Taste , Transducin/genetics , Transducin/immunology , Tritrichomonas/immunology
18.
J Feline Med Surg ; 15(7): 647-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23813833

ABSTRACT

OVERVIEW: Tritrichomonas foetus is a protozoan organism that is specific to cats and can cause large bowel diarrhoea. It is distinct from other Tritrichomonas species and not considered to be zoonotic. Infection is most common in young cats from multicat households, particularly pedigree breeding catteries. DISEASE SIGNS: Affected cats show frequent fetid diarrhoea, often with mucus, fresh blood and straining, but generally remain bright and do not lose weight. DIAGNOSIS: Diagnosis of infection is usually based on direct microscopic examination of freshly voided faeces. Polymerase chain reaction (PCR) testing is more sensitive but may detect infections unrelated to diarrhoea and, therefore, requires care in interpretation. TREATMENT: The treatment of choice is ronidazole, which should be used with care as it is an unlicensed drug for cats with a narrow safety margin. Clinical signs are generally self-limiting in untreated cases, but may take months to resolve.


Subject(s)
Cat Diseases/parasitology , Protozoan Infections, Animal/parasitology , Tritrichomonas , Animals , Cat Diseases/prevention & control , Cats , Protozoan Infections, Animal/prevention & control
19.
J Vet Intern Med ; 27(4): 760-70, 2013.
Article in English | MEDLINE | ID: mdl-23701212

ABSTRACT

This review assesses the efficacy of whole cell Tritrichomonas foetus vaccine to prevent and treat trichomoniasis in beef cattle. Three databases were searched in June 2012. Eligible studies compared infection risk, open risk, and abortion risk in heifers or infection risk in bulls that received vaccine compared with no vaccine. Study results were extracted, summary effect measures were calculated, and the quality of the evidence was assessed. From 334 citations identified, 10 were relevant to the review. For heifers, there was limited evidence of moderate quality to assess the impact of vaccination on infection risk (RR, 0.89; P = .16; 95% CI, 0.76-1.05; 6 randomized and 4 nonrandomized studies; 251 animals) and open risk (RR, 0.80; P = .06; 95% CI, 0.63-1.01; 6 randomized and 5 nonrandomized studies; 570 animals). The quality of the body of work describing the impact of vaccination on abortion risk was low (summary RR, 0.57; P = .0003; 95% CI, 0.42-0.78; 3 randomized and 2 nonrandomized studies; 176 animals). The quality of evidence was very low for duration of infection (mean difference, -23.42; P = .003; 95% CI, -38.36 to -7.85; 2 randomized and 3 nonrandomized studies; 163 animals). Although the summary effect measures suggest a benefit to vaccination, due to publication bias the effect reported here is likely an over estimate of efficacy. For bull-associated outcomes, the evidence base was low or very low quality.


Subject(s)
Cattle Diseases/prevention & control , Protozoan Infections, Animal/prevention & control , Protozoan Vaccines/immunology , Tritrichomonas/immunology , Animals , Cattle
20.
Vet Parasitol ; 196(3-4): 278-82, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23618514

ABSTRACT

Tritrichomonas foetus is the causative agent of venereal trichomonosis in cattle causing infertility, pyometra and abortions. The objectives of this study were to determine the positivity rate of Tritrichomonas spp. in abomasal content of aborted foetuses from Eastern Anatolian Region of Turkey, using staining, culture and PCR methods and to present the isolates found in the region. A total of 246 abomasal content of aborted foetuses were tested and 14 of 246 (5.7%) were Tritrichomonas spp. positive only by polymerase chain reaction (PCR). Positivity was not attained by staining or culture method. Four of the positive samples in PCR were confirmed to be T. foetus by sequencing of the amplified 5.8S rRNA gene and flanking ITS regions. Nucleotide sequences of TR-Erzurum T. foetus isolates have been entered into the GenBank sequence database under accession numbers KC236423 through KC236426. This preliminary study suggests that future studies are needed on the systematic relationships and epidemiology of T. foetus isolates in the region.


Subject(s)
Abortion, Veterinary/parasitology , Cattle Diseases/parasitology , Protozoan Infections, Animal/parasitology , Tritrichomonas , Abomasum/parasitology , Abortion, Veterinary/epidemiology , Animals , Cattle , Cattle Diseases/epidemiology , Female , Polymerase Chain Reaction/veterinary , Pregnancy , Protozoan Infections, Animal/epidemiology , Turkey/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL