Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Parasit Vectors ; 16(1): 171, 2023 May 28.
Article in English | MEDLINE | ID: mdl-37246221

ABSTRACT

BACKGROUND: Heligmosomoides bakeri (often mistaken for Heligmosomoides polygyrus) is a promising model for parasitic nematodes with the key advantage of being amenable to study and manipulation within a controlled laboratory environment. While draft genome sequences are available for this worm, which allow for comparative genomic analyses between nematodes, there is a notable lack of information on its gene expression. METHODS: We generated biologically replicated RNA-seq datasets from samples taken throughout the parasitic life of H. bakeri. RNA from tissue-dwelling and lumen-dwelling worms, collected under a dissection microscope, was sequenced on an Illumina platform. RESULTS: We find extensive transcriptional sexual dimorphism throughout the fourth larval and adult stages of this parasite and identify alternative splicing, glycosylation, and ubiquitination as particularly important processes for establishing and/or maintaining sex-specific gene expression in this species. We find sex-linked differences in transcription related to aging and oxidative and osmotic stress responses. We observe a starvation-like signature among transcripts whose expression is consistently upregulated in males, which may reflect a higher energy expenditure by male worms. We detect evidence of increased importance for anaerobic respiration among the adult worms, which coincides with the parasite's migration into the physiologically hypoxic environment of the intestinal lumen. Furthermore, we hypothesize that oxygen concentration may be an important driver of the worms encysting in the intestinal mucosa as larvae, which not only fully exposes the worms to their host's immune system but also shapes many of the interactions between the host and parasite. We find stage- and sex-specific variation in the expression of immunomodulatory genes and in anthelmintic targets. CONCLUSIONS: We examine how different the male and female worms are at the molecular level and describe major developmental events that occur in the worm, which extend our understanding of the interactions between this parasite and its host. In addition to generating new hypotheses for follow-up experiments into the worm's behavior, physiology, and metabolism, our datasets enable future more in-depth comparisons between nematodes to better define the utility of H. bakeri as a model for parasitic nematodes in general.


Subject(s)
Anthelmintics , Nematoda , Parasites , Trichostrongyloidea , Animals , Male , Female , Sex Characteristics , Nematoda/genetics , Larva/genetics
2.
Front Immunol ; 13: 1020056, 2022.
Article in English | MEDLINE | ID: mdl-36569914

ABSTRACT

Introduction: Intestinal roundworms cause chronic debilitating disease in animals, including humans. Traditional experimental models of these types of infection use a large single-dose infection. However, in natural settings, hosts are exposed to parasites on a regular basis and when mice are exposed to frequent, smaller doses of Heligmosomoides polygyrus, the parasites are cleared more quickly. Whether this more effective host response has any negative consequences for the host is not known. Results: Using a trickle model of infection, we found that worm clearance was associated with known resistance-related host responses: increased granuloma and tuft cell numbers, increased levels of granuloma IgG and decreased intestinal transit time, as well as higher serum IgE levels. However, we found that the improved worm clearance was also associated with an inflammatory phenotype in and around the granuloma, increased smooth muscle hypertrophy/hyperplasia, and elevated levels of Adamts gene expression. Discussion: To our knowledge, we are the first to identify the involvement of this protein family of matrix metalloproteinases (MMPs) in host responses to helminth infections. Our results highlight the delicate balance between parasite clearance and host tissue damage, which both contribute to host pathology. When continually exposed to parasitic worms, improved clearance comes at a cost.


Subject(s)
Nematospiroides dubius , Humans , Mice , Animals , Cicatrix , Immunity , Granuloma , Inflammation
3.
Cell Mol Gastroenterol Hepatol ; 11(1): 77-98, 2021.
Article in English | MEDLINE | ID: mdl-32659381

ABSTRACT

BACKGROUND & AIMS: Alterations in intestinal MUC2 mucin and microbial diversity are closely linked with important intestinal pathologies; however, their impact on each other and on intestinal pathogenesis has been vaguely characterized. Therefore, it was of interest in this study to delineate distinct and cooperative function of commensal microbiota and the Muc2 mucus barrier in maintaining intestinal epithelial barrier function. METHODS: Muc2 mucin deficient (Muc2-/-) and sufficient (Muc2+/+) littermates were used as a model for assessing the role of Muc2. To quantify the role of the microbiota in disease pathogenesis, Muc2+/+ and Muc2-/- littermates were treated with a cocktail of antibiotics that reduced indigenous bacteria, and then fecal transplanted with littermate stool and susceptibility to dextran sulphate sodium (DSS) quantified. RESULTS: Although, Muc2+/+ and Muc2-/- littermates share similar phyla distribution as evidenced by 16S sequencing they maintain their distinctive gastrointestinal phenotypes. Basally, Muc2-/- showed low-grade colonic inflammation with high populations of inflammatory and tolerogenic immune cells that became comparable to Muc2+/+ littermates following antibiotic treatment. Antibiotics treatment rendered Muc2+/+ but not Muc2-/- littermates highly susceptibility to DSS-induced colitis that was ILC3 dependent. Muc2-/- microbiota was colitogenic to Muc2+/+ as it worsened DSS-induced colitis. Microbiota dependent inflammation was confirmed by bone-marrow chimera studies, as Muc2-/- receiving Muc2+/+ bone marrow showed no difference in their susceptibility toward DSS induced colitis. Muc2-/- microbiota exhibited presence of characteristic OTUs of specific bacterial populations that were transferrable to Muc2+/+ littermates. CONCLUSIONS: These results highlight a distinct role for Muc2 mucin in maintenance of healthy microbiota critical in shaping innate host defenses to promote intestinal homeostasis.


Subject(s)
Colitis/immunology , Disease Resistance/immunology , Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Mucin-2/metabolism , Animals , Anti-Bacterial Agents/administration & dosage , Colitis/chemically induced , Colitis/pathology , Colon/drug effects , Colon/immunology , Colon/microbiology , Colon/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Gastrointestinal Microbiome/drug effects , Host Microbial Interactions/immunology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Mucin-2/genetics
4.
Nat Commun ; 11(1): 483, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980623

ABSTRACT

Inflammatory bowel disease is associated with changes in the mucosal barrier, increased intestinal permeability, and increased risk of infections and sepsis, but the underlying mechanisms are incompletely understood. Here, we show how continuous translocation of gut microbial components affects iron homeostasis and facilitates susceptibility to inflammation-associated sepsis. A sub-lethal dose of lipopolysaccharide results in higher mortality in Mucin 2 deficient (Muc2-/-) mice, and is associated with elevated circulatory iron load and increased bacterial translocation. Translocation of gut microbial components attenuates hepatic stearoyl CoA desaturase-1 activity, a key enzyme in hepatic de novo lipogenesis. The resulting reduction of hepatic saturated and unsaturated fatty acid levels compromises plasma membrane fluidity of red blood cells, thereby significantly reducing their life span. Inflammation in Muc2-/- mice alters erythrophagocytosis efficiency of splenic macrophages, resulting in an iron-rich milieu that promotes bacterial growth. Our study thus shows that increased intestinal permeability triggers a cascade of events resulting in increased bacterial growth and risk of sepsis.


Subject(s)
Intestinal Mucosa/metabolism , Iron/metabolism , Liver/metabolism , Sepsis/metabolism , Stearoyl-CoA Desaturase/metabolism , Animals , Cell Membrane Permeability , Cytophagocytosis , Disease Models, Animal , Female , Gastrointestinal Microbiome , Inflammation/etiology , Inflammation/metabolism , Inflammation/microbiology , Intestinal Mucosa/microbiology , Iron/blood , Lipogenesis , Macrophages/metabolism , Male , Mice , Mice, Knockout , Mucin-2/deficiency , Mucin-2/genetics , Sepsis/etiology , Sepsis/microbiology
5.
Gut Microbes ; 11(1): 118-125, 2020.
Article in English | MEDLINE | ID: mdl-31091163

ABSTRACT

Entamoeba histolytica (Eh) is a protozoan parasite of humans that colonizes the outer colonic mucus layer. Under conditions not fully understood, Eh breaches innate host defenses and invades the intestinal mucosa-causing amebic colitis and liver abscess. In asymptomatic infection, Eh interacts with and feeds on resident microbiota that forms biofilms on the outer colonic mucus layer. Despite the close association between Eh and commensal microbiota, we still lack basic knowledge on whether microbiota and/or their metabolites influence Eh virulence traits critical in disease pathogenesis. In the pathogenesis of intestinal amebiasis, Eh overcomes the protective mucus layer using a combination of mucinase/glycosidase and potent mucus secretagogue activity. In this addendum, we discuss the interconnected role of a healthy mucus barrier and the role commensal microbiota play in shaping innate host defense against Eh-induced pro-inflammatory and secretory responses critical in disease pathogenesis.


Subject(s)
Dysentery, Amebic , Entamoeba histolytica , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Mucins/metabolism , Cytokines/metabolism , Dysentery, Amebic/microbiology , Dysentery, Amebic/pathology , Entamoeba histolytica/pathogenicity , Entamoeba histolytica/physiology , Epithelial Cells/metabolism , Humans , Inflammation , Intestinal Mucosa/microbiology , Intestinal Mucosa/parasitology , Liver/parasitology , Liver/pathology , Virulence
6.
Mucosal Immunol ; 13(2): 344-356, 2020 03.
Article in English | MEDLINE | ID: mdl-31772322

ABSTRACT

Even though Entamoeba histolytica (Eh)-induced host pro-inflammatory responses play a critical role in disease, we know very little about the host factors that regulate this response. Direct contact between host cell and Eh signify the highest level of danger, and to eliminate this threat, the host immune system elicits an augmented immune response. To understand the mechanisms of this response, we investigated the induction and release of the endogenous alarmin molecule high-mobility group box 1 (HMGB1) that act as a pro-inflammatory cytokine and chemoattractant during Eh infection. Eh in contact with macrophage induced a dose- and time-dependent secretion of HMGB1 in the absence of cell death. Secretion of HMGB1 was facilitated by Eh surface Gal-lectin-activated phosphoinositide 3-kinase and nuclear factor-κB signaling and up-regulation of histone acetyltransferase activity to trigger acetylated HMGB1 translocation from the nucleus. Unlike lipopolysaccharide, Eh-induced HMGB1 release was independent of caspase-1-mediated inflammasome and gasdermin D pores. In vivo, Eh inoculation in specific pathogen-free but not germ-free mice was associated with high levels of pro-inflammatory cytokines such as tumor necrosis factor-α, interleukin-1ß, and keratinocyte-derived chemokine, which was suppressed with HMGB1 neutralization. This study reveals that Eh-induced active secretion of the HMGB1 plays a key role in shaping the pro-inflammatory landscape critical in innate host defense against amebiasis.


Subject(s)
Alarmins/metabolism , CD4-Positive T-Lymphocytes/immunology , Entamoeba histolytica/physiology , Entamoebiasis/immunology , HMGB1 Protein/metabolism , Macrophages/immunology , Alarmins/genetics , Animals , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Caspase 1/metabolism , Cytokines/metabolism , HMGB1 Protein/genetics , Host-Pathogen Interactions , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , THP-1 Cells
7.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31527129

ABSTRACT

Epidemiological studies suggest frequent association of enteropathogenic bacteria with Entamoeba histolytica during symptomatic infection. In this study, we sought to determine if the interaction with enteropathogenic (EPEC) or nonpathogenic Escherichia coli (strain DH5α) could modify the virulence of E. histolytica to cause disease in animal models of amebiasis. In vitro studies showed a 2-fold increase in CaCo2 monolayer destruction when E. histolytica interacted with EPEC but not with E. coli DH5α for 2.5 h. This was associated with increased E. histolytica proteolytic activity as revealed by zymogram analysis and degradation of the E. histolytica CP-A1/5 (EhCP-A1/5) peptide substrate Z-Arg-Arg-pNC and EhCP4 substrate Z-Val-Val-Arg-AMC. Additionally, E. histolytica-EPEC interaction increased EhCP-A1, -A2, -A4, and -A5, Hgl, Apa, and Cox-1 mRNA expression. Despite the marked upregulation of E. histolytica virulence factors, nonsignificant macroscopic differences in amebic liver abscess development were observed at early stages in hamsters inoculated with either E. histolytica-EPEC or E. histolytica-E. coli DH5α. Histopathology of livers of E. histolytica-EPEC-inoculated animals revealed foci of acute inflammation 3 h postinoculation that progressively increased, producing large inflammatory reactions, ischemia, and necrosis with high expression of il-1ß, ifn-γ, and tnf-α proinflammatory cytokine genes compared with that in livers of E. histolytica-E. coli DH5α-inoculated animals. In closed colonic loops from mice, intense inflammation was observed with E. histolytica-EPEC manifested by downregulation of Math1 mRNA with a corresponding increase in the expression of Muc2 mucin and proinflammatory cytokine genes il-6, il-12, and mcp-1 These results demonstrate that E. histolytica/EPEC interaction enhanced the expression and production of key molecules associated with E. histolytica virulence, critical in pathogenesis and progression of disease.


Subject(s)
Entamoeba histolytica/pathogenicity , Entamoebiasis/pathology , Enteropathogenic Escherichia coli/physiology , Host Microbial Interactions/physiology , Animals , Caco-2 Cells , Cell Line , Cricetinae , Cysteine Proteases/metabolism , Cytokines/metabolism , Entamoeba histolytica/microbiology , HT29 Cells , Humans , Inflammation , Mesocricetus , Mice , Mice, Inbred C57BL , Mucin-2/metabolism , Virulence Factors/biosynthesis
8.
Infect Immun ; 87(11)2019 11.
Article in English | MEDLINE | ID: mdl-31427448

ABSTRACT

Entamoeba histolytica is an anaerobic parasitic protozoan and the causative agent of amoebiasis. E. histolytica expresses proteins that are structurally homologous to human proteins and uses them as virulence factors. We have previously shown that E. histolytica binds exogenous interferon gamma (IFN-γ) on its surface, and in this study, we explored whether exogenous IFN-γ could modulate parasite virulence. We identified an IFN-γ receptor-like protein on the surface of E. histolytica trophozoites by using anti-IFN-γ receptor 1 (IFN-γR1) antibody and performing immunofluorescence, Western blot, protein sequencing, and in silico analyses. Coupling of human IFN-γ to the IFN-γ receptor-like protein on live E. histolytica trophozoites significantly upregulated the expression of E. histolytica cysteine protease A1 (EhCP-A1), EhCP-A2, EhCP-A4, EhCP-A5, amebapore A (APA), cyclooxygenase 1 (Cox-1), Gal-lectin (Hgl), and peroxiredoxin (Prx) in a time-dependent fashion. IFN-γ signaling via the IFN-γ receptor-like protein enhanced E. histolytica's erythrophagocytosis of human red blood cells, which was abrogated by the STAT1 inhibitor fludarabine. Exogenous IFN-γ enhanced chemotaxis of E. histolytica, its killing of Caco-2 colonic and Hep G2 liver cells, and amebic liver abscess formation in hamsters. These results demonstrate that E. histolytica expresses a surface IFN-γ receptor-like protein that is functional and may play a role in disease pathogenesis and/or immune evasion.


Subject(s)
Entamoeba histolytica/metabolism , Protozoan Proteins/metabolism , Receptors, Interferon/chemistry , Amebiasis/immunology , Amebiasis/parasitology , Animals , Caco-2 Cells , Cell Survival , Cricetinae , Hep G2 Cells , Humans , Interferon-gamma/pharmacology , Male , Phagocytosis , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Interferon gamma Receptor
9.
PLoS Pathog ; 14(11): e1007466, 2018 11.
Article in English | MEDLINE | ID: mdl-30500860

ABSTRACT

Amebiasis is caused by the protozoan parasite Entamoeba histolytica (Eh), a potentially fatal disease occurring mainly in developing countries. How Eh interacts with innate host factors in the gut is poorly understood. Eh resides and feed in/on the outer colonic mucus layer and thus share an ecological niche with indigenous microbiota. As gut microbiota regulates innate immune responses, in this study we characterized the cooperative roles that microbiota and the mucus layer play in Eh-induced pro-inflammatory responses in the colon. To study this, we used antibiotics treated and non-treated specific pathogen free Muc2-/- and Muc2+/+ littermates and germ-free mice inoculated with Eh in colonic loops as a short infection model. In antibiotic treated Muc2-/- and Muc2+/+ littermates, Eh elicited robust mucus and water secretions, enhanced pro-inflammatory cytokines and chemokine expression with elevated MPO activity and higher pathology scores as compared to the modest response observed in non-antibiotic treated littermates. Host responses were microbiota specific as mucus secretion and pro-inflammatory responses were attenuated following homologous fecal microbial transplants in antibiotic-treated Muc2+/+ quantified by secretion of 3H-glucosamine newly synthesized mucin, Muc2 mucin immunostaining and immunohistochemistry. Eh-elicited pro-inflammatory responses and suppressed goblet cell transcription factor Math1 as revealed by in vivo imaging of Eh-colonic loops in Math1GFP mice, and in vitro using Eh-stimulated LS174T human colonic goblet cells. Eh in colonic loops increased bacterial translocation of bioluminescent E. coli and indigenous bacteria quantified by FISH and quantitative PCR. In germ-free animals, Eh-induced mucus/water secretory responses, but acute pro-inflammatory responses and MPO activity were severely impaired, allowing the parasite to bind to and disrupt mucosal epithelial cells. These findings have identified key roles for intestinal microbiota and mucus in regulating innate host defenses against Eh, and implicate dysbiosis as a risk factor for amebiasis that leads to exacerbated immune responses to cause life-threatening disease.


Subject(s)
Entamoeba histolytica/metabolism , Gastrointestinal Microbiome/immunology , Mucin-2/immunology , Animals , Cell Line , Colon/metabolism , Colon/pathology , Entamoeba histolytica/immunology , Entamoeba histolytica/pathogenicity , Epithelial Cells/metabolism , Gene Expression Regulation , Goblet Cells/metabolism , Humans , Immunity, Innate/immunology , Inflammation/pathology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Microbiota , Mucin-1 , Mucins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...