Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell Microbiol ; 22(9): e13232, 2020 09.
Article in English | MEDLINE | ID: mdl-32452132

ABSTRACT

Plasmodium falciparum responsible for the most virulent form of malaria invades human erythrocytes through multiple ligand-receptor interactions. The P. falciparum reticulocyte binding protein homologues (PfRHs) are expressed at the apical end of merozoites and form interactions with distinct erythrocyte surface receptors that are important for invasion. Here using a range of monoclonal antibodies (mAbs) against different regions of PfRH1 we have investigated the role of PfRH processing during merozoite invasion. We show that PfRH1 gets differentially processed during merozoite maturation and invasion and provide evidence that the different PfRH1 processing products have distinct functions during invasion. Using in-situ Proximity Ligation and FRET assays that allow probing of interactions at the nanometre level we show that a subset of PfRH1 products form close association with micronemal proteins Apical Membrane Antigen 1 (AMA1) in the moving junction suggesting a critical role in facilitating junction formation and active invasion. Our data provides evidence that time dependent processing of PfRH proteins is a mechanism by which the parasite is able to regulate distinct functional activities of these large processes. The identification of a specific close association with AMA1 in the junction now may also provide new avenues to target these interactions to prevent merozoite invasion.


Subject(s)
Antigens, Protozoan/metabolism , Erythrocytes/parasitology , Host-Parasite Interactions , Membrane Proteins/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Reticulocytes/metabolism , Tight Junctions/metabolism , Antibodies, Monoclonal , Antigens, Protozoan/genetics , Erythrocytes/metabolism , Membrane Proteins/genetics , Merozoites/metabolism , Plasmodium falciparum/chemistry , Protozoan Proteins/genetics , Tight Junctions/parasitology
2.
Microvasc Res ; 131: 104024, 2020 09.
Article in English | MEDLINE | ID: mdl-32502488

ABSTRACT

Congenital toxoplasmosis is a parasitic disease that occurs due vertical transmission of the protozoan Toxoplasma gondii (T. gondii) during pregnancy. The parasite crosses the placental barrier and reaches the developing brain, infecting progenitor, glial, neuronal and vascular cell types. Although the role of Radial glia (RG) neural stem cells in the development of the brain vasculature has been recently investigated, the impact of T. gondii infection in these events is not yet understood. Herein, we studied the role of T. gondii infection on RG cell function and its interaction with endothelial cells. By infecting isolated RG cultures with T. gondii tachyzoites, we observed a cytotoxic effect with reduced numbers of RG populations together with decrease neuronal and oligodendrocyte progenitor populations. Conditioned medium (CM) from RG control cultures increased ZO-1 protein levels and organization on endothelial bEnd.3 cells membranes, which was impaired by CM from infected RG, accompanied by decreased trans-endothelial electrical resistance (TEER). ELISA assays revealed reduced levels of anti-inflammatory cytokine TGF-ß1 in CM from T. gondii-infected RG cells. Treatment with recombinant TGF-ß1 concomitantly with CM from infected RG cultures led to restoration of ZO-1 staining in bEnd.3 cells. Congenital infection in Swiss Webster mice led to abnormalities in the cortical microvasculature in comparison to uninfected embryos. Our results suggest that infection of RG cells by T. gondii negatively modulates cytokine secretion, which might contribute to endothelial loss of barrier properties, thus leading to impairment of neurovascular interaction establishment.


Subject(s)
Cell Differentiation , Cerebral Cortex/blood supply , Endothelial Cells/parasitology , Ependymoglial Cells/parasitology , Microvessels/parasitology , Neurovascular Coupling , Toxoplasma/pathogenicity , Toxoplasmosis, Cerebral/parasitology , Toxoplasmosis, Congenital/parasitology , Animals , Cell Line , Disease Models, Animal , Electric Impedance , Endothelial Cells/metabolism , Endothelial Cells/pathology , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Mice, Inbred C57BL , Microvessels/metabolism , Microvessels/pathology , Tight Junctions/metabolism , Tight Junctions/parasitology , Tight Junctions/pathology , Toxoplasmosis, Cerebral/metabolism , Toxoplasmosis, Cerebral/pathology , Toxoplasmosis, Congenital/metabolism , Toxoplasmosis, Congenital/pathology , Transforming Growth Factor beta1/metabolism , Zonula Occludens-1 Protein/metabolism
3.
Cell Microbiol ; 21(9): e13048, 2019 09.
Article in English | MEDLINE | ID: mdl-31099453

ABSTRACT

The apicomplexan parasite Toxoplasma gondii invades tissues and traverses non-permissive biological barriers in infected humans and other vertebrates. Following ingestion, the parasite penetrates the intestinal wall and disseminates to immune-privileged sites such as the brain parenchyma, after crossing the blood-brain barrier. In the present study, we have established a protocol for high-purification of primary mouse brain endothelial cells to generate stably polarised monolayers that allowed assessment of cellular barrier traversal by T. gondii. We report that T. gondii tachyzoites translocate across polarised monolayers of mouse brain endothelial cells and human intestinal Caco2 cells without significantly perturbing barrier impermeability and with minimal change in transcellular electrical resistance. In contrast, challenge with parasite lysate or LPS increased barrier permeability by destabilising intercellular tight junctions (TJs) and accentuated transmigration of T. gondii. Conversely, reduced phosphorylation of the TJ-regulator focal adhesion kinase (FAK) was observed dose-dependently upon challenge of monolayers with live T. gondii but not with parasite lysate or LPS. Pharmacological inhibition of FAK phosphorylation reversibly altered barrier integrity and facilitated T. gondii translocation. Finally, gene silencing of FAK by shRNA facilitated transmigration of T. gondii across epithelial and endothelial monolayers. Jointly, the data demonstrate that T. gondii infection transiently alters the TJ stability through FAK dysregulation to facilitate transmigration. This work identifies the implication of the TJ regulator FAK in the transmigration of T. gondii across polarised cellular monolayers and provides novel insights in how microbes overcome the restrictiveness of biological barriers.


Subject(s)
Blood-Brain Barrier/parasitology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Toxoplasma/pathogenicity , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain/parasitology , Caco-2 Cells , Cell Polarity/physiology , Endothelial Cells/parasitology , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Silencing , Host-Pathogen Interactions , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Small Interfering , Tight Junctions/metabolism , Tight Junctions/parasitology , Virulence/drug effects , Virulence/immunology
4.
Malar J ; 19(1): 370, 2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33066767

ABSTRACT

BACKGROUND: We present two conundra in the biology of intraerythrocytic malaria parasite: how an apparent open parasitophorous duct provide direct access of only a select set of serum proteins to the parasitophorous vacuole, and how proteases mediate membrane lysis to allow merozoite egress. SOLUTION: We posit the existence of a parasitophorous vacuolar duct plug that is originally formed from a tight junction (or parts thereof) between merozoite apical surface and red blood cell plasma membrane, which by moving over the parasite surface towards the posterior end draws the parasite into the host cell interior, and by remaining at the passage orifice provides a location of transporter(s) for import of serum proteins into parasitophorous vacuole and an opening for merozoite egress upon its dissolution/dismantling through protease(s) action. CONCLUSION: This notion obviates the need of a distinct intact parasitophorous vacuolar membrane, which in the proposed model is an extension of the red blood cell membrane but still forms an intracellular compartment for parasite growth and development. The model is testable using existing high-resolution electron and X-ray tomography tools.


Subject(s)
Blood Proteins/physiology , Erythrocytes/parasitology , Merozoites/physiology , Plasmodium falciparum/physiology , Humans , Tight Junctions/parasitology
5.
Cell Microbiol ; 20(6): e12830, 2018 06.
Article in English | MEDLINE | ID: mdl-29444370

ABSTRACT

Infection with the protozoan parasite Cryptosporidium parvum (CP) causes cryptosporidiosis, a widespread diarrhoeal disease. Impaired intestinal epithelial barrier function and increased permeability are most commonly associated with diarrhoeal diseases caused by enteric infections. However, studies on barrier disruption and underlying mechanisms in cryptosporidiosis are extremely limited. Epithelial tight junctions (TJs) and adherens junctions (AJs) are important in maintaining barrier integrity. Therefore, we examined the effects of CP infection on paracellular permeability and on the expression of the major TJ and AJ proteins utilising in vitro, ex vivo, and in vivo models. CP infection (0.5 × 106  oocysts/well in Transwell inserts, 24 hr) increased paracellular permeability (FITC-dextran flux) in Caco-2 cell monolayers and substantially decreased the protein levels of occludin, claudin 4, and E-cadherin. Claudin 3, zonula occludens-1 (ZO1) and α-catenin were also significantly decreased, whereas claudins 1 and 2 and ß-catenin were not altered. Substantial downregulation of occludin, claudin 4, and E-cadherin was also observed in response to CP infection ex vivo in mouse enteroid-derived monolayers and in vivo in the ileal and jejunal mocosa of C57BL/6 mice. The mRNA levels of these proteins were also significantly decreased in CP-infected mouse ileum and jejunum but were unaltered in Caco-2 cells. Further, bafilomycin-A, an inhibitor of lysosomal proton pump, partially abrogated CP effects on occludin expression in Caco-2 cells, suggesting a potential role of posttranslational mechanisms, such as induction of protein degradation pathways, in mediating the effects of the parasite. Our studies suggest that disruption of barrier function via downregulation of specific key components of TJ and AJ could be a major mechanism underlying CP infection-induced diarrhoea.


Subject(s)
Adherens Junctions/parasitology , Cell Adhesion Molecules/antagonists & inhibitors , Cryptosporidiosis/pathology , Cryptosporidium parvum/growth & development , Down-Regulation , Host-Pathogen Interactions , Tight Junctions/parasitology , Animals , Caco-2 Cells , Disease Models, Animal , Gene Expression Profiling , Humans , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Models, Biological , Permeability
6.
Exp Parasitol ; 183: 69-75, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29097064

ABSTRACT

Free-living amoebae of the genus Acanthamoeba are protozoa ubiquitously found in nature. Some species of the genus are potentially pathogenic for humans provoking keratitis in healthy individuals, often in contact lens wearers and opportunistic infections such as pneumonitis, fatal granulomatous encephalitis and skin infections, particularly in immunocompromised individuals. The pathogenic mechanisms of these amoebae are poorly understood, however it had been suggested that contact dependent mechanisms are important during invasion, regardless of the epithelia type, since amoebae penetrate epithelia separating tight junction (TJ). This study was undertaken to determine whether Acanthamoeba sp. (T4) damages the barrier function of the TJ in MDCK epithelial monolayers. Actin cytoskeleton staining and electron microscopy analyses were performed; paracellular permeability and TJ sealing were evaluated by apicobasolateral diffusion of ruthenium red and transepithelial resistance (TER) measurements; immunofluorescence and Western blot assays were performed to locate and estimate expression of TJ protein claudins 2 (Cldn2) and 4 (Cldn4). The results show that Acanthamoeba sp. crosses the MDCK monolayer without altering the actin cytoskeleton or the morphology of the cells. When trophozoites or conditioned medium interact with the monolayer, paracellular diffusion of ruthenium red increases. After 6 h, the amoebae, but not their conditioned medium, increase the TER, and Cldn2 is removed from the TJ, and its overall content in the cells diminishes, while Cldn4 is targeted to the TJ without changing its expression level. In conclusion Acanthamoeba (T4) crosses MDCK monolayer without damaging the cells, increasing permeability and TER through Cldn2 degradation, and redirecting Cldn4 to TJ. These results strongly suggest that contact-dependent mechanisms are relevant during amoebae invasion.


Subject(s)
Acanthamoeba/physiology , Madin Darby Canine Kidney Cells/parasitology , Tight Junctions/parasitology , Acanthamoeba/pathogenicity , Acanthamoeba/ultrastructure , Animals , Blotting, Western , Claudin-2/metabolism , Claudin-4/metabolism , Culture Media, Conditioned , Dogs , Electric Impedance , Fluorescent Antibody Technique , Indicators and Reagents/metabolism , Madin Darby Canine Kidney Cells/ultrastructure , Microscopy, Electron, Transmission , Permeability , Ruthenium Red/metabolism , Tight Junctions/chemistry , Tight Junctions/metabolism , Trophozoites/physiology , Trophozoites/ultrastructure
7.
Exp Parasitol ; 174: 31-41, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28011167

ABSTRACT

Trypanosoma brucei are extracellular hemoflagellate protozoan parasites and one of the causative agents of a devastating zoonotic disease called African Trypanosomiasis. In humans, the disease is caused by Trypanosoma brucei rhodensiense and Trypanosoma brucei gambiense, which cross the blood brain barrier (BBB) causing neurological disorders which culminate in death if untreated. In some domestic animals and laboratory rodents, Trypanosoma brucei brucei causes a disease similar to that in humans. The mechanism by which Trypanosoma brucei brucei invade biological barriers including the BBB has not been fully elucidated. To further address this issue, Mardin Dardy Canine Kidney II (MDCKII) and Human dermal microvascular endothelial cell (HDMEC) monolayers were grown to confluence on transwell inserts to constitute in vitro biological barriers. MDCKII cells were chosen for their ability to form tight junctions similar to those formed by the BBB endothelial cells. Labeled trypanosomes were placed in the upper chamber of transwell inserts layered with confluent MDCKII/HDMEC monolayers and their ability to cross the monolayer over time evaluated. Our results show that only 0.5-1.25% of Trypanosoma brucei brucei were able to migrate across the monolayers after 3 h. By employing immune-staining and confocal microscopic analysis we observed that trypanosomes were located at the tight junctions and inside the cell in the MDCK II monolayers indicating that they crossed the monolayer using both the paracellular and transcellular routes. Our observations also showed that there seemed to be no obvious degradation of junction proteins Zonula Ocludens-1, Occludin and Ecadherin. In the HDMEC cell monolayer, our scanning electron microscopy data showed that Trypanosoma brucei brucei is able to modulate the plasma membrane to form invaginations similar to cuplike structures formed by Tlymphocytes. However these structures seemed to be independent of vascular adhesion molecules suggesting that they could be more like the membrane ruffles formed by certain intracellular bacteria during invasion. Taken together, our data reveal a mechanism by which Trypanosoma brucei brucei is able to cross different biological barriers including the BBB without causing any obvious damage.


Subject(s)
Blood-Brain Barrier/parasitology , Madin Darby Canine Kidney Cells/parasitology , Trypanosoma brucei brucei/physiology , Trypanosomiasis, African/parasitology , Animals , Blood-Brain Barrier/ultrastructure , Cell Line , Cell Membrane/parasitology , Cell Membrane/ultrastructure , Dogs , Flagella/physiology , Flagella/ultrastructure , Fluorescent Antibody Technique , Humans , Madin Darby Canine Kidney Cells/ultrastructure , Mice , Microscopy, Confocal , Microscopy, Electron, Scanning , Rabbits , Rats , Tight Junction Proteins/chemistry , Tight Junctions/parasitology , Trypanosoma brucei brucei/ultrastructure , Trypanosomiasis, African/pathology
8.
PLoS Pathog ; 9(2): e1003162, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23408890

ABSTRACT

Apicomplexans are obligate intracellular parasites that actively penetrate their host cells to create an intracellular niche for replication. Commitment to invasion is thought to be mediated by the rhoptries, specialized apical secretory organelles that inject a protein complex into the host cell to form a tight-junction for parasite entry. Little is known about the molecular factors that govern rhoptry biogenesis, their subcellular organization at the apical end of the parasite and subsequent release of this organelle during invasion. We have identified a Toxoplasma palmitoyl acyltransferase, TgDHHC7, which localizes to the rhoptries. Strikingly, conditional knockdown of TgDHHC7 results in dispersed rhoptries that fail to organize at the apical end of the parasite and are instead scattered throughout the cell. While the morphology and content of these rhoptries appears normal, failure to tether at the apex results in a complete block in host cell invasion. In contrast, attachment and egress are unaffected in the knockdown, demonstrating that the rhoptries are not required for these processes. We show that rhoptry targeting of TgDHHC7 requires a short, highly conserved C-terminal region while a large, divergent N-terminal domain is dispensable for both targeting and function. Additionally, a point mutant lacking a key residue predicted to be critical for enzyme activity fails to rescue apical rhoptry tethering, strongly suggesting that tethering of the organelle is dependent upon TgDHHC7 palmitoylation activity. We tie the importance of this activity to the palmitoylated Armadillo Repeats-Only (TgARO) rhoptry protein by showing that conditional knockdown of TgARO recapitulates the dispersed rhoptry phenotype of TgDHHC7 knockdown. The unexpected finding that apicomplexans have exploited protein palmitoylation for apical organelle tethering yields new insight into the biogenesis and function of rhoptries and may provide new avenues for therapeutic intervention against Toxoplasma and related apicomplexan parasites.


Subject(s)
Acyltransferases/metabolism , Armadillo Domain Proteins/metabolism , Organelles/enzymology , Protozoan Infections/parasitology , Protozoan Proteins/metabolism , Toxoplasma/enzymology , Acyltransferases/genetics , Amino Acid Sequence , Animals , Armadillo Domain Proteins/genetics , Gene Knockdown Techniques , Host-Parasite Interactions , Humans , Lipoylation , Models, Biological , Molecular Sequence Data , Organelles/physiology , Organelles/ultrastructure , Phenotype , Protein Structure, Tertiary , Protein Transport , Protozoan Infections/metabolism , Protozoan Proteins/genetics , Rats , Recombinant Fusion Proteins , Sequence Alignment , Sequence Deletion , Tight Junctions/parasitology , Tight Junctions/ultrastructure , Toxoplasma/genetics , Toxoplasma/physiology , Toxoplasma/ultrastructure
9.
Am J Pathol ; 182(3): 852-65, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23357502

ABSTRACT

Human mucin-2 (MUC-2) is the first line of innate host defense in preventing pathogen-induced epithelial injury. Entamoeba histolytica (Eh) colonizes the mucus layer by binding of the parasite's surface galactose lectin to galactose and N-acetyl-d-galactosamine residues on colonic MUC-2, preventing parasite contact-dependent cytolysis of epithelial cells. We quantified early innate responses to Eh in wild-type and MUC-2-deficient mice (Muc2(-/-)) using closed colonic loops. Eh infection in wild-type but not Muc2(-/-) mice induced a time-dependent increase in (3)H-labeled mucin and nonmucin glycoprotein secretions. Immunohistochemical staining revealed intense MUC-2 secretion, which formed a thick, protective mucus plug overlying the surface epithelium, entrapping Eh. In Muc2(-/-) mice, Eh induced a pronounced time-dependent secretory exudate with increased gross pathology scores and serum albumin leakage. Colonic pathology, secretory responses, and increased proinflammatory cytokine secretions of TNF-α, IFN-γ, and IL-13 correlated with altered expression of the tight junction proteins claudin-2, occludin, and ZO-1. We identified the putative Eh virulence factor that elicits the proinflammatory responses and alters tight junction permeability as Eh cysteine protease A5 (EhCP-A5). The present findings demonstrate that colonic mucins confer both luminal and epithelial barrier functions and that, in the absence of MUC-2, mice are more susceptible to Eh-induced secretory and proinflammatory responses mediated by EhCP-A5.


Subject(s)
Entamoeba histolytica/physiology , Epithelial Cells/pathology , Epithelial Cells/parasitology , Inflammation Mediators/metabolism , Mucin-2/deficiency , Tight Junctions/parasitology , Animals , Blood Cells/metabolism , Colon/parasitology , Colon/pathology , Entamoeba histolytica/pathogenicity , Gene Expression Regulation , Goblet Cells/parasitology , Goblet Cells/pathology , Humans , Intestines/parasitology , Intestines/pathology , Mice , Mice, Inbred C57BL , Mucin-2/metabolism , Permeability , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Virulence Factors/metabolism
10.
PLoS Negl Trop Dis ; 17(12): e0011816, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38048314

ABSTRACT

BACKGROUND: Cathepsin L, a lysosomal enzyme, participates in diverse physiological processes. Recombinant Trichinella spiralis cathepsin L domains (rTsCatL2) exhibited natural cysteine protease activity and hydrolyzed host immunoglobulin and extracellular matrix proteins in vitro, but its functions in larval invasion are unknown. The aim of this study was to explore its functions in T. spiralis invasion of the host's intestinal epithelial cells. METHODOLOGY/PRINCIPAL FINDINGS: RNAi significantly suppressed the expression of TsCatL mRNA and protein with TsCatL specific siRNA-302. T. spiralis larval invasion of Caco-2 cells was reduced by 39.87% and 38.36%, respectively, when anti-TsCatL2 serum and siRNA-302 were used. Mice challenged with siRNA-302-treated muscle larvae (ML) exhibited a substantial reduction in intestinal infective larvae, adult worm, and ML burden compared to the PBS group, with reductions of 44.37%, 47.57%, and 57.06%, respectively. The development and fecundity of the females from the mice infected with siRNA-302-treated ML was significantly inhibited. After incubation of rTsCatL2 with Caco-2 cells, immunofluorescence test showed that the rTsCatL2 gradually entered into the cells, altered the localization of cellular tight junction proteins (claudin 1, occludin and zo-1), adhesion junction protein (e-cadherin) and extracellular matrix protein (laminin), and intercellular junctions were lost. Western blot showed a 58.65% reduction in claudin 1 expression in Caco-2 cells treated with rTsCatL2. Co-IP showed that rTsCatL2 interacted with laminin and collagen I but not with claudin 1, e-cadherin, occludin and fibronectin in Caco-2 cells. Moreover, rTsCatL2 disrupted the intestinal epithelial barrier by inducing cellular autophagy. CONCLUSIONS: rTsCatL2 disrupts the intestinal epithelial barrier and facilitates T. spiralis larval invasion.


Subject(s)
Cathepsin L , Tight Junctions , Trichinella spiralis , Trichinellosis , Animals , Female , Humans , Mice , Caco-2 Cells , Cadherins/metabolism , Cathepsin L/genetics , Cathepsin L/metabolism , Claudin-1/genetics , Claudin-1/metabolism , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Laminin/genetics , Laminin/metabolism , Larva/parasitology , Mice, Inbred BALB C , Occludin/genetics , Occludin/metabolism , RNA, Double-Stranded , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tight Junctions/parasitology , Tight Junctions/pathology , Trichinella spiralis/genetics
11.
Poult Sci ; 99(9): 4203-4216, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32867964

ABSTRACT

This study was conducted to evaluate graded Eimeria challenge on growth performance, apparent ileal digestibility, gastrointestinal permeability, intestinal morphology, gene expression of tight junction protein, and intestinal lesion scores in broiler chickens. There were 5 groups in this study, including a control and 4 different Eimeria treatment doses. A mixed Eimeria spp. solution with 50,000 Eimeria maxima, 50,000 Eimeria tenella, and 250,000 Eimeria acervulina per milliliter was prepared for the high-dose challenge treatment. The 2-fold serial dilution was used to make the medium-high (25,000 E. maxima; 25,000 E. tenella; 125,000 E. acervulina), the medium-low (12,500 E. maxima; 12,500 E. tenella; 62,500 E. acervulina), and the low challenge dose (6,250 E. maxima; 6,250 E. tenella; 31,250 E. acervulina). A total of three hundred sixty 13-day-old male broiler chickens were randomly allocated into 5 treatments with 6 replicated cages. Growth performance was calculated from 0 to 6 D postinfection (DPI). Intestine lesion was scored on 6 DPI. Gastrointestinal permeability was measured on 3, 5, 6, 7, and 9 DPI. The results indicated significant linear reduction in growth performance, intestinal villi height, and ileal nutrient digestibility in response to the increase of Eimeria challenge dose. Furthermore, gene expression of tight junction protein was linearly upregulated by the increasing challenge doses. Significant linear increases of gastrointestinal permeability were found on 5, 6, and 7 DPI (P < 0.01). On 9 DPI, the gastrointestinal permeability was recovered back to normal level in the challenge groups. In conclusion, the higher Eimeria doses birds received, the more severe intestine damage was observed in several gastrointestinal health parameters. The medium-low or medium-high levels of mixed Eimeria oocysts is suggested as an optimum Eimeria-challenge dose to establish a subclinical challenge model for future studies evaluating nutritional strategies. Moreover, it is recommended to measure gastrointestinal permeability on 5 DPI with higher oocysts doses and 6 DPI when using the lower oocysts doses.


Subject(s)
Coccidiosis , Eimeria , Gastrointestinal Tract , Poultry Diseases , Tight Junctions , Animals , Chickens , Coccidiosis/physiopathology , Coccidiosis/veterinary , Digestion , Gastrointestinal Tract/parasitology , Gastrointestinal Tract/physiopathology , Ilium/metabolism , Intestines/anatomy & histology , Intestines/parasitology , Male , Permeability , Poultry Diseases/parasitology , Poultry Diseases/physiopathology , Tight Junctions/parasitology
12.
J Histochem Cytochem ; 64(8): 459-69, 2016 08.
Article in English | MEDLINE | ID: mdl-27370796

ABSTRACT

After oral infection, Toxoplasma gondii invades intestinal cells, induces breakdown of intestinal physiology and barrier functions, and causes intestinal pathology in some animal species. Although parasites' invasion into host cells is a known phenomenon, the effects of T. gondii infection in the intestinal barrier are still not well established. To evaluate morphological and physiological modifications on the colorectal adenocarcinoma-derived Caco-2 cell line during T. gondii infection, microvilli, tight junction integrity, and transepithelial electrical resistance (TEER) were investigated under infection. It was observed that the dextran uptake (endocytosis) and distribution were smaller in infected than in noninfected Caco-2 cells. The infection leads to the partial loss of microvilli at the cell surface. Claudin-1, zonula occludens-1 (ZO-1), and occludin expressions were colocalized by immunofluorescence and presented discontinuous net patterns in infected cells. Immunoblotting analysis at 24 hr postinfection revealed decreasing expression of occludin and ZO-1 proteins, whereas claudin-1 presented similar expression level compared with noninfected cells. T. gondii decreased TEER in Caco-2 cells 24 hr after infection. Our results suggest that T. gondii infection may lead to the loss of integrity of intestinal mucosa, resulting in impaired barrier function.


Subject(s)
Intestinal Mucosa/parasitology , Toxoplasma/physiology , Actin Cytoskeleton/ultrastructure , Caco-2 Cells , Cell Polarity , Claudin-1/metabolism , Dextrans/metabolism , Electric Impedance , Endocytosis , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Microvilli/metabolism , Microvilli/parasitology , Microvilli/ultrastructure , Occludin/metabolism , Tight Junctions/metabolism , Tight Junctions/parasitology , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein/metabolism
13.
mBio ; 7(3)2016 06 07.
Article in English | MEDLINE | ID: mdl-27273825

ABSTRACT

UNLABELLED: Cerebral malaria (CM) is a disease of the vascular endothelium caused by Plasmodium falciparum It is characterized by parasite sequestration, inflammatory cytokine production, and vascular leakage. A distinguishing feature of P. falciparum infection is parasite production and secretion of histidine-rich protein II (HRPII). Plasma HRPII is a diagnostic and prognostic marker for falciparum malaria. We demonstrate that disruption of a human cerebral microvascular endothelial barrier by P. falciparum-infected erythrocytes depends on expression of HRPII. Purified recombinant or native HRPII can recapitulate these effects. HRPII action occurs via activation of the inflammasome, resulting in decreased integrity of tight junctions and increased endothelial permeability. We propose that HRPII is a virulence factor that may contribute to cerebral malaria by compromising endothelial barrier integrity within the central nervous system. IMPORTANCE: Cerebral malaria is a devastating disease. Patients have high levels of the protein HRPII in their blood. We have found that endothelial cell barriers become leaky when treated with concentrations of HRPII similar to those found in patients. This result suggests that HRPII may be important in cerebral malaria. Our finding that HRPII functions by causing inflammation suggests points of intervention for therapy or vaccination against this disease.


Subject(s)
Blood-Brain Barrier/physiology , Endothelial Cells/physiology , Erythrocytes/parasitology , Malaria, Cerebral/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/pathogenicity , Proteins/metabolism , Animals , Blood-Brain Barrier/parasitology , Brain/cytology , Brain/immunology , Brain/parasitology , Brain/physiopathology , Cell Death/drug effects , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Immunity, Innate/drug effects , Inflammasomes/drug effects , Inflammation/parasitology , Malaria, Cerebral/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Proteins/genetics , Proteins/isolation & purification , Proteins/pharmacology , Tight Junctions/drug effects , Tight Junctions/parasitology , Tight Junctions/physiology , Virulence Factors
14.
Int J Parasitol ; 34(7): 785-94, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15157761

ABSTRACT

Tight junctions and microvilli constitute an anti-invasive barrier at the luminal side of enteric cell layers. Both subcellular structures are disrupted following adhesion of Entamoeba histolytica trophozoites to enteric cell layers in vitro. It was our aim to analyse the molecular mechanism underlying this disruption. Therefore, we cocultured enteric T84 cell layers established on filter inserts with E. histolytica trophozoites and tested various modulators of enteric molecules, involved in the functional regulation of tight junctions, as well as inhibitors of trophozoite virulence factors on their capacity to maintain the transepithelial electrical resistance. Pretreatment of trophozoites with the proteinase inhibitor N-Tosyl-Phenylalanine chloromethyl ketone or N-Tosyl-l-Lysine chloromethyl ketone prevented the decrease in transepithelial electrical resistance whereas none of the modulators used to pretreat enterocytes were successful. Moreover, zymography and Western blot analysis revealed that both N-Tosyl-Phenylalanine chloromethyl ketone and N-Tosyl-l-Lysine chloromethyl ketone inhibited E. histolytica cysteine proteinases and prevented proteolysis of tight junction molecules ZO-1 and ZO-2 and of villin, the major actin bundling molecule in microvilli. Immunocytochemistry with an antibody against ezrin, an actin-binding molecule in microvilli, and phase contrast microscopy demonstrated that pretreatment of trophozoites with N-Tosyl-Phenylalanine chloromethyl ketone or N-Tosyl-l-Lysine chloromethyl ketone also prevented disturbance of microvilli and destruction of Caco-2 enteric cell layers in cocultures. Taken together, our results indicate that trophozoites use their proteinases to overcome microvilli and tight junction barriers during the invasion of enteric cell layers, that these phenomena could be prevented by pretreatment of trophozoites with N-Tosyl-Phenylalanine chloromethyl ketone or N-Tosyl-l-Lysine chloromethyl ketone, and that such pretreatment disabled trophozoites to destroy enteric cell layers in vitro.


Subject(s)
Entamoeba histolytica/physiology , Enterocytes/parasitology , Protein Synthesis Inhibitors/pharmacology , Tight Junctions/parasitology , Tosyllysine Chloromethyl Ketone/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Animals , Cells, Cultured , Cysteine Endopeptidases/metabolism , Electric Impedance , Entamoeba histolytica/drug effects , Entamoeba histolytica/enzymology , Enterocytes/drug effects , Enterocytes/physiology , Microfilament Proteins/metabolism , Microvilli/drug effects , Microvilli/parasitology , Microvilli/physiology , Tight Junctions/drug effects , Tight Junctions/physiology
15.
J Parasitol ; 99(2): 353-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22924932

ABSTRACT

In view of the interest in genotype-specific pathogenesis in Giardia duodenalis , the aim of the present study was to examine the effects of infection with different, or mixed, G. duodenalis assemblages on the integrity of human intestinal epithelia. To that end, human epithelial cells (HCT-8) were cultured and exposed to different G. duodenalis assemblages (A, B, and E) or a combination of these assemblages. Epithelial disruption and apoptosis were evaluated by fluorescent microscopy and apoptotic oligonucleosome quantification. The results indicate that infection with trophozoites disrupts epithelial tight junctions and induces varying degrees of enterocyte apoptosis, depending on the infecting assemblage. All disruptions were caspase-3 dependent and were more pronounced when caused by a non-host specific assemblage. Furthermore, infections by isolates in combination with isolates from another assemblage enhanced the epithelial disruption and apoptosis. Further studies in vitro and in vivo are required to confirm the mechanisms of enhanced pathogenicity of mixed or non-host specific (or both) G. duodenalis infections. Findings in the present study point to the potential pathogenic importance of intra-species polyparasitism in giardiasis.


Subject(s)
Apoptosis , Epithelial Cells/parasitology , Giardia lamblia/pathogenicity , Intestinal Mucosa/parasitology , Tight Junctions/parasitology , Adenocarcinoma , Caspase 3/drug effects , Caspase Inhibitors/pharmacology , Epithelial Cells/pathology , Giardia lamblia/classification , Humans , Ileal Neoplasms , Ileocecal Valve , Intestinal Mucosa/pathology , Tight Junctions/pathology , Trophozoites/classification , Trophozoites/growth & development , Tumor Cells, Cultured
16.
PLoS One ; 8(9): e73339, 2013.
Article in English | MEDLINE | ID: mdl-24058468

ABSTRACT

The exact mechanism by which Entamoeba histolytica disrupts the human colonic epithelium and invades the mucosa has yet to be clearly elucidated. E. histolytica produces a diverse array of putative virulent factors such as glycosidase, cysteine proteinases and amebapore that can modulate and/or disrupt epithelial barrier functions. However, it is currently thought that E. histolytica produces numerous other molecules and strategies to disrupt colonic mucosal defenses. In this study, we document a putative mechanism whereby the parasite alters the integrity of human epithelium by expressing a cognate tight junction protein of the host. We detected this protein as "occludin-like" as revealed by immunoblotting and immunoprecipitation studies and visualization by confocal microscopy using antibodies highly specific for human occludin. We propose that E. histolytica occludin-like protein might displace mucosal epithelial occludin-occludin tight junction interactions resulting in epithelial disruption analogous to sub mucosal human dendritic cells sampling luminal contents. These results indicate that E. histolytica occludin is a putative virulent component that can play a role in the pathogenesis of intestinal amebiasis.


Subject(s)
Entamoeba histolytica/genetics , Epithelial Cells/parasitology , Occludin/genetics , Protozoan Proteins/genetics , Tight Junctions/parasitology , Virulence Factors/genetics , Blotting, Western , Cell Line , Colon/metabolism , Colon/parasitology , Colon/pathology , Entamoeba histolytica/metabolism , Entamoeba histolytica/pathogenicity , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Host-Parasite Interactions , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Microscopy, Confocal , Occludin/metabolism , Permeability , Protozoan Proteins/metabolism , Species Specificity , Tight Junctions/metabolism , Tight Junctions/pathology , Virulence Factors/metabolism
17.
Parasitol Int ; 61(2): 280-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22146155

ABSTRACT

Giardia duodenalis is a parasitic protozoan that causes diarrhea and other symptoms which together constitute a disease known as giardiasis. Although the disease has been well defined, the mechanisms involving the establishment of the infection have not yet been fully elucidated. In this study, we show that after 24h of interaction between parasites and intestinal Caco-2 cells, there was an alteration of the paracellular permeability, as observed by an approximate 42% of reduction in the transepithelial electrical resistance and permeation to ruthenium red, which was concomitant with ultrastructural changes. Nevertheless, epithelium viability was not affected. We also demonstrate that there was no change in expression of junctional proteins (tight and adherens) but that the distribution of these proteins in Caco-2 cells after parasite adhesion was significantly altered, as observed via laser scanning confocal microscopy 3D reconstruction. The present work shows that adhesion of Giardia duodenalis trophozoites to intestinal cells in vitro induces disturbances of the tight, adherens and desmosomal junctions.


Subject(s)
Adherens Junctions/metabolism , Desmosomes/metabolism , Giardia/physiology , Giardiasis/parasitology , Tight Junctions/metabolism , Actin Cytoskeleton/metabolism , Adherens Junctions/parasitology , Adherens Junctions/ultrastructure , Animals , Caco-2 Cells , Cell Membrane Permeability , Cell Survival , Desmosomes/parasitology , Desmosomes/ultrastructure , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Host-Parasite Interactions , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Tight Junctions/parasitology , Tight Junctions/ultrastructure , Trophozoites
18.
Int J Parasitol ; 39(14): 1611-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19580812

ABSTRACT

Haematogenous spread is a key step in the development of Acanthamoeba granulomatous encephalitis, however it is not clear how circulating amoebae cross the blood-brain barrier to enter the CNS to produce disease. Using the primary human brain microvascular endothelial cells (HBMEC), which constitute the blood-brain barrier, here it is shown that Acanthamoeba abolishes the HBMEC transendothelial electrical resistance. Using traversal assays, it was observed that Acanthamoeba crosses the HBMEC monolayers. The primary interactions of Acanthamoeba with the HBMEC resulted in increased protein tyrosine phosphorylations and the activation of RhoA, suggesting host-parasite cross-talk. Furthermore, Western blot assays revealed that Acanthamoeba degraded occludin and zonula occludens-1 proteins in a Rho kinase-dependent manner. Overall, these findings suggest that Acanthamoeba affects the integrity of the monolayer and traverses the HBMEC by targeting the tight junction proteins.


Subject(s)
Acanthamoeba/pathogenicity , Amebiasis/parasitology , Blood-Brain Barrier/parasitology , Encephalitis/parasitology , Endothelial Cells/parasitology , Tight Junctions/parasitology , Amebiasis/physiopathology , Blood-Brain Barrier/physiology , Blotting, Western , Connexins/metabolism , Encephalitis/physiopathology , Endothelial Cells/physiology , Humans , Tight Junctions/physiology
20.
Infect Immun ; 72(10): 6076-86, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15385512

ABSTRACT

Infection with intestinal nematodes induces profound pathological changes to the gut that are associated with eventual parasite expulsion. We have applied expression profiling as an initial screening process with oligonucleotide microarrays (Affymetrix MG-U74AV2 gene chips) and time course kinetics to investigate gene transcription triggered by the intraepithelial nematode Trichinella spiralis in jejunal epithelium from BALB/c mice. Of the 4,114 genes detected, 2,617 were present in all uninfected and T. spiralis-infected replicates, 8% of which were notably upregulated, whereas 12% were downregulated at the time of worm expulsion (day 14 postinfection). Upregulation of goblet cell mucin gene transcripts intestinal mucin gene 3 (MUC3), calcium chloride channel 5 (CLCA5), and goblet cell gene 4 (GOB4) is consistent with enhanced production and alteration of mucus, whereas a 60- to 70-fold upregulation of transcripts for mast cell proteases 1 and 2 (MCPT-1 and -2) is consistent with intraepithelial mucosal mast cell recruitment. Importantly, there was novel expression of sialyltransferase 4C (SIAT4C), small proline-rich protein 2A (SPRR2A), and resistin-like molecule beta (RELMbeta) on day 14 postinfection. In contrast, DNase I and regenerating protein 3 (REG3) transcripts were substantially downregulated. Time course analyses revealed early (within 48 h of infection) induction of Siat4c, Sprr2A, and Relmbeta and later (within 120 h) induction of Mcpt-1 and -2. The findings demonstrate early innate responses and later inflammatory changes within the epithelium. The early epithelial responses may be associated both with repair (Sprr2A) and with the development of innate immunity (Siat4c and Relmbeta).


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/parasitology , Gene Expression Profiling , Inflammation/genetics , Jejunum/cytology , Trichinella spiralis/physiology , Trichinellosis/genetics , Animals , Antioxidants/metabolism , Cytoskeleton/genetics , Cytoskeleton/parasitology , Epithelial Cells/cytology , Epithelial Cells/enzymology , Female , Gene Expression Regulation , Glutathione/metabolism , Goblet Cells/metabolism , Goblet Cells/parasitology , Immunity/genetics , Inflammation/parasitology , Jejunum/enzymology , Jejunum/metabolism , Jejunum/parasitology , Male , Mast Cells/metabolism , Mast Cells/parasitology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mucins/biosynthesis , Oligonucleotide Array Sequence Analysis , Organ Specificity , Paneth Cells/metabolism , Paneth Cells/parasitology , Tight Junctions/genetics , Tight Junctions/parasitology , Transcription, Genetic/genetics , Trichinellosis/enzymology , Trichinellosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL