Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
mSphere ; 5(1)2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31996420

ABSTRACT

Toxoplasma gondii can infect and replicate in vascular endothelial cells prior to entering host tissues. However, little is known about the molecular interactions at the parasite-endothelial cell interface. We demonstrate that T. gondii infection of primary human umbilical vein endothelial cells (HUVEC) altered cell morphology and dysregulated barrier function, increasing permeability to low-molecular-weight polymers. T. gondii disrupted vascular endothelial cadherin (VE-cadherin) and ß-catenin localization to the cell periphery and reduced VE-cadherin protein expression. Notably, T. gondii infection led to reorganization of the host cytoskeleton by reducing filamentous actin (F-actin) stress fiber abundance under static and microfluidic shear stress conditions and by reducing planar cell polarity. RNA sequencing (RNA-Seq) comparing genome-wide transcriptional profiles of infected to uninfected endothelial cells revealed changes in gene expression associated with cell-cell adhesion, extracellular matrix reorganization, and cytokine-mediated signaling. In particular, genes downstream of Hippo signaling and the biomechanical sensor and transcriptional coactivator Yes-associated protein (YAP) were downregulated in infected endothelial cells. Interestingly, T. gondii infection activated Hippo signaling by increasing phosphorylation of LATS1, leading to cytoplasmic retention of YAP, and reducing YAP target gene expression. These findings suggest that T. gondii infection triggers Hippo signaling and YAP nuclear export, leading to an altered transcriptional profile of infected endothelial cells.IMPORTANCE Toxoplasma gondii is a foodborne parasite that infects virtually all warm-blooded animals and can cause severe disease in individuals with compromised or weakened immune systems. During dissemination in its infected hosts, T. gondii breaches endothelial barriers to enter tissues and establish the chronic infections underlying the most severe manifestations of toxoplasmosis. The research presented here examines how T. gondii infection of primary human endothelial cells induces changes in cell morphology, barrier function, gene expression, and mechanotransduction signaling under static conditions and under the physiological conditions of shear stress found in the bloodstream. Understanding the molecular interactions occurring at the interface between endothelial cells and T. gondii may provide insights into processes linked to parasite dissemination and pathogenesis.


Subject(s)
Cell Membrane Permeability , Human Umbilical Vein Endothelial Cells/parasitology , Mechanotransduction, Cellular , Toxoplasma/pathogenicity , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Polarity , Cells, Cultured , Cytoskeleton , Hippo Signaling Pathway , Human Umbilical Vein Endothelial Cells/cytology , Humans , Protein Serine-Threonine Kinases/metabolism , RNA-Seq , Stress Fibers/metabolism , Transcription Factors/metabolism , Transcriptome , YAP-Signaling Proteins , beta Catenin/metabolism
2.
Cell Biol Int ; 44(5): 1112-1123, 2020 May.
Article in English | MEDLINE | ID: mdl-31943572

ABSTRACT

Chagas disease is a vector-borne disease caused by the protozoan parasite Trypanosoma cruzi. Current therapy involves benznidazole. Benznidazole and other drugs can modify gene expression patterns, improving the response to the inflammatory influx induced by T. cruzi and decreasing the endothelial activation or immune cell recruitment, among other effects. Here, we performed a microarray analysis of human umbilical vein endothelial cells (HUVECs) treated with benznidazole and the anti-inflammatory drugs acetylsalicylic acid or simvastatin and infected with T. cruzi. Parasitic infection produces differential expression of a set of genes in HUVECs treated with benznidazole alone or a combination with simvastatin or acetylsalicylic acid. The differentially expressed genes were involved in inflammation, adhesion, cardiac function, and remodeling. Notch1 and high mobility group B1 were genes of interest in this analysis due to their importance in placental development, cardiac development, and inflammation. Quantitative polymerase chain reaction confirmation of these two genes indicated that both are upregulated in the presence of benznidazole.


Subject(s)
Aspirin/pharmacology , Gene Expression/drug effects , HMGB1 Protein/metabolism , Human Umbilical Vein Endothelial Cells/parasitology , Nitroimidazoles/pharmacology , Receptor, Notch1/metabolism , Simvastatin/pharmacology , Cells, Cultured , Chagas Disease/drug therapy , Humans , Trypanosoma cruzi
3.
J R Soc Interface ; 15(149): 20180773, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30958233

ABSTRACT

Malaria is associated with significant microcirculation disorders, especially when the infection reaches its severe stage. This can lead to a range of fatal conditions, from cerebral malaria to multiple organ failure, of not fully understood pathogenesis. It has recently been proposed that a breakdown of the glycocalyx, the carbohydrate-rich layer lining the vascular endothelium, plays a key role in severe malaria, but direct evidence supporting this hypothesis is still lacking. Here, the interactions between Plasmodium falciparum infected red blood cells ( PfRBCs) and endothelial glycocalyx are investigated by developing an in vitro, physiologically relevant model of human microcirculation based on microfluidics. Impairment of the glycocalyx is obtained by enzymatic removal of sialic acid residues, which, due to their terminal location and net negative charge, are implicated in the initial interactions with contacting cells. We show a more than twofold increase of PfRBC adhesion to endothelial cells upon enzymatic treatment, relative to untreated endothelial cells. As a control, no effect of enzymatic treatment on healthy red blood cell adhesion is found. The increased adhesion of PfRBCs is also associated with cell flipping and reduced velocity as compared to the untreated endothelium. Altogether, these results provide a compelling evidence of the increased cytoadherence of PfRBCs to glycocalyx-impaired vascular endothelium, thus supporting the advocated role of glycocalyx disruption in the pathogenesis of this disease.


Subject(s)
Cell Adhesion , Erythrocytes/metabolism , Glycocalyx/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Erythrocytes/parasitology , Erythrocytes/pathology , Human Umbilical Vein Endothelial Cells/parasitology , Human Umbilical Vein Endothelial Cells/pathology , Humans
4.
DNA Cell Biol ; 36(3): 212-218, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28092463

ABSTRACT

Autophagy is a main defense strategy by which infected host cells can virtually induce the killing of parasite, including Toxoplasma gondii. However, the regulatory mechanisms of autophagy in T. gondii-infected nonhematopoietic cells are still unknown. Emerging evidence indicates that CCAAT/enhancer-binding protein ß (C/EBP ß) is associated with the regulation of autophagy. Herein, we hypothesized that C/EBP ß plays roles in inducing autophagy in nonhematopoietic cells. Expression of C/EBP ß was aberrantly regulated in endothelial cells and retinal pigment epithelial cells challenged by T. gondii. Inhibition of C/EBP ß reduced the killing of T. gondii in nonhematopoietic cells, whereas C/EBP ß overexpression resulted in the enhancement of killing of T. gondii as well as the increase in autophagy in infected cells. Furthermore, the mammalian target of rapamycin (mTOR) activation was found to be reduced by C/EBP ß overexpression, but increased by C/EBP ß inhibition. The increase in T. gondii killing induced by C/EBP ß overexpression was blocked by the mTOR activator phosphatidic acid and was increased by the inhibitor AZD8055. In conclusion, we demonstrate that C/EBP ß expression is increased in nonhematopoietic cells infected by T. gondii, resulting in the activation of autophagy in host cells by inhibiting mTOR pathway.


Subject(s)
Autophagy/physiology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Epithelial Cells/parasitology , Toxoplasma/physiology , Autophagy/genetics , Blotting, Western , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Line , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression , Host-Parasite Interactions/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Microscopy, Fluorescence , Morpholines/pharmacology , Phosphatidic Acids/pharmacology , RNA Interference , Retinal Pigment Epithelium/cytology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
5.
Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi ; 29(3): 320-323, 2017 May 23.
Article in Chinese | MEDLINE | ID: mdl-29469522

ABSTRACT

OBJECTIVE: To preliminarily study the pro-angiogenic activity of Echinococcus granulosus hydatid cysts against human umbilical vein endothelial cells in vitro and the transcriptional level of potential pro-angiogenic factors. METHODS: The hydatid cysts and protoscolex derived from experimentally infected mice were collected and cultured in vitro, then the human umbilical vein endothelial cells were stimulated by the supernatant and cyst fluid respectively, and the angiogenesis was observed and analyzed through a microscope and the angiogenesis mode of the software NIH Image J. Meanwhile, the mouse homologous proteins of matrix metalloproteinase-9 (MMP-9) and high mobility group box B1 (HMGB1) were identified in E. granulosus genome through sequence alignment, and their transcriptional levels in the cyst wall and protoscolex were analyzed. RESULTS: The culture supernatant of hydatid cysts significantly promoted human umbilical vein endothelial cells into tubes (F = 73.03, P < 0.001), the transcriptions of MMP-9 and HMGB1 were detected in the cyst wall and protoscolex, and the transcriptional level of MMP-9 was higher in protoscolex (t = -11.65, P < 0.001), while the level of HMGB1 was higher in hydatid cysts (t = 6.43, P = 0.003). CONCLUSIONS: Some parasite-derived pro-angiogenic molecules may exist in the supernatant of E. granulosus hydatid cysts, while further researches are required into their exact mechanisms.


Subject(s)
Echinococcosis/pathology , Echinococcus granulosus , HMGB1 Protein/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic , Animals , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Mice , Sequence Alignment
6.
Parasitol Int ; 65(1): 20-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26384965

ABSTRACT

Hypervalent organotellurium compounds (organotelluranes) have shown several promising applications, including their use as potent and selective cysteine protease inhibitors and antiprotozoal agents. Here, we report the antimalarial activities of three organotellurane derivatives (RF05, RF07 and RF19) in two Plasmodium falciparum strains (CQS 3D7 and CQR W2), which demonstrated significant decreases in parasitemia in vitro. The inhibition of intracellular P. falciparum proteases by RF05, RF07 and RF19 was determined and the IC50 values were 3.7±1.0µM, 1.1±0.2µM and 0.2±0.01µM, respectively. Using an assay performed in the presence of the ER Ca(2+)-ATPase inhibitor we showed that the main enzymatic targets were cysteine proteases stimulated by calcium (calpains). None of the compounds tested caused haemolysis or a significant decrease in endothelial cell viability in the concentration range used for the inhibition assay. Taken together, the results suggest promising compounds for the development of antimalarial drugs.


Subject(s)
Antimalarials/pharmacology , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Organometallic Compounds/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Tellurium/pharmacology , Antimalarials/toxicity , Calcium/metabolism , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/toxicity , Drug Discovery , Erythrocytes/drug effects , Erythrocytes/parasitology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Inhibitory Concentration 50 , Malaria, Falciparum/drug therapy , Organometallic Compounds/toxicity , Tellurium/toxicity
8.
PLoS One ; 8(12): e81187, 2013.
Article in English | MEDLINE | ID: mdl-24312535

ABSTRACT

Chagas heart disease, the leading cause of heart failure in Latin America, results from infection with the parasite Trypanosoma cruzi. Although T. cruzi disseminates intravascularly, how the parasite contends with the endothelial barrier to escape the bloodstream and infect tissues has not been described. Understanding the interaction between T. cruzi and the vascular endothelium, likely a key step in parasite dissemination, could inform future therapies to interrupt disease pathogenesis. We adapted systems useful in the study of leukocyte transmigration to investigate both the occurrence of parasite transmigration and its determinants in vitro. Here we provide the first evidence that T. cruzi can rapidly migrate across endothelial cells by a mechanism that is distinct from productive infection and does not disrupt monolayer integrity or alter permeability. Our results show that this process is facilitated by a known modulator of cellular infection and vascular permeability, bradykinin, and can be augmented by the chemokine CCL2. These represent novel findings in our understanding of parasite dissemination, and may help identify new therapeutic strategies to limit the dissemination of the parasite.


Subject(s)
Capillary Permeability , Chagas Cardiomyopathy/metabolism , Endothelium, Vascular , Human Umbilical Vein Endothelial Cells , Trypanosoma cruzi/metabolism , Animals , Bradykinin/metabolism , Chemokine CCL2/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/parasitology , Female , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Male , Mice
9.
J Biol Chem ; 288(15): 10548-57, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23443665

ABSTRACT

Nicotinamide, a soluble compound of the vitamin B3 group, has antimicrobial activity against several microorganisms ranging from viruses to parasite protozoans. However, the mode of action of this antimicrobial activity is unknown. Here, we investigate the trypanocidal activity of nicotinamide on Trypanosoma brucei, the causative agent of African trypanosomiasis. Incubation of trypanosomes with nicotinamide causes deleterious defects in endocytic traffic, disruption of the lysosome, failure of cytokinesis, and, ultimately, cell death. At the same concentrations there was no effect on a cultured mammalian cell line. The effects on endocytosis and vesicle traffic were visible within 3 h and can be attributed to inhibition of lysosomal cathepsin b-like protease activity. The inhibitory effect of nicotinamide was confirmed by a direct activity assay of recombinant cathepsin b-like protein. Taken together, these data demonstrate that inhibition of the lysosomal protease cathepsin b-like blocks endocytosis, causing cell death. In addition, these results demonstrate for the first time the inhibitory effect of nicotinamide on a protease.


Subject(s)
Cathepsin B/antagonists & inhibitors , Niacinamide/pharmacology , Protozoan Proteins/antagonists & inhibitors , Trypanosoma brucei brucei/enzymology , Vitamin B Complex/pharmacology , Cathepsin B/metabolism , Endocytosis/drug effects , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Lysosomes/enzymology , Protein Transport/drug effects , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/cytology , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/enzymology
10.
PLoS One ; 7(2): e31567, 2012.
Article in English | MEDLINE | ID: mdl-22363675

ABSTRACT

The protozoan parasite Plasmodium falciparum, responsible for the most severe form of malaria, is able to sequester from peripheral circulation during infection. The asexual stage parasites sequester by binding to endothelial cell receptors in the microvasculature of various organs. P. falciparum gametocytes, the developmental stages responsible for parasite transmission from humans to Anopheles mosquitoes, also spend the almost ten days necessary for their maturation sequestered away from the peripheral circulation before they are released in blood mainstream. In contrast to those of asexual parasites, the mechanisms and cellular interactions responsible for immature gametocyte sequestration are largely unexplored, and controversial evidence has been produced so far on this matter. Here we present a systematic comparison of cell binding properties of asexual stages and immature and mature gametocytes from the reference P. falciparum clone 3D7 and from a patient parasite isolate on a panel of human endothelial cells from different tissues. This analysis includes assays on human bone marrow derived endothelial cell lines (HBMEC), as this tissue has been proposed as a major site of gametocyte maturation. Our results clearly demonstrate that cell adhesion of asexual stage parasites is consistently more efficient than that, virtually undetectable of immature gametocytes, irrespectively of the endothelial cell lines used and of parasite genotypes. Importantly, immature gametocytes of both lines tested here do not show a higher binding efficiency compared to asexual stages on bone marrow derived endothelial cells, unlike previously reported in the only study on this issue. This indicates that gametocyte-host interactions in this tissue are unlikely to be mediated by the same adhesion processes to specific endothelial receptors as seen with asexual forms.


Subject(s)
Endothelial Cells/parasitology , Life Cycle Stages , Organ Specificity/drug effects , Plasmodium falciparum/cytology , Plasmodium falciparum/growth & development , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/parasitology , Cell Adhesion/drug effects , Chemokine CXCL12/pharmacology , Dermis/cytology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Germ Cells/cytology , Germ Cells/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Interleukin-1beta/pharmacology , Life Cycle Stages/drug effects , Microvessels/cytology , Parasites/cytology , Parasites/drug effects , Parasites/growth & development , Parasites/isolation & purification , Plasmodium falciparum/drug effects , Plasmodium falciparum/isolation & purification , Tumor Necrosis Factor-alpha/pharmacology
11.
Br J Pharmacol ; 165(5): 1333-47, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21797847

ABSTRACT

BACKGROUND AND PURPOSE: Independent studies in experimental models of Trypanosoma cruzi appointed different roles for endothelin-1 (ET-1) and bradykinin (BK) in the immunopathogenesis of Chagas disease. Here, we addressed the hypothesis that pathogenic outcome is influenced by functional interplay between endothelin receptors (ET(A)R and ET(B)R) and bradykinin B(2) receptors (B(2)R). EXPERIMENTAL APPROACH: Intravital microscopy was used to determine whether ETR/B(2)R drives the accumulation of rhodamine-labelled leucocytes in the hamster cheek pouch (HCP). Inflammatory oedema was measured in the infected BALB/c paw of mice. Parasite invasion was assessed in CHO over-expressing ETRs, mouse cardiomyocytes, endothelium (human umbilical vein endothelial cells) or smooth muscle cells (HSMCs), in the presence/absence of antagonists of B(2)R (HOE-140), ET(A)R (BQ-123) and ET(B)R (BQ-788), specific IgG antibodies to each GPCRs; cholesterol or calcium-depleting drugs. RNA interference (ET(A)R or ET(B)R genes) in parasite infectivity was investigated in HSMCs. KEY RESULTS: BQ-123, BQ-788 and HOE-140 reduced leucocyte accumulation in HCP topically exposed to trypomastigotes and blocked inflammatory oedema in infected mice. Acting synergistically, ET(A)R and ET(B)R antagonists reduced parasite invasion of HSMCs to the same extent as HOE-140. Exogenous ET-1 potentiated T. cruzi uptake by HSMCs via ETRs/B(2)R, whereas RNA interference of ET(A)R and ET(B)R genes conversely reduced parasite internalization. ETRs/B(2)R-driven infection in HSMCs was reduced in HSMC pretreated with methyl-ß-cyclodextrin, a cholesterol-depleting drug, or in thapsigargin- or verapamil-treated target cells. CONCLUSIONS AND IMPLICATIONS: Our findings suggest that plasma leakage, a neutrophil-driven inflammatory response evoked by trypomastigotes via the kinin/endothelin pathways, may offer a window of opportunity for enhanced parasite invasion of cardiovascular cells.


Subject(s)
Chagas Disease/metabolism , Chagas Disease/parasitology , Receptor, Bradykinin B2/metabolism , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Trypanosoma cruzi/metabolism , Animals , Bradykinin B2 Receptor Antagonists , CHO Cells , Calcium/metabolism , Cells, Cultured , Chagas Disease/immunology , Chagas Disease/pathology , Cricetinae , Edema/metabolism , Edema/pathology , Endothelin A Receptor Antagonists , Endothelin B Receptor Antagonists , Endothelin-1/metabolism , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Inflammation/metabolism , Inflammation/pathology , Kinins/metabolism , Mice , Mice, Inbred BALB C , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Trypanosoma cruzi/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...