Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Antioxidants (Basel) ; 13(2)2024 Feb 19.
Article En | MEDLINE | ID: mdl-38397843

Reactive sulfur species (RSS) like hydrogen sulfide (H2S) and cysteine persulfide (Cys-SSH) emerged as key signaling molecules with diverse physiological roles in the body, depending on their concentration and the cellular environment. While it is known that H2S and Cys-SSH are produced by both colonocytes and by the gut microbiota through sulfur metabolism, it remains unknown how these RSS affect amebiasis caused by Entamoeba histolytica, a parasitic protozoan that can be present in the human gastrointestinal tract. This study investigates H2S and Cys-SSH's impact on E. histolytica physiology and explores potential therapeutic implications. Exposing trophozoites to the H2S donor, sodium sulfide (Na2S), or to Cys-SSH led to rapid cytotoxicity. A proteomic analysis of Cys-SSH-challenged trophozoites resulted in the identification of >500 S-sulfurated proteins, which are involved in diverse cellular processes. Functional assessments revealed inhibited protein synthesis, altered cytoskeletal dynamics, and reduced motility in trophozoites treated with Cys-SSH. Notably, cysteine proteases (CPs) were significantly inhibited by S-sulfuration, affecting their bacterial biofilm degradation capacity. Immunofluorescence microscopy confirmed alterations in actin dynamics, corroborating the proteomic findings. Thus, our study reveals how RSS perturbs critical cellular functions in E. histolytica, potentially influencing its pathogenicity and interactions within the gut microbiota. Understanding these molecular mechanisms offers novel insights into amebiasis pathogenesis and unveils potential therapeutic avenues targeting RSS-mediated modifications in parasitic infections.

2.
NPJ Biofilms Microbiomes ; 9(1): 77, 2023 Oct 09.
Article En | MEDLINE | ID: mdl-37813896

The human protozoan parasite Entamoeba histolytica is responsible for amebiasis, a disease endemic to developing countries. E. histolytica trophozoites colonize the large intestine, primarily feeding on bacteria. However, in the gastrointestinal tract, bacterial cells form aggregates or structured communities called biofilms too large for phagocytosis. Remarkably, trophozoites are still able to invade and degrade established biofilms, utilizing a mechanism that mimics digestive exophagy. Digestive exophagy refers to the secretion of digestive enzymes that promote the digestion of objects too large for direct phagocytosis by phagocytes. E. histolytica cysteine proteinases (CPs) play a crucial role in the degradation process of Bacillus subtilis biofilm. These proteinases target TasA, a major component of the B. subtilis biofilm matrix, also contributing to the adhesion of the parasite to the biofilm. In addition, they are also involved in the degradation of biofilms formed by Gram-negative and Gram-positive enteric pathogens. Furthermore, biofilms also play an important role in protecting trophozoites against oxidative stress. This specific mechanism suggests that the amoeba has adapted to prey on biofilms, potentially serving as an untapped reservoir for novel therapeutic approaches to treat biofilms. Consistently, products derived from the amoeba have been shown to restore antibiotic sensitivity to biofilm cells. In addition, our findings reveal that probiotic biofilms can act as a protective shield for mammalian cells, hindering the progression of the parasite towards them.


Amoeba , Entamoeba histolytica , Animals , Humans , Entamoeba histolytica/metabolism , Phagocytosis , Gastrointestinal Tract , Biofilms , Mammals
3.
Cells ; 11(16)2022 08 12.
Article En | MEDLINE | ID: mdl-36010587

Queuosine (Q) is a naturally occurring modified nucleoside that occurs in the first position of transfer RNA anticodons such as Asp, Asn, His, and Tyr. As eukaryotes lack pathways to synthesize queuine, the Q nucleobase, they must obtain it from their diet or gut microbiota. Previously, we described the effects of queuine on the physiology of the eukaryotic parasite Entamoeba histolytica and characterized the enzyme EhTGT responsible for queuine incorporation into tRNA. At present, it is unknown how E. histolytica salvages queuine from gut bacteria. We used liquid chromatography-mass spectrometry (LC-MS) and N-acryloyl-3-aminophenylboronic acid (APB) PAGE analysis to demonstrate that E. histolytica trophozoites can salvage queuine from Q or E. coli K12 but not from the modified E. coli QueC strain, which cannot produce queuine. We then examined the role of EhDUF2419, a protein with homology to DNA glycosylase, as a queuine salvage enzyme in E. histolytica. We found that glutathione S-transferase (GST)-EhDUF2419 catalyzed the conversion of Q into queuine. Trophozoites silenced for EhDUF2419 expression are impaired in their ability to form Q-tRNA from Q or from E. coli. We also observed that Q or E. coli K12 partially protects control trophozoites from oxidative stress (OS), but not siEhDUF2419 trophozoites. Overall, our data reveal that EhDUF2419 is central for the direct salvaging of queuine from bacteria and for the resistance of the parasite to OS.


Entamoeba histolytica , Parasites , Animals , Entamoeba histolytica/metabolism , Escherichia coli/metabolism , Guanine/analogs & derivatives , Humans , Parasites/metabolism , RNA, Transfer/genetics
4.
Antioxidants (Basel) ; 11(5)2022 Apr 22.
Article En | MEDLINE | ID: mdl-35624678

Amebiasis is an intestinal disease transmitted by the protist parasite, Entamoeba histolytica. Lactobacillus acidophilus is a common inhabitant of healthy human gut and a probiotic that has antimicrobial properties against a number of pathogenic bacteria, fungi, and parasites. The aim of this study was to investigate the amebicide activity of L. acidophilus and its mechanisms. For this purpose, E. histolytica and L. acidophilus were co-incubated and the parasite's viability was determined by eosin dye exclusion. The level of ozidized proteins (OXs) in the parasite was determined by resin-assisted capture RAC (OX-RAC). Incubation with L. acidophilus for two hours reduced the viability of E. histolytica trophozoites by 50%. As a result of the interaction with catalase, an enzyme that degrades hydrogen peroxide (H2O2) to water and oxygen, this amebicide activity is lost, indicating that it is mediated by H2O2 produced by L. acidophilus. Redox proteomics shows that L. acidophilus triggers the oxidation of many essential amebic enzymes such as pyruvate: ferredoxin oxidoreductase, the lectin Gal/GalNAc, and cysteine proteases (CPs). Further, trophozoites of E. histolytica incubated with L. acidophilus show reduced binding to mammalian cells. These results support L. acidophilus as a prophylactic candidate against amebiasis.

5.
Antioxidants (Basel) ; 10(8)2021 Aug 02.
Article En | MEDLINE | ID: mdl-34439488

Auranofin (AF), an antirheumatic agent, targets mammalian thioredoxin reductase (TrxR), an important enzyme controlling redox homeostasis. AF is also highly effective against a diversity of pathogenic bacteria and protozoan parasites. Here, we report on the resistance of the parasite Entamoeba histolytica to 2 µM of AF that was acquired by gradual exposure of the parasite to an increasing amount of the drug. AF-adapted E. histolytica trophozoites (AFAT) have impaired growth and cytopathic activity, and are more sensitive to oxidative stress (OS), nitrosative stress (NS), and metronidazole (MNZ) than wild type (WT) trophozoites. Integrated transcriptomics and redoxomics analyses showed that many upregulated genes in AFAT, including genes encoding for dehydrogenase and cytoskeletal proteins, have their product oxidized in wild type trophozoites exposed to AF (acute AF trophozoites) but not in AFAT. We also showed that the level of reactive oxygen species (ROS) and oxidized proteins (OXs) in AFAT is lower than that in acute AF trophozoites. Overexpression of E. histolytica TrxR (EhTrxR) did not protect the parasite against AF, which suggests that EhTrxR is not central to the mechanism of adaptation to AF.

6.
mBio ; 12(2)2021 03 09.
Article En | MEDLINE | ID: mdl-33688012

Queuosine is a naturally occurring modified ribonucleoside found in the first position of the anticodon of the transfer RNAs for Asp, Asn, His, and Tyr. Eukaryotes lack pathways to synthesize queuine, the nucleobase precursor to queuosine, and must obtain it from diet or gut microbiota. Here, we describe the effects of queuine on the physiology of the eukaryotic parasite Entamoeba histolytica, the causative agent of amebic dysentery. Queuine is efficiently incorporated into E. histolytica tRNAs by a tRNA-guanine transglycosylase (EhTGT) and this incorporation stimulates the methylation of C38 in [Formula: see text] Queuine protects the parasite against oxidative stress (OS) and antagonizes the negative effect that oxidation has on translation by inducing the expression of genes involved in the OS response, such as heat shock protein 70 (Hsp70), antioxidant enzymes, and enzymes involved in DNA repair. On the other hand, queuine impairs E. histolytica virulence by downregulating the expression of genes previously associated with virulence, including cysteine proteases, cytoskeletal proteins, and small GTPases. Silencing of EhTGT prevents incorporation of queuine into tRNAs and strongly impairs methylation of C38 in [Formula: see text], parasite growth, resistance to OS, and cytopathic activity. Overall, our data reveal that queuine plays a dual role in promoting OS resistance and reducing parasite virulence.IMPORTANCEEntamoeba histolytica is a unicellular parasite that causes amebiasis. The parasite resides in the colon and feeds on the colonic microbiota. The gut flora is implicated in the onset of symptomatic amebiasis due to alterations in the composition of bacteria. These bacteria modulate the physiology of the parasite and affect the virulence of the parasite through unknown mechanisms. Queuine, a modified nucleobase of queuosine, is exclusively produced by the gut bacteria and leads to tRNA modification at the anticodon loops of specific tRNAs. We found that queuine induces mild oxidative stress resistance in the parasite and attenuates its virulence. Our study highlights the importance of bacterially derived products in shaping the physiology of the parasite. The fact that queuine inhibits the virulence of E. histolytica may lead to new strategies for preventing and/or treating amebiasis by providing to the host queuine directly or via probiotics.


Entamoeba histolytica/drug effects , Entamoeba histolytica/pathogenicity , Guanine/analogs & derivatives , Oxidative Stress/drug effects , Animals , Entamoeba histolytica/genetics , Female , Guanine/metabolism , Guanine/pharmacology , HeLa Cells , Humans , Methylation , Mice , Mice, Inbred BALB C , RNA, Transfer/metabolism
7.
Cell Microbiol ; 22(6): e13174, 2020 06.
Article En | MEDLINE | ID: mdl-32017328

Metronidazole (MNZ), the first line drug for amoebiasis and auranofin (AF), an emerging antiprotozoan drug, are both inhibiting Entamoeba histolytica thioredoxin reductase. The nature of oxidised proteins (OXs) formed in AF- or MNZ-treated E. histolytica trophozoites is unknown. In order to fill this knowledge gap, we performed a large-scale identification and quantification of the OXs formed in AF- or MNZ-treated E. histolytica trophozoites using resin-assisted capture coupled to mass spectrometry (MS). We detected 661 OXs in MNZ-treated trophozoites and 583 OXs in AF-treated trophozoites. More than 50% of these OXs were shared, and their functions include hydrolases, enzyme modulators, transferases, nucleic acid binding proteins, oxidoreductases, cytoskeletal proteins, chaperones, and ligases. Here, we report that the formation of actin filaments (F-actin) is impaired in AF-treated trophozoites. Consequently, their erythrophagocytosis, cytopathic activity, and their motility are impaired. We also observed that less than 15% of OXs present in H2 O2 -treated trophozoites are also present in AF- or MNZ-treated trophozoites. These results strongly suggest that the formation of OXs in AF- or MNZ-treated trophozoites and in H2 O2 -treated trophozoites occurred by two different mechanisms.


Auranofin/metabolism , Entamoeba histolytica/metabolism , Parasites/metabolism , Protozoan Proteins/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Movement , Cytoskeletal Proteins/metabolism , Hydrogen Peroxide/pharmacology , Lethal Dose 50 , Oxidoreductases , Trophozoites/drug effects , Trophozoites/metabolism , Virulence
8.
PLoS Pathog ; 14(10): e1007295, 2018 10.
Article En | MEDLINE | ID: mdl-30308066

Amebiasis, a global intestinal parasitic disease, is due to Entamoeba histolytica. This parasite, which feeds on bacteria in the large intestine of its human host, can trigger a strong inflammatory response upon invasion of the colonic mucosa. Whereas information about the mechanisms which are used by the parasite to cope with oxidative and nitrosative stresses during infection is available, knowledge about the contribution of bacteria to these mechanisms is lacking. In a recent study, we demonstrated that enteropathogenic Escherichia coli O55 protects E. histolytica against oxidative stress. Resin-assisted capture (RAC) of oxidized (OX) proteins coupled to mass spectrometry (OX-RAC) was used to investigate the oxidation status of cysteine residues in proteins present in E. histolytica trophozoites incubated with live or heat-killed E. coli O55 and then exposed to H2O2-mediated oxidative stress. We found that the redox proteome of E. histolytica exposed to heat-killed E. coli O55 is enriched with proteins involved in redox homeostasis, lipid metabolism, small molecule metabolism, carbohydrate derivative metabolism, and organonitrogen compound biosynthesis. In contrast, we found that proteins associated with redox homeostasis were the only OX-proteins that were enriched in E. histolytica trophozoites which were incubated with live E. coli O55. These data indicate that E. coli has a profound impact on the redox proteome of E. histolytica. Unexpectedly, some E. coli proteins were also co-identified with E. histolytica proteins by OX-RAC. We demonstrated that one of these proteins, E. coli malate dehydrogenase (EcMDH) and its product, oxaloacetate, are key elements of E. coli-mediated resistance of E. histolytica to oxidative stress and that oxaloacetate helps the parasite survive in the large intestine. We also provide evidence that the protective effect of oxaloacetate against oxidative stress extends to Caenorhabditis elegans.


Entamoeba histolytica/drug effects , Entamoebiasis/drug therapy , Escherichia coli/physiology , Oxaloacetic Acid/pharmacology , Oxidative Stress/drug effects , Protozoan Proteins/metabolism , Amebiasis/drug therapy , Amebiasis/metabolism , Amebiasis/parasitology , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/parasitology , Cells, Cultured , Entamoebiasis/metabolism , Entamoebiasis/parasitology , HeLa Cells , Humans , Intestine, Large/drug effects , Intestine, Large/metabolism , Intestine, Large/parasitology , Macrophages/cytology , Macrophages/drug effects , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA
9.
Sci Rep ; 8(1): 9042, 2018 06 13.
Article En | MEDLINE | ID: mdl-29899530

Oxidative stress is one of the strongest toxic factors in nature: it can harm or even kill cells. Cellular means of subverting the toxicity of oxidative stress are important for the success of infectious diseases. Many types of bacterium inhabit the intestine, where they can encounter pathogens. During oxidative stress, we analyzed the interplay between an intestinal parasite (the pathogenic amoeba Entamoeba histolytica - the agent of amoebiasis) and enteric bacteria (microbiome residents, pathogens and probiotics). We found that live enteric bacteria protected E. histolytica against oxidative stress. By high-throughput RNA sequencing, two amoebic regulatory modes were observed with enteric bacteria but not with probiotics. The first controls essential elements of homeostasis, and the second the levels of factors required for amoeba survival. Characteristic genes of both modes have been acquired by the amoebic genome through lateral transfer from the bacterial kingdom (e.g. glycolytic enzymes and leucine-rich proteins). Members of the leucine-rich are homologous to proteins from anti-bacterial innate immune such as Toll-like receptors. The factors identified here suggest that despite its old age in evolutionary terms, the protozoan E. histolytica displays key characteristics of higher eukaryotes' innate immune systems indicating that components of innate immunity existed in the common ancestor of plants and animals.


Entamoeba histolytica/immunology , Enterobacteriaceae/immunology , Gastrointestinal Microbiome/immunology , Immunity, Innate/immunology , Oxidative Stress/immunology , Entamoeba histolytica/genetics , Entamoeba histolytica/physiology , Enterobacteriaceae/physiology , Escherichia coli/immunology , Escherichia coli/physiology , Gastrointestinal Microbiome/physiology , Homeostasis/immunology , Humans , Intestines/immunology , Intestines/microbiology , Intestines/parasitology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Transcriptome/immunology
10.
Article En | MEDLINE | ID: mdl-28589096

We have recently reported that Entamoeba histolytica trophozoites can adapt to toxic levels of the nitric oxide (NO) donor, S-nitrosoglutathione (GSNO). Even if the consequences of this adaptation on the modulation of gene expression in NO-adapted trophozoites (NAT) have been previously explored, insight on S-nitrosylated (SNO) proteins in NAT is missing. Our study aims to fill this knowledge gap by performing a screening of SNO proteins in NAT. Employing SNO resin-assisted capture (RAC), we identified 242 putative SNO proteins with key functions in calcium binding, enzyme modulation, redox homeostasis, and actin cytoskeleton. Of the SNO proteins in NAT, proteins that are associated with actin family cytoskeleton protein are significantly enriched. Here we report that the formation of actin filaments (F-actin) is impaired in NAT. Consequently, the ability of NAT to ingest erythrocytes and their motility and their cytopathic activity are impaired. These phenotypes can be imitated by treating control parasite with cytochalasin D (CytD), a drug that binds to F-actin polymer and prevent polymerization of actin monomers. Removal of GSNO from the culture medium of NAT restored the sensitivity of the parasite to nitrosative stress (NS) and its ability to form F-actin formation and its virulence. These results establish the central role of NO in shaping the virulence of the parasite through its effect on F-actin formation and highlight the impressive ability of this parasite to adapt to NS.


Actins/metabolism , Entamoeba histolytica/chemistry , Entamoeba histolytica/metabolism , Nitrosative Stress , Protozoan Proteins/metabolism , S-Nitrosothiols/chemistry , Virulence , Actin Cytoskeleton/metabolism , Actins/ultrastructure , Cell Movement/physiology , Cysteine/analogs & derivatives , Entamoeba histolytica/drug effects , Entamoeba histolytica/pathogenicity , Erythrocytes/parasitology , Gene Expression , Microscopy, Confocal , Nitric Oxide/pharmacology , Proteolysis , Protozoan Proteins/genetics , Trophozoites/metabolism , Virulence/drug effects
12.
Sci Rep ; 6: 36323, 2016 11 03.
Article En | MEDLINE | ID: mdl-27808157

Adaptation of the Entamoeba histolytica parasite to toxic levels of nitric oxide (NO) that are produced by phagocytes may be essential for the establishment of chronic amebiasis and the parasite's survival in its host. In order to obtain insight into the mechanism of E. histolytica's adaptation to NO, E. histolytica trophozoites were progressively adapted to increasing concentrations of the NO donor drug, S-nitrosoglutathione (GSNO) up to a concentration of 110 µM. The transcriptome of NO adapted trophozoites (NAT) was investigated by RNA sequencing (RNA-seq). N-acetyl ornithine deacetylase (NAOD) was among the 208 genes that were upregulated in NAT. NAOD catalyzes the deacetylation of N-acetyl-L-ornithine to yield ornithine and acetate. Here, we report that NAOD contributes to the better adaptation of the parasite to nitrosative stress (NS) and that this function does not depend on NAOD catalytic activity. We also demonstrated that glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is detrimental to E. histolytica exposed to NS and that this detrimental effect is neutralized by NAOD or by a catalytically inactive NAOD (mNAOD). These results establish NAOD as a moonlighting protein, and highlight the unexpected role of this metabolic enzyme in the adaptation of the parasite to NS.


Entamoeba histolytica/physiology , Nitrosative Stress , Ornithine Decarboxylase/genetics , S-Nitrosoglutathione/pharmacology , Animals , Dipeptides/metabolism , Entamoeba histolytica/enzymology , Entamoeba histolytica/genetics , Gene Expression Profiling , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , HeLa Cells , Humans , Mice , Ornithine Decarboxylase/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RAW 264.7 Cells , Sequence Analysis, RNA , Up-Regulation
13.
PLoS Negl Trop Dis ; 10(1): e0004340, 2016 Jan.
Article En | MEDLINE | ID: mdl-26735309

Entamoeba histolytica is an obligate protozoan parasite of humans, and amebiasis, an infectious disease which targets the intestine and/or liver, is the second most common cause of human death due to a protozoan after malaria. Although amebiasis is usually asymptomatic, E. histolytica has potent pathogenic potential. During host infection, the parasite is exposed to reactive oxygen species that are produced and released by cells of the innate immune system at the site of infection. The ability of the parasite to survive oxidative stress (OS) is essential for a successful invasion of the host. Although the effects of OS on the regulation of gene expression in E. histolytica and the characterization of some proteins whose function in the parasite's defense against OS have been previously studied, our knowledge of oxidized proteins in E. histolytica is lacking. In order to fill this knowledge gap, we performed a large-scale identification and quantification of the oxidized proteins in oxidatively stressed E. histolytica trophozoites using resin-assisted capture coupled to mass spectrometry. We detected 154 oxidized proteins (OXs) and the functions of some of these proteins were associated with antioxidant activity, maintaining the parasite's cytoskeleton, translation, catalysis, and transport. We also found that oxidation of the Gal/GalNAc impairs its function and contributes to the inhibition of E. histolytica adherence to host cells. We also provide evidence that arginase, an enzyme which converts L-arginine into L-ornithine and urea, is involved in the protection of the parasite against OS. Collectively, these results emphasize the importance of OS as a critical regulator of E. histolytica's functions and indicate a new role for arginase in E. histolytica's resistance to OS.


Arginase/metabolism , Entamoeba histolytica/metabolism , Oxidative Stress/physiology , Proteomics/methods , Cell Adhesion , Entamoeba histolytica/genetics , HeLa Cells , Humans , Lectins/metabolism , Resins, Synthetic , Trophozoites/physiology
14.
PLoS One ; 9(3): e91518, 2014.
Article En | MEDLINE | ID: mdl-24626316

Entamoeba histolytica is a gastrointestinal protozoan parasite that causes amebiasis, a disease which has a worldwide distribution with substantial morbidity and mortality. Nitrosative stress, which is generated by innate immune cells, is one of the various environmental challenges that E. histolytica encounters during its life cycle. Although the effects of nitric oxide (NO) on the regulation of gene expression in this parasite have been previously investigated, our knowledge on S-nitrosylated proteins in E.histolytica is lacking. In order to fill this knowledge gap, we performed a large-scale detection of S-nitrosylated (SNO) proteins in E.histolytica trophozoites that were treated with the NO donor, S-nitrosocysteine by resin-assisted capture (RAC). We found that proteins involved in glycolysis, gluconeogenesis, translation, protein transport, and adherence to target cells such as the heavy subunit of Gal/GalNac lectin are among the S-nitrosylated proteins that were enriched by SNO-RAC. We also found that the S-nitrosylated cysteine residues in the carbohydrate recognition domain (CRD) of Gal/GalNAc lectin impairs its function and contributes to the inhibition of E.histolytica adherence to host cells. Collectively, these results advance our understanding of the mechanism of reduced E.histolytica adherence to mammalian cells by NO and emphasize the importance of NO as a regulator of key physiological functions in E.histolytica.


Cysteine/analogs & derivatives , Entamoeba histolytica/chemistry , Lectins/chemistry , Nitric Oxide/chemistry , Nitrogen/chemistry , S-Nitrosothiols/chemistry , Carbohydrates/chemistry , Cell Adhesion , Chromatography, Affinity , Cysteine/chemistry , Entamoebiasis/immunology , Entamoebiasis/parasitology , Glycolysis , HeLa Cells , Humans , Protein Transport , Proteome , Proteomics , Protozoan Proteins/genetics
15.
Cell Microbiol ; 16(8): 1211-23, 2014 Aug.
Article En | MEDLINE | ID: mdl-24471581

The Entamoeba histolytica-methylated LINE-binding protein (EhMLBP) binds to methylated repetitive DNA and is a positive regulator of a reverse transcriptase of a long interspersed nucleotide element (LINE). This protein protects trophozoites against heat shock by reducing protein aggregation. The presence of EhMLBP and polyubiquitinated proteins in heat shock-induced protein aggregates raised the question whether these proteins interact. This assumption was confirmed by co-immunoprecipitation experiments: ubiquitinated proteins were detected in the perinuclear region of non-stressed E. histolytica trophozoites, whereas ubiquitinated proteins were detected in the perinuclear region and colocalized with EhMLBP in cytoplasmic granules in heat-shocked trophozoites. We also observed that overexpression of the reverse transcriptase of EhRLE3 induced the upregulation of EhMLBP expression and the formation of these EhMLBP-containing granules. Since (i) these EhMLBP-containing granules in the cytoplasm of heat-shocked E. histolytica trophozoites also contain polyubiquitinated proteins and poly(A)(+) mRNA and (ii) their formation is promoted by sodium arsenate, puromycin, and pateamine A and is inhibited by cycloheximide, we propose that these cytoplasmic EhMLBP-containing granules are stress granules. Our data also suggest that the formation of these granules is dependent upon EhMLBP and LINE.


Cytoplasmic Granules/metabolism , Entamoeba histolytica/pathogenicity , Long Interspersed Nucleotide Elements/genetics , RNA-Directed DNA Polymerase/genetics , Ubiquitinated Proteins/metabolism , Arsenates/pharmacology , DNA Methylation/genetics , DNA, Protozoan/genetics , Dysentery, Amebic/pathology , Entamoeba histolytica/genetics , Entamoebiasis/pathology , Epoxy Compounds/pharmacology , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Heat-Shock Response , Macrolides/pharmacology , Protein Synthesis Inhibitors/pharmacology , Protozoan Proteins/metabolism , Puromycin/pharmacology , RNA-Directed DNA Polymerase/biosynthesis , Thiazoles/pharmacology
16.
Connect Tissue Res ; 53(2): 169-79, 2012.
Article En | MEDLINE | ID: mdl-22149722

The familial disease of hereditary multiple exostoses is characterized by abnormal skeletal deformities requiring extensive surgical procedures. In hereditary multiple exostoses patients there is a shortage in the pericellular glycosaminoglycan (GAG) of heparan sulfate (HS), related to defective activity of HS glycosyltransferases, mainly in the pericellular regions of chondrocytes. This study searched for a novel approach employing xylosides with different aglycone groups priming a variety of GAG chains, in attempting to alter the GAG compositional profile. Cell cultures of patients with osteochondroma responded to p-nitrophenyl ß-D-xyloside by a significant increase in total GAG synthesis, expressed mainly in the extracellular domains, limited to chondroitin sulfate). The different ß-D-xylosides, in addition to increasing the synthesis of extracellular GAGs, led to a significant depletion of the intracellular GAG domains. In mouse chondrocyte cultures, ß-D-xylosides with different aglycones created a unique distribution of the GAG pools. Of special interest was the finding that the naphthalene methanol ß-D-xyloside showed the highest absolute levels of HS-GAGs in both extracellular and intra-pericellular moieties compared with other ß-D-xylosides and with controls without xyloside. In summary, ß-D-xylosides can be utilized in chondrocyte cultures to modify the distribution of GAGs between the extracellular and intracellular compartments. In addition, xylosides may alter the profile of specific GAG chains in each moiety.


Chondrocytes/drug effects , Chondroitin Sulfates/biosynthesis , Glycosides/pharmacology , Animals , Animals, Newborn , Cell Line , Chondrocytes/metabolism , Dose-Response Relationship, Drug , Humans , Mice , Naphthalenes/pharmacology , Osteochondroma , Tumor Cells, Cultured
17.
Int J Exp Pathol ; 89(5): 321-31, 2008 Oct.
Article En | MEDLINE | ID: mdl-18452536

The different clinical entities of osteochondromas, hereditary multiple exostoses (HME) and non-familial solitary exostosis, are known to express localized exostoses in their joint metaphyseal cartilage. In the current study biopsies of osteochondromas patients were screened with respect to a number of cellular and molecular parameters. Specifically, cartilaginous biopsy samples of nine HME patients, 10 solitary exostosis patients and 10 articular cartilages of control subjects were collected and cell cultures were established. Results obtained showed that one of the two HME samples that underwent DNA sequencing analysis (HME-1) had a novel mutation for an early stop codon, which led to an aberrant protein, migrating at a lower molecular weight position. The EXT-1 mRNA and protein levels in chondrocyte cultures derived from all nine HME patients were elevated, compared with solitary exostosis patients or control subjects. Furthermore, cell cultures of HME patients had significantly decreased pericellular heparan sulphate (HS) in comparison with cultures of solitary exostosis patients or control subjects. Immunohistochemical staining of tissue sections and Western blotting of cell cultures derived from HME patients revealed higher levels of heparanase compared with solitary exostosis patients and of control subjects. Further investigations are needed to determine whether the low pericellular HS levels in HME patients stem from decreased biosynthesis of HS, increased degradation or a combination of both. In conclusion, it appears that due to a mutated glycosyltransferase, the low content of pericellular HS in HME patients leads to the anatomical deformations with exostoses formation. Hence, elevation of HS content in the pericellular regions should be a potential molecular target for correction.


Chondrocytes/metabolism , Exostoses, Multiple Hereditary/genetics , Exostoses/genetics , N-Acetylglucosaminyltransferases/genetics , Antibody Specificity , Base Sequence , Case-Control Studies , Cells, Cultured , Chondrocytes/pathology , DNA Mutational Analysis , Exostoses/pathology , Gene Expression , Glucuronidase/analysis , Glucuronidase/genetics , Glycosaminoglycans/analysis , Glycosaminoglycans/genetics , Humans , Immunoblotting/methods , Immunohistochemistry , Molecular Sequence Data , N-Acetylglucosaminyltransferases/analysis , N-Acetylglucosaminyltransferases/immunology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods
18.
Exp Mol Pathol ; 74(1): 40-8, 2003 Feb.
Article En | MEDLINE | ID: mdl-12645631

Hereditary osteochondromas are often caused by mutation in the EXT1 gene. The lesions are typified by formation of a "pseudo" growth plate like lesion growing at 60 degrees to the normal growth direction of the bone. Such lesions can be mimicked surgically by reverting the position--the polarity of the zone of LaCroix. The current study attempts to compare the pathology between EXT1 gene expression in humans and surgically created osteochondroma pathology in a rat model. Tissues of human bunion, human embryonal tissue, and human adult cartilage as well as normal rat epiphyses served as controls. Rats were operated on and a 60 degree span of the ring of LaCroix was inverted as described by Delgado (Delgado, E., Rodriguez, J. I., Serada, A., Tellez, M., and Pariagoa, R.. Clin. Orthop. 201, 251-258 (1985)). The surgically created osteochondromas were assessed by histology, histochemistry, and immunohistochemistry. The findings show that the surgically created lesions contain only a small amount of FGF receptor 3 (FGFR3) expressed on mesenchymal stem cells located in the perichondrium, as compared to the cell population carrying FGFR3 in the contralateral limb. Indian hedgehog and Bcl2 are downregulated, while BMP-2 is overexpressed in the operated limb, compared to the LaCroix ring of the contralaetral limb. The shortage, as well as the disturbed migration routes of the residual mesenchymal stem cells in surgically created osteochondromas leads eventually to resorption of the pathological elements. In search of additional markers characterizing such pathological structures composed of mesenchymal stem cells and cartilaginous and bony cells, EXT1 gene was found to be expressed in the surgically created osteochondromas, like in normal growth plates. Nitric oxide synthase was also expressed like in adult cartilage, though tumor necrosis factor alpha typifying Bunion formation was absent. In summary, surgically created osteochondromas lack the massive and continuous population of mesenchymal stem cells with Bcl2 expression. However, the small residual mesenchymal cell population gives rise to short-lived EXT1-expressing cells that disappear eventually due to spontaneous resorption.


Cartilage/pathology , Exostoses, Multiple Hereditary/pathology , N-Acetylglucosaminyltransferases/genetics , Osteochondroma/pathology , Protein-Tyrosine Kinases , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Cartilage/metabolism , Disease Models, Animal , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/metabolism , Hindlimb/diagnostic imaging , Hindlimb/surgery , Humans , N-Acetylglucosaminyltransferases/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Osteochondroma/genetics , Osteochondroma/metabolism , Radiography , Rats , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/metabolism , Stem Cells/pathology , Stem Cells/physiology
...