Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
STAR Protoc ; 5(2): 103009, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38602869

ABSTRACT

Double labeling to identify different markers in the same tissue section represents a useful tool either for in situ diagnosis or characterization of molecular associations. Here, we present a protocol to detect senescence-associated ß-galactosidase (SA-ßGal) and immunoperoxidase (IPO) activity in fresh-frozen murine tissues. We describe steps for tissue collection, solution preparation, SA-ßGal staining, IPO staining, hematoxylin counterstaining, microscopic observation, and signal quantification. This protocol can be used to detect in situ proteins alongside SA-ßGal activity. For complete details on the use and execution of this protocol, please refer to Pacheco-Rivera et al.1.

2.
Antioxidants (Basel) ; 13(3)2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38539791

ABSTRACT

Aging is characterized by increased reactive species, leading to redox imbalance, oxidative damage, and senescence. The adverse effects of alcohol consumption potentiate aging-associated alterations, promoting several diseases, including liver diseases. Nucleoredoxin (NXN) is a redox-sensitive enzyme that targets reactive oxygen species and regulates key cellular processes through redox protein-protein interactions. Here, we determine the effect of chronic alcohol consumption on NXN-dependent redox interactions in the liver of aged mice. We found that chronic alcohol consumption preferentially promotes the localization of NXN either into or alongside senescent cells, declines its interacting capability, and worsens the altered interaction ratio of NXN with FLII, MYD88, CAMK2A, and PFK1 proteins induced by aging. In addition, carbonylated protein and cell proliferation increased, and the ratios of collagen I and collagen III were inverted. Thus, we demonstrate an emerging phenomenon associated with altered redox homeostasis during aging, as shown by the declining capability of NXN to interact with partner proteins, which is enhanced by chronic alcohol consumption in the mouse liver. This evidence opens an attractive window to elucidate the consequences of both aging and chronic alcohol consumption on the downstream signaling pathways regulated by NXN-dependent redox-sensitive interactions.

3.
Pathogens ; 13(1)2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38251351

ABSTRACT

Naegleria fowleri is a ubiquitous free-living amoeba that causes primary amoebic meningoencephalitis. As a part of the innate immune response at the mucosal level, the proteins lactoferrin (Lf) and lysozyme (Lz) are secreted and eliminate various microorganisms. We demonstrate that N. fowleri survives the individual and combined effects of bovine milk Lf (bLf) and chicken egg Lz (cLz). Moreover, amoebic proliferation was not altered, even at 24 h of co-incubation with each protein. Trophozoites' ultrastructure was evaluated using transmission electron microscopy, and these proteins did not significantly alter their organelles and cytoplasmic membranes. Protease analysis using gelatin-zymograms showed that secreted proteases of N. fowleri were differentially modulated by bLf and cLz at 3, 6, 12, and 24 h. The bLf and cLz combination resulted in the inhibition of N. fowleri-secreted proteases. Additionally, the use of protease inhibitors on bLf-zymograms demonstrated that secreted cysteine proteases participate in the degradation of bLf. Nevertheless, the co-incubation of trophozoites with bLf and/or cLz reduced the cytopathic effect on the MDCK cell line. Our study suggests that bLf and cLz, alone or together, inhibited secreted proteases and reduced the cytopathic effect produced by N. fowleri; however, they do not affect the viability and proliferation of the trophozoites.

4.
Int J Mol Sci ; 24(13)2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37446394

ABSTRACT

Entamoeba histolytica (E. histolytica) is a parasite in humans that provokes amoebiasis. The most employed drug is metronidazole (MTZ); however, some studies have reported that this drug induces genotoxic effects. Therefore, it is necessary to explore new compounds without toxicity that can eliminate E. histolytica. Flavonoids are polyphenolic compounds that have demonstrated inhibition of growth and dysregulation of amoebic proteins. Despite the knowledge acquired to date, action mechanisms are not completely understood. The present work evaluates the effect of kaempferol against E. histolytica trophozoites and in the interactions with neutrophils from hamster, which is a susceptibility model. Our study demonstrated a significant reduction in the amoebic viability of trophozoites incubated with kaempferol at 150 µM for 90 min. The gene expression analysis showed a significant downregulation of Pr (peroxiredoxin), Rr (rubrerythrin), and TrxR (thioredoxin reductase). In interactions with amoebae and neutrophils for short times, we observed a reduction in ROS (reactive oxygen species), NO (nitric oxide), and MPO (myeloperoxidase) neutrophil activities. In conclusion, we confirmed that kaempferol is an effective drug against E. histolytica through the decrease in E. histolytica antioxidant enzyme expression and a regulator of several neutrophil mechanisms, such as MPO activity and the regulation of ROS and NO.


Subject(s)
Amoeba , Entamoeba histolytica , Humans , Animals , Cricetinae , Neutrophils/metabolism , Trophozoites , Reactive Oxygen Species/metabolism , Kaempferols/pharmacology , Kaempferols/metabolism
5.
Front Cell Infect Microbiol ; 13: 1150054, 2023.
Article in English | MEDLINE | ID: mdl-37360530

ABSTRACT

The protozoan disease is a major global health concern. Amoebiasis, leishmaniasis, Chagas disease, and African sleeping sickness affect several million people worldwide, leading to millions of deaths annually and immense social and economic problems. Iron is an essential nutrient for nearly all microbes, including invading pathogens. The majority of iron in mammalian hosts is stored intracellularly in proteins, such as ferritin and hemoglobin (Hb). Hb, present in blood erythrocytes, is a very important source of iron and amino acids for pathogenic microorganisms ranging from bacteria to eukaryotic pathogens, such as worms, protozoa, yeast, and fungi. These organisms have developed adequate mechanisms to obtain Hb or its byproducts (heme and globin) from the host. One of the major virulence factors identified in parasites is parasite-derived proteases, essential for host tissue degradation, immune evasion, and nutrient acquisition. The production of Hb-degrading proteases is a Hb uptake mechanism that degrades globin in amino acids and facilitates heme release. This review aims to provide an overview of the Hb and heme-uptake mechanisms utilized by human pathogenic protozoa to survive inside the host.


Subject(s)
Parasites , Animals , Humans , Parasites/metabolism , Hemoglobins/metabolism , Heme/metabolism , Endopeptidases , Peptide Hydrolases , Iron/metabolism , Mammals/metabolism
6.
Microorganisms ; 11(3)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36985284

ABSTRACT

Acanthamoeba castellanii genotype T4 is a clinically significant free-living amoeba that causes granulomatous amoebic encephalitis and amoebic keratitis in human beings. During the initial stages of infection, trophozoites interact with various host immune responses, such as lactoferrin (Lf), in the corneal epithelium, nasal mucosa, and blood. Lf plays an important role in the elimination of pathogenic microorganisms, and evasion of the innate immune response is crucial in the colonization process. In this study, we describe the resistance of A. castellanii to the microbicidal effect of bovine apo-lactoferrin (apo-bLf) at different concentrations (25, 50, 100, and 500 µM). Acanthamoeba castellanii trophozoites incubated with apo-bLf at 500 µM for 12 h maintained 98% viability. Interestingly, despite this lack of effect on viability, our results showed that the apo-bLf inhibited the cytopathic effect of A. castellanii in MDCK cells culture, and analysis of amoebic proteases by zymography showed significant inhibition of cysteine and serine proteases by interaction with the apo-bLf. From these results, we conclude that bovine apo-Lf influences the activity of A. castellanii secretion proteases, which in turn decreases amoebic cytopathic activity.

7.
Curr Pharm Des ; 28(40): 3243-3260, 2022.
Article in English | MEDLINE | ID: mdl-36284379

ABSTRACT

The emergence of multidrug-resistant bacterial strains with respect to commercially available antimicrobial drugs has marked a watershed in treatment therapies to fight pathogens and has stimulated research on alternative remedies. Proteins of the innate immune system of mammals have been highlighted as potentially yielding possible treatment options for infections. Lactoferrin (Lf) is one of these proteins; interestingly, no resistance to it has been found. Lf is a conserved cationic nonheme glycoprotein that is abundant in milk and is also present in low quantities in mucosal secretions. Moreover, Lf is produced and secreted by the secondary granules of neutrophils at infection sites. Lf is a molecule of approximately 80 kDa that displays multiple functions, such as antimicrobial, anti-viral, anti-inflammatory, and anticancer actions. Lf can synergize with antibiotics, increasing its potency against bacteria. Lactoferricins (Lfcins) are peptides resulting from the N-terminal end of Lf by proteolytic cleavage with pepsin. They exhibit several anti-bacterial effects similar to those of the parental glycoprotein. Synthetic analog peptides exhibiting potent antimicrobial properties have been designed. The aim of this review is to update understanding of the structure and effects of Lf and Lfcins as anti-bacterial compounds, focusing on the mechanisms of action in bacteria and the use of Lf in treatment of infections in patients, including those studies where no significant differences were found. Lf could be an excellent option for prevention and treatment of bacterial diseases, mainly in combined therapies with antibiotics or other antimicrobials.


Subject(s)
Anti-Infective Agents , Bacterial Infections , Animals , Humans , Lactoferrin/pharmacology , Lactoferrin/therapeutic use , Anti-Infective Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacteria , Peptides/metabolism , Mammals/metabolism
8.
Pharmaceutics ; 14(8)2022 Aug 15.
Article in English | MEDLINE | ID: mdl-36015327

ABSTRACT

Parasites and other eventually pathogenic organisms require the ability to adapt to different environmental conditions inside the host to assure survival. Some host proteins have evolved as defense constituents, such as lactoferrin (Lf), which is part of the innate immune system. Lf in its iron-free form (apo-Lf) and its peptides obtained by cleavage with pepsin are microbicides. Parasites confront Lf in mucosae and blood. In this work, the activity of Lf against pathogenic and opportunistic parasites such as Cryptosporidium spp., Eimeria spp., Entamoeba histolytica, Giardia duodenalis, Leishmania spp., Trypanosoma spp., Plasmodium spp., Babesia spp., Toxoplasma gondii, Trichomonas spp., and the free-living but opportunistic pathogens Naegleria fowleri and Acanthamoeba castellani were reviewed. The major effects of Lf could be the inhibition produced by sequestering the iron needed for their survival and the production of oxygen-free radicals to more complicated mechanisms, such as the activation of macrophages to phagocytes with the posterior death of those parasites. Due to the great interest in Lf in the fight against pathogens, it is necessary to understand the exact mechanisms used by this protein to affect their virulence factors and to kill them.

9.
Molecules ; 25(24)2020 Dec 08.
Article in English | MEDLINE | ID: mdl-33302377

ABSTRACT

Due to the emergence of multidrug-resistant pathogens, it is necessary to develop options to fight infections caused by these agents. Lactoferrin (Lf) is a cationic nonheme multifunctional glycoprotein of the innate immune system of mammals that provides numerous benefits. Lf is bacteriostatic and/or bactericidal, can stimulate cell proliferation and differentiation, facilitate iron absorption, improve neural development and cognition, promote bone growth, prevent cancer and exert anti-inflammatory and immunoregulatory effects. Lactoferrin is present in colostrum and milk and is also produced by the secondary granules of polymorphonuclear leukocytes, which store this glycoprotein and release it at sites of infection. Lf is also present in many fluids and exocrine secretions, on the surfaces of the digestive, respiratory and reproductive systems that are commonly exposed to pathogens. Apo-Lf (an iron-free molecule) can be microbiostatic due to its ability to capture ferric iron, blocking the availability of host iron to pathogens. However, apo-Lf is mostly microbicidal via its interaction with the microbial surface, causing membrane damage and altering its permeability function. Lf can inhibit viral entry by binding to cell receptors or viral particles. Lf is also able to counter different important mechanisms evolved by microbial pathogens to infect and invade the host, such as adherence, colonization, invasion, production of biofilms and production of virulence factors such as proteases and toxins. Lf can also cause mitochondrial and caspase-dependent regulated cell death and apoptosis-like in pathogenic yeasts. All of these mechanisms are important targets for treatment with Lf. Holo-Lf (the iron-saturated molecule) can contain up to two ferric ions and can also be microbicidal against some pathogens. On the other hand, lactoferricins (Lfcins) are peptides derived from the N-terminus of Lf that are produced by proteolysis with pepsin under acidic conditions, and they cause similar effects on pathogens to those caused by the parental Lf. Synthetic analog peptides comprising the N-terminus Lf region similarly exhibit potent antimicrobial properties. Importantly, there are no reported pathogens that are resistant to Lf and Lfcins; in addition, Lf and Lfcins have shown a synergistic effect with antimicrobial and antiviral drugs. Due to the Lf properties being microbiostatic, microbicidal, anti-inflammatory and an immune modulator, it represents an excellent natural alternative either alone or as adjuvant in the combat to antibiotic multidrug-resistant bacteria and other pathogens. This review aimed to evaluate the data that appeared in the literature about the effects of Lf and its derived peptides on pathogenic bacteria, protozoa, fungi and viruses and how Lf and Lfcins inhibit the mechanisms developed by these pathogens to cause disease.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Lactoferrin/chemistry , Lactoferrin/pharmacology , Peptides/chemistry , Peptides/pharmacology , Animals , Anti-Infective Agents/chemical synthesis , Antiparasitic Agents/chemical synthesis , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacology , Bacteria/drug effects , Bacterial Adhesion/drug effects , Cell Wall/drug effects , Chemistry Techniques, Synthetic , Fungi/drug effects , Host-Pathogen Interactions , Humans , Peptides/chemical synthesis , Proteolysis/drug effects , Structure-Activity Relationship , Virulence/drug effects , Virulence Factors , Viruses/drug effects
10.
Eur J Cell Biol ; 99(5): 151085, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32646643

ABSTRACT

The Naegleria are ubiquitous free-living amoebae and are characterized by the presence of three phases in their biological cycle: trophozoite, cyst and flagellate. Of this genus, only Naegleria fowleri has been reported as pathogenic to humans. The proteasome is a multi-catalytic complex and is considered to be the most important structure responsible for the degradation of intracellular proteins. This structure is related to the maintenance of cellular homeostasis and, in pathogenic microorganisms, to the modulation of their virulence. Until now, the proteasome and its function have not been described for the Naegleria genus. In the current study, using bioinformatic analysis, protein sequences homologous to those reported for the subunits of the 20S proteasome in other organisms were found, and virtual modelling was used to determine their three-dimensional structure. The presence of structural and catalytic subunits of the 20S proteasome was detected by Western and dot blot assays. Its localization was observed by immunofluorescence microscopy to be mainly in the cytoplasm, and a leading role of the chymotrypsin-like catalytic activity was determined using fluorogenic peptidase assays and specific proteasome inhibitors. Finally, the role of the 20S proteasome in the proliferation and differentiation of Naegleria genus trophozoites was demonstrated.


Subject(s)
Naegleria fowleri/chemistry , Proteasome Endopeptidase Complex/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Cell Proliferation
11.
Future Microbiol ; 15: 919-936, 2020 07.
Article in English | MEDLINE | ID: mdl-32716210

ABSTRACT

Currently, there is growing interest in the identification and purification of microbial lectins due to their involvement in the pathogenicity mechanisms of pathogens, such as Entamoeba histolytica and free-living amoebae. The Gal/GalNAc lectin from E. histolytica participates in adhesion, cytotoxicity and regulation of immune responses. Furthermore, mannose- and galactose-binding protein have been described in Acanthamoeba castellanii and Balamuthia mandrillaris, respectively and they also contribute to host damage. Finally, in Naegleria fowleri, molecules containing mannose and fucose are implicated in adhesion and cytotoxicity. Considering their relevance in the pathogenesis of the diseases caused by these protozoa, lectins appear to be promising targets in the diagnosis, vaccination and treatment of these infections.


Subject(s)
Amoeba/drug effects , Entamoeba histolytica/drug effects , Lectins/pharmacology , Virulence Factors , Amebiasis/diagnosis , Animals , Balamuthia mandrillaris , Entamoebiasis/diagnosis , Entamoebiasis/drug therapy , Entamoebiasis/parasitology , Glycoconjugates , Glycoproteins , Host-Parasite Interactions , Humans , Naegleria fowleri , Vaccination
12.
J Leukoc Biol ; 108(3): 895-908, 2020 09.
Article in English | MEDLINE | ID: mdl-32531828

ABSTRACT

Naegleria fowleri produces a fatal disease called primary amebic meningoencephalitis (PAM), which is characterized by an extensive inflammatory reaction in the CNS. It is known that the immune response is orchestrated mainly by neutrophils, which activate several defense mechanisms in the host, including phagocytosis, the release of different enzymes such as myeloperoxidase (MPO), and the production of neutrophil extracellular traps. However, the mechanisms by which amoebas evade the neutrophil response are still unknown. In this study, we analyzed the ability of N. fowleri to respond to the stress exerted by MPO. Interestingly, after the interaction of trophozoites with neutrophils, the amoeba viability was not altered; however, ultrastructural changes were observed. To analyze the influence of MPO against N. fowleri and its participation in free radical production, we evaluated its enzymatic activity, expression, and localization with and without the specific 4-aminobenzoic acid hydrazide inhibitor. The production of oxidizing molecules is the principal mechanism used by neutrophils to eliminate pathogens. In this context, we demonstrated an increase in the production of NO, superoxide anion, and reactive oxygen species; in addition, the overexpression of several antioxidant enzymes present in the trophozoites was quantified. The findings strongly suggest that N. fowleri possesses antioxidant machinery that is activated in response to an oxidative environment, allowing it to evade the neutrophil-mediated immune response, which may contribute to the establishment of PAM.


Subject(s)
Host-Parasite Interactions/immunology , Naegleria fowleri/metabolism , Neutrophils/physiology , Oxidoreductases/biosynthesis , Peroxidase/physiology , Protozoan Proteins/biosynthesis , Aniline Compounds/pharmacology , Animals , Cell Shape , Cytoplasmic Granules/enzymology , Cytoplasmic Granules/ultrastructure , Enzyme Induction , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C , Naegleria fowleri/enzymology , Naegleria fowleri/growth & development , Naegleria fowleri/ultrastructure , Neutrophils/drug effects , Nitric Oxide/metabolism , Oxidation-Reduction , Oxidative Stress , Oxidoreductases/genetics , Peroxidase/antagonists & inhibitors , Protozoan Proteins/genetics , Reactive Oxygen Species , Superoxides/metabolism , Vacuoles/ultrastructure
13.
Int J Med Microbiol ; 310(1): 151358, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31587966

ABSTRACT

Amoebiasis is a parasitic disease caused by Entamoeba histolytica (E. histolytica), an extracellular enteric protozoan. This infection mainly affects people from developing countries with limited hygiene conditions, where it is endemic. Infective cysts are transmitted by the fecal-oral route, excysting in the terminal ileum and producing invasive trophozoites (amoebae). E. histolytica mainly lives in the large intestine without causing symptoms; however, possibly as a result of so far unknown signals, the amoebae invade the mucosa and epithelium causing intestinal amoebiasis. E. histolytica possesses different mechanisms of pathogenicity for the adherence to the intestinal epithelium and for degrading extracellular matrix proteins, producing tissue lesions that progress to abscesses and a host acute inflammatory response. Much information has been obtained regarding the virulence factors, metabolism, mechanisms of pathogenicity, and the host immune response against this parasite; in addition, alternative treatments to metronidazole are continually emerging. An accesible and low-cost diagnostic method that can distinguish E. histolytica from the most nonpathogenic amoebae and an effective vaccine are necessary for protecting against amoebiasis. However, research about the disease and its prevention has been a challenge due to the relationship between E. histolytica and the host during the distinct stages of the disease is multifaceted. In this review, we analyze the interaction between the parasite, the human host, and the colon microbiota or pathogenic microorganisms, which together give rise to intestinal amoebiasis.


Subject(s)
Amebiasis/parasitology , Developing Countries , Dysentery, Amebic/parasitology , Intestines/parasitology , Public Health , Amebiasis/drug therapy , Amebiasis/epidemiology , Animals , Antiprotozoal Agents/therapeutic use , Dysentery, Amebic/epidemiology , Entamoeba histolytica/immunology , Entamoeba histolytica/pathogenicity , Feces/parasitology , Gastrointestinal Microbiome , Host-Pathogen Interactions , Humans , Intestines/microbiology , Metronidazole/therapeutic use , Mice , Virulence
14.
Eur J Protistol ; 72: 125640, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31794894

ABSTRACT

The plasma membrane is essential in the pathogenicity of several microorganisms. However, to date, there are few studies related to the plasma membrane proteins in Naegleria fowleri; this amoeba produces a fatal disease called primary amoebic meningoencephalitis. In the present study, we analyzed the electrophoretic pattern of the membrane proteins of N. fowleri and compared it with the nonpathogenic N. lovaniensis and N. gruberi. We detected a 23-kDa protein (Nf23) present at a higher level in N. fowleri than in the nonpathogenic amoebae. The mass spectrometry analysis showed that the Nf23 protein has a sequence of 229 amino acids that corresponds to a membrane protein. The mRNA level of nf23 was overexpressed 4-fold and 40,000-fold in N. fowleri compared with N. lovaniensis and N. gruberi, respectively. Moreover, we found a 5-fold overexpression of nf23 in N. fowleri trophozoites recovered from mouse brains compared with trophozoites axenically cultivated. In addition, the cytopathic effect on Madin-Darby Canine Kidney cells coincubated with N. fowleri diminished in the presence of antibodies against Nf23; nevertheless, the nonpathogenic amoebae did not produce damage to the monolayer cells. These results suggest that the plasma membrane protein Nf23 is probably involved in the virulence of N. fowleri.


Subject(s)
Naegleria fowleri/metabolism , Naegleria fowleri/pathogenicity , Naegleria/metabolism , Naegleria/pathogenicity , Protozoan Proteins/metabolism , Virulence/genetics , Animals , Brain/metabolism , Brain/parasitology , Dogs , Gene Expression , Madin Darby Canine Kidney Cells , Mice , Naegleria fowleri/genetics , Protozoan Proteins/genetics , Sequence Analysis, Protein
15.
Parasitol Int ; 74: 102002, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31669294

ABSTRACT

Acanthamoeba spp. are free-living amoebae with a worldwide distribution. These amoebae can cause granulomatous amoebic encephalitis and amoebic keratitis in humans. Proteases are considered virulence factors in pathogenic Acanthamoeba. The objective of this study was to evaluate the behavior of Acanthamoeba mauritaniensis, a nonpathogenic amoeba. We analyzed the cytopathic effect of A. mauritaniensis on RCE1(5 T5) and MDCK cells and compared it to that of Acanthamoeba castellanii. A partial biochemical characterization of proteases was performed in total crude extracts (TCE) and conditioned medium (CM). Finally, we evaluated the effect of proteases on tight junction (TJ) proteins and the transepithelial electrical resistance of MDCK cells. The results showed that this amoeba can induce substantial damage to RCE1(5T5) and MDCK cells. Moreover, the zymograms and Azocoll assays of amoebic TCE and CM revealed different protease activities, with serine proteases being the most active. Furthermore, A. mauritaniensis induced the alteration and degradation of MDCK cell TJ proteins with serine proteases. After genotyping this amoeba, we determined that it is an isolate of Acanthamoeba genotype T4D. From these data, we suggest that A. mauritaniensis genotype T4D behaves similarly to the A. castellanii strain.


Subject(s)
Acanthamoeba/genetics , Acanthamoeba/pathogenicity , Genotype , Acanthamoeba/enzymology , Animals , Dogs , Epithelial Cells/parasitology , Epithelial Cells/pathology , Madin Darby Canine Kidney Cells , Serine Proteases/metabolism , Tight Junction Proteins/metabolism
16.
J Eukaryot Microbiol ; 65(6): 804-819, 2018 11.
Article in English | MEDLINE | ID: mdl-29655298

ABSTRACT

Naegleria fowleri causes a fatal disease known as primary amoebic meningoencephalitis. This condition is characterized by an acute inflammation that originates from the free passage of peripheral blood cells to the central nervous system through the alteration of the blood-brain barrier. In this work, we established models of the infection in rats and in a primary culture of endothelial cells from rat brains with the aim of evaluating the activation and the alterations of these cells by N. fowleri. We proved that the rat develops the infection similar to the mouse model. We also found that amoebic cysteine proteases produced by the trophozoites and the conditioned medium induced cytopathic effect in the endothelial cells. In addition, N. fowleri can decrease the transendothelial electrical resistance by triggering the destabilization of the tight junction proteins claudin-5, occludin, and ZO-1 in a time-dependent manner. Furthermore, N. fowleri induced the expression of VCAM-1 and ICAM-1 and the production of IL-8, IL-1ß, TNF-α, and IL-6 as well as nitric oxide. We conclude that N. fowleri damaged the blood-brain barrier model by disrupting the intercellular junctions and induced the presence of inflammatory mediators by allowing the access of inflammatory cells to the olfactory bulbs.


Subject(s)
Blood-Brain Barrier/parasitology , Central Nervous System Protozoal Infections/metabolism , Endothelial Cells/metabolism , Naegleria fowleri/metabolism , Naegleria fowleri/pathogenicity , Tight Junction Proteins/metabolism , Animals , Central Nervous System Protozoal Infections/parasitology , Central Nervous System Protozoal Infections/pathology , Claudin-5/metabolism , Cysteine Proteases/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Male , Meningoencephalitis/parasitology , Meningoencephalitis/pathology , Mice , Mucous Membrane/parasitology , Mucous Membrane/pathology , Occludin/metabolism , Rats , Rats, Wistar , Trophozoites/metabolism , Tumor Necrosis Factor-alpha/metabolism , Turbinates/pathology , Vascular Cell Adhesion Molecule-1/metabolism , Zonula Occludens-1 Protein/metabolism
17.
Parasitol Res ; 117(1): 75-87, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29128927

ABSTRACT

Naegleria fowleri is a protozoan that invades the central nervous system and causes primary amoebic meningoencephalitis. It has been reported that N. fowleri induces an important inflammatory response during the infection. In the present study, we evaluated the roles of Toll-like receptors in the recognition of N. fowleri trophozoites by human mucoepithelial cells, analyzing the expression and production of innate immune response mediators. After amoebic interactions with NCI-H292 cells, the expression and production levels of IL-8, TNF-α, IL-1ß, and human beta defensin-2 were evaluated by RT-PCR, ELISA, immunofluorescence, and dot blot assays, respectively. To determine whether the canonical signaling pathways were engaged, we used different inhibitors, namely, IMG-2005 for MyD88 and BAY 11-7085 for the nuclear factor NFkB. Our results showed that the expression and production of the pro-inflammatory cytokines and beta defensin-2 were induced by N. fowleri mainly through the canonical TLR4 pathway in a time-dependent manner.


Subject(s)
Naegleria fowleri/immunology , Naegleria fowleri/metabolism , Toll-Like Receptors/metabolism , Amebiasis , Animals , Cell Line , Cytokines/metabolism , Defensins/metabolism , Epithelial Cells/immunology , Humans , Immunity, Innate , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Nitriles/pharmacology , Signal Transduction/drug effects , Sulfones/pharmacology , Trophozoites/immunology , Trophozoites/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Microbiology (Reading) ; 163(7): 940-949, 2017 07.
Article in English | MEDLINE | ID: mdl-28721850

ABSTRACT

Naegleria fowleri and Naegleria gruberi belong to the free-living amoebae group. It is widely known that the non-pathogenic species N. gruberi is usually employed as a model to describe molecular pathways in this genus, mainly because its genome has been recently described. However, N. fowleri is an aetiological agent of primary amoebic meningoencephalitis, an acute and fatal disease. Currently, the most widely used drug for its treatment is amphotericin B (AmB). It was previously reported that AmB has an amoebicidal effect in both N. fowleri and N. gruberi trophozoites by inducing morphological changes that resemble programmed cell death (PCD). PCD is a mechanism that activates morphological, biochemical and genetic changes. However, PCD has not yet been characterized in the genus Naegleria. The aim of the present work was to evaluate the typical markers to describe PCD in both amoebae. These results showed that treated trophozoites displayed several parameters of apoptosis-like PCD in both species. We observed ultrastructural changes, an increase in reactive oxygen species, phosphatidylserine externalization and a decrease in intracellular potassium, while DNA degradation was evaluated using the TUNEL assay and agarose gels, and all of these parameters are related to PCD. Finally, we analysed the expression of apoptosis-related genes, such as sir2 and atg8, in N. gruberi. Taken together, our results showed that AmB induces the morphological, biochemical and genetic changes of apoptosis-like PCD in the genus Naegleria.


Subject(s)
Amphotericin B/pharmacology , Antiprotozoal Agents/pharmacology , Apoptosis/drug effects , Central Nervous System Protozoal Infections/parasitology , Naegleria fowleri/drug effects , Naegleria/drug effects , Naegleria/cytology , Naegleria/genetics , Naegleria/growth & development , Naegleria fowleri/cytology , Naegleria fowleri/genetics , Naegleria fowleri/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Reactive Oxygen Species/metabolism , Trophozoites/drug effects , Trophozoites/growth & development
19.
Future Microbiol ; 12: 781-799, 2017 07.
Article in English | MEDLINE | ID: mdl-28608712

ABSTRACT

AIM: The aim of this work was to identify, characterize and evaluate the pathogenic role of mucinolytic activity released by Naegleria fowleri. MATERIALS & METHODS: Zymograms, protease inhibitors, anion exchange chromatography, MALDI-TOF-MS, enzymatic assays, Western blot, and confocal microscopy were used to identify and characterize a secreted mucinase; inhibition assays using antibodies, dot-blots and mouse survival tests were used to evaluate the mucinase as a virulence factor. RESULTS: A 94-kDa protein with mucinolytic activity was inducible and abolished by p-hydroxymercuribenzoate. MALDI-TOF-MS identified a glycoside hydrolase. Specific antibodies against N. fowleri-glycoside hydrolase inhibit cellular damage and MUC5AC degradation, and delay mouse mortality. CONCLUSION: Our findings suggest that secretory products from N. fowleri play an important role in mucus degradation during the invasion process.


Subject(s)
Glycoside Hydrolases/metabolism , Mucins/metabolism , Naegleria fowleri/enzymology , Virulence Factors/metabolism , Animals , Blotting, Western , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/drug effects , Humans , Hydroxymercuribenzoates/pharmacology , Mice , Microscopy, Confocal , Naegleria fowleri/drug effects , Naegleria fowleri/metabolism , Naegleria fowleri/pathogenicity , Polysaccharide-Lyases/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
20.
Parasitol Res ; 116(1): 303-312, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27796560

ABSTRACT

Members of the Naegleria genus are free-living amoebae, and the only pathogenic specie described to date for humans is N. fowleri. However, as the complete genome of this specie has not been reported, non-pathogenic N. gruberi is employed to describe molecular pathways in N. fowleri. Regardless, certain mechanisms, such as autophagy, have not yet been characterized in N. gruberi. Autophagy is involved in different cellular processes in some protozoa, including the recycling of unnecessary organelles, development, and cell differentiation. In this work, we characterized autophagy in N. gruberi using the specific inducer rapamycin. The formation of autophagy vacuoles in treated trophozoites was observed by ultrastructural analysis, and real time quantitative PCR demonstrated overexpression of the atg8 gene. In addition, we detected an increase in the vacuolar acidification of treated amoebae using the LysoTracker. Finally, confocal microscopy was utilized to identify Atg8 protein signal in the cytoplasm of N. gruberi trophozoites induced with rapamycin and even in trophozoites induced to encyst. In conclusion, N. gruberi possesses an Atg8 protein homolog that is overexpressed during the autophagic mechanism induced by rapamycin and also during encystation of this free-living amoeba.


Subject(s)
Autophagy-Related Protein 8 Family/physiology , Autophagy/physiology , Gene Expression Regulation/physiology , Naegleria , Animals , Anti-Bacterial Agents/pharmacology , Naegleria/ultrastructure , Sirolimus/pharmacology , Trophozoites , Ubiquitin
SELECTION OF CITATIONS
SEARCH DETAIL
...