Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(15)2024 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-39125801

RESUMEN

Mannheimia haemolytica is the main etiological bacterial agent in ruminant respiratory disease. M. haemolytica secretes leukotoxin, lipopolysaccharides, and proteases, which may be targeted to treat infections. We recently reported the purification and in vivo detection of a 110 kDa Zn metalloprotease with collagenase activity (110-Mh metalloprotease) in a sheep with mannheimiosis, and this protease may be an important virulence factor. Due to the increase in the number of multidrug-resistant strains of M. haemolytica, new alternatives to antibiotics are being explored; one option is lactoferrin (Lf), which is a multifunctional iron-binding glycoprotein from the innate immune system of mammals. Bovine apo-lactoferrin (apo-bLf) possesses many properties, and its bactericidal and bacteriostatic effects have been highlighted. The present study was conducted to investigate whether apo-bLf inhibits the secretion and proteolytic activity of the 110-Mh metalloprotease. This enzyme was purified and sublethal doses of apo-bLf were added to cultures of M. haemolytica or co-incubated with the 110-Mh metalloprotease. The collagenase activity was evaluated using zymography and azocoll assays. Our results showed that apo-bLf inhibited the secretion and activity of the 110-Mh metalloprotease. Molecular docking and overlay assays showed that apo-bLf bound near the active site of the 110-Mh metalloprotease, which affected its enzymatic activity.


Asunto(s)
Lactoferrina , Mannheimia haemolytica , Metaloproteasas , Proteolisis , Lactoferrina/metabolismo , Lactoferrina/farmacología , Metaloproteasas/metabolismo , Metaloproteasas/antagonistas & inhibidores , Animales , Apoproteínas/metabolismo , Apoproteínas/química , Simulación del Acoplamiento Molecular , Ovinos , Bovinos , Colagenasas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Zinc/metabolismo
2.
Front Cell Infect Microbiol ; 13: 1150054, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37360530

RESUMEN

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.


Asunto(s)
Parásitos , Animales , Humanos , Parásitos/metabolismo , Hemoglobinas/metabolismo , Hemo/metabolismo , Endopeptidasas , Péptido Hidrolasas , Hierro/metabolismo , Mamíferos/metabolismo
3.
Pharmaceutics ; 14(8)2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-36015327

RESUMEN

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.

4.
Int J Parasitol ; 50(12): 959-967, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32822678

RESUMEN

Amoebiasis is a parasitic infection of the human large intestine caused by Entamoeba histolytica; this disease mainly affects people from developing countries. To survive, this primitive protozoan has a high demand for iron, and it uses host iron proteins upon invasion. Transferrin (Tf) is a plasma iron-binding protein that transports and delivers iron to all cells. Iron-loaded Tf (holoTf) in humans can support the proliferation of amoebae in vitro by binding to an amoebic TfR (EhTfR), and amoebae endocytose it inside clathrin-coated vesicles. In this study, it was found that EhTfR phosphorylation is required for human holoTf endocytosis by E. histolytica. Once this complex is endocytosed, human holoTf could be degraded with a nutritional purpose by cysteine proteases. HoloTf endocytosis initiates the activation of the mitogen-activated protein kinases (MAPKs) and focal adhesion kinase (FAK) pathways, which induce cell proliferation with phosphoinositide 3-kinase (PI-3 K) and Ca2+ involvement. In the first minutes after holoTf is endocytosed, several proteins are phosphorylated including transketolase, enolase, L-myo-inositol-1-phosphate synthase and phosphoglucomutase, which control carbohydrate metabolism, and heat shock protein-70. The study of these proteins and their signal transduction pathways could be useful for developing future therapies.


Asunto(s)
Endocitosis , Entamoeba histolytica , Transducción de Señal , Transferrina/química , Calcio , Quinasa 1 de Adhesión Focal , Humanos , Proteínas Quinasas Activadas por Mitógenos , Fosfatidilinositol 3-Quinasas
5.
Vet Res ; 51(1): 36, 2020 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-32138772

RESUMEN

Mannheimia haemolytica serotype A2 is the principal cause of pneumonic mannheimiosis in ovine and caprine livestock; this disease is a consequence of immune suppression caused by stress and associated viruses and is responsible for significant economic losses in farm production worldwide. Gram-negative bacteria such as M. haemolytica produce outer membrane (OM)-derived spherical structures named outer membrane vesicles (OMVs) that contain leukotoxin and other biologically active virulence factors. In the present study, the relationship between M. haemolytica A2 and bovine lactoferrin (BLf) was studied. BLf is an 80 kDa glycoprotein that possesses bacteriostatic and bactericidal properties and is part of the mammalian innate immune system. Apo-BLf (iron-free) showed a bactericidal effect against M. haemolytica A2, with an observed minimal inhibitory concentration (MIC) of 16 µM. Sublethal doses (2-8 µM) of apo-BLf increased the release of OMVs, which were quantified by flow cytometry. Apo-BLf modified the normal structure of the OM and OMVs, as observed through transmission electron microscopy. Apo-BLf also induced lipopolysaccharide (LPS) release from bacteria, disrupting OM permeability and functionality, as measured by silver staining and SDS and polymyxin B cell permeability assays. Western blot results showed that apo-BLf increased the secretion of leukotoxin in M. haemolytica A2 culture supernatants, possibly through its iron-chelating activity. In contrast, holo-BLf (with iron) did not have this effect, possibly due to differences in the tertiary structure between these proteins. In summary, apo-BLf affected the levels of several M. haemolytica virulence factors and could be evaluated for use in animals as an adjuvant in the treatment of ovine mannheimiosis.


Asunto(s)
Antibacterianos/farmacología , Exotoxinas , Lactoferrina/farmacología , Mannheimia haemolytica/efectos de los fármacos , Pasteurelosis Neumónica/tratamiento farmacológico , Enfermedades de las Ovejas/tratamiento farmacológico , Animales , Mannheimia haemolytica/fisiología , Ovinos
6.
Int J Med Microbiol ; 310(1): 151358, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31587966

RESUMEN

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.


Asunto(s)
Amebiasis/parasitología , Países en Desarrollo , Disentería Amebiana/parasitología , Intestinos/parasitología , Salud Pública , Amebiasis/tratamiento farmacológico , Amebiasis/epidemiología , Animales , Antiprotozoarios/uso terapéutico , Disentería Amebiana/epidemiología , Entamoeba histolytica/inmunología , Entamoeba histolytica/patogenicidad , Heces/parasitología , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Humanos , Intestinos/microbiología , Metronidazol/uso terapéutico , Ratones , Virulencia
8.
Microbiology (Reading) ; 163(9): 1329-1342, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28100304

RESUMEN

In Entamoeba histolytica, iron modulates virulence and gene expression via unknown regulatory mechanisms. The existence of a posttranscriptional iron regulatory system parallel with the iron-responsive element (IRE)/iron regulatory protein (IRP) system in the protozoan Trichomonas vaginalis has recently been reported. Due to their evolutionary closeness and the importance of iron for growth and virulence in these protozoa, we hypothesized the existence of an IRE/IRP-like mechanism in E. histolytica. To determine the presence of IRE-like elements in some mRNAs from this parasite, we performed in silico analyses of the 5'- and 3'-UTRs of mRNAs encoding virulence factors and cytoskeleton, ribosomal and metabolism proteins. The Zuker mfold software predicted IRE-like secondary structures in 52 of the 135 mRNAs analysed. However, only nine structures shared sequence similarity with the apical loop sequence (CAGUGN) of the previously reported human IRE-ferritin, whereas the GUU/UUG protozoan-specific motif was detected in 23 stem-loop structures. A new motif, AUU/AUUU, was also observed in 23 structures, suggesting the possible existence of an amoeba-specific motif. Additionally, cross-linking and RNA electrophoretic mobility shift assays showed specific RNA-protein interactions, using as a model two amoebic IRE-like elements from iron-regulated mRNAs and HeLa, T. vaginalis and E. histolytica cytoplasmic proteins. Our data suggest the presence of a posttranscriptional iron regulatory IRE/IRP-like mechanism in E. histolytica.


Asunto(s)
Entamoeba histolytica/genética , Entamoeba histolytica/metabolismo , Regulación de la Expresión Génica , Hierro/metabolismo , Elementos de Respuesta , Células Cultivadas , Entamebiasis/metabolismo , Entamebiasis/parasitología , Células HeLa , Interacciones Huésped-Parásitos , Humanos , Secuencias Invertidas Repetidas , Conformación de Ácido Nucleico , Unión Proteica , ARN Mensajero/química , ARN Mensajero/genética
9.
Front Microbiol ; 8: 2633, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29375503

RESUMEN

Vibrio is a genus of Gram-negative bacteria, some of which can cause serious infectious diseases. Vibrio infections are associated with the consumption of contaminated food and classified in Vibrio cholera infections and non-cholera Vibrio infections. In the present study, we investigate whether bovine lactoferrin (bLF) and several synthetic peptides corresponding to bLF sequences, are able to inhibit the growth or have bactericidal effect against V. cholerae and other Vibrio species. The antibacterial activity of LF and LF-peptides was assessed by kinetics of growth or determination of colony forming unit in bacteria treated with the peptides and antibiotics. To get insight in the mode of action, the interaction between bLF and bLF-peptides (coupled to FITC) and V. cholera was evaluated. The damage of effector-induced bacterial membrane permeability was measured by inclusion of the fluorescent dye propidium iodide using flow cytometry, whereas the bacterial ultrastructural damage in bacteria treated was observed by transmission electron microscopy. The results showed that bLF and LFchimera inhibited the growth of the V. cholerae strains; LFchimera permeabilized the bacteria which membranes were seriously damaged. Assays with a multidrug-resistant strain of Vibrio species indicated that combination of sub-lethal doses of LFchimera with ampicillin or tetracycline strongly reduced the concentration of the antibiotics to reach 95% growth inhibition. Furthermore, LFchimera were effective to inhibit the V. cholerae counts and damage due to this bacterium in a model mice. These data suggest that LFchimera and bLF are potential candidates to combat the V. cholerae and other multidrug resistant Vibrio species.

10.
Biomed Res Int ; 2015: 641392, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26090431

RESUMEN

Iron is the fourth most abundant element on Earth and the most abundant metal in the human body. This element is crucial for life because almost all organisms need iron for several biological activities. This is the case with pathogenic organisms, which are at the vanguard in the battle with the human host for iron. The latest regulates Fe concentration through several iron-containing proteins, such as transferrin. The transferrin receptor transports iron to each cell that needs it and maintains it away from pathogens. Parasites have developed several strategies to obtain iron as the expression of specific transferrin receptors localized on plasma membrane, internalized through endocytosis. Signal transduction pathways related to the activation of the receptor have functional importance in proliferation. The study of transferrin receptors and other proteins with action in the signaling networks is important because these proteins could be used as therapeutic targets due to their specificity or to differences with the human counterpart. In this work, we describe proteins that participate in signal transduction processes, especially those that involve transferrin endocytosis, and we compare these processes with those found in T. brucei, T. cruzi, Leishmania spp., and E. histolytica parasites.


Asunto(s)
Endocitosis/genética , Hierro/metabolismo , Parásitos/metabolismo , Transferrina/metabolismo , Animales , Antígenos CD/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Humanos , Parásitos/patogenicidad , Receptores de Transferrina/genética , Transducción de Señal/genética , Transferrina/genética
11.
Biochimie ; 107 Pt B: 223-34, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25223890

RESUMEN

Caveolin is the protein marker of caveola-mediated endocytosis. Previously, we demonstrated by immunoblotting and immunofluorescence that an anti-chick embryo caveolin-1 monoclonal antibody (mAb) recognizes a protein in amoeba extracts. Nevertheless, the caveolin-1 gene is absent in the Entamoeba histolytica genome database. In this work, the goal was to isolate, identify and characterize the protein that cross-reacts with chick embryo caveolin-1. We identified the protein using a proteomic approach, and the complete gene was cloned and sequenced. The identified protein, E. histolytica phosphatidylcholine transfer protein-like (EhPCTP-L), is a member of the StAR-related lipid transfer (START) protein superfamily. The human homolog binds and transfers phosphatidylcholine (PC) and phosphatidylethanolamine (PE) between model membranes in vitro; however, the physiological role of PCTP-L remains elusive. Studies in silico showed that EhPCTP-L has a central START domain and also contains a C-terminal intrinsically disordered region. The anti-rEhPCTP-L antibody demonstrated that EhPCTP-L is found in the plasma membrane and cytosol, which is in agreement with previous reports on the human counterpart. This result points to the plasma membrane as one possible target membrane for EhPCTP-L. Furthermore, assays using filipin and nystatin showed down regulation of EhPCTP-L, in an apparently cholesterol-independent way. Interestingly, EhPCTP-L binds primarily to anionic phospholipids phosphatidylserine (PS) and phosphatidic acid (PA), while its mammalian counterpart HsPCTP-L binds neutral phospholipids PC and PE. The present study provides information that helps reveal the possible function and regulation of PCTP-L expression in the primitive eukaryotic parasite E. histolytica.


Asunto(s)
Entamoeba histolytica/metabolismo , Proteínas Protozoarias/aislamiento & purificación , Proteínas Protozoarias/metabolismo , Acetilación , Secuencia de Aminoácidos , Animales , Caveolina 1/inmunología , Membrana Celular/metabolismo , Embrión de Pollo , Colesterol/metabolismo , Reacciones Cruzadas , Citoplasma/metabolismo , Entamoeba histolytica/efectos de los fármacos , Entamoeba histolytica/genética , Filipina/farmacología , Datos de Secuencia Molecular , Nistatina/farmacología , Fosfatidilcolinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/inmunología , Proteínas de Transferencia de Fosfolípidos/aislamiento & purificación , Proteínas de Transferencia de Fosfolípidos/metabolismo , Fosfoproteínas/química , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología
12.
Microbiologyopen ; 3(5): 711-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25065983

RESUMEN

Pasteurella multocida (Pm) is a gram-negative bacterium able to infect different animal species, including human beings. This bacterium causes economic losses to the livestock industry because of its high morbidity and mortality in animals. In this work, we report the characterization of outer membrane vesicles (OMVs) released into the culture medium by different Pm serogroups. Purified OMVs in the range of 50-300 nm were observed by electron microscopy. Serum obtained from chickens infected with Pm recognized several proteins from Pm OMVs. Additionally, rabbit antiserum directed against a secreted protease from Actinobacillus pleuropneumoniae recognized a similar protein in the Pm OVMs, suggesting that OMVs from these bacterial species contain common immunogenic proteins. OmpA, a multifunctional protein, was identified in OMVs from different Pm serogroups, and its concentration was twofold higher in OMVs from Pm serogroups B and D than in OMVs from other serogroups. Three outer membrane proteins were also identified: OmpH, OmpW, and transferrin-binding protein. Three bands of 65, 110, and 250 kDa with proteolytic activity were detected in Pm OMVs of serogroups A and E. Additionally, ß-lactamase activity was detected only in OMVs from Pm 12945 Amp(r) (serogroup A). Pm OMVs may be involved in different aspects of disease pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Pasteurella/microbiología , Infecciones por Pasteurella/veterinaria , Pasteurella multocida/metabolismo , Enfermedades de las Aves de Corral/microbiología , Vesículas Transportadoras/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Pollos , Humanos , Pasteurella multocida/genética , Vesículas Transportadoras/genética , Factores de Virulencia/genética
13.
Biometals ; 27(5): 969-80, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25053107

RESUMEN

Streptococcus pneumoniae (pneumococcus) is responsible for nearly one million child deaths annually. Pneumococcus causes infections such as pneumonia, otitis media, meningitis, and sepsis. The human immune system includes antibacterial peptides and proteins such as lactoferrin (LF), but its activity against pneumococcus is not fully understood. The aim of this work was to evaluate the bactericidal effect of bovine lactoferrin (bLF) and the synthetic LF-peptides lactoferricin (LFcin17-30), lactoferrampin (LFampin265-284), and LFchimera against S. pneumoniae planktonic cells. The mechanism of damage was also investigated, as well as the impact of these peptides on the transcription levels of genes known to encode important virulence factors. S. pneumoniae planktonic cells were treated with bLF, LFcin17-30, LFampin265-284 and LFchimera at different time points. The viability of treated planktonic cells was assessed by dilution and plating (in CFU/ml). The interaction between LF and LF-peptides coupled to fluorescein was visualized using a confocal microscope and flow cytometry, whereas the damage at structural levels was observed by electron microscopy. Damage to bacterial membranes was further evaluated by membrane permeabilization by use of propidium iodide and flow cytometry, and finally, the expression of pneumococcal genes was evaluated by qRT-PCR. bLF and LFchimera were the best bactericidal agents. bLF and peptides interacted with bacteria causing changes in the shape and size of the cell and membrane permeabilization. Moreover, the luxS gene was down-regulated in bacteria treated with LF. In conclusion, LF and LFchimera have a bactericidal effect, and LF down-regulates genes involved in the pathogenicity of pneumococcus, thus demonstrating potential as new agents for the treatment of pneumococcal infections.


Asunto(s)
Antibacterianos/farmacología , Lactoferrina/farmacología , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/genética , Animales , Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Bacterianas/genética , Liasas de Carbono-Azufre/genética , Bovinos , Niño , Expresión Génica/efectos de los fármacos , Genes Bacterianos/efectos de los fármacos , Humanos , Fragmentos de Péptidos/farmacología , Infecciones Neumocócicas/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Streptococcus pneumoniae/patogenicidad
14.
J Trop Med ; 2013: 890603, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23476670

RESUMEN

The standard reference for pathogenic and nonpathogenic amoebae is the human parasite Entamoeba histolytica; a direct correlation between virulence and protease expression has been demonstrated for this amoeba. Traditionally, proteases are considered virulence factors, including those that produce cytopathic effects in the host or that have been implicated in manipulating the immune response. Here, we expand the scope to other amoebae, including less-pathogenic Entamoeba species and highly pathogenic free-living amoebae. In this paper, proteases that affect mucin, extracellular matrix, immune system components, and diverse tissues and cells are included, based on studies in amoebic cultures and animal models. We also include proteases used by amoebae to degrade iron-containing proteins because iron scavenger capacity is currently considered a virulence factor for pathogens. In addition, proteases that have a role in adhesion and encystation, which are essential for establishing and transmitting infection, are discussed. The study of proteases and their specific inhibitors is relevant to the search for new therapeutic targets and to increase the power of drugs used to treat the diseases caused by these complex microorganisms.

15.
J Parasitol Res ; 2012: 748206, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792442

RESUMEN

Parasitic protozoa are among the most important pathogens worldwide. Diseases such as malaria, leishmaniasis, amoebiasis, giardiasis, trichomoniasis, and trypanosomiasis affect millions of people. Humans are constantly threatened by infections caused by these pathogens. Parasites engage a plethora of surface and secreted molecules to attach to and enter mammalian cells. The secretion of lytic enzymes by parasites into host organs mediates critical interactions because of the invasion and destruction of interstitial tissues, enabling parasite migration to other sites within the hosts. Extracellular matrix is a complex, cross-linked structure that holds cells together in an organized assembly and that forms the basement membrane lining (basal lamina). The extracellular matrix represents a major barrier to parasites. Therefore, the evolution of mechanisms for connective-tissue degradation may be of great importance for parasite survival. Recent advances have been achieved in our understanding of the biochemistry and molecular biology of proteases from parasitic protozoa. The focus of this paper is to discuss the role of protozoan parasitic proteases in the degradation of host ECM proteins and the participation of these molecules as virulence factors. We divide the paper into two sections, extracellular and intracellular protozoa.

16.
Microbiology (Reading) ; 157(Pt 1): 209-219, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20847004

RESUMEN

Transferrin (Tf) is a host glycoprotein capable of binding two ferric-iron ions to become holotransferrin (holoTf), which transports iron in to all cells. Entamoeba histolytica is a parasitic protozoan able to use holoTf as a sole iron source in vitro. The mechanism by which this parasite scavenges iron from holoTf is unknown. An E. histolytica holoTf-binding protein (EhTfbp) was purified by using an anti-human transferrin receptor (TfR) monoclonal antibody. EhTfbp was identified by MS/MS analysis and database searches as E. histolytica acetaldehyde/alcohol dehydrogenase-2 (EhADH2), an iron-dependent enzyme. Both EhTfbp and EhADH2 bound holoTf and were recognized by the anti-human TfR antibody, indicating that they correspond to the same protein. It was found that the amoebae internalized holoTf through clathrin-coated pits, suggesting that holoTf endocytosis could be important for the parasite during colonization and invasion of the intestinal mucosa and liver.


Asunto(s)
Alcohol Deshidrogenasa/metabolismo , Aldehído Oxidorreductasas/metabolismo , Clatrina/metabolismo , Endocitosis , Entamoeba histolytica/metabolismo , Interacciones Huésped-Patógeno , Transferrina/metabolismo , Alcohol Deshidrogenasa/aislamiento & purificación , Aldehído Oxidorreductasas/aislamiento & purificación , Vesículas Cubiertas por Clatrina/metabolismo , Humanos , Proteínas Protozoarias/aislamiento & purificación , Proteínas Protozoarias/metabolismo , Espectrometría de Masas en Tándem
17.
Biometals ; 23(3): 569-78, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20195887

RESUMEN

Increased prevalence of antibiotic-resistant bacteria has become a major threat to the health sector worldwide due to their virulence, limited therapeutic options and distribution in both hospital and community settings. Discovery and development of new agents to combat antibiotic-resistant bacteria is thus needed. This study therefore aimed to evaluate the ability of bovine lactoferrin (LF), peptides from two antimicrobial domains lactoferricin B (LFcin17-30) and lactoferrampin (LFampin265-284) and a chimeric construct (LFchimera) containing both peptides, as potential bactericidal agents against clinical isolates of antibiotic-resistant Staphylococcus aureus and Escherichia coli. Results in kinetics of growth show that LF chimera and peptides inhibited the growth of both bacterial species. By confocal microscopy and flow cytometry it was observed that LF and FITC-labeled peptides are able to interact with these bacteria and cause membrane permeabilization, as monitored by propidium iodide staining, these effects were decreased by preincubation with lipopolysaccharide in E. coli. By electron microscopy, a clear cellular damage was observed in bacteria after treatments with LFchimera and peptides, suggesting that interaction and membrane disruption are probably involved as a mechanism of action. In conclusion, results show that LFchimera, LF and peptides have potential as bactericidal agents in the antibiotic-resistant strains of S. aureus and E. coli and also the work strongly suggest that LFcin17-30 and LFampin265-284 acts synergistically with antibiotics against multidrug resistant EPEC and MRSA in vitro.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Escherichia coli/efectos de los fármacos , Lactoferrina/química , Lactoferrina/farmacología , Péptidos/farmacología , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/química , Bovinos , Pruebas de Sensibilidad Microbiana , Péptidos/química , Relación Estructura-Actividad
18.
Infect Genet Evol ; 9(6): 1038-50, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19539057

RESUMEN

Iron is essential for nearly all organisms; in mammals, it is part of proteins such as haemoglobin, and it is captured by transferrin and lactoferrin. Transferrin is present in serum, and lactoferrin is secreted by the mucosa and by neutrophils at infection sites, as a host iron-withholding response, sequestering iron away from invading microorganisms. Additionally, all cells contain ferritin, which sequesters iron when its intracellular levels are increased, detoxifying and preventing damage. Liver ferritin contains 50% of iron corporal reserves. During evolution, pathogens have evolved diverse strategies to obtain iron from their hosts in order to survive. The protozoan Entamoeba histolytica invades the intestinal mucosa, causing dysentery, and the trophozoites often travel to the liver producing hepatic abscesses; thus, intestine and liver proteins could be important iron supplies for E. histolytica. We found that E. histolytica trophozoites can grow in both ferrous and ferric iron, and that they can use haemoglobin, holo-transferrin, holo-lactoferrin, and ferritin as in vitro iron sources. These proteins supported the amoeba growth throughout consecutive passages, similarly to ferric citrate. By confocal microscopy and immunoblotting, iron-binding proteins were observed specifically bound to the amoeba surface, and they were endocytosed, trafficked through the endosomal/lysosomal route, and degraded by neutral and acidic cysteine-proteases. Transferrin and ferritin were mainly internalized through clathrin-coated vesicles, and holo-lactoferrin was mainly internalized by caveola-like structures. In contrast, apo-lactoferrin bound to membrane lipids and cholesterol, inducing cell death. The results suggest that in vivo trophozoites secrete products that can destroy enterocytes, erythrocytes, and hepatocytes, releasing transferrin, haemoglobin, ferritin, and other iron-containing proteins, which, together with lactoferrin derived from neutrophils and acinar cells, could be used as abundant iron supplies by amoebas.


Asunto(s)
Endocitosis , Entamoeba histolytica/fisiología , Entamebiasis/metabolismo , Entamebiasis/parasitología , Proteínas de Unión a Hierro/metabolismo , Trofozoítos/fisiología , Animales , Bacterias/metabolismo , Bacterias/patogenicidad , Entamoeba histolytica/citología , Entamoeba histolytica/patogenicidad , Entamebiasis/microbiología , Interacciones Huésped-Patógeno , Humanos , Hierro/metabolismo , Microscopía Confocal , Trofozoítos/citología , Virulencia
19.
Biochimie ; 91(1): 133-40, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18625283

RESUMEN

Infections caused by Vibrio parahaemolyticus, an halophilic member of the genus Vibrio, have increased globally in the last 5 years. Diarrhea caused by V. parahaemolyticus results from eating raw or undercooked seafood. The aim of this work was to investigate whether lactoferrin and some lactoferrin-peptides have bactericidal activity against Vibrio parahaemolyticus ATCC 17802, the pandemic strain O3:K6, and the multidrug resistant isolate 727, as well as against Vibrio cholerae strains O1 and non-O1. Whereas both peptides lactoferricin (17-30) and lactoferrampin (265-284) did not have bactericidal activity, 40 microM of lactoferrin chimera (a fusion of the two peptides) inhibited the growth of all Vibrio tested to the same extent as the antibiotic gentamicin. The cidal effect of LFchimera showed a clear concentration response in contrast to bovine lactoferrin which showed higher inhibition at 10 microM than at 40 microM. FITC-labeled LFchimera bound to the bacterial membranes. Moreover LFchimera permeabilized bacterial cells and membranes were seriously damaged. Finally, in experiments with the multidrug resistant isolate 727, sub-lethal doses of LFchimera strongly reduced the concentrations of ampicillin, gentamicin or kanamicin needed to reach more than 95% growth inhibition, suggesting synergistic effects. These data indicate that LFchimera is a potential candidate to combat the multidrug resistant pathogenic Vibrio species.


Asunto(s)
Lactoferrina/farmacología , Lactoglobulinas/farmacología , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Vibrio parahaemolyticus/efectos de los fármacos , Ampicilina/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Citometría de Flujo , Gentamicinas/farmacología , Lactoferrina/genética , Lactoglobulinas/genética , Pruebas de Sensibilidad Microbiana , Microscopía Confocal , Microscopía Electrónica de Transmisión , Fragmentos de Péptidos/genética , Proteínas Recombinantes de Fusión/genética , Vibrio cholerae/efectos de los fármacos , Vibrio cholerae/ultraestructura , Vibrio parahaemolyticus/ultraestructura
20.
Biochem Cell Biol ; 84(3): 327-36, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16936803

RESUMEN

Lactoferrin (Lf), in its iron-free form, has been shown to inhibit the growth of pathogenic microorganisms. In the light of new agents to control amoebiasis, the microbicidal activity of human and bovine Lf and bovine lactoferricin (bLfcin, fragment 4-14), and of each combined with metronidazole, the main drug used in amoebiasis, was evaluated in trophozoites of Entamoeba histolytica. Both lactoferrins and bLfcin were able to kill amoebas in a concentration-dependent manner. This killing effect was modulated according to the culture age, pH, and temperature. Parasites obtained from the stationary phase were more susceptible to Lf than those from the early exponential phase. The effect of Lf and its derived peptide, bLfcin, was prevented by both Fe2+ and Fe3+. However, the divalent cations Mg2+ and Ca2+ prevented the killing effect of Lf but not of bLfcin. A synergistic amoebicidal effect was found between metronidazole and human Lf, bovine Lf, or bLfcin. These data suggest that Lf and bLfcin might be used in amoebiasis if they are administered with a low dose of metronidazole to diminish the toxicity of this drug. Thus, Lf and bLfcin are therapeutically potential candidates for use as antiamoebics in patients.


Asunto(s)
Antibacterianos/farmacología , Entamoeba histolytica/efectos de los fármacos , Lactoferrina/farmacología , Metronidazol/farmacología , Animales , Apoproteínas/metabolismo , Cationes/metabolismo , Bovinos , Muerte Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Sinergismo Farmacológico , Entamoeba histolytica/citología , Entamoeba histolytica/crecimiento & desarrollo , Humanos , Concentración de Iones de Hidrógeno , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...