Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
BMC Complement Altern Med ; 16(1): 403, 2016 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-27770779

RESUMEN

BACKGROUND: Eugenia spp. are used in popular medicine in the treatment of pain, diabetes, intestinal disorders and cough. The aim of the work is to evaluate, ex vivo and in vivo, the anti-inflammatory activity of the hydroethanolic extracts of the leaves of Eugenia aurata (EA) and Eugenia punicifolia HBK (EP) upon neutrophils. METHODS: Ex vivo, isolated human neutrophils were sensitized by Eugenia extracts (0.1-1000 µg/mL) and stimulated by PMA. In these conditions, different neutrophil activities related to inflammatory process were measured: adhesion, degranulation and NET release. Neutrophil viability and tumor line cells were monitored. In vivo, neutrophil influx was evaluated by peritonitis model performed in mice pretreated with different concentrations of Eugenia extracts. Phytochemical profile was assessed by mass spectrometry. RESULTS: Ex vivo, EA and EP (1000 µg/mL) reduced cell adhesion and degranulation, respectively. NET release was inhibited by EA and EP. Anti-inflammatory activities occurred in the absence of cytotoxicity. In vivo, both EA as EP inhibited neutrophil migration. The phytochemical profile revealed that EA contains myricitrin, rutin, quinic acid and quercetin derivatives. EP presents gallic acid, quercetin derivatives, syringic acid, ellagic acid, monogalloyl-glucose, glycosyringic acid, mudanoside B, HHDP glucose isomer and digalloylglucose isomer. EA and EP inhibit neutrophil migration by different pathways. CONCLUSION: Different chemical compositions may explain the anti-inflammatory effects described herein for EA and EP. Both extracts inhibit NET release but only EA reduces cell adhesion whereas EP decreases elastase secretion. This work contributes to the elucidation of cellular mechanisms related to the anti-inflammatory activity for leaves of E. aurata and E. punicifolia HBK.


Asunto(s)
Antiinflamatorios/farmacología , Adhesión Celular/efectos de los fármacos , Degranulación de la Célula/efectos de los fármacos , Eugenia/química , Trampas Extracelulares/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Antiinflamatorios/química , Supervivencia Celular/efectos de los fármacos , Inflamación/metabolismo , Masculino , Ratones , Extractos Vegetales/química
2.
Artículo en Inglés | MEDLINE | ID: mdl-38652860

RESUMEN

Phototherapies are promising for noninvasive treatment of aggressive tumors, especially when combining heat induction and oxidative processes. Herein, we show enhanced phototoxicity of gold shell-isolated nanorods conjugated with toluidine blue-O (AuSHINRs@TBO) against human colorectal tumor cells (Caco-2) with synergic effects of photothermal (PTT) and photodynamic therapies (PDT). Mitochondrial metabolic activity tests (MTT) performed on Caco-2 cell cultures indicated a photothermal effect from AuSHINRs owing to enhanced light absorption from the localized surface plasmon resonance (LSPR). The phototoxicity against Caco-2 cells was further increased with AuSHINRs@TBO where oxidative processes, such as hydroperoxidation, were also present, leading to a cell viability reduction from 85.5 to 39.0%. The molecular-level mechanisms responsible for these effects were investigated on bioinspired tumor membranes using Langmuir monolayers of Caco-2 lipid extract. Polarization-modulation infrared reflection-absorption spectroscopy (PM-IRRAS) revealed that the AuSHINRs@TBO incorporation is due to attractive electrostatic interactions with negatively charged groups of the Caco-2 lipid extract, resulting in the expansion of surface pressure isotherms. Upon irradiation, Caco-2 lipid extract monolayers containing AuSHINRs@TBO (1:1 v/v) exhibited ca. 1.0% increase in surface area. This is attributed to the generation of reactive oxygen species (ROS) and their interaction with Caco-2 lipid extract monolayers, leading to hydroperoxide formation. The oxidative effects are facilitated by AuSHINRs@TBO penetration into the polar groups of the extract, allowing oxidative reactions with carbon chain unsaturations. These mechanisms are consistent with findings from confocal fluorescence microscopy, where the Caco-2 plasma membrane was the primary site of the cell death induction process.

3.
Int J Biol Macromol ; 280(Pt 1): 135387, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39260645

RESUMEN

The Human Respiratory Syncytial Virus (RSV) is the leading cause of acute respiratory infections in children. Currently, no safe, effective, or feasible option for pharmacological management of RSV exists. Hence, plant-derived natural compounds have been explored as promising antiviral agents. Mangifera indica is a globally distributed plant with reported anti-inflammatory, cardioprotective, and antiviral activities. Our study investigated the antiviral potential of a novel pectin from M. indica peels (PMi) and its chemically sulfated derivative (PSMi) against RSV in HEp-2 cells. The compounds were characterized using Fourier-transform infrared spectroscopy and nuclear magnetic resonance (NMR). NMR analysis revealed the presence of ester and carboxylic acid groups in PMi, and sulfation resulted in a sulfation degree of 0.5. PMi and PSMi showed no cytotoxic effects even at concentrations as high as 2000 µg/mL. PSMi completely inhibited RSV infectivity (100-1.56 µg/mL, 50 % inhibitory concentration of viral infectivity = 0.77 ± 0.11 µg/mL). The mechanism of action was investigated using the 50 % tissue culture infectious dose assay. PSMi displayed virucidal activity at concentrations from 100 to 6.25 µg/mL, and a significant reduction in viral infection was observed at all treatment times. Overall, PSMi is antiviral, cell-safe, and exhibits promising potential as an RSV treatment.

4.
BMC Complement Med Ther ; 23(1): 356, 2023 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-37805488

RESUMEN

BACKGROUND: Carboxymethylated Lasiodiplodan (LaEPS-C), Lasiodiplodia theobromae ß-glucan exopolysaccharide derivative, has a well-known range of biological activities. Compared to LaEPS-C, its fractions, Linear (LLaEPS-C) and Branched (BLaEPS-C), have biological potentialities scarcely described in the literature. So, in this study, we investigate the immunomodulatory, antiviral, antiproliferative, and anticoagulant activities of LLaEPS-C and BLaEPS-C and compare them to the LaEPS-C. METHODS: LaEPS was obtained from L. theobromae MMBJ. After carboxymethylation, LaEPS-C structural characteristics were confirmed by Elementary Composition Analysis by Energy Dispersive X-Ray Detector (EDS), Fourier Transform Infrared (FTIR), and Nuclear Magnetic Resonance (NMR). The immunomodulatory activity on cytokine secretion was evaluated in human monocyte-derived macrophage cultures. The antiviral activity was evaluated by Hep-2 cell viability in the presence or absence of hRSV (human respiratory syncytial virus). In vitro antiproliferative activity was tested by sulforhodamine B assay. The anticoagulant activity was determined by APTT (Activated Partial Thromboplastin Time) and PT (Prothrombin Time). RESULTS: LaEPS-C showed low macrophage cell viability only at 100 µg/mL (52.84 ± 24.06, 48 h), and LLaEPS-C presented no effect. Conversely, BLaEPS-C showed cytotoxicity from 25 to 100 µg/mL (44.36 ± 20.16, 40.64 ± 25.55, 33.87 ± 25.16; 48 h). LaEPS-C and LLaEPS-C showed anti-inflammatory activity. LaEPS-C presented this at 100 µg/mL (36.75 ± 5.53, 48 h) for IL-10, and LLaEPS-C reduces TNF-α cytokine productions at 100 µg/mL (18.27 ± 5.80, 48 h). LLaEPS-C showed an anti-hRSV activity (0.7 µg/ml) plus a low cytotoxic activity for Hep-2 cells (1.4 µg/ml). LaEPS-C presented an antiproliferative activity for NCI-ADR/RES (GI50 65.3 µg/mL). A better PT was achieved for LLaEPS-C at 5.0 µg/mL (11.85 ± 0.87s). CONCLUSIONS: These findings demonstrated that carboxymethylation effectively improves the biological potential of the LaEPS-C and their fractions. From those polysaccharides tested, LLaEPS provided the best results with low toxicity for anti-inflammatory, antiviral, and anticoagulant activities.


Asunto(s)
Citocinas , Polisacáridos , Humanos , Polisacáridos/farmacología , Polisacáridos/química , Antiinflamatorios/farmacología , Anticoagulantes/farmacología , Antivirales/farmacología
5.
Int Immunopharmacol ; 106: 108573, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35183035

RESUMEN

Human respiratory syncytial virus (hRSV) is an infectious agent in infants and young children which there are no vaccines or drugs for treatment. Neutrophils are recruited for airway, where they are stimulated by hRSV to release large amounts of neutrophil extracellular traps (NETs). NETs are compound by DNA and proteins, including microbicidal enzymes. They constitute a large part of the mucus accumulated in the lung of patients, compromising their breathing capacity. In contrast, NETs can capture/inactivate hRSV, but the molecules responsible for this effect are unknown. OBJECTIVES: We selected microbicidal NET enzymes (elastase, myeloperoxidase, cathepsin-G, and proteinase-3) to assess their anti-hRSV role. METHODS AND RESULTS: Through in vitro assays using HEp-2 cells, we observed that elastase, proteinase-3, and cathepsin-G, but not myeloperoxidase, showed virucidal effects even at non-cytotoxic concentrations. Elastase and proteinase-3, but not cathepsin-G, cleaved viral F-protein, which is responsible for viral adhesion and fusion with the target cells. Molecular docking analysis indicated the interaction of these macromolecules in the antigenic regions of F-protein through the active regions of the enzymes. CONCLUSIONS: Serine proteases from NETs interact and inactive hRSV. These results contribute to the understanding the role of NETs in hRSV infection and to designing treatment strategies for the inflammatory process during respiratory infections.


Asunto(s)
Trampas Extracelulares , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Serina Proteasas , Trampas Extracelulares/enzimología , Humanos , Simulación del Acoplamiento Molecular , Infecciones por Virus Sincitial Respiratorio/metabolismo , Serina Proteasas/metabolismo
6.
BMC Chem ; 14(1): 34, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32355912

RESUMEN

Eugenia punicifolia (Kunth) D. C. (Myrtaceae) has been showing interesting biological activities in the literature which was correlated to its phenolic compounds. In the sense of a better recovering of phenolics with the best antioxidant and antiproliferative activities, an extraction, based on multivariate analytical approach, was developed from E. punicifolia leaves. The different extractor solvents (ethanol, methanol and water) and their binary and ternary combinations were evaluated using a simplex-centroid mixture design and surface response methodology. The optimized crude extracts were investigated for phenol and flavonoid content and compared to their antioxidant (EC50) and antiproliferative properties against HEp-2 (cell line derived from the oropharyngeal carcinoma) and mononuclear viability cells. Ethanolic extracts showed the best phenolic content with the highest antioxidant activity and moderated activity antiproliferative to HEp-2. ESI-QTOF-MS revealed the presence of quercetin and myricetin derivatives, which was correlated to activities tested. Then, simplex-centroid design allowed us to correlate the Eugenia punicifolia biological activities with the extracts obtained from solvent different polarity mixtures.

7.
Virus Res ; 276: 197805, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31712123

RESUMEN

Human respiratory syncytial virus (hRSV) is one of the main etiological agents of diseases of the lower respiratory tract and is often responsible for the hospitalization of children and the elderly. To date, treatments are only palliative and there is no vaccine available. Natural products show exceptional structural diversity and they have played a vital role in drug research. Several investigations focused on applied structural modification of natural products to improved metabolic stability, solubility and biological actions them. Quercetin is a flavonoid that presents several biological activities, including anti-hRSV role. Some works criticize the pharmacological use of Quercetin because it has low solubility and low specificity. In this sense, we acetylated Quercetin structure and we used in vitro and in silico assays to compare anti-hRSV function between Quercetin (Q0) and its derivative molecule (Q1). Q1 shows lower cytotoxic effect than Q0 on HEp-2 cells. In addition, Q1 was more efficient than Q0 to protect HEp-2 cells infected with different multiplicity of infection (0.1-1 MOI). The virucidal effects of Q0 and Q1 suggest interaction between these molecules and viral particle. Dynamic molecular results suggest that Q0 and Q1 may interact with F-protein on hRSV surface in an important region to adhesion and viral infection. Q1 interaction with F-protein showed ΔG= -14.22 kcal/mol and it was more stable than Q0. Additional, MTT and plate assays confirmed that virucidal Q1 effects occurs during adhesion step of cycle hRSV replication. In conclusion, acetylation improves anti-hRSV Quercetin effects because Quercetin pentaacetate could interact with F-protein with lower binding energy and better stability to block viral adhesion. These results show alternative anti-hRSV strategy and contribute to drug discovery and development.


Asunto(s)
Antivirales/farmacología , Células Epiteliales/efectos de los fármacos , Quercetina/análogos & derivados , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Acoplamiento Viral/efectos de los fármacos , Acetilación , Línea Celular , Células Epiteliales/virología , Humanos , Simulación de Dinámica Molecular , Quercetina/farmacología , Virus Sincitial Respiratorio Humano/fisiología , Proteínas Virales de Fusión/metabolismo , Replicación Viral/efectos de los fármacos
8.
Biochem Biophys Res Commun ; 376(4): 764-9, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18812167

RESUMEN

The macrophage-derived neutrophil chemotactic factor (MNCF) is an alpha-galactoside-binding lectin, known to induce dexamethasone-insensitive neutrophil recruitment. We further characterized MNCF effects on neutrophils and showed that it shares with TNF-alpha the ability to delay apoptosis and to trigger degranulation. MNCF and TNF-alpha effects show similar kinetics and involve Src kinases and MAPKinases dependent pathways. They were, however, clearly distinguished, since the soluble TNF-receptor etanercept prevented TNF but not MNCF effects, while melibiose disaccharide inhibited MNCF but not TNF effects. Absorption of MNCF on detoxi-gel did not alter its properties, precluding an LPS contamination effect. By contrast, galectin-3 required LPS to activate neutrophils. Specific antibodies allowed to further demonstrate that MNCF and galectin-3 are two distinct molecules. Finally, MNCF- and IL-8-induced neutrophil activation differed by their kinetic and sensitivity to pertussis toxin. In conclusion, MNCF is a distinct neutrophil agonist, with pro-inflammatory activities involving its carbohydrate recognition domain.


Asunto(s)
Interleucina-8/inmunología , Lectinas/inmunología , Neutrófilos/inmunología , Animales , Degranulación de la Célula , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Galectina 3/inmunología , Galectina 3/farmacología , Humanos , Interleucina-8/farmacología , Lectinas/farmacología , Ratones , Neutrófilos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología
9.
Virus Res ; 251: 68-77, 2018 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-29621602

RESUMEN

Human respiratory syncytial virus (hRSV) is one of the main etiological agents of diseases of the lower respiratory tract, and is often responsible for the hospitalization of children and the elderly. To date, treatments are only palliative and there is no vaccine available. The airways of patients infected with hRSV exhibit intense neutrophil infiltration, which is responsible for the release of neutrophil extracellular traps (NETs). These are extracellular structures consisting of DNA associated with intracellular proteins, and are efficient in capturing and eliminating various microorganisms, including some viruses. hRSV induces the release of NETs into the lung tissue of infected individuals; however, the pathophysiological consequences of this event have not been elucidated. The objective of this study was to utilize in vitro and in silico assays to investigate the impact of NETs on hRSV infection. NETs, generated by neutrophils stimulated with phorbol myristate acetate (PMA), displayed long fragments of DNA and an electrophoretic profile suggestive of the presence of proteins that are classically associated with these structures (elastase, cathepsin G, myeloperoxidase, and histones). The presence of NETs (>2 µg/ml) in HEp-2 cell culture medium resulted in cellular cytotoxicity of less than 50%. Pre-incubation (1 h) of viral particles (multiplicity of infection (MOI) values of 0.1, 0.5, and 1.0) with NETs (2-32 µg/ml) resulted in cellular protection from virus-induced death of HEp-2 cells. Concurrently, there was a reduction in the formation of syncytia, which is related to decreased viral spread in infected tissue. Results from western blotting and molecular docking, suggest interactions between F protein of the hRSV viral envelope and BPI (bactericidal permeability-increasing protein), a microbicidal member of NETs. Interactions occurred at sites important for the neutralization and coordination of the hRSV infection/replication process. Our results showed that the presence of NETs decreases hRSV-induced cellular damage, possibly by directly affecting viral particle capture and/or interfering with the fusion activity of the F protein. These findings broaden the understanding of the role of NETs during hRSV infection.


Asunto(s)
Trampas Extracelulares/metabolismo , Interacciones Huésped-Patógeno , Neutrófilos/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/metabolismo , Células Cultivadas , ADN/análisis , Proteínas de Unión al ADN/análisis , Células Epiteliales/virología , Trampas Extracelulares/química , Humanos
10.
Int J Biol Macromol ; 95: 63-71, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27851930

RESUMEN

hRSV is the major causative agent of acute respiratory infections. Among its eleven proteins, M2-1 is a transcription antiterminator, making it an interesting target for antivirals. Quercetin is a flavonol which inhibits some virus infectivity and replication. In the present work, the M2-1 gene was cloned, expressed and the protein was purified. Thermal stability and secondary structure were analyzed by circular dichroism and the interaction with Quercetin was evaluated by fluorescence spectroscopy. Molecular docking experiments were performed to understand this mechanism of interaction. The purified protein is mainly composed of α-helix, with a melting temperature of 328.6K (≈55°C). M2-1 titration with Quercetin showed it interacts with two sites, one with a strong constant association K1 (site 1≈1.5×106M-1) by electrostatic interactions, and another with a weak constant association K2 (site 2≈1.1×105M-1) by a hydrophobic interaction. Ligand's docking shows it interacts with the N-terminus face in a more polar pocket and, between the domains of oligomerization and RNA and P protein interaction, in a more hydrophobic pocket, as predicted by experimental data. Therefore, we postulated this ligand could be interacting with important domains of the protein, avoiding viral replication and budding.


Asunto(s)
Fenómenos Biofísicos , Simulación del Acoplamiento Molecular , Quercetina/metabolismo , Virus Sincitial Respiratorio Humano , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Biología Computacional , Unión Proteica , Conformación Proteica , Proteínas Virales/química
11.
PLoS One ; 10(10): e0139569, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26431537

RESUMEN

Neutrophils release fibrous traps of DNA, histones, and granule proteins known as neutrophil extracellular traps (NETs), which contribute to microbicidal killing and have been implicated in autoimmunity. The role of NET formation in the host response to nonbacterial pathogens is not well-understood. In this study, we investigated the release of NETs by human neutrophils upon their interaction with Trypanosoma cruzi (Y strain) parasites. Our results showed that human neutrophils stimulated by T. cruzi generate NETs composed of DNA, histones, and elastase. The release occurred in a dose-, time-, and reactive oxygen species-dependent manner to decrease trypomastigote and increase amastigote numbers of the parasites without affecting their viability. NET release was decreased upon blocking with antibodies against Toll-like receptors 2 and 4. In addition, living parasites were not mandatory in the release of NETs induced by T. cruzi, as the same results were obtained when molecules from its soluble extract were tested. Our results increase the understanding of the stimulation of NETs by parasites, particularly T. cruzi. We suggest that contact of T. cruzi with NETs during Chagas's disease can limit infection by affecting the infectivity/pathogenicity of the parasite.


Asunto(s)
Trampas Extracelulares/metabolismo , Receptores Toll-Like/metabolismo , Trypanosoma cruzi/metabolismo , Animales , Antígenos/metabolismo , Línea Celular , Enfermedad de Chagas/metabolismo , Histonas/metabolismo , Humanos , Macaca mulatta , Activación Neutrófila/fisiología , Neutrófilos/metabolismo , Elastasa Pancreática/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
PLoS One ; 9(11): e112474, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25392933

RESUMEN

Dengue virus (DENV) is an enveloped RNA virus that is mosquito-transmitted and can infect a variety of immune and non-immune cells. Response to infection ranges from asymptomatic disease to a severe disorder known as dengue hemorrhagic fever. Despite efforts to control the disease, there are no effective treatments or vaccines. In our search for new antiviral compounds to combat infection by dengue virus type 1 (DENV-1), we investigated the role of galectin-1, a widely-expressed mammalian lectin with functions in cell-pathogen interactions and immunoregulatory properties. We found that DENV-1 infection of cells in vitro exhibited caused decreased expression of Gal-1 in several different human cell lines, suggesting that loss of Gal-1 is associated with virus production. In test of this hypothesis we found that exogenous addition of human recombinant Gal-1 (hrGal-1) inhibits the virus production in the three different cell types. This inhibitory effect was dependent on hrGal-1 dimerization and required its carbohydrate recognition domain. Importantly, the inhibition was specific for hrGal-1, since no effect was observed using recombinant human galectin-3. Interestingly, we found that hrGal-1 directly binds to dengue virus and acts, at least in part, during the early stages of DENV-1 infection, by inhibiting viral adsorption and its internalization to target cells. To test the in vivo role of Gal-1 in DENV infection, Gal-1-deficient-mice were used to demonstrate that the expression of endogenous Galectin-1 contributes to resistance of macrophages to in vitro-infection with DENV-1 and it is also important to physiological susceptibility of mice to in vivo infection with DENV-1. These results provide novel insights into the functions of Gal-1 in resistance to DENV infection and suggest that Gal-1 should be explored as a potential antiviral compound.


Asunto(s)
Virus del Dengue/clasificación , Dengue/metabolismo , Galectina 1/metabolismo , Adsorción , Animales , Antivirales/química , Carbohidratos/química , Muerte Celular , Línea Celular , Linaje de la Célula , Supervivencia Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Galectina 3/metabolismo , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Recombinantes/metabolismo
13.
PLoS One ; 6(10): e26004, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22031821

RESUMEN

Galectin-3 (Gal 3) is a glycan-binding protein that can be secreted by activated macrophages and mast cells at inflammation sites and plays an important role in inflammatory diseases caused by Bacteria and their products, such as lipopolysaccharides (LPS). Although it is well established that Gal 3 can interact with LPS, the pathophysiological importance of LPS/Gal 3 interactions is not fully understood. Data presented herein demonstrate for the first time that the interaction of Gal 3, either via its carbohydrate binding C-terminal domain or via its N-terminal part, with LPS from different bacterial strains, enhances the LPS-mediated neutrophil activation in vitro. Gal 3 allowed low LPS concentrations (1 µg/mL without serum, 1 ng/mL with serum) to upregulate CD11b expression and reactive oxygen species (ROS) generation on human neutrophils in vitro and drastically enhanced the binding efficiency of LPS to the neutrophil surface. These effects required LPS preincubation with Gal 3, before neutrophil stimulation and involved specific Gal 3/LPS interaction. A C-terminal Gal-3 fragment, which retains the lectin domain but lacks the N-terminal part, was still able to bind both to Escherichia coli LPS and to neutrophils, but had lost the ability to enhance neutrophil response to LPS. This result emphasizes the importance of an N-terminus-mediated Gal 3 oligomerization induced by its interaction with LPS. Finally we demonstrated that Balb/C mice were more susceptible to LPS-mediated shock when LPS was pretreated with Gal 3. Altogether, these results suggest that multimeric interactions between Gal 3 oligomers and LPS potentiate its pro-inflammatory effects on neutrophils.


Asunto(s)
Galectina 3/metabolismo , Lipopolisacáridos/toxicidad , Activación Neutrófila/efectos de los fármacos , Animales , Antígeno CD11b/metabolismo , Células Cultivadas , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Multimerización de Proteína/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
14.
Immunol Lett ; 123(1): 14-20, 2009 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-19428547

RESUMEN

The D-mannose binding lectin ArtinM from Artocarpus integrifolia, previously known as KM+ and artocarpin, is considered a stimulant of Th1-type immunity, which is able to confer resistance to some intracellular pathogens. In addition, ArtinM induces neutrophil migration by haptotaxis through simultaneous interactions of its carbohydrate recognition domains (CRDs) with glycans expressed on the extracellular matrix and the neutrophil surface. In the present study, we have expanded the characterization of ArtinM as a neutrophil activator. Exposure of neutrophils to ArtinM for 15 min resulted in tyrosine phosphorylation of intracellular proteins, a process that was selectively inhibited by d-mannose or mannotriose. Shortly after stimulation, neutrophils secreted high levels of LTB(4) and underwent shedding of L-selectin from their surface. Exposure to ArtinM enhanced neutrophil functions, such as respiratory burst and zymozan and Listeria monocytogenes phagocytosis. In addition, ArtinM-stimulated neutrophils displayed increased CXCL-8 secretion and TLR2 gene transcription. These results demonstrate that ArtinM is able to induce potent neutrophil activation, a feature that should be strongly considered in the assessment of the lectin capacity to confer resistance against infections.


Asunto(s)
Movimiento Celular , Lectinas de Unión a Manosa/farmacología , Activación Neutrófila , Neutrófilos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Lectinas de Plantas/farmacología , Humanos , Interleucina-8/biosíntesis , Interleucina-8/inmunología , Interleucina-8/farmacología , Selectina L/efectos de los fármacos , Selectina L/inmunología , Selectina L/metabolismo , Leucotrieno B4/biosíntesis , Leucotrieno B4/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Receptor Toll-Like 2/efectos de los fármacos , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA