Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Molecules ; 29(3)2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38338484

RESUMEN

The molecular events of protein misfolding and self-aggregation of tau and amylin are associated with the progression of Alzheimer's and diabetes, respectively. Recent studies suggest that tau and amylin can form hetero-tau-amylin oligomers. Those hetero-oligomers are more neurotoxic than homo-tau oligomers. So far, the detailed interactions between the hetero-oligomers and the neuronal membrane are unknown. Using multiscale MD simulations, the lipid binding and protein folding behaviors of hetero-oligomers on asymmetric lipid nanodomains or raft membranes were examined. Our raft membranes contain phase-separated phosphatidylcholine (PC), cholesterol, and anionic phosphatidylserine (PS) or ganglioside (GM1) in one leaflet of the lipid bilayer. The hetero-oligomers bound more strongly to the PS and GM1 than other lipids via the hydrophobic and hydrophilic interactions, respectively, in the raft membranes. The hetero-tetramer disrupted the acyl chain orders of both PC and PS in the PS-containing raft membrane, but only the GM1 in the GM1-containing raft membrane as effectively as the homo-tau-tetramer. We discovered that the alpha-helical content in the heterodimer was greater than the sum of alpha-helical contents from isolated tau and amylin monomers on both raft membranes, indicative of a synergetic effect of tau-amylin interactions in surface-induced protein folding. Our results provide new molecular insights into understanding the cross-talk between Alzheimer's and diabetes.


Asunto(s)
Enfermedad de Alzheimer , Diabetes Mellitus , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Péptidos beta-Amiloides/metabolismo , Gangliósido G(M1)/química , Membrana Dobles de Lípidos/química , Fosfatidilcolinas
2.
ACS Chem Neurosci ; 14(23): 4199-4207, 2023 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-37971427

RESUMEN

Alzheimer's disease is a progressive neurodegenerative disease and is the most common cause of dementia. It has been reported that the assembly of amyloid ß-protein (Aß) on the cell membrane is induced by the interaction of the Aß monomer with gangliosides such as GM1. The ganglioside-bound Aß (GAß) complex acts as a seed to promote the toxic assembly of the Aß fibrils. In a previous study, we found that a GM1 cluster-binding peptide (GCBP) specifically recognizes Aß-sensitive ganglioside nanoclusters and inhibits the assembly of Aß on a GM1-containing lipid membrane. In this study, cysteine-substituted double mutants of GCBP were designed and cyclized by intramolecular disulfide bond formation. Affinity assays indicated that one of the cyclic peptides had a higher affinity to a GM1-containing membrane compared to that of GCBP. Furthermore, surface topography analysis indicated that this peptide recognizes GM1 nanoclusters on the lipid membrane. An evaluation of the inhibitory kinetics indicated that the cyclic peptide could inhibit the formation of Aß fibrils with an IC50 value of 1.2 fM, which is 10,000-fold higher than that of GCBP. The cyclic peptide was also shown to have a clearance effect on Aß fibrils deposited on the lipid membrane and suppressed the formation of toxic Aß assemblies. Our results indicate that the cyclic peptide that binds to the Aß-sensitive ganglioside nanocluster is a potential novel inhibitor of ganglioside-induced Aß assembly.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Humanos , Péptidos beta-Amiloides/metabolismo , Gangliósido G(M1)/química , Ciclización , Enfermedad de Alzheimer/metabolismo , Gangliósidos/metabolismo , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/metabolismo
3.
Biophys Chem ; 301: 107091, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37549471

RESUMEN

Alzheimer's disease is a progressive degenerative condition that mainly affects cognition and memory. Recently, distinct clinical and neuropathological phenotypes have been identified in AD. Studies revealed that structural variation in Aß fibrillar aggregates correlates with distinct disease phenotypes. Moreover, environmental surroundings, including other biomolecules such as proteins and lipids, have been shown to interact and modulate Aß aggregation. Model membranes containing ganglioside (GM1) clusters are specifically known to promote Aß fibrillogenesis. This study unravels the modulatory effect of non-micellar GM1, a glycosphingolipid frequently released from the damaged neuronal membranes, on Aß42 amyloid fibril formation. Using far-UV circular dichroism experiments, we observed a change in the peptide secondary structure from random-coil to ß-turn structures with subsequent generation of predominantly ß-sheet-rich species upon interaction with GM1. Thioflavin-T (ThT) fluorescence assays further indicated that GM1 likely interacts with an amyloidogenic Aß42 intermediate species leading to a possible formation of GM1-modified Aß42 fibril. Statistically, no significant difference in toxicity to RA-differentiated SH-SY5Y cells was observed between Aß42 fibrils and GM1-tweaked Aß42 aggregates. Moreover, GM1-modified Aß42 aggregates exhibited prion-like properties in catalyzing the amyloid fibril formation of both major isomers of Aß, Aß40, and Aß42.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Humanos , Péptidos beta-Amiloides/química , Gangliósido G(M1)/química , Amiloide/química , Fragmentos de Péptidos/química , Enfermedad de Alzheimer/metabolismo
4.
Glycoconj J ; 38(1): 101-117, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33620588

RESUMEN

It is well over a century that glycosphingolipids are matter of interest in different fields of research. The hydrophilic oligosaccharide and the lipid moiety, the ceramide, both or separately have been considered in different moments as the crucial portion of the molecule, responsible for the role played by the glycosphingolipids associated to the plasma-membranes or to any other subcellular fraction. Glycosphingolipids are a family of compounds characterized by thousands of structures differing in both the oligosaccharide and the ceramide moieties, but among them, the nervous system monosialylated glycosphingolipid GM1, belonging to the group of gangliosides, has gained particular attention by a multitude of Scientists. In recent years, a series of studies have been conducted on the functional roles played by the hydrophilic part of GM1, its oligosaccharide, that we have named "OligoGM1". These studies allowed to shed new light on the mechanisms underlying the properties of GM1 defining the role of the OligoGM1 in determining precise interactions with membrane proteins instrumental for the neuronal functions, leaving to the ceramide the role of correctly positioning the GM1 in the membrane crucial for the oligosaccharide-protein interactions. In this review we aim to report the recent studies on the cascade of events modulated by OligoGM1, as the bioactive portion of GM1, to support neuronal differentiation and trophism together with preclinical studies on its potential to modify the progression of Parkinson's disease.


Asunto(s)
Gangliósido G(M1)/química , Gangliósido G(M1)/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Oligosacáridos/química , Animales , Diferenciación Celular , Gangliósido G(M1)/farmacología , Humanos , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Oligosacáridos/síntesis química , Oligosacáridos/metabolismo , Receptor trkA/metabolismo
5.
Sci Rep ; 10(1): 22102, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33328520

RESUMEN

SeviL is a recently isolated lectin found to bind to the linear saccharides of the ganglioside GM1b (Neu5Ac[Formula: see text](2-3)Gal[Formula: see text](1-3)GalNAc[Formula: see text](1-4)Gal[Formula: see text](1-4)Glc) and its precursor, asialo-GM1 (Gal[Formula: see text](1-3)GalNAc[Formula: see text](1-4)Gal[Formula: see text](1-4)Glc). The crystal structures of recombinant SeviL have been determined in the presence and absence of ligand. The protein belongs to the [Formula: see text]-trefoil family, but shows only weak sequence similarity to known structures. SeviL forms a dimer in solution, with one binding site per subunit, close to the subunit interface. Molecular details of glycan recognition by SeviL in solution were analysed by ligand- and protein-based NMR techniques as well as ligand binding assays. SeviL shows no interaction with GM1 due to steric hindrance with the sialic acid branch that is absent from GM1b. This unusual specificity makes SeviL of great interest for the detection and control of certain cancer cells, and cells of the immune system, that display asialo-GM1.


Asunto(s)
Gangliósido G(M1)/química , Gangliósidos/química , Lectinas/ultraestructura , Neoplasias/genética , Animales , Bivalvos/química , Secuencia de Carbohidratos , Gangliósido G(M1)/genética , Gangliósidos/genética , Humanos , Lectinas/química , Lectinas/genética , Neoplasias/patología
6.
Int J Mol Sci ; 21(12)2020 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-32599772

RESUMEN

Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) protein is expressed at the apical plasma membrane (PM) of different epithelial cells. The most common mutation responsible for the onset of cystic fibrosis (CF), F508del, inhibits the biosynthesis and transport of the protein at PM, and also presents gating and stability defects of the membrane anion channel upon its rescue by the use of correctors and potentiators. This prompted a multiple drug strategy for F508delCFTR aimed simultaneously at its rescue, functional potentiation and PM stabilization. Since ganglioside GM1 is involved in the functional stabilization of transmembrane proteins, we investigated its role as an adjuvant to increase the effectiveness of CFTR modulators. According to our results, we found that GM1 resides in the same PM microenvironment as CFTR. In CF cells, the expression of the mutated channel is accompanied by a decrease in the PM GM1 content. Interestingly, by the exogenous administration of GM1, it becomes a component of the PM, reducing the destabilizing effect of the potentiator VX-770 on rescued CFTR protein expression/function and improving its stabilization. This evidence could represent a starting point for developing innovative therapeutic strategies based on the co-administration of GM1, correctors and potentiators, with the aim of improving F508del CFTR function.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Aminofenoles/farmacología , Aminopiridinas/farmacología , Benzodioxoles/farmacología , Fibrosis Quística/tratamiento farmacológico , Gangliósido G(M1)/farmacología , Quinolonas/farmacología , Adyuvantes Inmunológicos/química , Aminofenoles/química , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Bronquios/patología , Agonistas de los Canales de Cloruro/química , Agonistas de los Canales de Cloruro/farmacología , Fibrosis Quística/genética , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Gangliósido G(M1)/química , Humanos , Mutación , Quinolonas/química , Terapias en Investigación
7.
FEBS J ; 287(12): 2612-2630, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31769916

RESUMEN

A 15-kDa lectin, termed SeviL, was isolated from Mytilisepta virgata (purplish bifurcate mussel). SeviL forms a noncovalent dimer that binds strongly to ganglio-series GM1b oligosaccharide (Neu5Acɑ2-3Galß1-3GalNAcß1-4Galß1-4Glc) and its precursor, asialo-GM1 (Galß1-3GalNAcß1-4Galß1-4Glc). SeviL also interacts weakly with the glycan moiety of SSEA-4 hexaose (Neu5Acα2-3Galß1-3GalNAcß1-3Galα1-4Galß1-4Glc). A partial protein sequence of the lectin was determined by mass spectrometry, and the complete sequence was identified from transcriptomic analysis. SeviL, consisting of 129 amino acids, was classified as an R(icin B)-type lectin, based on the presence of the QxW motif characteristic of this fold. SeviL mRNA is highly expressed in gills and, in particular, mantle rim tissues. Orthologue sequences were identified in other species of the family Mytilidae, including Mytilus galloprovincialis, from which lectin MytiLec-1 was isolated and characterized in our previous studies. Thus, mytilid species contain lectins belonging to at least two distinct families (R-type lectins and mytilectins) that have a common ß-trefoil fold structure but differing glycan-binding specificities. SeviL displayed notable cytotoxic (apoptotic) effects against various cultured cell lines (human breast, ovarian, and colonic cancer; dog kidney) that possess asialo-GM1 oligosaccharide at the cell surface. This cytotoxic effect was inhibited by the presence of anti-asialo-GM1 oligosaccharide antibodies. With HeLa ovarian cancer cells, SeviL showed dose- and time-dependent activation of kinase MKK3/6, p38 MAPK, and caspase-3/9. The transduction pathways activated by SeviL via the glycosphingolipid oligosaccharide were triggered apoptosis. DATABASE: Nucleotide sequence data have been deposited in the GenBank database under accession numbers MK434191, MK434192, MK434193, MK434194, MK434195, MK434196, MK434197, MK434198, MK434199, MK434200, and MK434201.


Asunto(s)
Gangliósido G(M1)/análogos & derivados , Lectinas/química , Lectinas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mytilidae/química , Oligosacáridos/metabolismo , Animales , Sitios de Unión , Gangliósido G(M1)/química , Gangliósido G(M1)/metabolismo , Células HeLa , Humanos , Lectinas/aislamiento & purificación , Proteínas Quinasas Activadas por Mitógenos/química , Mytilidae/metabolismo , Oligosacáridos/química , Especificidad de la Especie
8.
ACS Appl Mater Interfaces ; 11(28): 25017-25023, 2019 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-31265226

RESUMEN

Vibrio cholerae is a Gram-negative bacterium that causes secretory diarrhea and constitutes a major health threat in the industrialized world and even more in developing countries. Its main virulence factor is the cholera toxin, which is internalized by intestinal epithelial cells after binding to the glycosphingolipid receptor GM1a on their apical surface. A potential future solution to dampen complications of cholera infection is by scavenging the cholera toxin by presenting competitive binding motifs to diminish the in vivo toxicity of V. cholerae. Here, we generate GM1a-functionalized and biocompatible microgels with diameters of 20 µm using drop-based microfluidics. The microgels are designed to exhibit a mesoporous and widely meshed network structure, allowing diffusion of the toxin protein deep into the microgel scavengers. Flow cytometry demonstrates strong and multivalent binding at high capacity of these microgels to the binding domain of the cholera toxin. Cell culture-based assays reveal the ability of these microgels to scavenge and retain the cholera toxin in direct binding competition to colorectal cells. This ability is evidenced by suppressed cyclic adenosine monophosphate production as well as reduced vacuole formation in mucus-forming colorectal HT-29 cells. Therefore, glycan-functionalized microgels show great potential as a non-antibiotic treatment for toxin-mediated infectious disorders.


Asunto(s)
Toxina del Cólera , Gangliósido G(M1) , Microgeles/química , Vibrio cholerae/metabolismo , Toxina del Cólera/antagonistas & inhibidores , Toxina del Cólera/química , Toxina del Cólera/metabolismo , Gangliósido G(M1)/química , Gangliósido G(M1)/farmacología , Células HT29 , Humanos
9.
Langmuir ; 35(9): 3534-3544, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30802059

RESUMEN

The binding of the pentameric capsid protein VP1 of simian virus 40 to its glycosphingolipid receptor GM1 is a key step for the entry of the virus into the host cell. Recent experimental studies have shown that the interaction of variants of soluble VP1 pentamers with giant unilamellar vesicles composed of GM1, DOPC, and cholesterol leads to the formation of tubular membrane invaginations to the inside of the vesicles, mimicking the initial steps of endocytosis. We have used coarse-grained and atomistic molecular dynamics (MD) simulations to study the interaction of VP1 with GM1/DOPC/cholesterol bilayers. In the presence of one VP1 protein, we monitor the formation of small local negative curvature and membrane thinning at the protein binding site as well as reduction of area per lipid. These membrane deformations are also observed under cholesterol-free conditions. However, here, the number of GM1 molecules attached to the VP1 binding pockets increases. The membrane curvature is slightly increased for asymmetric GM1 distribution that mimics conditions in vivo, compared to symmetric GM1 distributions which are often applied in experiments. Slightly smaller inward curvature was observed in atomistic control simulations. Binding of four VP1 proteins leads to an increase of the average intrinsic area per lipid in the protein binding leaflet. Membrane fluctuations appear to be the driving force of VP1 aggregation, as was previously shown for membrane-adhering particles because no VP1 aggregation is observed in the absence of a lipid membrane.


Asunto(s)
Proteínas de la Cápside/metabolismo , Membrana Dobles de Lípidos/metabolismo , Receptores de Superficie Celular/metabolismo , Virus 40 de los Simios/química , Colesterol/química , Gangliósido G(M1)/química , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Fosfatidilcolinas/química
10.
Commun Biol ; 2: 52, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30729188

RESUMEN

EPDR1, a member of the ependymin-related protein family, is a relatively uncharacterized protein found in the lysosomes and secretomes of most vertebrates. Despite having roles in human disease and health, the molecular functions of EPDR1 remain unknown. Here, we present crystal structures of human EPDR1 and reveal that the protein adopts a fold previously seen only in bacterial proteins related to the LolA lipoprotein transporter. EPDR1 forms a homodimer with an overall shape resembling a half-shell with two non-overlapping hydrophobic grooves on the flat side of the hemisphere. EPDR1 can interact with membranes that contain negatively charged lipids, including BMP and GM1, and we suggest that EPDR1 may function as a lysosomal activator protein or a lipid transporter. A phylogenetic analysis reveals that the fold is more widely distributed than previously suspected, with representatives identified in all branches of cellular life.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Gangliósido G(M1)/química , Lisofosfolípidos/química , Monoglicéridos/química , Proteínas de Neoplasias/química , Proteínas de Unión Periplasmáticas/química , Secuencia de Aminoácidos , Animales , Baculoviridae/genética , Baculoviridae/metabolismo , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Gangliósido G(M1)/metabolismo , Expresión Génica , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Lisofosfolípidos/metabolismo , Lisosomas/metabolismo , Modelos Moleculares , Monoglicéridos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso , Proteínas de Unión Periplasmáticas/genética , Proteínas de Unión Periplasmáticas/metabolismo , Filogenia , Plantas/genética , Plantas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido
11.
Glycobiology ; 29(3): 242-259, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30535277

RESUMEN

The hepatitis B virus (HBV)-induced chronic liver diseases are serious health threats worldwide. There is evidence to display the alterations of salivary N-linked glycans related to the development of HBV-infected liver diseases. Here, we further investigated the alterations of fucosylated N/O-glycans recognized by LTL in saliva from 120 subjects (30 healthy volunteers (HV), 30 patients with hepatitis B (HB), 30 patients with hepatic cirrhosis (HC), and 30 patients with hepatocellular carcinoma (HCC)) using salivary microarrys and MALDI-TOF/TOF-MS. The results showed that the expression level of fucosylated glycans recognized by LTL was significantly increased in HCC compared with other subjects (P < 0.0001). Besides, the fucosylated glycoproteins were isolated from pooled saliva of HV, HB, HC, and HCC by LTL-magnetic particle conjugates. Then, N/O- glycans were released from the isolated glycoproteins with PNGase F and NaClO, and were identified by MALDI-TOF-MS, respectively. Totally, there were 21/20, 25/18, 29/19, and 28/24 N/O-glycan peaks that were identified and annotated with proposed structures in saliva of HV, HB, HC, and HCC. Among the total, there were 8 N-glycan peaks (e.g., m/z 1905.634, 2158.777 and 2905.036) and 15 O-glycan peaks (e.g., 1177.407, 1308.444 and 1322.444) that only presented in patients with HBV-induced liver diseases. One N-glycan peak (m/z 2205.766) was unique in HC, and 9 O-glycan peaks (e.g., m/z 1157.420, 1163.417 and 1193.402) were unique in HCC. This study could facilitate the discovery of biomarkers for HC and HCC based on precise alterations of fucosylated N/O-glycans in saliva.


Asunto(s)
Biomarcadores de Tumor/genética , Virus de la Hepatitis B/genética , Polisacáridos/genética , Análisis por Matrices de Proteínas , Biomarcadores de Tumor/química , Biomarcadores de Tumor/aislamiento & purificación , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Femenino , Fibrosis/genética , Fibrosis/virología , Gangliósido G(M1)/análogos & derivados , Gangliósido G(M1)/química , Gangliósido G(M1)/genética , Virus de la Hepatitis B/aislamiento & purificación , Virus de la Hepatitis B/patogenicidad , Hepatitis Crónica/genética , Hepatitis Crónica/virología , Humanos , Lectinas/química , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Masculino , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Saliva/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
12.
ACS Chem Neurosci ; 10(1): 563-572, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30346704

RESUMEN

The abnormal aggregation of amyloid ß-protein (Aß) is considered central in the pathogenesis of Alzheimer's disease. We focused on membrane-mediated amyloidogenesis and found that amyloid fibrils formed on monosialoganglioside GM1 clusters were more toxic than those formed in aqueous solution. In this study, we investigated the structure of the toxic fibrils by Aß-(1-40) in detail in comparison with less-toxic fibrils formed in aqueous solution. The less-toxic fibrils contain in-resister parallel ß-sheets, whereas the structure of the toxic fibrils is unknown. Atomic force microscopy revealed that the toxic fibrils had a flat, tape-like morphology composed of a single ß-sheet layer. Isotope-edited infrared spectroscopy indicated that almost the entire sequence of Aß is included in the ß-sheet. Chemical cross-linking experiments using Cys-substituted Aßs suggested that the fibrils mainly contained both in-resister parallel and two-residue-shifted antiparallel ß-sheet structures. Solid-state NMR experiments also supported this conclusion. Thus, the toxic fibrils were found to possess a novel unique structure.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Proteínas Amiloidogénicas/metabolismo , Gangliósido G(M1)/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/química , Amiloide/metabolismo , Péptidos beta-Amiloides/química , Amiloidosis/metabolismo , Gangliósido G(M1)/química , Humanos , Estructura Secundaria de Proteína/fisiología
13.
Microb Pathog ; 124: 332-336, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30145256

RESUMEN

Binding of cholera toxin subunit B (CTB) to its receptor and toxin transport into the intestinal epithelial cells are the causative events for the potentially lethal disease cholera. The five sugar mono-sialo ganglioside GM1 is the cell surface receptor for cholera toxin B-subunit. CTB binding was determined by use of immobilized GM1 to microtiter plates and by immunohistochemistry. Sections from the human colon and the human soft palate were incubated with FITC-conjugated CTB and with anti-MUC2. Both the luminal surface of the intestine and the secretory goblet cells exhibited strong binding. Addition of simple carbohydrates and milk to the incubation medium showed that a combination of lactose and non-fat dry milk was potent inhibitors of toxin- and mucin binding. Both CTB and ant-MUC2 stained to the cytoplasm (mucin granules) in the goblet cells from the human soft palate. In the colon CTB stained the entire cytoplasm of the goblet cells while anti-MUC2 detected only the supranuclear region of some cells, suggesting carbohydrate heterogeneity between goblet cell mucin granules in different regions of the human body. Both CTB- and MUC2 binding were inhibited when GM1 was added to the incubation medium. It is proposed that the human colonic goblet cells play a role in the secretory diarrhea in patients with cholera and that milk might have a prophylactic or therapeutic application in the management of cholera.


Asunto(s)
Toxina del Cólera/metabolismo , Cólera/microbiología , Intestino Grueso/microbiología , Vibrio cholerae/metabolismo , Cólera/metabolismo , Toxina del Cólera/química , Toxina del Cólera/genética , Células Epiteliales/química , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Gangliósido G(M1)/química , Gangliósido G(M1)/metabolismo , Humanos , Intestino Grueso/química , Intestino Grueso/metabolismo , Cinética , Unión Proteica , Vibrio cholerae/química , Vibrio cholerae/genética
14.
Elife ; 72018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29851380

RESUMEN

Transport of biologically active molecules across tight epithelial barriers is a major challenge preventing therapeutic peptides from oral drug delivery. Here, we identify a set of synthetic glycosphingolipids that harness the endogenous process of intracellular lipid-sorting to enable mucosal absorption of the incretin hormone GLP-1. Peptide cargoes covalently fused to glycosphingolipids with ceramide domains containing C6:0 or smaller fatty acids were transported with 20-100-fold greater efficiency across epithelial barriers in vitro and in vivo. This was explained by structure-function of the ceramide domain in intracellular sorting and by the affinity of the glycosphingolipid species for insertion into and retention in cell membranes. In mice, GLP-1 fused to short-chain glycosphingolipids was rapidly and systemically absorbed after gastric gavage to affect glucose tolerance with serum bioavailability comparable to intraperitoneal injection of GLP-1 alone. This is unprecedented for mucosal absorption of therapeutic peptides, and defines a technology with many other clinical applications.


Asunto(s)
Absorción Fisiológica , Glicoesfingolípidos/metabolismo , Membrana Mucosa/metabolismo , Péptidos/uso terapéutico , Animales , Transporte Biológico Activo , Glucemia/metabolismo , Núcleo Celular/metabolismo , Ceramidas/química , Perros , Células Epiteliales/metabolismo , Gangliósido G(M1)/química , Gangliósido G(M1)/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células de Riñón Canino Madin Darby , Masculino , Ratones Endogámicos C57BL , Oligosacáridos/química , Oligosacáridos/metabolismo , Reproducibilidad de los Resultados , Soluciones , Relación Estructura-Actividad , Transcitosis
15.
Rapid Commun Mass Spectrom ; 32(17): 1481-1490, 2018 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-29876976

RESUMEN

RATIONALE: Certain lung cancer patients express elevated Fucosyl Monosialoganglioside (Fuc-GM1) in circulation compared to control groups. Several sensitive methods involving characterization of Fuc-GM1 have been reported. However, a highly specific and sensitive method for quantifying multiple potential Fuc-GM1 biomarkers present in various biological matrices has not been reported to date. METHODS: Individual Fuc-GM1 analogs in a commercially obtained standard mixture were characterized using HPLC/UV/MS and high-resolution mass spectrometry (HRMS). Proprietary antibodies, mAb1 and mAb2, were used to selectively capture and pre-concentrate the soluble and drug-bound forms of Fuc-GM1 molecules present in human serum and whole blood, eliminating the background matrix components. Immunocapture extraction (ICE) followed by HPLC/MS/MS was used to quantify specific Fuc-GM1 analogs in biological matrices. RESULTS: The concentration of individual Fuc-GM1 analogs in the standard mixture was estimated to be 7-34%, using HPLC/UV/MS. Using the standard mixture spiked into the biological matrices (100 µL), the lower limit of quantification (LLOQ) of each analog was 0.2-0.4 ng/mL with a dynamic range of up to 200 ng/mL. The applicability of the ICE-HPLC/MS/MS method was demonstrated by detecting endogenous Fuc-GM1 analogs present in rat blood and in several lung cancer cell lines. CONCLUSIONS: This highly specific and sensitive HPLC/MS/MS method for quantifying individual potential Fuc-GM1 biomarkers in serum and whole blood can play a critical role in patient stratification strategies and during drug treatment. This method can be employed for monitoring both free (soluble) form and antibody drug-bound Fuc-GM1.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Gangliósido G(M1)/análogos & derivados , Neoplasias Pulmonares/sangre , Espectrometría de Masas en Tándem/métodos , Animales , Anticuerpos Monoclonales/análisis , Biomarcadores/sangre , Biomarcadores/química , Gangliósido G(M1)/sangre , Gangliósido G(M1)/química , Gangliósido G(M1)/aislamiento & purificación , Humanos , Ratas
16.
ACS Chem Neurosci ; 9(6): 1264-1268, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29558805

RESUMEN

In the search of a potent candidate for neurotherapy, we designed and synthesized various analogues of ganglioside Hp-s1. The modification includes the change in hydrophobicity by varying the carbon chain length, altering the number of hydrogen bonds, and replacing the anomeric atom. The chemical synthesis was carried out by using various methods and discussed in details. The neuritogenic activities of these analogues are confirmed in a human neuroblastoma cell line SH-SY5Y. A higher activity of ganglioside Hp-s1 analogue on IL-17A transcript upregulation than ganglioside Hp-s1 was found.


Asunto(s)
Bioensayo , Gangliósido G(M1)/metabolismo , Gangliósidos/metabolismo , Neurogénesis/fisiología , Bioensayo/métodos , Línea Celular , Gangliósido G(M1)/química , Gangliósidos/química , Humanos , Neuritas/fisiología , Neuroblastoma/metabolismo , Células Tumorales Cultivadas
17.
PLoS Negl Trop Dis ; 12(2): e0006266, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29470490

RESUMEN

Diarrheal diseases are a major cause of morbidity and mortality worldwide. In many cases, antibiotic therapy is either ineffective or not recommended due to concerns about emergence of resistance. The pathogenesis of several of the most prevalent infections, including cholera and enteroxigenic Escherichia coli, is dominated by enterotoxins produced by lumen-dwelling pathogens before clearance by intestinal defenses. Toxins gain access to the host through critical host receptors, making these receptors attractive targets for alternative antimicrobial strategies that do not rely on conventional antibiotics. Here, we developed a new nanotechnology strategy as a countermeasure against cholera, one of the most important and prevalent toxin-mediated enteric infections. The key host receptor for cholera toxin, monosialotetrahexosylganglioside (GM1), was coated onto the surface of polymeric nanoparticles. The resulting GM1-polymer hybrid nanoparticles were shown to function as toxin decoys by selectively and stably binding cholera toxin, and neutralizing its actions on epithelial cells in vitro and in vivo. Furthermore, the GM1-coated nanoparticle decoys attenuated epithelial 3',5'-cyclic adenosine monophosphate production and fluid responses to infection with live Vibrio cholera in cell culture and a murine infection model. Together, these studies illustrate that the new nanotechnology-based platform can be employed as a non-traditional antimicrobial strategy for the management of enteric infections with enterotoxin-producing pathogens.


Asunto(s)
Toxina del Cólera/metabolismo , Cólera/tratamiento farmacológico , Gangliósido G(M1)/metabolismo , Nanopartículas , Vibrio cholerae/patogenicidad , Animales , Sitios de Unión , Línea Celular Tumoral , Cólera/microbiología , Cólera/fisiopatología , Toxina del Cólera/química , AMP Cíclico/metabolismo , Femenino , Gangliósido G(M1)/química , Mucosa Intestinal/metabolismo , Intestino Delgado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Nanotecnología/métodos
18.
J Neurochem ; 143(6): 645-659, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28796418

RESUMEN

GM1 ganglioside (II3 NeuAc-Gg4 Cer) is known to promote neurite formation in neuroblastoma cells by activating TrkA-MAPK pathway. The molecular mechanism by which GM1 is involved in the neurodifferentiation process is still unknown, however, in vitro and in vivo evidences have suggested that the oligosaccharide portion of this ganglioside could be involved. Here, we report that, similarly to the entire GM1 molecule, its oligosaccharide II3 NeuAc-Gg4, rather than its ceramide (Cer) portion is responsible for the neurodifferentiation process by augmenting neurite elongation and increasing the neurofilament protein expression in murine neuroblastoma cells, Neuro2a. Conversely, asialo-GM1, GM2 and GM3 oligosaccharides are not effective in neurite elongation on Neuro2a cells, whereas the effect exerted by the Fuc-GM1 oligosaccharide (IV2 αFucII3 Neu5Ac-Gg4 ) is similar to that exerted by GM1 oligosaccharide. The neurotrophic properties of GM1 oligosaccharide are exerted by activating the TrkA receptor and the following phosphorylation cascade. By photolabeling experiments performed with a nitrophenylazide containing GM1 oligosaccharide, labeled with tritium, we showed a direct interaction between the GM1 oligosaccharide and the extracellular domain of TrkA receptor. Moreover, molecular docking analyses confirmed that GM1 oligosaccharide binds the TrkA-nerve growth factor complex leading to a binding free energy of approx. -11.5 kcal/mol, acting as a bridge able to increase and stabilize the TrkA-nerve growth factor molecular interactions.


Asunto(s)
Gangliósido G(M1)/metabolismo , Neuritas/metabolismo , Neuroblastoma , Receptor trkA/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular , Gangliósido G(M1)/química , Ratones , Simulación del Acoplamiento Molecular , Oligosacáridos/química , Oligosacáridos/metabolismo , Transducción de Señal/fisiología
19.
Int J Nanomedicine ; 12: 4879-4889, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28744122

RESUMEN

For the treatment of glioma and other central nervous system diseases, one of the biggest challenges is that most therapeutic drugs cannot be delivered to the brain tumor tissue due to the blood-brain barrier (BBB). The goal of this study was to construct a nanodelivery vehicle system with capabilities to overcome the BBB for central nervous system administration. Doxorubicin as a model drug encapsulated in ganglioside GM1 micelles was able to achieve up to 9.33% loading efficiency and 97.05% encapsulation efficiency by orthogonal experimental design. The in vitro study demonstrated a slow and sustainable drug release in physiological conditions. In the cellular uptake studies, mixed micelles could effectively transport into both human umbilical vein endothelial cells and C6 cells. Furthermore, biodistribution imaging of mice showed that the DiR/GM1 mixed micelles were accumulated sustainably and distributed centrally in the brain. Experiments on zebrafish confirmed that drug-loaded GM1 micelles can overcome the BBB and enter the brain. Among all the treatment groups, the median survival time of C6-bearing rats after administering DOX/GM1 micelles was significantly prolonged. In conclusion, the ganglioside nanomicelles developed in this work can not only penetrate BBB effectively but also repair nerves and kill tumor cells at the same time.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Doxorrubicina/farmacología , Gangliósido G(M1)/química , Glioma/tratamiento farmacológico , Animales , Animales Modificados Genéticamente , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacología , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Gangliósidos/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Micelas , Regeneración Nerviosa/efectos de los fármacos , Ratas Wistar , Distribución Tisular , Pez Cebra/genética
20.
Biophys J ; 113(6): 1194-1199, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28410623

RESUMEN

In this perspective we summarize current knowledge of the effect of monosialoganglioside GM1 on the membrane-mediated aggregation of the ß-amyloid (Aß) peptide. GM1 has been suggested to be actively involved in the development of Alzheimer's disease due to its ability to seed the aggregation of Aß. However, GM1 is known to be neuroprotective against Aß-induced toxicity. Here we suggest that the two scenarios are not mutually exclusive but rather complementary, and might depend on the organization of GM1 in membranes. Improving our understanding of the molecular details behind the role of gangliosides in neurodegenerative amyloidoses might help in developing disease-modifying treatments.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Gangliósido G(M1)/metabolismo , Agregación Patológica de Proteínas/metabolismo , Péptidos beta-Amiloides/química , Encéfalo/metabolismo , Gangliósido G(M1)/química , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA