Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 502
Filtrar
1.
Phys Chem Chem Phys ; 26(16): 12552-12563, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38595108

RESUMO

Ganglioside GM1 is a class of glycolipids predominantly located in the nervous system. Comprising a ceramide anchor and an oligosaccharide chain containing sialic acid, GM1 plays a pivotal role in various cellular processes, including signal transduction, cell adhesion, and membrane organization. Moreover, GM1 has been implicated in the pathogenesis of several neurological disorders, such as Parkinson's disease, Alzheimer's disease, and stroke. In this study, by creating a neural cell model membrane simulation system and employing rigorous molecular models, we utilize a coarse-grained molecular dynamics approach to explore the structural and dynamic characteristics of multi-component neuronal plasma membranes at varying GM1 ganglioside concentrations. The simulation results reveal that as GM1 concentration increases, a greater number of hydrogen bonds form between GM1 molecules, resulting in the formation of larger clusters, which leads to reduced membrane fluidity, increased lipid ordering, decreased membrane thickness and surface area and higher levels of GM1 dissociation. Through a meticulous analysis, while considering GM1's structural attributes, we offer valuable insights into the structural and dynamic traits of the cell membrane. This study provides a robust methodology for exploring membrane characteristics and enhances our comprehension of GM1 molecules, serving as a resource for both experimental and computational researchers in this field.


Assuntos
Membrana Celular , Gangliosídeo G(M1) , Simulação de Dinâmica Molecular , Gangliosídeo G(M1)/química , Gangliosídeo G(M1)/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Ligação de Hidrogênio , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo
2.
J Phys Chem B ; 128(11): 2745-2754, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38447189

RESUMO

Monosialoganglioside (GM1), a ubiquitous component of lipid rafts, and hemin, an integral part of heme proteins such as hemoglobin, are essential to the cell membranes of brain neurons and erythrocyte red blood cells for regulating cellular communication and oxygen transport. Protoporphyrin IX (PPIX) and its derivative hemin, on the contrary, show significant cytotoxic effects when in excess causing hematological diseases, such as thalassemia, anemia, malaria, and neurodegeneration. However, the in-depth molecular etiology of their interactions with the cell membrane has so far been poorly understood. Herein, the structure of the polymer cushion-supported lipid bilayer (SLB) of the binary mixture of phospholipid and GM1 in the presence of PPIX and its derivative hemin has been investigated to predict the molecular interactions in model phospholipid membranes. A high-resolution synchrotron-based X-ray scattering technique has been employed to explore the out-of-plane structure of the assembly at different compositions and concentrations. The structural changes have been complemented with the isobaric changes in the mean molecular area obtained from the Langmuir monolayer isotherm to predict the additive-induced membrane condensation and fluidization. PPIX-induced fluidization of phospholipid SLB without GM1 was witnessed, which was reversed to condensation with 2-fold higher structural changes in the presence of GM1. A hemin concentration-dependent linear condensing effect was observed in the pristine SLB. The effect was significantly reduced, and the linearity was observed to be lost in the mixed SLB containing GM1. Our study shows that GM1 alters the interaction of hemin and PPIX with the membrane, which could be explained with the aid of hydrophobic and electrostatic interactions. Our study indicates favorable and unfavorable interactions of GM1 with PPIX and hemin, respectively, in the membrane. The observed structural changes in both SLB and the underlying polymer cushion layer lead to the proposal of a molecule-specific interaction model that can benefit the pharmaceutical industries specialized for drug designing. Our study potentially enriches our fundamental biophysical understanding of neurodegenerative diseases and drug-membrane interactions.


Assuntos
Fosfolipídeos , Protoporfirinas , Hemina/metabolismo , Gangliosídeo G(M1)/química , Adsorção , Bicamadas Lipídicas/química , Polímeros
3.
Molecules ; 29(3)2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38338484

RESUMO

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.


Assuntos
Doença de Alzheimer , Diabetes Mellitus , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Peptídeos beta-Amiloides/metabolismo , Gangliosídeo G(M1)/química , Bicamadas Lipídicas/química , Fosfatidilcolinas
4.
ACS Chem Neurosci ; 14(24): 4335-4343, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38050745

RESUMO

Amyloid ß peptide (Aß) is the crucial protein component of extracellular plaques in Alzheimer's disease. The plaques also contain gangliosides lipids, which are abundant in membranes of neuronal cells and in cell-derived vesicles and exosomes. When present at concentrations above its critical micelle concentration (cmc), gangliosides can occur as mixed micelles. Here, we study the coassembly of the ganglioside GM1 and the Aß peptides Aß40 and 42 by means of microfluidic diffusional sizing, confocal microscopy, and cryogenic transmission electron microscopy. We also study the effects of lipid-peptide interactions on the amyloid aggregation process by fluorescence spectroscopy. Our results reveal coassembly of GM1 lipids with both Aß monomers and Aß fibrils. The results of the nonseeded kinetics experiments show that Aß40 aggregation is delayed with increasing GM1 concentration, while that of Aß42 is accelerated. In seeded aggregation reactions, the addition of GM1 leads to a retardation of the aggregation process of both peptides. Thus, while the effect on nucleation differs between the two peptides, GM1 may inhibit the elongation of both types of fibrils. These results shed light on glycolipid-peptide interactions that may play an important role in Alzheimer's pathology.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Peptídeos beta-Amiloides/metabolismo , Gangliosídeos/metabolismo , Micelas , Gangliosídeo G(M1)/química , Amiloide/metabolismo , Doença de Alzheimer/metabolismo , Fragmentos de Peptídeos/metabolismo
5.
Glycoconj J ; 40(6): 655-668, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38100017

RESUMO

Since the 1980s, it has been known that the administration of ganglioside GM1 to cultured cells induced or enhanced neuronal differentiation. GM1 mechanism of action relies on its direct interaction and subsequent activation of the membrane tyrosine kinase receptor, TrkA, which naturally serves as NGF receptor. This process is mediated by the sole oligosaccharide portion of GM1, the pentasaccharide ß-Gal-(1-3)-ß-GalNAc-(1-4)-[α-Neu5Ac-(2-3)]-ß-Gal-(1-4)-ß-Glc. Here we detailed the minimum structural requirements of the oligosaccharide portion of GM1 for mediating the TrkA dependent neuritogenic processing. By in vitro and in silico biochemical approaches, we demonstrated that the minimal portion of GM1 required for the TrkA activation is the inner core of the ganglioside's oligosaccharide ß-Gal-(1-3)-ß-GalNAc-(1-4)-[α-Neu5Ac-(2-3)]-ß-Gal. The addition of a sialic acid residue at position 3 of the outer galactose of the GM1 oligosaccharide, which forms the oligosaccharide of GD1a, prevented the interaction with TrkA and the resulting neuritogenesis. On the contrary, the addition of a fucose residue at position 2 of the outer galactose, forming the Fucosyl-GM1 oligosaccharide, did not prevent the TrkA-mediated neuritogenesis.


Assuntos
Gangliosídeo G(M1) , Galactose , Gangliosídeo G(M1)/química , Ácido N-Acetilneuramínico , Oligossacarídeos/química
6.
ACS Chem Neurosci ; 14(23): 4199-4207, 2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-37971427

RESUMO

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.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Humanos , Peptídeos beta-Amiloides/metabolismo , Gangliosídeo G(M1)/química , Ciclização , Doença de Alzheimer/metabolismo , Gangliosídeos/metabolismo , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/metabolismo
7.
Biophys Chem ; 301: 107091, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37549471

RESUMO

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.


Assuntos
Doença de Alzheimer , Neuroblastoma , Humanos , Peptídeos beta-Amiloides/química , Gangliosídeo G(M1)/química , Amiloide/química , Fragmentos de Peptídeos/química , Doença de Alzheimer/metabolismo
8.
J Phys Chem B ; 127(31): 6940-6948, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37523476

RESUMO

SARS-CoV-2 spike glycoprotein is anchored by gangliosides. The sialic acid in the ganglioside headgroup is responsible for virus attachment and entry into host cells. We used coarse-grained (CG) molecular dynamics simulations to expand on our previous study of GM1 interaction with two different orientations of the SARS-CoV-2 S1 subunit N-terminal domain (NTD) and to confirm the role of sialic acid receptors in driving the viral receptor; GM3 was used as another ganglioside on the membrane. Because of the smaller headgroup, sialic acid is crucial in GM3 interactions, whereas GM1 interacts with NTD via both the sialic acid and external galactose. In line with our previous findings for NTD orientations in GM1 binding, we identified two orientations, "compact" and "distributed", comprising sugar receptor-interacting residues in GM3-embedded lipid bilayers. Gangliosides in closer proximity to the compact NTD orientation might cause relatively greater restrictions to penetrate the bilayer. However, the attachment of a distributed NTD orientation with more negative interaction energies appears to facilitate GM1/GM3 to move quickly across the membrane. Our findings likely shed some light on the orientations that the NTD receptor acquires during the early phases of interaction with GM1 and GM3 in a membrane environment.


Assuntos
COVID-19 , Gangliosídeo G(M3) , Humanos , Gangliosídeo G(M1)/química , Gangliosídeo G(M3)/química , Gangliosídeos/química , Ácido N-Acetilneuramínico , SARS-CoV-2/metabolismo
9.
ACS Chem Neurosci ; 14(15): 2648-2657, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37482658

RESUMO

Alzheimer's disease (AD) is associated with progressive accumulation of amyloid-ß (Aß) cross-ß fibrils in the brain. Aß species tightly associated with GM1 ganglioside, a glycosphingolipid abundant in neuronal membranes, promote amyloid fibril formation; therefore, they could be attractive clinical targets. However, the active conformational state of Aß in GM1-containing lipid membranes is still unknown. The present solid-state nuclear magnetic resonance study revealed a nonfibrillar Aß assemblage characterized by a double-layered antiparallel ß-structure specifically formed on GM1 ganglioside clusters. Our data show that this unique assemblage was not transformed into fibrils on GM1-containing membranes but could promote conversion of monomeric Aß into fibrils, suggesting that a solvent-exposed hydrophobic layer provides a catalytic surface evoking Aß fibril formation. Our findings offer structural clues for designing drugs targeting catalytically active Aß conformational species for the development of anti-AD therapeutics.


Assuntos
Doença de Alzheimer , Gangliosídeo G(M1) , Humanos , Gangliosídeo G(M1)/química , Peptídeos beta-Amiloides/química , Doença de Alzheimer/patologia , Amiloide/química , Neurônios/patologia
10.
Artigo em Inglês | MEDLINE | ID: mdl-37330108

RESUMO

Fibrillary aggregated α-synuclein represents the neurologic hallmark of Parkinson's disease and is considered to play a causative role in the disease. Although the causes leading to α-synuclein aggregation are not clear, the GM1 ganglioside interaction is recognized to prevent this process. How GM1 exerts these functions is not completely clear, although a primary role of its soluble oligosaccharide (GM1-OS) is emerging. Indeed, we recently identified GM1-OS as the bioactive moiety responsible for GM1 neurotrophic and neuroprotective properties, specifically reverting the parkinsonian phenotype both in in vitro and in vivo models. Here, we report on GM1-OS efficacy against the α-synuclein aggregation and toxicity in vitro. By amyloid seeding aggregation assay and NMR spectroscopy, we demonstrated that GM1-OS was able to prevent both the spontaneous and the prion-like α-synuclein aggregation. Additionally, circular dichroism spectroscopy of recombinant monomeric α-synuclein showed that GM1-OS did not induce any change in α-synuclein secondary structure. Importantly, GM1-OS significantly increased neuronal survival and preserved neurite networks of dopaminergic neurons affected by α-synuclein oligomers, together with a reduction of microglia activation. These data further demonstrate that the ganglioside GM1 acts through its oligosaccharide also in preventing the α-synuclein pathogenic aggregation in Parkinson's disease, opening a perspective window for GM1-OS as drug candidate.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Humanos , alfa-Sinucleína/genética , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , Gangliosídeo G(M1)/farmacologia , Gangliosídeo G(M1)/química , Oligossacarídeos/farmacologia
11.
ACS Infect Dis ; 9(7): 1346-1361, 2023 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-37145972

RESUMO

Multiple recent reports indicate that the S protein of SARS-CoV-2 specifically interacts with membrane receptors and attachment factors other than ACE2. They likely have an active role in cellular attachment and entry of the virus. In this article, we examined the binding of SARS-CoV-2 particles to gangliosides embedded in supported lipid bilayers (SLBs), mimicking the cell membrane-like environment. We show that the virus specifically binds to sialylated (sialic acid (SIA)) gangliosides, i.e., GD1a, GM3, and GM1, as determined from the acquired single-particle fluorescence images using a time-lapse total internal reflection fluorescence (TIRF) microscope. The data of virus binding events, the apparent binding rate constant, and the maximum virus coverage on the ganglioside-rich SLBs show that the virus particles have a higher binding affinity toward the GD1a and GM3 compared to the GM1 ganglioside. Enzymatic hydrolysis of the SIA-Gal bond of the gangliosides confirms that the SIA sugar unit of GD1a and GM3 is essential for virus attachment to the SLBs and even the cell surface sialic acid is critical for the cellular attachment of the virus. The structural difference between GM3/GD1a and GM1 is the presence of SIA at the main or branched chain. We conclude that the number of SIA per ganglioside can weakly influence the initial binding rate of SARS-CoV-2 particles, whereas the terminal or more exposed SIA is critical for the virus binding to the gangliosides in SLBs.


Assuntos
COVID-19 , Gangliosídeos , Humanos , Gangliosídeos/química , Ácido N-Acetilneuramínico/metabolismo , Gangliosídeo G(M1)/química , Gangliosídeo G(M1)/metabolismo , SARS-CoV-2/metabolismo
12.
Biophys J ; 122(11): 1926-1937, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-35986516

RESUMO

Galectin-3 (Gal-3) is a ß-galactosidase-binding protein involved in various biological processes, including neuronal growth and adhesion. The pairing of Gal-3 with ganglioside GM1's pentasaccharide chain at the outer leaflet of the plasma membrane, which triggers downstream cell-signaling cascades, seems to be involved in these processes. A crucial feature of Gal-3 is its ability to form oligomers and supramolecular assemblies that connect various carbohydrate-decorated molecules. Although we know the atomistic structure of Gal-3 bound to small carbohydrate ligands, it remains unclear how Gal-3 binds GM1 in a membrane. Furthermore, the influence of this interaction on Gal-3's structure and oligomeric assembly has to be elucidated. In this study, we used X-ray reflectivity (XR) from a model membrane to determine the structure and surface coverage of Gal-3 bound to a membrane containing GM1. We observed that the carbohydrate recognition domain interacts with GM1's pentasaccharide, while the N-terminal domain is pointed away from the membrane, likely to facilitate protein-protein interactions. In a membrane containing 20 mol % GM1, Gal-3 covered ∼50% of the membrane surface with one Gal-3 molecule bound per 2130 Å2. We used molecular dynamics simulations and Voronoi tessellation algorithms to build an atomistic model of membrane-bound Gal-3, which is supported by the XR results. Overall, this work provides structural information describing how Gal-3 can bind GM1's pentasaccharide chain, a prerequisite for triggering regulatory processes in neuronal growth and adhesion.


Assuntos
Gangliosídeo G(M1) , Galectina 3 , Gangliosídeo G(M1)/química , Galectina 3/metabolismo , Gangliosídeos , Membrana Celular/metabolismo , Simulação de Dinâmica Molecular
13.
Anal Chem ; 94(46): 16042-16049, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36367338

RESUMO

Interactions between glycan-binding proteins (GBPs) and glycosphingolipids (GSLs) are involved in numerous physiological and pathophysiological processes. Many model membrane systems are available for studying GBP-GSL interactions, but a systematic investigation has not been carried out on how the nature of the model membrane affects binding. In this work, we use electrospray ionization mass spectrometry (ESI-MS), both direct and competitive assays, to measure the binding of cholera toxin B subunit homopentamer (CTB5) to GM1 ganglioside in liposomes, bilayer islands [styrene maleic acid lipid particles (SMALPs), nanodiscs (NDs), and picodiscs (PDs)], and micelles. We find that direct ESI-MS analysis of CTB5 binding to GM1 is unreliable due to non-uniform response factors, incomplete extraction of bound GM1 in the gas phase, and nonspecific CTB5-GM1 interactions. Conversely, indirect proxy ligand ESI-MS measurements show that the intrinsic (per binding site) association constants of CTB5 for PDs, NDs, and SMALPs are similar and comparable to the affinity of soluble GM1 pentasaccharide (GM1os). The observed affinity decreases with increasing GM1 content due to molecular crowding stemming from GM1 clustering. Unlike the smaller model membranes, the observed affinity of CTB5 toward GM1 liposomes is ∼10-fold weaker than GM1os and relatively insensitive to the GM1 content. GM1 glycomicelles exhibit the lowest affinity, ∼35-fold weaker than GM1os. Together, the results highlight experimental design considerations for quantitative GBP-GSL binding studies involving multisubunit GBPs and factors to consider when comparing results obtained with different membrane systems. Notably, they suggest that bilayer islands with a low percentage of GSL, wherein clustering is minimized, are ideal for assessing intrinsic strength of GBP-GSL interactions in a membrane environment, while binding to liposomes, which is sub-optimal due to extensive clustering, may be more representative of authentic cellular environments.


Assuntos
Gangliosídeo G(M1) , Glicoesfingolipídeos , Toxina da Cólera/química , Gangliosídeo G(M1)/química , Glicoesfingolipídeos/química , Lipossomos , Proteínas/química , Espectrometria de Massas por Ionização por Electrospray/métodos
14.
Biophys Chem ; 290: 106874, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36067650

RESUMO

Lipid binding kinetics and energetics of self-aggregated and disordered beta-amyloid oligomers of various sizes, from solution to lipid raft surfaces, were investigated using MD simulations. Our systems include small (monomers to tetramers) and larger (octamers and dodecamers) oligomers. Our lipid rafts contain saturated and unsaturated phosphatidylcholine (PC), cholesterol, and with or without asymmetrically distributed monosialotetrahexosylganglioside (GM1). All rafts exhibited dynamic and structurally diversified domains including liquid-ordered (Lo), liquid-disordered (Ld), and interfacial Lod domains. For rafts without GM1, all oligomers bound to the Lod domain. For GM1-containing rafts, all small oligomers and most larger oligomers bound specifically to the GM1-clusters embedded in the Lo domain. Lipid-protein binding energies followed an order of GM1 >> unsaturated PC > saturated PC > cholesterol for all rafts. In addition, protein-induced membrane structural disruption increased progressively with the size of the oligomer for the annular lipids surrounding the membrane-bound protein in non-GM1-containing rafts. We propose that the tight binding of beta-amyloid oligomers to the GM1-clusters and the structural perturbation of lipids surrounding the membrane-bound proteins at the Lod domain are early molecular events of the beta-amyloid aggregation process on neuronal membrane surfaces that trigger the onset of Alzheimer's.


Assuntos
Peptídeos beta-Amiloides , Gangliosídeos , Peptídeos beta-Amiloides/química , Colesterol/química , Gangliosídeo G(M1)/química , Gangliosídeos/análise , Gangliosídeos/metabolismo , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Fosfatidilcolinas/metabolismo
15.
Biochemistry ; 61(20): 2206-2220, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36173882

RESUMO

A major hallmark of Alzheimer's disease (AD) is the accumulation of extracellular aggregates of amyloid-ß (Aß). Structural polymorphism observed among Aß fibrils in AD brains seem to correlate with the clinical subtypes suggesting a link between fibril polymorphism and pathology. Since fibrils emerge from a templated growth of low-molecular-weight oligomers, understanding the factors affecting oligomer generation is important. Membrane lipids are key factors to influence early stages of Aß aggregation and oligomer generation, which cause membrane disruption. We have previously demonstrated that conformationally discrete Aß oligomers can be generated by modulating the charge, composition, and chain length of lipids and surfactants. Here, we extend our studies into liposomal models by investigating Aß oligomerization on large unilamellar vesicles (LUVs) of total brain extracts (TBE), reconstituted lipid rafts (LRs), or 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC). Varying the vesicle composition by specifically increasing the amount of GM1 gangliosides as a constituent, we found that only GM1-enriched liposomes induce the formation of toxic, low-molecular-weight oligomers. Furthermore, we found that the aggregation on liposome surface and membrane disruption are highly cooperative and sensitive to membrane surface characteristics. Numerical simulations confirm such a cooperativity and reveal that GM1-enriched liposomes form twice as many pores as those formed in the absence GM1. Overall, this study uncovers mechanisms of cooperativity between oligomerization and membrane disruption under controlled lipid compositional bias, and refocuses the significance of the early stages of Aß aggregation in polymorphism, propagation, and toxicity in AD.


Assuntos
Doença de Alzheimer , Gangliosídeo G(M1) , Peptídeos beta-Amiloides/química , Dimiristoilfosfatidilcolina , Gangliosídeo G(M1)/química , Gangliosídeos , Humanos , Lipídeos de Membrana , Fosfolipídeos , Fosforilcolina , Tensoativos , Lipossomas Unilamelares/química
16.
J Phys Chem Lett ; 13(40): 9303-9308, 2022 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-36174129

RESUMO

A recently proposed lipid-chaperone hypothesis suggests that free lipid molecules, not bound to membranes, affect the aggregation of amyloidogenic peptides such as amyloid-ß (Aß) peptides, whose aggregates are the hallmarks of Alzheimer's disease. Here, we combine experiments with all-atom molecular dynamics simulations in explicit solvent to explore the effects of neuronal ganglioside GM1, abundant in mammalian brains, on the aggregation of two principal isoforms of Aß, Aß40 and Aß42. Our simulations show that free GM1 forms stable, highly water-soluble complexes with both isoforms, and nuclear magnetic resonance experiments support the formation of well-ordered, structurally compact GM1+Aß complexes. By simulation, we also show that Aß40 monomers display a preference for binding to GM1-containing hetero-oligomers over GM1-lacking homo-oligomers, while Aß42 monomers have the opposite preference. These observations explain why GM1 dose-dependently inhibits Aß40 aggregation but has no effect on Aß42 aggregation, as assessed by thioflavin T fluorescence.


Assuntos
Doença de Alzheimer , Gangliosídeo G(M1) , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Animais , Gangliosídeo G(M1)/química , Gangliosídeo G(M1)/metabolismo , Gangliosídeos/metabolismo , Mamíferos/metabolismo , Simulação de Dinâmica Molecular , Fragmentos de Peptídeos/química , Solventes , Água
17.
Eur J Med Chem ; 241: 114636, 2022 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-35952400

RESUMO

Ganglioside GM1 is a glycosphingolipid found on mammalian cell membranes, and it is involved in ischemic encephalopathy, spinal cord injury and neurodegenerative diseases. Fatty acids, as a structure module of GM1, have been reported to affect its physiological function and neurite growth activity. Due to the limitation of preparation methods, the function of GM1 derivatives containing different fatty acids in nerve cells has not been systematically studied. To discover novel GM1 derivatives as nerve growth-promoting agents, we developed an efficient SA_SCDase enzymatic synthetic system of GM1 derivatives, yielding twenty GM1 derivatives with unsaturated fatty acid chains in high total yields (16-67%). Subsequently, the neurite outgrowth activities of GM1 derivatives were assessed on Neuro2a Cells. Among all the GM1 derivatives, GM1 (d18:1/C16:1) induced demonstrably neurite outgrowth activity. The subsequent RNA-sequencing (RNA-seq) and Western blot analysis was then performed and indicated that the mechanism of nerve cells growth involved cholesterol biosynthesis regulation by up-regulating SREBP2 expression or ERBB4 phosphorylation to activate the PI3K-mTOR pathway.


Assuntos
Gangliosídeo G(M1) , Neuritos , Animais , Ácidos Graxos/farmacologia , Gangliosídeo G(M1)/química , Gangliosídeo G(M1)/metabolismo , Gangliosídeo G(M1)/farmacologia , Mamíferos/metabolismo , Neuritos/fisiologia , Crescimento Neuronal , Neurônios/metabolismo
18.
ACS Chem Neurosci ; 13(13): 1979-1991, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35713284

RESUMO

The aggregation of the amyloid beta (Aß) peptide is associated with Alzheimer's disease (AD) pathogenesis. Cell membrane composition, especially monosialotetrahexosylganglioside (GM1), is known to promote the formation of Aß fibrils, yet little is known about the roles of GM1 in the early steps of Aß oligomer formation. Here, by using GM1-contained liposomes as a mimic of the neuronal cell membrane, we demonstrate that GM1 is a critical trigger of Aß oligomerization and aggregation. We find that GM1 not only promotes the formation of Aß fibrils but also facilitates the maintenance of Aß42 oligomers on liposome membranes. We structurally characterize the Aß42 oligomers formed on the membrane and find that GM1 captures Aß by binding to its arginine-5 residue. To interrogate the mechanism of Aß42 oligomer toxicity, we design a new liposome-based Ca2+-encapsulation assay and provide new evidence for the Aß42 ion channel hypothesis. Finally, we determine the toxicity of Aß42 oligomers formed on membranes. Overall, by uncovering the roles of GM1 in mediating early Aß oligomer formation and maintenance, our work provides a novel direction for pharmaceutical research for AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Gangliosídeo G(M1)/química , Humanos , Lipossomos , Fragmentos de Peptídeos/metabolismo
19.
Nat Commun ; 12(1): 3675, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34135326

RESUMO

Gangliosides in the outer leaflet of the plasma membrane of eukaryotic cells are essential for many cellular functions and pathogenic interactions. How gangliosides are dynamically organized and how they respond to ligand binding is poorly understood. Using fluorescence anisotropy imaging of synthetic, fluorescently labeled GM1 gangliosides incorporated into the plasma membrane of living cells, we found that GM1 with a fully saturated C16:0 acyl chain, but not with unsaturated C16:1 acyl chain, is actively clustered into nanodomains, which depends on membrane cholesterol, phosphatidylserine and actin. The binding of cholera toxin B-subunit (CTxB) leads to enlarged membrane domains for both C16:0 and C16:1, owing to binding of multiple GM1 under a toxin, and clustering of CTxB. The structure of the ceramide acyl chain still affects these domains, as co-clustering with the glycosylphosphatidylinositol (GPI)-anchored protein CD59 occurs only when GM1 contains the fully saturated C16:0 acyl chain, and not C16:1. Thus, different ceramide species of GM1 gangliosides dictate their assembly into nanodomains and affect nanodomain structure and function, which likely underlies many endogenous cellular processes.


Assuntos
Membrana Celular/química , Ceramidas/química , Actinas/química , Antígenos CD59/química , Membrana Celular/efeitos dos fármacos , Toxina da Cólera/química , Toxina da Cólera/farmacologia , Colesterol/química , Gangliosídeo G(M1)/química , Glicoesfingolipídeos/química , Glicosilfosfatidilinositóis/química , Modelos Biológicos , Simulação de Dinâmica Molecular , Fosfatidilserinas/química
20.
Glycoconj J ; 38(1): 101-117, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33620588

RESUMO

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.


Assuntos
Gangliosídeo G(M1)/química , Gangliosídeo G(M1)/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Oligossacarídeos/química , Animais , Diferenciação Celular , Gangliosídeo G(M1)/farmacologia , Humanos , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Oligossacarídeos/síntese química , Oligossacarídeos/metabolismo , Receptor trkA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA