Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 241
Filtrar
1.
Nanoscale Horiz ; 9(7): 1175-1189, 2024 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-38689531

RESUMEN

The virulence of Staphylococcus aureus, a multi-drug resistant pathogen, notably depends on the expression of the phenol soluble modulins α3 (PSMα3) peptides, able to self-assemble into amyloid-like cross-α fibrils. Despite remarkable advances evidencing the crucial, yet insufficient, role of fibrils in PSMα3 cytotoxic activities towards host cells, the relationship between its molecular structures, assembly propensities, and modes of action remains an open intriguing problem. In this study, combining atomic force microscopy (AFM) imaging and infrared spectroscopy, we first demonstrated in vitro that the charge provided by the N-terminal capping of PSMα3 alters its interactions with model membranes of controlled lipid composition without compromising its fibrillation kinetics or morphology. N-formylation eventually dictates PSMα3-membrane binding via electrostatic interactions with the lipid head groups. Furthermore, PSMα3 insertion within the lipid bilayer is favoured by hydrophobic interactions with the lipid acyl chains only in the fluid phase of membranes and not in the gel-like ordered domains. Strikingly, our real-time AFM imaging emphasizes how intermediate protofibrillar entities, formed along PSMα3 self-assembly and promoted at the membrane interface, likely disrupt membrane integrity via peptide accumulation and subsequent membrane thinning in a peptide concentration and lipid-dependent manner. Overall, our multiscale and multimodal approach sheds new light on the key roles of N-formylation and intermediate self-assembling entities, rather than mature fibrils, in dictating deleterious interactions of PSMα3 with membrane lipids, likely underscoring its ultimate cellular toxicity in vivo, and in turn S. aureus pathogenesis.


Asunto(s)
Membrana Dobles de Lípidos , Microscopía de Fuerza Atómica , Staphylococcus aureus , Microscopía de Fuerza Atómica/métodos , Membrana Dobles de Lípidos/química , Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidad , Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Amiloide/química , Amiloide/toxicidad , Interacciones Hidrofóbicas e Hidrofílicas
2.
Adv Sci (Weinh) ; 11(23): e2310314, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582521

RESUMEN

Understanding the environmental health and safety of nanomaterials (NanoEHS) is essential for the sustained development of nanotechnology. Although extensive research over the past two decades has elucidated the phenomena, mechanisms, and implications of nanomaterials in cellular and organismal models, the active remediation of the adverse biological and environmental effects of nanomaterials remains largely unexplored. Inspired by recent developments in functional amyloids for biomedical and environmental engineering, this work shows their new utility as metallothionein mimics in the strategically important area of NanoEHS. Specifically, metal ions released from CuO and ZnO nanoparticles are sequestered through cysteine coordination and electrostatic interactions with beta-lactoglobulin (bLg) amyloid, as revealed by inductively coupled plasma mass spectrometry and molecular dynamics simulations. The toxicity of the metal oxide nanoparticles is subsequently mitigated by functional amyloids, as validated by cell viability and apoptosis assays in vitro and murine survival and biomarker assays in vivo. As bLg amyloid fibrils can be readily produced from whey in large quantities at a low cost, the study offers a crucial strategy for remediating the biological and environmental footprints of transition metal oxide nanomaterials.


Asunto(s)
Amiloide , Cobre , Animales , Ratones , Amiloide/metabolismo , Amiloide/química , Amiloide/toxicidad , Cobre/toxicidad , Cobre/química , Nanopartículas del Metal/toxicidad , Nanopartículas del Metal/química , Óxido de Zinc/toxicidad , Óxido de Zinc/química , Lactoglobulinas/química , Supervivencia Celular/efectos de los fármacos , Simulación de Dinámica Molecular , Humanos , Óxidos/toxicidad , Óxidos/química
3.
Neurochem Res ; 49(2): 379-387, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37847330

RESUMEN

Oligomeric aggregates of the amyloid-beta (Aß) peptide have been implicated as the toxic species for Alzheimer's disease by contributing to oxidative cytotoxicity and physical disruption in cell membranes in the brain. Recent evidence points to the ability of the catecholamine neurotransmitter dopamine in the presence of copper ions to both stabilize oligomers and decrease the toxic effects of these oligomers. Based on these results, physical characterization of aggregates and subsequent cell studies with a neuroblastoma line were performed that show both dopamine and the related neurotransmitter, norepinephrine, can stabilize oligomers and decrease toxicity of Aß aggregates without copper present. To investigate this reduction of toxicity, structural characterization of oligomers in the presence of neurotransmitters was compared to aggregates formed with Aß alone. Gel electrophoresis and transmission electron microscopy show higher levels of oligomers in the presence of dopamine and norepinephrine, yet the oligomer structure is largely amorphous. Aß aggregated alone forms the predicted highly organized fibrillar species, with increased levels of dityrosine covalent linkages, which are largely absent in the presence of the neurotransmitters. A proposed mechanism for the observed decrease in cell death by Aß in the presence of dopamine and norepinephrine suggests the neurotransmitters both block the formation of organized oligomer structures and dityrosine stabilizing linkages while also behaving as antioxidants, providing a dual mechanism for increased cell viability.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Humanos , Péptidos beta-Amiloides/metabolismo , Cobre/metabolismo , Dopamina , Enfermedad de Alzheimer/metabolismo , Catecoles , Norepinefrina , Neurotransmisores , Fragmentos de Péptidos/metabolismo , Amiloide/toxicidad
4.
FASEB J ; 37(7): e22972, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37302013

RESUMEN

Docosahexaenoic (DHA) and arachidonic acids (ARA) are omega-3 and omega-6 long-chain polyunsaturated fatty acids (LCPUFAs). These molecules constitute a substantial portion of phospholipids in plasma membranes. Therefore, both DHA and ARA are essential diet components. Once consumed, DHA and ARA can interact with a large variety of biomolecules, including proteins such as insulin and α-synuclein (α-Syn). Under pathological conditions known as injection amyloidosis and Parkinson's disease, these proteins aggregate forming amyloid oligomers and fibrils, toxic species that exert high cell toxicity. In this study, we investigate the role of DHA and ARA in the aggregation properties of α-Syn and insulin. We found that the presence of both DHA and ARA at the equimolar concentrations strongly accelerated aggregation rates of α-Syn and insulin. Furthermore, LCPUFAs substantially altered the secondary structure of protein aggregates, whereas no noticeable changes in the fibril morphology were observed. Nanoscale Infrared analysis of α-Syn and insulin fibrils grown in the presence of both DHA and ARA revealed the presence of LCPUFAs in these aggregates. We also found that such LCPUFAs-rich α-Syn and insulin fibrils exerted significantly greater toxicities compared to the aggregates grown in the LCPUFAs-free environment. These findings show that interactions between amyloid-associated proteins and LCPUFAs can be the underlying molecular cause of neurodegenerative diseases.


Asunto(s)
Ácidos Grasos Omega-3 , Enfermedad de Parkinson , Humanos , alfa-Sinucleína/metabolismo , Insulina , Amiloide/toxicidad , Amiloide/química , Ácidos Grasos Insaturados , Proteínas Amiloidogénicas , Ácidos Araquidónicos
5.
Sci Rep ; 12(1): 11898, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35831343

RESUMEN

α-Synuclein (α-Syn) aggregates are key components of intracellular inclusion bodies characteristic of Parkinson's disease (PD) and other synucleinopathies. Metal ions have been considered as the important etiological factors in PD since their interactions with α-Syn alter the kinetics of fibrillation. In the present study, we have systematically explored the effects of Zn2+, Cu2+, Ca2+, and Mg2+ cations on α-Syn fibril formation. Specifically, we determined fibrillation kinetics, size, morphology, and secondary structure of the fibrils and their cytotoxic activity. While all cations accelerate fibrillation, we observed distinct effects of the different ions. For example, Zn2+ induced fibrillation by lower tlag and higher kapp and formation of shorter fibrils, while Ca2+ ions lead to formation of longer fibrils, as evidenced by dynamic light scattering and atomic force microscopy studies. Additionally, the morphology of formed fibrils was different. Circular dichroism and attenuated total reflection-Fourier transform infrared spectroscopies revealed higher contents of ß-sheets in fibrils. Interestingly, cell viability studies indicated nontoxicity of α-Syn fibrils formed in the presence of Zn2+ ions, while the fibrils formed in the presence of Cu2+, Ca2+, and Mg2+ were cytotoxic. Our results revealed that α-Syn fibrils formed in the presence of different divalent cations have distinct structural and cytotoxic features.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Amiloide/química , Amiloide/toxicidad , Humanos , Iones , Metales , alfa-Sinucleína/química
6.
Cell Mol Life Sci ; 79(3): 174, 2022 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-35244787

RESUMEN

Protein misfolding is a general hallmark of protein deposition diseases, such as Alzheimer's disease or Parkinson's disease, in which different types of aggregated species (oligomers, protofibrils and fibrils) are generated by the cells. Despite widespread interest, the relationship between oligomers and fibrils in the aggregation process and spreading remains elusive. A large variety of experimental evidences supported the idea that soluble oligomeric species of different proteins might be more toxic than the larger fibrillar forms. Furthermore, the lack of correlation between the presence of the typical pathological inclusions and disease sustained this debate. However, recent data show that the ß-sheet core of the α-Synuclein (αSyn) fibrils is unable to establish persistent interactions with the lipid bilayers, but they can release oligomeric species responsible for an immediate dysfunction of the recipient neurons. Reversibly, such oligomeric species could also contribute to pathogenesis via neuron-to-neuron spreading by their direct cell-to-cell transfer or by generating new fibrils, following their neuronal uptake. In this Review, we discuss the various mechanisms of cellular dysfunction caused by αSyn, including oligomer toxicity, fibril toxicity and fibril spreading.


Asunto(s)
Amiloide/metabolismo , Sinucleinopatías/patología , alfa-Sinucleína/metabolismo , Amiloide/toxicidad , Humanos , Cuerpos de Lewy/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Agregado de Proteínas , Pliegue de Proteína , Sinucleinopatías/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/genética
7.
Biochem Biophys Res Commun ; 593: 46-51, 2022 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-35051782

RESUMEN

Cytotoxicity of amyloid fibrils has been shown to depend on their structure. However, specific features of toxic and non-toxic amyloids remain unclear. Here we focus on the relationship between structural characteristics of the fibrils and their cytotoxicity. Bovine carbonic anhydrase B (BCAB) serves as the object of this study because its amyloids reduce cell viability. Limited proteolysis and mass spectrometry were used to determine BCAB regions forming the core of amyloid fibrils. Four BCAB mutants with substitutions reducing hydrophobicity in the regions important for amyloid formation were obtained to study the kinetics of aggregation, structural features, and cytotoxicity of the amyloids. We demonstrate that fibrils of WT BCAB, L78A, L139A, and M239A variants display a pronounced toxic effect on eukaryotic cells, while I208A mutation significantly reduces the cell-damaging effect of amyloids. The data obtained conclude that cytotoxicity of BCAB fibrils does not depend on their length, secondary structure, and exposure of hydrophobic groups to the solvent. A distinctive feature of the low-toxic I208A fibrils is their specific morphology characterized by the lateral protofilaments association and formation of fibril-ribbons.


Asunto(s)
Amiloide/toxicidad , Anhidrasas Carbónicas/metabolismo , Fibroblastos/patología , Mutación , Proteolisis , Sustitución de Aminoácidos , Animales , Anhidrasas Carbónicas/química , Anhidrasas Carbónicas/genética , Bovinos , Fibroblastos/enzimología , Ratones , Pruebas de Toxicidad
8.
Cells ; 10(10)2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34685723

RESUMEN

Neurodegenerative diseases (NDs) are increasingly positioned as leading causes of global deaths. The accelerated aging of the population and its strong relationship with neurodegeneration forecast these pathologies as a huge global health problem in the upcoming years. In this scenario, there is an urgent need for understanding the basic molecular mechanisms associated with such diseases. A major molecular hallmark of most NDs is the accumulation of insoluble and toxic protein aggregates, known as amyloids, in extracellular or intracellular deposits. Here, we review the current knowledge on how molecular chaperones, and more specifically a ternary protein complex referred to as the human disaggregase, deals with amyloids. This machinery, composed of the constitutive Hsp70 (Hsc70), the class B J-protein DnaJB1 and the nucleotide exchange factor Apg2 (Hsp110), disassembles amyloids of α-synuclein implicated in Parkinson's disease as well as of other disease-associated proteins such as tau and huntingtin. We highlight recent studies that have led to the dissection of the mechanism used by this chaperone system to perform its disaggregase activity. We also discuss whether this chaperone-mediated disassembly mechanism could be used to solubilize other amyloidogenic substrates. Finally, we evaluate the implications of the chaperone system in amyloid clearance and associated toxicity, which could be critical for the development of new therapies.


Asunto(s)
Amiloide/metabolismo , Chaperonas Moleculares/metabolismo , Agregado de Proteínas , Amiloide/toxicidad , Humanos , Modelos Biológicos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , alfa-Sinucleína/metabolismo
9.
Int J Mol Sci ; 22(12)2021 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-34199259

RESUMEN

Beta2-microglobulin (B2M) a key component of major histocompatibility complex class I molecules, which aid cytotoxic T-lymphocyte (CTL) immune response. However, the majority of studies of B2M have focused only on amyloid fibrils in pathogenesis to the neglect of its role of antimicrobial activity. Indeed, B2M also plays an important role in innate defense and does not only function as an adjuvant for CTL response. A previous study discovered that human aggregated B2M binds the surface protein structure in Streptococci, and a similar study revealed that sB2M-9, derived from native B2M, functions as an antibacterial chemokine that binds Staphylococcus aureus. An investigation of sB2M-9 exhibiting an early lymphocyte recruitment in the human respiratory epithelium with bacterial challenge may uncover previously unrecognized aspects of B2M in the body's innate defense against Mycobactrium tuberculosis. B2M possesses antimicrobial activity that operates primarily under pH-dependent acidic conditions at which B2M and fragmented B2M may become a nucleus seed that triggers self-aggregation into distinct states, such as oligomers and amyloid fibrils. Modified B2M can act as an antimicrobial peptide (AMP) against a wide range of microbes. Specifically, these AMPs disrupt microbe membranes, a feature similar to that of amyloid fibril mediated cytotoxicity toward eukaryotes. This study investigated two similar but nonidentical effects of B2M: the physiological role of B2M, in which it potentially acts against microbes in innate defense and the role of B2M in amyloid fibrils, in which it disrupts the membrane of pathological cells. Moreover, we explored the pH-governing antibacterial activity of B2M and acidic pH mediated B2M amyloid fibrils underlying such cytotoxicity.


Asunto(s)
Amiloide/toxicidad , Antibacterianos/farmacología , Microglobulina beta-2/metabolismo , Secuencia de Aminoácidos , Animales , Muerte Celular/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Microglobulina beta-2/química
10.
Vet Med Sci ; 7(5): 1938-1947, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33978313

RESUMEN

The formation of amyloid fibrils is critical for neurodegenerative diseases. Some physiochemical conditions can promote the conversion of proteins from soluble globular shapes into insoluble well-organized amyloid fibrils. The aim of this study was to investigate the effect of temperatures on amyloid fibrils formation in vitro using the protein model of hen egg-white lysozyme (HEWL). The HEWL fibrils were prepared at temperatures of 37, 45, 50 and 57°C in glycine solution of pH 2.2. Under transmission electron microscopy, we found the well-organized HEWL amyloid fibrils at temperatures of 45, 50 and 57°C after 10 days of incubation. Thioflavin T and Congo red florescence assays confirmed that the formation and growth of HEWL fibrils displayed a temperature-dependent increase, and 57°C produced the most amounts. Meanwhile, the surface hydrophobicity of aggregates was greatly increased by ANS binding assay, and ß-sheet contents by circular dichroism analysis were increased by 17.8%, 22.0% and 34.9%, respectively. Furthermore, the HEWL fibrils formed at 57°C caused significant cytotoxicity in SH-SY5Y cells after 48 hr exposure, and the cell viability determined by MTT assay was decreased, with 81.35 ± 0.29% for 1 µM, 61.45 ± 2.62% for 2 µM, and 11.58 ± 0.39% (p < .01) for 3 µM. Nuclear staining results also confirmed the apoptosis features. These results suggest that the elevated temperatures could accelerate protein unfolding of the native structure and formation of toxic amyloid fibrils, which can improve understanding the mechanisms of the unfolding and misfolding process of prion protein.


Asunto(s)
Amiloide , Muramidasa , Amiloide/química , Amiloide/metabolismo , Amiloide/toxicidad , Animales , Dicroismo Circular/veterinaria , Microscopía Electrónica de Transmisión/veterinaria , Muramidasa/química , Muramidasa/metabolismo , Temperatura
11.
Int J Mol Sci ; 22(9)2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925514

RESUMEN

Various amyloid aggregates, in particular, aggregates of amyloid ß-proteins, demonstrate in vitro and in vivo cytotoxic effects associated with impairment of cell adhesion. We investigated the effect of amyloid aggregates of smooth-muscle titin on smooth-muscle-cell cultures. The aggregates were shown to impair cell adhesion, which was accompanied by disorganization of the actin cytoskeleton, formation of filopodia, lamellipodia, and stress fibers. Cells died after a 72-h contact with the amyloid aggregates. To understand the causes of impairment, we studied the effect of the microtopology of a titin-amyloid-aggregate-coated surface on fibroblast adhesion by atomic force microscopy. The calculated surface roughness values varied from 2.7 to 4.9 nm, which can be a cause of highly antiadhesive properties of this surface. As all amyloids have the similar structure and properties, it is quite likely that the antiadhesive effect is also intrinsic to amyloid aggregates of other proteins. These results are important for understanding the mechanisms of the negative effect of amyloids on cell adhesion.


Asunto(s)
Amiloide/toxicidad , Adhesión Celular/efectos de los fármacos , Conectina/química , Conectina/toxicidad , Músculo Liso/química , Actinas/metabolismo , Animales , Aorta/citología , Células Cultivadas , Pollos , Conectina/aislamiento & purificación , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Electroforesis en Gel de Poliacrilamida , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Humanos , Microscopía de Fuerza Atómica , Músculo Liso/citología , Agregado de Proteínas , Ratas
12.
ACS Appl Mater Interfaces ; 13(15): 18089-18099, 2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33829756

RESUMEN

Fibrillogenesis of amyloid ß-protein (Aß) is pathologically associated with Alzheimer's disease (AD), so modulating Aß aggregation is crucial for AD prevention and treatment. Herein, a zwitterionic polymer with short dimethyl side chains (pID) is synthesized and conjugated with a heptapeptide inhibitor (Ac-LVFFARK-NH2, LK7) to construct zwitterionic polymer-inhibitor conjugates for enhanced inhibition of Aß aggregation. However, it is unexpectedly found that the LK7@pID conjugates remarkably promote Aß fibrillization to form more fibrils than the free Aß system but effectively eliminate Aß-induced cytotoxicity. Such an unusual behavior of the LK7@pID conjugates is unraveled by extensive mechanistic studies. First, the hydrophobic environment within the assembled micelles of LK7@pID promotes the hydrophobic interaction between Aß molecules and LK7@pID, which triggers Aß aggregation at the very beginning, making fibrillization occur at an earlier stage. Second, in the aggregation process, the LK7@pID micelles disassemble by the intensive interactions with Aß, and LK7@pID participates in the fibrillization by being embedded in the Aß fibrils, leading to the formation of hybrid and heterogeneous fibrillar aggregates with a different structure than normal Aß fibrils. This unique Trojan horse-like feature of LK7@pID conjugates has not been observed for any other inhibitors reported previously and may shed light on the design of new modulators against ß-amyloid cytotoxicity.


Asunto(s)
Amiloide/química , Amiloide/toxicidad , Citotoxinas/química , Citotoxinas/toxicidad , Oligopéptidos/química , Polímeros/química , Polímeros/farmacología , Secuencia de Aminoácidos , Diseño de Fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Agregado de Proteínas/efectos de los fármacos
13.
J Mol Biol ; 433(10): 166953, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33771571

RESUMEN

Aberrant aggregation and amyloid formation of tar DNA binding protein (TDP-43) and α-synuclein (αS) underlie frontotemporal dementia (FTD) and Parkinson's disease (PD), respectively. Amyloid inclusions of TDP-43 and αS are also commonly co-observed in amyotrophic lateral sclerosis (ALS), dementia with Lewy bodies (DLB) and Alzheimer disease (AD). Emerging evidence from cellular and animal models show colocalization of the TDP-43 and αS aggregates, raising the possibility of direct interactions and co-aggregation between the two proteins. In this report, we set out to answer this question by investigating the interactions between αS and prion-like pathogenic C-terminal domain of TDP-43 (TDP-43 PrLD). PrLD is an aggregation-prone fragment generated both by alternative splicing as well as aberrant proteolytic cleavage of full length TDP-43. Our results indicate that two proteins interact in a synergistic manner to augment each other's aggregation towards hybrid fibrils. While monomers, oligomers and sonicated fibrils of αS seed TDP-43 PrLD monomers, TDP-43 PrLD fibrils failed to seed αS monomers indicating selectivity in interactions. Furthermore, αS modulates liquid droplets formed by TDP-43 PrLD and RNA to promote insoluble amyloid aggregates. Importantly, the cross-seeded hybrid aggregates show greater cytotoxicity as compared to the individual homotypic aggregates suggesting that the interactions between the two proteins have a discernable impact on cellular functions. Together, these results bring forth insights into TDP-43 PrLD - αS interactions that could help explain clinical and pathological presentations in patients with co-morbidities involving the two proteins.


Asunto(s)
Amiloide/química , Proteínas de Unión al ADN/química , Neuronas/efectos de los fármacos , ARN/química , alfa-Sinucleína/química , Empalme Alternativo , Amiloide/genética , Amiloide/metabolismo , Amiloide/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/toxicidad , Humanos , Gotas Lipídicas/química , Gotas Lipídicas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Priones/química , Priones/genética , Priones/metabolismo , Priones/toxicidad , Agregado de Proteínas/genética , Unión Proteica , Dominios Proteicos , Proteolisis , ARN/genética , ARN/metabolismo , Sonicación , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , alfa-Sinucleína/toxicidad
14.
J Chem Inf Model ; 61(2): 966-975, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33445870

RESUMEN

Soluble low-molecular-weight oligomers formed during the early stage of amyloid aggregation are considered the major toxic species in amyloidosis. The structure-function relationship between oligomeric assemblies and the cytotoxicity in amyloid diseases are still elusive due to the heterogeneous and transient nature of these aggregation intermediates. To uncover the structural characteristics of toxic oligomeric intermediates, we compared the self-assembly dynamics and structures of SOD128-38, a cytotoxic fragment of the superoxide dismutase 1 (SOD1) associated with the amyotrophic lateral sclerosis, with its two nontoxic mutants G33V and G33W using molecular dynamics simulations. Single-point glycine substitutions in SOD128-38 have been reported to abolish the amyloid toxicity. Our simulation results showed that the toxic SOD128-38 and its nontoxic mutants followed different aggregation pathways featuring distinct aggregation intermediates. Specifically, wild-type SOD128-38 initially self-assembled into random-coil-rich oligomers, among which fibrillar aggregates composed of well-defined curved single-layer ß-sheets were nucleated via coil-to-sheet conversions and the formation of ß-barrels as intermediates. In contrast, the nontoxic G33V/G33W mutants readily assembled into small ß-sheet-rich oligomers and then coagulated with each other into cross-ß fibrils formed by two-layer ß-sheets without forming ß-barrels as the intermediates. The direct observation of ß-barrel oligomers during the assembly of toxic SOD128-38 fragments but not the nontoxic glycine-substitution mutants strongly supports ß-barrels as the toxic oligomers in amyloidosis, probably via interactions with the cell membrane and forming amyloid pores. With well-defined structures, the ß-barrel might serve as a novel therapeutic target against amyloid-related diseases.


Asunto(s)
Glicina , Superóxido Dismutasa , Amiloide/toxicidad , Péptidos beta-Amiloides , Glicina/toxicidad , Conformación Proteica en Lámina beta , Superóxido Dismutasa/genética , Superóxido Dismutasa-1/genética
15.
Int J Biol Macromol ; 169: 532-540, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33352154

RESUMEN

A wide range of biophysical and theoretical analysis were employed to explore the formation of (α-syn) amyloid fibril formation as a model of Parkinson's disease in the presence of silica oxide nanoparticles (SiO2 NPs). Also, different cellular and molecular assays such as MTT, LDH, caspase, ROS, and qPCR were performed to reveal the α-syn amyloid fibrils-associated cytotoxicity against SH-SY5Y cells. Fluorescence measurements showed that SiO2 NPs accelerate the α-syn aggregation and exposure of hydrophobic moieties. Congo red absorbance, circular dichroism (CD), and transmission electron microscopy (TEM) analysis depicted the SiO2 NPs accelerated the formation of α-syn amyloid fibrils. Molecular docking study showed that SiO2 clusters preferably bind to the N-terminal of α-syn as the helix folding site. We also realized that SiO2 NPs increase the cytotoxicity of α-syn amyloid fibrils through a significant decrease in cell viability, increase in membrane leakage, activation of caspase-9 and -3, elevation of ROS, and increase in the ratio of Bax/Bcl2 mRNA. The cellular assay indicated that α-syn amyloid fibrils formed in the presence of SiO2 NPs induce their cytotoxic effects through the mitochondrial-mediated intrinsic apoptosis pathway. We concluded that these data may reveal some adverse effects of NPs on the progression of Parkinson's disease.


Asunto(s)
Amiloide/química , Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/química , Amiloide/metabolismo , Amiloide/toxicidad , Apoptosis/efectos de los fármacos , Benzotiazoles/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Cinética , Modelos Biológicos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Nanopartículas/química , Enfermedades Neurodegenerativas/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Dióxido de Silicio/química , Dióxido de Silicio/farmacología , Espectrometría de Fluorescencia/métodos , alfa-Sinucleína/metabolismo , alfa-Sinucleína/fisiología
16.
Int J Biol Macromol ; 170: 33-41, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33352157

RESUMEN

Fast green FCF (FGF) is often used in foods, pharmaceuticals, and cosmetics. However, little is known about the interactions of FGF with amyloid-ß protein (Aß) associated with Alzheimer's disease. In this study, the inhibitory effects of FGF on Aß fibrillogenesis, the disruption of preformed Aß fibrils, the reduction of Aß-induced cytotoxicity, and the attenuation of Aß-induced learning and memory impairments in mice were investigated. FGF significantly inhibited Aß fibrillogenesis and disintegrated the mature fibrils as evidenced by thioflavin T fluorescence and atomic force microscopy studies. Co-incubation of Aß with FGF greatly reduced Aß-induced cytotoxicity in vitro. Moreover, FGF showed a protective effect against cognitive impairment in Aß-treated mice. Molecular dynamics simulations further showed that FGF could synergistically interact with the Aß17-42 pentamer via electrostatic interactions, hydrogen bonds and π-π interactions, which reduced the ß-sheet content, and disordered random coils and bend structures of the Aß17-42 pentamer. This study offers a comprehensive understanding of the inhibitory effects of FGF against Aß neurotoxicity, which is critical for the search of effective food additives that can combat amyloid-associated disease.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Amiloide/antagonistas & inhibidores , Disfunción Cognitiva/prevención & control , Aditivos Alimentarios/uso terapéutico , Colorantes Verde de Lisamina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Agregación Patológica de Proteínas/prevención & control , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Amiloide/efectos de los fármacos , Amiloide/toxicidad , Amiloide/ultraestructura , Péptidos beta-Amiloides/química , Animales , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Conducta Exploratoria/efectos de los fármacos , Aditivos Alimentarios/farmacología , Humanos , Enlace de Hidrógeno , Colorantes Verde de Lisamina/farmacología , Ratones , Microscopía de Fuerza Atómica , Modelos Moleculares , Simulación de Dinámica Molecular , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/efectos de los fármacos , Agregación Patológica de Proteínas/tratamiento farmacológico , Estructura Secundaria de Proteína/efectos de los fármacos , Distribución Aleatoria , Electricidad Estática
17.
Eur J Pharmacol ; 889: 173522, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-32866503

RESUMEN

The complications of Alzheimer's disease (AD) have made the development of its treatment a challenging task. Several studies have indicated the disruption of insulin receptor substrate-1 (IRS-1) signaling during the development and progression of AD. The role of a dipeptidyl peptidase-4 (DPP-4) inhibitor on hippocampal IRS-1 signaling has not been investigated before. In this study, we evaluated the efficacy of alogliptin (DPP-4 inhibitor) on hippocampal insulin resistance and associated AD complications. In the present study, amyloid-ß (1-42) fibrils were produced and administered intrahippocampally for inducing AD in Wistar rats. After 7 days of surgery, rats were treated with 10 and 20 mg/kg of alogliptin for 28 days. Morris water maze (MWM) test was performed in the last week of our experimental study. Post 24 h of final treatment, rats were euthanized and hippocampi were separated for biochemical and histopathological investigations. In-silico analysis revealed that alogliptin has a good binding affinity with Aß and beta-secretase-1 (BACE-1). Alogliptin significantly restored cognitive functions in Aß (1-42) fibrils injected rats during the MWM test. Alogliptin also significantly attenuated insulin level, IRS-1pS307 expression, Aß (1-42) level, GSK-3ß activity, TNF-α level and oxidative stress in the hippocampus. The histopathological analysis supported alogliptin mediated neuroprotective and anti-amyloidogenic effect. Immunohistochemical analysis also revealed a reduction in IRS-1pS307 expression after alogliptin treatment. The in-silico, behavioral, biochemical and histopathological analysis supports the protective effect of alogliptin against hippocampal insulin resistance and AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Resistencia a la Insulina/fisiología , Fragmentos de Péptidos/toxicidad , Piperidinas/uso terapéutico , Uracilo/análogos & derivados , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Amiloide/metabolismo , Amiloide/toxicidad , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Hipocampo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Piperidinas/farmacología , Distribución Aleatoria , Ratas , Ratas Wistar , Uracilo/farmacología , Uracilo/uso terapéutico
18.
FEBS Lett ; 594(21): 3490-3503, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32871611

RESUMEN

Amyloid fibrils found in plaques in Alzheimer's disease (AD) brains are composed of amyloid-ß peptides. Oligomeric amyloid-ß 1-42 (Aß42) is thought to play a critical role in neurodegeneration in AD. Here, we determine how size and conformation affect neurotoxicity and internalisation of Aß42 assemblies using biophysical methods, immunoblotting, toxicity assays and live-cell imaging. We report significant cytotoxicity of Aß42 oligomers and their internalisation into neurons. In contrast, Aß42 fibrils show reduced internalisation and no toxicity. Sonicating Aß42 fibrils generates species similar in size to oligomers but remains nontoxic. The results suggest that Aß42 oligomers have unique properties that underlie their neurotoxic potential. Furthermore, we show that incubating cells with Aß42 oligomers for 24 h is sufficient to trigger irreversible neurotoxicity.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Fragmentos de Péptidos/química , Fragmentos de Péptidos/toxicidad , Agregación Patológica de Proteínas , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/química , Amiloide/metabolismo , Amiloide/farmacología , Amiloide/toxicidad , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacocinética , Supervivencia Celular/efectos de los fármacos , Humanos , Peso Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacocinética , Conformación Proteica , Sonicación
19.
Biophys J ; 119(2): 326-336, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32579964

RESUMEN

Amyloid aggregates have been demonstrated to exert cytotoxic effects in several diseases. It is widely accepted that the complex and fascinating aggregation pathway involves a series of steps during which many heterogeneous intermediates are generated. This process may be greatly potentiated by the presence of amphipathic components of plasma membrane because they may serve as interaction, condensation, and nucleation points. However, there are few data regarding structural alterations induced by the binding between the amyloid fibrils and membrane components and its direct effects on cell integrity. In this study, we found, by 1-anilinonaphthalene 8-sulfonic acid and transmission electron microscopy/fast Fourier transform, that yeast prion Sup35 oligomers showed higher structural uniformity and altered surface properties when grown in the presence of monosialotetrahexosylganglioside, a component of the cell membrane. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and confocal/sensitized Förster resonance energy transfer analyses revealed that these fibrils showed low cytotoxicity and affinity to plasma membrane. Moreover, time-lapse analysis of Sup35 oligomer fibrillation on cells suggested that the amyloid aggregation process per se exerts cytotoxic effects through the interaction of amyloid intermediates with plasma membrane components. These data provide, to our knowledge, new insights to understand the mechanism of amyloid growth and cytotoxicity in the pathogenesis of amyloid diseases.


Asunto(s)
Amiloide , Proteínas de Saccharomyces cerevisiae , Amiloide/toxicidad , Membrana Celular , Gangliósido G(M1) , Factores de Terminación de Péptidos , Saccharomyces cerevisiae
20.
Biochim Biophys Acta Biomembr ; 1862(6): 183251, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32113849

RESUMEN

Extensive research has shown that assembling of α-synuclein amyloid aggregates on mitochondria is an important mechanistic feature of Parkinson's disease (PD) and other Lewy body disorders. However, the molecular mechanism(s) of its neuronal toxicity remain unclear. Type 1 Hexokinase (HKI), a key enzyme in the control of brain glucose metabolism, plays an important role in protecting against mitochondrially-regulated apoptosis through reducing generation of reactive oxygen species (ROS). The release of mitochondrially-bound HKI causes a significant decrease in enzyme activity and triggers oxidative stress. Here, we have investigated the potency of amyloid fibrillation products arising from α-synuclein and hen egg white lysozyme (HEWL) for the release of HKI and ROS content enhancement in mitochondria isolated from rat brain. Results clearly indicate the capacity of the fibrillation products of α-synuclein, and not HEWL, to trigger release of HKI from the Type A binding site of mitochondria for the enzyme and to induce mitochondrial ROS enhancement in a dose-dependent manner. Moreover, we found that curcumin was very effective in preventing mitochondrial HKI release and ROS enhancement induced by α-synuclein fibrillation products. The pathophysiological significance of mitochondrial HKI activity and localization in pathogenesis of neurodegenerative disorders including PD are discussed. Taken together, these results may offer insight into a possible mechanism by which disease-related peptides and proteins may exert their neuronal toxicity.


Asunto(s)
Amiloide/toxicidad , Curcumina/farmacología , Hexoquinasa/metabolismo , Mitocondrias/metabolismo , Enfermedad de Parkinson/etiología , alfa-Sinucleína/química , Amiloide/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Pollos , Humanos , Muramidasa , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , alfa-Sinucleína/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...