Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biol Chem ; 295(21): 7224-7234, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32241918

RESUMEN

A detailed understanding of the molecular pathways for amyloid-ß (Aß) peptide aggregation from monomers into amyloid fibrils, a hallmark of Alzheimer's disease, is crucial for the development of diagnostic and therapeutic strategies. We investigate the molecular details of peptide fibrillization in vitro by perturbing this process through addition of differently charged metal ions. Here, we used a monovalent probe, the silver ion, that, similarly to divalent metal ions, binds to monomeric Aß peptide and efficiently modulates Aß fibrillization. On the basis of our findings, combined with our previous results on divalent zinc ions, we propose a model that links the microscopic metal-ion binding to Aß monomers to its macroscopic impact on the peptide self-assembly observed in bulk experiments. We found that substoichiometric concentrations of the investigated metal ions bind specifically to the N-terminal region of Aß, forming a dynamic, partially compact complex. The metal-ion bound state appears to be incapable of aggregation, effectively reducing the available monomeric Aß pool for incorporation into fibrils. This is especially reflected in a decreased fibril-end elongation rate. However, because the bound state is significantly less stable than the amyloid state, Aß peptides are only transiently redirected from fibril formation, and eventually almost all Aß monomers are integrated into fibrils. Taken together, these findings unravel the mechanistic consequences of delaying Aß aggregation via weak metal-ion binding, quantitatively linking the contributions of specific interactions of metal ions with monomeric Aß to their effects on bulk aggregation.


Asunto(s)
Péptidos beta-Amiloides/química , Metales/química , Agregado de Proteínas , Enfermedad de Alzheimer , Humanos , Unión Proteica , Dominios Proteicos
2.
Biochem Biophys Res Commun ; 510(4): 520-524, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30737030

RESUMEN

Dysregulation and aggregation of the peptide hormone IAPP (islet amyloid polypeptide, a.k.a. amylin) into soluble oligomers that appear to be cell-toxic is a known aspect of diabetes mellitus (DM) Type 2 pathology. IAPP aggregation is influenced by several factors including interactions with metal ions such as Cu(II). Because Cu(II) ions are redox-active they may contribute to metal-catalyzed formation of oxidative tyrosyl radicals, which can generate dityrosine cross-links. Here, we show that such a process, which involves Cu(II) ions bound to the IAPP peptide together with H2O2, can induce formation of large amounts of IAPP dimers connected by covalent dityrosine cross-links. This cross-linking is less pronounced at low pH and for murine IAPP, likely due to less efficient Cu(II) binding. Whether IAPP can carry out its hormonal function as a cross-linked dimer is unknown. As dityrosine concentrations are higher in blood plasma of DM Type 2 patients - arguably due to disease-related oxidative stress - and as dimer formation is the first step in protein aggregation, generation of dityrosine-linked dimers may be an important factor in IAPP aggregation and thus relevant for DM Type 2 progression.


Asunto(s)
Cobre/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Multimerización de Proteína , Tirosina/análogos & derivados , Animales , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/química , Ratones , Tirosina/análisis , Tirosina/metabolismo
3.
J Biol Inorg Chem ; 24(8): 1189-1196, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31562546

RESUMEN

The amyloid-ß (Aß) peptides are key molecules in Alzheimer's disease (AD) pathology. They interact with cellular membranes, and can bind metal ions outside the membrane. Certain oligomeric Aß aggregates are known to induce membrane perturbations and the structure of these oligomers-and their membrane-perturbing effects-can be modulated by metal ion binding. If the bound metal ions are redox active, as e.g., Cu and Fe ions are, they will generate harmful reactive oxygen species (ROS) just outside the membrane surface. Thus, the membrane damage incurred by toxic Aß oligomers is likely aggravated when redox-active metal ions are present. The combined interactions between Aß oligomers, metal ions, and biomembranes may be responsible for at least some of the neuronal death in AD patients.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Membrana Celular/metabolismo , Cobre/metabolismo , Hierro/metabolismo , Enfermedad de Alzheimer/metabolismo , Secuencia de Aminoácidos , Cobre/química , Humanos , Hierro/química , Unión Proteica , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/metabolismo
4.
J Am Chem Soc ; 140(26): 8138-8146, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29708745

RESUMEN

In Alzheimer's disease, amyloid-ß (Aß) plaques and tau neurofibrillary tangles are the two pathological hallmarks. The co-occurrence and combined reciprocal pathological effects of Aß and tau protein aggregation have been observed in animal models of the disease. However, the molecular mechanism of their interaction remain unknown. Using a variety of biophysical measurements, we here show that the native full-length tau protein solubilizes the Aß40 peptide and prevents its fibrillation. The tau protein delays the amyloid fibrillation of the Aß40 peptide at substoichiometric ratios, showing different binding affinities toward the different stages of the aggregated Aß40 peptides. The Aß monomer structure remains random coil in the presence of tau, as observed by nuclear magnetic resonance (NMR), circular dichroism (CD) spectroscopy and photoinduced cross-linking methods. We propose a potential interaction mechanism for the influence of tau on Aß fibrillation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Péptidos beta-Amiloides/química , Dicroismo Circular , Humanos , Microscopía de Fuerza Atómica , Resonancia Magnética Nuclear Biomolecular , Tamaño de la Partícula , Propiedades de Superficie , Proteínas tau/química
5.
J Neuroinflammation ; 15(1): 172, 2018 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-29866153

RESUMEN

BACKGROUND: Chronic neuroinflammation is a hallmark of Parkinson's disease (PD) pathophysiology, associated with increased levels of pro-inflammatory factors in PD brain tissues. The pro-inflammatory mediator and highly amyloidogenic protein S100A9 is involved in the amyloid-neuroinflammatory cascade in Alzheimer's disease. This is the first report on the co-aggregation of α-synuclein (α-syn) and S100A9 both in vitro and ex vivo in PD brain. METHODS: Single and sequential immunohistochemistry, immunofluorescence, scanning electron and atomic force (AFM) microscopies were used to analyze the ex vivo PD brain tissues for S100A9 and α-syn location and aggregation. In vitro studies revealing S100A9 and α-syn interaction and co-aggregation were conducted by NMR, circular dichroism, Thioflavin-T fluorescence, AFM, and surface plasmon resonance methods. RESULTS: Co-localized and co-aggregated S100A9 and α-syn were found in 20% Lewy bodies and 77% neuronal cells in the substantia nigra; both proteins were also observed in Lewy bodies in PD frontal lobe (Braak stages 4-6). Lewy bodies were characterized by ca. 10-23 µm outer diameter, with S100A9 and α-syn being co-localized in the same lamellar structures. S100A9 was also detected in neurons and blood vessels of the aged patients without PD, but in much lesser extent. In vitro S100A9 and α-syn were shown to interact with each other via the α-syn C-terminus with an apparent dissociation constant of ca. 5 µM. Their co-aggregation occurred significantly faster and led to formation of larger amyloid aggregates than the self-assembly of individual proteins. S100A9 amyloid oligomers were more toxic than those of α-syn, while co-aggregation of both proteins mitigated the cytotoxicity of S100A9 oligomers. CONCLUSIONS: We suggest that sustained neuroinflammation promoting the spread of amyloidogenic S100A9 in the brain tissues may trigger the amyloid cascade involving α-syn and S100A9 and leading to PD, similar to the effect of S100A9 and Aß co-aggregation in Alzheimer's disease. The finding of S100A9 involvement in PD may open a new avenue for therapeutic interventions targeting S100A9 and preventing its amyloid self-assembly in affected brain tissues.


Asunto(s)
Encéfalo/metabolismo , Calgranulina B/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Agregado de Proteínas/fisiología , alfa-Sinucleína/metabolismo , Anciano , Anciano de 80 o más Años , Amiloide/metabolismo , Amiloide/ultraestructura , Autopsia , Encéfalo/diagnóstico por imagen , Encéfalo/ultraestructura , Calgranulina B/farmacología , Línea Celular Tumoral , Dicroismo Circular/métodos , Femenino , Humanos , Cuerpos de Lewy/patología , Cuerpos de Lewy/ultraestructura , Espectroscopía de Resonancia Magnética , Masculino , Microscopía Electrónica de Rastreo , Neuroblastoma/patología , Enfermedad de Parkinson/diagnóstico por imagen , Estadísticas no Paramétricas , Resonancia por Plasmón de Superficie , alfa-Sinucleína/farmacología
6.
Environ Health ; 16(1): 61, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28641573

RESUMEN

BACKGROUND: Polycyclic aromatic hydrocarbons (PAHs) are the main toxic compounds in natural bitumen, a fossil material used by modern and ancient societies around the world. The adverse health effects of PAHs on modern humans are well established, but their health impacts on past populations are unclear. It has previously been suggested that a prehistoric health decline among the native people living on the California Channel Islands may have been related to PAH exposure. Here, we assess the potential health risks of PAH exposure from the use and manufacture of bitumen-coated water bottles by ancient California Indian societies. METHODS: We replicated prehistoric bitumen-coated water bottles with traditional materials and techniques of California Indians, based on ethnographic and archaeological evidence. In order to estimate PAH exposure related to water bottle manufacture and use, we conducted controlled experiments to measure PAH contamination 1) in air during the manufacturing process and 2) in water and olive oil stored in a completed bottle for varying periods of time. Samples were analyzed with gas chromatography/mass spectrometry (GC/MS) for concentrations of the 16 PAHs identified by the US Environmental Protection Agency (EPA) as priority pollutants. RESULTS: Eight PAHs were detected in concentrations of 1-10 µg/m3 in air during bottle production and 50-900 ng/L in water after 2 months of storage, ranging from two-ring (naphthalene and methylnaphthalene) to four-ring (fluoranthene) molecules. All 16 PAHs analyzed were detected in olive oil after 2 days (2 to 35 µg/kg), 2 weeks (3 to 66 µg/kg), and 2 months (5 to 140 µg/kg) of storage. CONCLUSIONS: For ancient California Indians, water stored in bitumen-coated water bottles was not a significant source of PAH exposure, but production of such bottles could have resulted in harmful airborne PAH exposure.


Asunto(s)
Contaminantes Atmosféricos/historia , Agua Potable/análisis , Exposición a Riesgos Ambientales/historia , Hidrocarburos , Hidrocarburos Policíclicos Aromáticos/historia , Grupos de Población/historia , Contaminantes Químicos del Agua/historia , Contaminantes Atmosféricos/análisis , California , Exposición a Riesgos Ambientales/análisis , Historia Antigua , Humanos , Hidrocarburos Policíclicos Aromáticos/análisis , Medición de Riesgo , Contaminantes Químicos del Agua/análisis
7.
Chemistry ; 22(40): 14236-46, 2016 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-27539220

RESUMEN

Many peptides and proteins with large sequences and structural differences self-assemble into disease-causing amyloids that share very similar biochemical and biophysical characteristics, which may contribute to their cross-interaction. Here, we demonstrate how the self-assembled, cyclic d,l-α-peptide CP-2, which has similar structural and functional properties to those of amyloids, acts as a generic inhibitor of the Parkinson's disease associated α-synuclein (α-syn) aggregation to toxic oligomers by an "off-pathway" mechanism. We show that CP-2 interacts with the N-terminal and the non-amyloid-ß component region of α-syn, which are responsible for α-syn's membrane intercalation and self-assembly, thus changing the overall conformation of α-syn. CP-2 also remodels α-syn fibrils to nontoxic amorphous species and permeates cells through endosomes/lysosomes to reduce the accumulation and toxicity of intracellular α-syn in neuronal cells overexpressing α-syn. Our studies suggest that targeting the common structural conformation of amyloids may be a promising approach for developing new therapeutics for amyloidogenic diseases.


Asunto(s)
Enfermedad de Parkinson/tratamiento farmacológico , Péptidos Cíclicos/farmacología , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/tratamiento farmacológico , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Amiloide/ultraestructura , Animales , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células PC12 , Enfermedad de Parkinson/metabolismo , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacocinética , Agregación Patológica de Proteínas/metabolismo , Ratas , alfa-Sinucleína/ultraestructura
8.
Comput Struct Biotechnol J ; 20: 5672-5679, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36284704

RESUMEN

Amyloid ß-peptide (Aß) misfolding into ß-sheet structures triggers neurotoxicity inducing Alzheimer's disease (AD). Molecules able to reduce or to impair Aß aggregation are highly relevant as possible AD treatments since they should protect against Aß neurotoxicity. We have studied the effects of the interaction of dynorphins, a family of opioid neuropeptides, with Aß40 the most abundant species of Aß. Biophysical measurements indicate that Aß40 interacts with Big Dynorphin (BigDyn), lowering the amount of hydrophobic aggregates, and slowing down the aggregation kinetics. As expected, we found that BigDyn protects against Aß40 aggregates when studied in human neuroblastoma cells by cell survival assays. The cross-interaction between BigDyn and Aß40 provides insight into the mechanism of amyloid pathophysiology and may open up new therapy possibilities.

9.
J Mol Biol ; 433(2): 166717, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33220262

RESUMEN

Heat shock protein 90 (Hsp90) is a molecular chaperone that assists protein folding in an Adenosine triphosphate (ATP)-dependent way. Hsp90 has been reported to interact with Alzheimers disease associated amyloid-ß (Aß) peptides and to suppress toxic oligomer- and fibril formation. However, the mechanism remains largely unclear. Here we use a combination of atomic force microscopy (AFM) imaging, circular dichroism (CD) spectroscopy and biochemical analysis to quantify this interaction and put forward a microscopic picture including rate constants for the different transitions towards fibrillation. We show that Hsp90 binds to Aß40 monomers weakly but inhibits Aß40 from growing into fibrils at substoichiometric concentrations. ATP impedes this interaction, presumably by modulating Hsp90's conformational dynamics and reducing its hydrophobic surface. Altogether, these results might indicate alternative ways to prevent Aß40 fibrillation by manipulating chaperones that are already abundant in the brain.


Asunto(s)
Adenosina Trifosfato/química , Péptidos beta-Amiloides/química , Amiloide/química , Proteínas HSP90 de Choque Térmico/química , Fragmentos de Péptidos/química , Adenosina Trifosfato/metabolismo , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Microscopía de Fuerza Atómica , Modelos Moleculares , Fragmentos de Péptidos/metabolismo , Agregado de Proteínas , Unión Proteica , Conformación Proteica , Proteínas Recombinantes , Análisis Espectral , Relación Estructura-Actividad
10.
Sci Rep ; 10(1): 2376, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32047184

RESUMEN

Arginine residues of the antimicrobial peptide LL-37 can be citrullinated by peptidyl arginine deiminases, which reduce the positive charge of the peptide. Notably, citrullinated LL-37 has not yet been detected in human samples. In addition, functional and biophysical properties of citrullinated LL-37 are not fully explored. The aim of this study was to detect citrullinated LL-37 in human bronchoalveolar lavage (BAL) fluid and to determine antibacterial and biophysical properties of citrullinated LL-37. BAL fluid was obtained from healthy human volunteers after intra-bronchial exposure to lipopolysaccharide. Synthetic peptides were used for bacterial killing assays, transmission electron microscopy, isothermal titration calorimetry, mass-spectrometry and circular dichroism. Using targeted proteomics, we were able to detect both native and citrullinated LL-37 in BAL fluid. The citrullinated peptide did not kill Escherichia coli nor lysed human red blood cells. Both peptides had similar α-helical secondary structures but citrullinated LL-37 was more stable at higher temperatures, as shown by circular dichroism. In conclusion, citrullinated LL-37 is present in the human airways and citrullination impaired bacterial killing, indicating that a net positive charge is important for antibacterial and membrane lysing effects. It is possible that citrullination serves as a homeostatic regulator of AMP-function by alteration of key functions.


Asunto(s)
Antibacterianos/farmacología , Catelicidinas/farmacología , Antibacterianos/análisis , Antibacterianos/química , Péptidos Catiónicos Antimicrobianos , Líquido del Lavado Bronquioalveolar/química , Catelicidinas/análisis , Catelicidinas/química , Células Cultivadas , Citrulina/análogos & derivados , Eritrocitos/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Humanos , Conformación Proteica en Hélice alfa , Estabilidad Proteica
11.
Biomolecules ; 10(1)2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31892131

RESUMEN

Brains and blood of Alzheimer's disease (AD) patients have shown elevated mercury concentrations, but potential involvement of mercury exposure in AD pathogenesis has not been studied at the molecular level. The pathological hallmark of AD brains is deposition of amyloid plaques, consisting mainly of amyloid-ß (Aß) peptides aggregated into amyloid fibrils. Aß peptide fibrillization is known to be modulated by metal ions such as Cu(II) and Zn(II). Here, we study in vitro the interactions between Aß peptides and Hg(II) ions by multiple biophysical techniques. Fluorescence spectroscopy and atomic force microscopy (AFM) show that Hg(II) ions have a concentration-dependent inhibiting effect on Aß fibrillization: at a 1:1 Aß·Hg(II) ratio only non-fibrillar Aß aggregates are formed. NMR spectroscopy shows that Hg(II) ions interact with the N-terminal region of Aß(1-40) with a micromolar affinity, likely via a binding mode similar to that for Cu(II) and Zn(II) ions, i.e., mainly via the histidine residues His6, His13, and His14. Thus, together with Cu(II), Fe(II), Mn(II), Pb(IV), and Zn(II) ions, Hg(II) belongs to a family of metal ions that display residue-specific binding interactions with Aß peptides and modulate their aggregation processes.


Asunto(s)
Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Mercurio/farmacología , Agregado de Proteínas/efectos de los fármacos , Enfermedad de Alzheimer/diagnóstico , Péptidos beta-Amiloides/metabolismo , Sitios de Unión/efectos de los fármacos , Humanos , Iones/química , Iones/farmacología , Espectroscopía de Resonancia Magnética , Mercurio/química , Microscopía de Fuerza Atómica , Espectrometría de Fluorescencia
12.
ACS Chem Biol ; 14(7): 1410-1417, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31194501

RESUMEN

Amyloid cascade and neuroinflammation are hallmarks of neurodegenerative diseases, and pro-inflammatory S100A9 protein is central to both of them. Here, we have shown that NCAM1 peptide constructs carrying polycationic sequences derived from Aß peptide (KKLVFF) and PrP protein (KKRPKP) significantly promote the S100A9 amyloid self-assembly in a concentration-dependent manner by making transient interactions with individual S100A9 molecules, perturbing its native structure and acting as catalysts. Since the individual molecule misfolding is a rate-limiting step in S100A9 amyloid aggregation, the effects of the NCAM1 construct on the native S100A9 are so critical for its amyloid self-assembly. S100A9 rapid self-assembly into large aggregated clumps may prevent its amyloid tissue propagation, and by modulating S100A9 aggregation as a part of the amyloid cascade, the whole process may be effectively tuned.


Asunto(s)
Amiloide/inmunología , Antígeno CD56/inmunología , Calgranulina B/inmunología , Agregación Patológica de Proteínas/inmunología , Secuencia de Aminoácidos , Amiloide/química , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/inmunología , Antígeno CD56/química , Calgranulina B/química , Humanos , Inflamación/inmunología , Modelos Moleculares , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Priones/química , Priones/inmunología , Agregado de Proteínas
13.
J Mol Biol ; 430(3): 348-362, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29183787

RESUMEN

Proteolysis plays an important role in mitochondrial biogenesis, from the processing of newly imported precursor proteins to the degradation of mitochondrial targeting peptides. Disruption of peptide degradation activity in yeast, plant and mammalian mitochondria is known to have deleterious consequences for organism physiology, highlighting the important role of mitochondrial peptidases. In the present work, we show that the human mitochondrial peptidase neurolysin (hNLN) can degrade mitochondrial presequence peptides as well as other fragments up to 19 amino acids long. The crystal structure of hNLNE475Q in complex with the products of neurotensin cleavage at 2.7Å revealed a closed conformation with an internal cavity that restricts substrate length and highlighted the mechanism of enzyme opening/closing that is necessary for substrate binding and catalytic activity. Analysis of peptide degradation in vitro showed that hNLN cooperates with presequence protease (PreP or PITRM1) in the degradation of long targeting peptides and amyloid-ß peptide, Aß1-40, associated with Alzheimer disease, particularly cleaving the hydrophobic fragment Aß35-40. These findings suggest that a network of proteases may be required for complete degradation of peptides localized in mitochondria.


Asunto(s)
Metaloendopeptidasas/metabolismo , Mitocondrias/metabolismo , Péptidos/metabolismo , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Cristalografía por Rayos X , Células HeLa , Humanos , Metaloendopeptidasas/química , Ratones Endogámicos C57BL , Modelos Moleculares , Neurotensina/química , Neurotensina/metabolismo , Péptidos/química , Unión Proteica , Conformación Proteica , Proteolisis , Especificidad por Sustrato
14.
ACS Chem Neurosci ; 9(7): 1680-1692, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-29683649

RESUMEN

The amphiphilic nature of the amyloid-ß (Aß) peptide associated with Alzheimer's disease facilitates various interactions with biomolecules such as lipids and proteins, with effects on both structure and toxicity of the peptide. Here, we investigate these peptide-amphiphile interactions by experimental and computational studies of Aß(1-40) in the presence of surfactants with varying physicochemical properties. Our findings indicate that electrostatic peptide-surfactant interactions are required for coclustering and structure induction in the peptide and that the strength of the interaction depends on the surfactant net charge. Both aggregation-prone peptide-rich coclusters and stable surfactant-rich coclusters can form. Only Aß(1-40) monomers, but not oligomers, are inserted into surfactant micelles in this surfactant-rich state. Surfactant headgroup charge is suggested to be important as electrostatic peptide-surfactant interactions on the micellar surface seems to be an initiating step toward insertion. Thus, no peptide insertion or change in peptide secondary structure is observed using a nonionic surfactant. The hydrophobic peptide-surfactant interactions instead stabilize the Aß monomer, possibly by preventing self-interaction between the peptide core and C-terminus, thereby effectively inhibiting the peptide aggregation process. These findings give increased understanding regarding the molecular driving forces for Aß aggregation and the peptide interaction with amphiphilic biomolecules.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Tensoactivos/farmacología , Péptidos beta-Amiloides/química , Animales , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Micelas , Simulación de Dinámica Molecular , Agregación Patológica de Proteínas/tratamiento farmacológico , Agregación Patológica de Proteínas/metabolismo , Estructura Secundaria de Proteína , Electricidad Estática , Tensoactivos/química
15.
Sci Rep ; 7(1): 14423, 2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29089568

RESUMEN

Cigarette smoking is a significant risk factor for Alzheimer's disease (AD), which is associated with extracellular brain deposits of amyloid plaques containing aggregated amyloid-ß (Aß) peptides. Aß aggregation occurs via multiple pathways that can be influenced by various compounds. Here, we used AFM imaging and NMR, fluorescence, and mass spectrometry to monitor in vitro how Aß aggregation is affected by the cigarette-related compounds nicotine, polycyclic aromatic hydrocarbons (PAHs) with one to five aromatic rings, and the metal ions Cd(II), Cr(III), Pb(II), and Pb(IV). All PAHs and metal ions modulated the Aß aggregation process. Cd(II), Cr(III), and Pb(II) ions displayed general electrostatic interactions with Aß, whereas Pb(IV) ions showed specific transient binding coordination to the N-terminal Aß segment. Thus, Pb(IV) ions are especially prone to interact with Aß and affect its aggregation. While Pb(IV) ions affected mainly Aß dimer and trimer formation, hydrophobic toluene mainly affected formation of larger aggregates such as tetramers. The uncharged and hydrophilic nicotine molecule showed no direct interactions with Aß, nor did it affect Aß aggregation. Our Aß interaction results suggest a molecular rationale for the higher AD prevalence among smokers, and indicate that certain forms of lead in particular may constitute an environmental risk factor for AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/efectos de los fármacos , Fumar Cigarrillos/efectos adversos , Cadmio/química , Cromo/química , Humanos , Iones/química , Plomo/química , Nicotina/efectos adversos , Fragmentos de Péptidos/metabolismo , Placa Amiloide , Hidrocarburos Policíclicos Aromáticos/efectos adversos , Factores de Riesgo , Fumar/efectos adversos
16.
J Alzheimers Dis ; 54(3): 971-982, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27567855

RESUMEN

Aggregation of the amyloid-beta (Aß) peptide into insoluble plaques is a major factor in Alzheimer's disease (AD) pathology. Another major factor in AD is arguably metal ions, as metal dyshomeostasis is observed in AD patients, metal ions modulate Aß aggregation, and AD plaques contain numerous metals including redox-active Cu and Fe ions. In vivo, Aß is found in various cellular locations including membranes. So far, Cu(II)/Aß interactions and ROS generation have not been investigated in a membrane environment. Here, we study Cu(II) and Zn(II) interactions with Aß bound to SDS micelles or to engineered aggregation-inhibiting molecules (the cyclic peptide CP-2 and the ZAß3(12-58)Y18L Affibody molecule). In all studied systems the Aß N-terminal segment was found to be unbound, unstructured, and free to bind metal ions. In SDS micelles, Aß was found to bind Cu(II) and Zn(II) with the same ligands and the same KD as in aqueous solution. ROS was generated in all Cu(II)/Aß complexes. These results indicate that binding of Aß to membranes, drugs, and other entities that do not interact with the Aß N-terminal part, appears not to compromise the N-terminal segment's ability to bind metal ions, nor impede the capacity of N-terminally bound Cu(II) to generate ROS.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Cobre/metabolismo , Micelas , Fragmentos de Péptidos/metabolismo , Agregado de Proteínas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Péptidos beta-Amiloides/análisis , Sitios de Unión/fisiología , Cobre/análisis , Humanos , Peróxido de Hidrógeno/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Fragmentos de Péptidos/análisis
17.
J Trace Elem Med Biol ; 38: 183-193, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27085215

RESUMEN

Growing evidence links neurodegenerative diseases to metal exposure. Aberrant metal ion concentrations have been noted in Alzheimer's disease (AD) brains, yet the role of metals in AD pathogenesis remains unresolved. A major factor in AD pathogenesis is considered to be aggregation of and amyloid formation by amyloid-ß (Aß) peptides. Previous studies have shown that Aß displays specific binding to Cu(II) and Zn(II) ions, and such binding has been shown to modulate Aß aggregation. Here, we use nuclear magnetic resonance (NMR) spectroscopy to show that Mn(II) ions also bind to the N-terminal part of the Aß(1-40) peptide, with a weak binding affinity in the milli- to micromolar range. Circular dichroism (CD) spectroscopy, solid state atomic force microscopy (AFM), fluorescence spectroscopy, and molecular modeling suggest that the weak binding of Mn(II) to Aß may not have a large effect on the peptide's aggregation into amyloid fibrils. However, identification of an additional metal ion displaying Aß binding reveals more complex AD metal chemistry than has been previously considered in the literature.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Manganeso/química , Manganeso/metabolismo , Sitios de Unión , Humanos , Iones/química , Iones/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA