Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Phys Chem Chem Phys ; 24(1): 86-97, 2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-34878460

RESUMEN

Alzheimer's disease (AD) is a primary form of dementia with debilitating consequences, but no effective cure is available. While the pathophysiology of AD remains multifactorial, the aggregation of amyloid beta (Aß) mediated by the cell membrane is known to be the cause for the neurodegeneration associated with AD. Here we examined the effects of graphene quantum dots (GQDs) on the obstruction of the membrane axis of Aß in its three representative forms of monomers (Aß-m), oligomers (Aß-o), and amyloid fibrils (Aß-f). Specifically, we determined the membrane fluidity of neuroblastoma SH-SY5Y cells perturbed by the Aß species, especially by the most toxic Aß-o, and demonstrated their recovery by GQDs using confocal fluorescence microscopy. Our computational data through discrete molecular dynamics simulations further revealed energetically favorable association of the Aß species with the GQDs in overcoming peptide-peptide aggregation. Overall, this study positively implicated GQDs as an effective agent in breaking down the membrane axis of Aß, thereby circumventing adverse downstream events and offering a potential therapeutic solution for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Membrana Celular/metabolismo , Grafito/metabolismo , Puntos Cuánticos/metabolismo , Péptidos beta-Amiloides/química , Membrana Celular/química , Grafito/química , Humanos , Simulación de Dinámica Molecular , Agregado de Proteínas , Puntos Cuánticos/química
2.
Biomacromolecules ; 21(2): 988-998, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-31909987

RESUMEN

Alzheimer's disease (AD) is the most severe form of neurological disorder, characterized by the presence of extracellular amyloid-ß (Aß) plaques and intracellular tau tangles. For decades, therapeutic strategies against the pathological symptoms of AD have often relied on the delivery of monoclonal antibodies to target specifically Aß amyloid or oligomers, largely to no avail. Aß can be traced in the brain as well as in cerebrospinal fluid and the circulation, giving rise to abundant opportunities to interact with their environmental proteins. Using liquid chromatography tandem-mass spectrometry, here we identified for the first time the protein coronae of the two major amyloid forms of Aß-Aß1-42 and Aß1-40-exposed to human blood plasma. Out of the proteins identified in all groups, 58 proteins were unique to the Aß1-42 samples and 31 proteins unique to the Aß1-40 samples. Both fibrillar coronae consisted of proteins significant in complement activation, inflammation, and protein metabolic pathways involved in the pathology of AD. Structure-wise, the coronal proteins often possessed multidomains of high flexibility to maximize their association with the amyloid fibrils. The protein corona hindered recognition of Aß1-42 fibrils by their structurally specific antibodies and accelerated the aggregation but not the ß-cell toxicity of human islet amyloid polypeptide, the peptide associated with type 2 diabetes. This study highlights the importance of understanding the structural, functional, and pathological implications of the amyloid protein corona for the development of therapeutics against AD and a range of amyloid diseases.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Fragmentos de Péptidos/metabolismo , Corona de Proteínas/metabolismo , Mapas de Interacción de Proteínas/fisiología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Línea Celular , Humanos , Células Secretoras de Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/química , Fragmentos de Péptidos/química , Corona de Proteínas/química , Estructura Secundaria de Proteína
3.
Nano Lett ; 19(9): 6535-6546, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31455083

RESUMEN

Human amyloids and plaques uncovered post mortem are highly heterogeneous in structure and composition, yet literature concerning the heteroaggregation of amyloid proteins is extremely scarce. This knowledge deficiency is further exacerbated by the fact that peptide delivery is a major therapeutic strategy for targeting their full-length counterparts associated with the pathologies of a range of human diseases, including dementia and type 2 diabetes (T2D). Accordingly, here we examined the coaggregation of full-length human islet amyloid polypeptide (IAPP), a peptide associated with type 2 diabetes, with its primary and secondary amyloidogenic fragments 19-29 S20G and 8-20. Single-molecular aggregation dynamics was obtained by high-speed atomic force microscopy, augmented by transmission electron microscopy, X-ray diffraction, and super-resolution stimulated emission depletion microscopy. The coaggregation significantly prolonged the pause phase of fibril elongation, increasing its dwell time by 3-fold. Surprisingly, unidirectional elongation of mature fibrils, instead of protofilaments, was observed for the coaggregation, indicating a new form of tertiary protein aggregation unknown to existing theoretical models. Further in vivo zebrafish embryonic assay indicated improved survival and hatching, as well as decreased frequency and severity of developmental abnormalities for embryos treated with the heteroaggregates of IAPP with 19-29 S20G, but not with 8-20, compared to the control, indicating the therapeutic potential of 19-29 S20G against T2D.


Asunto(s)
Amiloidosis/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Agregación Patológica de Proteínas/tratamiento farmacológico , Amiloidosis/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Microscopía de Fuerza Atómica , Microscopía Electrónica de Transmisión , Agregación Patológica de Proteínas/metabolismo , Pez Cebra/metabolismo
4.
Small ; 15(18): e1805166, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30908844

RESUMEN

The self-assembly of human islet amyloid polypeptide (hIAPP) into ß-sheet-rich nanofibrils is associated with the pathogeny of type 2 diabetes. Soluble hIAPP is intrinsically disordered with N-terminal residues 8-17 as α-helices. To understand the contribution of the N-terminal helix to the aggregation of full-length hIAPP, here the oligomerization dynamics of the hIAPP fragment 8-20 (hIAPP8-20) are investigated with combined computational and experimental approaches. hIAPP8-20 forms cross-ß nanofibrils in silico from isolated helical monomers via the helical oligomers and α-helices to ß-sheets transition, as confirmed by transmission electron microscopy, atomic force microscopy, circular dichroism spectroscopy, Fourier transform infrared spectroscopy, and reversed-phase high performance liquid chromatography. The computational results also suggest that the critical nucleus of aggregation corresponds to hexamers, consistent with a recent mass-spectroscopy study of hIAPP8-20 aggregation. hIAPP8-20 oligomers smaller than hexamers are helical and unstable, while the α-to-ß transition starts from the hexamers. Converted ß-sheet-rich oligomers first form ß-barrel structures as intermediates before aggregating into cross-ß nanofibrils. This study uncovers a complete picture of hIAPP8-20 peptide oligomerization, aggregation nucleation via conformational conversion, formation of ß-barrel intermediates, and assembly of cross-ß protofibrils, thereby shedding light on the aggregation of full-length hIAPP, a hallmark of pancreatic beta-cell degeneration.


Asunto(s)
Amiloide/química , Polipéptido Amiloide de los Islotes Pancreáticos/química , Conformación Proteica en Lámina beta , Algoritmos , Cromatografía Líquida de Alta Presión/métodos , Humanos , Células Secretoras de Insulina/metabolismo , Microscopía de Fuerza Atómica , Microscopía Electrónica de Transmisión , Simulación de Dinámica Molecular , Análisis Espectral
5.
Nano Lett ; 18(9): 5797-5804, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30088935

RESUMEN

Amyloid diseases are global epidemics with no cure available. Herein, we report a first demonstration of in vivo mitigation of amyloidogenesis using biomimetic nanotechnology. Specifically, the amyloid fragments (ba) of ß-lactoglobulin, a whey protein, were deposited onto the surfaces of carbon nanotubes (baCNT), which subsequently sequestered human islet amyloid polypeptide (IAPP) through functional-pathogenic double-protein coronae. Conformational changes at the ba-IAPP interface were studied by Fourier transform infrared, circular dichroism, and X-ray scattering spectroscopies. baCNT eliminated the toxic IAPP species from zebrafish embryos, as evidenced by the assays of embryonic development, cell morphology, hatching, and survival as well as suppression of oxidative stress. In addition to IAPP, baCNT also displayed high potency against the toxicity of amyloid-ß, thereby demonstrating the broad applicability of this biomimetic nanotechnology and the use of an embryonic zebrafish model for the high-throughput screening of a range of amyloidogenesis and their inhibitors in vivo.


Asunto(s)
Amiloide/química , Polipéptido Amiloide de los Islotes Pancreáticos/química , Lactoglobulinas/química , Nanotubos de Carbono/química , Corona de Proteínas/química , Proteína de Suero de Leche/química , Amiloide/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Humanos , Estrés Oxidativo , Propiedades de Superficie , Pez Cebra/embriología
6.
Small ; 14(47): e1802825, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30369028

RESUMEN

Amyloid fibrils generally display chirality, a feature which has rarely been exploited in the development of therapeutics against amyloid diseases. This study reports, for the first time, the use of mesoscopic chiral silica nanoribbons against the in vivo amyloidogenesis of human islet amyloid polypeptide (IAPP), the peptide whose aggregation is implicated in type 2 diabetes. The thioflavin T assay and transmission electron microscopy show accelerated IAPP fibrillization through elimination of the nucleation phase and shortening of the elongation phase by the nanostructures. Coarse-grained simulations offer complementary molecular insights into the acceleration of amyloid aggregation through their nonspecific binding and directional seeding with the nanostructures. This accelerated IAPP fibrillization translates to reduced toxicity, especially for the right-handed silica nanoribbons, as revealed by cell viability, helium ion microscopy, as well as zebrafish embryo survival, developmental, and behavioral assays. This study has implicated the potential of employing chiral nanotechnologies against the mesoscopic enantioselectivity of amyloid proteins and their associated diseases.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/química , Nanotubos de Carbono/química , Dióxido de Silicio/química , Humanos , Estereoisomerismo
7.
Chem Soc Rev ; 46(21): 6492-6531, 2017 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-28702523

RESUMEN

Neurodegenerative disorders and type 2 diabetes are global epidemics compromising the quality of life of millions worldwide, with profound social and economic implications. Despite the significant differences in pathology - much of which are poorly understood - these diseases are commonly characterized by the presence of cross-ß amyloid fibrils as well as the loss of neuronal or pancreatic ß-cells. In this review, we document research progress on the molecular and mesoscopic self-assembly of amyloid-beta, alpha synuclein, human islet amyloid polypeptide and prions, the peptides and proteins associated with Alzheimer's, Parkinson's, type 2 diabetes and prion diseases. In addition, we discuss the toxicities of these amyloid proteins based on their self-assembly as well as their interactions with membranes, metal ions, small molecules and engineered nanoparticles. Through this presentation we show the remarkable similarities and differences in the structural transitions of the amyloid proteins through primary and secondary nucleation, the common evolution from disordered monomers to alpha-helices and then to ß-sheets when the proteins encounter the cell membrane, and, the consensus (with a few exceptions) that off-pathway oligomers, rather than amyloid fibrils, are the toxic species regardless of the pathogenic protein sequence or physicochemical properties. In addition, we highlight the crucial role of molecular self-assembly in eliciting the biological and pathological consequences of the amyloid proteins within the context of their cellular environments and their spreading between cells and organs. Exploiting such structure-function-toxicity relationship may prove pivotal for the detection and mitigation of amyloid diseases.


Asunto(s)
Proteínas Amiloidogénicas/biosíntesis , Diabetes Mellitus Tipo 2/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas Amiloidogénicas/química , Humanos
8.
Small ; 13(36)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28783260

RESUMEN

Polyethylene glycol (PEG) is widely used as an antifouling and stealth polymer in surface engineering and nanomedicine. However, recent research has revealed adverse effects of bioaccumulation and immunogenicity following the administration of PEG, prompting this proteomic examination of the plasma protein coronae association with superparamagnetic iron oxide nanoparticles (IONPs) grafted with brushed PEG (bPEG) and an alternative, brushed phosphorylcholine (bPC). Using label-free quantitation by liquid chromatography tandem-mass spectrometry, this study determines protein abundances for the in vitro hard coronae of bare, bPC-, and bPEG-grafted IONPs in human plasma. This study also shows unique protein compositions in the plasma coronae of each IONP, including enrichment of coagulation factors and immunogenic complement proteins with bPEG, and enhanced binding of apolipoproteins with bPC. Functional analysis reveals that plasma protein coronae elevate the horseradish peroxidase-like activities of the bPC- and bPEG-IONPs by approximately twofold, an effect likely mediated by the diverse composition and physicochemical properties of the polymers as well as their associated plasma proteins. Taken together, these observations support the rational design of stealth polymers based on a quantitative understanding of the interplay between IONPs and the plasma proteome, and should prove beneficial for the development of materials for nanomedicine, biosensing, and catalysis.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Compuestos Férricos/química , Nanopartículas/química , Polímeros/química , Proteoma/metabolismo , Catálisis , Ontología de Genes , Peroxidasa de Rábano Silvestre/metabolismo , Humanos , Nanopartículas/ultraestructura , Fosfatidilcolinas/química , Polietilenglicoles/química , Corona de Proteínas/química , Mapas de Interacción de Proteínas
9.
Biomacromolecules ; 18(12): 4316-4322, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29095600

RESUMEN

Biomimetic nanocomposites and scaffolds hold the key to a wide range of biomedical applications. Here we show, for the first time, a facile scheme of cofibrillizing pathogenic and functional amyloid fibrils via gold nanoparticles (AuNPs) and their applications against amyloidogenesis. This scheme was realized by ß-sheet stacking between human islet amyloid polypeptide (IAPP) and the ß-lactoglobulin "corona" of the AuNPs, as revealed by transmission electron microscopy, 3D atomic force microscopy, circular dichroism spectroscopy, and molecular dynamics simulations. The biomimetic AuNPs eliminated IAPP toxicity, enabled X-ray destruction of IAPP amyloids, and allowed dark-field imaging of pathogenic amyloids and their immunogenic response by human T cells. In addition to providing a viable new nanotechnology against amyloidogenesis, this study has implications for understanding the in vivo cross-talk between amyloid proteins of different pathologies.


Asunto(s)
Proteínas Amiloidogénicas/química , Oro/química , Nanopartículas del Metal/química , Amiloide/química , Dicroismo Circular/métodos , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Cinética , Lactoglobulinas/química , Microscopía de Fuerza Atómica/métodos , Microscopía Electrónica de Transmisión/métodos , Simulación de Dinámica Molecular , Nanotecnología/métodos , Conformación Proteica en Lámina beta , Linfocitos T/efectos de los fármacos
10.
Biomacromolecules ; 18(12): 4249-4260, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29035554

RESUMEN

Protein aggregation into amyloid fibrils is a ubiquitous phenomenon across the spectrum of neurodegenerative disorders and type 2 diabetes. A common strategy against amyloidogenesis is to minimize the populations of toxic oligomers and protofibrils by inhibiting protein aggregation with small molecules or nanoparticles. However, melanin synthesis in nature is realized by accelerated protein fibrillation to circumvent accumulation of toxic intermediates. Accordingly, we designed and demonstrated the use of star-shaped poly(2-hydroxyethyl acrylate) (PHEA) nanostructures for promoting aggregation while ameliorating the toxicity of human islet amyloid polypeptide (IAPP), the peptide involved in glycemic control and the pathology of type 2 diabetes. The binding of PHEA elevated the ß-sheet content in IAPP aggregates while rendering a new morphology of "stelliform" amyloids originating from the polymers. Atomistic molecular dynamics simulations revealed that the PHEA arms served as rodlike scaffolds for IAPP binding and subsequently accelerated IAPP aggregation by increased local peptide concentration. The tertiary structure of the star nanoparticles was found to be essential for driving the specific interactions required to impel the accelerated IAPP aggregation. This study sheds new light on the structure-toxicity relationship of IAPP and points to the potential of exploiting star polymers as a new class of therapeutic agents against amyloidogenesis.


Asunto(s)
Amiloide/química , Proteínas Amiloidogénicas/química , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polímeros/química , Agregación Patológica de Proteínas/patología , Amiloidosis/patología , Animales , Línea Celular , Diabetes Mellitus Tipo 2/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación de Dinámica Molecular , Nanopartículas/química
11.
Phys Chem Chem Phys ; 19(45): 30627-30635, 2017 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-29115353

RESUMEN

Amyloid aggregation of human islet amyloid polypeptide (IAPP) is a hallmark of type 2 diabetes (T2D), a metabolic disease and a global epidemic. Although IAPP is synthesized in pancreatic ß-cells, its fibrils and plaques are found in the extracellular space indicating a causative transmembrane process. Numerous biophysical studies have revealed that cell membranes as well as model lipid vesicles promote the aggregation of amyloid-ß (associated with Alzheimer's), α-synuclein (associated with Parkinson's) and IAPP, through electrostatic and hydrophobic interactions between the proteins/peptides and lipid membranes. Using a thioflavin T kinetic assay, transmission electron microscopy, circular dichroism spectroscopy, discrete molecular dynamics simulations as well as free energy calculations here we show that micellar lysophosphatidylcholine (LPC), the most abundant lysophospholipid in the blood, inhibited the amyloid aggregation of IAPP through nonspecific interactions while elevating the α-helical peptide secondary structure. This surprising finding suggests a native protective mechanism against IAPP aggregation in vivo.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Polipéptido Amiloide de los Islotes Pancreáticos/química , Lisofosfatidilcolinas/química , Simulación de Dinámica Molecular , Benzotiazoles , Humanos , Células Secretoras de Insulina/metabolismo , Cinética , Microscopía Electrónica de Transmisión , Tiazoles , alfa-Sinucleína/química
12.
Small ; 12(12): 1615-26, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-26808649

RESUMEN

Human islet amyloid polypeptide (hIAPP, or amylin) forms amyloid deposits in the islets of Langerhans, a phenomenon that is associated with type-2 diabetes impacting millions of people worldwide. Accordingly, strategies against hIAPP aggregation are essential for the prevention and eventual treatment of the disease. Here, it is shown that generation-3 OH-terminated poly(amidoamine) dendrimer, a polymeric nanoparticle, can effectively halt the aggregation of hIAPP and shut down hIAPP toxicity in pancreatic MIN6 and NIT-1 cells as well as in mouse islets. This finding is supported by high-throughput dynamic light scattering experiment and thioflavin T assay, where the rapid evolution of hIAPP nucleation and elongation processes is halted by the addition of the dendrimer up to 8 h. Discrete molecular dynamics simulations further reveal that hIAPP residues bound strongly with the dendrimer near the c-terminal portion of the peptide, where the amyloidogenic sequence (residues 22-29) locates. Furthermore, simulations of hIAPP dimerization reveal that binding with the dendrimer significantly reduces formation of interpeptide contacts and hydrogen bonds, thereby prohibiting peptide self-association and amyloidosis. This study points to a promising nanomedicinal strategy for combating type-2 diabetes and may have broader implications for targeting neurological disorders whose distinct hallmark is also amyloid fibrillation.


Asunto(s)
Amiloide/metabolismo , Dendrímeros/toxicidad , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Agregado de Proteínas/efectos de los fármacos , Benzotiazoles , Muerte Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Humanos , Hidroxilación , Células Secretoras de Insulina/efectos de los fármacos , Modelos Moleculares , Multimerización de Proteína/efectos de los fármacos , Tiazoles/metabolismo
13.
Int J Nanomedicine ; 19: 4857-4875, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38828195

RESUMEN

Brain diseases are the most devastating problem among the world's increasingly aging population, and the number of patients with neurological diseases is expected to increase in the future. Although methods for delivering drugs to the brain have advanced significantly, none of these approaches provide satisfactory results for the treatment of brain diseases. This remains a challenge due to the unique anatomy and physiology of the brain, including tight regulation and limited access of substances across the blood-brain barrier. Nanoparticles are considered an ideal drug delivery system to hard-to-reach organs such as the brain. The development of new drugs and new nanomaterial-based brain treatments has opened various opportunities for scientists to develop brain-specific delivery systems that could improve treatment outcomes for patients with brain disorders such as Alzheimer's disease, Parkinson's disease, stroke and brain tumors. In this review, we discuss noteworthy literature that examines recent developments in brain-targeted nanomedicines used in the treatment of neurological diseases.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Sistemas de Liberación de Medicamentos , Nanomedicina , Humanos , Nanomedicina/métodos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Animales , Nanopartículas/química , Encefalopatías/tratamiento farmacológico , Sistema de Administración de Fármacos con Nanopartículas/química , Sistema de Administración de Fármacos con Nanopartículas/farmacocinética , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Alzheimer/tratamiento farmacológico
14.
Nat Commun ; 15(1): 613, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38242873

RESUMEN

Alzheimer's disease (AD) is a major cause of dementia debilitating the global ageing population. Current understanding of the AD pathophysiology implicates the aggregation of amyloid beta (Aß) as causative to neurodegeneration, with tauopathies, apolipoprotein E and neuroinflammation considered as other major culprits. Curiously, vascular endothelial barrier dysfunction is strongly associated with Aß deposition and 80-90% AD subjects also experience cerebral amyloid angiopathy. Here we show amyloid protein-induced endothelial leakiness (APEL) in human microvascular endothelial monolayers as well as in mouse cerebral vasculature. Using signaling pathway assays and discrete molecular dynamics, we revealed that the angiopathy first arose from a disruption to vascular endothelial (VE)-cadherin junctions exposed to the nanoparticulates of Aß oligomers and seeds, preceding the earlier implicated proinflammatory and pro-oxidative stressors to endothelial leakiness. These findings were analogous to nanomaterials-induced endothelial leakiness (NanoEL), a major phenomenon in nanomedicine depicting the paracellular transport of anionic inorganic nanoparticles in the vasculature. As APEL also occurred in vitro with the oligomers and seeds of alpha synuclein, this study proposes a paradigm for elucidating the vascular permeation, systemic spread, and cross-seeding of amyloid proteins that underlie the pathogeneses of AD and Parkinson's disease.


Asunto(s)
Enfermedad de Alzheimer , Angiopatía Amiloide Cerebral , Humanos , Ratones , Animales , Péptidos beta-Amiloides/metabolismo , Agregado de Proteínas , Proteínas Amiloidogénicas/metabolismo , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo
15.
Nanotechnology ; 24(34): 345101, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23899823

RESUMEN

We report on the dose-dependent inhibition of firefly luciferase activity induced by exposure of the enzyme to 20 nm citrate-coated silver nanoparticles (AgNPs). The inhibition mechanism was examined by characterizing the physicochemical properties and biophysical interactions of the enzyme and the AgNPs. Consistently, binding of the enzyme induced an increase in zeta potential from -22 to 6 mV for the AgNPs, triggered a red-shift of 44 nm in the absorbance peak of the AgNPs, and rendered a 'protein corona' of 20 nm in thickness on the nanoparticle surfaces. However, the secondary structures of the enzyme were only marginally affected upon formation of the protein corona, as verified by circular dichroism spectroscopy measurement and multiscale discrete molecular dynamics simulations. Rather, inductively coupled plasma mass spectrometry measurement revealed a significant ion release from the AgNPs. The released silver ions could readily react with the cysteine residues and N-groups of the enzyme to alter the physicochemical environment of their neighboring catalytic site and subsequently impair the enzymatic activity.


Asunto(s)
Luciferasas de Luciérnaga/metabolismo , Nanopartículas del Metal/química , Plata/metabolismo , Oro/análisis , Iones , Luciferasas de Luciérnaga/antagonistas & inhibidores , Luciferasas de Luciérnaga/química , Nanopartículas del Metal/ultraestructura , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína , Plata/análisis , Espectrofotometría Ultravioleta , Electricidad Estática
16.
Adv Sci (Weinh) ; 10(29): e2301423, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37594661

RESUMEN

Alzheimer's disease (AD) is a leading form of dementia where the presence of extra-neuronal plaques of Amyloid-ß (Aß) is a pathological hallmark. However, Aß peptide is also observed in the intestinal tissues of AD patients and animal models. In this study, it is reported that Aß monomers can target and disintegrate microbial amyloids of FapC and CsgA formed by opportunistic gut pathogens, Pseudomonas aeruginosa and Escherichia coli, explaining a potential role of Aß in the gut-brain axis. Employing a zebrafish-based transparent in vivo system and whole-mount live-imaging, Aß is observed to diffuse into the vasculature and subsequently localize with FapC or CsgA fibrils that were injected into the tail muscles of the fish. FapC aggregates, produced after Aß treatment (Faß), present selective toxicity to SH-SY5Y neuronal cells while the intestinal Caco-2 cells are shown to phagocytose Faß in a non-toxic cellular process. After remodeling by Aß, microbial fibrils lose their native function of cell adhesion with intestinal Caco-2 cells and Aß dissolves and detaches the microbial fibrils already attached to the cell membrane. Taken together, this study strongly indicates an anti-biofilm role for Aß monomers that can help aid in the future development of selective anti-Alzheimer's and anti-infective medicine.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Animales , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Células CACO-2 , Pez Cebra/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Escherichia coli/metabolismo , Biopelículas
17.
Exploration (Beijing) ; : 20220012, 2022 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-35941991

RESUMEN

Coronavirus disease 2019 (COVID-19) is a highly contagious viral disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This deadly infection has resulted in more than 5.2 million deaths worldwide. The global rollout of COVID-19 vaccines has without doubt saved countless lives by reducing the severity of symptoms for patients. However, as the virus continues to evolve, there is a risk that the vaccines and antiviral designed to target the infection will no longer be therapeutically viable. Furthermore, there remain fears over both the short and long-term side effects of repeat exposure to currently available vaccines. In this review, we discuss the pros and cons of the vaccine rollout and promote the idea of a COVID medicinal toolbox made up of different antiviral treatment modalities, and present some of the latest therapeutic strategies that are being explored in this respect to try to combat the COVID-19 virus and other COVID viruses that are predicted to follow. Lastly, we review current literature on the use of siRNA therapeutics as a way to remain adaptable and in tune with the ever-evolving mutation rate of the COVID-19 virus.

18.
Sensors (Basel) ; 11(11): 10502-21, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22346655

RESUMEN

The analysis of (bio)available copper in complex environmental settings, including biological test media, is a challenging task. In this study, we demonstrated the potential of a recombinant Pseudomonas fluorescens-based biosensor for bioavailability analysis of CuSO4 and CuO nanoparticles (nano-CuO) in seventeen different ecotoxicological and microbiologial test media. In parallel, free Cu in these test media was analysed using Cu-ion selective electrode (Cu-ISE). In the case of CuSO4, both free and bioavailable Cu decreased greatly with increasing concentration of organics and phosphates in the tested media. A good correlation between free and bioavailable Cu was observed (r = 0.854, p < 0.01) indicating that the free Cu content in biological test media may be a reasonably good predictor for the toxicity of CuSO4. As a proof, it was demonstrated that when eleven EC50 values for CuSO4 from different organisms in different test media were normalized for the free Cu in these media, the difference in these EC50 values was decreased from 4 to 1.8 orders of magnitude. Thus, toxicity of CuSO4 to these organisms was attributed to the properties of the test media rather than to inherent differences in sensitivity between the test organisms. Differently from CuSO4, the amount of free and bioavailable Cu in nano-CuO spiked media was not significantly correlated with the concentration of organics in the test media. Thus, the speciation of nano-CuO in toxicological test systems was not only determined by the complexation of Cu ions but also by differential dissolution of nano-CuO in different test conditions leading to a new speciation equilibrium. In addition, a substantial fraction of nano-CuO that was not detectable by Cu-ISE (i.e., not present as free Cu-ions) was bioavailable to Cu-biosensor bacteria. Thus, in environmental hazard analysis of (nano) particulate materials, biosensor analysis may be more informative than other analytical techniques. Our results demonstrate that bacterial Cu-biosensors either in combination with other analytical/speciation techniques or on their own, may serve as a rapid (eco)toxicological screening method.


Asunto(s)
Técnicas Biosensibles/métodos , Sulfato de Cobre/química , Cobre/análisis , Medios de Cultivo/química , Electrodos de Iones Selectos , Nanopartículas del Metal/química , Aliivibrio fischeri/efectos de los fármacos , Animales , Anostraca/efectos de los fármacos , Disponibilidad Biológica , Tampones (Química) , Chlorophyta/efectos de los fármacos , Cobre/química , Cobre/farmacología , Cobre/toxicidad , Sulfato de Cobre/farmacología , Sulfato de Cobre/toxicidad , Daphnia/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Concentración 50 Inhibidora , Límite de Detección , Mediciones Luminiscentes , Ensayo de Materiales/métodos , Nanopartículas del Metal/toxicidad , Pseudomonas fluorescens/efectos de los fármacos , Pseudomonas fluorescens/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Tetrahymena thermophila/efectos de los fármacos , Pruebas de Toxicidad , Contaminantes Químicos del Agua/análisis , Contaminantes Químicos del Agua/química , Contaminantes Químicos del Agua/farmacología , Contaminantes Químicos del Agua/toxicidad
19.
Nanoscale Horiz ; 6(2): 95-119, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33438715

RESUMEN

Amyloid diseases are global epidemics characterized by the accumulative deposits of cross-beta amyloid fibrils and plaques. Despite decades of intensive research, few solutions are available for the diagnosis, treatment, and prevention of these debilitating diseases. Since the early work on the interaction of human ß2-microglobulin and nanoparticles by Linse et al. in 2007, the field of amyloidosis inhibition has gradually evolved into a new frontier in nanomedicine offering numerous interdisciplinary research opportunities, especially for materials, chemistry and biophysics. In this review we summarise, for the first time, the in vitro and in vivo models employed thus far in the field of anti-amyloidosis nanomedicines. Based on this systematic summary, we bring forth the notion that, due to the complex and often overlapping physiopathologies of amyloid diseases, there is a crucial need for the appropriate use of in vitro and in vivo models for validating novel anti-amyloidosis nanomedicines, and there is a crucial need for the development of new animal models that reflect the behavioural, symptomatic and cross-talk hallmarks of amyloid diseases such as Alzheimer's (AD), Parkinson's (PD) diseases and type 2 diabetes (T2DM).


Asunto(s)
Amiloidosis/tratamiento farmacológico , Modelos Animales de Enfermedad , Modelos Biológicos , Nanopartículas/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Nanomedicina/métodos , Nanopartículas/química
20.
J Phys Chem Lett ; 12(1): 368-378, 2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33356290

RESUMEN

Increasing experiments suggest that amyloid peptides can undergo liquid-liquid phase separation (LLPS) before the formation of amyloid fibrils. However, the exact role of LLPS in amyloid aggregation at the molecular level remains elusive. Here, we investigated the LLPS and amyloid fibrillization of a coarse-grained peptide, capable of capturing fundamental properties of amyloid aggregation over a wide range of concentrations in molecular dynamics simulations. On the basis of the Flory-Huggins theory of polymer solutions, we determined the binodal and spinodal concentrations of LLPS in the low-concentration regime, ϕBL and ϕSL, respectively. Only at concentrations above ϕBL, peptides formed metastable or stable oligomers corresponding to the high-density liquid phase (HDLP) in LLPS, out of which the nucleated conformational conversion to fibril seeds occurred. Below ϕSL, the HDLP was metastable and transient, and the subsequent fibrillization process followed the traditional nucleation and elongation mechanisms. Only above ϕSL, the HDLP became stable, and the initial fibril nucleation and growth were governed by the high local peptide concentrations. The predicted saturation of amyloid aggregation half-times with increasing peptide concentration to a constant, instead of the traditional power-law scaling to zero, was confirmed by simulations and by a thioflavin-T kinetic assay and the transmission electron microscopy of islet amyloid polypeptide (IAPP) aggregation. Our study provides a unified picture of amyloid aggregation for a wide range of concentrations within the framework of LLPS, which may help us better understand the etiology of amyloid diseases, where the amyloid protein concentration can vary by ∼9 orders of magnitude depending on the organ location and facilitate the engineering of novel amyloid-based functional materials.


Asunto(s)
Amiloide/química , Simulación de Dinámica Molecular , Péptidos/química , Agregado de Proteínas , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA