Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Int J Biol Macromol ; 259(Pt 2): 129303, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38216018

RESUMEN

Cellulose nanocrystals (CNC) conventionally involve highly concentrated sulphuric acid, which typically resulted in the formation of undesirable by-products. Although less corrosive mineral acids have been explored as alternatives, high concentrations are still required. In this study, CNC was successfully isolated from Leucaena leucocephala wood using mild sulphuric acid with acetic acid as protic solvent, and it was further studied with the addition of Lewis acids in the form of multivalent transition metal salts as co-catalyst. Selected divalent and trivalent transition metal salts including (Cr(NO3)3, Fe(NO3)3, Co(NO3)2, and Ni(NO3)2) were investigated. The morphology, chemical structure, particle size, and physicochemical properties of the CNCs were determined. Controlled depolymerization of cellulose was observed using transmission electron microscopy (TEM). Rod-like morphology for all CNCs was obtained during the hydrolysis process with the smallest CNC particles found at an average length of 278.1 ± 35.1 nm and a diameter of 13.4 ± 3.0 nm. The results showed that higher valence state metal ions resulted in better cellulose hydrolysis efficiency. In addition, the use of transition metal salt as a co-catalyst improved production efficiency and minimised carbonization of CNC while maintaining desired crystallinity and thermal properties.


Asunto(s)
Celulosa , Nanopartículas , Celulosa/química , Solventes , Ácido Acético , Sales (Química) , Nanopartículas/química , Ácidos Sulfúricos/química
2.
Nat Commun ; 14(1): 4278, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460561

RESUMEN

Current technologies to subtype glioblastoma (GBM), the most lethal brain tumor, require highly invasive brain biopsies. Here, we develop a dedicated analytical platform to achieve direct and multiplexed profiling of circulating RNAs in extracellular vesicles for blood-based GBM characterization. The technology, termed 'enzyme ZIF-8 complexes for regenerative and catalytic digital detection of RNA' (EZ-READ), leverages an RNA-responsive transducer to regeneratively convert and catalytically enhance signals from rare RNA targets. Each transducer comprises hybrid complexes - protein enzymes encapsulated within metal organic frameworks - to configure strong catalytic activity and robust protection. Upon target RNA hybridization, the transducer activates directly to liberate catalytic complexes, in a target-recyclable manner; when partitioned within a microfluidic device, these complexes can individually catalyze strong chemifluorescence reactions for digital RNA quantification. The EZ-READ platform thus enables programmable and reliable RNA detection, across different-sized RNA subtypes (miRNA and mRNA), directly in sample lysates. When clinically evaluated, the EZ-READ platform established composite signatures for accurate blood-based GBM diagnosis and subtyping.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , MicroARNs , Humanos , MicroARNs/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , ARN Mensajero , Hibridación de Ácido Nucleico , Glioblastoma/genética , Glioblastoma/patología
3.
Nat Nanotechnol ; 16(6): 734-742, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33686255

RESUMEN

Current technologies to measure drug-target interactions require complex processing and invasive tissue biopsies, limiting their clinical utility for cancer treatment monitoring. Here we develop an analytical platform that leverages circulating extracellular vesicles (EVs) for activity-based assessment of tumour-specific drug-target interactions in patient blood samples. The technology, termed extracellular vesicle monitoring of small-molecule chemical occupancy and protein expression (ExoSCOPE), utilizes bio-orthogonal probe amplification and spatial patterning of molecular reactions within matched plasmonic nanoring resonators to achieve in situ analysis of EV drug dynamics. It measures changes in drug occupancy and protein composition in molecular subpopulations of EVs. When used to monitor various targeted therapies, the ExoSCOPE revealed EV signatures that closely reflected cellular treatment efficacy. We further applied the technology for clinical cancer diagnostics and treatment monitoring. Using a small volume of blood, the ExoSCOPE accurately classified disease status and rapidly distinguished between targeted treatment outcomes, within 24 h after treatment initiation.


Asunto(s)
Antineoplásicos/farmacología , Vesículas Extracelulares/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Antineoplásicos/farmacocinética , Biomarcadores de Tumor/sangre , Técnicas Biosensibles/instrumentación , Técnicas Biosensibles/métodos , Estudios de Casos y Controles , Línea Celular Tumoral , Receptores ErbB/genética , Clorhidrato de Erlotinib/sangre , Clorhidrato de Erlotinib/uso terapéutico , Vesículas Extracelulares/química , Estudios de Factibilidad , Humanos , Neoplasias Pulmonares/sangre , Relación Señal-Ruido
4.
Eur J Neurol ; 28(5): 1479-1489, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33370497

RESUMEN

BACKGROUND AND PURPOSE: Various blood biomarkers reflecting brain amyloid-ß (Aß) load have recently been proposed with promising results. However, to date, no comparative study amongst blood biomarkers has been reported. Our objective was to examine the diagnostic performance and cost effectiveness of three blood biomarkers on the same cohort. METHODS: Using the same cohort (n = 68), the performances of the single-molecule array (Simoa) Aß40, Aß42, Aß42/Aß40 and the amplified plasmonic exosome (APEX) Aß42 blood biomarkers were compared using amyloid positron emission tomography (PET) as the reference standard. The extent to which these blood tests can reduce the recruitment cost of clinical trials was also determined by identifying amyloid positive (Aß+) participants. RESULTS: Compared to Simoa biomarkers, APEX-Aß42 showed significantly higher correlations with amyloid PET retention values and excellent diagnostic performance (sensitivity 100%, specificity 93.3%, area under the curve 0.995). When utilized for clinical trial recruitment, our simulation showed that pre-screening with blood biomarkers followed by a confirmatory amyloid PET imaging would roughly half the cost (56.8% reduction for APEX-Aß42 and 48.6% for Simoa-Aß42/Aß40) compared to the situation where only PET imaging is used. Moreover, with 100% sensitivity, APEX-Aß42 pre-screening does not increase the required number of initial participants. CONCLUSIONS: With its high diagnostic performance, APEX is an ideal candidate for Aß+ subject identification, monitoring and primary care screening, and could efficiently enrich clinical trials with Aß+ participants whilst halving recruitment costs.


Asunto(s)
Enfermedad de Alzheimer , Exosomas , Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides , Biomarcadores , Humanos , Inmunoensayo , Fragmentos de Péptidos
5.
Sci Adv ; 6(19): eaba2556, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32494726

RESUMEN

Exosomes are nanoscale vesicles distinguished by characteristic biophysical and biomolecular features; current analytical approaches, however, remain univariate. Here, we develop a dedicated platform for multiparametric exosome analysis-through simultaneous biophysical and biomolecular evaluation of the same vesicles-directly in clinical biofluids. Termed templated plasmonics for exosomes, the technology leverages in situ growth of gold nanoshells on vesicles to achieve multiselectivity. For biophysical selectivity, the nanoshell formation is templated by and tuned to distinguish exosome dimensions. For biomolecular selectivity, the nanoshell plasmonics locally quenches fluorescent probes only if they are target-bound on the same vesicle. The technology thus achieves multiplexed analysis of diverse exosomal biomarkers (e.g., proteins and microRNAs) but remains unresponsive to nonvesicle biomarkers. When implemented on a microfluidic, smartphone-based sensor, the platform is rapid, sensitive, and wash-free. It not only distinguished biomarker organizational states in native clinical samples but also showed that the exosomal subpopulation could more accurately differentiate patient prognosis.

6.
Adv Biosyst ; 4(12): e1900309, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32597034

RESUMEN

Neurodegenerative diseases are heterogeneous disorders characterized by a progressive loss of function and/or death of nerve cells, leading to severe cognitive and functional decline. Due to the complex pathology, early detection and intervention are critical to the development of successful treatments; however, current diagnostic approaches are limited to subjective, late-stage clinical findings. Extracellular vesicles (EVs) have recently emerged as a promising circulating biomarker for neurodegenerative diseases. Actively released by diverse cells, EVs are nanoscale membrane vesicles. They abound in blood, readily cross the blood-brain barrier, and carry diverse molecular cargoes in different organizational states: these molecular cargoes are inherited from the parent cells or bound to the EV membrane through surface associations. Specifically, EVs have been found to be associated with several important pathogenic proteins of neurodegenerative diseases, and their involvement could alter disease progression. This article provides an overview of EVs as circulating biomarkers of neurodegenerative diseases and introduces new technological advances to characterize the biophysical properties of EV-associated biomarkers for accurate, blood-based detection of neurodegenerative diseases.


Asunto(s)
Biomarcadores/sangre , Vesículas Extracelulares/química , Enfermedades Neurodegenerativas/diagnóstico , Humanos , Enfermedades Neurodegenerativas/sangre , Enfermedades Neurodegenerativas/metabolismo
7.
ACS Sens ; 5(1): 4-12, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31888329

RESUMEN

Extracellular vesicles (EVs) are diverse, nanoscale membrane vesicles released by cells into the circulation. As an emerging class of circulating biomarkers, EVs contain a trove of molecular information and play important roles in mediating intercellular communication. These EV molecular cargoes are differentially organized in the vesicles; they could be inherited from the parent cells or bound to the EV membrane through surface interactions. While the inherited constituents could serve as cell surrogate biomarkers, extravesicular association could reflect structural states of the bound molecules, revealing distinct subpopulations with different biophysical and/or biochemical properties. Despite the clinical potential of EVs and their diverse contents, conventional sensing technologies have limited compatibility to reveal nanoscale EV features. Complementary analytical platforms are being developed to address these technical challenges and expand the biomedical applications of EVs, to establish novel correlations and empower new diagnostics. This article provides a perspective on recent developments in sensor technologies to profile the diverse contents-different molecular types, quantities, and organizational states-in extracellular vesicles.


Asunto(s)
Biomarcadores/metabolismo , Técnicas Biosensibles/métodos , Vesículas Extracelulares/química , Humanos
8.
Bioorg Med Chem ; 27(20): 114962, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31307763

RESUMEN

The global emergence of antibiotic resistance is one of the most serious challenges facing modern medicine. There is an urgent need for validation of new drug targets and the development of small molecules with novel mechanisms of action. We therefore sought to inhibit bacterial DNA repair mediated by the AddAB/RecBCD protein complexes as a means to sensitize bacteria to DNA damage caused by the host immune system or quinolone antibiotics. A rational, hypothesis-driven compound optimization identified IMP-1700 as a cell-active, nanomolar potency compound. IMP-1700 sensitized multidrug-resistant Staphylococcus aureus to the fluoroquinolone antibiotic ciprofloxacin, where resistance results from a point mutation in the fluoroquinolone target, DNA gyrase. Cellular reporter assays indicated IMP-1700 inhibited the bacterial SOS-response to DNA damage, and compound-functionalized Sepharose successfully pulled-down the AddAB repair complex. This work provides validation of bacterial DNA repair as a novel therapeutic target and delivers IMP-1700 as a tool molecule and starting point for therapeutic development to address the pressing challenge of antibiotic resistance.


Asunto(s)
Antibacterianos/farmacología , ADN Bacteriano/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Quinolonas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Antibacterianos/síntesis química , Antibacterianos/química , Reparación del ADN , Relación Dosis-Respuesta a Droga , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Quinolonas/síntesis química , Quinolonas/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
9.
Nat Commun ; 10(1): 1144, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850633

RESUMEN

Despite intense interests in developing blood measurements of Alzheimer's disease (AD), the progress has been confounded by limited sensitivity and poor correlation to brain pathology. Here, we present a dedicated analytical platform for measuring different populations of circulating amyloid ß (Aß) proteins - exosome-bound vs. unbound - directly from blood. The technology, termed amplified plasmonic exosome (APEX), leverages in situ enzymatic conversion of localized optical deposits and double-layered plasmonic nanostructures to enable sensitive, multiplexed population analysis. It demonstrates superior sensitivity (~200 exosomes), and enables diverse target co-localization in exosomes. Employing the platform, we find that prefibrillar Aß aggregates preferentially bind with exosomes. We thus define a population of Aß as exosome-bound (Aß42+ CD63+) and measure its abundance directly from AD and control blood samples. As compared to the unbound or total circulating Aß, the exosome-bound Aß measurement could better reflect PET imaging of brain amyloid plaques and differentiate various clinical groups.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Encéfalo/patología , Exosomas/química , Neuronas/patología , Fragmentos de Péptidos/química , Placa Amiloide/patología , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/sangre , Técnicas Biosensibles , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Estudios de Casos y Controles , Línea Celular Tumoral , Exosomas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas Analíticas Microfluídicas , Neuronas/metabolismo , Neuronas/ultraestructura , Fragmentos de Péptidos/sangre , Placa Amiloide/diagnóstico por imagen , Placa Amiloide/metabolismo , Tomografía de Emisión de Positrones , Agregado de Proteínas , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/metabolismo , Resonancia por Plasmón de Superficie , Células THP-1 , Tetraspanina 30/química , Tetraspanina 30/metabolismo
10.
Angew Chem Int Ed Engl ; 58(1): 164-167, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30407697

RESUMEN

The PtIV prodrug strategy has emerged as an excellent alternative to tackle the problems associated with conventional PtII drug therapy. However, there is a lack of tools to study how this new class of PtIV drugs are processed at the cellular level. Herein, we report the first ratiometric probe for cisplatin detection and use it to investigate PtIV anticancer complexes in biological systems. The probe was able to distinguish between cisplatin and its PtIV derivatives, allowing us to probe the intracellular reduction of PtIV prodrug complexes. The correlation between the amount of active PtII species available after intracellular reduction of PtIV complexes and their cytotoxicity and the role glutathione plays in the reduction of PtIV complexes were investigated.


Asunto(s)
Cisplatino/química , Colorantes Fluorescentes/uso terapéutico , Platino (Metal)/química , Profármacos/uso terapéutico , Humanos
11.
J Biomed Mater Res B Appl Biomater ; 100(7): 1980-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22888018

RESUMEN

Alginate hydrogels possess tunable mechanical properties that can mimic soft marrow tissue and present three-dimensional (3D) cues. This study evaluates its utility for supporting leukemic cell growth in vitro and its impact on cell survival, growth, and differentiation. Our results showed that the standard viscosity alginates had compromised leukemia cell viability but lower viscosity alginates recovered cell viability and improved 3D cell proliferation (27 fold) compared to 2D cultures (18 fold). Conjugation with RGD peptides promoted further cell growth (43 folds). In general, 3D hydrogels supported high-density cultures better than 2D cultures. Leukemic cells formed densely packed cell clusters in alginate hydrogels and spontaneously differentiated into a more diverse myeloid population. The cell cycle data suggested that more cells go into active cycling with a G2/M arrest in alginate hydrogels and the presence of multiploidy confirmed maturation toward megakaryocytes. In summary, superior culture of leukemia cells in 3D hydrogels is demonstrated in this study accompanied by a potential role of physical cues influencing cell fate decision. Manipulation of biophysical and biochemical properties of alginate hydrogels permits the study of specific interactions and serves to provide a robust 3D platform for studying extrinsic contributions inside the bone marrow.


Asunto(s)
Alginatos/farmacología , Materiales Biomiméticos/farmacología , Médula Ósea , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Leucemia/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ácido Glucurónico/farmacología , Ácidos Hexurónicos/farmacología , Humanos , Células K562 , Leucemia/patología , Megacariocitos/metabolismo , Megacariocitos/patología , Poliploidía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA