Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Healthc Mater ; 13(3): e2301221, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37916912

RESUMO

Vascular damage and reduced tissue perfusion are expected to majorly contribute to the loss of neurons or neural signals around implanted electrodes. However, there are limited methods of controlling the vascular dynamics in tissues surrounding these implants. This work utilizes conducting polymer poly(ethylenedioxythiophene) and sulfonated silica nanoparticle composite (PEDOT/SNP) to load and release a vasodilator, sodium nitroprusside, to controllably dilate the vasculature around carbon fiber electrodes (CFEs) implanted in the mouse cortex. The vasodilator release is triggered via electrical stimulation and the amount of release increases with increasing electrical pulses. The vascular dynamics are monitored in real-time using two-photon microscopy, with changes in vessel diameters quantified before, during, and after the release of the vasodilator into the tissues. This work observes significant increases in vessel diameters when the vasodilator is electrically triggered to release, and differential effects of the drug release on vessels of different sizes. In conclusion, the use of nanoparticle reservoirs in conducting polymer-based drug delivery platforms enables the controlled delivery of vasodilator into the implant environment, effectively altering the local vascular dynamics on demand. With further optimization, this technology could be a powerful tool to improve the neural electrode-tissue interface and study neurovascular coupling.


Assuntos
Nanopartículas , Vasodilatadores , Camundongos , Animais , Dióxido de Silício , Polímeros/farmacologia , Eletrodos Implantados , Encéfalo/fisiologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia
2.
Antioxidants (Basel) ; 11(8)2022 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-36009346

RESUMO

Neural electrode insertion trauma impedes the recording and stimulation capabilities of numerous diagnostic and treatment avenues. Implantation leads to the activation of inflammatory markers and cell types, which is detrimental to neural tissue health and recording capabilities. Oxidative stress and inflammation at the implant site have been shown to decrease with chronic administration of antioxidant melatonin at week 16, but its effects on the acute landscape have not been studied. To assess the effect of melatonin administration in the acute phase, specifically the first week post-implantation, we utilized histological and q-PCR methods to quantify cellular and molecular indicators of inflammation and oxidative stress in the tissue surrounding implanted probes in C57BL/6 mice as well as two-photon microscopy to track the microglial responses to the probes in real-time in transgenic mice expressing GFP with CX3CR1 promotor. Histological results indicate that melatonin effectively maintained neuron density surrounding the electrode, inhibited accumulation and activation of microglia and astrocytes, and reduced oxidative tissue damage. The expression of the pro-inflammatory cytokines, TNF-α and IL-6, were significantly reduced in melatonin-treated animals. Additionally, microglial encapsulation of the implant surface was inhibited by melatonin as compared to control animals following implantation. Our results combined with previous research suggest that melatonin is a particularly suitable drug for modulating inflammatory activity around neural electrode implants both acutely and chronically, translating to more stable and reliable interfaces.

3.
Langmuir ; 38(24): 7512-7521, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35678760

RESUMO

While roughening the surface of neural implants has been shown to significantly improve their performance, the mechanism for this improvement is not understood, preventing systematic optimization of surfaces. Specifically, prior work has shown that the cellular response to a surface can be significantly enhanced by coating the implant surface with inorganic nanoparticles and neuroadhesion protein L1, and this improvement occurs even when the surface chemistry is identical between the nanoparticle-coated and uncoated electrodes, suggesting the critical importance of surface topography. Here, we use transmission electron microscopy to characterize the topography of bare and nanoparticle-coated implants across 7 orders of magnitude in size, from the device scale to the atomic scale. The results reveal multiscale roughness, which cannot be adequately described using conventional roughness parameters. Indeed, the topography is nearly identical between the two samples at the smallest scales and also at the largest scales but vastly different in the intermediate scales, especially in the range of 5-100 nm. Using a multiscale topography analysis, we show that the coating causes a 76% increase in the available surface area for contact and an order-of-magnitude increase in local surface curvature at characteristic sizes corresponding to specific biological structures. These are correlated with a 75% increase in bound proteins on the surface and a 134% increase in neurite outgrowth. The present investigation presents a framework for analyzing the scale-dependent topography of medical device-relevant surfaces, and suggests the most critical size scales that determine the biological response to implanted materials.


Assuntos
Nanopartículas , Titânio , Materiais Revestidos Biocompatíveis/química , Nanopartículas/química , Propriedades de Superfície , Titânio/química
4.
Adv Healthc Mater ; 10(16): e2002150, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34190425

RESUMO

Due to their ability to interface with neural tissues, neural electrodes are the key tool used for neurophysiological studies, electrochemical detection, brain computer interfacing, and countless neuromodulation therapies and diagnostic procedures. However, the long-term applications of neural electrodes are limited by the inflammatory host tissue response, decreasing detectable electrical signals, and insulating the device from the native environment. Surface modification methods are proposed to limit these detrimental responses but each has their own limitations. Here, a combinatorial approach is presented toward creating a stable interface between the electrode and host tissues. First, a thiolated nanoparticle (TNP) coating is utilized to increase the surface area and roughness. Next, the neural adhesion molecule L1 is immobilized to the nanoparticle modified substrate. In vitro, the combined nanotopographical and bioactive modifications (TNP+L1) elevate the bioactivity of L1, which is maintained for 28 d. In vivo, TNP+L1 modification improves the recording performance of the neural electrode arrays compared to TNP or L1 modification alone. Postmortem histology reveals greater neural cell density around the TNP+L1 coating while eliminating any inflammatory microglial encapsulation after 4 weeks. These results demonstrate that nanotopographical and bioactive modifications synergistically produce a seamless neural tissue interface for chronic neural implants.


Assuntos
Nanopartículas , Molécula L1 de Adesão de Célula Nervosa , Eletrodos Implantados , Humanos , Microeletrodos , Neurônios
5.
Front Bioeng Biotechnol ; 8: 602216, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33330433

RESUMO

Melatonin (MT) has been recently considered an excellent candidate for the treatment of sleep disorders, neural injuries, and neurological diseases. To better investigate the actions of MT in various brain functions, real-time detection of MT concentrations in specific brain regions is much desired. Previously, we have demonstrated detection of exogenously administered MT in anesthetized mouse brain using square wave voltammetry (SWV). Here, for the first time, we show successful detection of exogenous MT in the brain using fast scan cyclic voltammetry (FSCV) on electrochemically pre-activated carbon fiber microelectrodes (CFEs). In vitro evaluation showed the highest sensitivity (28.1 nA/µM) and lowest detection limit (20.2 ± 4.8 nM) ever reported for MT detection at carbon surface. Additionally, an extensive CFE stability and fouling assessment demonstrated that a prolonged CFE pre-conditioning stabilizes the background, in vitro and in vivo, and provides consistent CFE sensitivity over time even in the presence of a high MT concentration. Finally, the stable in vivo background, with minimized CFE fouling, allows us to achieve a drift-free FSCV detection of exogenous administered MT in mouse brain over a period of 3 min, which is significantly longer than the duration limit (usually < 90 s) for traditional in vivo FSCV acquisition. The MT concentration and dynamics measured by FSCV are in good agreement with SWV, while microdialysis further validated the concentration range. These results demonstrated reliable MT detection using FSCV that has the potential to monitor MT in the brain over long periods of time.

6.
Biosens Bioelectron ; 155: 112096, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32090868

RESUMO

Intracortical microelectrodes are being developed to both record and stimulate neurons to understand brain circuitry or restore lost functions. However, the success of these probes is hampered partly due to the inflammatory host tissue responses to implants. To minimize the foreign body reactions, L1, a brain derived neuronal specific cell adhesion molecule, has been covalently bound to the neural electrode array surface. Here we evaluated the chronic recording performance of L1-coated silicon based laminar neural electrode arrays implanted into V1m cortex of mice. The L1 coating enhanced the overall visually evoked single-unit (SU) yield and SU amplitude, as well as signal-to-noise-ratio (SNR) in the mouse brain compared to the uncoated arrays across the 0-1500 µm depth. The improvement in recording is most dramatic in the hippocampus region, where the control group showed severe recording yield decrease after one week, while the L1 implants maintained a high SU yield throughout the 16 weeks. Immunohistological analysis revealed significant increases of axonal and neuronal density along with significantly lowered microglia activation around the L1 probe after 16 weeks. These results collectively confirm the effectiveness of L1 based biomimetic coating on minimizing inflammatory tissue response and improving neural recording quality and longevity. Improving chronic recording will benefit the brain-computer interface technologies and neuroscience studies involving chronic tracking of neural activities.


Assuntos
Encéfalo/fisiologia , Moléculas de Adesão Celular , Materiais Revestidos Biocompatíveis , Eletrônica/métodos , Neurônios/fisiologia , Proteínas , Animais , Axônios , Barreira Hematoencefálica/metabolismo , Moléculas de Adesão Celular/química , Sobrevivência Celular , Espectroscopia Dielétrica , Eletrodos Implantados , Eletrônica/normas , Fenômenos Eletrofisiológicos , Imuno-Histoquímica , Camundongos , Microeletrodos , Permeabilidade , Proteínas/química
7.
Adv Healthc Mater ; 8(21): e1900622, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31583857

RESUMO

In order to address material limitations of biologically interfacing electrodes, modified silica nanoparticles are utilized as dopants for conducting polymers. Silica precursors are selected to form a thiol modified particle (TNP), following which the particles are oxidized to sulfonate modified nanoparticles (SNPs). The selective inclusion of hexadecyl trimethylammonium bromide allows for synthesis of both porous and nonporous SNPs. Nonporous nanoparticle doped polyethylenedioxythiophene (PEDOT) films possess low interfacial impedance, high charge injection (4.8 mC cm-2 ), and improved stability under stimulation compared to PEDOT/poly(styrenesulfonate). Porous SNP dopants can serve as drug reservoirs and greatly enhance the capability of conducting polymer-based, electrically controlled drug release technology. Using the SNP dopants, drug loading and release is increased up to 16.8 times, in addition to greatly expanding the range of drug candidates to include both cationic and electroactive compounds, all while maintaining their bioactivity. Finally, the PEDOT/SNP composite is capable of precisely modulating neural activity in vivo by timed release of a glutamate receptor antagonist from coated microelectrode sites. Together, this work demonstrates the feasibility and potential of doping conducting polymers with engineered nanoparticles, creating countless options to produce composite materials for enhanced electrical stimulation, neural recording, chemical sensing, and on demand drug delivery.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/química , Nanopartículas/química , Polímeros/química , Sistemas de Liberação de Medicamentos/métodos , Microeletrodos
8.
Adv Healthc Mater ; 8(9): e1801311, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30843365

RESUMO

State-of-the-art intraneural electrodes made from silicon or polyimide substrates have shown promise in selectively modulating efferent and afferent activity in the peripheral nervous system. However, when chronically implanted, these devices trigger a multiphase foreign body response ending in device encapsulation. The presence of encapsulation increases the distance between the electrode and the excitable tissue, which not only reduces the recordable signal amplitude but also requires increased current to activate nearby axons. Herein, this study reports a novel conducting polymer based intraneural electrode which has Young's moduli similar to that of nerve tissue. The study first describes material optimization of the soft wire conductive matrix and evaluates their mechanical and electrochemical properties. Second, the study demonstrates 3T3 cell survival when cultured with media eluted from the soft wires. Third, the study presents acute in vivo functionality for stimulation of peripheral nerves to evoke force and compound muscle action potential in a rat model. Furthermore, comprehensive histological analyses show that soft wires elicit significantly less scar tissue encapsulation, less changes to axon size, density and morphology, and reduced macrophage activation compared to polyimide implants in the sciatic nerves at 1 month postimplantation.


Assuntos
Elastômeros/química , Microeletrodos , Nervos Periféricos/citologia , Células 3T3 , Animais , Sobrevivência Celular/fisiologia , Eletroquímica , Ativação de Macrófagos/fisiologia , Camundongos , Microscopia Eletrônica de Varredura , Nanotubos de Carbono/química , Nanotubos de Carbono/ultraestrutura , Nervos Periféricos/ultraestrutura , Ratos
9.
J Nanobiotechnology ; 16(1): 13, 2018 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-29433522

RESUMO

BACKGROUND: Oxidative stress acts as a trigger in the course of neurodegenerative diseases and neural injuries. An antioxidant-based therapy can be effective to ameliorate the deleterious effects of oxidative stress. Resveratrol (RSV) has been shown to be effective at removing excess reactive oxygen species (ROS) or reactive nitrogen species generation in the central nervous system (CNS), but the delivery of RSV into the brain through systemic administration is inefficient. Here, we have developed a RSV delivery vehicle based on polylactic acid (PLA)-coated mesoporous silica nanoparticles (MSNPs), conjugated with a ligand peptide of low-density lipoprotein receptor (LDLR) to enhance their transcytosis across the blood-brain barrier (BBB). RESULTS: Resveratrol was loaded into MSNPs (average diameter 200 nm, pore size 4 nm) at 16 µg/mg (w/w). As a gatekeeper, the PLA coating prevented the RSV burst release, while ROS was shown to trigger the drug release by accelerating PLA degradation. An in vitro BBB model with a co-culture of rat brain microvascular endothelial cells (RBECs) and microglia cells using Transwell chambers was established to assess the RSV delivery across BBB. The conjugation of LDLR ligand peptides markedly enhanced the migration of MSNPs across the RBECs monolayer. RSV could be released and effectively reduce the activation of the microglia cells stimulated by phorbol-myristate-acetate or lipopolysaccharide. CONCLUSIONS: These ROS responsive LDLR peptides conjugated PLA-coated MSNPs have great potential for oxidative stress therapy in CNS.


Assuntos
Antioxidantes/administração & dosagem , Barreira Hematoencefálica/metabolismo , Preparações de Ação Retardada/química , Poliésteres/química , Receptores de LDL/química , Dióxido de Silício/química , Estilbenos/administração & dosagem , Animais , Antioxidantes/farmacocinética , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/metabolismo , Nanopartículas/química , Peptídeos/química , Ratos , Espécies Reativas de Oxigênio/metabolismo , Resveratrol , Estilbenos/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...