Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biosens Bioelectron ; 155: 112096, 2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32090868

RESUMEN

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.


Asunto(s)
Encéfalo/fisiología , Moléculas de Adhesión Celular , Materiales Biocompatibles Revestidos , Electrónica/métodos , Neuronas/fisiología , Proteínas , Animales , Axones , Barrera Hematoencefálica/metabolismo , Moléculas de Adhesión Celular/química , Supervivencia Celular , Espectroscopía Dieléctrica , Electrodos Implantados , Electrónica/normas , Fenómenos Electrofisiológicos , Inmunohistoquímica , Ratones , Microelectrodos , Permeabilidad , Proteínas/química
2.
Biomaterials ; 161: 117-128, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29421549

RESUMEN

Intracortical microelectrode arrays, especially the Utah array, remain the most common choice for obtaining high dimensional recordings of spiking neural activity for brain computer interface and basic neuroscience research. Despite the widespread use and established design, mechanical, material and biological challenges persist that contribute to a steady decline in recording performance (as evidenced by both diminished signal amplitude and recorded cell population over time) or outright array failure. Device implantation injury causes acute cell death and activation of inflammatory microglia and astrocytes that leads to a chronic neurodegeneration and inflammatory glial aggregation around the electrode shanks and often times fibrous tissue growth above the pia along the bed of the array within the meninges. This multifaceted deleterious cascade can result in substantial variability in performance even under the same experimental conditions. We track both impedance signatures and electrophysiological performance of 4 × 4 floating microelectrode Utah arrays implanted in the primary monocular visual cortex (V1m) of Long-Evans rats over a 12-week period. We employ a repeatable visual stimulation method to compare signal-to-noise ratio as well as single- and multi-unit yield from weekly recordings. To explain signal variability with biological response, we compare arrays categorized as either Type 1, partial fibrous encapsulation, or Type 2, complete fibrous encapsulation and demonstrate performance and impedance signatures unique to encapsulation type. We additionally assess benefits of a biomolecule coating intended to minimize distance to recordable units and observe a temporary improvement on multi-unit recording yield and single-unit amplitude.


Asunto(s)
Biomimética/métodos , Impedancia Eléctrica , Animales , Fenómenos Electrofisiológicos , Microelectrodos , Ratas
3.
Soft Matter ; 11(24): 4847-61, 2015 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-25993261

RESUMEN

Current designs for microelectrodes used for interfacing with the nervous system elicit a characteristic inflammatory response that leads to scar tissue encapsulation, electrical insulation of the electrode from the tissue and ultimately failure. Traditionally, relatively stiff materials like tungsten and silicon are employed which have mechanical properties several orders of magnitude different from neural tissue. This mechanical mismatch is thought to be a major cause of chronic inflammation and degeneration around the device. In an effort to minimize the disparity between neural interface devices and the brain, novel soft electrodes consisting of elastomers and intrinsically conducting polymers were fabricated. The physical, mechanical and electrochemical properties of these materials were extensively characterized to identify the formulations with the optimal combination of parameters including Young's modulus, elongation at break, ultimate tensile strength, conductivity, impedance and surface charge injection. Our final electrode has a Young's modulus of 974 kPa which is five orders of magnitude lower than tungsten and significantly lower than other polymer-based neural electrode materials. In vitro cell culture experiments demonstrated the favorable interaction between these soft materials and neurons, astrocytes and microglia, with higher neuronal attachment and a two-fold reduction in inflammatory microglia attachment on soft devices compared to stiff controls. Surface immobilization of neuronal adhesion proteins on these microwires further improved the cellular response. Finally, in vivo electrophysiology demonstrated the functionality of the elastomeric electrodes in recording single unit activity in the rodent visual cortex. The results presented provide initial evidence in support of the use of soft materials in neural interface applications.


Asunto(s)
Materiales Biocompatibles/química , Electrofisiología/instrumentación , Nanocables/química , Neuronas/fisiología , Elastómeros de Silicona/química , Animales , Materiales Biocompatibles/efectos adversos , Células Cultivadas , Módulo de Elasticidad , Conductividad Eléctrica , Electrofisiología/métodos , Microelectrodos , Nanocables/efectos adversos , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Elastómeros de Silicona/efectos adversos
4.
Biomaterials ; 32(3): 681-92, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20933270

RESUMEN

Brain tissue inflammatory responses, including neuronal loss and gliosis at the neural electrode/tissue interface, limit the recording stability and longevity of neural probes. The neural adhesion molecule L1 specifically promotes neurite outgrowth and neuronal survival. In this study, we covalently immobilized L1 on the surface of silicon-based neural probes and compared the tissue response between L1 modified and non-modified probes implanted in the rat cortex after 1, 4, and 8 weeks. The effect of L1 on neuronal health and survival, and glial cell reactions were evaluated with immunohistochemistry and quantitative image analysis. Similar to previous findings, persistent glial activation and significant decreases of neuronal and axonal densities were found at the vicinity of the non-modified probes. In contrast, the immediate area (100 µm) around the L1 modified probe showed no loss of neuronal bodies and a significantly increased axonal density relative to background. In this same region, immunohistochemistry analyses show a significantly lower activation of microglia and reaction of astrocytes around the L1 modified probes when compared to the control probes. These improvements in tissue reaction induced by the L1 coating are likely to lead to improved functionality of the implanted neural electrodes during chronic recordings.


Asunto(s)
Gliosis/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Gliosis/patología , Inmunohistoquímica , Masculino , Microglía/efectos de los fármacos , Modelos Biológicos , Molécula L1 de Adhesión de Célula Nerviosa/química , Ratas , Ratas Sprague-Dawley
5.
Acta Biomater ; 4(5): 1208-17, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18420473

RESUMEN

Silicon-based implantable neural electrode arrays are known to experience failure during long-term recording, partially due to host tissue responses. Surface modification and immobilization of biomolecules may provide a means to improve their biocompatibility and integration within the host brain tissue. Previously, the laminin biomolecule or laminin fragments have been used to modify the neural probe's silicon surface to promote neuronal attachment and growth. Here we report the successful immobilization of the L1 biomolecule on a silicon surface. L1 is a neuronal adhesion molecule that can specifically promote neurite outgrowth and neuronal survival. Silane chemistry and the heterobifunctional coupling agent 4-maleimidobutyric acid N-hydroxysuccinimide ester (GMBS) were used to covalently bind these two biomolecules onto the surface of silicon dioxide wafers, which mimic the surface of silicon-based implantable neural probes. After covalent binding of the biomolecules, polyethylene glycol (PEG)-NH(2) was used to cap the unreacted GMBS groups. Surface immobilization was verified by goniometry, dual polarization interferometry, and immunostaining techniques. Primary murine neurons or astrocytes were used to evaluate the modified silicon surfaces. Both L1- and laminin-modified surfaces promoted neuronal attachment, while the L1-modified surface demonstrated significantly enhanced levels of neurite outgrowth (p<0.05). In addition, the laminin-modified surface promoted astrocyte attachment, while the L1-modified surface showed significantly reduced levels of astrocyte attachment relative to the laminin-modified surface and other controls (p<0.05). These results demonstrate the ability of the L1-immobilized surface to specifically promote neuronal growth and neurite extension, while inhibiting the attachment of astrocytes, one of the main cellular components of the glial sheath. Such unique properties present vast potentials to improve the biocompatibility and chronic recording performance of neural probes.


Asunto(s)
Astrocitos/fisiología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Microelectrodos , Molécula L1 de Adhesión de Célula Nerviosa/farmacología , Neuronas/citología , Neuronas/fisiología , Adsorción , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Materiales , Molécula L1 de Adhesión de Célula Nerviosa/química , Neuronas/efectos de los fármacos , Polietilenglicoles/química , Unión Proteica , Ratas , Ratas Sprague-Dawley , Dióxido de Silicio/química , Propiedades de Superficie
6.
Biosens Bioelectron ; 22(8): 1723-32, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17015008

RESUMEN

There is continued interest in the development of conductive polymer coatings to improve the electrical properties and biocompatibility of electrodes for neural prostheses. We present here a new type of coating, based on mixed self-assembled monolayers (SAMs) of thiolated poly(alkylthiophene)s and functionalized alkanethiols. When assembled as a SAM on electrodes designed for in vitro electrophysiology, these polymers are able to significantly lower electrode impedance at 1 kHz. The same mixed formulation is able to promote the outgrowth of neurites from primary mouse cortical neurons when the alkanethiol component is functionalized with a neural cell adhesion molecule (NCAM) binding antibody. Atomic force microscopy of the SAMs shows that they exert their effect through the well-known mechanism of increasing electrode surface area. These new covalently bound films have the potential to be more robust and are more controllable in their composition than existing electrodeposited conductive polymer coatings.


Asunto(s)
Técnicas Biosensibles/instrumentación , Electrodos Implantados , Ensayo de Materiales , Neuronas/fisiología , Polímeros/química , Tiofenos/química , Animales , Células Cultivadas , Impedancia Eléctrica , Electrofisiología , Femenino , Ratones , Nanotecnología , Neuritas/fisiología , Prótesis e Implantes
7.
J Pharmacol Exp Ther ; 317(3): 1320-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16547166

RESUMEN

Methylisothiazolinone (MIT) is a biocide widely used in industrial and cosmetic products with potential as a neurotoxicant. We previously reported that short acute exposures to relatively high concentrations of MIT (100 microM) lead to widespread and selective neuronal death in vitro. To evaluate the biological properties of chronic exposures to MIT, freshly dissociated rat cortical neurons were continuously exposed to low concentrations (0.1-3 microM) of the biocide in serum-containing media. Although we observed minimal effects on cell viability, MIT induced a dramatic inhibition of neurite outgrowth. Immunoblotting and immunoprecipitation experiments revealed that focal adhesion kinase (FAK) phosphorylation was primarily affected by the MIT treatment. The phosphorylation level at tyrosines 576 and 861 of FAK was significantly decreased and likely contributed to the overall reduction of tyrosine phosphorylation of this protein. MIT inhibited Src family kinases (SFKs) in cell-free assays and led to the physical dissociation of FAK from the signaling complexes that it normally forms with c-Src and Fyn in developing neurons. High-density neuronal cultures were then employed to increase cell-to-cell contact. This approach resulted in an overall enhancement of SFKs and FAK phosphorylation and could overcome the deficits induced by MIT. This study suggests that a disruption of FAK-SFK complexes due to SFK inhibition leads to FAK dysfunction, with detrimental effects to immature neurons. Prolonged exposure to low levels of MIT and related compounds may have damaging consequences to the developing nervous system.


Asunto(s)
Corteza Cerebral/embriología , Desinfectantes/toxicidad , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neuronas/efectos de los fármacos , Tiazoles/toxicidad , Familia-src Quinasas/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Neuronas/enzimología , Neuronas/patología , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores
8.
J Control Release ; 110(3): 531-41, 2006 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-16360955

RESUMEN

Chronic recordings from micromachined neural electrode arrays often fail a few weeks after implantation primarily due to the formation of an astro-glial sheath around the implant. We propose a drug delivery system, from conducting polymer (CP) coatings on the electrode sites, to modulate the inflammatory implant-host tissue reaction. In this study, polypyrrole (PPy) based coatings for electrically controlled and local delivery of the ionic form of an anti-inflammatory drug, dexamethasone (Dex), was investigated. The drug was incorporated in PPy via electropolymerization of pyrrole and released in PBS using cyclic voltammetry (CV). FTIR analysis of the surface showed the presence of Dex and polypyrrole on the coated electrode. The thickness of the coated film was estimated to be approximately 50 nm by ellipsometry. We are able to release 0.5 mug/cm(2) Dex in 1 CV cycle and a total of almost 16 mug/cm(2) Dex after 30 CV cycles. In vitro studies and immunocytochemistry on murine glial cells suggest that the released drug lowers the count of reactive astrocytes to the same extent as the added drug. In addition, the released drug is not toxic to neurons as seen by healthy neuronal viability in the released drug treated cells.


Asunto(s)
Dexametasona/química , Polímeros/química , Pirroles/química , Animales , Células Cultivadas , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Dexametasona/farmacocinética , Electroquímica , Electrodos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Polímeros/farmacocinética , Pirroles/farmacocinética
9.
J Leukoc Biol ; 76(6): 1187-99, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15356097

RESUMEN

Activated, adherent natural killer (A-NK) cells represent a distinct subpopulation of interleukin (IL)-2-stimulated NK cells, which are selectively endowed with the increased expression of integrins and ability to adhere to solid surfaces, migrate into, infiltrate, and destroy cancerous tissues. The present study defines the phenotype and functions of precursors of A-NK (pre-A-NK) cells in humans. Peripheral blood pre-A-NK cells, in contrast to the rest of NK cells, express a novel epitope of CD56 neuronal cell adhesion molecule, termed ANK-1, and increased cell-surface levels of integrins. Pre-A-NK cells also express low levels of CD56 and CD161, and some express CD162 receptor, do not express CD25 or activation markers, and are effective mediators of NK cytotoxicity. Thus, pre-A-NK cells are generally similar to CD56(dim) NK cells. However, pre-A-NK cells differ from the main NK cell subpopulation by having a lower expression level of CD16 and a lower ability to mediate redirected antibody-dependent, cell-mediated cytotoxicity. More importantly, pre-A-NK cells are preferentially endowed with the ability to rapidly respond to IL-2 by integrin-mediated adherence to endothelial cells, extracellular matrix, and plastic. This early, specific response of pre-A-NK cells to IL-2 is followed by their activation, vigorous proliferation, and differentiation into phenotypically and functionally similar A-NK cells. Pre-A-NK cells represent only approximately 26% of peripheral blood NK cells but encompass the majority of NK cells in normal and cancerous, solid tissues. We conclude that pre-A-NK cells represent a distinct subset of resting, mature NK cells with the characteristics indicative of their ability to migrate and reside in solid tissues.


Asunto(s)
Antígeno CD56/inmunología , Epítopos/inmunología , Células Asesinas Naturales/metabolismo , Células Madre/metabolismo , Anticuerpos Monoclonales/inmunología , Antígenos de Superficie/inmunología , Antígenos de Superficie/metabolismo , Antígeno CD56/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/inmunología , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/inmunología , Citotoxicidad Inmunológica/inmunología , Humanos , Inmunofenotipificación , Integrinas/inmunología , Integrinas/metabolismo , Interleucina-2/inmunología , Interleucina-2/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Subfamilia B de Receptores Similares a Lectina de Células NK , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Células Madre/efectos de los fármacos , Células Madre/inmunología
10.
J Immunol ; 172(7): 4091-9, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15034021

RESUMEN

Src homology 2 domain-containing protein tyrosine phosphatase substrate 1 (SHPS-1) is a member of the signal regulatory protein family in which the extracellular region interacts with its ligand, CD47. Recent studies have demonstrated that SHPS-1 plays an important role in cell migration and cell adhesion. We demonstrate in this study, using immunohistochemical and flow cytometric analyses, that murine Langerhans cells (LCs) express SHPS-1. Treatment of mice ears with 2,4-dinitro-1-fluorobenzene significantly reduced the number of epidermal LCs, and that reduction could be reversed by pretreatment with mAb to SHPS-1 or the CD47-Fc fusion protein. Treatment with the SHPS-1 mAb in vivo reduced the number of FITC-bearing cells in the lesional lymph nodes after the application of FITC to the skin. The SHPS-1 mAb inhibited the in vivo TNF-alpha-induced migration of LCs. The emigration of dendritic cells expressing I-A(b+) from skin explants to the medium was also reduced by the SHPS-1 mAb. We further demonstrate that the chemotaxis of a murine dendritic cell line, XS52, by macrophage inflammatory protein-3beta was significantly inhibited by treatment with the SHPS-1 mAb or CD47-Fc recombinant protein. Finally, we show that migration of LCs was attenuated in mutant mice that lack the intracellular domain of SHPS-1. These observations show that the ligation of SHPS-1 with the SHPS-1 mAb or with CD47-Fc abrogates the migration of LCs in vivo and in vitro, which suggests that the SHPS-1-CD47 interaction may negatively regulate LC migration.


Asunto(s)
Antígenos de Diferenciación/fisiología , Movimiento Celular/inmunología , Células Epidérmicas , Epidermis/metabolismo , Células de Langerhans/citología , Células de Langerhans/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Glicoproteínas de Membrana/fisiología , Molécula L1 de Adhesión de Célula Nerviosa/fisiología , Receptores Inmunológicos/fisiología , Administración Tópica , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD/administración & dosificación , Antígenos CD/genética , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígeno CD11c/biosíntesis , Antígeno CD47 , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/genética , Recuento de Células , Línea Celular , Inhibición de Migración Celular , Medios de Cultivo , Dinitrofluorobenceno/administración & dosificación , Epidermis/inmunología , Femenino , Inhibidores de Crecimiento/administración & dosificación , Haptenos/administración & dosificación , Haptenos/biosíntesis , Antígenos de Histocompatibilidad Clase II/biosíntesis , Fragmentos Fc de Inmunoglobulinas/genética , Inyecciones Intradérmicas , Interleucina-4/farmacología , Células de Langerhans/inmunología , Ganglios Linfáticos/patología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Molécula L1 de Adhesión de Célula Nerviosa/biosíntesis , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/inmunología , Técnicas de Cultivo de Órganos , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA