Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuromodulation ; 25(6): 789-795, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33438369

RESUMO

OBJECTIVES: Microelectrode arrays offer a means to probe the functional circuitry of the brain and the promise of cortical neuroprosthesis for individuals suffering from paralysis or limb loss. These devices are typically comprised of one or more shanks incorporating microelectrode sites, where the shanks are positioned by inserting the devices along a straight path that is normal to the brain surface. The lack of consistent long-term chronic recording technology has driven interest in novel probe design and approaches that go beyond the standard insertion approach that is limited to a single velocity or axis. This review offers a description of typical approaches and associated limitations and surveys emergent methods for implantation of microelectrode arrays, in particular those new approaches that leverage embedded microactuators and extend the insertion direction beyond a single axis. MATERIALS AND METHODS: This review paper surveys the current technologies that enable probe implantation, repositioning, and the capability to record/stimulate from a tissue volume. A comprehensive literature search was performed using PubMed, Web of Science, and Google Scholar. RESULTS: There has been substantial innovation in the development of microscale and embedded technology that enables probe repositioning to maintain quality recordings in the brain. Innovations in material science have resulted in novel strategies for deployable structures that can record from or stimulate a tissue volume. Moreover, new developments involving magnetically steerable catheters and needles offer an alternative approach to "pull" rather than "push" a probe into the tissue. CONCLUSION: We envision the emergence of a new generation of probes and insertion methodologies for neuromodulation applications that enable reliable chronic performance from devices that can be positioned virtually anywhere in the brain.


Assuntos
Encéfalo , Eletrodos Implantados , Humanos , Microeletrodos
2.
Micromachines (Basel) ; 12(7)2021 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-34203234

RESUMO

Several recent studies have reported improved histological and electrophysiological outcomes with soft neural interfaces that have elastic moduli ranging from 10 s of kPa to hundreds of MPa. However, many of these soft interfaces use custom fabrication processes. We test the hypothesis that a readily adoptable fabrication process for only coating the tips of microelectrodes with soft brain-like (elastic modulus of ~5 kPa) material improves the long-term electrical performance of neural interfaces. Conventional tungsten microelectrodes (n = 9 with soft coatings and n = 6 uncoated controls) and Pt/Ir microelectrodes (n = 16 with soft coatings) were implanted in six animals for durations ranging from 5 weeks to over 1 year in a subset of rats. Electrochemical impedance spectroscopy was used to assess the quality of the brain tissue-electrode interface under chronic conditions. Neural recordings were assessed for unit activity and signal quality. Electrodes with soft, silicone coatings showed relatively stable electrical impedance characteristics over 6 weeks to >1 year compared to the uncoated control electrodes. Single unit activity recorded by coated electrodes showed larger peak-to-peak amplitudes and increased number of detectable neurons compared to uncoated controls over 6-7 weeks. We demonstrate the feasibility of using a readily translatable process to create brain-like soft interfaces that can potentially overcome variable performance associated with chronic rigid neural interfaces.

3.
J Neural Eng ; 18(4)2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34111852

RESUMO

Objective. Respiration and vascular pulsation cause relative micromotion of brain tissue against stationary implants resulting in repetitive displacements of 2-4µm (due to vascular pulsation) and 10-30µm (due to breathing) in rats. However, the direct functional impact of such tissue micromotion on the cells at the neural interface remains unknown. This study aims to test the hypothesis that micromotion in brain tissue causes changes in membrane potentials (MPs) through the activation of mechanosensitive ion channels.Approach. Intracellular MPs were recorded from Aplysia ganglion cells (n= 8) and cortical cells (n= 15)in vivoinn= 7 adult rats. Cyclic stresses between 0.2 and 4 kPa repeated at 1 Hz were tested in Aplysia ganglion cells. For thein vivoexperiments, 30µM of gadolinium chloride (Gd3+), a non-selective blocker of mechanosensitive ion channels, was used to assess the role of such ion channels.Main results. In Aplysia ganglion cells, there were no MP changes for <1.5 kPa, and action potentials were observed at >3.1 kPa. Drug studies utilizing 5-HT showed an 80% reduction in firing frequency from controls. Inin vivoexperiments, periodic pulsations (1-10 mV) were observed in the MPs of cells that corresponded to breathing and heart-rate. In response to the addition of 30µM Gd3+, we observed a significant reduction (0.5-3 mV) in the periodic pulsations in MP in all cortical cells across four different rats, suggesting the role of mechanosensitive ion channels in mediating MP fluctuations due to tissue micromotion at the neural interface.Significance.Under chronic conditions, the tissue at the interface stiffens due to scar tissue formation, which is expected to increase the likelihood of recruiting stretch-receptors due to tissue micromotion. It is speculated that such chronic sub-threshold pulsations in MPs might trigger the immune response at the neural interface.


Assuntos
Membranas Intracelulares , Próteses e Implantes , Animais , Ratos
4.
Biomed Phys Eng Express ; 6(2): 025003, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33438629

RESUMO

OBJECTIVE: There is a need for low power, scalable photoelectronic devices and systems for emerging optogenetic needs in neuromodulation. Conventional light emitting diodes (LEDs) are constrained by power and lead-counts necessary for scalability. Organic LEDs (OLEDs) offer an exciting approach to decrease power and lead-counts while achieving high channel counts on thin, flexible substrates that conform to brain surfaces or peripheral neuronal fibers. In this study, we investigate the potential for using OLEDs to modulate neuronal networks cultured in vitro on a transparent microelectrode array (MEA) and subsequently validate neurostimulation in vivo in a transgenic mouse model. APPROACH: Cultured mouse cortical neurons were transfected with light-sensitive opsins such as blue-light sensitive channel-rhodopsin (ChR2) and green-light sensitive chimeric channel-rhodopsin (C1V1tt) and stimulated using blue and green OLEDs (with 455 and 520 nm peak emission spectra respectively) at a power of ~1 mW mm-2 under pulsed conditions. MAIN RESULTS: We demonstrate neuromodulation and optostimulus-locked, single unit-neuronal activity in neurons expressing stimulating opsins (34 units on n = 4 MEAs, each with 16 recordable channels). We also validated the optostimulus-locked response in preliminary experiments in a channel-rhodopsin expressing transgenic mouse model, where at least three isolatable single neuronal cortical units respond to OLED stimulation. SIGNIFICANCE: The above results indicate the feasibility of generating sufficient luminance from OLEDs to perform neuromodulation both in vitro and in vivo. This opens up the possibility of developing thin, flexible OLED films with multiple stimulation sites that can conform to the shape of the neuronal targets in the brain or the peripheral nervous system. However, stability of these OLEDs under chronic conditions still needs to be carefully assessed with appropriate packaging approaches.


Assuntos
Channelrhodopsins/fisiologia , Eletrodos , Luz , Neurônios/fisiologia , Optogenética , Estimulação Luminosa/métodos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/efeitos da radiação
5.
Microsyst Nanoeng ; 6: 1, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34567616

RESUMO

Conventional electrodes and associated positioning systems for intracellular recording from single neurons in vitro and in vivo are large and bulky, which has largely limited their scalability. Further, acquiring successful intracellular recordings is very tedious, requiring a high degree of skill not readily achieved in a typical laboratory. We report here a robotic, MEMS-based intracellular recording system to overcome the above limitations associated with form factor, scalability, and highly skilled and tedious manual operations required for intracellular recordings. This system combines three distinct technologies: (1) novel microscale, glass-polysilicon penetrating electrode for intracellular recording; (2) electrothermal microactuators for precise microscale movement of each electrode; and (3) closed-loop control algorithm for autonomous positioning of electrode inside single neurons. Here we demonstrate the novel, fully integrated system of glass-polysilicon microelectrode, microscale actuators, and controller for autonomous intracellular recordings from single neurons in the abdominal ganglion of Aplysia californica (n = 5 cells). Consistent resting potentials (<-35 mV) and action potentials (>60 mV) were recorded after each successful penetration attempt with the controller and microactuated glass-polysilicon microelectrodes. The success rate of penetration and quality of intracellular recordings achieved using electrothermal microactuators were comparable to that of conventional positioning systems. Preliminary data from in vivo experiments in anesthetized rats show successful intracellular recordings. The MEMS-based system offers significant advantages: (1) reduction in overall size for potential use in behaving animals, (2) scalable approach to potentially realize multi-channel recordings, and (3) a viable method to fully automate measurement of intracellular recordings. This system will be evaluated in vivo in future rodent studies.

6.
Front Neurosci ; 13: 518, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31213969

RESUMO

The overall goal of this study is to develop thickness shear mode (TSM) resonators for the real-time, label-free, non-destructive sensing of biological adhesion events in small populations (hundreds) of neurons, in a cell culture medium and subsequently in vivo in the future. Such measurements will enable the discovery of the role of biomechanical events in neuronal function and dysfunction. Conventional TSM resonators have been used for chemical sensing and biosensing applications in media, with hundreds of thousands of cells in culture. However, the sensitivity and spatial resolution of conventional TSM devices need to be further enhanced for sensing smaller cell populations or molecules of interest. In this report, we focus on key challenges such as eliminating inharmonics in solution and maximizing Q-factor while simultaneously miniaturizing the active sensing (electrode) area to make them suitable for small populations of cells. We used theoretical expressions for sensitivity and electrode area of TSM sensors operating in liquid. As a validation of the above design effort, we fabricated prototype TSM sensors with resonant frequencies of 42, 47, 75, and 90 MHz and characterized their performance in liquid using electrode diameters of 150, 200, 400, 800, and 1,200 µm and electrode thicknesses of 33 and 230 nm. We validated a candidate TSM resonator with the highest sensitivity and Q-factor for real-time monitoring of the adhesion of cortical neurons. We reduced the size of the sensing area to 150-400 µm for TSM devices, improving the spatial resolution by monitoring few 100-1,000s of neurons. Finally, we modified the electrode surface with single-walled carbon nanotubes (SWCNT) to further enhance adhesion and sensitivity of the TSM sensor to adhering neurons (Marx, 2003).

7.
Micromachines (Basel) ; 9(11)2018 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-30441831

RESUMO

We demonstrate a method of neurostimulation using implanted, free-floating, inter-neural diodes. They are activated by volume-conducted, high frequency, alternating current (AC) fields and address the issue of instability caused by interconnect wires in chronic nerve stimulation. The aim of this study is to optimize the set of AC electrical parameters and the diode features to achieve wireless neurostimulation. Three different packaged Schottky diodes (1.5 mm, 500 µm and 220 µm feature sizes) were tested in vivo (n = 17 rats). A careful assessment of sciatic nerve activation as a function of diode⁻dipole lengths and relative position of the diode was conducted. Subsequently, free-floating Schottky microdiodes were implanted in the nerve (n = 3 rats) and stimulated wirelessly. Thresholds for muscle twitch responses increased non-linearly with frequency. Currents through implanted diodes within the nerve suffer large attenuations (~100 fold) requiring 1⁻2 mA drive currents for thresholds at 17 µA. The muscle recruitment response using electromyograms (EMGs) is intrinsically steep for subepineurial implants and becomes steeper as diode is implanted at increasing depths away from external AC stimulating electrodes. The study demonstrates the feasibility of activating remote, untethered, implanted microscale diodes using external AC fields and achieving neurostimulation.

8.
Biomed Microdevices ; 18(4): 72, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27457752

RESUMO

Emerging neural prosthetics require precise positional tuning and stable interfaces with single neurons for optimal function over a lifetime. In this study, we report an autonomous control to precisely navigate microscale electrodes in soft, viscoelastic brain tissue without visual feedback. The autonomous control optimizes signal-to-noise ratio (SNR) of single neuronal recordings in viscoelastic brain tissue while maintaining quasi-static mechanical stress conditions to improve stability of the implant-tissue interface. Force-displacement curves from microelectrodes in in vivo rodent experiments are used to estimate viscoelastic parameters of the brain. Using a combination of computational models and experiments, we determined an optimal movement for the microelectrodes with bidirectional displacements of 3:2 ratio between forward and backward displacements and a inter-movement interval of 40 s for minimizing mechanical stress in the surrounding brain tissue. A regulator with the above optimal bidirectional motion for the microelectrodes in in vivo experiments resulted in significant reduction in the number of microelectrode movements (0.23 movements/min) and longer periods of stable SNR (53 % of the time) compared to a regulator using a conventional linear, unidirectional microelectrode movement (with 1.48 movements/min and stable SNR 23 % of the time).


Assuntos
Encéfalo/fisiologia , Eletrodos Implantados , Fenômenos Mecânicos , Neurônios/fisiologia , Animais , Desenho de Equipamento , Masculino , Microeletrodos , Movimento (Física) , Ratos , Ratos Sprague-Dawley , Razão Sinal-Ruído , Estresse Mecânico
9.
J Neural Eng ; 12(3): 036002, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25834105

RESUMO

OBJECTIVE: The objective of this research is to characterize the mechanical interactions of (1) soft, compliant and (2) non-compliant implants with the surrounding brain tissue in a rodent brain. Understanding such interactions will enable the engineering of novel materials that will improve stability and reliability of brain implants. APPROACH: Acute force measurements were made using a load cell in n = 3 live rats, each with 4 craniotomies. Using an indentation method, brain tissue was tested for changes in force using established protocols. A total of 4 non-compliant, bare silicon microshanks, 3 non-compliant polyvinyl acetate (PVAc)-coated silicon microshanks, and 6 compliant, nanocomposite microshanks were tested. Stress values were calculated by dividing the force by surface area and strain was estimated using a linear stress-strain relationship. Micromotion effects from breathing and vascular pulsatility on tissue stress were estimated from a 5 s interval of steady-state measurements. Viscoelastic properties were estimated using a second-order Prony series expansion of stress-displacement curves for each shank. MAIN RESULTS: The distribution of strain values imposed on brain tissue for both compliant nanocomposite microshanks and PVAc-coated, non-compliant silicon microshanks were significantly lower compared to non-compliant bare silicon shanks. Interestingly, step-indentation experiments also showed that compliant, nanocomposite materials significantly decreased stress relaxation rates in the brain tissue at the interface (p < 0.05) compared to non-compliant silicon and PVAc-coated silicon materials. Furthermore, both PVAc-coated non-compliant silicon and compliant nanocomposite shanks showed significantly reduced (by 4-5 fold) stresses due to tissue micromotion at the interface. SIGNIFICANCE: The results of this study showed that soft, adaptive materials reduce strains and strain rates and micromotion induced stresses in the surrounding brain tissue. Understanding the material behavior at the site of tissue contact will help to improve neural implant design.


Assuntos
Encéfalo/fisiologia , Encéfalo/cirurgia , Eletrodos Implantados , Microeletrodos , Implantação de Prótese/métodos , Animais , Módulo de Elasticidade/fisiologia , Análise de Falha de Equipamento , Fricção , Dureza/fisiologia , Desenho de Prótese , Ratos , Ratos Sprague-Dawley , Resistência ao Cisalhamento , Estresse Mecânico , Viscosidade
10.
J Neural Eng ; 10(6): 066001, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24099854

RESUMO

OBJECTIVE: Brain tissue undergoes dramatic molecular and cellular remodeling at the implant-tissue interface that evolves over a period of weeks after implantation. The biomechanical impact of such remodeling on the interface remains unknown. In this study, we aim to assess the changes in the mechanical properties of the brain-electrode interface after chronic implantation of a microelectrode. APPROACH: Microelectrodes were implanted in the rodent cortex at a depth of 1 mm for different durations-1 day (n = 4), 10-14 days (n = 4), 4 weeks (n = 4) and 6-8 weeks (n = 7). After the initial duration of implantation, the microelectrodes were moved an additional 1 mm downward at a constant speed of 10 µm s(-1). Forces experienced by the microelectrode were measured during movement and after termination of movement. The biomechanical properties of the interfacial brain tissue were assessed from measured force-displacement curves using two separate models-a two-parameter Mooney-Rivlin hyperelastic model and a viscoelastic model with a second-order Prony series. MAIN RESULTS: Estimated shear moduli using a second-order viscoelastic model increased from 0.5-2.6 kPa (day 1 of implantation) to 25.7-59.3 kPa (after 4 weeks of implantation) and subsequently decreased to 0.8-7.9 kPa after 6-8 weeks of implantation in 6 of the 7 animals. The estimated elastic modulus increased from 4.1-7.8 kPa on the day of implantation to 24-44.9 kPa after 4 weeks. The elastic modulus was estimated to be 6.8-33.3 kPa in 6 of the 7 animals after 6-8 weeks of implantation. The above estimates suggest that the brain tissue surrounding the microelectrode evolves from a stiff matrix with maximal shear and elastic modulus after 4 weeks of implantation into a composite of two different layers with different mechanical properties-a stiff compact inner layer surrounded by softer brain tissue that is biomechanically similar to brain tissue-during the first week of implantation. Tissue micromotion-induced stresses on the microelectrode constituted 12-55% of the steady-state stresses on the microelectrode on the day of implantation (n = 4), 2-21% of the steady-state stresses after 4 weeks of implantation (n = 4), and 4-10% of the steady-state stresses after 6-8 weeks of implantation (n = 7). SIGNIFICANCE: Understanding biomechanical behavior at the brain-microelectrode interface is necessary for the long-term success of implantable neuroprosthetics and microelectrode arrays. Such quantitative physical characterization of the dynamic changes in the electrode-tissue interface will (a) drive the design and development of more mechanically optimal, chronic brain implants, and (b) lead to new insights into key cellular and molecular events such as neuronal adhesion, migration and function in the immediate vicinity of the brain implant.


Assuntos
Córtex Cerebral/fisiologia , Eletrodos Implantados/tendências , Estresse Mecânico , Animais , Microeletrodos/tendências , Próteses e Implantes/tendências , Ratos , Ratos Sprague-Dawley , Propriedades de Superfície , Fatores de Tempo , Viscosidade
11.
Mol Ther Nucleic Acids ; 2: e82, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23531602

RESUMO

We present here a high efficiency, high viability siRNA-delivery method using a voltage-controlled chemical transfection strategy to achieve modulated delivery of polyethylenimine (PEI) complexed with siRNA in an in vitro culture of neuro2A cells and neurons. Low voltage pulses were applied to adherent cells before the administration of PEI-siRNA complexes. Live assays of neuro2a cells transfected with fluorescently tagged siRNA showed an increase in transfection efficiency from 62 ± 14% to 98 ± 3.8% (after -1 V). In primary hippocampal neurons, transfection efficiencies were increased from 30 ± 18% to 76 ± 18% (after -1 V). Negligible or low-level transfection was observed after preconditioning at higher voltages, suggesting an inverse relationship with applied voltage. Experiments with propidium iodide ruled out the role of electroporation in the transfection of siRNAs suggesting an alternate electro-endocytotic mechanism. In addition, image analysis of preconditioned and transfected cells demonstrates siRNA uptake and loading that is tuned to preconditioning voltage levels. There is approximately a fourfold increase in siRNA loading after preconditioning at -1 V compared with the same at ±2-3 V. Modulated gene expression is demonstrated in a functional knockdown of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in neuro2A cells using siRNA. Cell density and dendritic morphological changes are also demonstrated in modulated knockdown of brain derived neurotrophic factor (BDNF) in primary hippocampal neurons. The method reported here has potential applications in the development of high-throughput screening systems for large libraries of siRNA molecules involving difficult-to-transfect cells like neurons.Molecular Therapy-Nucleic Acids (2013) 2, e82; doi:10.1038/mtna.2013.10; published online 26 March 2013.

12.
J Vis Exp ; (67): e4415, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-23007885

RESUMO

The discovery of RNAi pathway in eukaryotes and the subsequent development of RNAi agents, such as siRNA and shRNA, have achieved a potent method for silencing specific genes for functional genomics and therapeutics. A major challenge involved in RNAi based studies is the delivery of RNAi agents to targeted cells. Traditional non-viral delivery techniques, such as bulk electroporation and chemical transfection methods often lack the necessary spatial control over delivery and afford poor transfection efficiencies. Recent advances in chemical transfection methods such as cationic lipids, cationic polymers and nanoparticles have resulted in highly enhanced transfection efficiencies. However, these techniques still fail to offer precise spatial control over delivery that can immensely benefit miniaturized high-throughput technologies, single cell studies and investigation of cell-cell interactions. Recent technological advances in gene delivery have enabled high-throughput transfection of adherent cells, a majority of which use microscale electroporation. Microscale electroporation offers precise spatio-temporal control over delivery (up to single cells) and has been shown to achieve high efficiencies. Additionally, electroporation based approaches do not require a prolonged period of incubation (typically 4 hours) with siRNA and DNA complexes as necessary in chemical based transfection methods and lead to direct entry of naked siRNA and DNA molecules into the cell cytoplasm. As a consequence gene expression can be achieved as early as six hours after transfection. Our lab has previously demonstrated the use of microelectrode arrays (MEA) for site-specific transfection in adherent mammalian cell cultures. In the MEA based approach, delivery of genetic payload is achieved via localized micro-scale electroporation of cells. An application of electric pulse to selected electrodes generates local electric field that leads to electroporation of cells present in the region of the stimulated electrodes. The independent control of the micro-electrodes provides spatial and temporal control over transfection and also enables multiple transfection based experiments to be performed on the same culture increasing the experimental throughput and reducing culture-to-culture variability. Here we describe the experimental setup and the protocol for targeted transfection of adherent HeLa cells with a fluorescently tagged scrambled sequence siRNA using electroporation. The same protocol can also be used for transfection of plasmid vectors. Additionally, the protocol described here can be easily extended to a variety of mammalian cell lines with minor modifications. Commercial availability of MEAs with both pre-defined and custom electrode patterns make this technique accessible to most research labs with basic cell culture equipment.


Assuntos
RNA Interferente Pequeno/administração & dosagem , Transfecção/métodos , Adesão Celular , Eletroporação/métodos , Células HeLa , Humanos , Microeletrodos , RNA Interferente Pequeno/genética
13.
Lab Chip ; 12(16): 2930-41, 2012 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-22722746

RESUMO

Recent evidence suggests that integrin-mediated adhesion of neurons has immediate functional implications for learning and memory. In addition, adhesion of neurons to artificial substrates often determines the effectiveness and life of implants in the brain and peripheral nervous system. In this study, we present a novel biochip capable of simultaneous, quantitative, real-time monitoring of integrin-mediated adhesion and electrophysiology of primary neurons in vitro. The proposed technology combines acoustic micro-resonators capable of tracking changes in mechanics of the adhering neuronal layer, and microelectrode arrays for recording extracellular unit activity. Our results showed in four different experimental paradigms that the acoustic sensor response to adhering cells is correlated to integrin-mediated adhesion and that the micro-sensor is capable of monitoring the dynamics of neuronal adhesion over a period of 9 days. Finally, using our unique dual measurement platform, we performed simultaneous, real-time measurement of integrin-mediated adhesion and single cell electrophysiology in a neuronal culture. The sensitivities of the micro-resonators were 4-5 orders of magnitude greater than the sensitivity of the macro-scale resonators in response to adhering neurons. This multi-functional sensor platform offers insight into the interplay between integrin-mediated adhesion and neural function on a temporal resolution beyond any currently available experimental method and can therefore potentially lead to novel discoveries on the interactions between neuronal adhesion and function.


Assuntos
Técnicas Eletroquímicas/métodos , Neurônios/fisiologia , Células 3T3 , Animais , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Eletricidade , Técnicas Eletroquímicas/instrumentação , Fibronectinas/farmacologia , Integrinas/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Microeletrodos , Neurônios/citologia , Oligopeptídeos/farmacologia
14.
J Microelectromech Syst ; 21(4): 882-896, 2012 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24431925

RESUMO

We report here a successful demonstration of a flip-chip packaging approach for a microelectromechanical systems (MEMS) device with in-plane movable microelectrodes implanted in a rodent brain. The flip-chip processes were carried out using a custom-made apparatus that was capable of the following: 1) creating Ag epoxy microbumps for first-level interconnect; 2) aligning the die and the glass substrate; and 3) creating non-hermetic encapsulation (NHE). The completed flip-chip package had an assembled weight of only 0.5 g significantly less than the previously designed wire-bonded package of 4.5 g. The resistance of the Ag bumps was found to be negligible. The MEMS micro-electrodes were successfully tested for its mechanical movement with microactuators generating forces of 450 µN with a displacement resolution of 8.8 µm/step. An NHE on the front edge of the package was created by patterns of hydrophobic silicone microstructures to prevent contamination from cerebrospinal fluid while simultaneously allowing the microelectrodes to move in and out of the package boundary. The breakdown pressure of the NHE was found to be 80 cm of water, which is significantly (4.5-11 times) larger than normal human intracranial pressures. Bench top tests and in vivo tests of the MEMS flip-chip packages for up to 75 days showed reliable NHE for potential long-term implantation.

15.
J Microelectromech Syst ; 21(5): 1172-1186, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-24431926

RESUMO

This paper presents a new actuation scheme for in-plane bidirectional translation of polysilicon microelectrodes. The new Chevron-peg actuation scheme uses microelectromechanical systems (MEMS) based electrothermal microactuators to move microelectrodes for brain implant applications. The design changes were motivated by specific needs identified by the in vivo testing of an earlier generation of MEMS microelectrodes that were actuated by the Chevron-latch type of mechanism. The microelectrodes actuated by the Chevron-peg mechanism discussed here show improved performance in the following key areas: higher force generation capability (111 µN per heat strip compared to 50 µN), reduced power consumption (91 mW compared to 360 mW), and reliable performance with consistent forward and backward movements of microelectrodes. Failure analysis of the Chevron-latch and the Chevron-peg type of actuation schemes showed that the latter is more robust to wear over four million cycles of operation. The parameters for the activation waveforms for Chevron-peg actuators were optimized using statistical analysis. Waveforms with a 1-ms time period and a 1-Hz frequency of operation showed minimal error between the expected and the actual movement of the microelectrodes. The new generation of Chevron-peg actuators and microelectrodes are therefore expected to enhance the longevity and performance of implanted microelectrodes in the brain. [2011-0341].

16.
Front Neurosci ; 5: 94, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21927593

RESUMO

Implantable microelectrodes that are currently used to monitor neuronal activity in the brain in vivo have serious limitations both in acute and chronic experiments. Movable microelectrodes that adapt their position in the brain to maximize the quality of neuronal recording have been suggested and tried as a potential solution to overcome the challenges with the current fixed implantable microelectrodes. While the results so far suggest that movable microelectrodes improve the quality and stability of neuronal recordings from the brain in vivo, the bulky nature of the technologies involved in making these movable microelectrodes limits the throughput (number of neurons that can be recorded from at any given time) of these implantable devices. Emerging technologies involving the use of microscale motors and electrodes promise to overcome this limitation. This review summarizes some of the most recent efforts in developing movable neural interfaces using microscale technologies that adapt their position in response to changes in the quality of the neuronal recordings. Key gaps in our understanding of the brain-electrode interface are highlighted. Emerging discoveries in these areas will lead to success in the development of a reliable and stable interface with single neurons that will impact basic neurophysiological studies and emerging cortical prosthetic technologies.

17.
J Microelectromech Syst ; 21(1): 132-144, 2011 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-24504168

RESUMO

Flip-chip packaging is desirable for microelectro-mechanical systems (MEMS) devices because it reduces the overall package size and allows scaling up the number of MEMS chips through 3-D stacks. In this report, we demonstrate three novel techniques to create first-level interconnect (FLI) on MEMS: 1) Dip and attach technology for Ag epoxy; 2) Dispense technology for solder paste; 3) Dispense, pull, and attach technology (DPAT) for solder paste. The above techniques required no additional microfabrication steps, produced no visible surface contamination on the MEMS active structures, and generated high-aspect-ratio interconnects. The developed FLIs were successfully tested on MEMS moveable microelectrodes microfabricated by SUMMiTVTM process producing no apparent detrimental effect due to outgassing. The bumping processes were successfully applied on Al-deposited bond pads of 100 µm × 100 µm with an average bump height of 101.3 µm for Ag and 184.8 µm for solder (63Sn, 37Pb). DPAT for solder paste produced bumps with the aspect ratio of 1.8 or more. The average shear strengths of Ag and solder bumps were 78 MPa and 689 kPa, respectively. The electrical test on Ag bumps at 794 A/cm2 demonstrated reliable electrical interconnects with negligible resistance. These scalable FLI technologies are potentially useful for MEMS flip-chip packaging and 3-D stacking.

18.
Front Neuroeng ; 3: 10, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20617188

RESUMO

One of the critical requirements of the emerging class of neural prosthetic devices is to maintain good quality neural recordings over long time periods. We report here a novel MEMS (Micro Electro Mechanical Systems) based technology that can move microelectrodes in the event of deterioration in neural signal to sample a new set of neurons. Microscale electro-thermal actuators are used to controllably move microelectrodes post-implantation in steps of approximately 9 mum. In this study, a total of 12 movable microelectrode chips were individually implanted in adult rats. Two of the twelve movable microelectrode chips were not moved over a period of 3 weeks and were treated as control experiments. During the first 3 weeks of implantation, moving the microelectrodes led to an improvement in the average signal to noise ratio (SNR) from 14.61 +/- 5.21 dB before movement to 18.13 +/- 4.99 dB after movement across all microelectrodes and all days. However, the average root-mean-square values of noise amplitudes were similar at 2.98 +/- 1.22 muV and 3.01 +/- 1.16 muV before and after microelectrode movement. Beyond 3 weeks, the primary observed failure mode was biological rejection of the PMMA (dental cement) based skull mount resulting in the device loosening and eventually falling from the skull. Additionally, the average SNR for functioning devices beyond 3 weeks was 11.88 +/- 2.02 dB before microelectrode movement and was significantly different (p < 0.01) from the average SNR of 13.34 +/- 0.919 dB after movement. The results of this study demonstrate that MEMS based technologies can move microelectrodes in rodent brains in long-term experiments resulting in improvements in signal quality. Further improvements in packaging and surgical techniques will potentially enable movable microelectrodes to record cortical neuronal activity in chronic experiments.

19.
IEEE Trans Neural Syst Rehabil Eng ; 18(5): 489-97, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20667815

RESUMO

The aims of this study are to 1) experimentally validate for the first time the nonlinear current-potential characteristics of bulk doped polycrystalline silicon in the small amplitude voltage regimes (0-200 µV) and 2) test if noise amplitudes ( 0-15 µV ) from single neuronal electrical recordings get selectively attenuated in doped polycrystalline silicon microelectrodes due to the above property. In highly doped polycrystalline silicon, bulk resistances of several hundred kilo-ohms were experimentally measured for voltages typical of noise amplitudes and 9-10 kΩ for voltages typical of neural signal amplitudes ( > 150-200 µV). Acute multiunit measurements and noise measurements were made in n=6 and n=8 anesthetized adult rats, respectively, using polycrystalline silicon and tungsten microelectrodes. There was no significant difference in the peak-to-peak amplitudes of action potentials recorded from either microelectrode (p > 0.10). However, noise power in the recordings from tungsten microelectrodes (26.36 ±10.13 pW) was significantly higher than the corresponding value in polycrystalline silicon microelectrodes (7.49 ±2.66 pW). We conclude that polycrystalline silicon microelectrodes result in selective attenuation of noise power in electrical recordings compared to tungsten microelectrodes. This reduction in noise compared to tungsten microelectrodes is likely due to the exponentially higher bulk resistances offered by highly doped bulk polycrystalline silicon in the range of voltages corresponding to noise in multiunit measurements.


Assuntos
Potenciais de Ação/fisiologia , Artefatos , Encéfalo/fisiologia , Eletrodos Implantados , Microeletrodos , Neurônios/fisiologia , Silício/química , Animais , Cristalização/métodos , Condutividade Elétrica , Desenho de Equipamento , Análise de Falha de Equipamento , Ratos , Ratos Wistar , Sensibilidade e Especificidade
20.
Artigo em Inglês | MEDLINE | ID: mdl-19964241

RESUMO

In this study, we test the hypothesis that increased surface roughness due to carbon nanotubes (CNTs) enhances neuronal adhesion and consequently electrical excitability of single neurons. Neurons are grown on CNT modified microelectrode arrays (MEAs). Multi-unit activity was seen as early as 4 days after seeding compared to 7 days in control cultures on microelectrodes without CNTs. The results overall, show earlier onset and higher level of electrical activity in neurons seeded on CNT modified MEAs compared to non-modified control MEAs. We conclude that CNTs on microelectrodes enhance electrical excitability of single neurons in culture.


Assuntos
Potenciais de Ação/fisiologia , Técnicas de Cultura de Células/métodos , Hipocampo/fisiologia , Microeletrodos , Nanotubos de Carbono/química , Neurônios/fisiologia , Animais , Células Cultivadas , Materiais Revestidos Biocompatíveis , Hipocampo/citologia , Hipocampo/embriologia , Nanotubos de Carbono/ultraestrutura , Neurônios/citologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA