Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 5: 9669, 2015 May 18.
Article in English | MEDLINE | ID: mdl-25982506

ABSTRACT

Due to the limited regenerative ability of neural tissue, a diverse set of biochemical and biophysical cues for increasing nerve growth has been investigated, including neurotrophic factors, topography, and electrical stimulation. In this report, we explore optogenetic control of neurite growth as a cell-specific alternative to electrical stimulation. By investigating a broad range of optical stimulation parameters on dorsal root ganglia (DRGs) expressing channelrhodopsin 2 (ChR2), we identified conditions that enhance neurite outgrowth by three-fold as compared to unstimulated or wild-type (WT) controls. Furthermore, optogenetic stimulation of ChR2 expressing DRGs induces directional outgrowth in WT DRGs co-cultured within a 10 mm vicinity of the optically sensitive ganglia. This observed enhancement and polarization of neurite growth was accompanied by an increased expression of neural growth and brain derived neurotrophic factors (NGF, BDNF). This work highlights the potential for implementing optogenetics to drive nerve growth in specific cell populations.


Subject(s)
Light , Nerve Regeneration , Neurogenesis , Animals , Cell Culture Techniques , Channelrhodopsins , Coculture Techniques , Ganglia, Spinal/physiology , Ganglia, Spinal/radiation effects , Gene Expression , Genes, Reporter , Mice , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurites/physiology , Physical Stimulation , Schwann Cells/physiology , Schwann Cells/radiation effects
2.
Biomed Microdevices ; 16(5): 727-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24907052

ABSTRACT

While many advanced liver models support hepatic phenotypes necessary for drug and disease studies, these models are characterized by intricate features such as co-culture with one of more supporting cell types or advanced media perfusion systems. These systems have helped elucidate some of the critical biophysical features missing from standard well-plate based hepatocyte culture, but their advanced designs add to their complexity. Additionally, regardless of the culture system, primary hepatocyte culture systems suffer from reproducibility issues due to phenotypic variation and expensive, limited supplies of donor lots. Here we describe a microfluidic bilayer device that sustains primary human hepatocyte phenotypes, including albumin production, factor IX production, cytochrome P450 3A4 drug metabolism and bile canaliculi formation for at least 14 days in a simple monoculture format with static media. Using a variety of channel architectures, we describe how primary cell phenotype is promoted by spatial confinement within the microfluidic channel, without the need for perfusion or co-culture. By sourcing human hepatocytes expanded in the Fah, Rag2, and Il2rg-knockout (FRG™-KO) humanized mouse model, utilizing a few hundred hepatocytes within each channel, and maintaining hepatocyte function for weeks in vitro within a relatively simple model, we demonstrate a basic primary human hepatocyte culture system that addresses many of the major hurdles in human hepatocyte culture research.


Subject(s)
Cell Culture Techniques , Cell Proliferation , Hepatocytes/metabolism , Liver , Microfluidic Analytical Techniques , Animals , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Hep G2 Cells , Hepatocytes/cytology , Humans , Mice , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
3.
Lab Chip ; 13(4): 542-53, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23108480

ABSTRACT

In this work, we describe the fabrication and working of a modular microsystem that recapitulates the functions of the "Neurovascular Unit". The microdevice comprised a vertical stack of a poly(dimethylsiloxane) (PDMS) neural parenchymal chamber separated by a vascular channel via a microporous polycarbonate (PC) membrane. The neural chamber housed a mixture of neurons (~4%), astrocytes (~95%), and microglia (~1%). The vascular channel was lined with a layer of rat brain microvascular endothelial cell line (RBE4). Cellular components in the neural chamber and vascular channel showed viability (>90%). The neural cells fired inhibitory as well as excitatory potentials following 10 days of culture. The endothelial cells showed diluted-acetylated low density lipoprotein (dil-a-LDL) uptake, expressed von Willebrand factor (vWF) and zonula occludens (ZO-1) tight junctions, and showed decreased Alexafluor™-conjugated dextran leakage across their barriers significantly compared with controls (p < 0.05). When the vascular layer was stimulated with TNF-α for 6 h, about 75% of resident microglia and astrocytes on the neural side were activated significantly (p < 0.05 compared to controls) recapitulating tissue-mimetic responses resembling neuroinflammation. The impact of this microsystem lies in the fact that this biomimetic neurovascular platform might not only be harnessed for obtaining mechanistic insights for neurodegenerative disorders, but could also serve as a potential screening tool for central nervous system (CNS) therapeutics in toxicology and neuroinfectious diseases.


Subject(s)
Brain/blood supply , Coculture Techniques , Endothelial Cells/physiology , Microfluidic Analytical Techniques , Microvessels/physiology , Animals , Brain/cytology , Cell Differentiation , Cell Shape , Cell Survival , Cells, Cultured , Coculture Techniques/instrumentation , Endothelial Cells/cytology , Microfluidic Analytical Techniques/instrumentation , Microvessels/cytology , Neurons/cytology , Rats
4.
Macromol Biosci ; 10(8): 872-80, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20503195

ABSTRACT

Vapor-deposited silicone coatings are attractive candidates for providing insulation in neuroprosthetic devices owing to their excellent resistivity, adhesion, chemical inertness and flexibility. A biocompatibility assessment of these coatings is an essential part of the materials design process, but current techniques are limited to rudimentary cell viability assays or animal muscle implantation tests. This article describes how a recently developed in vitro model of glial scar formation can be utilized to assess the biocompatibility of vapor-deposited silicone coatings on micron-scale wires. A multi-cellular monolayer comprising mixed glial cells was obtained by culturing primary rat midbrain cells on poly(D-lysine)-coated well plates. Stainless steel microwires were coated with two novel insulating thin film silicone polymers, namely poly(trivinyltrimethylcyclotrisiloxane) (polyV(3)D(3)) and poly(trivinyltrimethylcyclotrisiloxane-hexavinyldisiloxane) (polyV(3)D(3)-HVDS) by initiated chemical vapor deposition (iCVD). The monolayer of midbrain cells was disrupted by placing segments of coated microwires into the culture followed by immunocytochemical analysis after 7 d of implantation. Microglial proximity to the microwires was observed to correlate with the amount of fibronectin adsorbed on the coating surface; polyV(3)D(3)-HVDS adsorbed the least amount of fibronectin compared to both stainless steel and polyV(3)D(3). Consequently, the relative number of microglia within 100 µm of the microwires was least on polyV(3)D(3)-HVDS coatings compared to steel and polyV(3)D(3). In addition, the astrocyte reactivity on polyV(3)D(3)-HVDS coatings was lower compared to stainless steel and polyV(3)D(3). The polyV(3)D(3)-HVDS coating was therefore deemed to be most biocompatible, least reactive and most preferable insulating coating for neural prosthetic devices.


Subject(s)
Biocompatible Materials , Microglia/metabolism , Polymers , Silicones , Adsorption , Animals , Female , Fibronectins/metabolism , Fluorescent Antibody Technique , Fluorescent Dyes , Immunohistochemistry , In Vitro Techniques , Pregnancy , Rats , Rats, Inbred F344 , Spectroscopy, Fourier Transform Infrared
5.
Langmuir ; 26(6): 4160-7, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20214394

ABSTRACT

Occlusion or blockage of silicone shunts utilized in the treatment of hydrocephalus is a major challenge that is currently addressed by multiple shunt replacements. Shunt occlusion is caused by the adhesion and proliferation of reactive cells, such as glial and vascular cells, into the lumen of the catheter and on valve components. This in vitro study describes how the adhesive behavior of four human cell types on poly(dimethylsiloxane) (PDMS) surfaces can be suppressed by functionalization with trypsin, a proteolytic enzyme. The covalently conjugated trypsin retained its proteolytic activity and acted in a dose-dependent manner. Trypsin-modified PDMS surfaces supported significantly lower adhesion of normal human astrocytes, human microglia, human dermal fibroblasts, and human umbilical vein endothelial cells compared to unmodified PDMS surfaces (p < 0.0001). Immunofluorescence imaging of cellular fibronectin and quantitative adsorption experiments with serum components indicated that the PDMS surfaces immobilized with trypsin inhibited surface remodeling by all cell types and resisted protein adsorption. The impact of this work lies in the recognition that the well-known proteolytic characteristics of trypsin can be harnessed by covalent surface immobilization to suppress cell adhesion and protein adsorption.


Subject(s)
Cell Adhesion , Dimethylpolysiloxanes , Enzymes, Immobilized , Trypsin , Adsorption , Blood Proteins/chemistry , Cells, Cultured , Fluorescent Antibody Technique , Humans
6.
Front Neuroeng ; 2: 18, 2010.
Article in English | MEDLINE | ID: mdl-20161810

ABSTRACT

Neuroprosthetic devices have made a major impact in the treatment of a variety of disorders such as paralysis and stroke. However, a major impediment in the advancement of this technology is the challenge of maintaining device performance during chronic implantation (months to years) due to complex intrinsic host responses such as gliosis or glial scarring. The objective of this review is to bring together research communities in neurobiology, tissue engineering, and neuroprosthetics to address the major obstacles encountered in the translation of neuroprosthetics technology into long-term clinical use. This article draws connections between specific challenges faced by current neuroprosthetics technology and recent advances in the areas of nerve tissue engineering and neurobiology. Within the context of the device-nervous system interface and central nervous system implants, areas of synergistic opportunity are discussed, including platforms to present cells with multiple cues, controlled delivery of bioactive factors, three-dimensional constructs and in vitro models of gliosis and brain injury, nerve regeneration strategies, and neural stem/progenitor cell biology. Finally, recent insights gained from the fields of developmental neurobiology and cancer biology are discussed as examples of exciting new biological knowledge that may provide fresh inspiration toward novel technologies to address the complexities associated with long-term neuroprosthetic device performance.

7.
Biotechnol Prog ; 25(1): 227-34, 2009.
Article in English | MEDLINE | ID: mdl-19224594

ABSTRACT

Immobilized extracellular matrix proteins and neurotrophins have been extensively studied to enhance neuronal adhesion and proliferation on surfaces for applications in nerve tissue engineering and neuroprosthetic devices. This article describes how the coimmobilization of laminin, an extracellular matrix protein and nerve growth factor (NGF), a neurotrophin can enhance neurite outgrowth observed separately with each type of molecule. In the absence of immobilized NGF, PC12 neurite outgrowth is influenced strongly by the presence of NGF in solution and unaffected by significant increases in laminin surface density (18.7-93.5 ng/mm(2)). However, when both laminin and NGF are immobilized together, the surface density of laminin is an important factor in determining whether or not the neurite outgrowth-promoting effect of NGF can be obtained. PC12 neurite outgrowth on surfaces with coimmobilized laminin and NGF with surface densities of 27.6 ng/mm(2) and 1.4 ng/mm(2), respectively, are similar to that observed on surfaces with immobilized laminin and dissolved NGF.


Subject(s)
Laminin/pharmacology , Nerve Growth Factor/pharmacology , Neurites/drug effects , Animals , Cell Line, Tumor , Drug Synergism , Laminin/chemistry , Nerve Growth Factor/chemistry , Rats
8.
Macromolecules ; 42(6): 1970-1978, 2009.
Article in English | MEDLINE | ID: mdl-21359171

ABSTRACT

Poly (trivinyl-trimethyl-cyclotrisiloxane) or polyV(3)D(3) is a promising insulating thin film known for its potential application in neural probe fabrication. However, its time-consuming synthesis rate renders it impractical for manufacturing standards. Previously, the growth mechanism of polyV(3)D(3) was shown to be affected by significant steric barriers. This article describes the synthesis of a copolymer of polyV(3)D(3) via initiated chemical vapor deposition (iCVD) using V(3)D(3) as the monomer, hexavinyl disiloxane (HVDS) as a spacer, and tert-butyl peroxide (TBP) as the initiator to obtain nearly a 4-fold increase in deposition rate. The film formation kinetics is limited by the adsorption of the reactive species on the surface of the substrate with an activation energy of -41.5 kJ/mol with respect to substrate temperature. The films deposited are insoluble in polar and non polar solvents due to their extremely crosslinked structure. They have excellent adhesion to silicon substrates and their adhesion properties are retained after soaking in a variety of solvents. Spectroscopic evidence shows that the films do not vary in structure after boiling in DI water for 1 hour, illustrating hydrolytic stability. PolyV(3)D(3)-HVDS has a bulk resistivity of 5.6 (±1) × 10(14) Ω-cm, which is comparable to that of parylene-C; the insulating thin film currently used in neuroprosthetic devices.

SELECTION OF CITATIONS
SEARCH DETAIL
...