Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biomed Phys Eng Express ; 10(2)2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38316040

ABSTRACT

The barrier function of the epidermis poses a significant challenge to nanoparticle-mediated topical delivery. A key factor in this barrier function is the thickness of the stratum corneum (SC) layer within the epidermis, which varies across different anatomical sites. The epidermis from the palms and soles, for instance, have thicker SC compared to those from other areas. Previous studies have attempted to bypass the SC layer for nanoparticle penetration by using physical disruption; however, these studies have mostly focused on non-thick skin. In this study, we investigate the role of SC-disrupting mechano-physical strategies (tape-stripping and microneedle abrasion) on thick and thin skin, in allowing transdermal penetration of topically applied nanoparticles using an ex-vivo skin model from rat. Our findings show that tape-stripping reduced the overall thickness of SC in thick skin by 87%, from 67.4 ± 17.3µm to 8.2 ± 8.5µm, whereas it reduced thin skin SC by only 38%, from 9.9 ± 0.6µm to 6.2 ± 3.2µm. Compared to non-thick skin, SC disruption in thick skin resulted in higher nanoparticle diffusion. Tape-stripping effectively reduces SC thickness of thick skin and can be potentially utilized for enhanced penetration of topically applied nanoparticles in skin conditions that affect thick skin.


Subject(s)
Nanoparticles , Skin Diseases , Rats , Animals , Skin Absorption , Epidermis/metabolism , Skin , Polymers
2.
Neurosci Res ; 196: 40-51, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37336292

ABSTRACT

Nerve terminals within the tumor microenvironment as potential pain-mitigating targets for local infiltration analgesia is relatively less explored. In this study, we examine the role of key analgesics administered as local infiltration analgesia in a model of cancer-induced bone pain (CIBP). CIBP was induced by administration of allogenic MRMT1 breast cancer cells in the proximal tibia of rats, and tumor mass characterized using radiogram, micro-CT, and histological analysis. In vitro responsiveness to key analgesics δ-opioid receptor agonist (DOPr), Ca2+ channel and TRPV1 antagonists was assessed using ratiometric Ca2+ imaging in sensory neurons innervating the tumor site. Effectiveness of locally infiltrated analgesics administered independently or in combination was assessed by quantifying evoked limb withdrawal thresholds at two distinct sites for up to 14 days. CIBP animals demonstrated DOPr, N-, and L-type and TRPV1 expression in lumbar dorsal root ganglion neurons (DRG), comparable to controls. Evoked Ca2+ transients in DRG neurons from CIBP animals were significantly reduced in response to treatment with compounds targeting DOPr, N-, L-type Ca2+ channels and TRPV1 proteins. Behaviourally, evoked hyperalgesia at the tumor site was strongly mitigated by peritumoral injection of the DOPr agonist and T-type calcium antagonist, via its activity on bone afferents. Results from this study suggest that nerve terminals at tumor site could be utilized as targets for specific analgesics, using local infiltration analgesia.


Subject(s)
Analgesia , Tumor Microenvironment , Rats , Animals , Rats, Sprague-Dawley , Pain/metabolism , Hyperalgesia/metabolism , Analgesics, Opioid/pharmacology , Sensory Receptor Cells , Analgesics/adverse effects , Analgesics/metabolism , Ganglia, Spinal/metabolism
3.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055003

ABSTRACT

Nerve axonal injury and associated cellular mechanisms leading to peripheral nerve damage are important topics of research necessary for reducing disability and enhancing quality of life. Model systems that mimic the biological changes that occur during human nerve injury are crucial for the identification of cellular responses, screening of novel therapeutic molecules, and design of neural regeneration strategies. In addition to in vivo and mathematical models, in vitro axonal injury models provide a simple, robust, and reductionist platform to partially understand nerve injury pathogenesis and regeneration. In recent years, there have been several advances related to in vitro techniques that focus on the utilization of custom-fabricated cell culture chambers, microfluidic chamber systems, and injury techniques such as laser ablation and axonal stretching. These developments seem to reflect a gradual and natural progression towards understanding molecular and signaling events at an individual axon and neuronal-soma level. In this review, we attempt to categorize and discuss various in vitro models of injury relevant to the peripheral nervous system and highlight their strengths, weaknesses, and opportunities. Such models will help to recreate the post-injury microenvironment and aid in the development of therapeutic strategies that can accelerate nerve repair.


Subject(s)
Guided Tissue Regeneration , In Vitro Techniques , Nerve Regeneration , Peripheral Nerve Injuries/etiology , Peripheral Nerve Injuries/metabolism , Animals , Axons/metabolism , Axons/pathology , Biomarkers , Cell Culture Techniques , Disease Susceptibility , Guided Tissue Regeneration/methods , Humans , In Vitro Techniques/instrumentation , In Vitro Techniques/methods , Peripheral Nerve Injuries/therapy , Peripheral Nervous System/physiology
4.
Toxicol In Vitro ; 74: 105175, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33865945

ABSTRACT

The use of gold nanoparticles (AuNps) in applications connected to the peripheral nervous system (PNS) holds much promise in terms of therapeutic and diagnostic strategies. Despite their extensive use, a clear understanding of their effects on neurons and glia in the PNS is lacking. In this study, we set out to examine the effects of AuNps on dorsal root ganglion (DRG) cells, and how such AuNp-exposed cells could in-turn affect neurite differentiation. DRG cultures were exposed to mono-dispersed spherical-shaped AuNps of diameter 24.3 ± 2.3, 109.2 ± 14.7 or 175 ± 19.2 nm at varying concentrations. Cellular uptake and viability were quantified using flow-cytometry. Neurite differentiation was quantified using neurite tracing analysis in PC-12 and DRG neurons exposed to conditioned media derived from AuNp-treated DRG cells. Both neurons and glia were found to internalize AuNps. DRG cell viability was significantly reduced upon treatment with higher concentration of 175 nm sized AuNps, while 24 nm and 109 nm sized AuNps had no effect. Further, conditioned media from AuNp-treated DRG cells produced comparable neurite outgrowth and neurite branching measurement as controls in PC-12 and DRG neurons. DRG cells were quite resilient to AuNp exposure in mild-moderate concentration. AuNp-exposed DRG cells, irrespective of size and concentration range tested, did not affect neuronal differentiation.


Subject(s)
Ganglia, Spinal/cytology , Gold/administration & dosage , Metal Nanoparticles/administration & dosage , Neurons/drug effects , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Endocytosis , Gold/chemistry , Metal Nanoparticles/chemistry , Neurons/physiology , PC12 Cells , Particle Size , Rats , Rats, Sprague-Dawley
5.
Sci Rep ; 11(1): 2566, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33510229

ABSTRACT

Accessing the peripheral nervous system (PNS) by topically applied nanoparticles is a simple and novel approach with clinical applications in several PNS disorders. Skin is richly innervated by long peripheral axons that arise from cell bodies located distally within ganglia. In this study we attempt to target dorsal root ganglia (DRG) neurons, via their axons by topical application of lectin-functionalized gold nanoparticles (IB4-AuNP). In vitro, 140.2 ± 1.9 nm IB4-AuNP were found to bind both axons and cell bodies of DRG neurons, and AuNP applied at the axonal terminals were found to translocate to the cell bodies. Topical application of IB4-AuNP on rat hind-paw resulted in accumulation of three to fourfold higher AuNP in lumbar DRG than in contralateral control DRGs. Results from this study clearly suggest that topically applied nanoparticles with neurotropic targeting ligands can be utilized for delivering nanoparticles to neuronal cell bodies via axonal transport mechanisms.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Neurons/chemistry , Skin/metabolism , Animals , Electrophysiology , Female , Ganglia, Spinal/metabolism , Microscopy, Electron, Transmission , Nerve Fibers/metabolism , Rats, Sprague-Dawley
6.
RSC Adv ; 11(8): 4623-4630, 2021 Jan 21.
Article in English | MEDLINE | ID: mdl-35424395

ABSTRACT

Microparticle shape, as a tunable design parameter, holds much promise for controlling drug-release kinetics from polymeric microparticulate systems. In this study we hypothesized that the intensity and duration of a local nerve block can be controlled by administration of bupivacaine-loaded stretch-induced anisotropic poly(lactic-co-glycolic acid) microparticles (MPs). MPs of size 27.3 ± 8.5 µm were synthesized by single emulsion method and subjected to controlled stretching force. The aspect ratio of the anisotropic-bupivacaine MPs was quantified, and bupivacaine release was measured in vitro. The anisotropic MPs were administered as local nerve block injections in rats, and the intensity and duration of local anesthesia was measured. Bupivacaine-loaded anisotropic MPs used in this study were ellipsoid in shape and exhibited increased surface pores in comparison to spherical MPs. Anisotropic MPs exhibited a higher rate of bupivacaine release in vitro, and showed significantly (P < 0.05) stronger sensory nerve blocking as compared to spherical bupivacaine MPs, even though the duration of the nerve block remained similar. This study demonstrates the utility of stretch-induced anisotropic MPs in controlling drug release profiles from polymeric MPs, under both in vitro and in vivo conditions. We show that shape, as a tunable design parameter, could play an important role in engineering drug-delivery systems.

7.
Biomed Mater ; 15(6): 065015, 2020 10 03.
Article in English | MEDLINE | ID: mdl-33016262

ABSTRACT

Electrically stimulable nerve conduits are implants that could potentially be utilized in patients with nerve injury for restoring function and limb mobility. Such conduits need to be developed from specialized scaffolds that are both electrically conductive and allow neuronal attachment and differentiation. In this study, we investigate neural cell attachment and axonal differentiation on scaffolds co-woven with poly-(L-lactic acid) (PLLA) yarns and conducting threads. Yarns obtained from electrospun PLLA were co-woven with polypyrrole (PPy)-coated PLLA yarns or ultrathin wires of copper or platinum using a custom built low-resistance semi-automated weaving machine. The conducting threads were first electrically characterized and tested for stability in cell growth media. Suitability of the conducting threads was further assessed via cell viability studies using PC12 cells. Neurite growth was then quantified after electrically stimulating rat dorsal root ganglion (DRG) sensory neurons cultured on the woven scaffolds. Electrical conductivity tests and cellular viability studies demonstrated better bio-tolerability of platinum wires over PPy-coated PLLA yarns and copper wires. Electrically stimulated DRG neurons cultured on platinum-PLLA co-woven scaffolds showed enhanced neurite outgrowth and length. We demonstrate that a woven scaffold design could be utilized to incorporate conducting materials into cell-tolerable polymer yarns for developing electrically stimulable nerve conduits.


Subject(s)
Cell Differentiation , Materials Testing , Neurites/drug effects , Peripheral Nerves/pathology , Tissue Engineering/methods , Animals , Automation , Cell Adhesion , Cell Survival , Electric Conductivity , Electric Stimulation Therapy , Ganglia, Spinal/metabolism , Male , Nanofibers , Neurons/metabolism , PC12 Cells , Polyesters/chemistry , Polymers/chemistry , Pyrroles/chemistry , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Textiles , Tissue Scaffolds
8.
J R Soc Interface ; 17(162): 20190505, 2020 01.
Article in English | MEDLINE | ID: mdl-31910776

ABSTRACT

Neural tissue engineering (NTE) is a rapidly progressing field that promises to address several serious neurological conditions that are currently difficult to treat. Selecting the right scaffolding material to promote neural and non-neural cell differentiation as well as axonal growth is essential for the overall design strategy for NTE. Among the varieties of scaffolds, hydrogels have proved to be excellent candidates for culturing and differentiating cells of neural origin. Considering the intrinsic resistance of the nervous system against regeneration, hydrogels have been abundantly used in applications that involve the release of neurotrophic factors, antagonists of neural growth inhibitors and other neural growth-promoting agents. Recent developments in the field include the utilization of encapsulating hydrogels in neural cell therapy for providing localized trophic support and shielding neural cells from immune activity. In this review, we categorize and discuss the various hydrogel-based strategies that have been examined for neural-specific applications and also highlight their strengths and weaknesses. We also discuss future prospects and challenges ahead for the utilization of hydrogels in NTE.


Subject(s)
Hydrogels , Tissue Engineering , Nervous System , Neurons , Tissue Scaffolds
9.
J Dermatol Sci ; 89(2): 146-154, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29154084

ABSTRACT

BACKGROUND: Transdermal particulate penetration across thick-skin, such as that of palms and sole, is particularly important for drug delivery for disorders such as small fiber neuropathies. Nanoparticle-based drug delivery across skin is believed to have much translational applications, but their penetration especially through thick-skin, is not clear. OBJECTIVE: This study specifically investigates the effectiveness of gold nanoparticles (AuNPs) for thick-skin penetration, especially across the stratum corneum (SC) as a function of particle size. METHODS: The thick-skinned hind-paw of rat was used to characterize depth and distribution of AuNPs of varying sizes, namely, 22±3, 105±11, and 186±20nm. Epidermal penetration of AuNPs was characterized both, in harvested skin from the hind-paw using a diffusion chamber, as well as in vivo. RESULTS: Harvested skin segments exposed to 22nm AuNPs for only 3h demonstrated higher penetration (p<0.05) as compared to the 105 and 186nm particles. In animal studies, hind-paw skin of adult rats exposed to AuNPs solution for the same time, demonstrated nanoparticles in blood on the 4th day, and histological analysis revealed AuNPs in epidermal layers just below the SC, with no apparent tissue response. CONCLUSION: We conclude that the thick-skin allows nanoparticle penetration and acts as a depot for release of AuNPs into circulation long after the initial exposure has ceased.


Subject(s)
Epidermis/metabolism , Gold/pharmacokinetics , Skin Absorption , Administration, Cutaneous , Animals , Gold/administration & dosage , Hindlimb , Male , Metal Nanoparticles/administration & dosage , Models, Animal , Particle Size , Permeability , Rats , Rats, Sprague-Dawley
10.
Proc Natl Acad Sci U S A ; 108(46): 18690-5, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22039220

ABSTRACT

Arrestins are regulatory molecules for G-protein coupled receptor function. In visual rhodopsin, selective binding of arrestin to the cytoplasmic side of light-activated, phosphorylated rhodopsin (P-Rh*) terminates signaling via the G-protein transducin. While the "phosphate-sensor" of arrestin for the recognition of receptor-attached phosphates is identified, the molecular mechanism of arrestin binding and the involvement of receptor conformations in this process are still largely hypothetic. Here we used fluorescence pump-probe and time-resolved fluorescence depolarization measurements to investigate the kinetics of arrestin conformational changes and the corresponding nanosecond dynamical changes at the receptor surface. We show that at least two sequential conformational changes of arrestin occur upon interaction with P-Rh*, thus providing a kinetic proof for the suggested multistep nature of arrestin binding. At the cytoplasmic surface of P-Rh*, the structural dynamics of the amphipathic helix 8 (H8), connecting transmembrane helix 7 and the phosphorylated C-terminal tail, depends on the arrestin interaction state. We find that a high mobility of H8 is required in the low-affinity (prebinding) but not in the high-affinity binding state. High-affinity arrestin binding is inhibited when a bulky, inflexible group is bound to H8, indicating close interaction. We further show that this close steric interaction of H8 with arrestin is mandatory for the transition from prebinding to high-affinity binding; i.e., for arrestin activation. This finding implies a regulatory role for H8 in activation of visual arrestin, which shows high selectivity to P-Rh* in contrast to the broad receptor specificity displayed by the two nonvisual arrestins.


Subject(s)
Arrestin/chemistry , Receptors, G-Protein-Coupled/chemistry , Rhodopsin/chemistry , Animals , Anisotropy , Cattle , Crystallography, X-Ray/methods , Kinetics , Microscopy, Fluorescence/methods , Molecular Conformation , Phosphorylation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Retina/metabolism , Signal Transduction , Spectrophotometry/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...