Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Bioconjug Chem ; 33(9): 1685-1697, 2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36017941

RESUMEN

Traumatic brain injury (TBI) is a major cause of disability and death among children and young adults in the United States, yet there are currently no treatments that improve the long-term brain health of patients. One promising therapeutic for TBI is brain-derived neurotrophic factor (BDNF), a protein that promotes neurogenesis and neuron survival. However, outstanding challenges to the systemic delivery of BDNF are its instability in blood, poor transport into the brain, and short half-life in circulation and brain tissue. Here, BDNF is encapsulated into an engineered, biodegradable porous silicon nanoparticle (pSiNP) in order to deliver bioactive BDNF to injured brain tissue after TBI. The pSiNP carrier is modified with the targeting ligand CAQK, a peptide that binds to extracellular matrix components upregulated after TBI. The protein cargo retains bioactivity after release from the pSiNP carrier, and systemic administration of the CAQK-modified pSiNPs results in effective delivery of the protein cargo to injured brain regions in a mouse model of TBI. When administered after injury, the CAQK-targeted pSiNP delivery system for BDNF reduces lesion volumes compared to free BDNF, supporting the hypothesis that pSiNPs mediate therapeutic protein delivery after systemic administration to improve outcomes in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Nanopartículas , Animales , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Matriz Extracelular , Ligandos , Ratones , Péptidos/uso terapéutico , Porosidad , Silicio
2.
Adv Funct Mater ; 30(25)2020 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-32982626

RESUMEN

Scaffolds made from biocompatible polymers provide physical cues to direct the extension of neurites and to encourage repair of damaged nerves. The inclusion of neurotrophic payloads in these scaffolds can substantially enhance regrowth and repair processes. However, many promising neurotrophic candidates are excluded from this approach due to incompatibilities with the polymer or with the polymer processing conditions. This work provides one solution to this problem by incorporating porous silicon nanoparticles (pSiNPs) that are pre-loaded with the therapeutic into a polymer scaffold during fabrication. The nanoparticle-drug-polymer hybrids are prepared in the form of oriented poly(lactic-co-glycolic acid) nanofiber scaffolds. We test three different therapeutic payloads: bpV(HOpic), a small molecule inhibitor of phosphatase and tensin homolog (PTEN); an RNA aptamer specific to tropomyosin-related kinase receptor type B (TrkB); and the protein nerve growth factor (NGF). Each therapeutic is loaded using a loading chemistry that is optimized to slow the rate of release of these water-soluble payloads. The drug-loaded pSiNP-nanofiber hybrids release approximately half of their TrkB aptamer, bpV(HOpic), or NGF payload in 2, 10, and >40 days, respectively. The nanofiber hybrids increase neurite extension relative to drug-free control nanofibers in a dorsal root ganglion explant assay.

3.
Cells Tissues Organs ; 205(5-6): 372-395, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30517922

RESUMEN

Over several decades, biomaterial scientists have developed materials to spur axonal regeneration and limit secondary injury and tested these materials within preclinical animal models. Rarely, though, are astrocytes examined comprehensively when biomaterials are placed into the injury site. Astrocytes support neuronal function in the central nervous system. Following an injury, astrocytes undergo reactive gliosis and create a glial scar. The astrocytic glial scar forms a dense barrier which restricts the extension of regenerating axons through the injury site. However, there are several beneficial effects of the glial scar, including helping to reform the blood-brain barrier, limiting the extent of secondary injury, and supporting the health of regenerating axons near the injury site. This review provides a brief introduction to the role of astrocytes in the spinal cord, discusses astrocyte phenotypic changes that occur following injury, and highlights studies that explored astrocyte changes in response to biomaterials tested within in vitro or in vivo environments. Overall, we suggest that in order to improve biomaterial designs for spinal cord injury applications, investigators should more thoroughly consider the astrocyte response to such designs.


Asunto(s)
Astrocitos/patología , Materiales Biocompatibles/uso terapéutico , Regeneración Nerviosa , Traumatismos de la Médula Espinal/terapia , Animales , Astrocitos/citología , Astrocitos/metabolismo , Materiales Biocompatibles/química , Proteína Ácida Fibrilar de la Glía/análisis , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Neurotransmisores/análisis , Neurotransmisores/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
4.
J Am Chem Soc ; 138(46): 15106-15109, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27933884

RESUMEN

Heterocyclic silanes containing Si-N or Si-S bonds in the ring undergo a ring opening reaction with -OH groups at the surface of porous Si nanostructures to generate -SH or -NH functional surfaces, grafted via O-Si bonds. The reaction is substantially faster (0.5-2 h at 25 °C) and more efficient than hydrolytic condensation of trialkoxysilanes on similar hydroxy-terminated surfaces, and the reaction retains the open pore structure and photoluminescence of the quantum-confined silicon nanostructures. The chemistry is sufficiently mild to allow trapping of the test protein lysozyme, which retains its enzymatic activity upon release from the modified porous nanostructure.

5.
Cells Tissues Organs ; 202(1-2): 102-115, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27701150

RESUMEN

Nanoparticles are increasingly being studied within experimental models of spinal cord injury (SCI). They are used to image cells and tissue, move cells to specific regions of the spinal cord, and deliver therapeutic agents locally. The focus of this article is to provide a brief overview of the different types of nanoparticles being studied for spinal cord applications and present data showing the capability of nanoparticles to deliver the chondroitinase ABC (chABC) enzyme locally following acute SCI in rats. Nanoparticles releasing chABC helped promote axonal regeneration following injury, and the nanoparticles also protected the enzyme from rapid degradation. In summary, nanoparticles are viable materials for diagnostic or therapeutic applications within experimental models of SCI and have potential for future clinical use.


Asunto(s)
Nanopartículas/química , Nanotecnología/métodos , Médula Espinal/patología , Animales , Condroitina ABC Liasa/metabolismo , Modelos Animales de Enfermedad , Humanos , Traumatismos de la Médula Espinal/terapia
6.
Front Cell Neurosci ; 17: 1266019, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37941607

RESUMEN

Central nervous system (CNS) glia, including astrocytes, microglia, and oligodendrocytes, play prominent roles in traumatic injury and degenerative disorders. Due to their importance, active pharmaceutical ingredients (APIs) are being developed to modulate CNS glia in order to improve outcomes in traumatic injury and disease. While many of these APIs show promise in vitro, the majority of APIs that are systemically delivered show little penetration through the blood-brain barrier (BBB) or blood-spinal cord barrier (BSCB) and into the CNS, rendering them ineffective. Novel nanomaterials are being developed to deliver APIs into the CNS to modulate glial responses and improve outcomes in injury and disease. Nanomaterials are attractive options as therapies for central nervous system protection and repair in degenerative disorders and traumatic injury due to their intrinsic capabilities in API delivery. Nanomaterials can improve API accumulation in the CNS by increasing permeation through the BBB of systemically delivered APIs, extending the timeline of API release, and interacting biophysically with CNS cell populations due to their mechanical properties and nanoscale architectures. In this review, we present the recent advances in the fields of both locally implanted nanomaterials and systemically administered nanoparticles developed for the delivery of APIs to the CNS that modulate glial activity as a strategy to improve outcomes in traumatic injury and disease. We identify current research gaps and discuss potential developments in the field that will continue to translate the use of glia-targeting nanomaterials to the clinic.

7.
ACS Biomater Sci Eng ; 8(10): 4140-4152, 2022 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-36210772

RESUMEN

Porous silicon (pSi) nanoparticles are loaded with Immunoglobulin A-2 (IgA2) antibodies, and the assembly is coated with pH-responsive polymers on the basis of the Eudragit family of enteric polymers (L100, S100, and L30-D55). The temporal release of the protein from the nanocomposite formulations is quantified following an in vitro protocol simulating oral delivery: incubation in simulated gastric fluid (SGF; at pH 1.2) for 2 h, followed by a fasting state simulated intestinal fluid (FasSIF; at pH 6.8) or phosphate buffer solution (PBS; at pH 7.4). The nanocomposite formulations display a negligible release in SGF, while more than 50% of the loaded IgA2 is released in solutions at a pH of 6.8 (FasSIF) or 7.4 (PBS). Between 21 and 44% of the released IgA2 retains its functional activity. A capsule-based system is also evaluated, where the IgA2-loaded particles are packed into a gelatin capsule and the capsule is coated with either EudragitL100 or EudragitS100 polymer for a targeted release in the small intestine or the colon, respectively. The capsule-based formulations outperform polymer-coated nanoparticles in vitro, preserving 45-54% of the activity of the released protein.


Asunto(s)
Nanopartículas , Polímeros , Gelatina , Concentración de Iones de Hidrógeno , Inmunoglobulina A , Intestino Delgado , Fosfatos , Porosidad , Silicio , Solubilidad
8.
ACS Biomater Sci Eng ; 8(10): 4123-4131, 2022 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-34468123

RESUMEN

Peptide nucleic acids (PNAs) are a class of artificial oligonucleotide mimics that have garnered much attention as precision biotherapeutics for their efficient hybridization properties and their exceptional biological and chemical stability. However, the poor cellular uptake of PNA is a limiting factor to its more extensive use in biomedicine; encapsulation in nanoparticle carriers has therefore emerged as a strategy for internalization and delivery of PNA in cells. In this study, we demonstrate that PNA can be readily loaded into porous silicon nanoparticles (pSiNPs) following a simple salt-based trapping procedure thus far employed only for negatively charged synthetic oligonucleotides. We show that the ease and versatility of PNA chemistry also allows for producing PNAs with different net charge, from positive to negative, and that the use of differently charged PNAs enables optimization of loading into pSiNPs. Differently charged PNA payloads determine different release kinetics and allow modulation of the temporal profile of the delivery process. In vitro silencing of a set of specific microRNAs using a pSiNP-PNA delivery platform demonstrates the potential for biomedical applications.


Asunto(s)
MicroARNs , Nanopartículas , Ácidos Nucleicos de Péptidos , MicroARNs/genética , Nanopartículas/química , Oligonucleótidos , Ácidos Nucleicos de Péptidos/química , Porosidad , Silicio/química
9.
Nanoscale ; 14(47): 17700-17713, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36416809

RESUMEN

Evaluation of Gastrointestinal Stromal Tumors (GIST) during initial clinical staging, surgical intervention, and postoperative management can be challenging. Current imaging modalities (e.g., PET and CT scans) lack sensitivity and specificity. Therefore, advanced clinical imaging modalities that can provide clinically relevant images with high resolution would improve diagnosis. KIT is a tyrosine kinase receptor overexpressed on GIST. Here, the application of a specific DNA aptamer targeting KIT, decorated onto a fluorescently labeled porous silicon nanoparticle (pSiNP), is used for the in vitro & in vivo imaging of GIST. This nanoparticle platform provides high-fidelity GIST imaging with minimal cellular toxicity. An in vitro analysis shows greater than 15-fold specific KIT protein targeting compared to the free KIT aptamer, while in vivo analyses of GIST-burdened mice that had been injected intravenously (IV) with aptamer-conjugated pSiNPs show extensive nanoparticle-to-tumor signal co-localization (>90% co-localization) compared to control particles. This provides an effective platform for which aptamer-conjugated pSiNP constructs can be used for the imaging of KIT-expressing cancers or for the targeted delivery of therapeutics.


Asunto(s)
Aptámeros de Nucleótidos , Tumores del Estroma Gastrointestinal , Animales , Ratones , Silicio , Tumores del Estroma Gastrointestinal/diagnóstico por imagen
10.
Crit Rev Biomed Eng ; 39(2): 125-80, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21488818

RESUMEN

With increasing regularity, biomaterials are being designed with the goal of promoting repair of the injured spinal cord. Most often, the efficacy of novel biomaterials is tested using in vitro models; however, their true potential will be realized only after they are applied and evaluated in standardized in vivo spinal cord injury (SCI) models. The purpose of this review is to (1) provide a primer on SCI research including an overview of common pathogenic mechanisms that may respond to biomaterials and the in vivo models and outcomes assessment tools used to evaluate therapeutic efficacy; (2) review the types of biomaterials that have been tested in these models; (3) discuss which biomaterials might be applied to these models in the future; and (4) recommend future engineering strategies to create better in vivo models and assessment tools.


Asunto(s)
Materiales Biocompatibles , Prótesis Neurales , Diseño de Prótesis , Traumatismos de la Médula Espinal/terapia , Animales , Femenino , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato , Masculino , Ensayo de Materiales , Ratas
11.
Curr Opin Biomed Eng ; 14: 67-74, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34296048

RESUMEN

Astrocytes are dynamic cells residing in the central nervous system exhibiting many diverse functions. Astrocytes quickly change and present unique phenotypes in response to injury or disease. Here, we briefly summarize recent information regarding astrocyte morphology and function and provide brief insight into their phenotypic changes following injury or disease. We also present the utility of in vitro astrocyte cultures and present recent advances in biomaterial development that enable better recapitulation of their in vivo behavior and morphology.

12.
Nanoscale ; 12(4): 2333-2339, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31930266

RESUMEN

Synthetic DNA-based oligonucleotides are loaded into porous silicon nanoparticles (pSiNPs) and incorporated into nanofibers of poly(lactide-co-glycolide) (PLGA), poly-l-lactic acid (PLA), or polycaprolactone (PCL). The resulting hybrid nanofibers are characterized for their ability to release the functional oligonucleotide payload under physiologic conditions. Under temperature and pH conditions mimicking physiological values, the PLGA-based nanofibers release >80% of their DNA cargo within 5 days, whereas the PLA and PCL-based fibers require 15 days to release >80% of their cargo. The quantity of DNA released scales with the quantity of DNA-loaded pSiNPs embedded in the nanofibers; mass loadings of between 2.4 and 9.1% (based on mass of DNA-pSiNP construct relative to mass of polymer composite) are investigated. When a responsive DNA-based nanodevice (i.e. molecular beacon) is used as a payload, it retains its functionality during the release period, independent of the polymer used for the formation of the nanofibers.


Asunto(s)
ADN/química , Nanopartículas/química , Oligonucleótidos/química , Polímeros/química , Silicio/química , Preparaciones de Acción Retardada/química , Concentración de Iones de Hidrógeno , Ensayo de Materiales , Nanofibras , Poliésteres/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Porosidad , Medicina Regenerativa , Relación Señal-Ruido
13.
ACS Appl Bio Mater ; 2(1): 104-117, 2019 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-31061987

RESUMEN

The topography of electrospun fiber scaffolds modifies astrocytes toward in vivo-like morphologies and behaviors. However, little is known about how electrospun fiber diameter influences astrocyte behavior. In this work, aligned fibers with two distinct nanoscale fiber diameters (808 and 386 nm) were prepared, and the astrocyte response was measured over time. Astrocytes on the large diameter fibers showed significantly increased elongation as early as 2 h after seeding and remained significantly more elongated for up to 4 days compared to those on small diameter fibers. Astrocytes extending along larger diameter fibers were better equipped to support long neurite outgrowth from dorsal root ganglia neurons, and neurite outgrowth along these astrocytes was less branched than outgrowth along astrocytes cultured on small diameter fibers. The differences in astrocyte shape observed on the small or large diameter fibers did not translate into differences in GLT-1, GFAP, or GLAST protein expression. Thus, different fiber diameters were unable to influence astrocyte protein expression uniquely. Nevertheless, astrocytes cultured in either small or large fibers significantly increased their expression of GLT-1 compared to astrocytes cultured on nonfiber (film) controls. Fibrous-induced increases in astrocyte GLT-1 expression protected astrocyte/neuron cocultures from toxicity generated by high extracellular glutamate. Alternatively, astrocytes/neurons cultured on films were less able to protect these cells from culture conditions consisting of high glutamate levels. Biomaterials, such as the fibrous materials presented here, may help stimulate astrocytes to increase GLT-1 expression and uptake more glutamate, since astrocytes are less likely to uptake glutamate in neurodegenerative pathologies or following central nervous system injury.

14.
ACS Appl Mater Interfaces ; 11(27): 23926-23937, 2019 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-31251556

RESUMEN

Silencing of aberrantly expressed microRNAs (miRNAs or miRs) has emerged as one of the strategies for molecular targeted cancer therapeutics. In particular, miR-21 is an oncogenic miRNA overexpressed in many tumors, including ovarian cancer. To achieve efficient administration of anti-miR therapeutics, delivery systems are needed that can ensure local accumulation in the tumor environment, low systemic toxicity, and reduced adverse side effects. In order to develop an improved anti-miR therapeutic agent for the treatment of ovarian cancer, a nanoformulation is engineered that leverages biodegradable porous silicon nanoparticles (pSiNPs) encapsulating an anti-miR-21 locked nucleic acid payload and displaying a tumor-homing peptide for targeted distribution. Targeting efficacy, miR-21 silencing, and anticancer activity are optimized in vitro on a panel of ovarian cancer cell lines, and a formulation of anti-miR-21 in a pSiNP displaying the targeting peptide CGKRK is identified for in vivo evaluation. When this nanoparticulate agent is delivered to mice bearing tumor xenografts, a substantial inhibition of tumor growth is achieved through silencing of miR-21. This study presents the first successful application of tumor-targeted anti-miR porous silicon nanoparticles for the treatment of ovarian cancer in a mouse xenograft model.


Asunto(s)
Portadores de Fármacos , MicroARNs , Nanopartículas , Neoplasias Ováricas , Silicio , Animales , Línea Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Femenino , Humanos , Ratones , Ratones Desnudos , MicroARNs/química , MicroARNs/genética , MicroARNs/farmacología , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Porosidad , Silicio/química , Silicio/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
ACS Appl Mater Interfaces ; 11(1): 356-372, 2019 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-30516370

RESUMEN

Magnetic electrospun fibers are of interest for minimally invasive biomaterial applications that also strive to provide cell guidance. Magnetic electrospun fibers can be injected and then magnetically positioned in situ, and the aligned fiber scaffolds provide consistent topographical guidance to cells. In this study, magnetically responsive aligned poly-l-lactic acid electrospun fiber scaffolds were developed and tested for neural applications. Incorporating oleic acid-coated iron oxide nanoparticles significantly increased neurite outgrowth, reduced the fiber alignment, and increased the surface nanotopography of the electrospun fibers. After verifying neuron viability on two-dimensional scaffolds, the system was tested as an injectable three-dimensional scaffold. Small conduits of aligned magnetic fibers were easily injected in a collagen or fibrinogen hydrogel solution and repositioned using an external magnetic field. The aligned magnetic fibers provided internal directional guidance to neurites within a three-dimensional collagen or fibrin model hydrogel, supplemented with Matrigel. Neurites growing from dorsal root ganglion explants extended 1.4-3× farther on the aligned fibers compared with neurites extending in the hydrogel alone. Overall, these results show that magnetic electrospun fiber scaffolds can be injected and manipulated with a magnetic field in situ to provide directional guidance to neurons inside an injectable hydrogel. Most importantly, this injectable guidance system increased both neurite alignment and neurite length within the hydrogel scaffold.


Asunto(s)
Ganglios Espinales/fisiología , Hidrogeles/química , Regeneración Nerviosa , Neuritas/metabolismo , Andamios del Tejido/química , Animales , Ganglios Espinales/citología , Ratas , Ratas Sprague-Dawley
16.
Adv Mater ; 30(27): e1800512, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29782671

RESUMEN

Photoacoustic (PA) imaging allows visualization of the physiology and pathology of tissues with good spatial resolution and relatively deep tissue penetration. The method converts near-infrared (NIR) laser excitation into thermal expansion, generating pressure transients that are detected with an acoustic transducer. Here, we find that the response of the PA contrast agent indocyanine green (ICG) can be enhanced 17-fold when it is sealed within a rigid nanoparticle. ICG encapsulated in particles composed of porous silicon (pSiNP), porous silica, or calcium silicate all show greater PA contrast relative to equivalent quantities of free ICG, with the pSiNPs showing the strongest enhancement. A liposomal formulation of ICG performs similar to free ICG, suggesting that a rigid host nanostructure is necessary to enhance ICG performance. The improved response of the nanoparticle formulations is attributed to the low thermal conductivity of the porous inorganic hosts and their ability to protect the ICG payload from photolytic and/or thermal degradation. The translational potential of ICG-loaded pSiNPs as photoacoustic probes is demonstrated via imaging of a whole mouse brain.

17.
Adv Mater ; 30(12): e1706785, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29363828

RESUMEN

Oriented composite nanofibers consisting of porous silicon nanoparticles (pSiNPs) embedded in a polycaprolactone or poly(lactide-co-glycolide) matrix are prepared by spray nebulization from chloroform solutions using an airbrush. The nanofibers can be oriented by an appropriate positioning of the airbrush nozzle, and they can direct growth of neurites from rat dorsal root ganglion neurons. When loaded with the model protein lysozyme, the pSiNPs allow the generation of nanofiber scaffolds that carry and deliver the protein under physiologic conditions (phosphate-buffered saline (PBS), at 37 °C) for up to 60 d, retaining 75% of the enzymatic activity over this time period. The mass loading of protein in the pSiNPs is 36%, and in the resulting polymer/pSiNP scaffolds it is 3.6%. The use of pSiNPs that display intrinsic photoluminescence (from the quantum-confined Si nanostructure) allows the polymer/pSiNP composites to be definitively identified and tracked by time-gated photoluminescence imaging. The remarkable ability of the pSiNPs to protect the protein payload from denaturation, both during processing and for the duration of the long-term aqueous release study, establishes a model for the generation of biodegradable nanofiber scaffolds that can load and deliver sensitive biologics.


Asunto(s)
Nanofibras , Animales , Nanopartículas , Polímeros , Porosidad , Ratas , Silicio , Ingeniería de Tejidos , Andamios del Tejido
18.
Adv Mater ; 29(39)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28833739

RESUMEN

A major obstacle in luminescence imaging is the limited penetration of visible light into tissues and interference associated with light scattering and autofluorescence. Near-infrared (NIR) emitters that can also be excited with NIR radiation via two-photon processes can mitigate these factors somewhat because they operate at wavelengths of 650-1000 nm where tissues are more transparent, light scattering is less efficient, and endogenous fluorophores are less likely to absorb. This study presents photolytically stable, NIR photoluminescent, porous silicon nanoparticles with a relatively high two-photon-absorption cross-section and a large emission quantum yield. Their ability to be targeted to tumor tissues in vivo using the iRGD targeting peptide is demonstrated, and the distribution of the nanoparticles with high spatial resolution is visualized.

19.
ACS Chem Neurosci ; 6(11): 1781-8, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26322376

RESUMEN

Nerve growth factor releasing composite nanoparticles (NGF-cNPs) were developed to direct the extension of neurite outgrowth from dorsal root ganglia (DRG). Iron oxide magnetic nanoparticles were incorporated into poly-l-lactic acid (PLLA) nanoparticles in order to position the NGF-cNPs in a culture dish. Neurites growing from DRG extended toward the NGF released from the NGF-cNPs. DRG were then cultured on aligned PLLA microfibers in the presence of NGF-cNPs, and these biomaterials combined to align DRG neurite extension along one axis and preferentially toward the NGF-cNPs. This combinatorial biomaterial approach shows promise as a strategy to direct the extension of regenerating neurites.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanopartículas de Magnetita , Factor de Crecimiento Nervioso/administración & dosificación , Neuritas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Animales , Aumento de la Célula/efectos de los fármacos , Embrión de Pollo , Sistemas de Liberación de Medicamentos/instrumentación , Compuestos Férricos/química , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Inmunohistoquímica , Ácido Láctico/química , Vértebras Lumbares , Nanopartículas de Magnetita/química , Microscopía Electrónica de Rastreo , Factor de Crecimiento Nervioso/farmacocinética , Neuritas/fisiología , Fármacos Neuroprotectores/farmacocinética , Poliésteres , Polímeros/química , Técnicas de Cultivo de Tejidos/métodos
20.
Biol Open ; 4(7): 885-92, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-26002930

RESUMEN

Integrin-mediated cell adhesion to the ECM regulates many physiological processes in part by controlling cell proliferation. It is well established that many normal cells require integrin-mediated adhesion to enter S phase of the cell cycle. Recent evidence indicates that integrins also regulate cytokinesis. Mechanical properties of the ECM can dictate entry into S phase; however, it is not known whether they also can affect the successful completion of cell division. To address this issue, we modulated substrate compliance using fibronectin-coated acrylamide-based hydrogels. Soft and hard substrates were generated with approximate elastic moduli of 1600 and 34,000 Pascals (Pa) respectively. Our results indicate that dermal fibroblasts successfully complete cytokinesis on hard substrates, whereas on soft substrates, a significant number fail and become binucleated. Cytokinesis failure occurs at a step following the formation of the intercellular bridge connecting presumptive daughter cells, suggesting a defect in abscission. Like dermal fibroblasts, mesenchymal stem cells require cell-matrix adhesion for successful cytokinesis. However, in contrast to dermal fibroblasts, they are able to complete cytokinesis on both hard and soft substrates. These results indicate that matrix stiffness regulates the successful completion of cytokinesis, and does so in a cell-type specific manner. To our knowledge, our study is the first to demonstrate that matrix stiffness can affect cytokinesis. Understanding the cell-type specific contribution of matrix compliance to the regulation of cytokinesis will provide new insights important for development, as well as tissue homeostasis and regeneration.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA