Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Nanobiomed Res ; 2(6)2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36051821

RESUMEN

Neuroinflammation is one of the hallmarks contributing to Parkinson's Disease (PD) pathology, where microglial activation occurs as one of the earliest events, triggered by extracellular alpha synuclein (aSYN) binding to the CD36 receptor. Here, CD36-binding nanoparticles (NPs) containing synthetic tartaric acid-based amphiphilic polymers (AMs) were rationally designed to inhibit this aSYN-CD36 binding. In silico docking revealed that four AMs with varying alkyl side chain lengths presented differential levels of CD36 binding affinity and that an optimal alkyl chain length would promote the strongest inhibitory activity towards aSYN-CD36 interactions. In vitro competitive binding assays indicated that the inhibitory activity of AM-based NPs plateaued at intermediate side chain lengths of 12- and 18-carbons, supporting the in silico docking predictions. These 12- and 18-carbon length AM NPs also had significantly stronger effects on reducing aSYN internalization and inhibiting the production of the proinflammatory molecules TNF-α and nitric oxide from aSYN-challenged microglia. All four NPs modulated the gene expression of aSYN-challenged microglia, downregulating the expression of the proinflammatory genes TNF, IL-6, and IL-1ß, and upregulating the expression of the anti-inflammatory genes TGF-ß and Arg1. Overall, this work represents a novel polymeric nanotechnology platform that can be used to modulate aSYN-induced microglial activation in PD.

2.
Front Oncol ; 11: 718408, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868914

RESUMEN

BACKGROUND: Late-stage diagnosis of ovarian cancer, a disease that originates in the ovaries and spreads to the peritoneal cavity, lowers 5-year survival rate from 90% to 30%. Early screening tools that can: i) detect with high specificity and sensitivity before conventional tools such as transvaginal ultrasound and CA-125, ii) use non-invasive sampling methods and iii) longitudinally significantly increase survival rates in ovarian cancer are needed. Studies that employ blood-based screening tools using circulating tumor-cells, -DNA, and most recently tumor-derived small extracellular vesicles (sEVs) have shown promise in non-invasive detection of cancer before standard of care. Our findings in this study show the promise of a sEV-derived signature as a non-invasive longitudinal screening tool in ovarian cancer. METHODS: Human serum samples as well as plasma and ascites from a mouse model of ovarian cancer were collected at various disease stages. Small extracellular vesicles (sEVs) were extracted using a commercially available kit. RNA was isolated from lysed sEVs, and quantitative RT-PCR was performed to identify specific metastatic gene expression. CONCLUSION: This paper highlights the potential of sEVs in monitoring ovarian cancer progression and metastatic development. We identified a 7-gene panel in sEVs derived from plasma, serum, and ascites that overlapped with an established metastatic ovarian carcinoma signature. We found the 7-gene panel to be differentially expressed with tumor development and metastatic spread in a mouse model of ovarian cancer. The most notable finding was a significant change in the ascites-derived sEV gene signature that overlapped with that of the plasma-derived sEV signature at varying stages of disease progression. While there were quantifiable changes in genes from the 7-gene panel in serum-derived sEVs from ovarian cancer patients, we were unable to establish a definitive signature due to low sample number. Taken together our findings show that differential expression of metastatic genes derived from circulating sEVs present a minimally invasive screening tool for ovarian cancer detection and longitudinal monitoring of molecular changes associated with progression and metastatic spread.

3.
APL Bioeng ; 4(3): 030902, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32923843

RESUMEN

Advances in nanotechnology have enabled the design of nanotherapeutic platforms that could address the challenges of targeted delivery of active therapeutic agents to the central nervous system (CNS). While the majority of previous research studies on CNS nanotherapeutics have focused on neurons and endothelial cells, the predominant resident immune cells of the CNS, microglia, are also emerging as a promising cellular target for neurodegeneration considering their prominent role in neuroinflammation. Under normal physiological conditions, microglia protect neurons by removing pathological agents. However, long-term exposure of microglia to stimulants will cause sustained activation and lead to neuronal damage due to the release of pro-inflammatory agents, resulting in neuroinflammation and neurodegeneration. This Perspective highlights criteria to be considered when designing microglia-targeting nanotherapeutics for the treatment of neurodegenerative disorders. These criteria include conjugating specific microglial receptor-targeting ligands or peptides to the nanoparticle surface to achieve targeted delivery, leveraging microglial phagocytic properties, and utilizing biocompatible and biodegradable nanomaterials with low immune reactivity and neurotoxicity. In addition, certain therapeutic agents for the controlled inhibition of toxic protein aggregation and for modulation of microglial activation pathways can also be incorporated within the nanoparticle structure without compromising stability. Overall, considering the multifaceted disease mechanisms of neurodegeneration, microglia-targeted nanodrugs and nanotherapeutic particles may have the potential to resolve multiple pathological determinants of the disease and to guide a shift in the microglial phenotype spectrum toward a more neuroprotective state.

4.
Artículo en Inglés | MEDLINE | ID: mdl-32154238

RESUMEN

Parkinson's Disease is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta, the extracellular accumulation of toxic α-synuclein (αSYN) aggregates, and neuroinflammation. Microglia, resident macrophages of the brain, are one of the critical cell types involved in neuroinflammation. Upon sensing extracellular stimuli or experiencing oxidative stress, microglia become activated, which further exacerbates neuroinflammation. In addition, as the first line of defense in the central nervous system, microglia play a critical role in αSYN clearance and degradation. While the role of microglia in neurodegenerative pathologies is widely recognized, few therapeutic approaches have been designed to target both microglial activation and αSYN aggregation. Here, we designed nanoparticles (NPs) to deliver aggregation-inhibiting antioxidants to ameliorate αSYN aggregation and attenuate activation of a pro-inflammatory microglial phenotype. Ferulic acid diacid with an adipic acid linker (FAA) and tannic acid (TA) were used as shell and core molecules to form NPs via flash nanoprecipitation. These NPs showed a strong inhibitory effect on αSYN fibrillization, significantly diminishing αSYN fibrillization in vitro compared to untreated αSYN using a Thioflavin T assay. Treating microglia with NPs decreased overall αSYN internalization and intracellular αSYN oligomer formation. NP treatment additionally lowered the in vitro secretion of pro-inflammatory cytokines TNF-α and IL-6, and also attenuated nitric oxide and reactive oxygen species production induced by αSYN. NP treatment also significantly decreased Iba-1 expression in αSYN-challenged microglia and suppressed nuclear translocation of nuclear factor kappa B (NF-κB). Overall, this work lays the foundation for an antioxidant-based nanotherapeutic candidate to target pathological protein aggregation and neuroinflammation in neurodegenerative diseases.

5.
Tissue Eng Part A ; 26(3-4): 193-205, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31537172

RESUMEN

Cell replacement therapy is a promising treatment strategy for Parkinson's disease (PD); however, the poor survival rate of transplanted neurons is a critical barrier to functional recovery. In this study, we used self-assembling peptide nanofiber scaffolds (SAPNS) based on the peptide RADA16-I to support the in vitro maturation and in vivo post-transplantation survival of encapsulated human dopaminergic (DA) neurons derived from induced pluripotent stem cells. Neurons encapsulated within the SAPNS expressed mature neuronal and midbrain DA markers and demonstrated in vitro functional activity similar to neurons cultured in two dimensions. A microfluidic droplet generation method was used to encapsulate cells within monodisperse SAPNS microspheres, which were subsequently used to transplant adherent, functional networks of DA neurons into the striatum of a 6-hydroxydopamine-lesioned PD mouse model. SAPNS microspheres significantly increased the in vivo survival of encapsulated neurons compared with neurons transplanted in suspension, and they enabled significant recovery in motor function compared with control lesioned mice using approximately an order of magnitude fewer neurons than have been previously needed to demonstrate behavioral recovery. These results indicate that such biomaterial scaffolds can be used as neuronal transplantation vehicles to successfully improve the outcome of cell replacement therapies for PD. Impact Statement Transplantation of dopaminergic (DA) neurons holds potential as a treatment for Parkinson's disease (PD), but low survival rates of transplanted neurons is a barrier to successfully improving motor function. In this study, we used hydrogel scaffolds to transplant DA neurons into PD model mice. The hydrogel scaffolds enhanced survival of the transplanted neurons compared with neurons that were transplanted in a conventional manner, and they also improved recovery of motor function by using significantly fewer neurons than have typically been transplanted to see functional benefits. This cell transplantation technology has the capability to improve the outcome of neuron transplantation therapies.


Asunto(s)
Neuronas Dopaminérgicas/citología , Células Madre Pluripotentes Inducidas/citología , Péptidos/química , Andamios del Tejido/química , Materiales Biocompatibles/química , Neuronas Dopaminérgicas/trasplante , Humanos , Hidrogeles/química , Células Madre Pluripotentes Inducidas/trasplante , Trasplante de Células Madre
6.
Med One ; 42019.
Artículo en Inglés | MEDLINE | ID: mdl-31592196

RESUMEN

As a nascent and emerging field that holds great potential for precision oncology, nanotechnology has been envisioned to improve drug delivery and imaging capabilities through precise and efficient tumor targeting, safely sparing healthy normal tissue. In the clinic, nanoparticle formulations such as the first-generation Abraxane® in breast cancer, Doxil® for sarcoma, and Onivyde® for metastatic pancreatic cancer, have shown advancement in drug delivery while improving safety profiles. However, effective accumulation of nanoparticles at the tumor site is sub-optimal due to biological barriers that must be overcome. Nanoparticle delivery and retention can be altered through systematic design considerations in order to enhance passive accumulation or active targeting to the tumor site. In tumor niches where passive targeting is possible, modifications in the size and charge of nanoparticles play a role in their tissue accumulation. For niches in which active targeting is required, precision oncology research has identified targetable biomarkers, with which nanoparticle design can be altered through bioconjugation using antibodies, peptides, or small molecule agonists and antagonists. This review is structured to provide a better understanding of nanoparticle engineering design principles with emphasis on overcoming tumor-specific biological barriers.

7.
J Tissue Eng Regen Med ; 11(1): 285-294, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-24889394

RESUMEN

Freeze casting, or controlled unidirectional solidification, can be used to fabricate chitosan-alginate (C-A) scaffolds with highly aligned porosity that are suitable for use as nerve-guidance channels. To augment the guidance of growth across a spinal cord injury lesion, these scaffolds are now evaluated in vitro to assess their ability to release neurotrophin-3 (NT-3) and chondroitinase ABC (chABC) in a controlled manner. Protein-loaded microcapsules were incorporated into C-A scaffolds prior to freeze casting without affecting the original scaffold architecture. In vitro protein release was not significantly different when comparing protein loaded directly into the scaffolds with release from scaffolds containing incorporated microcapsules. NT-3 was released from the C-A scaffolds for 8 weeks in vitro, while chABC was released for up to 7 weeks. Low total percentages of protein released from the scaffolds over this time period were attributed to limitation of diffusion by the interpenetrating polymer network matrix of the scaffold walls. NT-3 and chABC released from the scaffolds retained bioactivity, as determined by a neurite outgrowth assay, and the promotion of neurite growth across an inhibitory barrier of chondroitin sulphate proteoglycans. This demonstrates the potential of these multifunctional scaffolds for enhancing axonal regeneration through growth-inhibiting glial scars via the sustained release of chABC and NT-3. Copyright © 2014 John Wiley & Sons, Ltd.


Asunto(s)
Condroitina ABC Liasa/metabolismo , Neuroglía/patología , Neuronas/fisiología , Neurotrofina 3/metabolismo , Ingeniería de Tejidos/métodos , Andamios del Tejido , Alginatos/química , Animales , Axones/patología , Quitosano/química , Composición de Medicamentos , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Polímeros/química , Traumatismos de la Médula Espinal/terapia
8.
Nat Commun ; 7: 10862, 2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26983594

RESUMEN

Cell replacement therapy with human pluripotent stem cell-derived neurons has the potential to ameliorate neurodegenerative dysfunction and central nervous system injuries, but reprogrammed neurons are dissociated and spatially disorganized during transplantation, rendering poor cell survival, functionality and engraftment in vivo. Here, we present the design of three-dimensional (3D) microtopographic scaffolds, using tunable electrospun microfibrous polymeric substrates that promote in situ stem cell neuronal reprogramming, neural network establishment and support neuronal engraftment into the brain. Scaffold-supported, reprogrammed neuronal networks were successfully grafted into organotypic hippocampal brain slices, showing an ∼ 3.5-fold improvement in neurite outgrowth and increased action potential firing relative to injected isolated cells. Transplantation of scaffold-supported neuronal networks into mouse brain striatum improved survival ∼ 38-fold at the injection site relative to injected isolated cells, and allowed delivery of multiple neuronal subtypes. Thus, 3D microscale biomaterials represent a promising platform for the transplantation of therapeutic human neurons with broad neuro-regenerative relevance.


Asunto(s)
Encéfalo/citología , Reprogramación Celular , Imagenología Tridimensional , Neuronas/citología , Neuronas/trasplante , Andamios del Tejido/química , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Humanos , Polímeros/química , Factores de Transcripción/metabolismo
9.
ACS Biomater Sci Eng ; 2(6): 1030-1038, 2016 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-32582837

RESUMEN

While cell transplantation presents a potential strategy to treat the functional deficits of neurodegenerative diseases or central nervous system injuries, the poor survival rate of grafted cells in vivo is a major barrier to effective therapeutic treatment. In this study, we investigated the role of a peptide-based nanofibrous scaffold composed of the self-assembling peptide RADA16-I to support the reprogramming and maturation of human neurons in vitro and to transplant these neurons in vivo. The induced human neurons were generated via the single transcriptional factor transduction of induced pluripotent stem cells (iPSCs), which are a promising cell source for regenerative therapies. These neurons encapsulated within RADA16-I scaffolds displayed robust neurite outgrowth and demonstrated high levels of functional activity in vitro compared to that of 2-D controls, as determined by live cell calcium imaging. When evaluated in vivo as a transplantation vehicle for adherent, functional networks of neurons, monodisperse RADA16-I microspheres significantly increased survival (over 100-fold greater) compared to the conventional transplantation of unsupported neurons in suspension. The scaffold-encapsulated neurons integrated well in vivo within the injection site, extending neurites several hundred microns long into the host brain tissue. Overall, these results suggest that this biomaterial platform can be used to successfully improve the outcome of cell transplantation and neuro-regenerative therapies.

10.
J Biomed Mater Res A ; 101(12): 3493-503, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23596011

RESUMEN

Several strategies have been investigated to enhance axonal regeneration after spinal cord injury, however, the resulting growth can be random and disorganized. Bioengineered scaffolds provide a physical substrate for guidance of regenerating axons towards their targets, and can be produced by freeze casting. This technique involves the controlled directional solidification of an aqueous solution or suspension, resulting in a linearly aligned porous structure caused by ice templating. In this study, freeze casting was used to fabricate porous chitosan-alginate (C/A) scaffolds with longitudinally oriented channels. Chick dorsal root ganglia explants adhered to and extended neurites through the scaffold in parallel alignment with the channel direction. Surface adsorption of a polycation and laminin promoted significantly longer neurite growth than the uncoated scaffold (poly-L-ornithine + Laminin = 793.2 ± 187.2 µm; poly-L-lysine + Laminin = 768.7 ± 241.2 µm; uncoated scaffold = 22.52 ± 50.14 µm) (P < 0.001). The elastic modulus of the hydrated scaffold was determined to be 5.08 ± 0.61 kPa, comparable to reported spinal cord values. The present data suggested that this C/A scaffold is a promising candidate for use as a nerve guidance scaffold, because of its ability to support neuronal attachment and the linearly aligned growth of DRG neurites.


Asunto(s)
Alginatos/farmacología , Quitosano/farmacología , Hielo , Sistema Nervioso/efectos de los fármacos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Congelación , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ácido Glucurónico/farmacología , Ácidos Hexurónicos/farmacología , Ensayo de Materiales , Fenómenos Mecánicos/efectos de los fármacos , Microscopía Electrónica de Rastreo , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Microtomografía por Rayos X
11.
J Microencapsul ; 28(5): 353-62, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21736520

RESUMEN

Transplantation of cells genetically modified to produce neurotrophins is a promising spinal cord repair strategy. Previously we showed that fibroblasts engineered to produce brain-derived neurotrophic factor (Fb/BDNF) microencapsulated in alginate survive, continue to grow and express bioactive BDNF. We here compared the effects of different alginate crosslinkers on dorsal root ganglia (DRG) neurite growth using alginate-encapsulated Fb/BDNF. Aqueous sodium alginate (±Fb/BDNF) was contacted with different calcium salts, and used as substrate for DRG growth. Length, number and orientation of neurites were measured. Chloride or carbonate salts promoted significantly more neurite growth than sulphate, with or without Fb/BDNF, although encapsulated Fb/BDNF stimulated significantly more neurite growth than cell-free. An Fb/BDNF-derived neurotrophin concentration gradient directionally guided DRG neurite growth. This positive effect of alginate-encapsulated Fb/BDNF on neurite growth/guidance shows promise for enhanced regeneration and guidance of axons towards a specific target in the injured spinal cord.


Asunto(s)
Alginatos/farmacología , Composición de Medicamentos , Fibroblastos/metabolismo , Factores de Crecimiento Nervioso/administración & dosificación , Neuritas/efectos de los fármacos , Regeneración de la Medula Espinal , Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Reactivos de Enlaces Cruzados , Ganglios Espinales , Ácido Glucurónico/farmacología , Ácidos Hexurónicos/farmacología , Humanos , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/uso terapéutico , Regeneración Nerviosa
12.
Biomacromolecules ; 11(11): 2936-43, 2010 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-20942395

RESUMEN

Engineered tissue strategies for central nervous system (CNS) repair have the potential for localizing treatment using a wide variety of cells or growth factors. However, these strategies are often limited by their ability to address only one aspect of the injury. Here we report the development of a novel alginate construct that acts as a multifunctional tissue scaffold for CNS repair, and as a localized growth factor delivery vehicle. We show that the surface of this alginate construct acts as an optimal growth environment for neural progenitor cell (NPC) attachment, survival, migration, and differentiation. Importantly, we show that tailor-made alginate constructs containing brain-derived neurotrophic factor or neurotrophin-3 differentially direct lineage fates of NPCs and may therefore be useful in treating a wide variety of injuries. It is this potential for directed differentiation of a scaffold prior to implantation at the injury site that we explore here.


Asunto(s)
Fibroblastos/metabolismo , Ingeniería Genética , Hidrogeles/química , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Células-Madre Neurales/citología , Animales , Cápsulas/química , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Fibroblastos/química , Fibroblastos/citología , Tamaño de la Partícula , Ratas , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...