Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Control Release ; 358: 43-58, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37088306

RESUMO

Demyelinating disorders, with a particular focus on multiple sclerosis (MS), have a multitude of detrimental cognitive and physical effects on the patients. Current treatment options that involve substances promoting remyelination fail in the clinics due to difficulties in reaching the central nervous system (CNS). Here, the dual encapsulation of retinoic acid (RA) into lipid nanocapsules with a nominal size of 70 nm, and a low PdI of 0.1, coupled with super paramagnetic iron oxide nanoparticles (SPIONs) was accomplished, and joined by an external functionalization process with a transferrin-receptor binding peptide. This nanosystem showed a 3-fold improved internalization by endothelial cells compared to the free drug, ability to interact with oligodendrocyte progenitor cells and microglia, and improvements in the permeability through the blood-brain barrier by 5-fold. The lipid nanocapsules also induced the differentiation of oligodendrocyte progenitor cells into more mature, myelin producing oligodendrocytes, as evaluated by high-throughput image screening, by 3-5-fold. Furthermore, the ability to tame the inflammatory response was verified in lipopolysaccharide-stimulated microglia, suppressing the production of pro-inflammatory cytokines by 50-70%. Overall, the results show that this nanosystem can act in both the inflammatory microenvironment present at the CNS of affected patients, but also stimulate the differentiation of new oligodendrocytes, paving the way for a promising platform in the therapy of MS.


Assuntos
Doenças Desmielinizantes , Esclerose Múltipla , Nanocápsulas , Doenças Neurodegenerativas , Animais , Camundongos , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/metabolismo , Nanocápsulas/uso terapêutico , Tretinoína/farmacologia , Células Endoteliais/metabolismo , Esclerose Múltipla/tratamento farmacológico , Bainha de Mielina , Inflamação/tratamento farmacológico , Oligodendroglia , Diferenciação Celular , Doenças Neurodegenerativas/tratamento farmacológico , Lipídeos/farmacologia , Camundongos Endogâmicos C57BL
2.
Biomater Adv ; 146: 213312, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36736264

RESUMO

Spinal cord injury (SCI) is characterized by neuroinflammatory processes that are marked by an uncontrolled activation of microglia, which directly damages neurons. Natural and synthetic melanins represent an effective tool to treat neuroinflammation because they possess immunomodulatory properties. Here, the main objective was to evaluate the effect of eumelanin-coated poly(lactic acid) (EU@PLA) aligned microfibers on in vitro model of neuroinflammation related to spinal cord injury in terms of inflammatory mediators' modulation. Aligned fibers were chosen to provide physical cues to guide axonal growth in a specific direction thus restoring the synaptic connection. Eumelanin decorated PLA electrospun substrates were produced combining electrospinning, spin coating and solid-state polymerization processes (oxidative coupling under oxygen atmosphere). Biological response in terms of antioxidant and anti-inflammatory activity was analyzed on an in vitro model of neuroinflammation [microglial cells stimulated with lipopolysaccharide (LPS)]. Cell morphology and EU@PLA mechanism of action, in terms of toll-like receptor-4 (TLR-4) involvement were assessed. The results show that EU@PLA fibers were able to decrease reactive oxygen species, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-кB) expression >50 % compared to PLA + LPS and interleukin 6 (IL-6) secretion about 20 %. Finally, the mechanism of action of EU@PLA in microglia was found to be dependent on the TLR-4 signaling. Protein expression analysis revealed a decreased in TLR-4 production induced by LPS stimulation in presence of EU@PLA. Overall, our results show that EU@PLA represents an innovative and effective strategy for the control of inflammatory response in central nervous system.


Assuntos
Melaninas , Traumatismos da Medula Espinal , Ratos , Animais , Receptor 4 Toll-Like , Ratos Sprague-Dawley , Doenças Neuroinflamatórias , Lipopolissacarídeos/farmacologia , Traumatismos da Medula Espinal/terapia , Traumatismos da Medula Espinal/metabolismo , Poliésteres
3.
Biomater Sci ; 11(4): 1499-1516, 2023 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-36602540

RESUMO

Nanomedicines based on nanoparticles as carriers of therapeutics are expected to drastically influence the future of healthcare. However, clinical translation of these technologies can be very challenging. The development process of nanoparticles for biological applications encompasses the analysis and understanding of several steps in vitro, before in vivo, and subsequent clinical applications, namely, the in-depth study of biosafety, cellular interaction, and intracellular trafficking. Recently, we proposed a new family of fully biodegradable PEG-GATGE (Poly(Ethylene Glycol)-Gallic Acid-Triethylene Glycol Ester) dendritic block copolymers to act as versatile delivery vectors in nanomedicine. These nanosystems showed great promise in complexing, protecting, and delivering nucleic acids to cells, forming nanoscaled complexes called dendriplexes. Due to these favourable features, in the present study, the dendriplexes' characterization was expanded and, in addition, their biocompatibility, cellular uptake, and cellular path in neuronal cells from the peripheral and central nervous systems were assessed. Our fully biodegradable dendritic nanosystem was found to be biocompatible in all the studied neuronal cells and mediates fast cellular interaction and endocytosis in both cell line tested and primary mouse cortical neurons. Nevertheless, the mechanism of dendriplex cell entry and intracellular fate was found to be different in cell lines and primary cultures. Dendriplexes' internalization was observed to be mediated by clathrin in ND7/23 and HT22 cells, while caveolin-mediated endocytosis occurred in primary mouse cortical neurons, in which, after internalization, dendriplexes were not colocalized with lysosomes or autophagosomes. Taken together, these results further point to PEG-GATGE dendrimers as biosafe delivery vectors of nucleic acids to neuronal cells in vitro, suggesting their feasibility as carriers in the context of nervous system applications. Furthermore, our data reinforce the importance of testing the performance of new vectors in different models to verify their potential applicability in vitro and/or in vivo.


Assuntos
Nanopartículas , Ácidos Nucleicos , Camundongos , Animais , Polietilenoglicóis/metabolismo , Polímeros , Nanopartículas/metabolismo , Linhagem Celular , Endocitose , Neurônios/metabolismo
4.
Front Neurosci ; 16: 861613, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35573316

RESUMO

Cells are continuously exposed to physical forces and the central nervous system (CNS) is no exception. Cells dynamically adapt their behavior and remodel the surrounding environment in response to forces. The importance of mechanotransduction in the CNS is illustrated by exploring its role in CNS pathology development and progression. The crosstalk between the biochemical and biophysical components of the extracellular matrix (ECM) are here described, considering the recent explosion of literature demonstrating the powerful influence of biophysical stimuli like density, rigidity and geometry of the ECM on cell behavior. This review aims at integrating mechanical properties into our understanding of the molecular basis of CNS disease. The mechanisms that mediate mechanotransduction events, like integrin, Rho/ROCK and matrix metalloproteinases signaling pathways are revised. Analysis of CNS pathologies in this context has revealed that a wide range of neurological diseases share as hallmarks alterations of the tissue mechanical properties. Therefore, it is our belief that the understanding of CNS mechanotransduction pathways may lead to the development of improved medical devices and diagnostic methods as well as new therapeutic targets and strategies for CNS repair.

5.
Biomaterials ; 281: 121356, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35032909

RESUMO

The repeated administration of non-degradable dendrimers can lead to toxicity due to their bioaccumulation. Furthermore, in drug delivery applications, carrier stability can result in low biological performance due to insufficient intracellular cargo release. A novel family of versatile, biosafe, water-soluble, and fully biodegradable PEG-dendritic nanosystems is proposed, which overcomes the limitations of the most used dendrimers. Their novelty relies on the full and adjustable degradability thanks to the presence of tunable ester bonds in every dendritic arm. These dendritic nanosystems present peripheral azides that allow their easy multivalent functionalization, by "click" chemistry, with a vast range of ligands to act as versatile carriers. Here, their amine-functionalization to serve as nucleic acid vectors for gene therapy is explored. These nanosystems complex and protect efficiently siRNA in very small dendriplexes (<60 nm), being successfully cell-internalized, including in hard-to-transfect neuronal cells even when in full tissue explants (dorsal root ganglia). Importantly, full biodegradability was crucial for an efficient nucleic acid intracellular release and the attainment of excellent transfection efficiencies. The reported fully biodegradable dendritic nanosystems can act as multi-function nanotherapeutics for gene therapy, and also for broader applications in nanomedicine. Therefore, they represent top-notch and clinically translatable health facilitating nanotechnologies for further developments in theranostics.


Assuntos
Dendrímeros , Ácidos Nucleicos , Dendrímeros/química , Sistemas de Liberação de Medicamentos , RNA Interferente Pequeno , Transfecção
6.
Antioxidants (Basel) ; 10(7)2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34356328

RESUMO

Parkinson's disease's etiology is unknown, although evidence suggests the involvement of oxidative modifications of intracellular components in disease pathobiology. Despite the known involvement of the extracellular matrix in physiology and disease, the influence of oxidative stress on the matrix has been neglected. The chemical modifications that might accumulate in matrix components due to their long half-live and the low amount of extracellular antioxidants could also contribute to the disease and explain ineffective cellular therapies. The enriched striatal extracellular matrix from a mouse model of Parkinson's disease was characterized by Raman spectroscopy. We found a matrix fingerprint of increased oxalate content and oxidative modifications. To uncover the effects of these changes on brain cells, we morphologically characterized the primary microglia used to repopulate this matrix and further quantified the effects on cellular mechanical stress by an intracellular fluorescence resonance energy transfer (FRET)-mechanosensor using the U-2 OS cell line. Our data suggest changes in microglia survival and morphology, and a decrease in cytoskeletal tension in response to the modified matrix from both hemispheres of 6-hydroxydopamine (6-OHDA)-lesioned animals. Collectively, these data suggest that the extracellular matrix is modified, and underscore the need for its thorough investigation, which may reveal new ways to improve therapies or may even reveal new therapies.

7.
Front Neurosci ; 15: 787518, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975386

RESUMO

Platinum nanoparticles (PtNPs) have unique physico-chemical properties that led to their use in many branches of medicine. Recently, PtNPs gathered growing interest as delivery vectors for drugs, biosensors and as surface coating on chronically implanted biomedical devices for improving electrochemical properties. However, there are contradictory statements about their biocompatibility and impact on target organs such as the brain tissue, where these NPs are finding many applications. Furthermore, many of the reported studies are conducted in homeostasis conditions and, consequently, neglect the impact of the pathologic conditions on the tissue response. To expand our knowledge on the effects of PtNPs on neuronal and glial cells, we investigated the acute effects of monodisperse sodium citrate-coated PtNPs on rat organotypic hippocampal cultures in physiological or neuronal excitotoxic conditions induced by kainic acid (KA). The cellular responses of the PtNPs were evaluated through cytotoxic assays and confocal microscopy analysis. To mimic a pathologic scenario, 7-day organotypic hippocampal cultures were exposed to KA for 24 h. Subsequently, PtNPs were added to each slice. We show that incubation of the slices with PtNPs for 24 h, does not severely impact cell viability in normal conditions, with no significant differences when comparing the dentate gyrus (DG), as well as CA3 and CA1 pyramidal cell layers. Such effects are not exacerbated in KA-treated slices, where the presence of PtNPs does not cause additional neuronal propidium iodide (PI) uptake in CA3 and CA1 pyramidal cell layers. However, PtNPs cause microglial cell activation and morphological alterations in CA3 and DG regions indicating the establishment of an inflammatory reaction. Morphological analysis revealed that microglia acquire activated ameboid morphology with loss of ramifications, as a result of their response to PtNPs contact. Surprisingly, this effect is not increased in pathological conditions. Taken together, these results show that PtNPs cause microglia alterations in short-term studies. Additionally, there is no worsening of the tissue response in a neuropathological induced scenario. This work highlights the need of further research to allow for the safe use of PtNPs. Also, it supports the demand of the development of novel and more biocompatible NPs to be applied in the brain.

8.
J Control Release ; 322: 390-400, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32247807

RESUMO

The central nervous system (CNS), namely the brain, still remains as the hardest area of the human body to achieve adequate concentration levels of most drugs, mainly due to the limiting behavior of its physical and biological defenses. Lipid nanocapsules emerge as a versatile platform to tackle those barriers, and efficiently delivery different drug payloads due to their numerous advantages. They can be produced in a fast, solvent-free and scalable-up process, and their properties can be fine-tuned for to make an optimal brain drug delivery vehicle. Moreover, lipid nanocapsule surface modification can further improve their bioavailability towards the central nervous system. Coupling these features with alternative delivery methods that stem to disrupt or fully circumvent the blood-brain barrier may fully harness the therapeutic advance that lipid nanocapsules can supply to current treatment options. Thus, this review intends to critically address the development of lipid nanocapsules, as well as to highlight the key features that can be modulated to ameliorate their properties towards the central nervous system delivery, mainly through intravenous methods, and how the pathological microenvironment of the CNS can be taken advantage of. The different routes to promote drug delivery towards the brain parenchyma are also discussed, as well as the synergetic effect that can be obtained by combining modified lipid nanocapsules with new/smart administration routes.


Assuntos
Nanocápsulas , Disponibilidade Biológica , Barreira Hematoencefálica , Sistema Nervoso Central , Sistemas de Liberação de Medicamentos , Humanos , Lipídeos
9.
J Pharm Sci ; 109(4): 1594-1604, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31935391

RESUMO

Current treatment options for bone-related disorders rely on a systemic administration of therapeutic agents that possess low solubility and intracellular bioavailability, as well as a high pharmacokinetic variability, which in turn lead to major off-target side effects. Hence, there is an unmet need of developing drug delivery systems that can improve the clinical efficacy of such therapeutic agents. Nanoparticle delivery systems might serve as promising carriers of hydrophobic molecules. Here, we propose 2 nanoparticle-based delivery systems based on monomethoxy poly(ethylene glycol)-poly(trimethyl carbonate) (mPEG-PTMC) and poly(lactide-co-glycolide) for the intracellular controlled release of a small hydrophobic drug (dexamethasone) to osteoblast cells in vitro. mPEG-PTMC self-assembles into stable nanoparticles in the absence of surfactant and shows a greater entrapment capacity of dexamethasone, while assuring bioactivity in MC3T3-E1 and bone marrow stromal cells cultured under apoptotic and osteogenic conditions, respectively. The mPEG-PTMC nanoparticles represent a potential vector for the intracellular delivery of hydrophobic drugs in the framework of bone-related diseases.


Assuntos
Nanopartículas , Preparações Farmacêuticas , Dioxanos , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Tamanho da Partícula , Polietilenoglicóis , Polímeros
10.
Front Neurosci ; 13: 689, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31333407

RESUMO

The development of implantable neuroelectrodes is advancing rapidly as these tools are becoming increasingly ubiquitous in clinical practice, especially for the treatment of traumatic and neurodegenerative disorders. Electrodes have been exploited in a wide number of neural interface devices, such as deep brain stimulation, which is one of the most successful therapies with proven efficacy in the treatment of diseases like Parkinson or epilepsy. However, one of the main caveats related to the clinical application of electrodes is the nervous tissue response at the injury site, characterized by a cascade of inflammatory events, which culminate in chronic inflammation, and, in turn, result in the failure of the implant over extended periods of time. To overcome current limitations of the most widespread macroelectrode based systems, new design strategies and the development of innovative materials with superior biocompatibility characteristics are currently being investigated. This review describes the current state of the art of in vitro, ex vivo, and in vivo models available for the study of neural tissue response to implantable microelectrodes. We particularly highlight new models with increased complexity that closely mimic in vivo scenarios and that can serve as promising alternatives to animal studies for investigation of microelectrodes in neural tissues. Additionally, we also express our view on the impact of the progress in the field of neural tissue engineering on neural implant research.

11.
Biomaterials ; 192: 601-611, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30509501

RESUMO

Laminin immobilization into diverse biological and synthetic matrices has been explored to replicate the microenvironment of stem cell niches and gain insight into the role of extracellular matrix (ECM) on stem cell behavior. However, the site-specific immobilization of this heterotrimeric glycoprotein and, consequently, control over its orientation and bioactivity has been a challenge that has limited many of the explored strategies to date. In this work, we established an affinity-based approach that takes advantage of the native high affinity interaction between laminin and the human N-terminal agrin (hNtA) domain. This interaction is expected to promote the site-selective immobilization of laminin to a specific substrate, while preserving the exposure of its key bioactive epitopes. Recombinant hNtA (rhNtA) domain was produced with high purity (>90%) and successfully conjugated at its N-terminal with a thiol-terminated poly(ethylene glycol) (PEG) without affecting its affinity to laminin. Self-assembled monolayers (SAMs) of mono-PEGylated rhNtA on gold (mPEG rhNtA-SAMs) were then prepared to evaluate the effectiveness of this strategy. The site-specific immobilization of laminin onto mPEG rhNtA-SAMs was shown to better preserve protein bioactivity in comparison to laminin immobilized on SAMs of thiol-PEG-succinimidyl glutaramide (HS-PEG-SGA), used for the non-selective covalent immobilization of laminin, as evidenced by its enhanced ability to efficiently self-polymerize and mediate cell adhesion and spreading of human neural stem cells. These results highlight the potential of this novel strategy to be used as an alternative to the conventional immobilization approaches in a wide range of applications, including engineered coatings for neuroelectrodes and cell culture, as well as biofunctionalization of 3D matrices.


Assuntos
Agrina/química , Materiais Biocompatíveis/química , Proteínas Imobilizadas/química , Laminina/química , Adesão Celular , Linhagem Celular , Microambiente Celular , Humanos , Células-Tronco Neurais/citologia , Polietilenoglicóis/química , Proteínas Recombinantes/química , Compostos de Sulfidrila/química
12.
Sci Adv ; 4(8): eaar4008, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30116776

RESUMO

Muscle satellite cells (MuSCs) play a central role in muscle regeneration, but their quantity and function decline with comorbidity of trauma, aging, and muscle diseases. Although transplantation of MuSCs in traumatically injured muscle in the comorbid context of aging or pathology is a strategy to boost muscle regeneration, an effective cell delivery strategy in these contexts has not been developed. We engineered a synthetic hydrogel-based matrix with optimal mechanical, cell-adhesive, and protease-degradable properties that promotes MuSC survival, proliferation, and differentiation. Furthermore, we establish a biomaterial-mediated cell delivery strategy for treating muscle trauma, where intramuscular injections may not be applicable. Delivery of MuSCs in the engineered matrix significantly improved in vivo cell survival, proliferation, and engraftment in nonirradiated and immunocompetent muscles of aged and dystrophic mice compared to collagen gels and cell-only controls. This platform may be suitable for treating craniofacial and limb muscle trauma, as well as postoperative wounds of elderly and dystrophic patients.


Assuntos
Envelhecimento , Hidrogéis/química , Músculo Esquelético/citologia , Distrofias Musculares/terapia , Células Satélites de Músculo Esquelético/transplante , Ferimentos e Lesões/terapia , Animais , Diferenciação Celular , Comorbidade , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Regeneração , Células Satélites de Músculo Esquelético/citologia , Engenharia Tecidual , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia
13.
Acta Biomater ; 78: 247-259, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30092376

RESUMO

Poly(ethylene glycol) (PEG) has been extensively used to coat the surface of nanocarriers to improve their physicochemical properties and allow the grafting of targeting moieties. Still, to date there is no common agreement on the ideal PEG coverage-density or length to be used for optimum vector performance. In this study, we aimed to investigate the impact of both PEG density and length on the vectoring capacity of neuron-targeted gene-carrying trimethyl chitosan nanoparticles. The non-toxic fragment from the tetanus toxin (HC) was coupled to a 5 kDa heterobifunctional PEG (HC-PEG5k) reactive for the thiol groups inserted into the polymer backbone and grafted at different densities onto the nanoparticles. Internalization and transfection studies on neuronal versus non-neuronal cell lines allowed to determine the PEG density of 2 mol% of PEG chains per mol of primary amine groups as the one with superior biological performance. To enhance HC exposure and maximize cell-nanoparticle specific interaction, NPs containing different ratios of HC-PEG5k and 2 kDa methoxy-PEG at the same grafting density were produced. By intercalating HC-PEG5k with methoxy-PEG2k we attained the best performance in terms of internalization (higher payload delivery into cells) and transfection efficiency, using twice lower amount of HC. This outcome highlights the need for fine-tuning of PEG-modified nanoparticles towards the achievement of optimal targeting. STATEMENT OF SIGNIFICANCE: The amount and exposure of targeting moieties at a nanoparticle surface are critical parameters regarding the targeting potential of nanosized delivery vectors. However, to date, few studies have considered fundamental aspects impacting the ligand-receptor pair interaction, such as the effect of spacer chain length, flexibility or conformation. By optimizing the PEG spacer density and chain length grafted into nanoparticles, we were able to establish the formulation that maximizes cell-nanoparticle specific interaction and has superior biological performance. Our work shows that the precise adjustment of the PEG coverage-density presents a significant impact on the selectivity and bioactivity of the developed formulation, emphasizing the need for the fine-tuning of PEG-modified nanoparticles for the successful development of the next-generation nanomedicines.


Assuntos
Nanopartículas/química , Neurônios/metabolismo , Polietilenoglicóis/química , Animais , Linhagem Celular Tumoral , Quitosana/química , Endocitose , Ligantes , Camundongos , Ratos , Toxina Tetânica/metabolismo
14.
J Mater Sci Mater Med ; 28(10): 157, 2017 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-28894995

RESUMO

It is now widely accepted that a therapeutic strategy for spinal cord injury (SCI) demands a multi-target approach. Here we propose the use of an easily implantable bilayer polymeric patch based on poly(trimethylene carbonate-co-ε-caprolactone) (P(TMC-CL)) that combines physical guidance cues provided by electrospun aligned fibres and the delivery of ibuprofen, as a mean to reduce the inhibitory environment at the lesion site by taming RhoA activation. Bilayer patches comprised a solvent cast film onto which electrospun aligned fibres have been deposited. Both layers were loaded with ibuprofen. In vitro release (37°C, in phosphate buffered saline) of the drug from the loaded scaffolds under sink condition was found to occur in the first 24 h. The released ibuprofen was shown to retain its bioactivity, as indicated by the reduction of RhoA activation when the neuronal-like cell line ND7/23 was challenged with lysophosphatidic acid. Ibuprofen-loaded P(TMC-CL) bilayer scaffolds were successfully implanted in vivo in a dorsal hemisection rat SCI model mediating the reduction of RhoA activation after 5 days of implantation in comparison to plain P(TMC-CL) scaffolds. Immunohistochemical analysis of the tissue shows ßIII tubulin positive cells close to the ibuprofen-loaded patches further supporting the use of this strategy in the context of regeneration after a lesion in the spinal cord.


Assuntos
Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Ibuprofeno/administração & dosagem , Traumatismos da Medula Espinal/terapia , Alicerces Teciduais/química , Animais , Células Cultivadas , Dioxanos/química , Camundongos , Microtecnologia , Nanofibras/química , Regeneração Nervosa/efeitos dos fármacos , Regeneração Nervosa/fisiologia , Poliésteres/química , Polímeros/química , Ratos , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiologia , Engenharia Tecidual , Adesivo Transdérmico
15.
Curr Gene Ther ; 17(2): 105-119, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28494741

RESUMO

Over the last decades, gene therapy has emerged as a pioneering therapeutic approach to treat or prevent several diseases. Among the explored strategies, the short-term silencing of protein coding genes mediated by siRNAs has a good therapeutic potential in a clinical setting. However, the widespread use of siRNA will require the development of clinically suitable, safe and effective vehicles with the ability to complex and deliver siRNA into target cells with minimal toxicity. Lately, dendrimers have gained considerable attention as non-viral vectors in nucleic acid delivery due to their unique structural characteristics (globular, well defined and highly branched structure, multivalency, low polydispersity and tunable nanosize), along with their relevant capacity to complex and protect nucleic acids in compact nanostructures, which can be functionalized with targeting moieties in order to get cell specificity. Here, we present an overview of the state-of-the-art of the most significant and recent advances on the use of dendrimers as siRNA delivery vectors, with particular focus on the in vivo applications. We will cover the use of different dendrimers, distinct administration routes, toxicity issues, as well as the target tissue or disease, highlighting the potential of dendrimers as nanocarriers for therapeutic and biomedical applications.


Assuntos
Dendrímeros/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Técnicas de Transferência de Genes , Terapia Genética/métodos , RNA Interferente Pequeno/administração & dosagem , Animais , Dendrímeros/química , Portadores de Fármacos/química , Humanos , Nanomedicina/métodos , Interferência de RNA , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética
16.
Nucleic Acid Ther ; 26(6): 392-400, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27658045

RESUMO

ATM (ataxia-telangiectasia, mutated) is an important cancer susceptibility gene that encodes a key apical kinase in the DNA damage response pathway. ATM mutations in the germ line result in ataxia-telangiectasia (A-T), a rare genetic syndrome associated with hypersensitivity to double-strand DNA breaks and predisposition to lymphoid malignancies. ATM expression is limited by a tightly regulated nonsense-mediated RNA decay (NMD) switch exon (termed NSE) located in intron 28. In this study, we identify antisense oligonucleotides that modulate NSE inclusion in mature transcripts by systematically targeting the entire 3.1-kb-long intron. Their identification was assisted by a segmental deletion analysis of transposed elements, revealing NSE repression upon removal of a distant antisense Alu and NSE activation upon elimination of a long terminal repeat transposon MER51A. Efficient NSE repression was achieved by delivering optimized splice-switching oligonucleotides to embryonic and lymphoblastoid cells using chitosan-based nanoparticles. Together, these results provide a basis for possible sequence-specific radiosensitization of cancer cells, highlight the power of intronic antisense oligonucleotides to modify gene expression, and demonstrate transposon-mediated regulation of NSEs.


Assuntos
Elementos Alu , Proteínas Mutadas de Ataxia Telangiectasia/genética , Éxons , Oligonucleotídeos Antissenso/genética , Splicing de RNA , RNA Mensageiro/genética , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Sequência de Bases , Linhagem Celular Transformada , Quitosana/química , Elementos de DNA Transponíveis , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células HEK293 , Humanos , Íntrons , Linfócitos , Mutação , Nanopartículas/química , Nanopartículas/metabolismo , Oligonucleotídeos Antissenso/síntese química , Oligonucleotídeos Antissenso/metabolismo , Clivagem do RNA , Estabilidade de RNA , RNA Mensageiro/metabolismo
17.
Int J Nanomedicine ; 11: 2675-83, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27354797

RESUMO

A major challenge in neuronal gene therapy is to achieve safe, efficient, and minimally invasive transgene delivery to neurons. In this study, we report the use of a nonviral neurotropic poly(ethylene imine)-based nanoparticle that is capable of mediating neuron-specific transfection upon a subcutaneous injection. Nanoparticles were targeted to peripheral neurons by using the nontoxic carboxylic fragment of tetanus toxin (HC), which, besides being neurotropic, is capable of being retrogradely transported from neuron terminals to the cell bodies. Nontargeted particles and naked plasmid DNA were used as control. Five days after treatment by subcutaneous injection in the footpad of Wistar rats, it was observed that 56% and 64% of L4 and L5 dorsal root ganglia neurons, respectively, were expressing the reporter protein. The delivery mediated by HC-functionalized nanoparticles spatially limited the transgene expression, in comparison with the controls. Histological examination revealed no significant adverse effects in the use of the proposed delivery system. These findings demonstrate the feasibility and safety of the developed neurotropic nanoparticles for the minimally invasive delivery of genes to the peripheral nervous system, opening new avenues for the application of gene therapy strategies in the treatment of peripheral neuropathies.


Assuntos
Nanopartículas/administração & dosagem , Transfecção/métodos , Transgenes , Animais , Gânglios Espinais/citologia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Iminas/química , Injeções/métodos , Masculino , Nanopartículas/química , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Plasmídeos/administração & dosagem , Polietilenos/química , Ratos , Ratos Wistar , Toxina Tetânica/administração & dosagem , Toxina Tetânica/genética
18.
Drug Discov Today ; 21(9): 1355-1366, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27178019

RESUMO

Despite the recent progress in the understanding of neurodegenerative disorders, a lack of solid fundamental knowledge on the etiology of many of the major neurodegenerative diseases has made it difficult to obtain effective therapies to treat these conditions. Scientists have been looking to carry out more-human-relevant studies, with strong statistical power, to overcome the limitations of preclinical animal models that have contributed to the failure of numerous therapeutics in clinical trials. Here, we identify currently existing platforms to mimic central nervous system tissues, healthy and diseased, mainly focusing on cell-based platforms and discussing their strengths and limitations in the context of the high-throughput screening of new therapeutic targets and drugs.


Assuntos
Ensaios de Triagem em Larga Escala , Doenças Neurodegenerativas , Animais , Técnicas de Cultura de Células , Humanos
19.
J Tissue Eng Regen Med ; 10(3): E154-66, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23950030

RESUMO

The development of scaffolds that combine the delivery of drugs with the physical support provided by electrospun fibres holds great potential in the field of nerve regeneration. Here it is proposed the incorporation of ibuprofen, a well-known non-steroidal anti-inflammatory drug, in electrospun fibres of the statistical copolymer poly(trimethylene carbonate-co-ε-caprolactone) [P(TMC-CL)] to serve as a drug delivery system to enhance axonal regeneration in the context of a spinal cord lesion, by limiting the inflammatory response. P(TMC-CL) fibres were electrospun from mixtures of dichloromethane (DCM) and dimethylformamide (DMF). The solvent mixture applied influenced fibre morphology, as well as mean fibre diameter, which decreased as the DMF content in solution increased. Ibuprofen-loaded fibres were prepared from P(TMC-CL) solutions containing 5% ibuprofen (w/w of polymer). Increasing drug content to 10% led to jet instability, resulting in the formation of a less homogeneous fibrous mesh. Under the optimized conditions, drug-loading efficiency was above 80%. Confocal Raman mapping showed no preferential distribution of ibuprofen in P(TMC-CL) fibres. Under physiological conditions ibuprofen was released in 24 h. The release process being diffusion-dependent for fibres prepared from DCM solutions, in contrast to fibres prepared from DCM-DMF mixtures where burst release occurred. The biological activity of the drug released was demonstrated using human-derived macrophages. The release of prostaglandin E2 to the cell culture medium was reduced when cells were incubated with ibuprofen-loaded P(TMC-CL) fibres, confirming the biological significance of the drug delivery strategy presented. Overall, this study constitutes an important contribution to the design of a P(TMC-CL)-based nerve conduit with anti-inflammatory properties.


Assuntos
Dioxanos/química , Ibuprofeno/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Poliésteres/química , Engenharia Tecidual/métodos , Anti-Inflamatórios/farmacologia , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Dinoprostona/metabolismo , Liberação Controlada de Fármacos , Humanos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Microscopia Eletrônica de Varredura , Espectroscopia de Infravermelho com Transformada de Fourier
20.
Soft Matter ; 11(41): 8113-25, 2015 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-26335751

RESUMO

Cationic polymers have recently attracted attention due to their proven potential for nonviral gene delivery. In this study, we report novel biocompatible nanocomplexes produced using chemically functionalized N,N,N-trimethyl chitosan (TMC) with different N-acyl chain lengths (C5-C18) associated with single-stranded oligonucleotides. The TMC derivatives were synthesized by covalent coupling reactions of quaternized chitosan with n-pentanoic (C5), n-decanoic (C10), and n-octadecanoic (C18) fatty acids, which were extensively characterized by Fourier transform-infrared spectroscopy (FT-IR) and proton nuclear magnetic resonance ((1)H NMR). These N-acylated TMC derivatives (TMCn) were used as cationic polymeric matrices for encapsulating anionic 18-base single-stranded thiophosphorylated oligonucleotides (ssONs), leading to the formation of polyplexes further characterized by zeta potential (ZP), dynamic light scattering (DLS), binding affinity, transfection efficiency and in vitro cytotoxicity assays. The results demonstrated that the length of the grafted hydrophobic N-acyl chain and the relative amino:phosphate groups ratio (N/P ratio) between the TMC derivatives and ssON played crucial roles in determining the physicochemical properties of the obtained nanocomplexes. While none of the tested derivatives showed appreciable cytotoxicity, the type of acyl chain had a remarkable influence on the cell transfection capacity of TMC-ssON nanocomplexes with the derivatives based on stearic acid showing the best performance based on the results of in vitro assays using a model cell line expressing luciferase (HeLa/Luc705).


Assuntos
Quitosana/química , Nanopartículas/química , Oligonucleotídeos/química , Sobrevivência Celular/efeitos dos fármacos , Quitosana/metabolismo , Quitosana/toxicidade , Difusão Dinâmica da Luz , Ácidos Graxos/química , Células HeLa , Humanos , Espectroscopia de Ressonância Magnética , Nanopartículas/toxicidade , Oligonucleotídeos/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...