Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomater Sci ; 11(12): 4238-4253, 2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-36987922

RESUMO

Traumatic brain injury (TBI) affects millions of people worldwide, yet there are currently no therapeutics that address the long-term impairments that develop in a large portion of survivors. Lipid nanoparticles (LNPs) are a promising therapeutic strategy that may address the molecular basis of TBI pathophysiology. LNPs are the only non-viral gene delivery platform to achieve clinical success, but systemically administered formulations have only been established for targets in the liver. In this work, we evaluated the pharmacokinetics and activity of LNPs formulated with polyethylene glycol (PEG)-lipids of different anchor lengths when systemically administered to a mouse model of TBI. We observed an increase in LNP accumulation and activity in the injured brain hemisphere compared to the uninjured contralateral brain hemisphere. Interestingly, transgene expression mediated by LNPs was more durable in injured brain tissue compared to off-target organs when compared between 4 and 24 hours. The PEG-lipid is an important component of LNP formulation necessary for the stable formation and storage of LNPs, but the PEG-lipid structure and content also has an impact on LNP function. LNP formulations containing various ratios of PEG-lipid with C18 (DSPE-PEG) and C14 (DMG-PEG) anchors displayed similar physicochemical properties, independent of the PEG-lipid compositions. As the proportion of DSPE-PEG was increased in formulations, blood circulation times of LNPs increased and the duration of expression increased. We also evaluated diffusion of LNPs after convection enhanced delivery (CED) in healthy brains and found LNPs distributed >1 mm away from the injection site. Understanding LNP pharmacokinetics and activity in TBI models and the impact of PEG-lipid anchor length informs the design of LNP-based therapies for TBI after systemic administration.


Assuntos
Lesões Encefálicas Traumáticas , Nanopartículas , Camundongos , Animais , Polietilenoglicóis/química , Lipossomos , Nanopartículas/química , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lipídeos/química , RNA Interferente Pequeno/genética
2.
Bioconjug Chem ; 33(9): 1685-1697, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36017941

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas , Nanopartículas , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Matriz Extracelular , Ligantes , Camundongos , Peptídeos/uso terapêutico , Porosidade , Silício
3.
AAPS J ; 23(5): 100, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34401968

RESUMO

Peptides are used to control the pharmacokinetic profiles of nanoparticles due to their ability to influence tissue accumulation and cellular interactions. However, beyond the study of specific peptides, there is a lack of understanding of how peptide physicochemical properties affect nanoparticle pharmacokinetics, particularly in the context of traumatic brain injury (TBI). We engineered nanoparticle surfaces with peptides that possess a range of physicochemical properties and evaluated their distribution after two routes of administration: direct injection into a healthy mouse brain and systemic delivery in a mouse model of TBI. In both administration routes, we found that peptide-modified nanoparticle pharmacokinetics were influenced by the charge characteristics of the peptide. When peptide-modified nanoparticles are delivered directly into the brain, nanoparticles modified with positively charged peptides displayed restricted distribution from the injection site compared to nanoparticles modified with neutral, zwitterionic, or negatively charged peptides. After intravenous administration in a TBI mouse model, positively charged peptide-modified nanoparticles accumulated more in off-target organs, including the heart, lung, and kidneys, than zwitterionic, neutral, or negatively charged peptide-modified nanoparticles. The increase in off-target organ accumulation of positively charged peptide-modified nanoparticles was concomitant with a relative decrease in accumulation in the injured brain compared to zwitterionic, neutral, or negatively charged peptide-modified nanoparticles. Understanding how nanoparticle pharmacokinetics are influenced by the physicochemical properties of peptides presented on the nanoparticle surface is relevant to the development of nanoparticle-based TBI therapeutics and broadly applicable to nanotherapeutic design, including synthetic nanoparticles and viruses.


Assuntos
Barreira Hematoencefálica/metabolismo , Lesões Encefálicas Traumáticas/tratamento farmacológico , Sistemas de Liberação de Fármacos por Nanopartículas/farmacocinética , Fármacos Neuroprotetores/administração & dosagem , Peptídeos/química , Animais , Disponibilidade Biológica , Lesões Encefálicas Traumáticas/patologia , Engenharia Química , Química Farmacêutica , Modelos Animais de Doenças , Feminino , Meia-Vida , Humanos , Camundongos , Sistemas de Liberação de Fármacos por Nanopartículas/química , Peptídeos/farmacocinética , Distribuição Tecidual
4.
Mol Pharm ; 18(2): 522-538, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32584042

RESUMO

Acute brain injuries such as traumatic brain injury and stroke affect 85 million people a year worldwide, and many survivors suffer from long-term physical, cognitive, or psychosocial impairments. There are few FDA-approved therapies that are effective at preventing, halting, or ameliorating the state of disease in the brain after acute brain injury. To address this unmet need, one potential strategy is to leverage the unique physical and biological properties of nanomaterials. Decades of cancer nanomedicine research can serve as a blueprint for innovation in brain injury nanomedicines, both to emulate the successes and also to avoid potential pitfalls. In this review, we discuss how shared disease physiology between cancer and acute brain injuries can inform the design of novel nanomedicines for acute brain injuries. These disease hallmarks include dysregulated vasculature, an altered microenvironment, and changes in the immune system. We discuss several nanomaterial strategies that can be engineered to exploit these disease hallmarks, for example, passive accumulation, active targeting of disease-associated signals, bioresponsive designs that are "smart", and immune interactions.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Portadores de Fármacos/química , Nanopartículas/química , Fármacos Neuroprotetores/administração & dosagem , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Disponibilidade Biológica , Barreira Hematoencefálica/metabolismo , Encéfalo/imunologia , Encéfalo/patologia , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Fármacos Neuroprotetores/farmacocinética , Permeabilidade , Acidente Vascular Cerebral/patologia , Distribuição Tecidual , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
5.
ACS Sens ; 5(3): 686-692, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32100994

RESUMO

Currently, traumatic brain injury (TBI) is detected by medical imaging; however, medical imaging requires expensive capital equipment, is time- and resource-intensive, and is poor at predicting patient prognosis. To date, direct measurement of elevated protease activity has yet to be utilized to detect TBI. In this work, we engineered an activity-based nanosensor for TBI (TBI-ABN) that responds to increased protease activity initiated after brain injury. We establish that a calcium-sensitive protease, calpain-1, is active in the injured brain hours within injury. We then optimize the molecular weight of a nanoscale polymeric carrier to infiltrate into the injured brain tissue with minimal renal filtration. A calpain-1 substrate that generates a fluorescent signal upon cleavage was attached to this nanoscale polymeric carrier to generate an engineered TBI-ABN. When applied intravenously to a mouse model of TBI, our engineered sensor is observed to locally activate in the injured brain tissue. This TBI-ABN is the first demonstration of a sensor that responds to protease activity to detect TBI.


Assuntos
Técnicas Biossensoriais , Lesões Encefálicas Traumáticas/enzimologia , Encéfalo/enzimologia , Calpaína/metabolismo , Animais , Calpaína/química , Feminino , Camundongos Endogâmicos C57BL , Nanopartículas/química , Polímeros/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...