Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Exp Brain Res ; 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38789796

RESUMO

Traumatic brain injury (TBI) mechanism and severity are heterogenous clinically, resulting in a multitude of physical, cognitive, and behavioral deficits. Impact variability influences the origin, spread, and classification of molecular dysfunction which limits strategies for comprehensive clinical intervention. Indeed, there are currently no clinically approved therapeutics for treating the secondary consequences associated with TBI. Thus, examining pathophysiological changes from heterogeneous impacts is imperative for improving clinical translation and evaluating the efficacy of potential therapeutic strategies. Here we utilized TBI models that varied in both injury mechanism and severity including severe traditional controlled cortical impact (CCI), modified mild CCI (MTBI), and multiple severities of closed-head diffuse TBI (DTBI), and assessed pathophysiological changes. Severe CCI induced cortical lesions and necrosis, while both MTBI and DTBI lacked lesions or significant necrotic damage. Autophagy was activated in the ipsilateral cortex following CCI, but acutely impaired in the ipsilateral hippocampus. Additionally, autophagy was activated in the cortex following DTBI, and autophagic impairment was observed in either the cortex or hippocampus following impact from each DTBI severity. Thus, we provide evidence that autophagy is a therapeutic target for both mild and severe TBI. However, dramatic increases in necrosis following CCI may negatively impact the clinical translatability of therapeutics designed to treat acute dysfunction in TBI. Overall, these results provide evidence that injury sequalae affiliated with TBI heterogeneity is linked through autophagy activation and/or impaired autophagic flux. Thus, therapeutic strategies designed to intervene in autophagy may alleviate pathophysiological consequences, in addition to the cognitive and behavioral deficits observed in TBI.

2.
iScience ; 26(6): 106760, 2023 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-37235059

RESUMO

Blood flow is a key regulator of atherosclerosis. Disturbed blood flow promotes atherosclerotic plaque development, whereas normal blood flow protects against plaque development. We hypothesized that normal blood flow is also therapeutic, if it were able to be restored within atherosclerotic arteries. Apolipoprotein E-deficient (ApoE-/-) mice were initially instrumented with a blood flow-modifying cuff to induce plaque development and then five weeks later the cuff was removed to allow restoration of normal blood flow. Plaques in decuffed mice exhibited compositional changes that indicated increased stability compared to plaques in mice with the cuff maintained. The therapeutic benefit of decuffing was comparable to atorvastatin and the combination had an additive effect. In addition, decuffing allowed restoration of lumen area, blood velocity, and wall shear stress to near baseline values, indicating restoration of normal blood flow. Our findings demonstrate that the mechanical effects of normal blood flow on atherosclerotic plaques promote stabilization.

3.
J Neurosci Methods ; 392: 109866, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37116622

RESUMO

BACKGROUND: The Barnes Maze (BM) is a common method of testing cognitive deficits in rodents. Adapting BM protocols for specific neurological disorders could potentially aid in more effective testing, reduce research time, and help decrease variability between studies. NEW METHOD: We tested differences an updated, shortened BM consisting of 6 days, 3 trials per day, only covering the equivalent of the spatial acquisition week BM protocol and a probe trial day consisting of one trial (7 total days). RESULTS: Kaplan-Meier plots of escape percentage as a function of total latency showed a significant difference between control and CCI mice in the updated protocol on days 3 through 6. Additionally, probe trial data showed significant differences in primary latency, primary errors, and returns to goal. COMPARISON WITH EXISTING METHODS: We tested differences between a traditional 5 days per week, 2 trials per day, spatial acquisition and reversal weeks BM protocol (12 total days with probe trials) with an updated 6-day BM protocol (7 total days with probe trial). In the probe trial, the updated protocol control mice showed an over 5-fold decrease in primary latency and primary errors and a 4.6-fold increase in returns to goal compared to the traditional protocol. Additionally, mice in both protocols performed similarly on a trial-by-trial basis suggesting that the changes made for the updated protocol increased learning and memory and was not simply an easier task. CONCLUSION: The updated BM protocol showed an improved ability to distinguish between control and CCI mice and promoted improved and more consistent learning for both the control and CCI groups.


Assuntos
Lesões Encefálicas Traumáticas , Camundongos , Animais , Aprendizagem em Labirinto , Motivação , Roedores , Modelos Animais de Doenças
4.
J Control Release ; 355: 149-159, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36720285

RESUMO

Following traumatic brain injury (TBI), reactive oxygen species (ROS) are released in excess, causing oxidative stress, carbonyl stress, and cell death, which induce the additional release of ROS. The limited accumulation and retention of small molecule antioxidants commonly used in clinical trials likely limit the target engagement and therapeutic effect in reducing secondary injury. Small molecule drugs also need to be administered every several hours to maintain bioavailability in the brain. Therefore, there is a need for a burst and sustained release system with high accumulation and retention in the injured brain. Here, we utilized Pro-NP™ with a size of 200 nm, which was designed to have a burst and sustained release of encapsulated antioxidants, Cu/Zn superoxide dismutase (SOD1) and catalase (CAT), to scavenge ROS for >24 h post-injection. Here, we utilized a controlled cortical impact (CCI) mouse model of TBI and found the accumulation of Pro-NP™ in the brain lesion was highest when injected immediately after injury, with a reduction in the accumulation with delayed administration of 1 h or more post-injury. Pro-NP™ treatment with 9000 U/kg SOD1 and 9800 U/kg CAT gave the highest reduction in ROS in both male and female mice. We found that Pro-NP™ treatment was effective in reducing carbonyl stress and necrosis at 1 d post-injury in the contralateral hemisphere in male mice, which showed a similar trend to untreated female mice. Although we found that male and female mice similarly benefit from Pro-NP™ treatment in reducing ROS levels 4 h post-injury, Pro-NP™ treatment did not significantly affect markers of post-traumatic oxidative stress in female CCI mice as compared to male CCI mice. These findings of protection by Pro-NP™ in male mice did not extend to 7 d post-injury, which suggests subsequent treatments with Pro-NP™ may be needed to afford protection into the chronic phase of injury. Overall, these different treatment effects of Pro-NP™ between male and female mice suggest important sex-based differences in response to antioxidant nanoparticle delivery and that there may exist a maximal benefit from local antioxidant activity in injured brain.


Assuntos
Lesões Encefálicas Traumáticas , Nanopartículas , Camundongos , Masculino , Feminino , Animais , Antioxidantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase-1/farmacologia , Preparações de Ação Retardada/uso terapêutico , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/complicações , Estresse Oxidativo
5.
Adv Ther (Weinh) ; 6(12)2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38464558

RESUMO

Following a traumatic brain injury (TBI), excess reactive oxygen species (ROS) and lipid peroxidation products (LPOx) are generated and lead to secondary injury beyond the primary insult. A major limitation of current treatments is poor target engagement, which has prevented success in clinical trials. Thus, nanoparticle-based treatments have received recent attention because of their ability to increase accumulation and retention in damaged brain. Theranostic neuroprotective copolymers (NPC3) containing thiol functional groups can neutralize ROS and LPOx. Immediate administration of NPC3 following injury in a controlled cortical impact (CCI) mouse model provides a therapeutic window in reducing ROS levels at 2.08-20.83 mg/kg in males and 5.52-27.62 mg/kg in females. This NPC3-mediated reduction in oxidative stress improves spatial learning and memory in males, while females show minimal improvement. Notably, NPC3-mediated reduction in oxidative stress prevents the bilateral spread of necrosis in male mice, which was not observed in female mice and likely accounts for the sex-based spatial learning and memory differences. Overall, these findings suggest sex-based differences to oxidative stress scavenger nanoparticle treatments, and a possible upper threshold of antioxidant activity that provides therapeutic benefit in injured brain since female mice benefit from NPC3 treatment to a lesser extent than male mice.

6.
ACS Appl Mater Interfaces ; 14(49): 54389-54400, 2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36449986

RESUMO

Here, we report the synthesis of robust hybrid iodinated silica-lipid nanoemulsions (HSLNEs) for use as a contrast agent for ultrasound and X-ray applications. We engineered iodinated silica nanoparticles (SNPs), lipid nanoemulsions, and a series of HSLNEs by a low-energy spontaneous nanoemulsification process. The formation of a silica shell requires sonication to hydrolyze and polymerize/condensate the iodomethyltrimethoxysilane at the oil/water interface of the nanoemulsion droplets. The resulting nanoemulsions (NEs) exhibited a homogeneous spherical morphology under transmission electron microscopy. The particles had diameters ranging from 20 to 120 nm with both negative and positive surface charges in the absence and presence of cetyltrimethylammonium bromide (CTAB), respectively. Unlike CTAB-coated nanoformulations, the CTAB-free NEs showed excellent biocompatibility in murine RAW macrophages and human U87-MG cell lines in vitro. The maximum tolerated dose assessment was evaluated to verify their safety profiles in vivo. In vitro X-ray and ultrasound imaging and in vivo computed tomography were used to monitor both iodinated SNPs and HSLNEs, validating their significant contrast-enhancing properties and suggesting their potential as dual-modality clinical agents in the future.


Assuntos
Meios de Contraste , Nanopartículas , Humanos , Camundongos , Animais , Meios de Contraste/farmacologia , Raios X , Dióxido de Silício , Cetrimônio , Ultrassonografia , Lipídeos
7.
Pharmacol Ther ; 238: 108182, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35390422

RESUMO

Triggering receptor expressed on myeloid cells-1 (TREM-1) is a transmembrane protein expressed on endothelial cells, white blood cells, smooth muscle cells and platelets. TREM-1 plays an important role in innate immunity. TREM-1 activation pathways are implicated both in sepsis and in non-infectious inflammatory conditions, including atherosclerosis. TREM-1 enhances the subendothelial lipid accumulation and expression of pro-inflammatory cytokines and matrix-degrading enzymes, thereby promoting inflammation and plaque destabilization. TREM-1 inhibitors attenuate the inflammatory process in the atherosclerotic plaque, leading to plaque stabilization. This review focuses on the role of TREM-1 in the pathophysiology of atherosclerosis and the effects of TREM-1 inhibition in the natural history of the disease.


Assuntos
Aterosclerose , Placa Aterosclerótica , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Citocinas/metabolismo , Células Endoteliais/metabolismo , Humanos , Lipídeos , Placa Aterosclerótica/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo
8.
Biomacromolecules ; 23(4): 1703-1712, 2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35316025

RESUMO

Traumatic brain injury (TBI) results in the generation of reactive oxygen species (ROS) and lipid peroxidation product (LPOx), including acrolein and 4-hydroxynonenal (4HNE). The presence of these biochemical derangements results in neurodegeneration during the secondary phase of the injury. The ability to rapidly neutralize multiple species could significantly improve outcomes for TBI patients. However, the difficulty in creating therapies that target multiple biochemical derangements simultaneously has greatly limited therapeutic efficacy. Therefore, our goal was to design a material that could rapidly bind and neutralize both ROS and LPOx following TBI. To do this, a series of thiol-functionalized biocompatible copolymers based on lipoic acid methacrylate and polyethylene glycol monomethyl ether methacrylate (FW ∼ 950 Da) (O950) were prepared. A polymerizable gadolinium-DOTA methacrylate monomer (Gd-MA) was also synthesized starting from cyclen to facilitate direct magnetic resonance imaging and in vivo tracking of accumulation. These neuroprotective copolymers (NPCs) were shown to rapidly and effectively neutralize both ROS and LPOx. Horseradish peroxidase absorbance assays showed that the NPCs efficiently neutralized H2O2, while R-phycoerythrin protection assays demonstrated their ability to protect the fluorescent protein from oxidative damage. 1H NMR studies indicated that the thiol-functional NPCs rapidly form covalent bonds with acrolein, efficiently removing it from solution. In vitro cell studies with SH-SY5Y-differentiated neurons showed that NPCs provide unique protection against toxic concentrations of both H2O2 and acrolein. NPCs rapidly accumulate and are retained in the injured brain in controlled cortical impact mice and reduce post-traumatic oxidative stress. Therefore, these materials show promise for improved target engagement of multiple biochemical derangements in hopes of improving TBI therapeutic outcomes.


Assuntos
Acroleína , Lesões Encefálicas Traumáticas , Acroleína/farmacologia , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Humanos , Peróxido de Hidrogênio/farmacologia , Peroxidação de Lipídeos/fisiologia , Metacrilatos/farmacologia , Camundongos , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Compostos de Sulfidrila/farmacologia , Nanomedicina Teranóstica
9.
PLoS One ; 16(12): e0260606, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34882722

RESUMO

Atherosclerosis is a lipid-driven chronic inflammatory disease that leads to the formation of plaques in the inner lining of arteries. Plaques form over a range of phenotypes, the most severe of which is vulnerable to rupture and causes most of the clinically significant events. In this study, we evaluated the efficacy of nanoparticles (NPs) to differentiate between two plaque phenotypes based on accumulation kinetics in a mouse model of atherosclerosis. This model uses a perivascular cuff to induce two regions of disturbed wall shear stress (WSS) on the inner lining of the instrumented artery, low (upstream) and multidirectional (downstream), which, in turn, cause the development of an unstable and stable plaque phenotype, respectively. To evaluate the influence of each WSS condition, in addition to the final plaque phenotype, in determining NP uptake, mice were injected with NPs at intermediate and fully developed stages of plaque growth. The kinetics of artery wall uptake were assessed in vivo using dynamic contrast-enhanced magnetic resonance imaging. At the intermediate stage, there was no difference in NP uptake between the two WSS conditions, although both were different from the control arteries. At the fully-developed stage, however, NP uptake was reduced in plaques induced by low WSS, but not multidirectional WSS. Histological evaluation of plaques induced by low WSS revealed a significant inverse correlation between the presence of smooth muscle cells and NP accumulation, particularly at the plaque-lumen interface, which did not exist with other constituents (lipid and collagen) and was not present in plaques induced by multidirectional WSS. These findings demonstrate that NP accumulation can be used to differentiate between unstable and stable murine atherosclerosis, but accumulation kinetics are not directly influenced by the WSS condition. This tool could be used as a diagnostic to evaluate the efficacy of experimental therapeutics for atherosclerosis.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/diagnóstico por imagem , Ácido Fólico/administração & dosagem , Gadolínio/química , Miócitos de Músculo Liso/química , Placa Aterosclerótica/diagnóstico por imagem , Animais , Aterosclerose/genética , Velocidade do Fluxo Sanguíneo , Meios de Contraste/administração & dosagem , Meios de Contraste/química , Meios de Contraste/farmacocinética , Diagnóstico Diferencial , Modelos Animais de Doenças , Feminino , Ácido Fólico/química , Ácido Fólico/farmacocinética , Gadolínio/farmacocinética , Imageamento por Ressonância Magnética , Camundongos , Camundongos Knockout , Nanopartículas , Placa Aterosclerótica/genética , Resistência ao Cisalhamento , Estresse Mecânico
10.
ACS Nano ; 15(11): 18520-18531, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34748307

RESUMO

Aging-induced alterations to the blood-brain barrier (BBB) are increasingly being seen as a primary event in chronic progressive neurological disorders that lead to cognitive decline. With the goal of increasing delivery into the brain in hopes of effectively treating these diseases, a large focus has been placed on developing BBB permeable materials. However, these strategies have suffered from a lack of specificity toward regions of disease progression. Here, we report on the development of a nanoparticle (C1C2-NP) that targets regions of increased claudin-1 expression that reduces BBB integrity. Using dynamic contrast enhanced magnetic resonance imaging, we find that C1C2-NP accumulation and retention is significantly increased in brains from 12 month-old mice as compared to nontargeted NPs and brains from 2 month-old mice. Furthermore, we find C1C2-NP accumulation in brain endothelial cells with high claudin-1 expression, suggesting target-specific binding of the NPs, which was validated through fluorescence imaging, in vitro testing, and biophysical analyses. Our results further suggest a role of claudin-1 in reducing BBB integrity during aging and show altered expression of claudin-1 can be actively targeted with NPs. These findings could help develop strategies for longitudinal monitoring of tight junction protein expression changes during aging as well as be used as a delivery strategy for site-specific delivery of therapeutics at these early stages of disease development.


Assuntos
Barreira Hematoencefálica , Nanopartículas , Animais , Camundongos , Barreira Hematoencefálica/metabolismo , Claudina-1/metabolismo , Claudina-1/farmacologia , Células Endoteliais/metabolismo , Junções Íntimas/metabolismo , Envelhecimento
11.
Biomaterials ; 272: 120766, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33819812

RESUMO

The secondary phase of traumatic brain injury (TBI) is partly caused by the release of excess reactive oxygen species (ROS) from the primary injury. However, there are currently no therapies that have been shown to reduce the secondary spread of injury beyond the primary insult. Nanoparticles offer the ability to rapidly accumulate and be retained in injured brain for improved target engagement. Here, we utilized systemically administered antioxidant thioether core-cross-linked nanoparticles (NP1) that scavenge and inactivate ROS to reduce this secondary spread of injury in a mild controlled cortical impact (CCI) mouse model of TBI. We found that NP1 treatment protected CCI mice from injury induced learning and memory deficits observed in the Morris water maze (MWM) test at 1-month post-CCI. This protection was likely a result of NP1-mediated reduction in oxidative stress in the ipsilateral hemisphere as determined by immunofluorescence imaging of markers of oxidative stress and the spread of neuroinflammation into the contralateral hippocampus as determined by immunofluorescence imaging of activated microglia and neuron-astrocyte-microglia triad formation. These data suggest NP1-mediated reduction in post-traumatic oxidative stress correlates with the reduction in the spread of injury to the contralateral hippocampus to protect spatial memory and learning in CCI mice. Therefore, these materials may offer an improved treatment strategy to reduce the secondary spread of TBI.


Assuntos
Lesões Encefálicas Traumáticas , Nanopartículas , Animais , Antioxidantes , Lesões Encefálicas Traumáticas/tratamento farmacológico , Modelos Animais de Doenças , Aprendizagem em Labirinto , Camundongos , Aprendizagem Espacial , Sulfetos
12.
Adv Funct Mater ; 31(6)2021 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-33708035

RESUMO

Temozolomide (TMZ) is the standard of care chemotherapy drug for treating glioblastomas (GBMs), the most aggressive cancer that affects people of all ages. However, its therapeutic efficacy is limited by the drug resistance mediated by a DNA repair protein, O6-methylguanine-DNA methyltransferase (MGMT), which eliminates the TMZ-induced DNA lesions. Here we report the development of an iron oxide nanoparticle (NP) system for targeted delivery of siRNAs to suppress the TMZ-resistance gene (MGMT). We show that our NP is able to overcome biological barriers, bind specifically to tumor cells, and reduce MGMT expression in tumors of mice bearing orthotopic GBM serially-passaged patient-derived xenografts. The treatment with sequential administration of this NP and TMZ resulted in increased apoptosis of GBM stem-like cells, reduced tumor growth, and significantly-prolonged survival as compared to mice treated with TMZ alone. This study introduces an approach that holds great promise to improve the outcomes of GBM patients.

13.
J Nanotheranostics ; 2(4): 224-268, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35655793

RESUMO

Traumatic brain injury (TBI) is currently the leading cause of injury-related morbidity and mortality worldwide, with an estimated global cost of USD 400 billion annually. Both clinical and preclinical behavioral outcomes associated with TBI are heterogeneous in nature and influenced by the mechanism and frequency of injury. Previous literature has investigated this relationship through the development of animal models and behavioral tasks. However, recent advancements in these methods may provide insight into the translation of therapeutics into a clinical setting. In this review, we characterize various animal models and behavioral tasks to provide guidelines for evaluating the therapeutic efficacy of treatment options in TBI. We provide a brief review into the systems utilized in TBI classification and provide comparisons to the animal models that have been developed. In addition, we discuss the role of behavioral tasks in evaluating outcomes associated with TBI. Our goal is to provide those in the nanotheranostic field a guide for selecting an adequate TBI animal model and behavioral task for assessment of outcomes to increase research in this field.

14.
ACS Omega ; 5(26): 16220-16227, 2020 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-32656444

RESUMO

Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. TBI can have a long-term impact on the quality of life for survivors of all ages. However, there remains no approved treatment that improves outcomes following TBI, which is partially due to poor delivery of therapies into the brain. Therefore, there is a significant unmet need to develop more effective delivery strategies that increase the accumulation and retention of potentially efficacious treatments in the injured brain. Recent work has revealed that nanoparticles (NPs) may offer a promising approach for site-specific delivery; however, a detailed understanding of the specific NP properties that promote brain accumulation and retention are still being developed. Multimodal imaging plays a vital role in the understanding of physicochemical properties that initiate the uptake and accumulation of NPs in the brain at both high spatial (e.g., fluorescence imaging) and temporal (e.g., magnetic resonance imaging, MRI) frequency. However, many NP systems that are currently used in TBI only provide contrast in a single imaging modality limiting the imaging data that can be obtained, and those that offer multimodal imaging capabilities have complicated multistep synthesis methods. Therefore, the goal of this work was to develop an ultrasmall NP with simple fabrication capable of multimodal imaging. Here, we describe the development, characterization, accumulation, and retention of poly(ethylene glycol) (PEG)-coated europium-gadolinium (Eu-Gd) mixed magnetic NPs (MNPs) in a controlled cortical impact mouse model of TBI. We find that these NPs having an ultrasmall core size of 2 nm and a small hydrodynamic size of 13.5 nm can be detected in both fluorescence and MR imaging modalities and rapidly accumulate and are retained in injured brain parenchyma. These NPs should allow for further testing of NP physicochemical properties that promote accumulation and retention in TBI and other disease models.

15.
Sci Rep ; 9(1): 16099, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31695100

RESUMO

Traumatic brain injury (TBI) is a leading cause of injury-related death worldwide, yet there are no approved neuroprotective therapies that improve neurological outcome post-injury. Transient opening of the blood-brain barrier following injury provides an opportunity for passive accumulation of intravenously administered nanoparticles through an enhanced permeation and retention-like effect. However, a thorough understanding of physicochemical properties that promote optimal uptake and retention kinetics in TBI is still needed. In this study, we present a robust method for magnetic resonance imaging of nanoparticle uptake and retention kinetics following intravenous injection in a controlled cortical impact mouse model of TBI. Three contrast-enhancing nanoparticles with different hydrodynamic sizes and relaxivity properties were compared. Accumulation and retention were monitored by modelling the permeability coefficient, Ktrans, for each nanoparticle within the reproducible mouse model. Quantification of Ktrans for different nanoparticles allowed for non-invasive, multi-time point assessment of both accumulation and retention kinetics in the injured tissue. Using this method, we found that 80 nm poly(lactic-co-glycolic acid) nanoparticles had maximal Ktrans in a TBI when injected 3 hours post-injury, showing significantly higher accumulation kinetics than the small molecule, Gd-DTPA. This robust method will enable optimization of administration time and nanoparticle physicochemical properties to achieve maximum delivery.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/metabolismo , Animais , Lesões Encefálicas Traumáticas/diagnóstico por imagem , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/instrumentação , Feminino , Gadolínio DTPA/administração & dosagem , Gadolínio DTPA/química , Gadolínio DTPA/metabolismo , Humanos , Cinética , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Nanopartículas/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/administração & dosagem , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/metabolismo
16.
Pharmaceutics ; 11(9)2019 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-31540234

RESUMO

Traumatic brain injury (TBI) is one of the main causes of disability in children and young adults, as well as a significant concern for elderly individuals. Depending on the severity, TBI can have a long-term impact on the quality of life for survivors of all ages. The primary brain injury can result in severe disability or fatality, and secondary brain damage can increase the complexities in cellular, inflammatory, neurochemical, and metabolic changes in the brain, which can last decades post-injury. Thus, survival from a TBI is often accompanied by lifelong disabilities. Despite the significant morbidity, mortality, and economic loss, there are still no effective treatment options demonstrating an improved outcome in a large multi-center Phase III trial, which can be partially attributed to poor target engagement of delivered therapeutics. Thus, there is a significant unmet need to develop more effective delivery strategies to overcome the biological barriers that would otherwise inhibit transport of materials into the brain to prevent the secondary long-term damage associated with TBI. The complex pathology of TBI involving the blood-brain barrier (BBB) has limited the development of effective therapeutics and diagnostics. Therefore, it is of great importance to develop novel strategies to target the BBB. The leaky BBB caused by a TBI may provide opportunities for therapeutic delivery via nanoparticles (NP). The focus of this review is to provide a survey of NP-based strategies employed in preclinical models of TBI and to provide insights for improved NP based diagnostic or treatment approaches. Both passive and active delivery of various NPs for TBI are discussed. Finally, potential therapeutic targets where improved NP-mediated delivery could increase target engagement are identified with the overall goal of providing insight into open opportunities for NP researchers to begin research in TBI.

17.
ACS Nano ; 11(9): 9514-9524, 2017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28885825

RESUMO

Despite the preponderance of iron oxide nanoparticles (NPs) designed for theranostic applications, widespread clinical translation of these NPs lags behind. A better understanding of how NP pharmacokinetics vary between small and large animal models is needed to rapidly customize NPs for optimal performance in humans. Here we use noninvasive magnetic resonance imaging (MRI) to track iron oxide NPs through a large number of organ systems in vivo to investigate NP biokinetics in both mice and nonhuman primates. We demonstrate that pharmacokinetics are similar between mice and macaques in the blood, liver, spleen, and muscle, but differ in the kidneys, brain, and bone marrow. Our study also demonstrates that full-body MRI is practical, rapid, and cost-effective for tracking NPs noninvasively with high spatiotemporal resolution. Our techniques using a nonhuman primate model may provide a platform for testing a range of NP formulations.


Assuntos
Compostos Férricos/farmacocinética , Imageamento por Ressonância Magnética , Nanopartículas/análise , Animais , Compostos Férricos/administração & dosagem , Compostos Férricos/análise , Compostos Férricos/toxicidade , Macaca , Imageamento por Ressonância Magnética/métodos , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Distribuição Tecidual , Imagem Corporal Total/métodos
18.
ACS Nano ; 11(9): 8600-8611, 2017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28783305

RESUMO

Traumatic brain injury (TBI) is the leading cause of death and disability in children and young adults, yet there are currently no treatments available that prevent the secondary spread of damage beyond the initial insult. The chronic progression of this secondary injury is in part caused by the release of reactive oxygen species (ROS) into surrounding normal brain. Thus, treatments that can enter the brain and reduce the spread of ROS should improve outcome from TBI. Here a highly versatile, reproducible, and scalable method to synthesize core-cross-linked nanoparticles (NPs) from polysorbate 80 (PS80) using a combination of thiol-ene and thiol-Michael chemistry is described. The resultant NPs consist of a ROS-reactive thioether cross-linked core stabilized in aqueous solution by hydroxy-functional oligoethylene oxide segments. These NPs show narrow molecular weight distributions and have a high proportion of thioether units that reduce local levels of ROS. In a controlled cortical impact mouse model of TBI, the NPs are able to rapidly accumulate and be retained in damaged brain as visualized through fluorescence imaging, reduce neuroinflammation and the secondary spread of injury as determined through magnetic resonance imaging and histopathology, and improve functional outcome as determined through behavioral analyses. Our findings provide strong evidence that these NPs may, upon further development and testing, provide a useful strategy to help improve the outcome of patients following a TBI.


Assuntos
Antioxidantes/uso terapêutico , Lesões Encefálicas Traumáticas/terapia , Nanopartículas/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Sulfetos/uso terapêutico , Animais , Antioxidantes/química , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Inflamação/metabolismo , Inflamação/patologia , Inflamação/terapia , Camundongos , Camundongos Endogâmicos C57BL , Nanomedicina/métodos , Nanopartículas/química , Estresse Oxidativo/efeitos dos fármacos , Polissorbatos/química , Polissorbatos/uso terapêutico , Células RAW 264.7 , Sulfetos/química
19.
Nanomedicine ; 13(7): 2131-2139, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28614736

RESUMO

Glioblastoma (GBM) remains incurable, and recurrent tumors rarely respond to standard-of-care radiation and chemo-therapies. Therefore, strategies that enhance the effects of these therapies should provide significant benefits to GBM patients. We have developed a nanoparticle delivery vehicle that can stably bind and protect nucleic acids for specific delivery into brain tumor cells. These nanoparticles can deliver therapeutic siRNAs to sensitize GBM cells to radiotherapy and improve GBM treatment via systemic administration. We show that nanoparticle-mediated knockdown of the DNA repair protein apurinic endonuclease 1 (Ape1) sensitizes GBM cells to radiotherapy and extend survival in a genetic mouse model of GBM. Specific knockdown of Ape1 activity by 30% in brain tumor tissue doubled the extended survival achieved with radiotherapy alone. Ape1 is a promising target for increasing the effectiveness of radiotherapy, and nanoparticle-mediated delivery of siRNA is a promising strategy for tumor specific knockdown of Ape1.


Assuntos
Neoplasias Encefálicas/radioterapia , Reparo do DNA , Portadores de Fármacos/química , Glioblastoma/radioterapia , Nanopartículas/química , RNA Interferente Pequeno/uso terapêutico , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/efeitos da radiação , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Modelos Animais de Doenças , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/terapia , Camundongos , RNA Interferente Pequeno/administração & dosagem , Terapêutica com RNAi/métodos
20.
Pharmacol Res ; 114: 56-66, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27771464

RESUMO

Compared to conventional treatments, gene therapy offers a variety of advantages for cancer treatment including high potency and specificity, low off-target toxicity, and delivery of multiple genes that concurrently target cancer tumorigenesis, recurrence, and drug resistance. In the past decades, gene therapy has undergone remarkable progress, and is now poised to become a first line therapy for cancer. Among various gene delivery systems, nanoparticles have attracted much attention because of their desirable characteristics including low toxicity profiles, well-controlled and high gene delivery efficiency, and multi-functionalities. This review provides an overview on gene therapeutics and gene delivery technologies, and highlight recent advances, challenges and insights into the design and the utility of nanoparticles in gene therapy for cancer treatment.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Nanopartículas/química , Neoplasias/genética , Neoplasias/terapia , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoterapia/métodos , Nanomedicina/métodos , Nanopartículas/administração & dosagem , Nanotecnologia/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA