Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Exp Brain Res ; 242(7): 1645-1658, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38789796

RESUMEN

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.


Asunto(s)
Autofagia , Lesiones Traumáticas del Encéfalo , Modelos Animales de Enfermedad , Animales , Autofagia/fisiología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Masculino , Muerte Celular/fisiología , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Ratas Sprague-Dawley , Ratas , Hipocampo/patología , Hipocampo/fisiopatología
2.
Biomacromolecules ; 23(4): 1703-1712, 2022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-35316025

RESUMEN

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.


Asunto(s)
Acroleína , Lesiones Traumáticas del Encéfalo , Acroleína/farmacología , Animales , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Humanos , Peróxido de Hidrógeno/farmacología , Peroxidación de Lípido/fisiología , Metacrilatos/farmacología , Ratones , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Sulfhidrilo/farmacología , Nanomedicina Teranóstica
3.
Adv Funct Mater ; 31(6)2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-33708035

RESUMEN

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.

4.
Nanomedicine ; 13(7): 2131-2139, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28614736

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Reparación del ADN , Portadores de Fármacos/química , Glioblastoma/radioterapia , Nanopartículas/química , ARN Interferente Pequeño/uso terapéutico , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/efectos de la radiación , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Modelos Animales de Enfermedad , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/terapia , Ratones , ARN Interferente Pequeño/administración & dosificación , Tratamiento con ARN de Interferencia/métodos
5.
Small ; 12(4): 477-87, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26641029

RESUMEN

Hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide. Small interfering RNA (siRNA) holds promise as a new class of therapeutics for HCC, as it can achieve sequence-specific gene knockdown with low cytotoxicity. However, the main challenge in the clinical application of siRNA lies in the lack of effective delivery approaches that need to be highly specific and thus incur low or no systemic toxicity. Here, a nonviral nanoparticle-based gene carrier is presented that can specifically deliver siRNA to HCC. The nanovector (NP-siRNA-GPC3 Ab) is made of an iron oxide core coated with chitosan-polyethylene glycol (PEG) grafted polyethyleneimine copolymer, which is further functionalized with siRNA and conjugated with a monoclonal antibody (Ab) against human glypican-3 (GPC3) receptor highly expressed in HCC. A rat RH7777 HCC cell line that coexpresses human GPC3 and firefly luciferase (Luc) is established to evaluate the nanovector. The nanoparticle-mediated delivery of siRNA against Luc effectively suppresses Luc expression in vitro without notable cytotoxicity. Significantly, NP-siLuc-GPC3 Ab administered intravenously in an orthotopic model of HCC is able to specifically bound to tumor and induce remarkable inhibition of Luc expression. The findings demonstrate the potential of using this nanovector for targeted delivery of therapeutic siRNA to HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Compuestos Férricos/química , Técnicas de Transferencia de Gen , Vectores Genéticos/metabolismo , Neoplasias Hepáticas/metabolismo , Nanopartículas/química , ARN Interferente Pequeño/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Glipicanos/metabolismo , Humanos , Neoplasias Hepáticas/patología , Luciferasas/metabolismo , Ratones , Espectroscopía de Protones por Resonancia Magnética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Pharmacol Res ; 114: 56-66, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27771464

RESUMEN

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.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Nanopartículas/química , Neoplasias/genética , Neoplasias/terapia , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia/métodos , Nanomedicina/métodos , Nanopartículas/administración & dosificación , Nanotecnología/métodos
7.
Biomacromolecules ; 16(10): 3362-72, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26347946

RESUMEN

Cationic nanoparticles (NPs) for targeted gene delivery are conventionally evaluated using 2D in vitro cultures. However, this does not translate well to corresponding in vivo studies because of the marked difference in NP behavior in the presence of the tumor microenvironment. In this study, we investigated whether prostate cancer (PCa) cells cultured in three-dimensional (3D) chitosan-alginate (CA) porous scaffolds could model cationic NP-mediated gene targeted delivery to tumors in vitro. We assessed in vitro tumor cell proliferation, formation of tumor spheroids, and expression of marker genes that promote tumor malignancy in CA scaffolds. The efficacy of NP-targeted gene delivery was evaluated in PCa cells in 2D cultures, PCa tumor spheroids grown in CA scaffolds, and PCa tumors in a mouse TRAMP-C2 flank tumor model. PCa cells cultured in CA scaffolds grew into tumor spheroids and displayed characteristics of higher malignancy as compared to those in 2D cultures. Significantly, targeted gene delivery was only observed in cells cultured in CA scaffolds, whereas cells cultured on 2D plates showed no difference in gene delivery between targeted and nontarget control NPs. In vivo NP evaluation confirmed targeted gene delivery, indicating that only CA scaffolds correctly modeled NP-mediated targeted delivery in vivo. These findings suggest that CA scaffolds serve as a better in vitro platform than 2D cultures for evaluation of NP-mediated targeted gene delivery to PCa.


Asunto(s)
Alginatos/química , Quitosano/química , Terapia Genética , Nanopartículas , Neoplasias de la Próstata/terapia , Animales , Femenino , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Humanos , Técnicas In Vitro , Masculino , Ratones , Porosidad
8.
Mol Pharm ; 11(7): 2134-42, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24779767

RESUMEN

Breast cancer is a major health problem for women worldwide. Although in vitro culture of established breast cancer cell lines is the most widely used model for preclinical assessment, it poorly represents the behavior of breast cancers in vivo. Acceleration of the development of effective therapeutic strategies requires a cost-efficient in vitro model that can more accurately resemble the in vivo tumor microenvironment. Here, we report the use of a thermoreversible poly(ethylene glycol)-g-chitosan hydrogel (PCgel) as an in vitro breast cancer model. We hypothesized that PCgel could provide a tumor microenvironment that promotes cultured cancer cells to a more malignant phenotype with drug and immune resistance. Traditional tissue culture plates and Matrigel were applied as controls in our studies. In vitro cellular proliferation and morphology, the secretion of angiogenesis-related growth factors and cytokines, and drug and immune resistance were assessed. Our results show that PCgel cultures promoted tumor aggregate formation, increased secretion of various angiogenesis- and metastasis-related growth factors and cytokines, and increased tumor cell resistance to chemotherapeutic drugs and immunotherapeutic T cells. This PCgel platform may offer a valuable strategy to bridge the gap between standard in vitro and costly animal studies for a wide variety of experimental designs.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quitosano/administración & dosificación , Hidrogel de Polietilenoglicol-Dimetacrilato/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Polietilenglicoles/administración & dosificación
9.
Biomacromolecules ; 15(7): 2656-62, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24890220

RESUMEN

The outcome for glioblastoma patients remains dismal for its invariably recrudesces within 2 cm of the resection cavity. Local immunotherapy has the potential to eradicate the residual infiltrative component of these tumors. Here, we report the development of a biodegradable hydrogel containing therapeutic T lymphocytes for localized delivery to glioblastoma cells for brain tumor immunotherapy. Thermoreversible poly(ethylene glycol)-g-chitosan hydrogels (PCgels) were optimized for steady T lymphocyte release. Nuclear magnetic resonance spectroscopy confirmed the chemical structure of poly(ethylene glycol)-g-chitosan, and rheological studies revealed that the sol-to-gel transition of the PCgel occurred around ≥32 °C. T lymphocyte invasion through the PCgel and subsequent cytotoxicity to glioblastoma were assessed in vitro. The PCgel was shown to be cellular compatible with T lymphocytes, and the T lymphocytes retain their anti-glioblastoma activity after being encapsulated in the PCgel. T lymphocytes in the PCgel were shown to be more effective in killing glioblastoma than those in the Matrigel control. This may be attributed to the optimal pore size of the PCgel allowing better invasion of T lymphocytes. Our study suggests that this unique PCgel depot may offer a viable approach for localized immunotherapy for glioblastoma.


Asunto(s)
Neoplasias Encefálicas/terapia , Quitosano/análogos & derivados , Quitosano/química , Glioblastoma/terapia , Polietilenglicoles/química , Linfocitos T/fisiología , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Medios de Cultivo , Citotoxicidad Inmunológica , Humanos , Hidrogeles/química , Inmunoterapia Adoptiva , Andamios del Tejido , Temperatura de Transición
10.
Mol Pharm ; 10(11): 4099-106, 2013 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-24099143

RESUMEN

Safety and efficacy are of critical importance to any nanomaterial-based diagnostic and therapy. The innocuity and functionality of a nanomaterial in vivo is largely dependent on the physicochemical properties of the material, particularly its surface coating. Here, we evaluated the influence of polycationic coating on the efficacy, clearance organ uptake, and safety of magnetic nanovectors designed for siRNA delivery. Polyethylene glycol (PEG) coated superparamagnetic iron oxide nanoparticles (NPs) of 12 nm in core diameter were modified with a polycationic coating of either poly-l-arginine (pArg) or polyethylenimine (PEI) and further covalently functionalized with siRNA oligonucleotides. The produced NP-pArg-siRNA and NP-PEI-siRNA nanovectors were similar in hydrodynamic size (21 and 22 nm, respectively) but significantly differed in zeta potentials (+2.1 mV and +29.8 mV, respectively). Fluorescence quantification assays revealed that the NP-pArg-siRNA nanovector was 3-fold more potent than NP-PEI-siRNA in delivering siRNA and 1.8-fold more effective in gene silencing when tested in rat C6 glioblastoma cells. In vivo, both nanovector formulations were similarly taken up by the spleen and liver as determined by histopathological and hemopathological assays. However, PEI coated nanovectors elicited severe hemoincompatibility and damage to the liver and spleen, while pArg coated nanovectors were found to be safe and tolerable. Combined, our findings suggest that polycationic coatings of pArg were more effective and safer than commonly used PEI coatings for preparation of nanovectors. The NP-pArg-siRNA nanovector formulation developed here shows great potential for in vivo based biomedical applications.


Asunto(s)
Nanotecnología/métodos , Péptidos/química , Compuestos Férricos/química , Terapia Genética , Nanopartículas/química , Polietilenglicoles/química , Polietileneimina/química , ARN Interferente Pequeño
11.
Biomacromolecules ; 14(5): 1330-7, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23517456

RESUMEN

In the tumor microenvironment, the signals from tumor-associated fibroblasts (TAF) that suppress antitumor immunity remain unclear. Here, we develop and investigate an in vitro three-dimensional (3D) scaffold model for the novel evaluation of TAF interaction with breast tumor cells and breast specific, neu antigen (p98) reactive T cells. Breast cancer cells seeded on 3D chitosan-alginate (CA) scaffolds showed productive growth and formed distinct tumor spheroids. Antigen specific p98 T cells, but not naïve T cells, bound significantly better to tumor cells on scaffolds. The p98 T cells induced potent tumor cell killing but T helper cell cytokine function was impaired in the presence of TAF coseeding on scaffolds. We found that the immunosuppression was mediated, in part, by transforming growth factor beta (TGF-b) and interleukin-10 (IL-10). Therefore, TAF appear capable of inducing potent T cell suppression. CA scaffolds can provide clinically relevant findings prior to preclinical testing of novel immunotherapies.


Asunto(s)
Antígenos de Neoplasias/inmunología , Carcinoma/inmunología , Fibroblastos/inmunología , Inmunomodulación , Neoplasias Mamarias Animales/inmunología , Andamios del Tejido , Microambiente Tumoral/inmunología , Alginatos/química , Animales , Antígenos de Neoplasias/genética , Carcinoma/genética , Carcinoma/patología , Adhesión Celular , Comunicación Celular , Línea Celular Tumoral , Proliferación Celular , Quitosano/química , Técnicas de Cocultivo , Femenino , Fibroblastos/patología , Expresión Génica , Interleucina-10/inmunología , Interleucina-10/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones , Transducción de Señal , Linfocitos T/citología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
12.
J Neurosci Methods ; 392: 109866, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37116622

RESUMEN

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.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Ratones , Animales , Aprendizaje por Laberinto , Motivación , Roedores , Modelos Animales de Enfermedad
13.
J Control Release ; 355: 149-159, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36720285

RESUMEN

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.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Nanopartículas , Ratones , Masculino , Femenino , Animales , Antioxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa-1/farmacología , Preparaciones de Acción Retardada/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Estrés Oxidativo
14.
Adv Ther (Weinh) ; 6(12)2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38464558

RESUMEN

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.

15.
iScience ; 26(6): 106760, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37235059

RESUMEN

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.

16.
Acc Chem Res ; 44(10): 853-62, 2011 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-21528865

RESUMEN

Nanotechnology provides a flexible platform for the development of effective therapeutic nanomaterials that can interact specifically with a target in a biological system and provoke a desired response. Of the nanomaterials studied, iron oxide nanoparticles have emerged as one of top candidates for cancer therapy. Their intrinsic superparamagnetism enables noninvasive magnetic resonance imaging (MRI), and their biodegradability is advantageous for in vivo applications. A therapeutic superparamagnetic iron oxide nanoparticle (SPION) typically consists of three primary components: an iron oxide nanoparticle core that serves as both a carrier for therapeutics and contrast agent for MRI, a coating on the iron oxide nanoparticle that promotes favorable interactions between the SPION and the biological system, and a therapeutic payload that performs the designated function in vivo. Often, the design may include a targeting ligand that recognizes the receptors over-expressed on the exterior surface of cancer cells. The body is a highly complex system that imposes multiple physiological and cellular barriers to foreign objects. Thus, the success of a therapeutic SPION largely relies on the design of the iron oxide core to ensure its detection in MRI and the coatings that allow the nanoparticles to bypass these barriers. Strategies to bypass the physiological barriers, such as liver, kidneys, and spleen, involve tuning the overall size and surface chemistry of the SPION to maximize blood half-life and facilitate the navigation in the body. Strategies to bypass cellular barriers include the use of targeting agents to maximize uptake of the SPION by cancer cells and the employment of materials that promote desired intracellular trafficking and enable controlled drug release. The payload can be genes, proteins, chemotherapy drugs, or a combination of these molecules. Each type of therapeutic molecule requires a specific coating design to maximize the loading and to achieve effective delivery and release. In this Account, we discuss the primary design parameters in developing therapeutic SPIONs with a focus on surface coating design to overcome the barriers imposed by the body's defense system. We provide examples of how these design parameters have been implemented to produce SPIONs for specific therapeutic applications. Although there are still challenges to be addressed, SPIONs show great promise in the successful diagnosis and treatment of the most devastating cancers. Once the critical design parameters have been optimized, these nanoparticles, combined with imaging modalities, can serve as truly multifunctional theranostic agents that not only perform a therapeutic function but also provide instant clinical feedback, allowing the physician to adjust the treatment plan.


Asunto(s)
Compuestos Férricos/química , Compuestos Férricos/síntesis química , Terapia Molecular Dirigida/métodos , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias/terapia , Animales , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Propiedades de Superficie
17.
Pharmacol Ther ; 238: 108182, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35390422

RESUMEN

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.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Citocinas/metabolismo , Células Endoteliales/metabolismo , Humanos , Lípidos , Placa Aterosclerótica/metabolismo , Receptor Activador Expresado en Células Mieloides 1/metabolismo
18.
ACS Appl Mater Interfaces ; 14(49): 54389-54400, 2022 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-36449986

RESUMEN

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.


Asunto(s)
Medios de Contraste , Nanopartículas , Humanos , Ratones , Animales , Medios de Contraste/farmacología , Rayos X , Dióxido de Silicio , Cetrimonio , Ultrasonografía , Lípidos
19.
Mol Imaging ; 10(1): 69-77, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21303616

RESUMEN

Imaging is essential in accurately detecting, staging, and treating primary liver cancer (hepatocellular carcinoma [HCC]), one of the most prevalent and lethal malignancies. We developed a novel multifunctional nanoparticle (NP) specifically targeting glypican-3 (GPC3), a proteoglycan implicated in promotion of cell growth that is overexpressed in most HCCs. Quantitative real-time polymerase chain reaction was performed to confirm the differential GPC3 expression in two human HCC cells, Hep G2 (high) and HLF (negligible). These cells were treated with biotin-conjugated GPC3 monoclonal antibody (αGPC3) and subsequently targeted using superparamagnetic iron oxide NPs conjugated to streptavidin and Alexa Fluor 647. Flow cytometry demonstrated that only GPC3-expressing Hep G2 cells were specifically targeted using this αGPC3-NP conjugate (fourfold mean fluorescence over nontargeted NP), and magnetic resonance imaging (MRI) experiments showed similar findings (threefold R2 relaxivity). Confocal fluorescence microscopy localized the αGPC3 NPs only to the cell surface of GPC3-expressing Hep G2 cells. Further characterization of this construct demonstrated a negatively charged, monodisperse, 50 nm NP, ideally suited for tumor targeting. This GPC3-specific NP system, with dual-modality imaging capability, may enhance pretreatment MRI, enable refined intraoperative HCC visualization by near-infrared fluorescence, and be potentially used as a carrier for delivery of tumor-targeted therapies, improving patient outcomes.


Asunto(s)
Glipicanos/metabolismo , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/metabolismo , Nanopartículas/química , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , AMP Cíclico/análogos & derivados , AMP Cíclico/química , Citometría de Flujo , Glipicanos/inmunología , Células Hep G2 , Humanos , Imagen por Resonancia Magnética , Nanotecnología , Estreptavidina/química
20.
Mater Today (Kidlington) ; 14(7-8): 330-338, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22389583

RESUMEN

Nanotechnology has given scientists new tools for the development of advanced materials for the detection and diagnosis of disease. Iron oxide nanoparticles (SPIONs) in particular have been extensively investigated as novel magnetic resonance imaging (MRI) contrast agents due to a combination of favorable superparamagnetic properties, biodegradability, and surface properties of easy modification for improved in vivo kinetics and multifunctionality. This review discusses the basics of MR imaging, the origin of SPION's unique magnetic properties, recent developments in MRI acquisition methods for detection of SPIONs, synthesis and post-synthesis processes that improve SPION's imaging characteristics, and an outlook on the translational potential of SPIONs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA