Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Allergy Clin Immunol ; 151(2): 509-525.e8, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36494212

RESUMEN

BACKGROUND: The human upper respiratory tract is the first site of contact for inhaled respiratory viruses and elaborates an array of innate immune responses. Seasonal variation in respiratory viral infections and the importance of ambient temperature in modulating immune responses to infections have been well recognized; however, the underlying biological mechanisms remain understudied. OBJECTIVE: We investigated the role of nasal epithelium-derived extracellular vesicles (EVs) in innate Toll-like receptor 3 (TLR3)-dependent antiviral immunity. METHODS: We evaluated the secretion and composition of nasal epithelial EVs after TLR3 stimulation in human autologous cells and fresh human nasal mucosal surgical specimens. We also explored the antiviral activity and mechanisms of TLR3-stimulated EVs against respiratory viruses as well as the effect of cool ambient temperature on TLR3-dependent antiviral immunity. RESULTS: We found that polyinosinic:polycytidylic acid, aka poly(I:C), exposure induced a swarm-like increase in the secretion of nasal epithelial EVs via the TLR3 signaling. EVs participated in TLR3-dependent antiviral immunity, protecting the host from viral infections through both EV-mediated functional delivery of miR-17 and direct virion neutralization after binding to virus ligands via surface receptors, including LDLR and ICAM-1. These potent antiviral immune defense functions mediated by TLR3-stimulated EVs were impaired by cold exposure via a decrease in total EV secretion as well as diminished microRNA packaging and antiviral binding affinity of individual EV. CONCLUSION: TLR3-dependent nasal epithelial EVs exhibit multiple innate antiviral mechanisms to suppress respiratory viral infections. Furthermore, our study provides a direct quantitative mechanistic explanation for seasonal variation in upper respiratory tract infection prevalence.


Asunto(s)
Vesículas Extracelulares , Virosis , Humanos , Receptor Toll-Like 3 , Inmunidad Innata , Antivirales/farmacología , Poli I-C
2.
Mol Pharm ; 20(3): 1729-1736, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36744718

RESUMEN

Verapamil is a calcium channel blocker that holds promise for the therapy of chronic rhinosinusitis (CRS) with and without nasal polyps. The verapamil-induced side effects limit its tolerated dose via the oral route, underscoring the usefulness of localized intranasal administration. However, the challenge to intranasal administration is mucociliary clearance, which diminishes localized dose availability. To overcome this challenge, verapamil was loaded into a mucoadhesive cationic poly(ethylene glycol)-modified (PEGylated) liposomal carrier. Organotypic nasal explants were exposed to verapamil liposomes under flow conditions to mimic mucociliary clearance. The liposomes resulted in significantly higher tissue residence compared with the free verapamil control. These findings were further confirmed in vivo in C57BL/6 mice following intranasal administration. Liposomes significantly increased the accumulation of verapamil in nasal tissues compared with the control group. The developed tissue-retentive verapamil liposomal formulation is considered a promising intranasal delivery system for CRS therapy.


Asunto(s)
Liposomas , Sinusitis , Animales , Ratones , Liposomas/uso terapéutico , Verapamilo , Polietilenglicoles/uso terapéutico , Ratones Endogámicos C57BL , Administración Intranasal , Sinusitis/tratamiento farmacológico , Administración Tópica
3.
Pharm Res ; 40(1): 77-105, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36380168

RESUMEN

Nucleic acid-based therapeutic molecules including small interfering RNA (siRNA), microRNA(miRNA), antisense oligonucleotides (ASOs), messenger RNA (mRNA), and DNA-based gene therapy have tremendous potential for treating diseases in the central nervous system (CNS). However, achieving clinically meaningful delivery to the brain and particularly to target cells and sub-cellular compartments is typically very challenging. Mediating cell-specific delivery in the CNS would be a crucial advance that mitigates off-target effects and toxicities. In this review, we describe these challenges and provide contemporary evidence of advances in cellular and sub-cellular delivery using a variety of delivery mechanisms and alternative routes of administration, including the nose-to-brain approach. Strategies to achieve subcellular localization, endosomal escape, cytosolic bioavailability, and nuclear transfer are also discussed. Ultimately, there are still many challenges to translating these experimental strategies into effective and clinically viable approaches for treating patients.


Asunto(s)
Sistemas de Liberación de Medicamentos , MicroARNs , Ácidos Nucleicos , ARN Interferente Pequeño , Humanos , Barrera Hematoencefálica , Encéfalo , MicroARNs/uso terapéutico , Ácidos Nucleicos/uso terapéutico , Oligonucleótidos Antisentido/uso terapéutico , ARN Interferente Pequeño/uso terapéutico
4.
J Allergy Clin Immunol ; 150(4): 872-881, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35660375

RESUMEN

BACKGROUND: Cystatin SN (CST1) and cystatin SA (CST2) are cysteine protease inhibitors that protect against allergen, viral, and bacterial proteases. Cystatins are overexpressed in the setting of allergic rhinitis and chronic rhinosinusitis with nasal polyps (CRSwNP); however, their role in promoting type 2 inflammation remains poorly characterized. OBJECTIVE: The purpose of this study was to use integrated poly-omics and a murine exposure model to explore the link between cystatin overexpression in CRSwNP and type 2 inflammation. METHODS: In this institutional review board- and institutional animal care and use committee-approved study, we compared tissue, exosome, and mucus CST1 and CST2 between CRSwNP and controls (n = 10 per group) by using matched whole exome sequencing, transcriptomic, proteomic, posttranslational modification, histologic, functional, and bioinformatic analyses. C57/BL6 mice were dosed with 3.9 µg/mL of CST1 or PBS intranasally for 5 to 18 days in the presence or absence of epithelial ABCB1a knockdown. Inflammatory cytokines were quantified by using Quansys multiplex assays or ELISAs. RESULTS: Of the 1305 proteins quantified, CST1 and CST2 were among the most overexpressed protease inhibitors in tissue, exosome, and mucus samples; they were localized to the epithelial layer. Multiple posttranslational modifications were identified in the polyp tissue. Exosomal CST1 and CST2 were strongly and significantly correlated with eosinophils and Lund-Mackay scores. Murine type 2 cytokine secretion and TH2 cell infiltration increased in a time-dependent manner following CST1 exposure and was abrogated by epithelial knockdown of ABCB1a, a regulator of epithelial cytokine secretion. CONCLUSION: CST1 is a potent upstream initiator of epithelial-derived type 2 inflammation in CRSwNP. Therapeutic strategies targeting CST activity and its associated posttranslational modifications deserve further interrogation.


Asunto(s)
Pólipos Nasales , Rinitis , Cistatinas Salivales , Sinusitis , Alérgenos , Animales , Enfermedad Crónica , Inhibidores de Cisteína Proteinasa , Citocinas , Inflamación , Ratones , Pólipos Nasales/patología , Péptido Hidrolasas , Proteómica , Rinitis/metabolismo , Cistatinas Salivales/genética , Cistatinas Salivales/metabolismo , Sinusitis/patología
5.
Mol Pharm ; 19(11): 4067-4079, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36226722

RESUMEN

Extracellular vesicle (EV)-mediated microRNA transfer and propagation from the donor cell to the recipient cell in the tumor microenvironment have significant implications, including the development of multidrug resistance (MDR). Although miRNA-encapsulated EV have been shown to have functional effects on recipient cells, the quantitative aspects of transfer kinetics and functional effects remain poorly understood. Intracellular events such as degradation of miRNA, loading of miRNA into EVs, cellular release of EVs, and their uptake by recipient cells govern the transfer and functional effect of encapsulated miRNA. Based on these rate-limiting steps, we developed a mathematical model using ordinary differential equations (model 1). We performed coculture experiments using ID8-VEGF ovarian cancer cells to demonstrate EV-mediated propagation of tumor suppressor miRNA Let7b administered with hyaluronic acid-poly(ethyleneimine) (HA-PEI) nanoparticles. Using the experimental data and model fitting, we determined the rate constants for the kinetic events involved in the transfer from the donor cells to the recipient cells. In model 2, we performed Let7b transfection experiments in ID8-VEGF cells with HA-PEI nanoparticles to determine the concentration-effect relationship on HMGA2 mRNA levels. Lastly, in model 3, we combined model 1 and model 2 parameters to describe the kinetics and effect relationship of EV-Let7b in recipient cells to predict the minimum number of miRNA copies needed to show functional effects.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Neoplasias Ováricas , Humanos , Femenino , MicroARNs/genética , MicroARNs/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vesículas Extracelulares/metabolismo , Neoplasias Ováricas/metabolismo , Modelos Teóricos , Microambiente Tumoral
6.
Mol Pharm ; 19(12): 4411-4427, 2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36154128

RESUMEN

Increased life expectancy has led to a rise in age-related disorders including neurological diseases such as Alzheimer's disease and Parkinson's disease. Limited progress has been made in the development of clinically translatable therapies for these central nervous system (CNS) diseases. Challenges including the blood-brain barrier, brain complexity, and comorbidities in the elderly population are some of the contributing factors toward lower success rates. Various invasive and noninvasive ways are being employed to deliver small and large molecules across the brain. Biodegradable, implantable drug-delivery systems have gained lot of interest due to advantages such as sustained and targeted delivery, lower side effects, and higher patient compliance. 3D printing is a novel additive manufacturing technique where various materials and printing techniques can be used to fabricate implants with the desired complexity in terms of mechanical properties, shapes, or release profiles. This review discusses an overview of various types of 3D-printing techniques and illustrative examples of the existing literature on 3D-printed systems for CNS drug delivery. Currently, there are various technical and regulatory impediments that need to be addressed for successful translation from the bench to the clinical stage. Overall, 3D printing is a transformative technology with great potential in advancing customizable drug treatment in a high-throughput manner.


Asunto(s)
Implantes Absorbibles , Sistemas de Liberación de Medicamentos , Anciano , Humanos , Sistemas de Liberación de Medicamentos/métodos , Impresión Tridimensional , Medicina de Precisión , Fármacos del Sistema Nervioso Central
7.
Mol Pharm ; 18(8): 3099-3107, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34228470

RESUMEN

Celiac disease is a chronic inflammatory condition characterized by activation of the immune system in response to deamidation of gluten peptides brought about by tissue transglutaminase-2 (TG2). Overexpression of interleukin-15 (IL-15) in the intestinal epithelium and the lamina propria leads to the dysregulation of the immune system, leading to epithelial damage. The goal of this study was to develop an RNA interference therapeutic strategy for celiac disease using a combination of TG2 and IL-15 gene silencing in the inflamed intestine. TG2 and IL-15 silencing siRNA sequences, along with scrambled control, were encapsulated in a nanoparticle-in-microsphere oral system (NiMOS) and administered in a poly(I:C) mouse model of celiac disease. Single TG2 and IL-15 siRNA therapy and the combination showed effective gene silencing in vivo. Additionally, it was found that IL-15 gene silencing alone and combination in the NiMOS significantly reduced other proinflammatory cytokines. The tissue histopathology data also confirmed a reduction in immune cell infiltration and restoration of the mucosal architecture and barrier function in the intestine upon treatment. Overall, the results of this study show evidence that celiac disease can be potentially treated with an oral microsphere formulation using a combination of TG2 and IL-15 RNA interference therapeutic strategies.


Asunto(s)
Enfermedad Celíaca/tratamiento farmacológico , Enfermedad Celíaca/genética , Gastroenteritis/tratamiento farmacológico , Gastroenteritis/genética , Interleucina-15/genética , Microesferas , Sistema de Administración de Fármacos con Nanopartículas/química , Nanopartículas/química , Proteína Glutamina Gamma Glutamiltransferasa 2/genética , Interferencia de ARN , Administración Oral , Animales , Enfermedad Celíaca/inducido químicamente , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Gastroenteritis/inducido químicamente , Interleucina-15/administración & dosificación , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Poli I-C/efectos adversos , Proteína Glutamina Gamma Glutamiltransferasa 2/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Resultado del Tratamiento
8.
Mol Pharm ; 17(6): 1996-2005, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32365295

RESUMEN

Nucleic acid-based therapeutics, including the use of messenger RNA (mRNA) as a drug molecule, has tremendous potential in the treatment of chronic diseases, such as age-related neurodegenerative diseases. In this study, we have developed a cationic liposomal formulation of mRNA and evaluated the potential of intranasal delivery to the brain in murine model. Preliminary in vitro studies in J774A.1 murine macrophages showed GFP expression up to 24 h and stably expressed GFP protein in the cytosol. Upon intranasal administration of GFP-mRNA/cationic liposomes (3 mg/kg dose) in mice, there was significantly higher GFP-mRNA expression in the brain post 24 h as compared to either naked mRNA or the vehicle-treated group. Luciferase mRNA encapsulated in cationic liposomes was used for quantification of mRNA expression distribution in the brain. The results showed increased luciferase activity in the whole brain in a dose-dependent manner. Specifically, the luciferase-mRNA/cationic liposome group (3 mg/kg dose) showed significantly higher luciferase activity in the cortex, striatum, and midbrain regions as compared with the control groups, with minimal systemic exposure. Overall, the results of this study demonstrate the feasibility of brain-specific, nonviral mRNA delivery for the treatment of various neurological disorders.


Asunto(s)
Encéfalo/metabolismo , Cationes/química , ARN Mensajero/administración & dosificación , ARN Mensajero/metabolismo , Administración Intranasal , Animales , Línea Celular , Sistemas de Liberación de Medicamentos , Liposomas/química , Masculino , Ratones
9.
J Allergy Clin Immunol ; 143(4): 1525-1535.e1, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30442371

RESUMEN

BACKGROUND: Nasal mucosa-derived exosomes (NMDEs) harbor immunodefensive proteins and are capable of rapid interepithelial protein transfer. OBJECTIVES: We sought to determine whether mucosal exposure to inhaled pathogens stimulates a defensive swarm of microbiocidal exosomes, which also donate their antimicrobial cargo to adjacent epithelial cells. METHODS: We performed an institutional review board-approved study of healthy NMDE secretion after Toll-like receptor (TLR) 4 stimulation by LPS (12.5 µg/mL) in the presence of TLR4 inhibitors. Interepithelial transfer of exosomal nitric oxide (NO) synthase and nitric oxide was measured by using ELISAs and NO activity assays. Exosomal antimicrobial assays were performed with Pseudomonas aeruginosa. Proteomic analyses were performed by using SOMAscan. RESULTS: In vivo and in vitro LPS exposure induced a 2-fold increase in NMDE secretion along with a 2-fold increase in exosomal inducible nitric oxide synthase expression and function through TLR4 and inhibitor of nuclear factor κB kinase activation. LPS stimulation increased exosomal microbiocidal activity against P aeruginosa by almost 2 orders of magnitude. LPS-stimulated exosomes induced a 4-fold increase in NO production within autologous epithelial cells with protein transfer within 5 minutes of contact. Pathway analysis of the NMDE proteome revealed 44 additional proteins associated with NO signaling and innate immune function. CONCLUSIONS: We provide direct in vivo evidence for a novel exosome-mediated innate immunosurveillance and defense mechanism of the human upper airway. These findings have implications for lower airway innate immunity, delivery of airway therapeutics, and host microbiome regulation.


Asunto(s)
Exosomas/inmunología , Inmunidad Innata/inmunología , Mucosa Nasal/inmunología , Humanos , Mucosa Nasal/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Infecciones por Pseudomonas/inmunología
10.
Small ; 15(36): e1902232, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31328877

RESUMEN

Chronic wounds are characterized by impaired healing and uncontrolled inflammation, which compromise the protective role of the immune system and may lead to bacterial infection. Upregulation of miR-223 microRNAs (miRNAs) shows driving of the polarization of macrophages toward the anti-inflammatory (M2) phenotype, which could aid in the acceleration of wound healing. However, local-targeted delivery of microRNAs is still challenging, due to their low stability. Here, adhesive hydrogels containing miR-223 5p mimic (miR-223*) loaded hyaluronic acid nanoparticles are developed to control tissue macrophages polarization during wound healing processes. In vitro upregulation of miR-223* in J774A.1 macrophages demonstrates increased expression of the anti-inflammatory gene Arg-1 and a decrease in proinflammatory markers, including TNF-α, IL-1ß, and IL-6. The therapeutic potential of miR-223* loaded adhesive hydrogels is also evaluated in vivo. The adhesive hydrogels could adhere to and cover the wounds during the healing process in an acute excisional wound model. Histological evaluation and quantitative polymerase chain reaction (qPCR) analysis show that local delivery of miR-223* efficiently promotes the formation of uniform vascularized skin at the wound site, which is mainly due to the polarization of macrophages to the M2 phenotype. Overall, this study demonstrates the potential of nanoparticle-laden hydrogels conveying miRNA-223* to accelerate wound healing.


Asunto(s)
Hidrogeles/química , Inmunomodulación/fisiología , MicroARNs/química , Nanopartículas/química , Cicatrización de Heridas/fisiología , Animales , Línea Celular , Ácido Hialurónico/química , Macrófagos/metabolismo , Macrófagos/ultraestructura , Espectroscopía de Resonancia Magnética , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Microscopía Electrónica de Rastreo , Cicatrización de Heridas/genética
11.
Mol Pharm ; 16(1): 128-140, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30525660

RESUMEN

Polymeric systems have been extensively studied as polyelectrolyte complexes to enhance the cellular delivery and transfection efficiency of genetic materials, such as plasmid DNA (pDNA). Here, self-assembled nanoparticles were formulated by complexation of hyaluronic acid (HA)-conjugated poly(ethylene glycol) (HA-PEG) and poly(ethylenimine) (HA-PEI), respectively, with pDNA creating relatively small, stable, and multifunctional nanoparticle complex formulations with high transfection efficiency. This formulation strategy offers high gene expression efficiency and negligible cytotoxicity in HeLa and A549 human lung cancer cell lines. To develop the ideal formulation, in vitro transfection efficiency was studied for three different nanoparticle formulations (HA-PEI/HA-PEG, HA-PEI, and HA-PEG) with different concentrations. The combination of the three polymers (HA, PEG, and PEI) was significant for the formulation to achieve the maximum gene expression results. The nanoparticles were found to be stable for up to a week at 4 °C conditions. Overall, these HA-based nanoparticles showed promising aspects that can be utilized in the designing of gene delivery vectors for cancer therapy.


Asunto(s)
Ácido Hialurónico/química , Nanopartículas/química , Plásmidos/genética , Transfección/métodos , Células A549 , Sulfato de Dextran/química , Células HeLa , Humanos , Polietilenglicoles/química , Polietileneimina/análogos & derivados , Polietileneimina/química
12.
Pharm Res ; 36(11): 153, 2019 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-31482243

RESUMEN

The purpose of this review is to discuss the challenges associated with the development of nanoparticle-based quality drug products in adhering to the principles of quality by design (QbD) and defining appropriate quality parameters towards successful product development. With the advent of nanotechnology into the pharmaceutical field, the novel field of nanomedicine was born. Due to their unique properties in terms of size, conformation and targeted delivery, nanomedicines are able to overcome many drawbacks of conventional medicine. As nano-sized formulations have made their way into more and more therapies, it has became clear that these very unique properties create hurdles for nanomedicines in successfully traversing the regulatory pathways and there is a need to develop nanomedicines in a more controlled and consistent fashion. The elements of a QbD methodology explained in this review enable the development of nano-based formulations in a way that maximizes the possibility of success. The identification of critical quality attributes (CQA) of the drug product and its intermediates are discussed in detail with a focus on nanomaterial-based formulations. In conclusion, QbD and the identification and specification of CQAs at its core are critical to the design, development and growth of nanomaterials in pharmaceuticals.


Asunto(s)
Desarrollo de Medicamentos/métodos , Nanocápsulas/química , Nanotecnología/métodos , Animales , Preparaciones de Acción Retardada/química , Composición de Medicamentos , Estabilidad de Medicamentos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Nanomedicina , Resultado del Tratamiento
13.
Nanomedicine ; 22: 102091, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31626992

RESUMEN

Iron is a nutrient metal, but excess iron promotes tissue damage. Since iron chelation therapies exhibit multiple off-target toxicities, there is a substantial demand for more specific approaches to decrease iron burden in iron overload. While the divalent metal transporter 1 (DMT1) plays a well-established role in the absorption of dietary iron, up-regulation of intestinal DMT1 is associated with iron overload in both humans and rodents. Hence, we developed a novel pH-sensitive multi-compartmental particulate (MCP) oral delivery system that encapsulates DMT1 siRNA and validated its efficacy in mice. Using the gelatin NPs coated with Eudragit® L100-55, we demonstrated that DMT1 siRNA-loaded MCPs down-regulated DMT1 mRNA levels in the duodenum, which was consistent with decreased intestinal absorption of orally-administered 59Fe. Together, the Eudragit® L100-55-based oral siRNA delivery system could provide an effective strategy to specifically down-regulate duodenal DMT1 and mitigate iron absorption.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Sistemas de Liberación de Medicamentos , Silenciador del Gen , Absorción Intestinal , Intestinos/fisiología , Hierro/metabolismo , Nanopartículas/administración & dosificación , Resinas Acrílicas/química , Administración Oral , Animales , Células CACO-2 , Gelatina/química , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Hierro/administración & dosificación , Masculino , Ratones , Nanopartículas/ultraestructura , Tamaño de la Partícula , ARN Interferente Pequeño/metabolismo
14.
Nanomedicine ; 15(1): 274-284, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30343013

RESUMEN

A woven nanotextile implant was developed and optimized for long-term continuous drug delivery for potential oncological applications. Electrospun polydioxanone (PDS) nanoyarns, which are twisted bundles of PDS nanofibres, were loaded with paclitaxel (PTX) and woven into nanotextiles of different packing densities. A mechanistic modeling of in vitro drug release proved that a combination of diffusion and matrix degradation controlled the slow PTX-release from a nanoyarn, emphasizing the role of nanostructure in modulating release kinetics. Woven nanotextiles, through variations in its packing density and thereby architecture, demonstrated tuneable PTX-release. In vivo PTX-release, pharmacokinetics and biodistribution were evaluated in healthy BALB/c mice by suturing the nanotextile to peritoneal wall. The slow and metronomic PTX-release for 60 days from the loosely woven implant was extremely effective in enhancing its residence in peritoneum, in contrast to intraperitoneal injections. Such an implantable matrix offers a novel platform for therapy of solid tumors over prolonged durations.


Asunto(s)
Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Nanoestructuras/química , Paclitaxel/farmacocinética , Peritoneo/metabolismo , Textiles , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Proliferación Celular , Células Cultivadas , Implantes de Medicamentos , Liberación de Fármacos , Ratones , Ratones Endogámicos BALB C , Nanoestructuras/administración & dosificación , Paclitaxel/administración & dosificación , Polímeros/química , Distribución Tisular
15.
Nano Lett ; 18(6): 3571-3579, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29722542

RESUMEN

Tumor-associated macrophages (TAMs) acquire a pro-tumor (M2) phenotype, which promotes tumor growth, angiogenesis, and metastasis. Certain microRNAs (miRs), such as miR-125b, can reprogram TAMs into an antitumor/pro-inflammatory (M1) phenotype. Using CD44 targeting hyaluronic acid-poly(ethylenimine) (HA-PEI)-based nanoparticles encapsulating miR-125b, we have herein shown macrophage-specific delivery and transfection upon intraperitoneal (i.p.) administration. We have exploited the inherent ability of peritoneal macrophages to migrate toward the inflammation/injury and demonstrated that following intraperitoneal administration of HA-PEI nanoparticles, there is an accumulation of HA-PEI nanoparticles in the macrophage-ablated lung tissues of both naïve and KRAS/p53 double mutant genetically engineered (KP-GEM) nonsmall cell lung cancer (NSCLC) mouse model. Additionally, upon transfection with miR-125b, we observed a >6-fold increase in the M1 to M2 macrophage ratio and 300-fold increase in the iNOS (M1 marker)/Arg-1 (M2 marker) ratio in TAMs as compared to the untreated control group. The results of these studies show that i.p. administered macrophage-specific HA-PEI nanoparticles can successfully transfect TAMs in lung tissues of both naïve mice and a KP-GEM NSCLC mouse model. Successful TAM repolarization toward the M1 phenotype has significant implication in anticancer immunotherapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Macrófagos Peritoneales/patología , MicroARNs/administración & dosificación , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Polaridad Celular , Modelos Animales de Enfermedad , Ingeniería Genética , Humanos , Ácido Hialurónico/administración & dosificación , Neoplasias Pulmonares/genética , Macrófagos Peritoneales/metabolismo , Ratones , MicroARNs/genética , Nanopartículas/administración & dosificación , Transfección
16.
Mol Pharm ; 14(9): 3036-3044, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28737923

RESUMEN

In this study, we have developed a type B gelatin nanoparticle based siRNA delivery system for silencing of intestinal transglutaminase-2 (TG2) and interleukin-15 (IL-15) genes in cultured human intestinal epithelial cells (Caco-2) and murine alveolar macrophage cells (J774A.1). Small interfering RNA (siRNA) targeting the TG2 or IL-15 gene was encapsulated within gelatin nanoparticles using ethanol-water solvent displacement method. Size, charge, and morphology of gelatin nanoparticles were evaluated using a Zetasizer instrument and transmission electron microscopy. siRNA encapsulation efficiency was determined using an siRNA specific stem-loop quantitative polymerase chain reaction (qPCR) assay. Cellular uptake of siRNA-containing gelatin nanoparticles was determined using fluorescent microscopy and stem-loop qPCR assay. siRNA loading in the RISC (RNA-induced silencing complex) was determined by immunoprecipitation of argonaute 2 (AGO2) protein followed by stem-loop qPCR for siRNA quantification. Gene expression analysis of TG2, IL-15, and the proinflammatory cytokines, tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ), was performed using qPCR assays. Efficacy of silencing TG2 and IL-15 knockdown was evaluated in an in vitro model of celiac disease by utilizing immunogenic α-gliadin peptide p31-43 in cultured J774A.1 cells. siRNA-containing gelatin nanoparticles were spherical in shape with mean particle size and charge of 217 ± 8.39 nm and -6.2 ± 0.95 mV, respectively. siRNA loading efficiency within gelatin nanoparticles was found to be 89.3 ± 3.05%. Evaluations of cellular uptake using fluorescent microscopy showed rapid internalization of gelatin nanoparticles within 2 h of dosing, with cytosolic localization of delivered siRNA in Caco-2 cells. Gelatin nanoparticles showed greater intracellular siRNA exposure with a longer half-life, when compared to Lipofectamine-mediated siRNA delivery. Approximately 0.1% of total intracellular siRNA was associated in the RISC complex. A maximum knockdown of 60% was observed at 72 h post siRNA treatment for both TG2 and IL-15 genes, which corresponded to ∼200 copies of RISC associated siRNA. Further, efficacy of gelatin nanoparticle mediated knockdown of TG2 and IL-15 mRNA was tested in an in vitro model of celiac disease. Significant suppression in the levels of proinflammatory cytokines (TNF-α and IFN-γ) was observed in p31-43 stimulated J774A.1 cells upon either IL-15 or IL-15 + TG2 siRNA treatment. The results from this study indicate that gelatin nanoparticle mediated TG2 and IL-15 siRNA gene silencing is a very promising approach for the treatment of celiac disease.


Asunto(s)
Enfermedad Celíaca/genética , Proteínas de Unión al GTP/metabolismo , Gelatina/química , Interleucina-15/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/enzimología , Nanopartículas/química , Transglutaminasas/metabolismo , Animales , Células CACO-2 , Línea Celular , Proteínas de Unión al GTP/genética , Silenciador del Gen , Humanos , Interferón gamma/metabolismo , Interleucina-15/genética , Ratones , Proteína Glutamina Gamma Glutamiltransferasa 2 , ARN Interferente Pequeño/metabolismo , Transglutaminasas/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
Pharm Res ; 34(7): 1416-1427, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28389708

RESUMEN

PURPOSE: Nanoemulsions have shown potential in delivering drug across epithelial and endothelial cell barriers, which express efflux transporters. However, their transport mechanisms are not entirely understood. Our goal was to investigate the cellular permeability of nanoemulsion-encapsulated drugs and apply mathematical modeling to elucidate transport mechanisms and sensitive nanoemulsion attributes. METHODS: Transport studies were performed in Caco-2 cells, using fish oil nanoemulsions and a model substrate, rhodamine-123. Permeability data was modeled using a semi-mechanistic approach, capturing the following cellular processes: endocytotic uptake of the nanoemulsion, release of rhodamine-123 from the nanoemulsion, efflux and passive permeability of rhodamine-123 in aqueous solution. RESULTS: Nanoemulsions not only improved the permeability of rhodamine-123, but were also less sensitive to efflux transporters. The model captured bidirectional permeability results and identified sensitive processes, such as the release of the nanoemulsion-encapsulated drug and cellular uptake of the nanoemulsion. CONCLUSIONS: Mathematical description of cellular processes, improved our understanding of transport mechanisms, such as nanoemulsions don't inhibit efflux to improve drug permeability. Instead, their endocytotic uptake, results in higher intracellular drug concentrations, thereby increasing the concentration gradient and transcellular permeability across biological barriers. Modeling results indicated optimizing nanoemulsion attributes like the droplet size and intracellular drug release rate, may further improve drug permeability.


Asunto(s)
Emulsiones/metabolismo , Modelos Biológicos , Nanopartículas/metabolismo , Rodaminas/metabolismo , Transporte Biológico , Células CACO-2 , Química Farmacéutica , Portadores de Fármacos , Liberación de Fármacos , Emulsiones/química , Endocitosis , Aceites de Pescado , Humanos , Nanopartículas/química , Tamaño de la Partícula , Permeabilidad , Rodaminas/química , Soluciones
18.
Mol Ther ; 24(4): 759-69, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26686386

RESUMEN

Mutations in KRAS and p53 signaling pathways contribute to loss of responsiveness to current therapies and a decreased survival in lung cancer. In this study, we have investigated the delivery and transfection of wild-type (wt-) p53 and microRNA-125b (miR-125b) expressing plasmid DNA, in SK-LU-1 human lung adenocarcinoma cells as well as in Kras(G12D)/p53(fl/fl) (KP) genetically engineered mouse model of lung cancer. Systemic plasmid DNA delivery with dual CD44/EGFR-targeted hyaluronic acid (HA)-based nanoparticles (NPs) resulted in a 2- to 20-fold increase in wt-p53 and miR-125b gene expression in SK-LU-1 cells. This resulted in enhanced apoptotic activity as seen with increased APAF-1 and caspase-3 gene expression. Similarly, in vivo evaluations in KP mouse model indicated successful CD44/EGFR-targeted delivery. Tumor growth inhibition and apoptotic induction were also observed with (wt-p53+miR125b) combination therapy in KP tumor model. Lastly, J774.A1 murine macrophages co-cultured with transfected SK-LU-1 cells showed a 14- to 35-fold increase in the iNOS-Arg-1 ratio, supportive of previous results demonstrating a role of miR-125b in macrophage repolarization. Overall, these results show tremendous promise of wt-p53 and miR-125b gene therapy using dual CD44/EGFR-targeting HA NP vector for effective treatment of lung cancer.


Asunto(s)
Ácido Hialurónico/administración & dosificación , Neoplasias Pulmonares/terapia , MicroARNs/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Receptores ErbB/metabolismo , Ingeniería Genética , Terapia Genética , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/química , Neoplasias Pulmonares/genética , Ratones , Nanopartículas/química , Neoplasias Experimentales , Plásmidos/genética , Transfección
19.
Mol Pharm ; 13(10): 3404-3416, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27552547

RESUMEN

The main goal of this study was to evaluate tumor necrosis factor-alpha (TNF-α) gene silencing in peritoneal macrophages upon activation with lipopolysaccharide (LPS), using CD44-targeting hyaluronic acid (HA)-based nanoparticles encapsulating TNF-α-specific small interfering RNA (siTNF-α). HA nanoparticles were formulated by blending hyaluronic acid-poly(ethylene imine) (HA-PEI), hyaluronic acid-hexyl fatty acid (HA-C6), and hyaluronic acid-poly(ethylene glycol) (HA-PEG) in 3:2:1 weight ratio, and encapsulating siTNF-α to form spherical particles of 78-90 nm diameter. Following intraperitoneal (IP) administration in LPS-treated C57BL/6 mice, the nanoparticles were actively taken up by macrophages and led to a significant downregulation of peritoneal TNF-α level. Downregulation of peritoneal macrophage-specific TNF-α also had a significant impact on other pro-inflammatory cytokine and chemokine levels in the serum. The C57BL/6 group of mice challenged with 5 mg/kg LPS had a significantly higher survival rate when they were treated with 3 mg/kg siTNF-α, either prior or simultaneously with the LPS administration, as compared to the LPS-challenged mice, which were treated with controls including the scrambled siRNA formulation. Overall, the results of this study demonstrate that CD44 targeting HA nanoparticles can selectively deliver siTNF-α to peritoneal macrophages leading to downregulation of pro-inflammatory cytokines in the peritoneal fluid and in the serum. This RNAi strategy could potentially provide an important therapeutic modality for acute inflammatory diseases, such as septic shock.


Asunto(s)
Receptores de Hialuranos/metabolismo , Ácido Hialurónico/química , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/metabolismo , Nanopartículas/química , Factor de Necrosis Tumoral alfa/genética , Animales , Receptores de Hialuranos/genética , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Nanopartículas/metabolismo , ARN Interferente Pequeño/genética
20.
Nanomedicine ; 12(4): 987-1002, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26767514

RESUMEN

Neuroinflammation is a hallmark of acute and chronic neurodegenerative disorders. The main aim of this study was to evaluate the therapeutic efficacy of intranasal cationic nanoemulsion encapsulating an anti-TNFα siRNA, for potential anti-inflammatory therapy. TNFα siRNA nanoemulsions were prepared and characterized for particle size, surface charge, morphology, and stability and encapsulation efficiency. Qualitative and quantitative intracellular uptake studies by confocal imaging and flow cytometry, respectively, showed higher uptake compared to Lipofectamine® transfected siRNA. Nanoemulsion significantly lowered TNFα levels in LPS-stimulated cells. Upon intranasal delivery of cationic nanoemulsions almost 5 fold higher uptake was observed in the rat brain compared to non-encapsulated siRNA. More importantly, intranasal delivery of TNFα siRNA nanoemulsions in vivo markedly reduced the unregulated levels of TNFα in an LPS-induced model of neuroinflammation. These results indicate that intranasal delivery of cationic nanoemulsions encapsulating TNFα siRNA offered an efficient means of gene knockdown and this approach has significant potential in prevention of neuroinflammation. FROM THE CLINICAL EDITOR: Neuroinflammation is often seen in patients with neurodegenerative disorders and tumor necrosis factor-alpha (TNFα) plays a significant role in contributing to neuronal dysfunction. As a result, inhibition of TNFα may alleviate disease severity. In this article, the authors investigated using a cationic nanoemulsion system carrying TNFα siRNA intra-nasally to protect against neuroinflammation. This new method may provide a future approach in this clinical setting.


Asunto(s)
Encéfalo/efectos de los fármacos , Inflamación/tratamiento farmacológico , ARN Interferente Pequeño/genética , Factor de Necrosis Tumoral alfa/genética , Administración Intranasal , Animales , Antiinflamatorios/administración & dosificación , Encéfalo/patología , Emulsiones/administración & dosificación , Citometría de Flujo , Silenciador del Gen , Humanos , Inflamación/genética , Lípidos , Macrófagos , Nanopartículas/administración & dosificación , Nanopartículas/química , Neuronas/efectos de los fármacos , Neuronas/patología , Tamaño de la Partícula , ARN Interferente Pequeño/administración & dosificación , Ratas , Factor de Necrosis Tumoral alfa/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA