Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 272
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Allergy Clin Immunol ; 151(2): 509-525.e8, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36494212

RESUMO

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.


Assuntos
Vesículas Extracelulares , Viroses , Humanos , Receptor 3 Toll-Like , Imunidade Inata , Antivirais/farmacologia , Poli I-C
2.
Mol Pharm ; 20(3): 1729-1736, 2023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36744718

RESUMO

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.


Assuntos
Lipossomos , Sinusite , Animais , Camundongos , Lipossomos/uso terapêutico , Verapamil , Polietilenoglicóis/uso terapêutico , Camundongos Endogâmicos C57BL , Administração Intranasal , Sinusite/tratamento farmacológico , Administração Tópica
3.
Pharm Res ; 40(1): 77-105, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36380168

RESUMO

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.


Assuntos
Sistemas de Liberação de Medicamentos , MicroRNAs , Ácidos Nucleicos , RNA Interferente Pequeno , Humanos , Barreira Hematoencefálica , Encéfalo , MicroRNAs/uso terapêutico , Ácidos Nucleicos/uso terapêutico , Oligonucleotídeos Antissenso/uso terapêutico , RNA Interferente Pequeno/uso terapêutico
4.
J Allergy Clin Immunol ; 150(4): 872-881, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35660375

RESUMO

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.


Assuntos
Pólipos Nasais , Rinite , Cistatinas Salivares , Sinusite , Alérgenos , Animais , Doença Crônica , Inibidores de Cisteína Proteinase , Citocinas , Inflamação , Camundongos , Pólipos Nasais/patologia , Peptídeo Hidrolases , Proteômica , Rinite/metabolismo , Cistatinas Salivares/genética , Cistatinas Salivares/metabolismo , Sinusite/patologia
5.
Mol Pharm ; 19(12): 4411-4427, 2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36154128

RESUMO

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.


Assuntos
Implantes Absorvíveis , Sistemas de Liberação de Medicamentos , Idoso , Humanos , Sistemas de Liberação de Medicamentos/métodos , Impressão Tridimensional , Medicina de Precisão , Fármacos do Sistema Nervoso Central
6.
Mol Pharm ; 19(11): 4067-4079, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36226722

RESUMO

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.


Assuntos
Vesículas Extracelulares , MicroRNAs , Neoplasias Ovarianas , Humanos , Feminino , MicroRNAs/genética , MicroRNAs/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vesículas Extracelulares/metabolismo , Neoplasias Ovarianas/metabolismo , Modelos Teóricos , Microambiente Tumoral
7.
Nanomedicine ; 40: 102488, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34748964

RESUMO

The goal of this study was to evaluate hepatocyte-specific gene editing, via systemic administration of hyaluronic acid (HA)-based nanoparticles in naïve CD-1 mice. Using HA-poly(ethylene imine) (HA-PEI) and HA-PEI-mannose nanoparticles with differential mannose density (1X and 2X), we have evaluated systemic biodistribution and hepatocyte-specific delivery using IVIS imaging and flow cytometry. Additionally, we have investigated hepatocyte-specific delivery and transfection of CRISPR/Cas9 gene editing plasmid and eGFP gene payload to integrate at the Rosa26 locus. IVIS imaging showed uptake of HA-PEI nanoparticles primarily by the liver, and with addition of mannose at different concentrations, the nanoparticles showed increased uptake in both the liver and spleen. HA-PEI-mannose nanoparticles showed 55-65% uptake by hepatocytes, along with uptake by resident macrophage regardless of the mannose concentration. One of two gRNA targets showed 15% genome editing and obtained similar results for all three nanoparticle formulations. Cells positive for our gene payload were greatest with HA-PEI-mannose-1X nanoparticles where 16.2% of cells were GFP positive. The results were encouraging as proof of concept for the development of a non-viral biodegradable and biocompatible polymeric delivery system for gene editing specifically targeting hepatocytes upon systemic administration.


Assuntos
Edição de Genes , Nanopartículas , Animais , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Hepatócitos , Ácido Hialurônico , Camundongos , Distribuição Tecidual
8.
Mol Pharm ; 18(8): 3099-3107, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34228470

RESUMO

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.


Assuntos
Doença Celíaca/tratamento farmacológico , Doença Celíaca/genética , Gastroenterite/tratamento farmacológico , Gastroenterite/genética , Interleucina-15/genética , Microesferas , Sistemas de Liberação de Fármacos por Nanopartículas/química , Nanopartículas/química , Proteína 2 Glutamina gama-Glutamiltransferase/genética , Interferência de RNA , Administração Oral , Animais , Doença Celíaca/induzido quimicamente , Modelos Animais de Doenças , Composição de Medicamentos/métodos , Gastroenterite/induzido quimicamente , Interleucina-15/administração & dosagem , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Poli I-C/efeitos adversos , Proteína 2 Glutamina gama-Glutamiltransferase/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Resultado do Tratamento
9.
Nanomedicine ; 37: 102422, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34175455

RESUMO

As mitochondria network together to act as the master sensors and effectors of apoptosis, ATP production, reactive oxygen species management, mitophagy/autophagy, and homeostasis; this organelle is an ideal target for pharmaceutical manipulation. Mitochondrial dysfunction contributes to many diseases, for example, ß-amyloid has been shown to interfere with mitochondrial protein import and induce apoptosis in Alzheimer's Disease while some forms of Parkinson's Disease are associated with dysfunctional mitochondrial PINK1 and Parkin proteins. Mitochondrial medicine has applications in the treatment of an array of pathologies from cancer to cardiovascular disease. A challenge of mitochondrial medicine is directing therapies to a subcellular target. Nanotechnology based approaches combined with mitochondrial targeting strategies can greatly improve the clinical translation and effectiveness of mitochondrial medicine. This review discusses mitochondrial drug delivery approaches and applications of mitochondrial nanomedicines. Nanomedicine approaches have the potential to drive the success of mitochondrial therapies into the clinic.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Nanomedicina , Doença de Parkinson/tratamento farmacológico , Trifosfato de Adenosina/biossíntese , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Autofagia/efeitos dos fármacos , Autofagia/genética , Humanos , Mitocôndrias/genética , Mitofagia/efeitos dos fármacos , Mitofagia/genética , Doença de Parkinson/genética , Doença de Parkinson/patologia , Espécies Reativas de Oxigênio
10.
Mol Pharm ; 17(6): 1996-2005, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32365295

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Cátions/química , RNA Mensageiro/administração & dosagem , RNA Mensageiro/metabolismo , Administração Intranasal , Animais , Linhagem Celular , Sistemas de Liberação de Medicamentos , Lipossomos/química , Masculino , Camundongos
11.
J Allergy Clin Immunol ; 143(4): 1525-1535.e1, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30442371

RESUMO

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.


Assuntos
Exossomos/imunologia , Imunidade Inata/imunologia , Mucosa Nasal/imunologia , Humanos , Mucosa Nasal/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Infecções por Pseudomonas/imunologia
12.
Small ; 15(36): e1902232, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31328877

RESUMO

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.


Assuntos
Hidrogéis/química , Imunomodulação/fisiologia , MicroRNAs/química , Nanopartículas/química , Cicatrização/fisiologia , Animais , Linhagem Celular , Ácido Hialurônico/química , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Microscopia Eletrônica de Varredura , Cicatrização/genética
13.
J Pharmacol Exp Ther ; 370(3): 843-854, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30591529

RESUMO

The main objective of this study was to develop and evaluate the CNS delivery efficiency, distribution, therapeutic efficacy, and safety of cyclosporine A (CSA) using a cationic oil-in-water nanoemulsion system upon intranasal administration. An omega-3 fatty acid-rich, flaxseed oil-based nanoemulsion was used for intranasal delivery to the brain, and further magnetic resonance imaging (MRI) was used to evaluate and confirm the transport of the positively charged CSA nanoemulsion (CSA-NE) in CNS. Furthermore, the anti-inflammatory potential of CSA peptide was evaluated using the lipopolysaccharide (LPS) model of neuroinflammation in rats. CSA-NE showed a good safety profile when tested in vitro in RPMI 2650 cells. Upon intranasal administration in rats, the nanoemulsion delivery system showed higher uptake in major regions of the brain based on changes in MRI T1 (longitudinal relaxation time) values. Additionally, CSA nanoemulsion showed improved therapeutic efficacy by inhibiting proinflammatory cytokines in the LPS-stimulated rat model of neuroinflammation compared with solution formulation. Preliminary safety evaluations show that the nanoemulsion system was well tolerated and did not cause any acute negative effects in rats. Based on these results, intranasal delivery of CSA and other "neuroprotective peptides" may provide a clinically translatable strategy for treating neurologic diseases.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Sistema Nervoso Central , Ciclosporina/administração & dosagem , Ciclosporina/farmacologia , Sistemas de Liberação de Medicamentos , Administração Intranasal , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Linhagem Celular , Ciclosporina/efeitos adversos , Citocinas/metabolismo , Composição de Medicamentos , Emulsões , Ácidos Graxos Ômega-3 , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Óleo de Semente do Linho , Lipopolissacarídeos , Imageamento por Ressonância Magnética , Masculino , Nanoestruturas , Ratos , Ratos Sprague-Dawley
14.
Mol Pharm ; 16(1): 128-140, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30525660

RESUMO

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.


Assuntos
Ácido Hialurônico/química , Nanopartículas/química , Plasmídeos/genética , Transfecção/métodos , Células A549 , Sulfato de Dextrana/química , Células HeLa , Humanos , Polietilenoglicóis/química , Polietilenoimina/análogos & derivados , Polietilenoimina/química
15.
Pharm Res ; 36(5): 75, 2019 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-30923914

RESUMO

PURPOSE: Despite encouraging preclinical results, mechanisms of CNS drug delivery following intranasal dosing of nanoemulsions remain incompletely understood. Herein, the transport characteristics of intranasally administered nanoemulsions are investigated using mathematical modeling and simulation. METHODS: A compartmental model was developed to describe systemic and brain pharmacokinetics of drug solutions following intranasal dosing in rodents. The association between transport processes and CNS drug delivery was predicted using sensitivity analysis. Published pharmacokinetic data for four drugs; dosed as a nanoemulsion and aqueous solution were modeled to characterize differences in transport processes across formulations. RESULTS: The intranasal model structure performed in a drug agnostic fashion. Sensitivity analysis suggested that though the extent of CNS drug delivery depends on nasal bioavailability, the CNS targeting efficiency is only sensitive to changes in drug permeability across the nasal epithelium. Modeling results indicated that nanoemulsions primarily improve nasal bioavailability and drug permeability across the olfactory epithelium, with minimal effect on drug permeability across the non-olfactory epithelium. CONCLUSIONS: Using mathematical modeling we outlined dominant transport pathways following intranasal dosing, predicted the association between transport pathways and CNS drug delivery, predicted human CNS delivery after accounting for inter-species differences in nasal anatomy, and quantified the CNS delivery potential of different formulations in rodents.


Assuntos
Sistema Nervoso Central/metabolismo , Simulação por Computador , Sistemas de Liberação de Medicamentos/métodos , Modelos Biológicos , Nanopartículas/administração & dosagem , Preparações Farmacêuticas/administração & dosagem , Administração Intranasal , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Emulsões , Humanos , Nanopartículas/química , Mucosa Olfatória/metabolismo , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Ratos , Pesquisa Translacional Biomédica
16.
Pharm Res ; 36(11): 153, 2019 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-31482243

RESUMO

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.


Assuntos
Desenvolvimento de Medicamentos/métodos , Nanocápsulas/química , Nanotecnologia/métodos , Animais , Preparações de Ação Retardada/química , Composição de Medicamentos , Estabilidade de Medicamentos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Nanomedicina , Resultado do Tratamento
17.
Pharm Res ; 36(9): 134, 2019 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-31297653

RESUMO

PURPOSE: Despite extensive preclinical investigations, in-vivo properties and formulation characteristics that improve CNS drug delivery following systemic dosing of nanoemulsions remain incompletely understood. METHODS: The CNS targeting potential of systemically administered nanoemulsions was evaluated by formulating rapamycin containing fish oil nanoemulsions, and testing the combined effect of formulation characteristics such as the circulation half-life and particle size distribution, on CNS delivery of rapamycin containing fish oil nanoemulsions in mice. RESULTS: Results generated with rapamycin nanoemulsions suggested that circulation half-life and particle size distribution did not impact the brain targeting efficiency of rapamycin containing fish oil nanoemulsions. Further, in the absence of any improvement in the systemic exposures of rapamycin, nanoemulsions did not outperform their aqueous counterpart with respect to the extent of CNS drug delivery. CONCLUSIONS: Our findings confirm that BBB penetration, which primarily depends on intrinsic drug-related properties, may not be significantly improved following encapsulation of drugs in nanoemulsions. Graphical Abstract The CNS targeting potential of systemically administered nanoemulsions was investigated by formulating various rapamycin containing fish oil nanoemulsions associated with different formulation characteristics such as the circulation half-life and particle size distribution. The targeting efficiency (TE) defined as the ratio of the brain exposures to the accompanying systemic exposures of rapamycin was estimated for each formulation following IV dosing in mice.


Assuntos
Encéfalo/metabolismo , Óleos de Peixe/química , Nanopartículas/química , Sirolimo/administração & dosagem , Animais , Barreira Hematoencefálica/metabolismo , Permeabilidade da Membrana Celular , Emulsões , Camundongos , Tamanho da Partícula , Polietilenoglicóis/química , Sirolimo/farmacocinética , Distribuição Tecidual
18.
Nanomedicine ; 22: 102091, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31626992

RESUMO

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.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Sistemas de Liberação de Medicamentos , Inativação Gênica , Absorção Intestinal , Intestinos/fisiologia , Ferro/metabolismo , Nanopartículas/administração & dosagem , Resinas Acrílicas/química , Administração Oral , Animais , Células CACO-2 , Gelatina/química , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Ferro/administração & dosagem , Masculino , Camundongos , Nanopartículas/ultraestrutura , Tamanho da Partícula , RNA Interferente Pequeno/metabolismo
19.
Nanomedicine ; 15(1): 274-284, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30343013

RESUMO

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.


Assuntos
Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Nanoestruturas/química , Paclitaxel/farmacocinética , Peritônio/metabolismo , Têxteis , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacologia , Proliferação de Células , Células Cultivadas , Implantes de Medicamento , Liberação Controlada de Fármacos , Camundongos , Camundongos Endogâmicos BALB C , Nanoestruturas/administração & dosagem , Paclitaxel/administração & dosagem , Polímeros/química , Distribuição Tecidual
20.
Nano Lett ; 18(6): 3571-3579, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29722542

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Macrófagos Peritoneais/patologia , MicroRNAs/administração & dosagem , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Polaridade Celular , Modelos Animais de Doenças , Engenharia Genética , Humanos , Ácido Hialurônico/administração & dosagem , Neoplasias Pulmonares/genética , Macrófagos Peritoneais/metabolismo , Camundongos , MicroRNAs/genética , Nanopartículas/administração & dosagem , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA