Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 14(5): 344, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37253771

RESUMEN

Cardiac hypertrophy is a common structural remodeling in many cardiovascular diseases. Recently, long non-coding RNAs (LncRNAs) were found to be involved in the physiological and pathological processes of cardiac hypertrophy. In this study, we found that LncRNA KCND1 (LncKCND1) was downregulated in both transverse aortic constriction (TAC)-induced hypertrophic mouse hearts and Angiotensin II (Ang II)-induced neonatal mouse cardiomyocytes. Further analyses showed that the knockdown of LncKCND1 impaired cardiac mitochondrial function and led to hypertrophic changes in cardiomyocytes. In contrast, overexpression of LncKCND1 inhibited Ang II-induced cardiomyocyte hypertrophic changes. Importantly, enhanced expression of LncKCND1 protected the heart from TAC-induced pathological cardiac hypertrophy and improved heart function in TAC mice. Subsequent analyses involving mass spectrometry and RNA immunoprecipitation assays showed that LncKCND1 directly binds to YBX1. Furthermore, overexpression of LncKCND1 upregulated the expression level of YBX1, while silencing LncKCND1 had the opposite effect. Furthermore, YBX1 was downregulated during cardiac hypertrophy, whereas overexpression of YBX1 inhibited Ang II-induced cardiomyocyte hypertrophy. Moreover, silencing YBX1 reversed the effect of LncKCND1 on cardiomyocyte mitochondrial function and its protective role in cardiac hypertrophy, suggesting that YBX1 is a downstream target of LncKCND1 in regulating cardiac hypertrophy. In conclusion, our study provides mechanistic insights into the functioning of LncKCND1 and supports LncKCND1 as a potential therapeutic target for pathological cardiac hypertrophy.


Asunto(s)
ARN Largo no Codificante , Factores de Transcripción , Animales , Ratones , Angiotensina II/farmacología , Cardiomegalia/metabolismo , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Canales de Potasio Shal/metabolismo , Factores de Transcripción/metabolismo
2.
Cell Death Dis ; 13(6): 525, 2022 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-35661695

RESUMEN

Long non-coding RNA (lncRNA) was reported to be a critical regulator of cellular homeostasis, but poorly understood in idiopathic pulmonary fibrosis (IPF). Here, we systematically identified a crucial lncRNA, p53-induced long non-coding RNA TP53 target 1 (TP53TG1), which was the dysregulated hub gene in IPF regulatory network and one of the top degree genes and down-regulated in IPF-drived fibroblasts. Functional experiments revealed that overexpression of TP53TG1 attenuated the increased expression of fibronectin 1 (Fn1), Collagen 1α1, Collagen 3α1, ACTA2 mRNA, Fn1, and Collagen I protein level, excessive fibroblasts proliferation, migration and differentiation induced by TGF-ß1 in MRC-5 as well as PMLFs. In vivo assays identified that forced expression of TP53TG1 by adeno-associated virus 5 (AAV5) not only prevented BLM-induced experimental fibrosis but also reversed established lung fibrosis in the murine model. Mechanistically, TP53TG1 was found to bind to amount of tight junction proteins. Importantly, we found that TP53TG1 binds to the Myosin Heavy Chain 9 (MYH9) to inhibit its protein expression and thus the MYH9-mediated activation of fibroblasts. Collectively, we identified the TP53TG1 as a master suppressor of fibroblast activation and IPF, which could be a potential hub for targeting treatment of the disease.


Asunto(s)
Fibrosis Pulmonar Idiopática , ARN Largo no Codificante , Animales , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibrosis , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
3.
Acta Pharmacol Sin ; 43(11): 2862-2872, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35396533

RESUMEN

Aberrant activation of cardiac fibroblasts is the main cause and character of cardiac fibrosis, and inhibition of cardiac fibrosis becomes a promising treatment for cardiac diseases. Platelet-activating factor (PAF) and Hippo pathway is recently recognized as key signaling mechanisms in cardiovascular diseases. In this study we explored the potential roles of PAF and Hippo signaling pathway in cardiac fibrosis. Myocardial infarction (MI) was induced in mice by left anterior descending artery ligation. After 28 days, the mice were sacrificed, and the hearts were collected for analyses. We showed that PAF receptor (PAFR) and yes-associated protein 1 (YAP1, a key effector in the Hippo pathway) were significantly increased in the heart of MI mice. Increased expression of PAFR and YAP1 was also observed in angiotensin II (Ang II)-treated mouse cardiac fibroblasts. In mouse cardiac fibroblasts, forced expression of YAP1 increased cell viability, resulted in collagen deposition and promoted fibroblast-myofibroblast transition. We showed that PAF induced fibrogenesis through activation of YAP1 and promoted its nuclear translocation via interacting with PAFR, while YAP1 promoted the expression of PAFR by binding to and activating transcription factor TEAD1. More importantly, silencing PAFR or YAP1 by shRNA, or using transgenic mice to induce the conditional deletion of YAP1 in cardiac fibroblasts, impeded cardiac fibrosis and improved cardiac function in MI mice. Taken together, this study elucidates the role and mechanisms of PAFR/YAP1 positive feedback loop in cardiac fibrosis, suggesting a potential role of this pathway as novel therapeutic targets in cardiac fibrosis.


Asunto(s)
Infarto del Miocardio , Factor de Activación Plaquetaria , Ratones , Animales , Retroalimentación , Transducción de Señal/fisiología , Fibroblastos/metabolismo , Infarto del Miocardio/metabolismo , Ratones Transgénicos , Fibrosis
4.
Adv Sci (Weinh) ; 7(1): 1901904, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31921563

RESUMEN

Organophosphate nerve agents rapidly inhibit cholinesterases thereby destroying the ability to sustain life. Strong nucleophiles, such as oximes, have been used as therapeutic reactivators of cholinesterase-organophosphate complexes, but suffer from short half-lives and limited efficacy across the broad spectrum of organophosphate nerve agents. Cholinesterases have been used as long-lived therapeutic bioscavengers for unreacted organophosphates with limited success because they react with organophosphate nerve agents with one-to-one stoichiometries. The chemical power of nucleophilic reactivators is coupled to long-lived bioscavengers by designing and synthesizing cholinesterase-polymer-oxime conjugates using atom transfer radical polymerization and azide-alkyne "click" chemistry. Detailed kinetic studies show that butyrylcholinesterase-polymer-oxime activity is dependent on the electrostatic properties of the polymers and the amount of oxime within the conjugate. The covalent coupling of oxime-containing polymers to the surface of butyrylcholinesterase slows the rate of inactivation of paraoxon, a model nerve agent. Furthermore, when the enzyme is covalently inhibited by paraoxon, the covalently attached oxime induced inter- and intramolecular reactivation. Intramolecular reactivation will open the door to the generation of a new class of nerve agent scavengers that couple the speed and selectivity of biology to the ruggedness and simplicity of synthetic chemicals.

5.
Acta Biomater ; 101: 422-435, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31669698

RESUMEN

The global and economic success of immunoglobulin-based therapeutics in treating a wide range of diseases has heightened the need to further enhance their efficacy and lifetime while diminishing deleterious side effects. The three most ubiquitous challenges of therapeutic immunoglobulin delivery are their relatively short lifetimes in vivo, the immunologic consequences of soluble antibody-antigen complexes, and the emergence of anti-drug antibodies. We describe the rapid, cell-tolerated chemical engineering of the erythrocyte membrane in order to display any antibody, our model system being the display of anti-Tumor Necrosis Factor (anti-TNFα), on the surface of long-lived red blood cells (RBCs) while masking the antibody's Fc region. We developed four synthetic approaches to generate RBC-Staphylococcal protein A (RBC-SpA) complexes: amino group targeting through N-hydrosuccinidyl ester-functionalized homobifunctional poly(ethylene glycol) (NHS-PEG-NHS), direct thiol group targeting using heterobifunctional NHS-PEG-maleimide (NHS-PEG-MAL), converted thiol targeting using heterobifunctional NHS-PEG-MAL, and click chemistry using heterobifunctional NHS-PEG-azido (NHS-PEG-N3) and NHS-PEG-alkyne (NHS-PEG-alk). The RBC-PEG-SpA complexes were formed within minutes, followed by the attachment of over 105 antibodies per RBC to the accessible RBC-bound SpA via Fc-Protein A coupling. The RBC-PEG-SpA-antibody arrays were shown to be stable for more than 60 days in PBS and for more than 42 days in serum containing buffer. RBC-PEG-SpA-antibody complexes were shown to remove TNFα from physiological buffer and had similar mechanical properties to unmodified RBCs. Out of the four approaches, the converted thiol method provided the most controlled chemistry and construct stability. We are now ideally positioned to determine the long-term in vivo efficacy of chemically membrane-engineered RBCs to remove antigens, like TNFα, from serum. STATEMENT OF SIGNIFICANCE: The global and economic success of immunoglobulin-based therapeutics in treating a wide range of diseases has heightened the need to further enhance their efficacy and lifetime while diminishing deleterious side effects. The three most ubiquitous challenges of therapeutic immunoglobulin delivery are their relatively short lifetimes in vivo, the immunologic consequences of soluble antibody-antigen complexes, and the emergence of anti-drug antibodies. We describe the rapid, cell-tolerated chemical engineering of the erythrocyte membrane to display any antibody, our model system being the display of anti-Tumor Necrosis Factor (anti-TNFα), on the surface of long-lived red blood cells (RBCs) while masking the antibody's Fc region. Conversion of RBCs into therapeutic delivery vehicles, we argue, would enhance the circulation life of immunoglobulin-based therapeutics while simultaneously evading deleterious immune response.


Asunto(s)
Portadores de Fármacos/química , Eritrocitos/metabolismo , Inmunoglobulinas/uso terapéutico , Anticuerpos/metabolismo , Antígenos/metabolismo , Química Clic , Membrana Eritrocítica/metabolismo , Humanos , Polietilenglicoles/síntesis química , Polietilenglicoles/química , Unión Proteica , Proteína Estafilocócica A/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Nano Lett ; 19(7): 4237-4249, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-30868883

RESUMEN

Among approaches of current cancer immunotherapy, a dendritic cell (DC)-targeted vaccine based on nanotechnology could be a promising way to efficiently induce potent immune responses. To enhance DC targeting and vaccine efficiency, we included imiquimod (IMQ), a toll-like receptor 7/8 (TLR 7/8) agonist, and monophosphoryl lipid A (MPLA), a TLR4 agonist, to synthesize lipid-polymer hybrid nanoparticles using PCL-PEG-PCL and DOTAP (IMNPs) as well as DSPE-PEG-mannose (MAN-IMNPS). The spatiotemporal delivery of MPLA (within the outer lipid layer) to extracellular TLR4 and IMQ (in the hydrophobic core of NPs) to intracellular TLR7/8 can activate DCs synergistically to improve vaccine efficacy. Ovalbumin (OVA) as a model antigen was readily absorbed by positively charged DOTAP and showed a quick release in vitro. Our results demonstrated that this novel nanovaccine enhanced cellular uptake, cytokine production, and maturation of DCs. Compared with the quick metabolism of free OVA-agonists, the depot effect of OVA-IMNPs was observed, whereas MAN-OVA-IMNPs promoted trafficking to secondary lymphoid organs. After immunization with a subcutaneous injection, the nanovaccine, especially MAN-OVA-IMNPs, induced more antigen-specific CD8+ T cells, greater lymphocyte activation, stronger cross-presentation, and more generation of memory T cells, antibody, IFN-γ, and granzyme B. Prophylactic vaccination of MAN-OVA-IMNPs significantly delayed tumor development and prolonged the survival in mice. The therapeutic tumor challenge indicated that MAN-OVA-IMNPs prohibited tumor progression more efficiently than other formulations, and the combination with an immune checkpoint blockade further enhanced antitumor effects. Hence, the DC-targeted vaccine codelivery with IMQ and MPLA adjuvants by hybrid cationic nanoparticles in a spatiotemporal manner is a promising multifunctional antigen delivery system in cancer immunotherapy.


Asunto(s)
Antígenos de Neoplasias , Vacunas contra el Cáncer , Células Dendríticas/inmunología , Sistemas de Liberación de Medicamentos , Imiquimod , Inmunoterapia , Lípido A/análogos & derivados , Nanopartículas , Neoplasias Experimentales , Receptores Toll-Like/agonistas , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/farmacología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Vacunas contra el Cáncer/farmacología , Células Dendríticas/patología , Imiquimod/inmunología , Imiquimod/farmacocinética , Imiquimod/farmacología , Lípido A/inmunología , Lípido A/farmacocinética , Lípido A/farmacología , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Receptores Toll-Like/inmunología
7.
Biomacromolecules ; 18(8): 2583-2593, 2017 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-28657716

RESUMEN

Antibacterial polymers are potentially powerful biocides that can destroy bacteria on contact. Debate in the literature has surrounded the mechanism of action of polymeric biocides and the propensity for bacteria to develop resistance to them. There has been particular interest in whether surfaces with covalently coupled polymeric biocides have the same mechanism of action and resistance profile as similar soluble polymeric biocides. We designed and synthesized a series of poly(quaternary ammonium) polymers, with tailorable molecular structures and architectures, to engineer their antibacterial specificity and their ability to delay the development of bacterial resistance. These linear poly(quaternary ammonium) homopolymers and block copolymers, generated using atom transfer radical polymerization, had structure-dependent antibacterial specificity toward Gram positive and negative bacterial species. When single block copolymers contained two polymer segments of differing antibacterial specificity, the polymer combined the specificities of its two components. Nanoparticulate human serum albumin-poly(quaternary ammonium) conjugates of these same polymers, synthesized via "grafting from" atom transfer radical polymerization, were strongly biocidal and also exhibited a marked decrease in the rate of bacterial resistance development relative to linear polymers. These protein-biocide conjugates mimicked the behavior of surface-presented polycationic biocides rather than their nonproteinaceous counterparts.


Asunto(s)
Antibacterianos , Bacterias/crecimiento & desarrollo , Polímeros , Compuestos de Amonio Cuaternario , Albúmina Sérica Humana , Adsorción , Antibacterianos/química , Antibacterianos/farmacología , Células HEK293 , Humanos , Polímeros/química , Polímeros/farmacología , Compuestos de Amonio Cuaternario/química , Compuestos de Amonio Cuaternario/farmacología , Albúmina Sérica Humana/química , Albúmina Sérica Humana/farmacología
8.
Nat Nanotechnol ; 12(8): 813-820, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28416815

RESUMEN

An emerging approach for treating cancer involves programming patient-derived T cells with genes encoding disease-specific chimeric antigen receptors (CARs), so that they can combat tumour cells once they are reinfused. Although trials of this therapy have produced impressive results, the in vitro methods they require to generate large numbers of tumour-specific T cells are too elaborate for widespread application to treat cancer patients. Here, we describe a method to quickly program circulating T cells with tumour-recognizing capabilities, thus avoiding these complications. Specifically, we demonstrate that DNA-carrying nanoparticles can efficiently introduce leukaemia-targeting CAR genes into T-cell nuclei, thereby bringing about long-term disease remission. These polymer nanoparticles are easy to manufacture in a stable form, which simplifies storage and reduces cost. Our technology may therefore provide a practical, broadly applicable treatment that can generate anti-tumour immunity 'on demand' for oncologists in a variety of settings.


Asunto(s)
ADN/química , Portadores de Fármacos , Técnicas de Transferencia de Gen , Inmunidad Celular/efectos de los fármacos , Leucemia/terapia , Nanopartículas/química , Receptores Quiméricos de Antígenos , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Inmunidad Celular/genética , Leucemia/genética , Leucemia/inmunología , Leucemia/patología , Ratones , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología
9.
Biomaterials ; 115: 9-18, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27871003

RESUMEN

Immobilization of a ligand that selectively interacts with cancer cells to nanomaterials can enhance their diagnostic and therapeutic efficiency. In this study, we firstly demonstrate the high expression of receptor for cyclic nine-amino acid peptide LyP-1 (Cys-Gly-Asn-Lys-Arg-Thr-Arg-Gly-Cys) in both mouse and human pancreatic cancer. Based on these findings, sub-50 nm multifunctional superparamagnetic mesoporous nanospheres with surface modified with LyP-1 are rationally designed. Theses nanospheres have a core of silica-protected magnetite nanoparticle and a shell of FITC-labeled mesoporous silica, and they are able to specifically recognize and conjugate with the pancreatic cancer cell in vitro, as verified by the combined techniques of fluorescent imaging and T2 weight magnetic resonance imaging. After systematic administration, these LyP-1 immobilized nanospheres are found to actively target to mouse orthotopic xenograft of pancreatic cancer, which opens up the door for applications in early probing and diagnosis of pancreatic cancer by the multimodal imaging.


Asunto(s)
Nanopartículas de Magnetita/química , Técnicas de Sonda Molecular , Nanosferas/química , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/metabolismo , Péptidos Cíclicos/farmacocinética , Animales , Línea Celular Tumoral , Medios de Contraste/química , Femenino , Nanopartículas de Magnetita/ultraestructura , Ratones , Ratones Endogámicos BALB C , Sondas Moleculares/química , Imagen Multimodal/métodos , Nanocápsulas/química , Nanocápsulas/ultraestructura , Nanoconjugados/química , Nanoconjugados/ultraestructura , Nanosferas/ultraestructura , Neoplasias Pancreáticas/patología , Péptidos Cíclicos/química , Porosidad , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
10.
ACS Appl Mater Interfaces ; 7(23): 12649-54, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-25996162

RESUMEN

Because of the noninvasive, locally selective potential of thermal energy, considerable effort has been focused on the use of an external, alternating magnetic field for conversion of magnetic work to heat with iron oxide nanoparticles. However, proper regulation of thermal energy remains a challenge because of the lack of feedback from the local temperature change to the external power supply. Here, we show development of smart magnetic nanoparticles composed of Fe and Si with intrinsically tunable heat generation capability. They were engineered to possess an adjustable magnetic transition temperature through tuning the exchange between Fe atoms by incorporation of silicon atoms. They show relatively high magnetic moment. Moreover, their biocompatibility was established in several cell lines. The nanoparticles were also combined with a thermosensitive polymer, which had the capability to release of molecules with a magnetic stimulus, thereby providing a platform for locally controlled, drug release.

11.
Mol Pharm ; 12(3): 932-40, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25658873

RESUMEN

Nonviral gene transfection mediated by cationic polymer/DNA polyplexes often imposes stress and toxicity to cells. To better understand the relationship between cellular stress responses and polyplex-mediated transfection, polyplex-induced early autophagy in mouse fibroblasts was characterized and the impact of autophagy modulation on transgene expression evaluated. Transmission electron microscopy revealed the formation of double-membraned autophagosome in the cytoplasm of polyplex-transfected cells. Immunofluorescence staining and microscopy revealed intracellular LC3 punctation that was characteristic of early autophagy activation. Elevated expression of autophagosome-associated LC3 II protein was also detected by Western blot. When cells were treated with small-molecule modulators of autophagy, polyplex-mediated gene transfection efficiency was significantly affected. 3-Methyladenine (3-MA), an early autophagy inhibitor, reduced transfection efficiency, whereas rapamycin, an autophagy inducer, enhanced transgene expression. Importantly, the observed functional impact on gene transfection by autophagy modulation was decoupled from that of other modes of cellular stress response (apoptosis/necrosis). Treatment of cells by 3-MA or rapamycin did not affect the level of intracellular reactive oxygen species (ROS) but did decrease or increase, respectively, nuclear localization of polyplex-delivered plasmid DNA. These findings suggest new possibilities of enhancing polyplex-mediated gene delivery by codelivery of small-molecule regulators of autophagy.


Asunto(s)
Autofagia/genética , Transfección/métodos , Transporte Activo de Núcleo Celular , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia/efectos de los fármacos , Biofarmacia , ADN/genética , Expresión Génica , Técnicas de Transferencia de Gen , Ratones , Microscopía Electrónica de Transmisión , Células 3T3 NIH , Fagosomas/genética , Fagosomas/ultraestructura , Polímeros , Especies Reactivas de Oxígeno/metabolismo , Sirolimus/farmacología
12.
Macromol Biosci ; 15(5): 668-81, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25630277

RESUMEN

An injectable hybrid hydrogel system was developed consisting of hyaluronic acid (HA) crosslinked by well-defined block copolymers of the cationic poly(2-aminoethyl methacrylate) (PAEM) and polyethylene glycol (PEG). Robust, shear-thinning hybrid hydrogel was produced by mixing HA and 4-arm star PEG-PAEM block copolymer at 1:1 charge ratio. The encapsulation and release of highly viable human mesenchymal stem cells in physiological media was demonstrated. After subcutaneous injection of the hybrid gel in mice, mild but resolvable inflammatory response was observed. This hybrid gel could serve as a model system for studying structure-function relationship of polyelectrolyte hydrogels and as a practical injectable biomaterial for medical applications.


Asunto(s)
Reactivos de Enlaces Cruzados/farmacología , Ácido Hialurónico/farmacología , Hidrogeles/farmacología , Poliaminas/farmacología , Animales , Materiales Biocompatibles/farmacología , Cromatografía en Gel , Humanos , Ácido Hialurónico/química , Hidrogeles/síntesis química , Hidrogeles/química , Inyecciones , Inyecciones Subcutáneas , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Microesferas , Peso Molecular , Poliaminas/síntesis química , Poliaminas/química , Polielectrolitos , Polimerizacion , Reología , Solventes , Distribución Tisular/efectos de los fármacos
13.
Nanoscale Res Lett ; 9(1): 397, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25177217

RESUMEN

Carbon dots exhibit great potential in applications such as molecular imaging and in vivo molecular tracking. However, how to enhance fluorescence intensity of carbon dots has become a great challenge. Herein, we report for the first time a new strategy to synthesize fluorescent carbon dots (C-dots) with high quantum yields by using ribonuclease A (RNase A) as a biomolecular templating agent under microwave irradiation. The synthesized RNase A-conjugated carbon dots (RNase A@C-dots) exhibited quantum yields of 24.20%. The fluorescent color of the RNase A@C-dots can easily be adjusted by varying the microwave reaction time and microwave power. Moreover, the emission wavelength and intensity of RNase A@C-dots displayed a marked excitation wavelength-dependent character. As the excitation wavelength alters from 300 to 500 nm, the photoluminescence (PL) peak exhibits gradually redshifts from 450 to 550 nm, and the intensity reaches its maximum at an excitation wavelength of 380 nm. Its Stokes shift is about 80 nm. Notably, the PL intensity is gradually decreasing as the pH increases, almost linearly dependent, and it reaches the maximum at a pH = 2 condition; the emission peaks also show clearly a redshift, which may be caused by the high activity and perfective dispersion of RNase A in a lower pH solution. In high pH solution, RNase A tends to form RNase A warped carbon dot nanoclusters. Cell imaging confirmed that the RNase A@C-dots could enter into the cytoplasm through cell endocytosis. 3D confocal imaging and transmission electron microscopy observation confirmed partial RNase A@C-dots located inside the nucleus. MTT and real-time cell electronic sensing (RT-CES) analysis showed that the RNase A@C-dots could effectively inhibit the growth of MGC-803 cells. Intra-tumor injection test of RNase A@C-dots showed that RNase A@C-dots could be used for imaging in vivo gastric cancer cells. In conclusion, the as-prepared RNase A@C-dots are suitable for simultaneous therapy and in vivo fluorescence imaging of nude mice loaded with gastric cancer or other tumors.

14.
J Biomater Sci Polym Ed ; 25(10): 965-84, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24830505

RESUMEN

A new type of amphiphilic block copolymers, poly(ethylene glycol)-block-poly(2-methyl-acrylicacid 2-methoxy-5-methyl-[1,3]dioxin-5-ylmethyl ester) (PEG-b-PMME), bearing acid-labile six-membered ortho ester rings in side chains was synthesized by reversible addition-fragmentation chain-transfer polymerization, and the influence of chain length of the hydrophobic PMME block on micelle properties was investigated. The PEG-b-PMME micelles were stable in aqueous buffer at physiological pH with a low critical micelle concentration. Nile Red as a model drug was encapsulated into the micelles to explore the release profiles. The Nile Red-loaded polymeric micelles showed rapid release of Nile Red in weakly acidic environments (pH 5) but slow release under physiological condition (pH 7.4), due to different hydrolysis rate of ortho ester side chains of PEG-b-PMME. The Paclitaxel (PTX)-loaded micelles retained potency in killing lung cancer cells (A549), compared with the free PTX. No obvious toxicity was found in vitro and in vivo after intraperitoneal injection of the micelles, which confirms that the PEG-b-PMME micelles with unique acid-labile characteristic have great potential as nano-scaled carriers for drug delivery.


Asunto(s)
Portadores de Fármacos/síntesis química , Portadores de Fármacos/toxicidad , Interacciones Hidrofóbicas e Hidrofílicas , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Pruebas de Toxicidad , Animales , Peso Corporal/efectos de los fármacos , Línea Celular Tumoral , Técnicas de Química Sintética , Portadores de Fármacos/química , Ésteres , Femenino , Humanos , Ratones , Micelas , Células 3T3 NIH , Oxazinas/química , Paclitaxel/química , Paclitaxel/farmacología , Polimerizacion
15.
Macromol Biosci ; 14(7): 1015-24, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24700554

RESUMEN

A new type of homopolymers, PMAOE, bearing acid-cleavable cationic side-chains is synthesized and characterized. PMAOE is obtained via free radical polymerization, and they could efficiently bind and condense plasmid DNA at neutral pH. The strength of DNA binding is dependent on the length of PMAOE chains. NMR analysis reveals that hydrolysis of the ortho ester group of PMAOE follows an exocyclic mechanism and the rate of hydrolysis is much accelerated at mildly acidic pH, leading to accelerated disruption of polyplexes and release of DNA in mildly acidic environment. PMAOE is not toxic to cultured NIH 3T3 and COS-7 cells measured by MTT. This study demonstrates a unique mechanism of achieving pH-modulated binding and release of DNA from polymers with potential applications for gene delivery.


Asunto(s)
Ácidos/química , ADN/metabolismo , Polímeros/síntesis química , Acrilamidas/síntesis química , Acrilamidas/química , Animales , Células COS , Espectroscopía de Resonancia Magnética con Carbono-13 , Cationes , Muerte Celular/efectos de los fármacos , Chlorocebus aethiops , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Ratones , Células 3T3 NIH , Polimerizacion , Polímeros/química , Polímeros/toxicidad , Espectroscopía de Protones por Resonancia Magnética , Factores de Tiempo
16.
J Control Release ; 159(2): 232-9, 2012 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-22300619

RESUMEN

DNA vaccination using cationic polymers as carriers has the potential to be a very powerful method of immunotherapy, but typical immune responses generated have been less than robust. To better understand the details of DNA vaccine delivery in vivo, we prepared polymer/DNA complexes using three structurally distinct cationic polymers and fluorescently labeled plasmid DNA and injected them intradermally into mice. We analyzed transgene expression (luciferase) and the local tissue distribution of the labeled plasmid at the injection site at various time points (from hours to days). Comparable numbers of luciferase expressing cells were observed in the skin of mice receiving naked plasmid or polyplexes one day after transfection. At day 4, however, the polyplexes appeared to result in more transfected skin cells than naked plasmid. Live animal imaging revealed that naked plasmid dispersed quickly in the skin of mice after injection and had a wider distribution than any of the three types of polyplexes. However, naked plasmid level dropped to below detection limit after 24h, whereas polyplexes persisted for up to 2 weeks. The PEGylated polyplexes had a significantly wider distribution in the tissue than the nonPEGylated polyplexes. PEGylated polyplexes also distributed more broadly among dermal fibroblasts and allowed greater interaction with antigen-presenting cells (APCs) (dendritic cells and macrophages) starting at around 24h post-injection. By day 4, co-localization of polyplexes with APCs was observed at the injection site regardless of polymer structure, whereas small amounts of polyplexes were found in the draining lymph nodes. These in vivo findings demonstrate the superior stability of PEGylated polyplexes in physiological milieu and provide important insight on how cationic polymers could be optimized for DNA vaccine delivery.


Asunto(s)
ADN/administración & dosificación , Portadores de Fármacos/química , Plásmidos/administración & dosificación , Polímeros/química , Transgenes , Vacunas de ADN/administración & dosificación , Resinas Acrílicas/química , Animales , Cationes , ADN/genética , ADN/farmacocinética , Estabilidad de Medicamentos , Expresión Génica , Inyecciones Intradérmicas , Luciferasas/genética , Masculino , Metacrilatos/química , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Fluorescente , Estructura Molecular , Plásmidos/genética , Plásmidos/farmacocinética , Polietilenglicoles/química , Polietileneimina/química , Piel/metabolismo , Distribución Tisular , Transgenes/genética , Vacunas de ADN/genética , Vacunas de ADN/farmacocinética
17.
Biomacromolecules ; 12(12): 4373-85, 2011 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-22082257

RESUMEN

Poly(2-aminoethyl methacrylate) (PAEM) homopolymers with defined chain length and narrow molecular weight distribution were synthesized using atom transfer radical polymerization (ATRP), and a comprehensive study was conducted to evaluate the colloidal properties of PAEM/plasmid DNA polyplexes, the uptake and subcellular trafficking of polyplexes in antigen-presenting dendritic cells (DCs), and the biological performance of PAEM as a potential DNA vaccine carrier. PAEM of different chain length (45, 75, and 150 repeating units) showed varying strength in condensing plasmid DNA into narrowly dispersed nanoparticles with very low cytotoxicity. Longer polymer chain length resulted in higher levels of overall cellular uptake and nuclear uptake of plasmid DNA, but shorter polymer chains favored intracellular and intranuclear release of free plasmid from the polyplexes. Despite its simple chemical structure, PAEM transfected DCs very efficiently in vitro in media with or without serum and led to phenotypic maturation of DCs. When a model antigen-encoding ovalbumin plasmid was used, transfected DCs stimulated the activation of naïve CD8(+) T cells to produce high levels of interferon-γ. The efficiency of transfection, DC maturation, and CD8(+) T cell activation showed varying degrees of polymer chain-length dependence. These structurally defined cationic polymers may have much potential as efficient DNA vaccine carriers and immunostimulatory adjuvants. They may also serve as a model material system for elucidating structural and intracellular mechanisms of polymer-mediated DNA vaccine delivery.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Metacrilatos/síntesis química , Vacunas de ADN/administración & dosificación , Transporte Biológico/fisiología , Linfocitos T CD8-positivos/metabolismo , ADN/química , ADN/genética , Células Dendríticas/citología , Células Dendríticas/metabolismo , Interferón gamma/biosíntesis , Activación de Linfocitos , Metacrilatos/química , Nanopartículas , Plásmidos , Polímeros/síntesis química , Polímeros/química , Transfección/métodos , Vacunas de ADN/farmacología
18.
Polymer (Guildf) ; 52(4): 921-932, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21479119

RESUMEN

A new type of pH-labile cationic polymers, poly(ortho ester amidine) (POEAmd) copolymers, has been synthesized and characterized with potential future application as gene delivery carriers. The acid-labile POEAmd copolymer was synthesized by polycondensation of a new ortho ester diamine monomer with dimethylaliphatimidates, and a non-acid-labile polyamidine (PAmd) copolymer was also synthesized for comparison using a triethylene glycol diamine monomer. Both copolymers were easily dissolved in water, and can efficiently bind and condense plasmid DNA at neutral pH, forming nano-scale polyplexes. The physico-chemical properties of the polyplexes have been studied using dynamic light scattering, gel electrophoresis, ethidium bromide exclusion, and heparin competition. The average size of the polyplexes was dependent on the amidine: phosphate (N:P) ratio of the polymers to DNA. Polyplexes containing the acid-labile POEAmd or the non-acid-labile PAmd showed similar average particle size, comparable strength of condensing DNA, and resistance to electrostatic destabilization. They also share similar metabolic toxicity to cells as measured by MTT assay. Importantly, the acid-labile polyplexes undergo accelerated polymer degradation at mildly-acid-pHs, resulting in increasing particle size and the release of intact DNA plasmid. Polyplexes from both types of polyamidines caused distinct changes in the scattering properties of Baby Hamster Kidney (BHK-21) cells, showing swelling and increasing intracellular granularity. These cellular responses are uniquely different from other cationic polymers such as polyethylenimine and point to stress-related mechanisms specific to the polyamidines. Gene transfection of BHK-21 cells was evaluated by flow cytometry. The positive yet modest transfection efficiency by the polyamidines (acid-labile and non-acid-labile alike) underscores the importance of balancing polymer degradation and DNA release with endosomal escape. Insights gained from studying such acid-labile polyamidine-based DNA carriers and their interaction with cells may contribute to improved design of practically useful gene delivery systems.

19.
J Control Release ; 151(1): 18-27, 2011 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-21194551

RESUMEN

A new type of block copolymer micelles for pH-triggered delivery of poorly water-soluble anticancer drugs has been synthesized and characterized. The micelles were formed by the self-assembly of an amphiphilic diblock copolymer consisting of a hydrophilic poly(ethylene glycol) (PEG) block and a hydrophobic polymethacrylate block (PEYM) bearing acid-labile ortho ester side-chains. The diblock copolymer was synthesized by atom transfer radical polymerization (ATRP) from a PEG macro-initiator to obtain well-defined polymer chain-length. The PEG-b-PEYM micelles assumed a stable core-shell structure in aqueous buffer at physiological pH with a low critical micelle concentration as determined by proton NMR and pyrene fluorescence spectroscopy. The hydrolysis of the ortho ester side-chain at physiological pH was minimal yet much accelerated at mildly acidic pHs. Doxorubicin (Dox) was successfully loaded into the micelles at pH 7.4 and was released at a much higher rate in response to slight acidification to pH 5. Interestingly, the release of Dox at pH 5 followed apparently a biphasic profile, consisting of an initial fast phase of several hours followed by a sustained release period of several days. Dox loaded in the micelles was rapidly taken up by human glioma (T98G) cells in vitro, accumulating in the endolysosome and subsequently in the nucleus in a few hours, in contrast to the very low uptake of free drug at the same dose. The dose-dependent cytotoxicity of the Dox-loaded micelles was determined by the MTT assay and compared with that of the free Dox. While the empty micelles themselves were not toxic, the IC(50) values of the Dox-loaded micelles were approximately ten-times (by 24h) and three-times (by 48h) lower than the free drug. The much enhanced potency in killing the multi-drug-resistant human glioma cells by Dox loaded in the micelles could be attributed to high intracellular drug concentration and the subsequent pH-triggered drug release. These results establish the PEG-b-PEYM block copolymer with acid-labile ortho ester side-chains as a novel and effective pH-responsive nano-carrier for enhancing the delivery of drugs to cancer cells.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Preparaciones de Acción Retardada/química , Doxorrubicina/administración & dosificación , Glioma/tratamiento farmacológico , Polietilenglicoles/química , Ácidos Polimetacrílicos/química , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Humanos , Concentración de Iones de Hidrógeno , Micelas
20.
Macromol Biosci ; 10(2): 192-201, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19904722

RESUMEN

A new type of pH-responsive block copolymer nanoparticle has been synthesized and characterized. The amphiphilic diblock copolymer, PEG-b-PMYM, contains acid-labile ortho ester side-chains in the hydrophobic block and can self-assemble into micelle-like nanoparticles in water at neutral pH. Hydrolysis of the ortho ester side-chains follows a distinct exocyclic mechanism and shows pH-dependent kinetics, which triggers changes in nanoparticle size and morphology. The nanoparticles have been found to be non-toxic to cells in vitro. The ability to tune the size and morphology of biocompatible block copolymer nanoparticles by controlling the pH-sensitive side-chain hydrolysis represents a unique approach that may be exploited to improve the efficacy of nanometer-scale drug delivery.


Asunto(s)
Ésteres/química , Polímeros/química , Agua/química , Acrilamidas/química , Acrilamidas/toxicidad , Animales , Muerte Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Hidrólisis/efectos de los fármacos , Cinética , Luz , Espectroscopía de Resonancia Magnética , Ratones , Peso Molecular , Células 3T3 NIH , Nanopartículas/toxicidad , Nanopartículas/ultraestructura , Tamaño de la Partícula , Polietilenglicoles/química , Polietilenglicoles/toxicidad , Polímeros/toxicidad , Dispersión de Radiación , Espectrometría de Fluorescencia , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA