Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Pharm ; 12(1): 150-61, 2015 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25384057

RESUMEN

The applicability of small interfering RNA (siRNA) in future therapies depends on the availability of safe and efficient carrier systems. Ideally, siRNA delivery requires a system that is stable in the circulation but upon specific uptake into target cells can rapidly release its cargo into the cytoplasm. Previously, we evaluated a novel generation of carrier systems ("decationized" polyplexes) for DNA delivery, and it was shown that folate targeted decationized polyplexes had an excellent safety profile and showed intracellular triggered release upon cell specific uptake. Targeted decationized polyplexes consist of a core of disulfide cross-linked poly(hydroxypropyl methacrylamide) (pHPMA) stably entrapping nucleic acids and a shell of poly(ethylene glycol) (PEG) decorated with folate molecules. In the present study, the applicability of folate targeted decationized polyplexes for siRNA delivery was investigated. This required optimization of the carrier system particularly regarding the cross-linking density of the core of the polyplexes. Stable and nanosized siRNA decationized polyplexes were successfully prepared by optimizing the cross-link density of their core. Upon incubation in human plasma, a significant portion of siRNA remained entrapped in the decationized polyplexes as determined by fluorescence correlation spectroscopy (FCS). When tested in a folate receptor overexpressing cell line stably expressing luciferase, Skov3-luc, sequence specific gene silencing was observed. As expected, neither interference on the intrinsic luciferase expression nor on the cell metabolic activity (determined by XTT) was induced by the free-polymer or the siRNA polyplexes. In conclusion, targeted decationized polyplexes are safe and stable carriers that interact with the targeted cells and rapidly disassemble upon cell entry making them promising siRNA delivery systems.


Asunto(s)
Técnicas de Transferencia de Gen , Metacrilatos/química , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , Materiales Biocompatibles/química , Línea Celular Tumoral , Ácido Fólico/química , Silenciador del Gen , Humanos , Espectroscopía de Resonancia Magnética , Microscopía Electrónica de Transmisión , Nanotecnología , Ácidos Nucleicos/química , Polietilenglicoles/química , Polímeros/química , Espectrofotometría Ultravioleta
2.
Macromol Biosci ; 18(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29152858

RESUMEN

The in vivo stability and biodegradability of nanocarriers crucially determine therapeutic efficacy as well as safety when used for drug delivery. This study aims to evaluate optimized in vitro techniques predictive for in vivo nanocarrier behavior. Polymeric biodegradable nanogels based on hydroxyethyl methacrylamide-oligoglycolates-derivatized poly(hydroxyethyl methacrylamide-co-N-(2-azidoethyl)methacrylamide) and with various degrees of PEGylation and crosslinking densities are prepared. Three techniques are chosen and refined for specific in vitro evaluation of the nanocarrier performance: (1) fluorescence single particle tracking (fSPT) to study the stability of nanogels in human plasma, (2) tangential flow filtration (TFF) to study the degradation and filtration of nanogel degradation products, and (3) fluorescence fluctuation spectroscopy (FFS) to evaluate and compare the degradation behavior of nanogels in buffer and plasma. fSPT results demonstrate that nanogels with highest PEGylation content show the least aggregation. The TFF results reveal that nanogels with higher crosslink density have slower degradation and removal by filtration. FFS results indicate a similar degradation behavior in human plasma as compared to that in phosphate buffered saline. In conclusion, three methods can be used to compare and select the optimal nanogel composition, and these methods hold potential to predict the in vivo performance of nanocarriers.


Asunto(s)
Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Polietilenglicoles/química , Polietileneimina/química , Polímeros/química , Plásticos Biodegradables/química , Plásticos Biodegradables/farmacología , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos/farmacología , Humanos , Nanogeles , Nanopartículas/química , Nanopartículas/uso terapéutico , Plasma/efectos de los fármacos , Polietilenglicoles/farmacología , Polietilenglicoles/uso terapéutico , Polietileneimina/farmacología , Polietileneimina/uso terapéutico , Polímeros/farmacología , Polímeros/uso terapéutico , Espectrometría de Fluorescencia
3.
J Control Release ; 284: 73-83, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-29908893

RESUMEN

Potent adjuvants are highly demanded for most protein and peptides based vaccine candidates in clinical development. Recognition of viral single stranded (ss)RNA by innate toll-like receptors 7/8 in dendritic cells results in a cytokine environment supportive to the establishment of long lasting antibody responses and Th1 oriented T cell immunity. To fully exploit the immunestimulatory properties of ssRNA, it needs to be adequately formulated to ensure its optimal delivery to dendritic cells in the vaccine draining lymph nodes. In the present paper, we report on the design of ssRNA nanocomplexes formed by complexation of the cationic poly(carbonic acid 2-dimethylamino-ethyl ester 1-methyl-2-(2-methacryloylamino)-ethyl ester) (pHPMA-DMAE) based polymeric carrier and ssRNA. The resulting ssRNA nanocomplexes were subsequently PEGylated through copper-free click chemistry using PEG-bicyclo[6.1.0]nonyne (PEG-BCN) and cross-linked via disulfide bonds to increase their stability. The obtained near-neutral charged PEGylated ssRNA nanocomplexes (~150 nm) combined ssRNA protection with highly efficient delivery of ssRNA to DCs in the vaccine draining lymph nodes after subcutanuously administration. When co-administrated with a model antigen (soluble ovalbumin (OVA)), ssRNA nanocomplexes were far more efficient at inducing CD8 cytolytic T cells when compared to OVA co-adminstarted with naked ssRNA. Furthermore, IgG2c antibody titers, indicative of Th1 skewed T cell responses, were >10 times increased by complexing ssRNA into the PEGylated nanocomplexes. This study highlights the potential of post-functionalizing ssRNA nanocomplexes by copper-free click chemistry and these findings indcate that this potent ssRNA adjuvant may profoundly improve the efficacy of a variety of vaccines requiring Th1-type immunity.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Portadores de Fármacos/química , Polietilenglicoles/química , ARN/administración & dosificación , Linfocitos T Citotóxicos/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Animales , Formación de Anticuerpos/efectos de los fármacos , Reactivos de Enlaces Cruzados/química , Células Dendríticas/efectos de los fármacos , Femenino , Ganglios Linfáticos/efectos de los fármacos , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/farmacología , ARN/farmacología
4.
Adv Drug Deliv Rev ; 108: 13-24, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27422808

RESUMEN

Intraperitoneal (IP) drug delivery represents an attractive strategy for the local treatment of peritoneal carcinomatosis (PC). Over the past decade, a lot of effort has been put both in the academia and clinic in developing IP therapeutic approaches that maximize local efficacy while limiting systemic side effects. Also nanomedicines are under investigation for the treatment of tumors confined to the peritoneal cavity, due to their potential to increase the peritoneal retention and to target drugs to the tumor sites as compared to free drugs. Despite the progress reported by multiple clinical studies, there are no FDA approved drugs or formulations for specific use in the IP cavity yet. This review discusses the current clinical management of PC, as well as recent advances in nanomedicine-based IP delivery. We address important challenges to be overcome towards designing optimal nanocarriers for IP therapy in vivo.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanomedicina , Neoplasias Peritoneales/terapia , Femenino , Humanos , Embarazo
5.
Drug Deliv Transl Res ; 7(2): 241-251, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27631392

RESUMEN

Delivery of small interfering RNA (siRNA) is recently gaining tremendous attention for the treatment of ovarian cancer. The present study investigated the potential of different liposomal formulations composed of (2,3-dioleoyloxy-propyl)-trimethylammonium (DOTAP) and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) encapsulating siRNA (hydration method) for their ability to knockdown luciferase (Luc) activity in human ovarian cancer SKOV-3 cells. Fluorescence single particle tracking (fSPT) and fluorescence correlation spectroscopy (FCS) in human-undiluted ascites fluid obtained from a peritoneal carcinomatosis patient revealed that cationic hydra-lipoplexes (HYDRA-LPXs) and HYDRA-LPXs decorated with stable DSPE-PEG (DSPE HYDRA-LPXs) showed high stability during at least 24 h. HYDRA-LPXs decorated with sheddable C8 and C16 PEG-Ceramides (Cer HYDRA-LPXs) resulted in rapid and premature release of siRNA already in the first hours. Despite their role in preventing aggregation in vivo, liposomes decorated with stable PEG residues resulted in a poor transfection compared to the ones decorated with sheddable PEG residues in reduced serum conditions. Yet, the transfection efficiency of both Cer HYDRA-LPXs significantly decreased following 1 h of incubation in ascites fluid due to a drastic drop in the cellular uptake, while DSPE HYDRA-LPXs are still taken up by cells, but too stable to induce efficient gene silencing.


Asunto(s)
Ascitis , ARN Interferente Pequeño , Línea Celular Tumoral , Supervivencia Celular , Ácidos Grasos Monoinsaturados/química , Humanos , Liposomas , Luciferasas/genética , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Compuestos de Amonio Cuaternario/química , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química
6.
Biomaterials ; 138: 1-12, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28550752

RESUMEN

The rising antimicrobial resistance contributes to 25000 annual deaths in Europe. This threat to the public health can only be tackled if novel antimicrobials are developed, combined with a more precise use of the currently available antibiotics through the implementation of fast, specific, diagnostic methods. Nucleic acid mimics (NAMs) that are able to hybridize intracellular bacterial RNA have the potential to become such a new class of antimicrobials and additionally could serve as specific detection probes. However, an essential requirement is that these NAMs should be delivered into the bacterial cytoplasm, which is a particular challenge given the fact that they are charged macromolecules. We consider these delivery challenges in relation to the gastric pathogen Helicobacter pylori, the most frequent chronic infection worldwide. In particular, we evaluate if cationic fusogenic liposomes are suitable carriers to deliver NAMs across the gastric mucus barrier and the bacterial envelope. Our study shows that DOTAP-DOPE liposomes post-PEGylated with DSPE-PEG (DSPE Lpx) can indeed successfully deliver NAMs into Helicobacter pylori, while offering protection to the NAMs from binding and inactivation in gastric mucus isolated from pigs. DSPE Lpx thus offer exciting new possibilities for in vivo diagnosis and treatment of Helicobacter pylori infections.


Asunto(s)
Antiinfecciosos/administración & dosificación , Sistemas de Liberación de Medicamentos , Farmacorresistencia Microbiana , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/metabolismo , Moco/química , Oligonucleótidos Antisentido/administración & dosificación , ARN Bacteriano/antagonistas & inhibidores , ARN Ribosómico/antagonistas & inhibidores , Animales , Antiinfecciosos/síntesis química , Antiinfecciosos/metabolismo , Citoplasma/metabolismo , Ácidos Grasos Monoinsaturados/química , Colorantes Fluorescentes/química , Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Hibridación Fluorescente in Situ , Liposomas , Imitación Molecular , Moco/microbiología , Oligonucleótidos/administración & dosificación , Oligonucleótidos/síntesis química , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Oligonucleótidos Antisentido/síntesis química , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Compuestos de Amonio Cuaternario/química , ARN Bacteriano/genética , ARN Ribosómico/genética , Estómago/microbiología , Porcinos
7.
Macromol Biosci ; 17(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28614632

RESUMEN

Peritoneal carcinomatosis is a severe form of cancer in the abdomen, currently treated with cytoreductive surgery and intravenous chemotherapy. Recently, nebulization has been proposed as a less invasive strategy for the local delivery of chemotherapeutic drugs. Also, RNA interference has been considered as a potential therapeutic approach for treatment of cancer. In this study, Lipofectamine RNAiMAX/siRNA complexes and cyclodextrin/siRNA complexes are evaluated before and after nebulization. Nebulization of the siRNA complexes does not significantly lower transfection efficiency when compared to non-nebulized complexes. After incubation in ascites fluid, however, the cyclodextrin/siRNA complexes show a drastic decrease in transfection efficiency. For the Lipofectamine RNAiMAX/siRNA complexes, this decrease is less pronounced. It is concluded that nebulization is an interesting technique to distribute siRNA complexes into the peritoneal cavity, providing the complexes are stable in ascites fluid which might be present in the peritoneal cavity.


Asunto(s)
Ciclodextrinas/química , Lípidos/química , ARN Interferente Pequeño/genética , Transfección/métodos , Aerosoles , Ascitis/genética , Ascitis/metabolismo , Ascitis/terapia , Línea Celular Tumoral , Femenino , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/terapia , Plásmidos/química , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico
8.
ACS Appl Mater Interfaces ; 9(3): 2181-2195, 2017 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-28029254

RESUMEN

Guanidine and morpholine functionalized aliphatic polycarbonate polymers are able to deliver efficiently histone deacetylase 5 (HDAC5) siRNA into the cytoplasm of cancer cells in vitro leading to a decrease of cell proliferation were previously developed. To allow these biodegradable and biocompatible polyplex nanoparticles to overcome the extracellular barriers and be effective in vivo after an intravenous injection, polyethylene glycol chains (PEG750 or PEG2000) were grafted on the polymer structure. These nanoparticles showed an average size of about 150 nm and a slightly positive ζ-potential with complete siRNA complexation. Behavior of PEGylated and non-PEGylated polyplexes were investigated in the presence of serum, in terms of siRNA complexation (fluorescence correlation spectroscopy), size (dynamic light scattering and single-particle tracking), interaction with proteins (isothermal titration calorimetry) and cellular uptake. Surprisingly, both PEGylated and non-PEGylated formulations presented relatively good behavior in the presence of fetal bovine serum (FBS). Hemocompatibility tests showed no effect of these polyplexes on hemolysis and coagulation. In vivo biodistribution in mice was performed and showed a better siRNA accumulation at the tumor site for PEGylated polyplexes. However, cellular uptake in protein-rich conditions showed that PEGylated polyplex lost their ability to interact with biological membranes and enter into cells, showing the importance to perform in vitro investigations in physiological conditions closed to in vivo situation. In vitro, the efficiency of PEGylated nanoparticles decreases compared to non-PEGylated particles, leading to the loss of the antiproliferative effect on cancer cells.


Asunto(s)
Nanopartículas , Administración Intravenosa , Animales , Histona Desacetilasas , Ratones , Neoplasias , Cemento de Policarboxilato , Polietilenglicoles , ARN Interferente Pequeño , Distribución Tisular
9.
J Control Release ; 236: 68-78, 2016 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-27329774

RESUMEN

Topical vaginal sustained delivery of siRNA presents a significant challenge due to the short residence time of formulations. Therefore, a drug delivery system capable to adhere to the vaginal mucosa is desirable, as it could allow a prolonged delivery and increase the effectiveness of the therapy. The aim of this project is to develop a polymeric solid mucoadhesive system, loaded with lipoplexes, able to be progressively rehydrated by the vaginal fluids to form a hydrogel and to deliver siRNA to vaginal tissues. To minimize adhesive interactions with vaginal mucus components, lipoplexes were coated with different derivatives of polyethylene glycol: DPSE-PEG2000, DPSE-PEG750 and ceramide-PEG2000. Based on stability and diffusion properties in simulated vaginal fluids, lipoplexes containing DSPE-PEG2000 were selected and incorporated in hydroxyethyl cellulose (HEC) hydrogels. Solid systems, called sponges, were then obtained by freeze-drying. Sponges meet acceptable mechanical characteristics and their hardness, deformability and mucoadhesive properties are not influenced by the presence of lipoplexes. Finally, mobility and stability of lipoplexes inside sponges rehydrated with vaginal mucus, mimicking in situ conditions, were evaluated by advanced fluorescence microscopy. The release rate was found to be influenced by the HEC concentration and consequently by the viscosity after rehydration. This study demonstrates the feasibility of entrapping pegylated lipoplexes into a solid matrix system for a prolonged delivery of siRNA into the vagina.


Asunto(s)
Liposomas/química , Membrana Mucosa/efectos de los fármacos , Polietilenglicoles/química , ARN Interferente Pequeño/química , Vagina/efectos de los fármacos , Adhesividad , Administración Intravaginal , Celulosa/análogos & derivados , Celulosa/química , Ceramidas/química , Química Farmacéutica , Femenino , Liofilización , Técnicas de Transferencia de Gen , Humanos , Hidrogeles , Nanopartículas/química , Tamaño de la Partícula , Fosfatidiletanolaminas/química , ARN Interferente Pequeño/administración & dosificación , Propiedades de Superficie
10.
Int J Pharm ; 511(2): 1112-23, 2016 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-27511710

RESUMEN

Active drug targeting and controlled release of hydrophilic macromolecular drugs represent crucial points in designing efficient polymeric drug delivery nanoplatforms. In the present work EGFR-targeted polylactide-co-glycolide (PLGA) nanoparticles were made by a blend of two different PLGA-based polymers. The first, GE11-PLGA, in which PLGA was functionalized with GE11, a small peptide and EGFR allosteric ligand, able to give nanoparticles selective targeting features. The second polymer was a PEGylated PLGA (PEG-PLGA) aimed at improving nanoparticles hydrophilicity and stealth features. GE11 and GE11-PLGA were custom synthetized through a simple and inexpensive method. The nanoprecipitation technique was exploited for the preparation of polymeric nanoparticles composed by a 1:1weight ratio between GE11-PLGA and PEG-PLGA, obtaining smart nanoplatforms with proper size for parenteral administration (143.9±5.0nm). In vitro cellular uptake in EGFR-overexpressing cell line (A549) demonstrated an active internalization of GE11-functionalized nanoparticles. GE11-PLGA/PEG-PLGA blend nanoparticles were loaded with Myoglobin, a model hydrophilic macromolecule, reaching a good loading (2.42% respect to the theoretical 4.00% w/w) and a prolonged release over 60days. GE11-PLGA/PEG-PLGA blend nanoparticles showed good in vitro stability for 30days in physiological saline solution at 4°C and for 24h in pH 7.4 or pH 5.0 buffer at 37°C respectively, giving indications about potential storage and administration conditions. Furthermore ex vivo stability study in human plasma using fluorescence Single Particle Tracking (fSPT) assessed good GE11-PLGA/PEG-PLGA nanoparticles dimensional stability after 1 and 4h. Thanks to the versatility in polymeric composition and relative tunable nanoparticles features in terms of drug incorporation and release, GE11-PLGA/PEG-PLGA blend NPs can be considered highly promising as smart nanoparticulate platforms for the treatment of diseases characterized by EGFR overexpression by parenteral administration .


Asunto(s)
Diseño de Fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Ácido Láctico/síntesis química , Nanopartículas/química , Péptidos/síntesis química , Polietilenglicoles/síntesis química , Ácido Poliglicólico/síntesis química , Células A549 , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/fisiología , Química Farmacéutica , Humanos , Infusiones Parenterales , Ácido Láctico/administración & dosificación , Sustancias Macromoleculares/administración & dosificación , Sustancias Macromoleculares/síntesis química , Nanopartículas/administración & dosificación , Péptidos/administración & dosificación , Polietilenglicoles/administración & dosificación , Ácido Poliglicólico/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
11.
ACS Appl Mater Interfaces ; 7(43): 24322-9, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26470057

RESUMEN

Small interfering RNA (siRNA) offers a great potential for the treatment of various diseases and disorders. Nevertheless, inefficient in vivo siRNA delivery hampers its translation into the clinic. While numerous successful in vitro siRNA delivery stories exist in reduced-protein conditions, most studies so far overlook the influence of the biological fluids present in the in vivo environment. In this study, we compared the transfection efficiency of liposomal formulations in Opti-MEM (low protein content, routinely used for in vitro screening) and human undiluted ascites fluid obtained from a peritoneal carcinomatosis patient (high protein content, representing the in vivo situation). In Opti-MEM, all formulations are biologically active. In ascites fluid, however, the biological activity of all lipoplexes is lost except for lipofectamine RNAiMAX. The drop in transfection efficiency was not correlated to the physicochemical properties of the nanoparticles, such as premature siRNA release and aggregation of the nanoparticles in the human ascites fluid. Remarkably, however, all of the formulations except for lipofectamine RNAiMAX lost their ability to be taken up by cells following incubation in ascites fluid. To take into account the possible effects of a protein corona formed around the nanoparticles, we recommend always using undiluted biological fluids for the in vitro optimization of nanosized siRNA formulations next to conventional screening in low-protein content media. This should tighten the gap between in vitro and in vivo performance of nanoparticles and ensure the optimal selection of nanoparticles for further in vivo studies.


Asunto(s)
Ascitis/metabolismo , Liposomas/química , Nanopartículas/química , ARN Interferente Pequeño/metabolismo , Línea Celular Tumoral , Femenino , Terapia Genética/métodos , Humanos , Lípidos/química , Nanotecnología/métodos , Metástasis de la Neoplasia , Neoplasias Ováricas/metabolismo , Tamaño de la Partícula , Proteínas/química , Interferencia de ARN , Espectrometría de Fluorescencia , Transfección
12.
Mol Ther Nucleic Acids ; 4: e269, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26645765

RESUMEN

Helicobacter pylori infects more than 50% of the worldwide population. It is mostly found deep in the gastric mucus lining of the stomach, being a major cause of peptic ulcers and gastric adenocarcinoma. To face the increasing resistance of H. pylori to antibiotics, antimicrobial nucleic acid mimics are a promising alternative. In particular, locked nucleic acids (LNA)/2'-OMethyl RNA (2'OMe) have shown to specifically target H. pylori, as evidenced by in situ hybridization. The success of in vivo hybridization depends on the ability of these nucleic acids to penetrate the major physical barriers-the highly viscoelastic gastric mucus and the bacterial cell envelope. We found that LNA/2'OMe is capable of diffusing rapidly through native, undiluted, gastric mucus isolated from porcine stomachs, without degradation. Moreover, although LNA/2'OMe hybridization was still successful without permeabilization and fixation of the bacteria, which is normally part of in vitro studies, the ability of LNA/2'OMe to efficiently hybridize with H. pylori was hampered by the presence of mucus. Future research should focus on developing nanocarriers that shield LNA/2'OMe from components in the gastric mucus, while remaining capable of diffusing through the mucus and delivering these nucleic acid mimics directly into the bacteria.

13.
Structure ; 22(4): 636-45, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24613488

RESUMEN

Our understanding of protein evolution would greatly benefit from mapping of binding landscapes, i.e., changes in protein-protein binding affinity due to all single mutations. However, experimental generation of such landscapes is a tedious task due to a large number of possible mutations. Here, we use a simple computational protocol to map the binding landscape for two homologous high-affinity complexes, involving a snake toxin fasciculin and acetylcholinesterase from two different species. To verify our computational predictions, we experimentally measure binding between 25 Fas mutants and the 2 enzymes. Both computational and experimental results demonstrate that the Fas sequence is close to the optimum when interacting with its targets, yet a few mutations could further improve Kd, kon, and koff. Our computational predictions agree well with experimental results and generate distributions similar to those observed in other high-affinity PPIs, demonstrating the potential of simple computational protocols in capturing realistic binding landscapes.


Asunto(s)
Acetilcolinesterasa/química , Inhibidores de la Colinesterasa/química , Venenos Elapídicos/química , Mapeo Peptídico/estadística & datos numéricos , Acetilcolinesterasa/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Termodinámica , Torpedo
14.
Acta Biomater ; 10(7): 2965-75, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24657672

RESUMEN

Intraperitoneal (IP) administration of nano-sized delivery vehicles containing small interfering RNA (siRNA) has recently gained attention as an alternative route for the efficient treatment of peritoneal carcinomatosis. The colloidal stability of nanomatter following IP administration has, however, not been thoroughly investigated yet. Here, enabled by advanced microscopy methods such as single particle tracking and fluorescence correlation spectroscopy, we follow the aggregation and cargo release of nano-scaled systems directly in peritoneal fluids from healthy mice and ascites fluid from a patient diagnosed with peritoneal carcinomatosis. The colloidal stability in the peritoneal fluids was systematically studied as a function of the charge (positive or negative) and poly(ethylene glycol) (PEG) degree of liposomes and polystyrene nanoparticles, and compared to human serum. Our data demonstrate strong aggregation of cationic and anionic nanoparticles in the peritoneal fluids, while only slight aggregation was observed for the PEGylated ones. PEGylated liposomes, however, lead to a fast and premature release of siRNA cargo in the peritoneal fluids. Based on our observations, we reflect on how to tailor improved delivery systems for IP therapy.


Asunto(s)
Líquido Ascítico/química , Coloides , Sistemas de Liberación de Medicamentos , Nanopartículas , Animales , Infusiones Parenterales , Liposomas , Ratones
15.
J Control Release ; 195: 162-175, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25204289

RESUMEN

Many polycation-based gene delivery vectors show high transfection in vitro, but their cationic nature generally leads to significant toxicity and poor in vivo performance which significantly hampers their clinical applicability. Unlike conventional polycation-based systems, decationized polyplexes are based on hydrophilic and neutral polymers. They are obtained by a 3-step process: charge-driven condensation followed by disulfide crosslinking stabilization and finally polyplex decationization. They consist of a disulfide-crosslinked poly(hydroxypropyl methacrylamide) (pHPMA) core stably entrapping plasmid DNA (pDNA), surrounded by a shell of poly(ethylene glycol) (PEG). In the present paper the applicability of decationized polyplexes for systemic administration was evaluated. Cy5-labeled decationized polyplexes were evaluated for stability in plasma by fluorescence single particle tracking (fSPT), which technique showed stable size distribution for 48 h unlike its cationic counterpart. Upon the incubation of the polymers used for the formation of polyplexes with HUVEC cells, MTT assay showed excellent cytocompatibility of the neutral polymers. The safety was further demonstrated by a remarkable low teratogenicity and mortality activity of the polymers in a zebrafish assay, in great contrast with their cationic counterpart. Near infrared (NIR) dye-labeled polyplexes were evaluated for biodistribution and tumor accumulation by noninvasive optical imaging when administered systemically in tumor bearing mice. Decationized polyplexes exhibited an increased circulation time and higher tumor accumulation, when compared to their cationic precursors. Histology of tumors sections showed that decationized polyplexes induced reporter transgene expression in vivo. In conclusion, decationized polyplexes are a platform for safer polymeric vectors with improved biodistribution properties when systemically administered.


Asunto(s)
ADN/administración & dosificación , Técnicas de Transferencia de Gen , Neoplasias/metabolismo , Polímeros/administración & dosificación , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , ADN/química , ADN/farmacocinética , ADN/toxicidad , Estabilidad de Medicamentos , Embrión no Mamífero/efectos de los fármacos , Femenino , Proteínas Fluorescentes Verdes/genética , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Ratones Desnudos , Tamaño de la Partícula , Plásmidos , Polímeros/química , Polímeros/farmacocinética , Polímeros/toxicidad , Distribución Tisular , Pez Cebra/embriología
16.
Int J Pharm ; 450(1-2): 241-9, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23623794

RESUMEN

Bolaamphiphiles - amphiphilic molecules consisting of two hydrophilic headgroups linked by a hydrophobic chain - form highly stable vesicles consisting of a monolayer membrane that can be used as vehicles to deliver drugs across biological membranes, particularly the blood-brain barrier (BBB). We prepared new vesicles comprising bolaamphiphiles (bolavesicles) that encapsulate iron oxide nanoparticles (IONPs) and investigated their suitability for targeted drug delivery. Bolavesicles displaying different headgroups were studied, and the effect of IONP encapsulation upon membrane interactions and cell uptake were examined. Experiments revealed more pronounced membrane interactions of the bolavesicles assembled with IONPs. Furthermore, enhanced internalization and stability of the IONP-bolavesicles were observed in b.End3 brain microvessel endothelial cells - an in vitro model of the blood-brain barrier. Our findings indicate that embedded IONPs modulate bolavesicles' physicochemical properties, endow higher vesicle stability, and enhance their membrane permeability and cellular uptake. IONP-bolavesicles thus constitute a promising drug delivery platform, potentially targeted to the desired location using external magnetic field.


Asunto(s)
Portadores de Fármacos/química , Furanos/química , Nanopartículas de Magnetita/química , Piridonas/química , Animales , Línea Celular , Portadores de Fármacos/administración & dosificación , Endocitosis , Furanos/administración & dosificación , Membrana Dobles de Lípidos/metabolismo , Fenómenos Magnéticos , Nanopartículas de Magnetita/administración & dosificación , Ratones , Piridonas/administración & dosificación
17.
J Control Release ; 160(2): 315-21, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22261280

RESUMEN

Bolaamphiphilic cationic vesicles with acetylcholine (ACh) surface groups were investigated for their ability to deliver a model protein-bovine serum albumin conjugated to fluorescein isothiocyanate (BSA-FITC) across biological barriers in vitro and in vivo. BSA-FITC-loaded vesicles were internalized into cells in culture, including brain endothelial b.End3 cells, at 37 °C, but not at 4 °C, indicating an active uptake process. To examine if BSA-FITC-loaded vesicles were stable enough for in vivo delivery, we tested vesicle stability in whole serum. The half-life of cationic BSA-FITC-loaded vesicles with ACh surface groups that are hydrolyzed by choline esterase (ChE) was about 2 h, whereas the half-life of vesicles with similar surface groups, but which are not hydrolyzed by choline esterase (ChE), was over 5 h. Pyridostigmine, a choline esterase inhibitor that does not penetrate the blood-brain barrier (BBB), increased the stability of the ChE-sensitive vesicles to 6 h but did not affect the stability of vesicles with ACh surface groups that are not hydrolyzed by ChE. Following intravenous administration to pyridostigmine-pretreated mice, BSA-FITC encapsulated in ChE-sensitive vesicles was distributed into various tissues with marked accumulation in the brain, whereas non-encapsulated (free) BSA-FITC was detected only in peripheral tissues, but not in the brain. These results show that cationic bolaamphiphilic vesicles with ACh head groups are capable of delivering proteins across biological barriers, such as the cell membrane and the blood-brain barrier (BBB). Brain ChE activity destabilizes the vesicles and releases the encapsulated protein, enabling its accumulation in the brain.


Asunto(s)
Encéfalo/metabolismo , Portadores de Fármacos/química , Furanos/química , Nanopartículas/química , Piridonas/química , Albúmina Sérica Bovina/administración & dosificación , Animales , Encéfalo/irrigación sanguínea , Bovinos , Composición de Medicamentos , Estabilidad de Medicamentos , Células Endoteliales/metabolismo , Humanos , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos ICR , Albúmina Sérica Bovina/farmacocinética , Distribución Tisular
18.
Int J Pharm ; 434(1-2): 20-7, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22643027

RESUMEN

In vitro safety assessment of disposable medical devices, including infusion sets, is usually performed using L-929 mouse keratinocytes. However, cells of different origin (endothelial, lymphoid and myeloid cells) are also exposed to infusion sets' extractables during their clinical use. We studied whether the cEND mouse brain endothelial cells can be suitable for in vitro safety assessment of infusion sets. We analyzed infusion sets from different manufacturers that varied in design and storage time. cEND cells were incubated with extracts of individual parts of the infusion sets (tube, cup, latex), and relative toxicities were analyzed using MTT test, DCFH-DA-based analysis of reactive oxygen species formation, apoptosis and cell cycle analyses. We identified a pattern of yellowing of the infusion sets upon storage and revealed that it originated from the latex part. Extracts of the individual parts of the infusion sets, primarily of the latex, were toxic to the cEND cells leading to induction of apoptosis and cell death. We conclude that infusion sets release extractables that can be toxic to the endothelial cells of the patients that receive infusion. We suggest to use cEND cells for in vitro safety assessment of infusion sets and other medical devices that release extractables to the bloodstream.


Asunto(s)
Células Endoteliales/metabolismo , Bombas de Infusión/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Pruebas de Toxicidad/métodos , Animales , Apoptosis , Encéfalo/citología , Encéfalo/metabolismo , Ciclo Celular , Línea Celular , Equipos Desechables , Seguridad de Equipos , Látex/química , Látex/toxicidad , Ratones , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA