Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Control Release ; 367: 76-92, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38262488

RESUMEN

Glucose oxidase (GOx)-based enzyme therapeutics are potential alternatives for colorectal cancer (CRC) treatment via glucose consumption and accumulation of hydrogen peroxide (H2O2). Given that H2O2 can be eliminated by cytoprotective autophagy, autophagy inhibitors that can interrupt autolysosome-induced H2O2 elimination are promising combination drugs of GOx. Here, we developed a multifunctional biomimetic nanocarrier for effective co-delivery of an autophagy inhibitor-chloroquine phosphate (CQP) and GOx to exert their synergistic effect by irreversibly upregulating intracellular reactive oxygen species (ROS) levels. Poly (D, l-lactide-co-glycolide) (PLGA) nanoparticles (NPs) were used to encapsulate both GOx and CQP using a W/O/W multi-emulsion method. Calcium phosphate (CaP) was used to "fix" CQP to GOx in the internal water phase, where it served as a pH-sensitive unit to facilitate intracellular drug release. Folic acid-modified red blood cell membranes (FR) were used to camouflage the GOx/CQP/CaP encapsulated PLGA NPs (referred to as PLGA/GCC@FR). In an AOM/DSS-induced CRC mouse model, PLGA/GCC@FR exhibited improved antitumor effects, in which the number of tumor nodes were only a quarter of that in the free drug combination group. The enhanced therapeutic effects of PLGA/GCC@FR were attributed to the prolonged tumor retention which was verified by both dynamic in vivo imaging and drug biodistribution. This multifunctional biomimetic nanocarrier facilitated combined enzyme therapeutics by depleting glucose and augmenting intracellular ROS levels in tumor cells, which exerted a synergistic inhibitory effect on tumor growth. Therefore, this study proposed a novel strategy for the enhancement of combined enzyme therapeutics, which provided a promising method for effective CRC treatment.


Asunto(s)
Neoplasias Colorrectales , Nanopartículas , Neoplasias , Animales , Ratones , Óxidos , Glucosa/metabolismo , Biomimética , Peróxido de Hidrógeno/metabolismo , Especies Reactivas de Oxígeno , Distribución Tisular , Neoplasias/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Terapia Enzimática , Neoplasias Colorrectales/tratamiento farmacológico , Glucosa Oxidasa , Línea Celular Tumoral
2.
Nano Lett ; 23(23): 10971-10982, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-37991895

RESUMEN

Nanoparticles have emerged as potential transporters of drugs targeting Alzheimer's disease (AD), but their design should consider the blood-brain barrier (BBB) integrity and neuroinflammation of the AD brain. This study presents that aging is a significant factor for the brain localization and retention of nanoparticles, which we engineered to bind with reactive astrocytes and activated microglia. We assembled 200 nm-diameter particles using a block copolymer of poly(lactic-co-glycolic acid) (PLGA) and CD44-binding hyaluronic acid (HA). The resulting PLGA-b-HA nanoparticles displayed increased binding to CD44-expressing reactive astrocytes and activated microglia. Upon intravascular injection, nanoparticles were localized to the hippocampi of both APP/PS1 AD model mice and their control littermates at 13-16 months of age due to enhanced transvascular transport through the leaky BBB. No particles were found in the hippocampi of young adult mice. These findings demonstrate the brain localization of nanoparticles due to aging-induced BBB breakdown regardless of AD pathology.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas , Ratones , Animales , Enfermedad de Alzheimer/metabolismo , Ratones Transgénicos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo
3.
Macromol Biosci ; 23(9): e2300181, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37399543

RESUMEN

Pure positive electrostatic charges (PPECs) show suppressive effect on the proliferation and metabolism of invasive cancer cells without affecting normal tissues. PPECs are used for the delivery of drug-loaded polymeric nanoparticles (DLNs) capped with negatively charged poly(lactide-co-glycolide) (PLGA) and Poly(vinyl-alcohol) PVA into the tumor site of mouse models. The charged patch is installed on top of the skin in the mouse models' tumor region, and the controlled selective release of the drug is assayed by biochemical, radiological, and histological experiments on both tumorized models and normal rats' livers. It is found that DLNs synthesized by PLGA show great attraction to PPECs due to their stable negative charges, which would not degrade immediately in blood. The burst and drug release after less than 48h of this synthesized DLNs are 10% and 50%, respectively. These compounds can deliver the loaded-drug into the tumor site with the assistance of PPECs, and the targeted-retarded release will take place. Hence, local therapy can be achieved with much lower drug concentration (conventional chemotherapy [2 mg kg-1 ] versus DLNs-based chemotherapy [0.75 mg kg-1 ]) with negligible side effects in non-targeted organs. PPECs have many potential clinical applications for advanced-targeted chemotherapy with the lowest discernible side effects.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Ratones , Ratas , Animales , Sistemas de Liberación de Medicamentos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Electricidad Estática , Antineoplásicos/química , Polímeros/uso terapéutico , Neoplasias/tratamiento farmacológico , Nanopartículas/química , Portadores de Fármacos/química , Liberación de Fármacos
4.
Phytochem Anal ; 34(8): 950-958, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37463671

RESUMEN

OBJECTIVES: Oxidative stress is one of the carcinogenic mechanisms underlying the development of glioblastoma multiforme (GBM), a highly aggressive brain tumor type associated with poor prognosis. Curcumin is known to be an efficient antioxidant, anti-inflammatory, and anticancer compound. However, its poor solubility in water, inappropriate pharmacokinetics, and low bioavailability limit its use as an antitumor drug. We prepared PLGA-based curcumin nanoparticles changed with folic acid and chitosan (curcumin-PLGA-CS-FA) and evaluated its effects on GBM tumor cells' redox status. METHODS: The nanoprecipitation method was used to synthesize CU nanoparticles (CU-NPs). The size, morphology, and stability were characterized by DLS, SEM, and zeta potential analysis, respectively. The CU-NPs' toxic properties were studied by MTT assay and measuring the intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) concentrations. The study was completed by measuring the gene expression levels and activity of superoxide dismutase, catalase, glutaredoxin, and thioredoxin antioxidant enzymes. RESULTS: The size, polydispersity index, and zeta potential of CU-NPs were 77.27 nm, 0.29, and -22.45 mV, respectively. The encapsulation efficiency was approximately 98%. Intracellular ROS and MDA levels decreased after CU-NP treatment. Meanwhile, the CU-NPs increased gene expression and activity of superoxide dismutase, catalase, glutaredoxin, and thioredoxin antioxidant enzymes. CONCLUSION: CU-NPs might be effective in the prevention and treatment of glioblastoma cancer by modulating the antioxidant-oxidant balance.


Asunto(s)
Quitosano , Curcumina , Glioblastoma , Nanopartículas , Curcumina/farmacología , Curcumina/uso terapéutico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/uso terapéutico , Glioblastoma/tratamiento farmacológico , Catalasa , Quitosano/metabolismo , Quitosano/uso terapéutico , Glutarredoxinas/metabolismo , Glutarredoxinas/uso terapéutico , Antioxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/uso terapéutico , Ácido Fólico/uso terapéutico , Oxidación-Reducción , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/uso terapéutico , Tiorredoxinas/metabolismo , Tiorredoxinas/uso terapéutico
5.
J Control Release ; 359: 257-267, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37290723

RESUMEN

The clinical application of EDV, a potent antioxidant drug approved for amyotrophic lateral sclerosis (ALS), is limited by its short biological half-life and poor water solubility necessitating hospitalization during intravenous infusion. Nanotechnology-based drug delivery constitutes a powerful tool through inferring drug stability and targeted drug delivery improving drug bioavailability at the diseased site. Nose-to-brain drug delivery offers direct access to the brain bypassing the blood brain barrier and reducing systemic biodistribution. In this study, we designed EDV-loaded poly(lactic-co-glycolic acid) (PLGA)-based polymeric nanoparticles (NP-EDV) for intranasal administration. NPs were formulated by the nanoprecipitation method. Morphology, EDV loading, physicochemical properties, shelf-life stability, in vitro release and pharmacokinetic assessment in mice were conducted. EDV was efficiently loaded into ∼90 nm NPs, stable up to 30 days of storage, at ∼3% drug loading. NP-EDV reduced H2O2-induced oxidative stress toxicity in mouse microglial cell line BV-2. Optical imaging and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) showed that intranasal delivery of NP-EDV offered higher and more sustained brain uptake of EDV compared to intravenous administration. This study is the first of its kind to develop an ALS drug in a nanoparticulate formulation for nose-to-brain delivery raising hope to ALS patients where currently treatment options are limited to two clinically approved drugs only.


Asunto(s)
Esclerosis Amiotrófica Lateral , Nanopartículas , Ratones , Animales , Administración Intranasal , Edaravona/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Portadores de Fármacos/química , Disponibilidad Biológica , Distribución Tisular , Cromatografía Liquida , Peróxido de Hidrógeno/metabolismo , Espectrometría de Masas en Tándem , Encéfalo/metabolismo , Sistemas de Liberación de Medicamentos , Tamaño de la Partícula
6.
Adv Sci (Weinh) ; 10(19): e2301107, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37097746

RESUMEN

Cancer vaccine has been considered as a promising immunotherapy by inducing specific anti-tumor immune response. Rational vaccination at suitable time to efficiently present tumor associated antigen will boost tumor immunity and is badly needed. Here, a poly (lactic-co-glycolic acid) (PLGA)-based cancer vaccine of nanoscale is designed, in which engineered tumor cell membrane proteins, mRNAs, and sonosensitizer chlorin e6 (Ce6) are encapsulated at high efficiency. The nanosized vaccine can be efficiently delivered into antigen presentation cells (APCs) in lymph nodes after subcutaneous injection. In the APCs, the encapsulated cell membrane and RNA from engineered cells, which have disturbed splicing resembling the metastatic cells, provide neoantigens of metastatic cancer in advance. Moreover, the sonosensitizer Ce6 together with ultrasound irradiation promotes mRNA escape from endosome, and augments antigen presentation. Through 4T1 syngeneic mouse model, it has been proved that the proposed nanovaccine is efficient to elicit antitumor immunity and thus prevent cancer metastasis.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Animales , Ratones , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Células Dendríticas , ARN/metabolismo , Neoplasias/terapia , Neoplasias/metabolismo , Presentación de Antígeno , Membrana Celular/metabolismo , Antígenos de Neoplasias/metabolismo
7.
ACS Biomater Sci Eng ; 9(3): 1672-1681, 2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36796355

RESUMEN

Osteoarthritis (OA) is an inflammatory disease accompanied by synovial joint inflammation, and IL-36 plays an important role in this process. Local application of IL-36 receptor antagonist (IL-36Ra) can effectively control the inflammatory response, thereby protecting cartilage and slowing down the development of OA. However, its application is limited by the fact that it is rapidly metabolized locally. We designed and prepared a temperature-sensitive poly(lactic-co-glycolic acid)-poly(ethylene glycol)-poly(lactic-co-glycolic acid) (PLGA-PEG-PLGA) hydrogel (IL-36Ra@Gel) system carrying IL-36Ra and evaluated its basic physicochemical characteristics. The drug release curve of IL-36Ra@Gel indicated that this system could slowly release the drug over a longer period. Furthermore, degradation experiments showed that it could be largely degraded from the body within 1 month. The biocompatibility-related results showed that it had no significant effect on cell proliferation compared to the control group. In addition, the expression of MMP-13 and ADAMTS-5 was lower in IL-36Ra@Gel-treated chondrocytes than in the control group, and the opposite results appeared in aggrecan and collagen X. After 8 weeks of treatment with IL-36Ra@Gel by joint cavity injection, HE and Safranin O/Fast green staining showed that the degree of cartilage tissue destruction in the IL-36Ra@Gel-treated group was less than those in other groups. Meanwhile, the joints of mice in the IL-36Ra@Gel group had the most intact cartilage surface, the smallest thickness of cartilage erosion, and the lowest OARSI and Mankins score among all groups. Consequently, the combination of IL-36Ra and PLGA-PLEG-PLGA temperature-sensitive hydrogels can greatly improve the therapeutic effect and prolong the drug duration time, thus effectively delaying the progression of degenerative changes in OA, providing a new feasible nonsurgical treatment for OA.


Asunto(s)
Hidrogeles , Osteoartritis , Ratones , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/uso terapéutico , Hidrogeles/metabolismo , Temperatura , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Condrocitos/metabolismo
8.
Int J Pharm ; 632: 122573, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36592892

RESUMEN

Polymeric nanoparticles are widely used drug delivery systems for cancer treatment due to their properties such as ease of passing through biological membranes, opportunity to modify drug release, specifically targeting drugs to diseased areas, and potential of reducing side effects. Here, we formulated irinotecan and Stattic co-loaded PLGA nanoparticles targeted to small cell lung cancer. Nanoparticles were successfully conjugated with CD56 antibody with a conjugation efficiency of 84.39 ± 1.01%, and characterization of formulated nanoparticles was conducted with in-vitro and in-vivo studies. Formulated particles had sizes in the range of 130-180 nm with PDI values smaller than 0.3. Encapsulation and active targeting of irinotecan and Stattic resulted in increased cytotoxicity and anti-cancer efficiency in-vitro. Furthermore, it was shown with ex-vivo biodistribution studies that conjugated nanoparticles were successfully targeted to CD56-expressing SCLC cells and distributed mainly to tumor tissue and lungs. Compliant with our hypothesis and literature, the STAT3 pathway was successfully inhibited with Stattic solution and Stattic loaded nanoparticles. Additionally, intravenous injection of conjugated co-loaded nanoparticles resulted in decreased side effects and better anti-tumor activity than individual solutions of drugs in SCLC tumor-bearing mice. These results may indicate a new treatment option for clinically aggressive small cell lung cancer.


Asunto(s)
Neoplasias Pulmonares , Nanopartículas , Carcinoma Pulmonar de Células Pequeñas , Ratones , Animales , Irinotecán , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Distribución Tisular , Línea Celular Tumoral , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Portadores de Fármacos/uso terapéutico
9.
ACS Nano ; 17(3): 2341-2355, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36688797

RESUMEN

Immunotherapy has had a profound positive effect on certain types of cancer but has not improved the outcomes of glioma because of the blood-brain barrier (BBB) and immunosuppressive tumor microenvironment. In this study, we developed an activated mature dendritic cell membrane (aDCM)-coated nanoplatform, rapamycin (RAPA)-loaded poly(lactic-co-glycolic acid) (PLGA), named aDCM@PLGA/RAPA, which is a simple, efficient, and individualized strategy to cross the BBB and improve the immune microenvironment precisely. In vitro cells uptake and the transwell BBB model revealed that the aDCM@PLGA/RAPA can enhance homotypic-targeting and BBB-crossing efficiently. According to the in vitro and in vivo immune response efficacy of aDCM@PLGA/RAPA, the immature dendritic cells (DCs) could be stimulated into the matured status, which leads to further activation of immune cells, such as tumor-infiltrating T cells and natural killer cells, and can induce the subsequent immune responses through direct and indirect way. The aDCM@PLGA/RAPA treatment can not only inhibit glioma growth significantly but also has favorable potential ability to induce glial differentiation in the orthotopic glioma. Moreover, the aDCM@PLGA could induce a robust CD8+ effector and therefore suppress orthotopic glioma growth in a prophylactic setup, which indicates certain tumor immunity. Overall, our work provides an effective antiglioma drug delivery system which has great potential for tumor combination immunotherapy.


Asunto(s)
Glioma , Nanopartículas , Humanos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Biomimética , Glioma/tratamiento farmacológico , Antígenos de Neoplasias/metabolismo , Inmunidad , Células Dendríticas , Microambiente Tumoral
10.
Drug Deliv ; 29(1): 3111-3122, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36131565

RESUMEN

Mitochondria play an important role in regulating tumor cell death and metabolism so that they can be potential therapeutic targets. Sonodynamic therapy (SDT) represents an attractive antitumor method that induces apoptosis by producing highly toxic reactive oxygen species (ROS). Mitochondria-targeting SDT can cause oxidative damage and improve the efficiency of tumor therapy. However, due to the nonselective distribution of nanosystems and the anti-apoptotic mechanism of cancer cells, the therapeutic effect of SDT is not ideal. Therefore, we proposed a novel mitochondria-targeting nanosystem ('Mito-Bomb') for ferroptosis-boosted SDT. Sonosensitizer IR780 and ferroptosis activator RSL-3 were both encapsulated in biocompatible poly(lactic-co-glycolic acid) (PLGA) nanoparticles to form 'Mito-Bomb' (named IRP NPs). IR780 in this nanosystem was used to mediate mitochondria-targeting SDT. RSL-3 inhibited the activity of GPX4 in the antioxidant system to induce ferroptosis of tumor cells, which could rewire tumor metabolism and make tumor cells extremely sensitive to SDT-induced apoptosis. Notably, we also found that RSL-3 can inhibit hypoxia inducible factor-1α (HIF-1α) and induce ROS production to improve the efficacy of SDT to synergistically antitumor. RSL-3 was applied as a 'One-Stone-Three-Birds' agent for cooperatively enhanced SDT against triple-negative breast cancer. This study presented the first example of RSL-3 boosting mitochondria-targeting SDT as a ferroptosis activator. The 'Mito-Bomb' biocompatible nanosystem was expected to become an innovative tumor treatment method and clinical transformation.


Asunto(s)
Bombas (Dispositivos Explosivos) , Ferroptosis , Terapia por Ultrasonido , Antioxidantes/metabolismo , Línea Celular Tumoral , Mitocondrias , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Terapia por Ultrasonido/métodos
11.
Colloids Surf B Biointerfaces ; 219: 112834, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36152599

RESUMEN

Modification of the cell surface with artificial nano- and microparticles (also termed "cellular backpacks") containing biologically active payloads usually enables drug targeting via harnessing intrinsic cell tropism to the sites of injury. In some cases, using cells as delivery vehicles leads to improved pharmacokinetics due to extended circulation time of cell-immobilized formulations. Another rationale for particle attachment to cells is augmentation of desirable cellular functions and cell proliferation in response to release of the particle contents. In this study, we conjugated poly(lactic-co-glycolic acid) (PLGA) microparticles loaded with multifunctional antioxidant enzyme peroxiredoxin-1 (Prx1) to the surface of fibroblasts. The obtained microparticles were uniform in size and demonstrated sustained protein release. We found that the released Prx1 maintains its signaling activity resulting in macrophage activation, as indicated by TNFα upregulation and increase in ROS generation. Functionalization of fibroblasts with PLGA/Prx1 microparticles via EDC/sulfo-NHS coupling reaction did not affect cell viability but increased cell migratory properties and collagen I production. Moreover, PLGA/Prx1 backpacks increased resistance of fibroblasts to oxidative stress and attenuated cell senescence. In summary, we have developed a novel approach of fibroblast modification to augment their biological properties, which can be desirable for wound repair, cosmetic dermatology, and tissue engineering.


Asunto(s)
Ácido Láctico , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Ácido Láctico/metabolismo , Fibroblastos/metabolismo , Colágeno Tipo I/metabolismo , Estrés Oxidativo , Tamaño de la Partícula
12.
J Mater Chem B ; 10(40): 8282-8294, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36155711

RESUMEN

Polymeric nanocarriers (NCs) are efficient vehicles to prevent drug unspecific biodistribution and increase the drug amounts delivered to tumor tissues. However, some toxicological aspects of NCs still lack a comprehensive assessment, such as their effects on cellular processes that lead to toxicity. We evaluate the interaction of poly(lactic-co-glycolic acid) (PLGA) NCs prepared using dextran (Dex) and Pluronic®-F127 as stabilizing agents with myocardial cells (H9C2), breast adenocarcinoma cells (MCF-7) and macrophages (RAW 264.7) to address the effect of Dex in PLGA NC formulations. By an emulsion diffusion method, doxorubicin-loaded NCs were prepared with no Dex (PLGA-DOX), 1% (w/v) Dex (Dex1/PLGA-DOX) and 5% (w/v) Dex (Dex5/PLGA-DOX). Uptake analyses revealed a significant reduction in Dex5/PLGA-DOX NC uptake by H9C2 and MCF-7, as in the case of Dex1/PLGA-DOX NCs in the absence of in vitro protein corona, revealing an effect of dextran concentration on the formation of protein corona. RAW 264.7 cells presented a greater uptake of Dex5/PLGA-DOX NCs than the other NCs likely because of receptor mediated endocytosis, since C-type lectins like SIGN-R1, mannose receptors and scavenger receptor type 1 that are expressed in RAW 264.7 can mediate Dex uptake. Despite the lower uptake, Dex5/PLGA-DOX NCs promote the generation of reactive oxygen species and oxidative membrane damage in MCF-7 and H9C2 even though cellular metabolic activity assessed by MTT was comparable among all the NCs. Our results highlight the importance of an in-depth investigation of the NC-cell interaction considering additional mechanisms of damage apart from metabolic variations, as nanoparticle-induced damage is not limited to imbalance in metabolic processes, but also associated with other mechanisms, e.g., membrane and DNA damage.


Asunto(s)
Antineoplásicos , Corona de Proteínas , Humanos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Dextranos , Portadores de Fármacos/metabolismo , Antineoplásicos/farmacología , Distribución Tisular , Poloxámero/metabolismo , Emulsiones/metabolismo , Excipientes/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Doxorrubicina/farmacología , Doxorrubicina/metabolismo , Membrana Celular/metabolismo , Lectinas Tipo C/metabolismo
13.
Int J Pharm ; 627: 122162, 2022 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-36122617

RESUMEN

Retinoic acid (RA) is an approved treatment for skin photoaging induced by ultraviolet (UVA). Topically applied RA is mainly located in the stratum corneum (SC) with limited diffusion into the deeper strata. A delivery system capable of facilitating dermal delivery and cellular internalization for RA is critical for a successful photoaging therapy. Two delivery approaches, namely nanoparticles and laser ablation, were combined to improve RA's absorption efficacy and safety. The nanoparticle absorption enhancement by the lasers was compared between full-ablative (Er:YAG) and fractional (CO2) modalities. We fabricated poly-L-lactic acid (PLA) and PLA/poly(lactic-co-glycolic acid) (PLGA) nanoparticles by an emulsion-solvent evaporation technique. The mean size of PLA and PLA/PLGA nanocarriers was 237 and 222 nm, respectively. The RA encapsulation percentage in both nanosystems was > 96 %. PLA and PLA/PLGA nanocarriers promoted RA skin deposition by 5- and 3-fold compared to free control. The ablative lasers further enhanced the skin deposition of RA-loaded nanoparticles, with the full-ablative laser showing greater permeation enhancement than the fractional mode. The skin biodistribution assay evaluated by confocal and fluorescence microscopies demonstrated that the laser-assisted nanoparticle delivery achieved a significant dermis and follicular accumulation. The cell-based study indicated a facile uptake of the nanoparticles into the human dermal fibroblasts. The nanoparticulate RA increased type I collagen and elastin production in the UVA-treated fibroblasts. A reduction of matrix metalloproteinase (MMP)-1 was also highlighted in the photoaging cells. The calculation of therapeutic index (TI) by multiplying collagen/elastin elevation percentage and skin deposition predicted better anti-photoaging performance in Er:YAG laser-assisted nanoparticle delivery than CO2 laser. Nanoencapsulation of RA decreased the cytotoxicity against skin fibroblasts. In vivo skin tolerance test on a nude mouse showed less skin damage after topical application of the nanoparticles than free RA. Our results hypothesized that the laser-mediated nanoparticle delivery provided an efficient and safe use for treating photoaging.


Asunto(s)
Láseres de Estado Sólido , Nanopartículas , Enfermedades de la Piel , Ratones , Animales , Humanos , Absorción Cutánea , Elastina/metabolismo , Tretinoina , Administración Cutánea , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Colágeno Tipo I/metabolismo , Distribución Tisular , Emulsiones/metabolismo , Dióxido de Carbono/metabolismo , Piel/metabolismo , Enfermedades de la Piel/metabolismo , Ratones Desnudos , Solventes/metabolismo , Metaloproteinasas de la Matriz/metabolismo
14.
Cartilage ; 13(3): 19476035221113959, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36040157

RESUMEN

OBJECTIVE: Intervertebral disk degeneration is a prevalent postoperative complication after discectomy, underscoring the need to develop preventative and bioactive treatment strategies that decelerate degeneration and seal annulus fibrosus (AF) defects. Human mesenchymal stem cell-derived exosomes (MSC-Exos) hold promise for cell-free bioactive repair; however, their ability to promote AF repair is poorly understood. The objective of this study was to evaluate the ability of MSC-Exos to promote endogenous AF repair processes and integrate MSC-Exos within a biomaterial delivery system. DESIGN: We characterize biophysical and biochemical properties of normoxic (Nx) and hypoxic (Hx) preconditioned MSC-Exos from young, healthy donors and examine their effects on AF cell proliferation, migration, and gene expression. We then integrate a poly(lactic-co-glycolic acid) microsphere (PLGA µSphere) delivery platform within an interpenetrating network hydrogel to facilitate sustained MSC-Exo delivery. RESULTS: Hx MSC-Exos led to a more robust response in AF cell proliferation and migration than Nx MSC-Exos and was selected for a downstream protection experiment. Hx MSC-Exos maintained a healthy AF cell phenotype under a TNFα challenge in vitro and attenuated catabolic responses. In all functional assays, AF cell responses were more sensitive to Hx MSC-Exos than Nx MSC-Exos. PLGA µSpheres released MSC-Exos over a clinically relevant timescale without affecting hydrogel modulus or pH upon initial embedment and µSphere degradation. CONCLUSIONS: This MSC-Exo treatment strategy may offer benefits of stem cell therapy without the need for exogenous stem cell transplantation by stimulating cell proliferation, promoting cell migration, and protecting cells from the degenerative proinflammatory microenvironment.


Asunto(s)
Anillo Fibroso , Exosomas , Células Madre Mesenquimatosas , Exosomas/genética , Exosomas/metabolismo , Glicoles/metabolismo , Humanos , Hidrogeles , Células Madre Mesenquimatosas/metabolismo , Microesferas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo
15.
Adv Sci (Weinh) ; 9(29): e2202039, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35988145

RESUMEN

Recent evidence has indicated that overexpression of the epigenetic reader bromodomain-containing protein 4 (BRD4) contributes to a poor prognosis of lung cancers, and the suppression of its expression promotes cell apoptosis and leads to tumor shrinkage. Proteolysis targeting chimera (PROTAC) has recently emerged as a promising therapeutic strategy with the capability to precisely degrade targeted proteins. Herein, a novel style of versatile nano-PROTAC (CREATE (CRV-LLC membrane/DS-PLGA/dBET6)) is developed, which is constructed by using a pH/GSH (glutathione)-responsive polymer (disulfide bond-linked poly(lactic-co-glycolic acid), DS-PLGA) to load BRD4-targeted PROTAC (dBET6), followed by the camouflage with engineered lung cancer cell membranes with dual targeting capability. Notably, CREATE remarkably confers simultaneous targeting ability to lung cancer cells and tumor-associated macrophages (TAMs). The pH/GSH-responsive design improves the release of dBET6 payload from nanoparticles to induce pronounced apoptosis of both cells, which synergistically inhibits tumor growth in both subcutaneous and orthotopic tumor-bearing mouse model. Furthermore, the efficient tumor inhibition is due to the direct elimination of lung cancer cells and TAMs, which remodels the tumor microenvironment. Taken together, the results elucidate the construction of a versatile nano-PROTAC enables to eliminate both lung cancer cells and TAMs, which opens a new avenue for efficient lung cancer therapy via PROTAC.


Asunto(s)
Neoplasias Pulmonares , Factores de Transcripción , Animales , Ratones , Disulfuros/metabolismo , Epigénesis Genética , Glutatión/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Polímeros , Proteolisis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Microambiente Tumoral
16.
Cancer Immunol Immunother ; 71(12): 2969-2983, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35546204

RESUMEN

Heparanase has been identified as a universal tumor-associated antigen, but heparanase epitope peptides are difficult to recognize. Therefore, it is necessary to explore novel strategies to ensure efficient delivery to antigen-presenting cells. Here, we established a novel immunotherapy model targeting antigens to dendritic cell (DC) receptors using a combination of heparanase CD4+ and CD8+ T-cell epitope peptides to achieve an efficient cytotoxic T-cell response, which was associated with strong activation of DCs. First, pegylated poly(lactic-coglycolic acid) (PLGA) nanoparticles (NPs) were used to encapsulate a combined heparanase CD4+ and CD8+ T-cell epitope alone or in combination with Toll-like receptor 3 and 7 ligands as a model antigen to enhance immunogenicity. The ligands were then targeted to DC cell-surface molecules using a DEC-205 antibody. The binding and internalization of these PLGA NPs and the activation of DCs, the T-cell response and the tumor-killing effect were assessed. The results showed that PLGA NPs encapsulating epitope peptides (mHpa399 + mHpa519) could be targeted to and internalized by DCs more efficiently, stimulating higher levels of IL-12 production, T-cell proliferation and IFN-γ production by T cells in vitro. Moreover, vaccination with DEC-205-targeted PLGA NPs encapsulating combined epitope peptides exhibited higher tumor-killing efficacy both in vitro and in vivo. In conclusion, delivery of PLGA NP vaccines targeting DEC-205 based on heparanase CD4+ and CD8+ T-cell epitopes are suitable immunogens for antitumor immunotherapy and have promising potential for clinical applications.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Epítopos de Linfocito T/metabolismo , Ácido Poliglicólico/química , Ácido Poliglicólico/metabolismo , Receptor Toll-Like 3 , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Ácido Láctico/química , Ácido Láctico/metabolismo , Ligandos , Células Dendríticas , Inmunoterapia/métodos , Linfocitos T CD8-positivos , Interleucina-12/metabolismo , Péptidos/metabolismo , Linfocitos T CD4-Positivos , Polietilenglicoles
17.
Macromol Biosci ; 22(7): e2100472, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35261175

RESUMEN

Environmental accumulation of non-degradable polystyrene (PS) microparticles from plastic waste poses potential adverse impact on marine life and human health. Herein, microparticles from a degradable PS analogue (dePS) are formulated and their immuno-modulatory characteristics are comprehensively evaluated. Both dePS copolymer and microparticles are chemically degradable under accelerated hydrolytic condition. In vitro studies show that dePS microparticles are non-toxic to three immortalized cell lines. While dePS microparticles do not induce macrophage polarization in vitro, dePS microparticles induce in vivo upregulation of both pro-inflammatory and anti-inflammatory biomarkers in immuno-competent mice, suggesting the coexistence of mixed phenotypes of macrophages in the host immune response to these microparticles. Interestingly, on day 7 following subcutaneous in mice, dePS microparticles induce a lower level of several immuno-modulatory biomarkers (matrix metallo-proteinases (MMPs), tumor necrosis factor (TNF-α), and arginase activity) compared to that of reference poly(lactic-co-glycolic acid) microparticles. Remarkably, compared to PS microparticles, dePS microparticles exhibit similar in vitro and in vivo bioactivity while acquiring additional chemical degradability. Overall, this study gains new insights into the host immune response to dePS microparticles and suggests that this dePS analogue might be explored as an alternative material choice for biomedical and consumer care applications.


Asunto(s)
Macrófagos , Poliestirenos , Animales , Humanos , Inmunidad , Macrófagos/metabolismo , Ratones , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Poliestirenos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
18.
ACS Appl Mater Interfaces ; 13(38): 45244-45258, 2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34524806

RESUMEN

Cationic polymeric nanoformulations have been explored to increase the transfection efficiency of small molecules and nucleic acid-based drugs. However, an excessive positive charge density often leads to severe cell and tissue-based toxicity that restricts the clinical translation of cationic polymeric nanoformulations. Herein, we investigate a series of cationic poly(lactic-co-glycolic acid) (PLGA)-histidine-based nanoformulations for enhanced cytoplasmic delivery with minimal toxicity. PLGA/poly-l-histidine nanoparticles show promising physico-biochemical features and transfection efficiency in a series of in vitro and cell culture-based studies. Further, the use of acetone/dichloromethane as a solvent mixture during the formulation process significantly improves the morphology and size distribution of PLGA/poly-l-histidine nanoparticles. PLGA/poly-l-histidine nanoformulations undergo clathrin-mediated endocytosis. A contrast-matched small-angle neutron scattering experiment confirmed poly-l-histidine's distribution on the PLGA nanoformulations. PLGA/poly-l-histidine formulations containing paclitaxel as a small molecule-based drug and peptide nucleic acids targeting microRNA-155 as nucleic acid analog are efficacious in in vitro and in vivo studies. PLGA/poly-l-histidine NPs significantly decrease tumor growth in PNA-155 (∼6 fold) and paclitaxel (∼6.5 fold) treatment groups in a lymphoma cell line derived xenograft mice model without inducing any toxicity. Hence, PLGA/poly-l-histidine nanoformulations exhibit substantial transfection efficiency and are safe to deliver reagents ranging from small molecules to synthetic nucleic acid analogs and can serve as a novel platform for drug delivery.


Asunto(s)
Antineoplásicos/uso terapéutico , Portadores de Fármacos/química , Linfoma/tratamiento farmacológico , Nanopartículas/química , Paclitaxel/uso terapéutico , Ácidos Nucleicos de Péptidos/uso terapéutico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Portadores de Fármacos/metabolismo , Endocitosis/fisiología , Femenino , Histidina/química , Histidina/metabolismo , Humanos , Ratones Endogámicos NOD , MicroARNs/antagonistas & inhibidores , Nanopartículas/metabolismo , Paclitaxel/farmacología , Ácidos Nucleicos de Péptidos/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Carbohydr Polym ; 273: 118592, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34560993

RESUMEN

N-trimethyl chitosan (TMC) is a multifunctional polymer that can be used in various nanoparticle forms in the pharmaceutical, nutraceutical and biomedical fields. In this study, TMC was used as a mucoadhesive adjuvant to enhance the oral bioavailability and hence antitumour effects of gemcitabine formulated into nanocomplexes composed of poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) conjugated with d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS). A central composite design was applied to achieve the optimal formulation. Cellular uptake and drug transportation studies revealed the nanocomplexes permeate over the intestinal cells via adsorptive-mediated and caveolae-mediated endocytosis. Pharmacokinetic studies demonstrated the oral drug bioavailability of the nanocomplexes was increased 5.1-fold compared with drug solution. In pharmacodynamic studies, the formulation reduced tumour size 3.1-fold compared with the drug solution. The data demonstrates that TMC modified nanocomplexes can enhance gemcitabine oral bioavailability and promote the anticancer efficacy.


Asunto(s)
Antineoplásicos/uso terapéutico , Desoxicitidina/análogos & derivados , Portadores de Fármacos/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Quitosano/síntesis química , Quitosano/química , Quitosano/metabolismo , Desoxicitidina/química , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Portadores de Fármacos/síntesis química , Portadores de Fármacos/metabolismo , Liberación de Fármacos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/síntesis química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Ratas Sprague-Dawley , Vitamina E/síntesis química , Vitamina E/química , Vitamina E/metabolismo , Gemcitabina
20.
J Ocul Pharmacol Ther ; 37(7): 399-411, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34227869

RESUMEN

Purpose: To determine the impact of hypothermia on the barrier function of donor corneal endothelium, thereby enhancing the success of corneal transplantation. Methods: Primary cultures of porcine endothelial cells were subjected to hypothermia (15 h; 4°C). The impact on microtubule assembly, peri-junctional actomyosin ring (PAMR), and ZO-1 was assessed by immunocytochemistry with and without pretreatment with a microtubule-stabilizing agent (Epothilone B; EpoB; 100 nM) and a p38 MAP kinase inhibitor (SB-203580; 20 µM). In addition, EpoB-loaded PLGA nanoparticles (ENPs) prepared by nanoprecipitation technique and coated with poly-L-lysine (PLL-ENPs) were administered one-time for sustained intracellular delivery of EpoB. Results: Exposure to hypothermia led to microtubule disassembly concomitant with the destruction of PAMR and the displacement of ZO-1 at the cellular periphery, suggesting a loss in barrier integrity. These adverse effects were attenuated by pretreatment with EpoB or SB-203580. PLL-ENPs possessed a zeta potential of ∼26 mV and a size of ∼110 nm. Drug loading and entrapment efficiency were 5% (w/w) and ∼87%, respectively, and PLL-ENPs showed a biphasic release in vitro: burst phase (1 day), followed by a sustained phase (∼4 weeks). Pretreatment with PLL-ENPs (0.4 mg/mL) for 24 h stabilized the microtubules and opposed the hypothermia-induced damage to PAMR and the redistribution of ZO-1. Conclusions: Hypothermia induces microtubule disassembly via activation of p38 MAP kinase and subsequently breaks down the barrier function of the endothelium. Sustained intracellular delivery of EpoB using nanoparticles has the potential to overcome endothelial barrier failure during prolonged cold storage of donor cornea.


Asunto(s)
Citoesqueleto/metabolismo , Endotelio Corneal/metabolismo , Hipotermia/metabolismo , Microtúbulos/metabolismo , Animales , Células Cultivadas , Epotilonas/química , Epotilonas/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo , Porcinos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...