Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nano Lett ; 23(11): 5083-5091, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37220198

RESUMEN

Pyroptosis is a proinflammatory form of programmed cell death that results in the release of cellular contents and activation of immune responses. However, GSDME (a pyroptosis-executed protein) is suppressed in many cancers. Herein, we constructed a nanoliposome (GM@LR) for codelivering the GSDME-expressing plasmid and manganese carbonyl (MnCO) into TNBC cells. MnCO generated Mn2+ and carbon monoxide (CO) in the presence of H2O2. The CO-activated caspase-3, which cleaved the expressed GSDME, converting apoptosis to pyroptosis in 4T1 cells. In addition, Mn2+ promoted maturation of dendritic cells (DCs) by the activation of STING signaling pathway. The increased proportion of intratumoral mature DCs brought about massive infiltration of cytotoxic lymphocytes, leading to a robust immune response. Besides, Mn2+ could be applied for magnetic resonance imaging (MRI)-guided metastasis detection. Taken together, our study showed that GM@LR nanodrug could effectively inhibit tumor growth via pyroptosis and STING activation combined immunotherapy.


Asunto(s)
Nanopartículas , Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Peróxido de Hidrógeno/farmacología , Nanopartículas/uso terapéutico , Nucleotidiltransferasas/farmacología , Piroptosis , Femenino , Animales , Ratones
2.
Small ; 19(30): e2300280, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37060227

RESUMEN

Anticancer immunotherapy is hampered by the poor tumor immunogenicity and immunosuppressive tumor microenvironment (TME). Herein, a liposome nanodrug co-encapsulating doxycycline hydrochloride (Doxy) and chlorin e6 (Ce6) to simultaneously induce autophagy inhibition and mitochondria dysfunction for potentiating tumor photo-immunotherapy is developed. Under near infrared laser irradiation, Ce6 generates cytotoxic reactive oxygen species (ROS) and elicits robust photodynamic therapy (PDT)-induced immunogenic cell death (ICD) for immunosuppressive TME remodeling. In addition, Doxy induced mitochondria dysfunction, which increases ROS generation and enhances PDT to exert more potent killing effect and more powerful ICD. Meanwhile, Doxy increases MHC-I expression on tumor cells surface by efficient autophagy inhibition, leading to more efficient antigen presentation and CTLs recognition to increase tumor immunogenicity. The nanodrugs elicit remarkable antitumor therapy by combining Ce6-mediated PDT and Doxy-induced autophagy inhibition and mitochondria dysfunction. The developed nanodrugs represent a highly efficient strategy for improving cancer immunotherapy.


Asunto(s)
Nanopartículas , Fotoquimioterapia , Porfirinas , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Inmunoterapia , Nanopartículas/uso terapéutico , Autofagia , Línea Celular Tumoral , Porfirinas/farmacología , Porfirinas/uso terapéutico , Microambiente Tumoral
3.
Small ; 19(37): e2301420, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37154213

RESUMEN

The current immunotherapy strategies for triple negative breast cancer (TNBC) are greatly limited due to the immunosuppressive tumor microenvironment (TME). Immunization with cancer vaccines composed of tumor cell lysates (TCL) can induce an effective antitumor immune response. However, this approach also has the disadvantages of inefficient antigen delivery to the tumor tissues and the limited immune response elicited by single-antigen vaccines. To overcome these limitations, a pH-sensitive nanocalcium carbonate (CaCO3 ) carrier loaded with TCL and immune adjuvant CpG (CpG oligodeoxynucleotide 1826) is herein constructed for TNBC immunotherapy. This tailor-made nanovaccine, termed CaCO3 @TCL/CpG, not only neutralizes the acidic TME through the consumption of lactate by CaCO3 , which increases the proportion of the M1/M2 macrophages and promotes infiltration of effector immune cells but also activates the dendritic cells in the tumor tissues and recruits cytotoxic T cells to further kill the tumor cells. In vivo fluorescence imaging study shows that the pegylated nanovaccine could stay longer in the blood circulation and extravasate preferentially into tumor site. Besides, the nanovaccine exhibits high cytotoxicity in 4T1 cells and significantly inhibits tumor growth of tumor-bearing mice. Overall, this pH-sensitive nanovaccine is a promising nanoplatform for enhanced immunotherapy of TNBC.


Asunto(s)
Vacunas contra el Cáncer , Nanopartículas , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/patología , Inmunoterapia/métodos , Adyuvantes Inmunológicos , Linfocitos T Citotóxicos , Concentración de Iones de Hidrógeno , Microambiente Tumoral
4.
Pharmacol Res ; 187: 106585, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36455814

RESUMEN

Disturbed endoplasmic reticulum (ER) stress response driven by the excessive lipid accumulation in the liver is a characteristic feature in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Restoring metabolic homeostasis by targeting ER stress is a potentially therapeutic strategy for NAFLD. Here we aim to identify novel proteins or pathways involved in regulating ER stress response and therapeutic targets for alleviating NAFLD. Proteomic and transcriptomic analysis demonstrated that major urinary proteins (MUPs) were significantly reduced in the livers from NAFLD mouse models. Then we confirmed that MUP1, the major secreted form of MUPs, was reduced at mRNA and protein expression levels in hepatocytes both in vivo and in vitro under ER stress. We further illustrated that MUP1 protein levels in the urine were reduced in mice with NAFLD, which was reversed by GLP-1 receptor agonist treatment. To study the relationship between ER stress and MUP1 biology, our analysis demonstrated that MUP1 was misfolded and trapped in the ER under ER stress in vivo. Interestingly, we discovered that recombinant MUP1 treatment in hepatocytes increased calcium efflux from the ER, which resulted in transient ER stress response, including reduced protein synthesis. These responses facilitated the alleviation of chemical induced ER stress in hepatocytes, which was suggested as "pre-adaptive ER stress". Besides, recombinant MUP1 pretreatment also improved ER stress-induced insulin resistance in hepatocytes. Our findings revealed a novel and critical role of MUP1, and recombinant MUP1 or its potential derivates may serve as a promising therapeutic target for alleviating NAFLD.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Estrés del Retículo Endoplásmico , Hepatocitos , Metabolismo de los Lípidos , Hígado , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteómica
5.
Nano Lett ; 22(7): 3095-3103, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35357839

RESUMEN

Recent breakthroughs in cell membrane-fabricated nanovaccine offer innovateive therapeutic options for preventing tumor metastasies and recurrence, yet the treatment of patient-specific solid tumor remained challenging owing to the immunosuppressive tumor microenvironment. Herein, we developed a personalized photothermal nanovaccine based on the surgical tumor-derived cell membranes (CMs) coating resiquimod (R848) loaded mesoporous polydopamine (MPDA) nanoparticles for targeting tumor photothermal immunotherapy and prevention. The fabricated photothermal nanovaccine MPDA-R848@CM (MR@C) demonstrates outstanding imaging-guided photothermal immunotherapy efficacy to eradicate solid tumors under near-IR laser irradiation and further inhibiting metastasis tumors by the resulted antitumor immunities, especially in combination with programmed death-ligand 1 antibody therapy (aPD-L1). Furthermore, from in vivo prophylactic testing results, it is confirmed that the 4T1 cells rechallenge can be prevented 100% in postsurgical tumor model after vaccination of the photothermal nanovaccine. Our work fabricates a personalized photothermal nanovaccine that possesses great potential for tumor-specific treatment and for preventing postoperative tumor recurrence.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Inmunoterapia , Nanopartículas/uso terapéutico , Neoplasias/prevención & control , Fototerapia/métodos , Microambiente Tumoral
6.
Small ; 18(15): e2107732, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35218310

RESUMEN

Immunotherapy brings great benefits for tumor therapy in clinical treatments but encounters the severe challenge of low response rate mainly because of the immunosuppressive tumor microenvironment. Multifunctional nanoplatforms integrating effective drug delivery and medical imaging offer tremendous potential for cancer treatment, which may play a critical role in combinational immunotherapy to overcome the immunosuppressive microenvironment for efficient tumor therapy. Here, a nanodrug (BMS-SNAP-MOF) is prepared using glutathione (GSH)-sensitive metal-organic framework (MOF) to encapsulate an immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO) inhibitor BMS-986205, and the nitric oxide (NO) donor s-nitrosothiol groups. The high T1 relaxivity allows magnetic resonance imaging to monitor nanodrug distribution in vivo. After the nanodrug accumulation in tumor tissue via the EPR effect and subsequent internalization into tumor cells, the enriched GSH therein triggers cascade reactions with MOF, which disassembles the nanodrug to rapidly release the IDO-inhibitory BMS-986205 and produces abundant NO. Consequently, the IDO inhibitor and NO synergistically modulate the immunosuppressive tumor microenvironment with increase CD8+ T cells and reduce Treg cells to result in highly effective immunotherapy. In an animal study, treatment using this theranostic nanodrug achieves obvious regressions of both primary and distant 4T1 tumors, highlighting its application potential in advanced tumor immunotherapy.


Asunto(s)
Estructuras Metalorgánicas , Animales , Linfocitos T CD8-positivos , Inhibidores Enzimáticos , Glutatión , Inmunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenasa , Óxido Nítrico , Microambiente Tumoral
7.
Nano Lett ; 21(1): 806-815, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33395306

RESUMEN

Transplantation of neural stem cells (NSCs) is a promising treatment paradigm to replace lost neurons and reconstruct the damaged neural circuit after ischemic stroke. However, most transplanted NSCs often differentiate into astrocytes rather than functional neurons, and the poor neuronal differentiation adversely affects the therapeutic outcome of NSCs and limits their clinical translation for stroke therapy. Herein, a theranostic nanomedicine is developed to codeliver superparamagnetic iron oxide nanoparticles (SPIO) and small interfering RNA/antisense oligonucleotides (siRNA/ASO) against Pnky long noncoding RNA (lncRNA) into NSCs. This nanomedicine not only directs neuronal differentiation of NSCs through silencing the Pnky lncRNA but also allows an in vivo tracking of NSCs with magnetic resonance imaging. The enhanced neuronal differentiation of NSCs significantly improved the structural and functional recovery of the damaged brain after a stroke. The results demonstrate the great potential of the multifunctional nanomedicine targeting lncRNA to enhance stem cell-based therapies for a stroke.


Asunto(s)
Células-Madre Neurales , ARN Largo no Codificante , Accidente Cerebrovascular , Diferenciación Celular , Humanos , Nanomedicina , ARN Largo no Codificante/genética , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/terapia
8.
Nano Lett ; 21(7): 3225-3236, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33764789

RESUMEN

Developing strategies for efficient expansion of cancer stem-like cells (CSCs) in vitro will help investigate the mechanism underlying tumorigenesis and cancer recurrence. Herein, we report a dynamic culture substrate tethered with integrin ligand-bearing magnetic nanoparticles via a flexible polymeric linker to enable magnetic manipulation of the nanoscale ligand tether mobility. The cancer cells cultured on the substrate with high ligand tether mobility develop into large semispherical colonies with CSCs features, which can be abrogated by magnetically restricting the ligand tether mobility. Mechanistically, the substrate with high ligand tether mobility suppresses integrin-mediated mechanotransduction and histone-related methylation, thereby enhancing cancer cell stemness. The culture-derived high-stemness cells can generate tumors both locally and at the distant lung and uterus much more efficiently than the low-stemness cells. We believe that this magnetic nanoplatform provides a promising strategy for investigating the dynamic interaction between CSCs and the microenvironment and establishing a cost-effective tumor spheroid model.


Asunto(s)
Mecanotransducción Celular , Neoplasias , Línea Celular Tumoral , Femenino , Humanos , Integrinas , Ligandos , Células Madre Neoplásicas , Microambiente Tumoral
9.
Small ; 17(25): e2100609, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34032365

RESUMEN

The tumor hypoxic microenvironment not only induces genetic and epigenetic changes in tumor cells, immature vessels formation for oxygen demand, but also compromises the efficiency of therapeutic interventions. On the other hand, conventional therapeutic approaches which kill tumor cells or destroy tumor blood vessels to block nutrition and oxygen supply usually facilitate even harsher microenvironment. Thus, simultaneously relieving the strained response of tumor cells and blood vessels represents a promising strategy to reverse the adverse tumor hypoxic microenvironment. In the present study, an integrated amphiphilic system (RSCD) is designed based on Angiotensin II receptor blocker candesartan for siRNA delivery against the hypoxia-inducible factor-1 alpha (HIF-1α), aiming at both vascular and cellular "relaxation" to reconstruct a tumor normoxic microenvironment. Both in vitro and in vivo studies have confirmed that the hypoxia-inducible HIF-1α expression is down-regulated by 70% and vascular growth is inhibited by 60%. The "relaxation" therapy enables neovascularization with more complete and organized structures to obviously increase the oxygen level inside tumor, which results in a 50% growth inhibition. Moreover, reconstruction of tumor microenvironment enhances tumor-targeted drug delivery, and significantly improves the chemotherapeutic and photodynamic anticancer treatments.


Asunto(s)
Hipoxia , Microambiente Tumoral , Hipoxia de la Célula , Línea Celular Tumoral , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Oxígeno , ARN Interferente Pequeño
10.
J Nanobiotechnology ; 19(1): 341, 2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702273

RESUMEN

Effectively promoting corneal allograft survival remains a challenge in corneal transplantation. The emerging therapeutic agents with high pharmacological activities and their appropriate administration routes provide attractive solutions. In the present study, a celastrol-loaded positive nanomedicine (CPNM) was developed to enhance corneal penetration and to promote corneal allograft survival. The in vitro, in vivo and ex vivo results demonstrated the good performance of CPNM prolonging the retention time on ocular surface and opening the tight junction in cornea, which resulted in enhanced corneal permeability of celastrol. Both in vitro and in vivo results demonstrated that celastrol inhibited the recruitment of M1 macrophage and the expression of TLR4 in corneal allografts through the TLR4/MyD88/NF-κB pathway, thereby significantly decreasing secretion of multiple pro-inflammatory cytokines to promote corneal allograft survival. This is the first celastrol-based topical instillation against corneal allograft rejection to provide treatment more potent than conventional eye drops for ocular anterior segment diseases.


Asunto(s)
Trasplante de Córnea , Supervivencia de Injerto/efectos de los fármacos , Nanomedicina/métodos , Triterpenos Pentacíclicos/farmacología , Animales , Citocinas/metabolismo , Femenino , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Soluciones Oftálmicas/administración & dosificación , Soluciones Oftálmicas/farmacología , Triterpenos Pentacíclicos/administración & dosificación , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Ratas Wistar
11.
Nano Lett ; 20(10): 7728-7736, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32965124

RESUMEN

Rheumatoid arthritis (RA) is a systemic inflammatory disorder which can cause bone and cartilage damage leading to disability, yet the treatment remains unsatisfactory nowadays. Celastrol (Cel) has shown antirheumatic activity against RA. However, the frequent parenteral delivery and poor water solubility of Cel restrict its further therapeutic applications. Here, aiming at effectively overcoming the poor water solubility and short half-life of Cel to boost its beneficial effects for treating RA, we developed a polymeric micelle for Cel delivery based on a reactive oxygen species (ROS) sensitive polymer. Our results demonstrated that Cel may inhibit the repolarization of macrophages toward the pro-inflammatory M1 pheno-type via regulating the NF-κB and Notch1 pathways, which resulted in significantly decreased secretion of multiple pro-inflammatory cytokines to suppress the RA progression. Consequently, the Cel-loaded micelle effectively alleviated the major RA-associated symptoms including articular scores, ankle thickness, synovial inflammation, bone erosion, and cartilage degradation.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Inflamación/tratamiento farmacológico , FN-kappa B/genética , Triterpenos Pentacíclicos
12.
Small ; 16(7): e1906832, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31990457

RESUMEN

Immune checkpoint blockade (ICB) is demonstrating great potential in cancer immunotherapy nowadays. Yet, the low response rate to ICB remains an urgent challenge for tumor immunotherapy. A pH and matrix metalloproteinase dual-sensitive micellar nanocarrier showing spatio-temporally controlled release of anti-PD-1 antibody (aPD-1) and paclitaxel (PTX) in solid tumors is prepared to realize synergistic cancer chemoimmunotherapy. Antitumor immunity can be activated by PTX-induced immunogenic cell death (ICD), while aPD-1 blocks the PD-1/PD-L1 axis to suppress the immune escape due to PTX-induced PD-L1 up-regulation, thus resulting in a synergistic antitumor chemoimmunotherapy. Through decoration with a sheddable polyethylene glycol (PEG) shell, the nanodrug may better accumulate in tumors to boost the synergistic antitumor treatment in a mouse melanoma model. The present study demonstrates a potent antitumor chemoimmunotherapy utilizing tumor microenvironment-sensitive micelles bearing a sheddable PEG layer to mediate site-specific sequential release of aPD-1 and PTX.


Asunto(s)
Anticuerpos , Antineoplásicos , Quimioterapia , Inmunoterapia , Metaloproteinasas de la Matriz , Micelas , Paclitaxel , Animales , Anticuerpos/administración & dosificación , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Concentración de Iones de Hidrógeno , Inmunoterapia/métodos , Metaloproteinasas de la Matriz/administración & dosificación , Ratones , Paclitaxel/administración & dosificación , Polietilenglicoles/química
13.
Small ; 16(33): e2001251, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32677157

RESUMEN

Ferroptosis is attracting significant attention due to its effectiveness in tumor treatment. The efficiency to produce toxic lipid peroxides (LPOs) at the tumor site plays a key role in ferroptosis. A hybrid PFP@Fe/Cu-SS metal organic framework (MOF) is synthesized and shown to increase intratumoral LPO content through redox reactions generating ·OH. In addition, glutathione (GSH) depletion through disulfide-thiol exchange leads to the inactivation of glutathione peroxide 4 (GPX4), which results in a further increase in LPO content. This MOF exhibits high inhibitory effect on the growth of xenografted Huh-7 tumors in mice. The coadministration of a ferroptosis inhibitor reduces the antitumor effect of the MOF, leading to a restoration of GPX4 activity and an increase in tumor growth. Moreover, the construction of Cu into mesoporous PFP@Fe/Cu-SS not only allows the MOF to be used as a contrast agent for T1 -weighted magnetic resonance imaging, but also renders its photothermal conversion capacity. Thus, near-infrared irradiation is able to induce photothermal therapy and transform the encapsulated liquid perfluoropentane into microbubbles for ultrasound imaging.


Asunto(s)
Ferroptosis , Estructuras Metalorgánicas , Nanopartículas , Neoplasias , Animales , Línea Celular Tumoral , Ratones , Neoplasias/tratamiento farmacológico , Oxidación-Reducción
14.
Mol Pharm ; 17(3): 817-826, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31910019

RESUMEN

Biomedical nanoplatforms have been widely investigated for ultrasound (US) imaging and cancer therapy. Herein, perfluorocarbon (PFC) is encapsulated into biocompatible polydopamine (PDA) to form a theranostic nanosystem, followed by the modification of polyethylene glycol (PEG) to stabilize the nanoparticle via a facile one-pot method. Under 808 nm near-infrared laser irradiation, PDA can generate hyperthermia to transform PFC droplets to bubbles with high US imaging sensitivity. The US imaging detection of the PFC-PDA-PEG nanosystem is achievable in a time span of up to 25 min in vitro at a low US frequency and mechanical index, manifesting a US imaging performance for in vivo application. Moreover, tumor cells incubated with the nanosystem are ablated effectively under laser irradiation at 808 nm. The results illustrate the potential of the PDA-based theranostic agent in US imaging-guided photothermal therapy of tumor.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fluorocarburos/administración & dosificación , Hipertermia Inducida/métodos , Indoles/administración & dosificación , Rayos Infrarrojos/uso terapéutico , Nanopartículas/química , Terapia Fototérmica/métodos , Polímeros/administración & dosificación , Animales , Cápsulas , Supervivencia Celular/efectos de los fármacos , Medios de Contraste , Femenino , Fluorocarburos/química , Células HCT116 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indoles/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Polietilenglicoles/química , Polímeros/química , Carga Tumoral/efectos de los fármacos , Ultrasonografía/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nanotechnology ; 30(1): 015101, 2019 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-30370902

RESUMEN

Various nanoformulations of perfluorocarbon have been developed thus far, to achieve ultrasound imaging of tumors and tumor-targeted therapy. However, their application has been greatly limited by their short sonographic duration and large size distribution. A novel theranostic agent was constructed based on gold nanoshell cerasome-encapsulated L-menthol (GNC-LM). Owing to the sustained and controllable generation of L-menthol bubbles under near-infrared laser irradiation, GNC-LM showed good performance in contrast enhancement of ultrasound imaging in vivo. GNC-LM could be imaged for 30 min, which is much longer than the imaging time of SonoVue (commercially used microbubbles). Moreover, photothermal therapy (PTT) based on the light-to-heat conversion of the nanosystem effectively ablated the tumor. Our study demonstrated the promising potential of the obtained GNC-LM to serve as a therapeutic nanoprobe for ultrasound contrast imaging and PTT of tumors.


Asunto(s)
Medios de Contraste/química , Oro/química , Hipertermia Inducida , Lípidos/química , Mentol/química , Nanocáscaras/química , Neoplasias/terapia , Fototerapia , Animales , Muerte Celular , Ratones Endogámicos BALB C , Nanocáscaras/ultraestructura , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Ultrasonografía
17.
Biomacromolecules ; 19(6): 2248-2256, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29690766

RESUMEN

Drug resistance, developed through multiple mechanisms, is a major hindrance to successful chemotherapy of tumor. Combination therapy of chemotherapeutic drugs and siRNA represents an emerging strategy which may improve anticancer effect by synergistic actions. In this study, triblock copolymer of poly(ethylene glycol)- block-poly(l-lysine)- block-poly aspartyl ( N-( N', N'-diisopropylaminoethyl)) (PEG-PLL-PAsp(DIP)) was synthesized for the first time to enable the codelivery of BCL-2 siRNA and DOX. The system is supposed to not only bypass drug efflux but also down-regulate the antiapoptotic gene and consequently confronting against chemoresistance as well. Moreover, the pH responsive ability of the codelivery system can prevent drug leakage during circulation and guarantee swift drug release at tumors. The codelivered siRNA serves to suppress the expression of antiapoptotic BCL-2 and hence sensitize the cancer cells to anticancer drugs and produce improved therapeutic effect. Consequently, the codelivery of BCL-2 siRNA and anticancer drug DOX serves as a promising strategy against drug resistance in chemotherapy.


Asunto(s)
Carcinoma Hepatocelular/terapia , Doxorrubicina , Resistencia a Antineoplásicos , Técnicas de Transferencia de Gen , Vectores Genéticos , Neoplasias Hepáticas Experimentales/terapia , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , ARN Interferente Pequeño , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Vectores Genéticos/química , Vectores Genéticos/genética , Vectores Genéticos/farmacocinética , Vectores Genéticos/farmacología , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Polímeros/química , Polímeros/farmacocinética , Polímeros/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Int J Med Sci ; 15(2): 129-141, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29333097

RESUMEN

Purpose: To establish small-sized superparamagnetic polymeric micelles for magnetic resonance and fluorescent dual-modal imaging, we investigated the feasibility of MR imaging (MRI) and macrophage-targeted in vitro. Methods: A new class of superparamagnetic iron oxide nanoparticles (SPIONs) and Nile red-co-loaded mPEG-Lys3-CA4-NR/SPION polymeric micelles was synthesized to label Raw264.7 cells. The physical characteristics of the polymeric micelles were assessed, the T2 relaxation rate was calculated, and the effect of labeling on the cell viability and cytotoxicity was also determined in vitro. In addition, further evaluation of the application potential of the micelles was conducted via in vitro MRI. Results: The diameter of the mPEG-Lys3-CA4-NR/SPION polymeric micelles was 33.8 ± 5.8 nm on average. Compared with the hydrophilic SPIO, mPEG-Lys3-CA4-NR/SPION micelles increased transversely (r2), leading to a notably high r2 from 1.908 µg/mL-1S-1 up to 5.032 µg/mL-1S-1, making the mPEG-Lys3-CA4-NR/SPION micelles a highly sensitive MRI T2 contrast agent, as further demonstrated by in vitro MRI. The results of Confocal Laser Scanning Microscopy (CLSM) and Prussian blue staining of Raw264.7 after incubation with micelle-containing medium indicated that the cellular uptake efficiency is high. Conclusion: We successfully synthesized dual-modal MR and fluorescence imaging mPEG-Lys3-CA4-NR/SPION polymeric micelles with an ultra-small size and high MRI sensitivity, which were effectively and quickly uptaken into Raw 264.7 cells. mPEG-Lys3-CA4-NR/SPION polymeric micelles might become a new MR lymphography contrast agent, with high effectiveness and high MRI sensitivity.


Asunto(s)
Medios de Contraste/química , Macrófagos/efectos de los fármacos , Imagen por Resonancia Magnética/métodos , Micelas , Polímeros/química , Animales , Supervivencia Celular/efectos de los fármacos , Medios de Contraste/farmacología , Compuestos Férricos/química , Colorantes Fluorescentes/química , Espectroscopía de Resonancia Magnética , Nanopartículas de Magnetita/química , Ratones , Oxazinas/química , Tamaño de la Partícula , Polímeros/síntesis química , Células RAW 264.7
19.
Nanomedicine ; 14(7): 2215-2226, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29964220

RESUMEN

Inflammation and oxidative stress are two major factors that are involved in the pathogenesis of atherosclerosis. A smart drug delivery system that responds to the oxidative microenvironment of atherosclerotic plaques was constructed in the present study. Andrographolide-loaded micelle was assembled from the block copolymer of poly(ethylene glycol) and poly(propylene sulphide) (PEG-PPS) for the purpose of simultaneously decreasing inflammatory response and the level of reactive oxygen species (ROS) to treat atherosclerosis. Owing to the ROS-responsive nature of PEG-PPS, the micelle not only serves as a stimuli-responsive drug carrier to quickly release the encapsulated drug, andrographolide, but also consumes ROS by itself at the pathologic sites, upon which the expressions of pro-inflammatory cytokines are effectively suppressed and oxidative stress is alleviated. Consequently, the andrographolide-loaded micelle demonstrated remarkable therapeutic effects both in vitro and in vivo. In conclusion, the andrographolide-loaded PEG-PPS micelle can synchronically alleviate inflammation and oxidative stress, providing a promising and innovative strategy against atherosclerosis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Diterpenos/farmacología , Sistemas de Liberación de Medicamentos , Inflamación/tratamiento farmacológico , Placa Aterosclerótica/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Animales , Aorta/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Diterpenos/administración & dosificación , Diterpenos/química , Portadores de Fármacos/química , Femenino , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones Noqueados para ApoE , Micelas , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Polietilenglicoles/química , Polímeros/administración & dosificación , Polímeros/química
20.
Hepatology ; 63(4): 1240-55, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26680504

RESUMEN

UNLABELLED: Because the primary surgical treatment options for hepatocellular carcinoma (HCC)-including hepatic resection and liver transplantation-often fail due to recurrence and metastasis, identifying early prognostic biomarkers and therapeutic targets for HCC is of great importance. This study shows that transducin ß-like protein 1-related protein (TBLR1) is a key HCC oncogene that plays important roles in HCC proliferation, antiapoptosis, and angiogenesis by regulating the Wnt/ß-catenin pathway. The folate-targeted theranostic small interfering RNA (siRNA) nanomedicine Fa-PEG-g-PEI-SPION/psiRNA-TBLR1 effectively silences the TBLR1 gene in different human HCC cell lines in vitro and in human HCC samples in vivo, resulting in the simultaneous suppression of HCC cell proliferation, antiapoptosis, and angiogenesis. Because of its multi-anticancer functions against HCC, intravenous injection of the folate-targeted siRNA nanomedicine into nude mice bearing intrahepatic or subcutaneous xenografts of human HCC has a significant therapeutic effect. Tumor growth in those animals was almost completely inhibited by treatment with Fa-PEG-g-PEI-SPION/psiRNA-TBLR1. Moreover, the SPION-encapsulated polyplexes possess high magnetic resonance imaging (MRI) detection sensitivity, which makes tumor-targeted siRNA delivery easily trackable using the clinical MRI technique. CONCLUSION: The theranostic siRNA nanomedicine examined here possesses great theranostic potential for combined gene therapy and MRI diagnosis of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Terapia Molecular Dirigida/métodos , Proteínas Nucleares/genética , ARN Interferente Pequeño/administración & dosificación , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Represoras/genética , Anciano , Análisis de Varianza , Animales , Apoptosis/genética , Carcinoma Hepatocelular/mortalidad , Modelos Animales de Enfermedad , Femenino , Terapia Genética/métodos , Xenoinjertos , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/mortalidad , Imagen por Resonancia Magnética/métodos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Análisis Multivariante , Oncogenes , Modelos de Riesgos Proporcionales , Distribución Aleatoria , Resultado del Tratamiento , Células Tumorales Cultivadas , Vía de Señalización Wnt/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA