Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Int J Biol Macromol ; 274(Pt 2): 133186, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38885858

RESUMEN

Ligand-receptor recognition serves as the fundamental driving force for active targeting, yet it is still constrained by off-target effects. Herein, we demonstrate that circumventing or blocking the mononuclear phagocyte system (MPS) are both viable strategies to address off-target effects. Naturally derived lignin nanoparticles (LNPs) show great potential to block MPS due to its good stability, low toxicity, and degradability. We further demonstrate the impact of LNPs dosage on in vivo tumor targeting and antitumor efficacy. Our results show that a high dose of LNPs (300 mg/kg) leads to significant accumulation at the tumor site for a duration of 14 days after intravenous administration. In contrast, the low-dose counterparts (e.g., 50, 150 mg/kg) result in almost all LNPs accumulating in the liver. This discovery indicates that the liver is the primary site of LNP capture, leaving only the surplus LNPs the chance to reach the tumor. In addition, although cell membrane-engineered LNPs can rapidly penetrate tumors, they are still prone to capture by the liver during subsequent circulation in the bloodstream. Excitingly, comparable therapeutic efficacy is obtained for the above two strategies. Our findings may offer valuable insights into the targeted delivery of drugs for disease treatment.


Asunto(s)
Macrófagos del Hígado , Lignina , Hígado , Nanopartículas , Fagocitosis , Animales , Lignina/farmacología , Lignina/química , Nanopartículas/química , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Ratones , Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Fagocitosis/efectos de los fármacos , Humanos , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología
2.
Chem Biol Drug Des ; 103(3): e14481, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38458969

RESUMEN

Studies have shown that saikosaponin D (SSD) has favorable neurotherapeutic effects. Therefore, the objective of this study was to explore the efficacy and possible molecular mechanisms of SSD on pilocarpine (PP)-induced astrocyte injury. Primary astrocytes were isolated from juvenile rats and identified using immunofluorescence. The cells were treated with PP and/or SSD for 6 h and 12 h, respectively, followed by measurement of their viability through 3-(4,5-dimethylthiazol)-2,5-diphenyl-tetrazolium bromide (MTT) assay. Next, quantitative real-time polymerase chain reaction (qRT-PCR) was used to measure the expression levels of Glial fibrillary acidic protein (GFAP), C3, S100 calcium binding protein A10 (S100a10), pentraxin 3 (Ptx3), toll-like receptor 4 (TLR4), and RAG in astrocytes after different treatments. Enzyme-linked immunosorbent assay and biochemical tests were utilized to evaluate the level of inflammatory factors [interleukin (IL)-1ß, IL-6, and tumor necrosis factor alpha (TNF-α)] secreted by cells and the content of oxidative stress-related factors (malondialdehyde [MDA] and glutathione [GSH]) or enzyme activity (catalase [CAT] and glutathione peroxidase [GPX]) in cells. The JC-1 mitochondrial membrane potential (MMP) fluorescence probe was used to measure the MMP in astrocytes. Additionally, western blot was applied to test the expression of proteins related to the nod-like receptor protein 3 (NLRP3)/caspase-1 signaling pathway. PP treatment (1 mM) induced cell injury by significantly reducing the viability of astrocytes and expression of cellular markers. SSD treatment (4 µM) had no toxicity to astrocytes. Besides, SSD (4 µM) treatment could significantly up-regulate the cell viability and marker expression of PP-induced astrocytes. Furthermore, SSD could be employed to inhibit inflammation (reduce IL-1ß, IL-6, and TNF-α levels) and oxidative stress (decrease MDA level, elevate GSH level, the activity of CAT and GPX), and ameliorate mitochondrial dysfunction (upregulate JC-1 ratio) in PP-induced astrocytes. Moreover, further mechanism exploration revealed that SSD treatment significantly reduced the activity of the NLRP3/caspase-1 signaling pathway activated by PP induction. SSD increased cell viability, inhibited inflammation and oxidative stress response, and ameliorated mitochondrial dysfunction in PP-induced astrocyte injury model, thus playing a neuroprotective role. The mechanism of SSD may be related to the inhibition of the NLRP3/caspase-1 inflammasome.


Asunto(s)
Bencimidazoles , Carbocianinas , Enfermedades Mitocondriales , Proteína con Dominio Pirina 3 de la Familia NLR , Ácido Oleanólico/análogos & derivados , Saponinas , Ratas , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Pilocarpina/toxicidad , Factor de Necrosis Tumoral alfa/genética , Caspasas/metabolismo , Interleucina-6 , Transducción de Señal , Inflamación/metabolismo
3.
Adv Healthc Mater ; 13(11): e2303779, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38288884

RESUMEN

Nanomaterials that generate reactive oxygen species (ROS) upon light irradiation have significant applications in various fields, including photodynamic therapy (PDT) that is widely recognized as a highly momentous strategy for the eradication of cancer cells. However, the ROS production rate of photosensitizers, as well as the tumor hypoxia environment, are two major challenges that restrict the widespread application of PDT. In this study, a cancer-thylakoid hybrid membrane-camouflaged thulium oxide nanoparticles (Tm2O3) for tumor-homing phototherapy through dual-stage-light-guided ROS generation and oxygen self-supply is developed. Tm2O3 as a type II photosensitizer are viable for NIR-stimulated ROS generation due to the unique energy levels, large absorption cross section, and long lifetime of the 3H4 state of Tm ions. The thylakoid membrane (TK) plays a catalase-like role in converting hydrogen peroxide into oxygen and also acts as a natural photosensitizer that can generate lethal ROS through electron transfer when exposed to light. In addition, fluorescence dye DiR is embedded in the hybrid membrane for in vivo tracing as well as photothermal therapy. Results show that tumors in Tm2O3@TK-M/DiR group are effectively ablated following dual-stage-light irradiation, highlighting the promising potential of rare-earth element-based type II photosensitizers in various applications.


Asunto(s)
Nanopartículas , Oxígeno , Fotoquimioterapia , Fármacos Fotosensibilizantes , Especies Reactivas de Oxígeno , Tulio , Animales , Tulio/química , Especies Reactivas de Oxígeno/metabolismo , Ratones , Humanos , Oxígeno/química , Oxígeno/metabolismo , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Nanopartículas/química , Fotoquimioterapia/métodos , Óxidos/química , Línea Celular Tumoral , Ratones Endogámicos BALB C , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/metabolismo , Fototerapia/métodos
4.
Exp Eye Res ; 219: 109053, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35351462

RESUMEN

PURPOSE: Dry eye syndrome (DES) is a multifactorial ocular disorder. The possible pathogens and pathogenic mechanisms for virus-related dry eye disease are largely unknown. The current study aimed to provide evidence for mechanisms contributing to DES induced by herpes simplex virus (HSV) infection in the harderian gland (HG) and lacrimal gland (LG). METHODS: We recorded the dry eye-like cornea pathology of irf3-/- mice infected with HSV-1 till 8 months of age. The slit-lamp and confocal microscopy was used to observe the corneal defects. TUNEL was used to detect the corneal apoptosis. Human corneas suffered from herpes stromal keratitis (HSK) were also analyzed as a comparison. Then, we measure the aqueous tear production with a phenol red thread test in irf3-/-mice, and recorded their tear film breakup time. HGs and LGs were sectioned and analyzed using HE and oil-red-O staining. For molecular signaling pathway analysis, we used mRNA sequencing to explore the related gene ontology. Western blotting (WB) and real-time reverse transcription-quantitative polymerase chain reaction were used to verify the level of the Akt signaling pathway and related inflammatory factors. RESULTS: Inoculated irf3-/- mice tended to develop dry eye-like symptoms, such as corneal keratinization, corneal cell apoptosis, and tear reduction. The HGs and LGs of irf3-/- mice showed increased level of HSV-1, and exhibited inflammatory pathological changes and impaired function, which explained the damaged tear film. WB and mRNA sequencing indicated that enhanced PI3K-Akt pathway in irf3-/- mice might account for the higher susceptibility to HSV infection. CONCLUSIONS: We observed evidence of DES in irf3-/- mice induced by HSV-1 infection in the HGs and LGs, which may introduce a potential novel target for DES treatment.


Asunto(s)
Síndromes de Ojo Seco , Glándula de Harder , Herpes Simple , Herpesvirus Humano 1 , Queratitis Herpética , Aparato Lagrimal , Animales , Córnea/metabolismo , Modelos Animales de Enfermedad , Síndromes de Ojo Seco/metabolismo , Glándula de Harder/metabolismo , Glándula de Harder/patología , Herpes Simple/metabolismo , Herpes Simple/patología , Factor 3 Regulador del Interferón/metabolismo , Aparato Lagrimal/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo
5.
Invest Ophthalmol Vis Sci ; 61(4): 1, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32271890

RESUMEN

Purpose: Purpose The role of endothelial Yes-associated protein 1 (YAP) in the pathogenesis of retinal angiogenesis and the astrocyte network in the mouse oxygen-induced retinopathy (OIR) model is unknown. Methods: For in vivo studies, OIR was induced in conditional endothelial YAP knockout mice and their wild-type littermates. Retinal vascularization and the astrocyte network were evaluated by whole-mount fluorescence and Western blotting. In vitro experiments were performed in astrocytes cultured with human microvascular endothelial cell-1-conditioned medium to analyze the mechanisms underlying the effect of endothelial YAP on astrocytes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Astrocitos/patología , Proteínas de Ciclo Celular/fisiología , Células Endoteliales/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Neovascularización Retiniana/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Proliferación Celular , Células Cultivadas , Medios de Cultivo Condicionados , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Oxígeno/toxicidad , Neovascularización Retiniana/inducido químicamente , Neovascularización Retiniana/patología , Vasos Retinianos/citología , Retinopatía de la Prematuridad/inducido químicamente , Retinopatía de la Prematuridad/metabolismo , Retinopatía de la Prematuridad/patología , Proteínas Señalizadoras YAP
6.
Cell Prolif ; 53(2): e12757, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31916327

RESUMEN

OBJECTIVES: To testify that endothelial cells (ECs) induce astrocyte maturation by leukaemia inhibitory factor (LIF) secretion. MATERIALS AND METHODS: In vivo experiments, mice bearing floxed alleles of YAP were crossed with mice expressing a Cre recombinase driven by the endothelial Tek promoter (Tek-Cre) to finally obtain the following three genotypes: YAPf/f , Tek-Cre; YAPf/w , Tek-Cre; and YAPf/f . Retinal vascularization and astrocyte network were evaluated by whole-mount fluorescence and Western blotting. In vitro, experiments were performed in an astrocyte and human microvascular endothelial cell (HMEC-1) coculture model to analyse the mechanisms underlying the effect of endothelial YAP on astrocytes. RESULTS: In vivo, YAPf/f ;Tek-Cre mice showed delayed angiogenesis, sparse vessels and decreased glial fibrillary acidic protein (GFAP)+ astrocytes but aberrant growth of endothelial networks and immature astrocytes (platelet-derived growth factor A, PDGFRA+ astrocytes) overgrowth. In vitro, Yap deletion attenuated the LIF release that delayed the maturation of retinal astrocyte which was consistent with the results of HMEC-1-astrocyte coculture. The effect of YAP overexpression on LIF-LIFR axis in HMEC-1 interferes the GFAP expression of astrocyte. In contrast, LIF protein rescues the astrocytic GFAP expression when EC YAP was inhibited by siRNAs. CONCLUSIONS: We show that EC yes-associated protein (YAP) is not only a critical coactivator of Hippo signalling in retinal vessel development but also plays an essential role in retinal astrocyte maturation by regulating LIF production.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Astrocitos/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Retina/metabolismo , Vasos Retinianos/metabolismo , Factores de Transcripción/metabolismo , Animales , Astrocitos/fisiología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Técnicas de Cocultivo/métodos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Fisiológica/fisiología , Neurogénesis/fisiología , Retina/fisiología , Vasos Retinianos/fisiología , Proteínas Señalizadoras YAP
7.
J Cell Physiol ; 234(1): 246-258, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30094836

RESUMEN

Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are transcriptional coactivators established as a nexus in numerous signaling pathways, notably in Hippo signaling. Previous research revealed multifarious function of YAP and TAZ in oncology and cardiovasology. Recently, the focus has been laid on their pivotal role in eye morphogenesis and homeostasis. In this review, we synthesize advances of YAP and TAZ function during eye development in different model organisms, introduce their function in different ocular tissues and eye diseases, and highlight the potential for therapeutic interventions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Oftalmopatías/genética , Ojo/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intracelular/genética , Fosfoproteínas/genética , Factores de Transcripción/genética , Ojo/metabolismo , Oftalmopatías/patología , Oftalmopatías/terapia , Vía de Señalización Hippo , Homeostasis/genética , Humanos , Morfogénesis/genética , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/genética , Transactivadores , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP
8.
Adv Mater ; 30(22): e1707459, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29675900

RESUMEN

Many viruses have a lipid envelope derived from the host cell membrane that contributes much to the host specificity and the cellular invasion. This study puts forward a virus-inspired technology that allows targeted genetic delivery free from man-made materials. Genetic therapeutics, metal ions, and biologically derived cell membranes are nanointegrated. Vulnerable genetic therapeutics contained in the formed "nanogene" can be well protected from unwanted attacks by blood components and enzymes. The surface envelope composed of cancer cell membrane fragments enables host-specific targeting of the nanogene to the source cancer cells and homologous tumors while effectively inhibiting recognition by macrophages. High transfection efficiency highlights the potential of this technology for practical applications. Another unique merit of this technology arises from the facile combination of special biofunction of metal ions with genetic therapy. Typically, Gd(III)-involved nanogene generates a much higher T1 relaxation rate than the clinically used Gd magnetic resonance imaging agent and harvests the enhanced MRI contrast at tumors. This virus-inspired technology points out a distinctive new avenue for the disease-specific transport of genetic therapeutics and other biomacromolecules.

9.
Small ; 14(20): e1800292, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29665292

RESUMEN

This study reports a double-targeting "nanofirework" for tumor-ignited imaging to guide effective tumor-depth photothermal therapy (PTT). Typically, ≈30 nm upconversion nanoparticles (UCNP) are enveloped with a hybrid corona composed of ≈4 nm CuS tethered hyaluronic acid (CuS-HA). The HA corona provides active tumor-targeted functionality together with excellent stability and improved biocompatibility. The dimension of UCNP@CuS-HA is specifically set within the optimal size window for passive tumor-targeting effect, demonstrating significant contributions to both the in vivo prolonged circulation duration and the enhanced size-dependent tumor accumulation compared with ultrasmall CuS nanoparticles. The tumors featuring hyaluronidase (HAase) overexpression could induce the escape of CuS away from UCNP@CuS-HA due to HAase-catalyzed HA degradation, in turn activating the recovery of initially CuS-quenched luminescence of UCNP and also driving the tumor-depth infiltration of ultrasmall CuS for effective PTT. This in vivo transition has proven to be highly dependent on tumor occurrence like a tumor-ignited explosible firework. Together with the double-targeting functionality, the pathology-selective tumor ignition permits precise tumor detection and imaging-guided spatiotemporal control over PTT operation, leading to complete tumor ablation under near infrared (NIR) irradiation. This study offers a new paradigm of utilizing pathological characteristics to design nanotheranostics for precise detection and personalized therapy of tumors.


Asunto(s)
Hipertermia Inducida , Nanofibras/química , Neoplasias/patología , Fototerapia , Animales , Muerte Celular , Cobre/química , Células Hep G2 , Humanos , Ácido Hialurónico/química , Hialuronoglucosaminidasa/metabolismo , Luminiscencia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células 3T3 NIH , Nanofibras/ultraestructura , Nanopartículas/química , Nanopartículas/ultraestructura , Células RAW 264.7 , Esferoides Celulares/patología , Esferoides Celulares/ultraestructura , Sulfuros/química , Temperatura
10.
ACS Appl Mater Interfaces ; 9(49): 42622-42632, 2017 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-29148707

RESUMEN

A facile and targeted gene delivery system was prepared by conjugating ß-cyclodextrin modified polyethylenimine (PEI-CD) and adamantyl peptide (AdGRGDS) based on host-guest interaction. With the rational design between PEI-CD and AdGRGDS, the PEI-CD/AdGRGDS gene delivery system showed excellent DNA binding capability and exhibited good ability to compact DNA into uniform spherical nanoparticles. In vitro luciferase assay showed that gene expression transfected by PEI-CD/AdGRGDS was stronger than that by PEI-CD in HeLa cells, whereas gene expression transfected by PEI-CD/AdGRGDS and PEI-CD was similar to each other in COS7 cells. Internalization of complexes was qualitatively studied using a confocal laser scanning microscope (CLSM) and quantitatively analyzed by flow cytometry, respectively, and targeting specificity was also evaluated by CLSM. Results of CLSM and flow cytometry indicated that PEI-CD/AdGRGDS had good targeting specificity to tumor cells with integrin αvß3 overexpression. To further evaluate the targeting specificity and transfection efficiency in vivo, a rat model with murine hepatic carcinoma cell line H22 was used. PEI-CD/AdGRGDS showed stronger gene expression efficiency than PEI-CD via in vivo transfection of pORF-LacZ and pGL-3 plasmids after subcutaneous injection. Interestingly, PEI-CD/AdGRGDS also showed high targeting specificity and transfection distribution to tumor xenograft after tail-vein injection. In vitro and in vivo assays highlighted the importance of GRGDS targeting specificity to tumor cells with integrin αvß3 overexpression and demonstrated that the PEI-CD/AdGRGDS gene delivery system would have great potential for targeted tumor therapy.


Asunto(s)
Técnicas de Transferencia de Gen , Animales , Células HeLa , Humanos , Ratones , Plásmidos , Polietileneimina , Ratas , Transfección
11.
Biomaterials ; 139: 116-126, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28600977

RESUMEN

Tumor metastasis is the leading cause of death in cancer patients, and epithelial-mesenchymal transition (EMT) is an essential step in tumor metastasis. Unfortunately, during the chemotherapy, EMT could be induced under the selective pressure of clinical cytotoxic drugs. Here, to solve this problem, we have synthesized multi-functional epigallocatechin gallate/iron nano-complexes (EIN) as a versatile coating material to improve conventional therapies. In vitro studies showed that this strategy could eliminate EMT-type cancer cells. Mechanism studies also revealed that EIN was able to down-regulate the downstream expression of metastasis-associated factors, decrease the migration ability of cancer cells and prevent cancer cells from gaining drug resistance. In vivo investigation revealed that EIN had superior ability to enhance the therapeutic effect of conventional nanomedicines and inhibit the EMT process. Our study indicates the promising use of EIN to make up for the deficiencies of chemotherapy may provide insights into systematic cancer therapy to overcome tumor metastasis and drug resistance.


Asunto(s)
Anticarcinógenos/uso terapéutico , Catequina/análogos & derivados , Transición Epitelial-Mesenquimal/efectos de los fármacos , Nanopartículas del Metal/uso terapéutico , Metástasis de la Neoplasia/prevención & control , Animales , Anticarcinógenos/síntesis química , Anticarcinógenos/farmacología , Catequina/síntesis química , Catequina/farmacología , Catequina/uso terapéutico , Línea Celular Tumoral , Movimiento Celular , Doxorrubicina/química , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hierro/química , Nanopartículas del Metal/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Int J Clin Exp Pathol ; 10(9): 9427-9435, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-31966815

RESUMEN

BACKGROUND: The distribution and functional integrity of members of the tripartite motif (TRIM) protein family are essential for cell proliferation, development and apoptosis, and TRIM proteins have been linked to various cancers. To explore the diagnostic potential and mechanisms of TRIM27 in human spermatogenesis and oogenesis, we analyzed its localization pattern and putative roles in human testes and ovaries. METHODS: TRIM27 mRNA and protein levels in human testes and ovaries were investigated using RT-PCR and western blotting, respectively. TRIM27 was abundantly transcribed in human testes and ovaries, particularly during the early stages of spermatogenesis, and localized in the nuclei of primary spermatocytes. Immunofluorescence also revealed a diffuse distribution in the cytoplasm of round spermatids, and the protein was abundant in ovary tissue during various stages of oogenesis development. RESULTS: TRIM27 mRNA and protein was abundantly transcribed in male and female human germ cells by RT-PCR and western blotting in the human testes followed by the ovary. Immunohistochemical results revealed TRIM27 protein was abundant in the sex body of primary spermatocytes undergoing meiotic prophase during the first cycle of spermatogenesis. Moreover, Trim27 was diffusely localized in the cytoplasm of spermatids and round spermatids. Furthermore, TRIM27 was localized to both the nucleus and cytoplasm of human ovary cells. CONCLUSIONS: TRIM27 as a gametogenesis-related protein could play multiple roles in the regulation of sex body formation and germ cell proliferation during spermatogenesis and oogenesis. The identification and characterization of TRIM27 enhances our understanding of the molecular mechanisms underpinning its functions, and provides insight into its potential role in the pathogenesis of germ cell differentiation and infertility.

13.
ACS Appl Mater Interfaces ; 9(1): 255-265, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-27966867

RESUMEN

Intracellularly biotriggered decomposition of gene vectors is generally thought to benefit transfection. However, the bioresponsiveness is far from satisfactory, and the exact role of biodecomposition in the transfection process remains unclear to date. To overcome the challenges, highly rapid bioresponse of vectors has to be achieved so as to greatly amplify the intracellular deviation compared with the noncontrolled pattern. To this end, a supramolecular polyrotaxane has been elaborately designed by integrating reversible dynamics of supramolecular assembly and chemically labile bonds, in order to effectively propel intracellular decomposition. Inside tumor cells, the redox-responsive bulk dissociation of the supramolecular vector readily took place and was further accelerated by the lysosomal-acidity-triggered terminal decomposition. Both the in vitro and in vivo experiments have demonstrated that this supramolecule could mediate considerably more rapid gene accumulation in nuclei than the nonresponsive controls including PEI25K, the gold standard of nonviral vectors. Along with the structural decomposition, the supramolecule simultaneously underwent the transition of fluorescence quenching, favoring the evaluation over the bioresponsiveness inside cells. Based on the resulting data, it is suggested that the biotriggered volume expansion of supramolecule/DNA complexes may be the major factor accounting for that dramatically accelerated transnuclear gene transport during cellular mitosis, thus affecting the transfection. This study offers an understanding of the intracellular gene transport from a new viewpoint.


Asunto(s)
Vectores Genéticos/genética , Núcleo Celular , ADN , Humanos , Oxidación-Reducción , Transfección
14.
Nano Lett ; 17(1): 284-291, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28027643

RESUMEN

Discovering advanced materials for regulating cell death is of great importance in the development of anticancer therapy. Herein, by harnessing the recently discovered oxidative stress regulation ability of p53 and the Fenton reaction inducing capability of metal-organic network (MON), MON encapsulated with p53 plasmid (MON-p53) was designed to eradicate cancer cells via ferroptosis/apoptosis hybrid pathway. After confirming the detailed mechanism of MON-p53 in evoking ferroptosis, we further discovered that MON-p53 mediated a "bystander effect" to further sensitize cancer cells toward the MON-p53 induced ferroptosis. A 75-day anticancer experiment indicated that MON-p53 treatment not only suppressed the tumor growth but also prolonged the life-span of tumor bearing mice. Owing to its ability to promote intracellular oxidative stress, MON-p53 decreased the blood metastasis, lung metastasis, and liver metastasis. As a consequence, discovering methods to induce cell ferroptosis would provide a new insight in designing anticancer materials.


Asunto(s)
Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Estructuras Metalorgánicas/administración & dosificación , Neoplasias/terapia , Polifenoles/química , Proteína p53 Supresora de Tumor/genética , Antineoplásicos/farmacología , Muerte Celular , Línea Celular Tumoral , Genes p53 , Terapia Genética , Humanos , Estructuras Metalorgánicas/farmacología , Nanoestructuras , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Plásmidos , Propiedades de Superficie
15.
Nanoscale ; 8(37): 16702-16709, 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27714082

RESUMEN

In this study, we report a novel redox-responsive mesoporous silica nanoparticle (MSN)-based nanocarrier, capping with a therapeutic peptide ((RGDWWW)2KC) containing a RGD target motif, for tumor targeting synergistic therapy, which is designated as TTSTMSN. The MSN was decorated with a tumor-targeting therapeutic peptide as a potential gatekeeper. The two branched peptides containing rich tryptophans allowed the pores to be blocked via π-π stacking and hydrophobic interactions. Once the drug loaded nanoparticles were taken up by the cancer cells through integrin-mediated endocytosis, the therapeutic peptide capping shells on the surface of MSNs were released, inducing the loaded drug to diffuse into the cytoplasm after breaking of the disulfide bonds, triggered by the high concentration of glutathione (GSH) in cancer cells. At the same time, the falling therapeutic rich tryptophans in the branched chains interacted with DNA due to the indole rings, leading to disturbance of the DNA structure through the strong π interactions and causing cell apoptosis. There is no such report on capping of drug loaded porous silica with a therapeutic peptide shell, co-delivering an anticancer drug and therapeutic agent for tumor targeting synergistic therapy, which will have great potential in developing multifunctional nanocarriers based on therapeutic peptides for synergistic treatment.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas , Péptidos/administración & dosificación , Dióxido de Silicio , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Doxorrubicina , Humanos , Neoplasias/tratamiento farmacológico , Oligopéptidos , Oxidación-Reducción , Péptidos/farmacología , Porosidad
16.
ACS Appl Mater Interfaces ; 8(38): 25060-8, 2016 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-27595983

RESUMEN

In this report, an amphiphilic mitochondria-targeted chimeric peptide-based drug delivery system (DDS) was designed to overcome drug resistance. In vitro studies revealed that chimeric peptide could encapsulate doxorubicin (DOX) with high efficacy and target tumor mitochondria, realizing controlled release of DOX and in situ photodynamic therapy (PDT) in mitochondria. Importantly, reactive oxygen species (ROS) during PDT significantly disrupted mitochondria, leading to a dramatic decrease of intracellular adenosine 5'-triphophate (ATP). As a result, ATP-dependent efflux of DOX was remarkably inhibited. Trinitarian therapeutic strategy was developed to ablation of drug-resistant cells, that is, (1) enhanced cellular uptake of hydrophobic DOX via encapsulation in DDS, (2) combined chemo-/photodynamic therapies, and (3) suppressed generation of intracellular ATP as well as drug efflux via in situ PDT in mitochondria. This trinitarian strategy may open a new window in the fabrication of subcellular organelle destructive DDS in overcoming drug resistance.


Asunto(s)
Mitocondrias , Línea Celular Tumoral , Doxorrubicina , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Humanos , Péptidos
17.
ACS Appl Mater Interfaces ; 8(35): 22892-9, 2016 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-27513690

RESUMEN

Activatable photosensitizers (aPSs) have emerged as promising photodynamic therapy (PDT) agents for simultaneous imaging and selective ablation of cancer. However, traditional synthetic aPSs are limited by complex design and tedious synthesis. Here, aPS regulated by cucurbit[8]uril (CB[8]) for targeted cancer imaging and PDT is reported. This system is based on the host-guest interaction between biotinylated toluidine blue (TB-B) and CB[8] to form 2TB-B@CB[8]. Moreover, a facile strategy to turn off/on the fluorescence and photodynamic activity of TB-B is developed through the reversible assembly/disassembly of 2TB-B@CB[8]. This established system can achieve selective accumulation in tumor, light-up cancer imaging, and enhanced anticancer behavior. Therefore, this work provides a novel and promising strategy for the aPS build via simple and facile regulation of supramolecular chemistry.


Asunto(s)
Fármacos Fotosensibilizantes/química , Hidrocarburos Aromáticos con Puentes , Fluorescencia , Imidazoles , Neoplasias , Fotoquimioterapia
18.
Nano Lett ; 16(9): 5895-901, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27513184

RESUMEN

The ultimate goal in cancer therapy and diagnosis is to achieve highly specific targeting to cancer cells. Coated with the source cancer cell membrane specifically derived from the homologous tumors, the nanoparticles are identified with the self-recognition internalization by the source cancer cell lines in vitro and the highly tumor-selective targeting "homing" to the homologous tumor in vivo even in the competition of another heterologous tumor. As the result, MNP@DOX@CCCM nanovehicle showed strong potency for tumor treatment in vivo and the MR imaging. This bioinspired strategy shows great potential for precise therapy/diagnosis of various tumors merely by adjusting the cell membrane source accordingly on the nanoparticle surface.


Asunto(s)
Membrana Celular/química , Sistemas de Liberación de Medicamentos , Nanopartículas , Animales , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Compuestos Férricos/química , Humanos , Imagen por Resonancia Magnética , Magnetismo , Ratones , Neoplasias/tratamiento farmacológico
19.
Nano Lett ; 16(7): 4341-7, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27327876

RESUMEN

Fighting metastasis is a major challenge in cancer therapy, and stimulation of the immune system is of particular importance in the treatment of metastatic cancers. Here, an integrated theranostic nanoplatform was developed for the efficient treatment of highly metastatic tumors. Versatile functions including "And" logically controlled drug release, prolonged circulation time, tumor targeting, and anti-metastasis were integrated into doxorubicin (DOX) loaded, highly integrated mesoporous silica nanoparticles (DOX@HIMSNs) for a systemic treatment of highly metastatic triple negative breast cancer (TNBC). It was found that the good therapeutic effect of DOX@HIMSN was only partially attributed to its anticancer cytotoxicity. Most importantly, DOX@HIMSN could induce anticancer immune responses including dendritic cell (DC) maturation and antitumor cytokine release. Compared with the traditional tumor chemotherapy, the integrated theranostic nanoplatform we developed not only improved the tumor specific cytotoxicity but also stimulated antitumor immune responses during the treatment.


Asunto(s)
Doxorrubicina/administración & dosificación , Inmunoterapia , Nanopartículas , Nanomedicina Teranóstica , Línea Celular Tumoral , Humanos , Dióxido de Silicio
20.
Biomaterials ; 77: 149-63, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26599622

RESUMEN

To improve the tumor therapeutic efficiency and reduce undesirable side effects, ternary FK/p53/PEG-PLL(DA) complexes with a detachable surface shielding layer were designed. The FK/p53/PEG-PLL(DA) complexes were fabricated by coating the folate incorporated positively charged FK/p53 complexes with charge-switchable PEG-shield (PEG-PLL(DA)) through electrostatic interaction. At the physiological pH 7.4 in the bloodstream, PEG-PLL(DA) could extend the circulating time by shielding the positively charged FK/p53 complexes. After the accumulation of the FK/p53/PEG-PLL(DA) complexes in tumor sites, tumor-acidity-triggered charge switch led to the detachment of PEG-PLL(DA) from the FK/p53 complexes, and resulted in efficient tumor cell entry by folate-mediated uptake and electrostatic attraction. Stimulated by the high content glutathione (GSH) in cytoplasm, the cleavage of disulfide bond resulted in the liberation of proapoptosis peptide C-KLA(TPP) and the p53 gene, which exerted the combined tumor therapy by regulating both intrinsic and extrinsic apoptotic pathways. Both in vitro and in vivo studies confirmed that the ternary detachable complexes FK/p53/PEG-PLL(DA) could enhance antitumor efficacy and reduce adverse effects to normal cells. These findings indicate that the tumor-triggered decomplexation of FK/p53/PEG-PLL(DA) supplies a useful strategy for targeting delivery of different therapeutic agents in synergetic anticancer therapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Péptidos/administración & dosificación , Polietilenglicoles/administración & dosificación , Polilisina/análogos & derivados , Profármacos/administración & dosificación , Proteína p53 Supresora de Tumor/administración & dosificación , Activación Metabólica , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Portadores de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Receptores de Folato Anclados a GPI/metabolismo , Genes p53 , Glutatión/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Neoplasias Hepáticas Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH , Tamaño de la Partícula , Péptidos/farmacocinética , Péptidos/uso terapéutico , Péptidos/toxicidad , Polilisina/administración & dosificación , Profármacos/farmacocinética , Electricidad Estática , Propiedades de Superficie , Transfección , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA