Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Acta Biomater ; 136: 456-472, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34562660

RESUMEN

The synergistic manipulation of autophagy blocking with tumor targeting and penetration effects to enhance cancer cell killing during photothermal therapy (PTT) remains a substantial challenge. Herein, we fabricated a biomimetic nanoplatform by precisely coating homologous prostate cancer cell membranes (CMs) onto the surface of mesoporous polydopamine nanoparticles (mPDA NPs) encapsulating the autophagy inhibitor chloroquine (CQ) for synergistically manipulating PTT and autophagy for anticancer treatment. The resulting biomimetic mPDA@CMs NPs-CQ system could escape macrophage phagocytosis, overcome the vascular barrier, and home in the homologous prostate tumor xenograft with high tumor targeting and penetrating efficiency. The mPDA NPs core endowed the mPDA@CMs NPs-CQ with good photothermal capability to mediate PTT killing of prostate cancer cells, while NIR-triggered CQ release from the nanosystem further arrested PTT-induced protective autophagy of cancer cells, thus weakening the resistance of prostate cancer cells to PTT. This combined PTT killing and autophagy blocking anticancer strategy could induce significant autophagosome accumulation, ROS generation, mitochondrial damage, endoplasmic reticulum stress, and apoptotic signal transduction, which finally results in synergistic prostate tumor ablation in vivo. This prostate cancer biomimetic nanosystem with synergistically enhanced anticancer efficiency achieved by manipulating PTT killing and autophagy blocking is expected to serve as a more effective anticancer strategy against prostate cancer. STATEMENT OF SIGNIFICANCE: Autophagy is considered as one of the most efficient rescuer and reinforcement mechanisms of cancer cells against photothermal therapy (PTT)-induced cancer cell eradication. How to synergistically manipulate autophagy blocking with significant tumor targeting and penetration to enhance PTT-mediated cancer cell killing remains a substantial challenge. Herein, we fabricated a biomimetic nanoplatform by precisely coating homologous cancer cell membranes onto the surface of mesoporous polydopamine nanoparticles with encapsulation of the autophagy inhibitor chloroquine for synergistic antitumor treatment with high tumor targeting and penetrating efficiency both in vitro and in vivo. This biomimetic nanosystem with synergistically enhanced anticancer efficiency by manipulating PTT killing and autophagy blocking is expected to serve as a more effective anticancer strategy.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Autofagia , Biomimética , Humanos , Indoles , Masculino , Fototerapia , Polímeros
2.
Bioinorg Chem Appl ; 2021: 5534870, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868396

RESUMEN

How to actively target tumor sites manipulating the controllable release of the encapsulated anticancer drugs and photosensitizers for synergistic anticancer therapy remains a big challenge. In this study, a cancer cell-targeted, near-infrared (NIR) light-triggered and anticancer drug loaded liposome system (LPs) was developed for synergistic cancer therapy. Photosensitizer indocyanine green (ICG) and chemotherapy drug Curcumin (CUR) were coencapsulated into the liposomes, followed by the surface conjugation of GE11 peptide for epidermal growth factor receptor (EGFR) targeting on the cancer cell surface. Strictly controlled by NIR light, GE11 peptide modified and CUR/ICG-loaded LPs (GE11-CUR/ICG-LPs) could introduce hyperthermia in EGFR overexpressed A549 cancer cells for photothermal therapy, which could also trigger the increased release of CUR for enhanced cancer cell inhibition. GE11-CUR/ICG-LPs synergized photochemotherapy could induce reactive oxygen species (ROS) generation and cytoskeleton disruption to activate stronger apoptotic signaling events than the photothermal therapy or chemotherapy alone by regulating Bax/Bcl-2 and PI3K/AKT pathways. This EGFR-targeted drug-delivery nanosystem with NIR sensitivity may potentially serve in more effective anticancer therapeutics with reduced off-target effects.

3.
Acta Biomater ; 121: 605-620, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33259958

RESUMEN

How to enable protein degradation pathways including the autophagy-lysosome pathway (ALP) and the ubiquitin-proteasome system (UPS) to enhance the efficacy of anticancer treatments remains a substantial challenge. Cobalt oxide nanoparticles (Co3O4 NPs) have attracted interest in recent years for their potential use as a synergistic anticancer treatment, although their therapeutic mechanisms of action are still poorly understood. Here, we describe the synergistic use of Co3O4 NPs as an autophagy inhibitor, chemosensitizer and photosensitizer, which manipulate protein degradation pathways (ALP and UPS) and photothermal therapy for enhanced anticancer treatments both in vitro and in vivo. We show that Co3O4 NPs can induce autolysosome accumulation and lysosomal functions damage by inhibiting lysosomal proteolytic activity and reducing intracellular ATP levels. Notably, Co3O4 NPs can be combined with the proteasome inhibitor, Carfilzomib (Cfz), to promote the accumulation of autophagic substrates, protein ubiquitination, and endoplasmic reticulum stress, and in doing so, inhibit cancer progression. By taking advantage of their photothermal conversion efficiency, Co3O4 NPs can also serve as photothermal sensitizer, which synergistically enhances the anticancer efficacy of Cfz both in vitro and in vivo. In summary, we provide evidence of a nanomaterial-synergized, photothermal anticancer strategy that synergistically targets cancer cell survival pathways and may eventually serve to enhance the anticancer efficacy of established cancer therapeutics.


Asunto(s)
Nanopartículas , Fototerapia , Línea Celular Tumoral , Cobalto/farmacología , Óxidos , Proteolisis
4.
Angew Chem Int Ed Engl ; 59(8): 3226-3234, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31756258

RESUMEN

Pathogenesis hallmarks for tuberculosis (TB) are the Mycobacterium tuberculosis (Mtb) escape from phagolysosomal destruction and limited drug delivery into infected cells. Several nanomaterials can be entrapped in lysosomes, but the development of functional nanomaterials to promote phagolysosomal Mtb clearance remains a big challenge. Here, we report on the bactericidal effects of selenium nanoparticles (Se NPs) against Mtb and further introduce a novel nanomaterial-assisted anti-TB strategy manipulating Ison@Man-Se NPs for synergistic drug-induced and phagolysosomal destruction of Mtb. Ison@Man-Se NPs preferentially entered macrophages and accumulated in lysosomes releasing Isoniazid. Surprisingly, Ison@Man-Se/Man-Se NPs further promoted the fusion of Mtb into lysosomes for synergistic lysosomal and Isoniazid destruction of Mtb. Concurrently, Ison@Man-Se/Man-Se NPs also induced autophagy sequestration of Mtb, evolving into lysosome-associated autophagosomal Mtb degradation linked to ROS-mitochondrial and PI3K/Akt/mTOR signaling pathways. This novel nanomaterial-assisted anti-TB strategy manipulating antimicrobial immunity and Mtb clearance may potentially serve in more effective therapeutics against TB and drug-resistant TB.


Asunto(s)
Antibacterianos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Isoniazida/química , Macrófagos/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Nanopartículas/química , Selenio/química , Tuberculosis/tratamiento farmacológico , Humanos , Tuberculosis/patología
5.
Biomed Pharmacother ; 103: 1592-1601, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29864947

RESUMEN

Atomic force microscopy (AFM) is appropriately applied to the examination of hard surfaces and soft samples with extremely high resolution and ultrasensitive force, which cannot be obtained by other imaging techniques, including optical and electron microscopy. In the current study, AFM was employed to evaluate the anti-arthritic effect of licochalcone A (LCA), a flavonoid isolated from the root of Chinese medicinal herb Glycyrrhiza inflate, on rheumatoid arthritis synovial fibroblasts (RASFs) at the nanoscale for the first time. The morphology, ultrastructure and stiffness of RASFs was modified by LCA as determined by AFM, suggesting that LCA most likely exerts an anti-arthritic effect based on the key role of RASFs in the progression of RA. Further studies showed that the inhibitory effect of LCA on IκBα phosphorylation and degradation as well as on p65 nuclear translocation and phosphorylation contributed to altering the morphology, ultrastructure and stiffness of the RASF membrane. Interestingly, IKKß phosphorylation was not detectable in RASFs, indicating that LCA altered the morphology, ultrastructure and stiffness of the RASF membrane by inhibiting NF-κB activation independent of IKKß phosphorylation. Antigen-induced arthritis (AIA) was established in Sprague Dawley (SD) rats to validate the anti-arthritic effect of LCA, and LCA significantly decreased both the arthritis scores and paw swelling in the AIA rats, suggesting that LCA inhibits the progression and development of arthritis in vivo. Collectively, AFM provides evidence at the nanoscale to predict the anti-arthritic effect of drugs on RASFs, and LCA should be further investigated as a candidate agent for the treatment of arthritis.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Chalconas/uso terapéutico , Microscopía de Fuerza Atómica , FN-kappa B/metabolismo , Transducción de Señal , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Artritis Reumatoide/patología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Chalconas/química , Chalconas/farmacología , Módulo de Elasticidad , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibroblastos/ultraestructura , Masculino , Inhibidor NF-kappaB alfa/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Ratas Sprague-Dawley , Membrana Sinovial/patología
6.
Pathol Res Pract ; 214(5): 691-699, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29567333

RESUMEN

Matrine, as a natural alkaloid isolated from the traditional herb medicine sophora flavescens, has been proved to possess excellent biological activities, including anticancer effects. Now, this research aims to assess the anticancer activities and the mechanism of matrine against esophageal cancer cells, we investigated the proliferative inhibition, apoptosis induction, as well as the underlying mechanism of matrine on esophageal cancer KYSE-150 cells. It was found that matrine could suppress KYSE-150 cell proliferation and significantly mediate cell apoptosis in a dose-dependent relation by increasing intracellular reactive oxygen species level and triggering mitochondrial membrane potential disruption. More precise mechanism studies demonstrated that matrine could up-regulate the expression of Bax proteins and down-regulate the expression of Bcl-2 proteins, as well as the activation about caspase-3, 8 and 9 in KYSE-150 cells. The morphological analysis of KYSE-150 cells exhibited that matrine could destroy the F-actin and nuclei structures and induce morphological damage with increased surface height distribution and roughness of cell membrane. These results not only demonstrated the potential anticancer activity mechanism of matrine at nanoscale, but also provide preliminary guidance for the treatment of esophageal cancer using matrine.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Mitocondrias/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Alcaloides , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Quinolizinas , Proteína X Asociada a bcl-2/metabolismo , Matrinas
7.
Nanoscale ; 9(42): 16365-16374, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29052674

RESUMEN

Poor bioavailability and non-specificity of chemotherapeutic agents are major challenges in breast cancer treatment. Antibodies and small molecules that block cell signaling pathways have shown promise in the clinic, but their application is also limited by the high costs and treatment dosages required. Novel therapies that aim to rapidly and specifically target malignant cells with long-lasting impact in the tumor microenvironment may ultimately improve clinical outcome in cancer patients. Here, we demonstrate that epidermal growth factor receptor (EGFR)-targeting GE11 peptides conjugated with PEGylated polylactic-co-glycolic acid (PLGA) nanoparticles can be used to effectively deliver an anti-cancer agent, curcumin, into EGFR-expressing MCF-7 cells in vitro and in vivo. Treatment of breast cancer cells and tumor-bearing mice with these curcumin-loaded nanoparticles gave rise to reduced phosphoinositide 3-kinase signaling, decreased cancer cell viability, attenuated drug clearance from the circulation, and suppressed tumor burden compared with free curcumin or non-EGFR targeting nanoparticles. The targeted nanoscale drug delivery system we describe here may provide a new strategy for the design of targeted cancer therapy vectors. Our study provides evidence that the efficacy of pharmacologic anti-cancer agents can be enhanced through their delivery in the form of modified nanoparticles that effectively target specific malignant cell types.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Curcumina/administración & dosificación , Portadores de Fármacos , Receptores ErbB/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Animales , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas , Fosfatidilinositol 3-Quinasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Drug Deliv ; 24(1): 1549-1564, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29019267

RESUMEN

Selenium nanoparticles (Se NPs) have attracted increasing interest in recent decades because of their anticancer, immunoregulation, and drug carrier functions. In this study, GE11 peptide-conjugated Se NPs (GE11-Se NPs), a nanosystem targeting EGFR over-expressed cancer cells, were synthesized for oridonin delivery to achieve enhanced anticancer efficacy. Oridonin loaded and GE11 peptide conjugated Se NPs (GE11-Ori-Se NPs) were found to show enhanced cellular uptake in cancer cells, which resulted in enhanced cancer inhibition against cancer cells and reduced toxicity against normal cells. After accumulation into the lysosomes of cancer cells and increase of oridonin release under acid condition, GE11-Ori-Se NPs were further transported into cytoplasm after the damage of lysosomal membrane integrity. GE11-Ori-Se NPs were found to induce cancer cell apoptosis by inducting reactive oxygen species (ROS) production, activating mitochondria-dependent pathway, inhibiting EGFR-mediated PI3K/AKT and inhibiting Ras/Raf/MEK/ERK pathways. GE11-Se NPs were also found to show active targeting effects against the tumor tissue in esophageal cancer bearing mice. And in nude mice xenograft model, GE11-Ori-Se NPs significantly inhibited the tumor growth via inhibition of tumor angiogenesis by reducing the angiogenesis-marker CD31 and activation of the immune system by enhancing IL-2 and TNF-α production. The selenium contents in mice were found to accumulate into liver, tumor, and kidney, but showed no significant toxicity against liver and kidney. This cancer-targeted design of Se NPs provides a new strategy for synergistic treating of cancer with higher efficacy and reduced side effects, introducing GE11-Ori-Se NPs as a candidate for further evaluation as a chemotherapeutic agent for EGFR over-expressed esophageal cancers.


Asunto(s)
Antineoplásicos/farmacología , Diterpenos de Tipo Kaurano/farmacología , Receptores ErbB/antagonistas & inhibidores , Péptidos/farmacología , Selenio/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Diterpenos de Tipo Kaurano/administración & dosificación , Diterpenos de Tipo Kaurano/farmacocinética , Portadores de Fármacos/química , Liberación de Fármacos , Humanos , Interleucina-2/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Nanopartículas/química , Péptidos/administración & dosificación , Péptidos/farmacocinética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Selenio/farmacocinética , Factor de Necrosis Tumoral alfa/biosíntesis
9.
Scanning ; 38(6): 901-912, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27487564

RESUMEN

Dihydromyricetin (DMY) a flavonoid derived from medicinal plant Ampelopsis grossedentata, possesses anti-oxidative and anti-inflammatory effects in vitro, however, the in vivo anti-inflammatory action of DMY remains unknown. In the current study, carrageenan-induced paw edema in rat, an acute inflammation model, and RAW264.7 macrophages activated by LPS were employed to evaluate the anti-inflammatory potency of DMY in vivo and in vitro. Results showed that DMY significantly attenuated rat paw edema induced by carrageenan. Also, DMY markedly inhibited NO secretion, iNOS, and COX-2 protein expression, as well as p65 phosphorylation via suppression of IKKß activity and IKKα/ß phosphorylation in RAW264.7 cells. And using high resolution Atomic Force Microscope (AFM), we also proved that DMY prevented morphological change and membrane alterations of RAW 264.7 macrophages caused by LPS stimulation. As activation of macrophages is one of major factors in carrageenan-induced paw edema of rats, the anti-inflammatory action of DMY is suggested to be closely associated with suppression of macrophage activation. These findings indicate that DMY is valuable of being further investigated as a candidate new agent for treating inflammatory conditions, and suggest that AFM could be a powerful nanotool for anti-inflammatory investigations. SCANNING 38:901-912, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Antiinflamatorios/farmacología , Flavonoles/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Animales , Femenino , Células HEK293 , Humanos , Macrófagos/inmunología , Ratones , Microscopía de Fuerza Atómica , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Células RAW 264.7 , Ratas , Ratas Wistar , Factor de Transcripción ReIA/antagonistas & inhibidores
10.
Scanning ; 38(6): 792-801, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27280953

RESUMEN

Polysaccharide compounds (PCs), which composed of different kinds of polysaccharides always isolated from different kinds of traditional Chinese medicine, are now attracting more and more attentions due to their strong immunomodulatory activities beyond the corresponding one-component polysaccharides. In this study, we demonstrated for the first time that PCs-1 and PCs-2 had strong immunomodulatory effects on macrophages both in in vitro and in vivo models by atomic force microscopy (AFM). By high resolution AFM imaging, PCs-1 and PCs-2 were found to inhibit LPS induced cell surface particle size and roughness increase in RAW264.7 macrophages, demonstrating the anti-inflammatory effects of PCs-1 and PCs-2 on macrophages. PCs-1 and PCs-2 were also proved to increase the particle size and roughness of resting RAW264.7 macrophages, which suggested that PCs could activate resting RAW264.7 macrophages. And additionally, PCs-1 and PCs-2 were also found to reverse the surface particle size and roughness decrease of peritoneal macrophages isolated from cyclophosphamide induced immunosuppressive mice, suggesting the activation effects of PCs-1 and PCs-2 on immunosuppressive macrophages. These results further enhanced our understanding of macrophage activations by direct imaging of cell surface ultrastructure and also highlighted AFM as a novel nanotool for macrophage detections. And most importantly, these results also indicated the outstanding immunomodulatory effects of PCs on macrophages, which therefore suggested that PCs could be served as a kind of novel immunomodulatory agents that would benefit human health. SCANNING 38:792-801, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Factores Inmunológicos/farmacología , Macrófagos/efectos de los fármacos , Polisacáridos/farmacología , Animales , Células Cultivadas , Activación de Macrófagos/efectos de los fármacos , Macrófagos/ultraestructura , Ratones , Ratones Endogámicos BALB C , Microscopía de Fuerza Atómica
11.
Bioorg Med Chem Lett ; 23(23): 6296-303, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24140445

RESUMEN

Selenium nanoparticles (Se NPs) have been served as promising materials for biomedical applications, especially for cancer treatment. The anti-cancer effects of Se NPs against cancer cells have been widely studied in recent years, but whether Se NPs can induce the changes of cell membrane bio-mechanical properties in cancer cells still remain unexplored. In this Letter, we prepared Se NPs for investigating the intracellular localization of Se NPs in MCF-7 cells and determined the effects of Se NPs on apoptosis and necrosis in MCF-7 cells. Especially, we reported for the first time about the effects of Se NPs on the bio-mechanical properties of cancer cells and found that Se NPs could remarkably decrease the adhesion force and Young's modulus of MCF-7 cells. To further understand the potential mechanisms about how Se NPs affect the bio-mechanical properties of MCF-7 cells, we also investigated the expression of CD44 molecules, the structure and the amounts of F-actin. The results indicated that the decreased adhesion force between AFM tip and cell membrane was partially due to the changes of membrane molecules induced by Se NPs, such as the down-regulation of trans-membrane CD44 molecules. Additionally, the decrease of Young's modulus of MCF-7 cells was due to the dis-organization and down-regulation of F-actin induced by Se NPs. These results collectively suggested that cell membrane was of vital importance in Se NPs induced toxicity in cancer cells, which could be served as a potential target for cancer treatment by Se NPs.


Asunto(s)
Actinas/metabolismo , Nanopartículas/química , Nanopartículas/uso terapéutico , Selenio/química , Selenio/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Humanos , Receptores de Hialuranos/biosíntesis , Lisosomas/química , Lisosomas/metabolismo , Células MCF-7 , Microscopía de Fuerza Atómica
12.
Bioorg Med Chem Lett ; 23(10): 2902-8, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23570787

RESUMEN

Germanium (Ge) is considered to play a key role in the pharmacological effects of some medicinal plants. Here, two new Ge(IV)-polyphenol complexes were synthesized and measured for their potential biological activities. The results indicated that these Ge(IV)-polyphenol complexes possessed great anti-oxidative activities, both showing stronger hydroxyl scavenging effects than their corresponding ligands. We also demonstrated the strong intercalating abilities of Ge(IV)-polyphenol complexes into calf thymus-DNA molecules. In addition, these two Ge(IV)-polyphenol complexes showed strong proliferative inhibition effect on HepG2 cancer cells. Moreover, the morphological changes in HepG2 cells induced by Ge(IV)-polyphenol complexes were detected by atomic force microscopy. All these results collectively suggested that Ge(IV)-polyphenol complexes could be served as promising pharmacologically active substances against cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Germanio/química , Compuestos Organometálicos/farmacología , Polifenoles/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células Hep G2 , Humanos , Estructura Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Relación Estructura-Actividad
13.
Appl Microbiol Biotechnol ; 97(3): 1051-62, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22945264

RESUMEN

Selenium nanoparticles (Se NPs) have been recognized as promising materials for biomedical applications. To prepare Se NPs which contained cancer targeting methods and to clarify the cellular localization and cytotoxicity mechanisms of these Se NPs against cancer cells, folic acid protected/modified selenium nanoparticles (FA-Se NPs) were first prepared by a one-step method. Some morphologic and spectroscopic methods were obtained to prove the successfully formation of FA-Se NPs while free folate competitive inhibition assay, microscope, and several biological methods were used to determine the in vitro uptake, subcellular localization, and cytotoxicity mechanism of FA-Se NPs in MCF-7 cells. The results indicated that the 70-nm FA-Se NPs were internalized by MCF-7 cells through folate receptor-mediated endocytosis and targeted to mitochondria located regions through endocytic vesicles transporting. Then, the FA-Se NPs entered into mitochondria; triggered the mitochondria-dependent apoptosis of MCF-7 cells which involved oxidative stress, Ca(2)+ stress changes, and mitochondrial dysfunction; and finally caused the damage of mitochondria. FA-Se NPs released from broken mitochondria were transported into nucleus and further into nucleolus which then induced MCF-7 cell cycle arrest. In addition, FA-Se NPs could induce cytoskeleton disorganization and induce MCF-7 cell membrane morphology alterations. These results collectively suggested that FA-Se NPs could be served as potential therapeutic agents and organelle-targeted drug carriers in cancer therapy.


Asunto(s)
Antineoplásicos/toxicidad , Ácido Fólico/metabolismo , Nanopartículas/toxicidad , Selenio/toxicidad , Antineoplásicos/metabolismo , Apoptosis , Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Endocitosis , Humanos , Células MCF-7 , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Selenio/metabolismo
14.
Scanning ; 35(5): 316-26, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23239560

RESUMEN

Integrin-mediated human umbilical vein endothelial cells (HUVECs) adhesion to the extracellular matrix plays a fundamental role in tumor-induced angiogenesis. Celastrol, a traditional Chinese medicine plant, has possessed anticancer and suppressed angiogenesis activities. Here, the mechanism underling the antiangiogenesis capacity of celastrol was investigated by exploring the effect of celastrol on ß1(CD29) integrin-mediated cell adhesion and migration. Flow cytometry results showed that the HUVECs highly expressed CD29 and cell adhesion assay indicated that celastrol specifically inhibited the adhesion of HUVECs to fibronectin (FN) without affecting nonspecific adhesion to poly-L-lysine (PLL). After cell FN adhesion being inhibited, the cell surface nanoscale structure and adhesion force were detected by atomic force microscope (AFM). High-resolution imaging revealed that cell morphology and ultrastructure changed a lot after being treated with celastrol. The membrane average roughness (Ra) and the major forces were decreased from 31.34 ± 4.56 nm, 519.60 ± 82.86 pN of 0 µg/ml celastrol to 18.47 ± 6.53 nm, 417.79 ± 53.35 pN of 4.0 µg/ml celastrol, 10.54 ± 2.85 nm, 258.95 ± 38.98 pN of 8.0 µg/ml celastrol, respectively. Accompanying with the decrease of adhesion force, the actin cytoskeleton in the cells was obviously disturbed by the celastrol. All of these changes influenced the migration of HUVECs from the wound-healing migration assay. Taken together, our results suggest that celastrol can be as an inhibitor of HUVEC adhesion to FN. This work provides a novel approach to inhibition of tumor angiogenesis and tumor growth.


Asunto(s)
Antineoplásicos/metabolismo , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Integrina beta1/metabolismo , Triterpenos/metabolismo , Membrana Celular/ultraestructura , Forma de la Célula , Fibronectinas/metabolismo , Humanos , Microscopía de Fuerza Atómica , Triterpenos Pentacíclicos , Propiedades de Superficie/efectos de los fármacos
15.
Eur J Pharm Sci ; 47(1): 28-34, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22579957

RESUMEN

Quercetin is a popular flavonoid in plant foods, herbs, and dietary supplement. Germanium, a kind of trace elements, can enhance the body immunity. This study investigated the hydroxyl-radical-scavenging mechanism of the quercertin-germanium (IV) (Qu-Ge) complex to human erythrocytes, especially the effects on ultrastructure and mechanical properties of cell membrane, plasma membrane potential and intracellular free Ca(2+) concentration. Results showed that QuGe(2), a kind of the Qu-Ge complex, could reduce the oxidative damage of erythrocytes, change the cell-surface morphology, and partly recover the disruption of plasma membrane potential and intracellular free Ca(2+) level. Atomic force microscopy (AFM) was used to characterize the changes of the cell morphology, cell-membrane ultrastructure and biophysical properties at nanoscalar level. QuGe(2) has triggered the antioxidative factor to inhibit cellular damage. These results can improve the understanding of hydroxyl-radical-scavenging mechanism of human erythrocytes induced by the Qu-Ge complex, which can be potentially developed as a new antioxidant for treatment of oxidative damage.


Asunto(s)
Complejos de Coordinación/farmacología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Depuradores de Radicales Libres/farmacología , Germanio/farmacología , Radical Hidroxilo/sangre , Quercetina/farmacología , Antioxidantes/farmacología , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Células Cultivadas , Membrana Eritrocítica/efectos de los fármacos , Membrana Eritrocítica/metabolismo , Citometría de Flujo , Humanos , Potenciales de la Membrana/efectos de los fármacos , Microscopía de Fuerza Atómica , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
16.
Appl Microbiol Biotechnol ; 94(5): 1199-208, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22406860

RESUMEN

Photothermal cancer therapy, as a prospective approach for local cancer treatment, is attracting increasing interests. In this paper, gold nanorods were conjugated with folate (folate/AuNRs), and their photothermal effects on hepatocellular carcinoma cell line (HepG2) using MTT assay, flow cytometry, as well as on the cellular morphology, cytoskeleton, cell surface adhesion, and stiffness detected at subcellular level by an atomic force microscope (AFM) were investigated. The results indicated that near-infrared laser-induced hyperthermia of folate/AuNRs could break the cell membrane integrity and homeostasis and then lead to the depolymerization of cytoskeleton and influx of intracellular Ca(2+). Thus, folate/AuNRs can be as effective and promising nanomaterials for photothermal therapy of folate receptor bearing tumor.


Asunto(s)
Ácido Fólico/metabolismo , Oro/metabolismo , Hepatocitos/fisiología , Hepatocitos/efectos de la radiación , Nanotubos , Supervivencia Celular/efectos de la radiación , Citometría de Flujo , Células Hep G2 , Calor , Humanos , Luz , Microscopía de Fuerza Atómica , Fototerapia/métodos , Coloración y Etiquetado/métodos , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo
17.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 26(3): 561-5, 2009 Jun.
Artículo en Chino | MEDLINE | ID: mdl-19634673

RESUMEN

Several complexes with different mass ratios of hyaluronic acid to pectin were studied using AFM and IR at the room temperature kept by air conditioning. The results showed that hyaluronic acid and pectin were in the state of being complex and were consistent when the concentrations of hyaluronic acid and pectin were above 1 mg/ml and 5 mg/ml respectively, and the mass ratio was 1 : 5. The complex self-assembled to composite grain-shaped film. In comparison with simple hyaluronic acid, the viscosity of hylauronic acid and pectin complex was stronger, and water-solubility was lower. The complex has the bio-function of both hylauronic acid and pectin and has wide application potential in the field of biomedical engineering.


Asunto(s)
Fenómenos Químicos , Ácido Hialurónico/química , Pectinas/química , Solubilidad , Ingeniería de Tejidos , Viscosidad
18.
Scanning ; 26(4): 155-61, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15473266

RESUMEN

Information obtained by atomic force microscopy (AFM) depends strongly on the kind of probe or tip used; therefore, probe and tip effects have to be taken into account when verifying or interpreting the data acquired. In many papers, double-tip effects have been mentioned while other research was done; however, there are only a few special reports on double- or triple-tip effects, especially double-probe effects. In our paper, metaphase chromosomes of Chinese hamster ovary (CHO) cells, aggregates of pectin molecules, membrane surface of mouse embryonic stem cells, and R-phycoerythrin-conjugated immunoglobulin G complexes were imaged by AFM with high-quality probes, double-probe cantilever, and double-tip and triple-tip probes, respectively, in order to determine double-probe, double-tip, and triple-tip effects during AFM scanning. We found that the double-probe, double-tip, and triple-tip effects share the same principle, and that these effects correlate with distance and height differences between probes of double-probe cantilever or tips of double-tip or multiple-tip probes. Since many other factors influence double-probe or double-tip effects, more in-depth studies must be undertaken. However, this initial research will make all users of AFM techniques aware of double-probe and double-tip or triple-tip effects during AFM scanning and aid in verifying or interpreting the data acquired.


Asunto(s)
Microscopía de Fuerza Atómica/instrumentación , Animales , Membrana Celular , Cromosomas/ultraestructura , Cricetinae , Inmunoconjugados/ultraestructura , Inmunoglobulina G/química , Inmunoglobulina G/ultraestructura , Metafase , Ratones , Microscopía de Fuerza Atómica/métodos , Pectinas/ultraestructura , Ficoeritrina/química , Ficoeritrina/ultraestructura , Células Madre/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA