Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cancer Cell Int ; 24(1): 59, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38321552

RESUMEN

Peroxiredoxin 3 (PRDX3), a mitochondrial hydrogen peroxide scavenger, is known to be upregulated during tumorigenesis and cancer progression. In this study, we provide evidence for the first time that PRDX3 could regulate cellular signaling pathways associated with Matrix Metalloproteinase-1 (MMP-1) expression and activity in breast cancer progression. We show that shRNA-mediated gene silencing of PRDX3 inhibits cell migration and invasion in two triple-negative breast cancer cell lines. Reciprocal experiments show that PRDX3 overexpression promotes invasion and migration of the cancer cells, processes which are important in the metastatic cascade. Notably, this phenomenon may be attributed to the activation of MMP-1, which is observed to be upregulated by PRDX3 in the breast cancer cells. Moreover, immunohistochemical staining of breast cancer tissues revealed a positive correlation between PRDX3 and MMP-1 expression in both epithelial and stromal parts of the tissues. Further pathway reporter array and luciferase assay demonstrated that activation of ERK signaling is responsible for the transcriptional activation of MMP-1 in PRDX3-overexpressed cells. These findings suggest that PRDX3 could mediate cancer spread via ERK-mediated activation of MMP-1. Targeted inhibition of ERK signaling may be able to inhibit tumor metastasis in triple-negative breast cancer.

2.
Breast Cancer Res Treat ; 186(3): 655-665, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33625616

RESUMEN

PURPOSE: Breast cancer is the most common type of cancer affecting women worldwide. Phosphoglycerate dehydrogenase (PHGDH) is an oxidoreductase in the serine biosynthesis pathway. Although it has been reported to affect growth of various tumors, its role in breast cancer is largely unknown. This study aimed to analyze the expression of PHGDH in breast cancer tissue samples and to determine if PHGDH regulates breast cancer cell proliferation. METHODS: Tissue microarrays consisting of 305 cases of breast invasive ductal carcinoma were used for immunohistochemical evaluation of PHGDH expression. The role of PHGDH in breast cancer was investigated in vitro by knocking down its expression and determining the effect on cell proliferation and cell cycling, and in ovo by using a chorioallantoic membrane (CAM) assay. RESULTS: Immunohistochemical examination showed that PHGDH is mainly localized in the cytoplasm of breast cancer cells and significantly associated with higher cancer grade, larger tumor size, increased PCNA expression, and lymph node positivity. Analysis of the GOBO dataset of 737 patients demonstrated that increased PHGDH expression was associated with poorer overall survival. Knockdown of PHGDH expression in breast cancer cells in vitro resulted in a decrease in cell proliferation, reduction in cells entering the S phase of the cell cycle, and downregulation of various cell cycle regulatory genes. The volume of breast tumor in an in ovo CAM assay was found to be smaller when PHGDH was silenced. CONCLUSION: The findings suggest that PHGDH has a regulatory role in breast cancer cell proliferation and may be a potential prognostic marker and therapeutic target in breast cancer.


Asunto(s)
Neoplasias de la Mama , Fosfoglicerato-Deshidrogenasa , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Fosfoglicerato-Deshidrogenasa/genética , Pronóstico , Serina
3.
Ecotoxicol Environ Saf ; 208: 111702, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33396033

RESUMEN

Cellular models exhibiting human physiological features of pseudostratified columnar epithelia, provide a more realistic approach for elucidating detailed mechanisms underlying PM2.5-induced pulmonary toxicity. In this study, we characterized the barrier and mucociliary functions of differentiated human small airway epithelial cells (SAECs), cultured at the air-liquid interface (ALI). Due to the presence of mucociliary protection, particle internalization was reduced, with a concomitant decrease in cytotoxicity in differentiated S-ALI cells, as compared to conventional submerged SAEC cultures. After 24-hour exposure to PM2.5 surrogates, 117 up-regulated genes and 156 down-regulated genes were detected in S-ALI cells, through transcriptomic analysis using the Affymetrix Clariom™ S Human Array. Transcription-level changes in >60 signaling pathways, were revealed by functional annotation of the 273 differentially expressed genes, using the PANTHER Gene List Analysis. These pathways are involved in multiple cellular processes, that include inflammation and apoptosis. Exposure to urban PM2.5 led to complex responses in airway epithelia, including a net induction of downstream pro-inflammatory and pro-apoptotic responses. Collectively, this study highlights the importance of using the more advanced ALI model rather than the undifferentiated submerged model, to avoid over-assessment of inhaled particle toxicity in human. The results of our study also suggest that reduction of ambient PM2.5 concentrations would have a protective effect on respiratory health in humans.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Células Epiteliales/efectos de los fármacos , Material Particulado/toxicidad , Transcriptoma/efectos de los fármacos , Contaminantes Atmosféricos/química , Apoptosis/efectos de los fármacos , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/patología , Perfilación de la Expresión Génica , Humanos , Tamaño de la Partícula , Material Particulado/química , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
4.
Ecotoxicol Environ Saf ; 202: 110932, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32800216

RESUMEN

Adverse health effects arising from exposure to fine particulates have become a major concern. Angiogenesis is a vital physiological process for the growth and development of cells and structures in the human body, whereby excessive or insufficient vessel growth could contribute to pathogenesis of diseases. We therefore evaluated indirect effects of carbon black (CB) and inhalable airborne particles on the angiogenic ability of unexposed Human Umbilical Vein Endothelial Cells (HUVECs) by co-culturing HUVECs with pre-exposed Small Airway Epithelial Cells (SAECs). As endothelial cells are major components of blood vessels and potential targets of fine particles, we investigated if lung epithelial cells exposed to ambient PM2.5 surrogates could induce bystander effects on neighboring unexposed endothelial cells in an alveolar-capillary co-culture lung model. Epithelial exposure to CB at a non-toxic dose of 25 µg/mL reduced endothelial tube formation and cell adhesion in co-cultured HUVECs, and decreased expression of angiogenic genes in SAECs. Similarly, exposure of differentiated SAECs to PM2.5 surrogates reduced cell reproductive ability, adhesion and tube formation of neighboring HUVECs. This indicates epithelial exposure to CB and urban PM2.5 surrogates both compromised the angiogenic ability of endothelial cells through bystander effects, thereby potentially perturbing the ventilation-perfusion ratio and affecting lung function.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Material Particulado/toxicidad , Pruebas de Toxicidad , Técnicas de Cocultivo , Células Epiteliales , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Pulmón/metabolismo , Neovascularización Patológica , Hollín
5.
J Vis Exp ; (151)2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31609321

RESUMEN

Zinc oxide nanoparticles (ZnO NPs) have a wide range of applications, but the number of reports on ZnO NP-associated toxicity has grown rapidly in recent years. However, studies that elucidate the underlying mechanisms for ZnO NP-induced toxicity are scanty. We determined the toxicity profiles of ZnO NPs using both in vitro and in vivo experimental models. A significant decrease in cell viability was observed in ZnO NP-exposed MRC5 lung fibroblasts, showing that ZnO NPs exert cytotoxic effects. Similarly, interestingly, gut exposed to ZnO NPs exhibited a dramatic increase in reactive oxygen species levels (ROS) in the fruit fly Drosophila. More in-depth studies are required to establish a risk assessment for the increased usage of ZnO NPs by consumers.


Asunto(s)
Pulmón/efectos de los fármacos , Pulmón/metabolismo , Nanopartículas del Metal/toxicidad , Nanopartículas/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Óxido de Zinc/toxicidad , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Drosophila melanogaster , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Humanos , Pulmón/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
6.
Nanotoxicology ; 13(8): 1117-1132, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31272252

RESUMEN

Engineered nanomaterials are of public health concern. Recently, there has been an increasing attention on the toxicity of nanoplastics and nanoZnO because of their increasing utilization and presence in the environment. However, knowledge of their toxicological behavior and metabolic interactions with the cellular machinery that determine their potential health effects are extremely limited. In this study, the cellular uptake, cytotoxic effects, and metabolic responses of bronchus epithelial (BEAS-2B) cells exposed to nanopolystyrene (nanoPS) and a widely used metallic nanoparticle, nanoZnO, were investigated using a tandem mass spectrometry-based metabolomics approach. The results revealed that even with low cytotoxicity, these nanoparticles (NPs) affected cell metabolism. NanoPS exposure showed autophagic- and endoplasmic reticulum (ER) stress-related metabolic changes such as increased in amino acids and tricarboxylic acid cycle (TCA) intermediate metabolites, a process known to play a critical role in regulating cell resistance to cytotoxic effects. Both metabolomics profiling and ER-stress pathway, together with quantitative real-time RT-polymerase chain reaction (qRT-PCR) analyses, demonstrated that autophagy was reciprocally regulated to couple metabolic and transcriptional reprograming. In contrast, nanoZnO-induced ROS-mediated cell death was associated with mitochondrial dysfunction and interference in regulating energy metabolism. Collectively, these two types of NPs were observed to cause perturbations albeit differential in cellular metabolism associated with their cytotoxic effects. Our findings provided an in depth understanding of metabolic changes influenced by two different types of NPs, with contrasting molecular mechanisms for the adverse effects observed.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Pulmón/citología , Metabolómica/métodos , Nanopartículas/toxicidad , Óxido de Zinc/toxicidad , Aminoácidos , Transporte Biológico , Línea Celular , Metabolismo Energético/efectos de los fármacos , Humanos , Metaboloma/efectos de los fármacos , Nanopartículas/química , Óxido de Zinc/química
7.
Anal Chem ; 90(10): 6193-6198, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29608843

RESUMEN

Cell surface receptors play an important role in mediating cell communication and are used as disease biomarkers and therapeutic targets. We present a one-pot molecular toolbox, which we term the split proximity circuit (SPC), for the autonomous detection and visualization of cell surface receptor clusters. Detection was powered by antibody recognition and a series of autonomous DNA hybridization to achieve localized, enzyme-free signal amplification. The system under study was the human epidermal growth factor receptor (HER) family, that is, HER2:HER2 homodimer and HER2:HER3 heterodimer, both in cell lysate and in situ on fixed whole cells. The detection and imaging of receptors were carried out using standard microplate scans and confocal microscopy, respectively. The circuit operated specifically with minimal leakages and successfully captured the receptor expression profiles on three cell types without any intermediate washing steps.


Asunto(s)
ADN/química , Receptor ErbB-2/análisis , Receptor ErbB-3/análisis , Ensayo de Inmunoadsorción Enzimática , Humanos , Microscopía Confocal , Células Tumorales Cultivadas
8.
Chem Biol Interact ; 289: 81-89, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29705079

RESUMEN

Gold nanoparticles (AuNPs) have emerging applications in biomedicine and the industry. Exposure to AuNPs has previously been shown to alter the transcriptional activity of nuclear factor kappa B (NF-kB), which is known to mediate physiological and pathological processes. This study seeks to provide mechanistic insights into AuNP-induced NF-kB activation in Small Airway Epithelial Cells (SAECs) in vitro. Increased NF-kB transcriptional activity (quantified by the luciferase reporter assay) was observed in AuNP-treated SAECs. Transcriptomic analysis revealed differential expression of 42 genes, which regulate functional processes that include cellular response to stimulus, chemicals and stress as well as immune response. Notably, the gene expression of serum amyloid A1 (SAA1), an acute phase protein and Toll-like receptor 2 (TLR2) were found to be up-regulated. As TLR2 is known to be a functional receptor of SAA1, a co-immunoprecipitation assay was performed. SAA1 was observed to be co-immunoprecipitated with the TLR2 protein and this protein-protein interaction was further supported by in silico computer based protein modeling. The present study suggests that AuNPs may potentially induce SAA1-TLR2-mediated NF-kB transcription factor activation in lung epithelial cells, highlighting that nano-bio interactions could result in biological effects that may affect cells.


Asunto(s)
Oro/química , Pulmón/metabolismo , Nanopartículas del Metal/química , FN-kappa B/metabolismo , Proteína Amiloide A Sérica/metabolismo , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Perfilación de la Expresión Génica , Humanos , Nanopartículas del Metal/ultraestructura , Modelos Biológicos , Unión Proteica
9.
Metabolomics ; 14(9): 118, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30830374

RESUMEN

INTRODUCTION: Histologically lung cancer is classified into four major types: adenocarcinoma (Ad), squamous cell carcinoma (SqCC), large cell carcinoma (LCC), and small cell lung cancer (SCLC). Presently, our understanding of cellular metabolism among them is still not clear. OBJECTIVES: The goal of this study was to assess the cellular metabolic profiles across these four types of lung cancer using an untargeted metabolomics approach. METHODS: Six lung cancer cell lines, viz., Ad (A549 and HCC827), SqCC (NCl-H226 and NCl-H520), LCC (NCl-H460), and SCLC (NCl-H526), were analyzed using liquid chromatography quadrupole time-of-flight mass spectrometry, with normal human small airway epithelial cells (SAEC) as the control group. The principal component analysis (PCA) was performed to identify the metabolic signatures that had characteristic alterations in each histological type. Further, a metabolite set enrichment analysis was performed for pathway analysis. RESULTS: Compared to the SAEC, 31, 27, 34, 34, 32, and 39 differential metabolites mainly in relation to nucleotides, amino acid, and fatty acid metabolism were identified in A549, HCC827, NCl-H226, NCl-H520, NCl-H460, and NCl-H526 cells, respectively. The metabolic signatures allowed the six cancerous cell lines to be clearly separated in a PCA score plot. CONCLUSION: The metabolic signatures are unique to each histological type, and appeared to be related to their cell-of-origin and mutation status. The changes are useful for assessing the metabolic characteristics of lung cancer, and offer potential for the establishment of novel diagnostic tools for different origin and oncogenic mutation of lung cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metabolómica , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Adenocarcinoma/patología , Carcinoma de Células Escamosas/patología , Cromatografía Liquida , Humanos , Espectrometría de Masas , Carcinoma Pulmonar de Células Pequeñas/patología , Células Tumorales Cultivadas
10.
Adv Healthc Mater ; 5(16): 2092-104, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27297640

RESUMEN

Uniform wüstite Fe0.6 Mn0.4 O nanoflowers have been successfully developed as an innovative theranostic agent with T1 -T2 dual-mode magnetic resonance imaging (MRI), for diagnostic applications and therapeutic interventions via magnetic hyperthermia. Unlike their antiferromagnetic bulk counterpart, the obtained Fe0.6 Mn0.4 O nanoflowers show unique room-temperature ferromagnetic behavior, probably due to the presence of an exchange coupling effect. Combined with the flower-like morphology, ferromagnetic Fe0.6 Mn0.4 O nanoflowers are demonstrated to possess dual-modal MRI sensitivity, with longitudinal relaxivity r1 and transverse relaxivity r2 as high as 4.9 and 61.2 mm(-1) s(-1) [Fe]+[Mn], respectively. Further in vivo MRI carried out on the mouse orthotopic glioma model revealed gliomas are clearly delineated in both T1 - and T2 -weighted MR images, after administration of the Fe0.6 Mn0.4 O nanoflowers. In addition, the Fe0.6 Mn0.4 O nanoflowers also exhibit excellent magnetic induction heating effects. Both in vitro and in vivo magnetic hyperthermia experimentation has demonstrated that magnetic hyperthermia by using the innovative Fe0.6 Mn0.4 O nanoflowers can induce MCF-7 breast cancer cell apoptosis and a complete tumor regression without appreciable side effects. The results have demonstrated that the innovative Fe0.6 Mn0.4 O nanoflowers can be a new magnetic theranostic platform for in vivo T1 -T2 dual-mode MRI and magnetic thermotherapy, thereby achieving a one-stop diagnosis cum effective therapeutic modality in cancer management.


Asunto(s)
Neoplasias de la Mama , Medios de Contraste , Compuestos Férricos , Hipertermia Inducida/métodos , Imagen por Resonancia Magnética , Imanes/química , Compuestos de Manganeso , Nanopartículas , Óxidos , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Medios de Contraste/síntesis química , Medios de Contraste/química , Medios de Contraste/farmacología , Femenino , Compuestos Férricos/síntesis química , Compuestos Férricos/química , Compuestos Férricos/farmacología , Humanos , Células MCF-7 , Compuestos de Manganeso/síntesis química , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Nanopartículas/química , Nanopartículas/uso terapéutico , Óxidos/síntesis química , Óxidos/química , Óxidos/farmacología , Nanomedicina Teranóstica/métodos
12.
Biomaterials ; 39: 31-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25477169

RESUMEN

Despite the availability of toxicity studies on cellular exposure to gold nanoparticles (AuNPs), there is scarcity of information with regard to the bystander effects induced by AuNPs on neighboring cells not exposed to the NPs. In this study, we showed that exposure of small airway epithelial cells (SAECs) to AuNPs induced changes in protein expression associated with functional effects in neighboring MRC5 lung fibroblasts in a co-culture system. Uptake of 20 nm size AuNPs by SAECs was first verified by focused ion beam scanning electron microscopy. Subsequently, pretreated SAECs were co-cultured with unexposed MRC5 lung fibroblasts, which then underwent proteome profiling using a quantitative proteomic approach. Stable-isotope labeling by amino acids in cell culture (SILAC)-based mass spectrometry identified 109 proteins (which included 47 up-regulated and 62 down-regulated proteins) that were differentially expressed in the lung fibroblasts co-cultured with AuNP pretreated SAECs. There was altered expression of proteins such as Paxillin, breast cancer anti-estrogen resistance 1 and Caveolin-1, which are known to be involved in the cell adhesion process. Morphological studies revealed that there was a concomitant increase in cell adhesion and altered F-actin stress fiber arrangement involving vinculin in the lung fibroblasts. It is likely that phenotypic changes observed in the underlying lung fibroblasts were mediated by AuNP-induced downstream signals in the pretreated SAECs and cell-cell cross talk.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Oro/química , Oro/farmacología , Pulmón/citología , Nanopartículas del Metal/química , Adhesión Celular/efectos de los fármacos , Línea Celular , Técnicas de Cocultivo , Humanos , Microscopía Confocal , Microscopía Electrónica de Rastreo
13.
Exp Biol Med (Maywood) ; 238(12): 1355-61, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24157586

RESUMEN

Gold nanoparticles (AuNPs) have diverse applications in the biomedical industry such as in diagnosis, labeling, delivering and sensing. Despite their prevalent medical use, nanotoxicity induced by AuNPs is still largely unknown. We have previously shown that AuNPs could exert cytotoxic effects on lung fibroblasts. In this study, we investigated the in vitro toxicological effects of AuNPs in small airway epithelial cells (SAECs) which are the first cells of contact for inhaled NPs and compared expression of metallothionein (MT), a reactive oxygen species scavenger, in SAECs and lung fibroblasts in vitro. Transmission electron microscopy (TEM) and energy-dispersive X-ray (EDX) spectroscopy study revealed cellular uptake of aggregates of AuNPs into the cytoplasm at the ultrastructural level. A significant increase in lipid peroxide as well as substantial DNA damage and cytotoxicity was observed in AuNP-treated cells. For MT expression, AuNPs induced down-regulation of the MT-1X isoform in SAECs, but up-regulation of the MT-1X and MT-2 A isoforms in MRC5 lung fibroblasts. The present study suggests that AuNPs could induce oxidative stress-related cytotoxicity and genotoxicity in SAECs.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Oro/toxicidad , Pulmón/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ensayo Cometa , Fibroblastos/efectos de los fármacos , Humanos , Pulmón/citología , Metalotioneína/metabolismo , Microscopía Electrónica de Transmisión , Estrés Oxidativo/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Exp Biol Med (Maywood) ; 237(6): 635-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22715432

RESUMEN

Carboxylate microspheres (CMs) are mainly used in industrial, biomedical and various household products. In this study, we assessed the cytotoxic effects of CMs on human MRC-5 lung fibroblasts by using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Oxidative stress was determined by measurements of reactive oxygen species and antioxidant (superoxide dismutase and catalase) levels and proinflammatory cytokines quantified by enzyme-linked immunosorbent assay. Morphological changes were examined by light microscopy, confocal microscopy and transmission electron microscopy. The lung fibroblasts were exposed to increasing concentrations of CMs (0.1-1000 µmol/L) for 24 h. The results showed significant changes in cell morphology with induction of cytotoxicity and oxidative stress observed in 10-1000 µmol/L concentrations of CM-treated fibroblasts. Ultrastructural examination revealed the presence of CMs inside the cytoplasm of treated lung fibroblasts. CMs also induced elevated interleukin (IL)-1, IL-6, IL-8, IL-10 and tumor necrosis factor α levels at higher concentrations. We have demonstrated that CMs significantly reduce cell viability in a dose-dependant manner in lung fibroblasts at 0.1-1000 µmol/L doses. The findings suggest that high doses of CMs have the potential to induce cellular toxicity to the lung in vitro.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Carboxílicos/farmacología , Fibroblastos/patología , Pulmón/patología , Microesferas , Catalasa/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Técnicas In Vitro , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
15.
Acta Pharmacol Sin ; 32(8): 983-90, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21743485

RESUMEN

The rapid advancement of nanotechnology in recent years has fuelled a burgeoning interest in the field of nanoparticle research, in particular, its application in the medical arena. A constantly expanding knowledge based on a better understanding of the properties of gold nanoparticles (AuNPs) coupled with relentless experimentation means that the frontiers of nanotechnology are constantly being challenged. At present, there seems to be heightened interest in the application of AuNPs to the management of cancer, encompassing diagnosis, monitoring and treatment of the disease. These efforts are undertaken in the hope of revolutionizing current methods of treatment and treatment strategies for a multifactorial disease such as cancer. This review will focus on the current applications of AuNPs in cancer management.


Asunto(s)
Oro/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Neoplasias/tratamiento farmacológico , Animales , Oro/química , Humanos , Nanotecnología/métodos , Neoplasias/diagnóstico
16.
Anat Rec (Hoboken) ; 294(2): 267-72, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21235001

RESUMEN

Gastrointestinal stromal tumors (GISTs) are mesenchymal tumors that account for about 2% of gastric tumors. Metallothioneins (MTs) are multifunctional proteins associated with carcinogenesis and known to be coded by 10 functional MT genes. This study evaluated MT mRNA and protein expression in GISTs and compared the expression levels with gastric carcinomas. An immunohistochemical study of MT protein expression was performed in 15 GISTs (specifically located in the stomach) and 38 early stage gastric carcinomas. The percentage of cells stained and intensity of staining were determined. MT-2A mRNA expression was investigated in 6 GISTs and 6 early stage gastric carcinoma patients. All GISTs displayed positive nuclear immunostaining, with most GISTs having predominantly mildly stained nuclei (93.3%). On the other hand, 37 out of 38 gastric carcinoma cases were positively stained for nuclear MT with 24 cases (63.2%) exhibiting predominantly mild nuclear staining, 7 cases (18.4%) moderate nuclear staining, and 6 cases (15.8%) strong nuclear staining. Nuclear MT expression was found to be significantly lower in GIST samples when compared with gastric carcinoma tissues based on the percentage stained and immunoreactive score. We then established that the MT-2A gene transcript was the most abundant MT isoform in MKN28 gastric cancer cells and analyzed its expression in GIST and gastric carcinoma tissues. We found that GISTs had significantly lower MT-2A mRNA levels than gastric carcinoma tissues. Lower MT-2A gene expression and nuclear MT protein expression in GISTs when compared with gastric carcinomas may reflect their different underlying biology and divergent histogenesis.


Asunto(s)
Adenocarcinoma/metabolismo , Biomarcadores de Tumor/metabolismo , Tumores del Estroma Gastrointestinal/metabolismo , Metalotioneína/metabolismo , Neoplasias Gástricas/metabolismo , Adenocarcinoma/patología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Tumores del Estroma Gastrointestinal/patología , Humanos , Estadificación de Neoplasias , ARN Mensajero/metabolismo , Neoplasias Gástricas/patología
17.
J Am Chem Soc ; 132(42): 14803-11, 2010 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-20882976

RESUMEN

In the present study, quantum dot (QD) capped magnetite nanorings (NRs) with a high luminescence and magnetic vortex core have been successfully developed as a new class of magnetic-fluorescent nanoprobe. Through electrostatic interaction, cationic polyethylenimine (PEI) capped QD have been firmly graft into negatively charged magnetite NRs modified with citric acid on the surface. The obtained biocompatible multicolor QD capped magnetite NRs exhibit a much stronger magnetic resonance (MR) T2* effect where the r2* relaxivity and r2*/r1 ratio are 4 times and 110 times respectively larger than those of a commercial superparamagnetic iron oxide. The multiphoton fluorescence imaging and cell uptake of QD capped magnetite NRs are also demonstrated using MGH bladder cancer cells. In particular, these QD capped magnetite NRs can escape from endosomes and be released into the cytoplasm. The obtained results from these exploratory experiments suggest that the cell-penetrating QD capped magnetite NRs could be an excellent dual-modality nanoprobe for intracellular imaging and therapeutic applications. This work has shown great potential of the magnetic vortex core based multifunctional nanoparticle as a high performance nanoprobe for biomedical applications.


Asunto(s)
Óxido Ferrosoférrico , Imagen por Resonancia Magnética/métodos , Nanoestructuras , Puntos Cuánticos , Espectrometría de Fluorescencia/métodos , Humanos , Microscopía Electrónica de Transmisión , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA