Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 12(1): 16399, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36180511

RESUMEN

Malignant brain tumors consist of malignancies originated primarily within the brain and the metastatic lesions disseminated from other organs. In spite of intensive studies, malignant brain tumors remain to be a medical challenge. Patient-derived organoid (PDO) can recapitulate the biological features of the primary tumor it was derived from and has emerged as a promising drug-screening model for precision therapy. Here we show a proof-of-concept based on early clinical study entailing the organoids derived from the surgically resected tumors of 26 patients with advanced malignant brain tumors enrolled during December 2020 to October 2021. The tumors included nine glioma patients, one malignant meningioma, one primary lymphoma patient, and 15 brain metastases. The primary tumor sites of the metastases included five from the lungs, three from the breasts, two from the ovaries, two from the colon, one from the testis, one of melanoma origin, and one of chondrosarcoma. Out of the 26 tissues, 13 (50%) organoids were successfully generated with a culture time of about 2 weeks. Among these patients, three were further pursued to have the organoids derived from their tumor tissues tested for the sensitivity to different therapeutic drugs in parallel to their clinical care. Our results showed that the therapeutic effects observed by the organoid models were consistent to the responses of these patients to their treatments. Our study suggests that PDO can recapitulate patient responses in the clinic with high potential of implementation in personalized medicine of malignant brain tumors.


Asunto(s)
Neoplasias Encefálicas , Organoides , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Humanos , Masculino , Medicina de Precisión/métodos
2.
Front Oncol ; 12: 851795, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35992877

RESUMEN

The cGAS-STING axis is one of the key mechanisms guarding cells from pathogen invasion in the cytoplasmic compartment. Sensing of foreign DNA in the cytosol by the cGAS-STING axis triggers a stress cascade, culminating at stimulation of the protein kinase TBK1 and subsequently activation of inflammatory response. In cancer cells, aberrant metabolism of the genomic DNA induced by the hostile milieu of tumor microenvironment or stresses brought about by cancer therapeutics are the major causes of the presence of nuclear DNA in the cytosol, which subsequently triggers a stress response. However, how the advanced tumors perceive and tolerate the potentially detrimental effects of cytosolic DNA remains unclear. Here we show that growth limitation by serum starvation activated the cGAS-STING pathway in breast cancer cells, and inhibition of cGAS-STING resulted in cell death through an autophagy-dependent mechanism. These results suggest that, instead of being subject to growth inhibition, tumors exploit the cGAS-STING axis and turn it to a survival advantage in the stressful microenvironment, providing a new therapeutic opportunity against advanced cancer. Concomitant inhibition of the cGAS-STING axis and growth factor signaling mediated by the epidermal growth factor receptor (EGFR) synergistically suppressed the development of tumor organoids derived from primary tumor tissues of triple-negative breast cancer (TNBC). The current study unveils an unexpected function of the cGAS-STING axis in promoting cancer cell survival and the potential of developing the stress-responding pathway as a therapeutic target, meanwhile highlights the substantial concerns of enhancing the pathway's activity as a means of anti-cancer treatment.

3.
Int J Mol Sci ; 23(10)2022 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-35628489

RESUMEN

Invasion is the most prominent lethal feature of malignant cancer. However, how cell proliferation, another important feature of tumor development, is integrated with tumor invasion and the subsequent cell dissemination from primary tumors is not well understood. Proliferating cell nuclear antigen (PCNA) is essential for DNA replication in cancer cells. Loss of phosphorylation at tyrosine 211 (Y211) in PCNA (pY211-PCNA) mitigates PCNA function in proliferation, triggers replication fork arrest/collapse, which in turn sets off an anti-tumor inflammatory response, and suppresses distant metastasis. Here, we show that pY211-PCNA is important in stromal activation in tumor tissues. Loss of the phosphorylation resulted in reduced expression of mesenchymal proteins as well as tumor progenitor markers, and of the ability of invasion. Spontaneous mammary tumors that developed in mice lacking Y211 phosphorylation contained fewer tumor-initiating cells compared to tumors in wild-type mice. Our study demonstrates a novel function of PCNA as an essential factor for maintaining cancer stemness through Y211 phosphorylation.


Asunto(s)
Invasividad Neoplásica , Neoplasias , Células Madre Neoplásicas , Antígeno Nuclear de Célula en Proliferación , Animales , Proliferación Celular , Replicación del ADN , Ratones , Fosforilación , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo
4.
Am J Cancer Res ; 12(1): 123-137, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35141008

RESUMEN

The atezolizumab (Tecentriq), a humanized antibody against human programmed death ligand 1 (PD-L1), combined with nab-paclitaxel was granted with accelerated approval to treat unresectable locally advanced or metastatic triple-negative breast cancer (TNBC) due to the encouraging positive results of the phase 3 IMpassion130 trial using PD-L1 biomarker from immune cells to stratify patients. However, the post-market study IMpassion131 did not support the original observation, resulting in the voluntary withdrawal of atezolizumab from the indication in breast cancer by Genentech in 2021. Emerging evidence has revealed a high frequency of false negative result using the standard immunohistochemical (IHC) staining due to heavy glycosylation of PD-L1. The removal of glycosylation prevents from the false negative staining, enabling more accurate assessment of PD-L1 levels and improving prediction for response to immune checkpoint therapy. In the present study, the natural and de-glycosylated PD-L1 expression in tumor and immune cells from nine TNBC patients were analyzed by using clone 28-8 monoclonal antibody to correlate with treatment outcome. Our results demonstrate that: (1) Removal of the glycosylation indeed enhances the detection of PD-L1 by IHC staining, (2) The PD-L1 levels on tumor cell surface after removal of the glycosylation correlates well with clinical responses for atezolizumab treatment; (3) The criteria used in the IMpassion130 and IMpassion131 trials which scored the natural PD-L1 in the immune cells failed to correlate with the clinical response. Taken together, tumor cell surface staining of PD-L1 with de-glycosylation has a significant correlation with the clinical response for atezolizumab treatment, suggesting that treatment of atezolizumab may be worthy of further consideration with de-glycosylation procedure as a patient stratification strategy. A larger cohort to validate this important issue is warranted to ensure right patient population who could benefit from the existing FDA-approved drugs.

5.
Comput Struct Biotechnol J ; 20: 241-251, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35024096

RESUMEN

Programmed cell death protein 1 (PD-1)/ programmed cell death protein ligand 1 (PD-L1) is the key immune checkpoint governing evasion of advanced cancer from immune surveillance. Immuno-oncology (IO) therapy targeting PD-1/PD-L1 with traditional antibodies is a promising approach to multiple cancer types but to which the response rate remains moderate in breast cancer, calling for the need of exploring alternative IO targeting approaches. A miRNA-gene network was integrated by a bioinformatics approach and corroborated with The Cancer Genome Atlas (TCGA) to screen miRNAs regulating immune checkpoint genes and associated with patient survival. Here we show the identification of a novel microRNA miR-4759 which repressed RNA expression of the PD-L1 gene. miR-4759 targeted the PD-L1 gene through two binding motifs in the 3' untranslated region (3'-UTR) of PD-L1. Reconstitution of miR-4759 inhibited PD-L1 expression and sensitized breast cancer cells to killing by immune cells. Treatment with miR-4759 suppressed tumor growth of orthotopic xenografts and promoted tumor infiltration of CD8+ T lymphocytes in immunocompetent mice. In contrast, miR-4759 had no effect to tumor growth in immunodeficient mice. In patients with breast cancer, expression of miR-4759 was preferentially downregulated in tumors compared to normal tissues and was associated with poor overall survival. Together, our results demonstrated miR-4759 as a novel non-coding RNA which promotes anti-tumor immunity of breast cancer.

6.
Cell Rep ; 36(8): 109537, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433039

RESUMEN

Increased DNA replication and metastasis are hallmarks of cancer progression, while deregulated proliferation often triggers sustained replication stresses in cancer cells. How cancer cells overcome the growth stress and proceed to metastasis remains largely elusive. Proliferating cell nuclear antigen (PCNA) is an indispensable component of the DNA replication machinery. Here, we show that phosphorylation of PCNA on tyrosine 211 (pY211-PCNA) regulates DNA metabolism and tumor microenvironment. Abrogation of pY211-PCNA blocks fork processivity, resulting in biogenesis of single-stranded DNA (ssDNA) through a MRE11-dependent mechanism. The cytosolic ssDNA subsequently induces inflammatory cytokines through a cyclic GMP-AMP synthetase (cGAS)-dependent cascade, triggering an anti-tumor immunity by natural killer (NK) cells to suppress distant metastasis. Expression of pY211-PCNA is inversely correlated with cytosolic ssDNA and associated with poor survival in patients with cancer. Our results pave the way to biomarkers and therapies exploiting immune responsiveness to target metastatic cancer.


Asunto(s)
Neoplasias Experimentales/inmunología , Antígeno Nuclear de Célula en Proliferación/inmunología , Escape del Tumor , Microambiente Tumoral/inmunología , Animales , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/mortalidad , Fosforilación , Antígeno Nuclear de Célula en Proliferación/genética , Microambiente Tumoral/genética , Tirosina/genética , Tirosina/inmunología
7.
Int J Mol Sci ; 22(13)2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203261

RESUMEN

A group of clinically approved cancer therapeutic tyrosine kinase inhibitors was screened to test their effects on the expression of angiotensin-converting enzyme 2 (ACE2), the cell surface receptor for SARS-CoV-2. Here, we show that the receptor tyrosine kinase inhibitor imatinib (also known as STI571, Gleevec) can inhibit the expression of the endogenous ACE2 gene at both the transcript and protein levels. Treatment with imatinib resulted in inhibition of cell entry of the viral pseudoparticles (Vpps) in cell culture. In FVB mice orally fed imatinib, tissue expression of ACE2 was reduced, specifically in the lungs and renal tubules, but not in the parenchyma of other organs such as the heart and intestine. Our finding suggests that receptor tyrosine kinases play a role in COVID-19 infection and can be therapeutic targets with combined treatments of the best conventional care of COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Mesilato de Imatinib/farmacología , SARS-CoV-2/fisiología , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/genética , Animales , COVID-19/patología , COVID-19/virología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Femenino , Genes Reporteros , Humanos , Ratones , Regiones Promotoras Genéticas , SARS-CoV-2/aislamiento & purificación
8.
Cancers (Basel) ; 12(9)2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32882870

RESUMEN

Chalcone flavokawain B (FKB) possesses a chemopreventive and anti-cancer activity. Doxorubicin is a chemotherapeutic DNA intercalating agent widely used in malignancy treatment. The present study investigated whether synergistic effects exist between the combination of FKB (1.25-5 µg/mL) and doxorubicin (0.5 µg/mL) on the apoptosis and autophagy in human gastric cancer (AGS) cells, and the possible in vitro and in vivo mechanisms. The MTT assay measured cell viability. Various apoptotic-, autophagy-associated protein expression was determined by the Western blot technique. FKB+doxorubicin synergy was estimated by the Chou-Talalay combination index (CI) method. In vivo studies were performed on BALB/c mice. Results showed that compared to FKB/doxorubicin treatments, low doses of FKB+doxorubicin suppressed AGS cell growth. FKB potentiated doxorubicin-induced DNA fragmentation, apoptotic cell death, and enhanced doxorubicin-mediated mitochondrial, death receptor pathways. FKB+doxorubicin activated increased LC3-II accumulation, p62/SQSTM1 expression, and AVO formation as compared to the FKB/doxorubicin alone treatments indicating autophagy in these cells. The death mechanism in FKB+doxorubicin-treated AGS cells is due to the activation of autophagy. FKB+doxorubicin-mediated dysregulated Bax/Bcl-2, Beclin-1/Bcl-2 ratios suggested apoptosis, autophagy induction in AGS cells. FKB+doxorubicin-induced LC3-II/AVOs downregulation was suppressed due to an apoptotic inhibitor Z-VAD-FMK. Whereas, 3-methyladenine/chloroquine weakened FKB+doxorubicin-induced apoptosis (decreased DNA fragmentation/caspase-3). Activation of ERK/JNK may be involved in FKB+doxorubicin-induced apoptosis and autophagy. FKB+doxorubicin-triggered ROS generation, but NAC attenuated FKB+doxorubicin-induced autophagic (LC3 accumulation) and apoptotic (caspase-3 activation and PARP cleavage) cell death. FKB+doxorubicin blocked gastric cancer cell xenografts in nude mice in vivo as compared to FKB/doxorubicin alone treatments. FKB and doxorubicin wielded synergistic anti-tumor effects in gastric cancer cells and is a promising therapeutic approach.

9.
Am J Cancer Res ; 10(12): 4538-4546, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33415017

RESUMEN

The cell surface protein TMPRSS2 (transmembrane protease serine 2) is an androgen-responsive serine protease important for prostate cancer progression and therefore an attractive therapeutic target. Besides its role in tumor biology, TMPRSS2 is also a key player in cellular entry by the SARS-CoV viruses. The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 has resulted in huge losses in socio-economy, culture, and human lives for which safe and effective cures are highly demanded. The main protease (Mpro/3CLpro) of SARS-CoV-2 is a critical enzyme for viral propagation in host cells and, like TMPRSS2, has been exploited for treatment of the infectious disease. Numerous natural compounds abundant in common fruits have been suggested with anti-coronavirus infection in the previous outbreaks of SARS-CoV. Here we show that screening of these compounds identified tannic acid a potent inhibitor of both SARS-CoV-2 Mpro and TMPRSS2. Molecular analysis demonstrated that tannic acid formed a thermodynamically stable complex with the two proteins at a KD of 1.1 mM for Mpro and 1.77 mM for TMPRSS2. Tannic acid inhibited the activities of the two proteases with an IC50 of 13.4 mM for Mpro and 2.31 mM for TMPRSS2. Mpro protein. Consistently, functional assays using the virus particles pseudotyped (Vpp) of SARS-CoV2-S demonstrated that tannic acid suppressed viral entry into cells. Thus, our results demonstrate that tannic acid has high potential of developing anti-COVID-19 therapeutics as a potent dual inhibitor of two independent enzymes essential for SARS-CoV-2 infection.

10.
Food Chem Toxicol ; 124: 30-44, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30465897

RESUMEN

The medicinal fungus Ganoderma, known in Chinese as Lingzhi or Reishi, traditionally has various medicinal uses and has been employed in cancer treatment in Asia for centuries. This study used ethanol-extracted Ganoderma tsugae (GT) and examined its antitumor activities on human chronic myeloid leukemia cells as well as its molecular mechanism of action. Treatment with GT (200-400 µg/mL) significantly reduced cell viability and caused G2/M arrest in K562 cells. In addition, GT induced mitochondrial and death receptor mediated apoptosis, correlated with DNA fragmentation, followed by cytochrome c release, caspase-3/8/9 activation, PARP cleavage, Fas activation, Bid cleavage, and Bax/Bcl-2 dysregulation. Cytoprotective autophagy was found to be induced by GT, as was revealed by increased LC3-II accumulation, Beclin-1/Bcl-2 dysregulation, acidic vesicular organelle formation, and p62/SQSTM1 activation. Notably, pretreatment of cells with the autophagy inhibitors 3-MA and CQ enhanced GT-induced apoptosis. Interestingly, reactive oxygen species production in cells was not triggered by GT administration; equally, the antioxidant N-acetylcysteine was found to be incapable of preventing apoptosis and autophagy induced by GT treatment. Finally, this study discovered that cytoprotective autophagy induced by GT was associated with EGFR and PI3K/AKT/mTOR signaling cascade suppression. In summary, GT demonstrated antitumor activity against human chronic myeloid leukemia.


Asunto(s)
Antineoplásicos/farmacología , Medicamentos Herbarios Chinos/farmacología , Ganoderma/química , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Adenina/análogos & derivados , Adenina/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cloroquina/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Sci Rep ; 7(1): 8062, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28808311

RESUMEN

Coenzyme Q0 (CoQ0, 2,3-dimethoxy-5-methyl-1,4-benzoquinone) has been reported to exert anticancer properties against human breast/lung cancer cells. This study investigated the in vitro and in vivo anticancer properties of CoQ0 on human ovarian carcinoma (SKOV-3) cells and xenografted nude mice, and revealed the underlying molecular mechanism. CoQ0 induced G2/M arrest through downregulation of cyclin B1/A and CDK1/K2 expressions. CoQ0-induced autophagy as a survival mechanism was evidenced by increased accumulation of LC3-II, GFP-LC3 puncta, AVOs formation and Beclin-1/Bcl-2 dysregulation. Increased TUNEL-positive cells and Annexin-V/PI stained cells indicated CoQ0-induced late apoptosis. Both mitochondrial (caspase-3, PARP and Bax/Bcl-2 dysregulation) and ER stress (caspase-12 and Hsp70) signals are involved in execution of apoptosis. Interestingly, CoQ0-induced apoptosis/autophagy is associated with suppression of HER-2/neu and PI3K/AKT signalling cascades. CoQ0 triggered intracellular ROS production, whereas antioxidant N-acetylcysteine prevented CoQ0-induced apoptosis, but not autophagy. Inhibition of apoptosis by Z-VAD-FMK suppressed CoQ0-induced autophagy (diminished LC3-II/AVOs), indicates CoQ0-induced apoptosis led to evoke autophagy. Contrary, inhibition of autophagy by 3-MA/CQ potentiated CoQ0-induced apoptosis (increased DNA fragmentation/PARP cleavage). Furthermore, CoQ0 treatment to SKOV-3 xenografted nude mice reduced tumor incidence and burden. Histopathological analyses confirmed that CoQ0 modulated xenografted tumor progression by apoptosis induction. Our findings emphasize that CoQ0 triggered ROS-mediated apoptosis and cytoprotective autophagy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Coenzimas/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Sustancias Protectoras/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Beclina-1/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neoplasias Ováricas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Molecules ; 19(7): 8820-39, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24972270

RESUMEN

Increasing evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play a key role in neurodegeneration. Fisetin, a plant flavonol commonly found in fruits and vegetables, is frequently added to nutritional supplements due to its antioxidant properties. In the present study, treatment with fisetin inhibited microglial cell migration and ROS (reactive oxygen species) production. Treatment with fisetin also effectively inhibited LPS plus IFN-γ-induced nitric oxide (NO) production, and inducible nitric oxide synthase (iNOS) expression in microglial cells. Furthermore, fisetin also reduced expressions of iNOS and NO by stimulation of peptidoglycan, the major component of the Gram-positive bacterium cell wall. Fisetin also inhibited the enhancement of LPS/IFN-γ- or peptidoglycan-induced inflammatory mediator IL (interlukin)-1 ß expression. Besides the antioxidative and anti-inflammatory effects of fisetin, our study also elucidates the manner in fisetin-induced an endogenous anti-oxidative enzyme HO (heme oxygenase)-1 expression. Moreover, the regulatory molecular mechanism of fisetin-induced HO-1 expression operates through the PI-3 kinase/AKT and p38 signaling pathways in microglia. Notably, fisetin also significantly attenuated inflammation-related microglial activation and coordination deficit in mice in vivo. These findings suggest that fisetin may be a candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.


Asunto(s)
Antiinflamatorios/farmacología , Flavonoides/farmacología , Microglía/inmunología , Fármacos Neuroprotectores/farmacología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Flavonoles , Hemo-Oxigenasa 1/metabolismo , Lipopolisacáridos/farmacología , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos ICR , Microglía/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
13.
Biosens Bioelectron ; 54: 306-10, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24291267

RESUMEN

We demonstrate a biosensor structure consisting of an IGZO (Indium-Gallium-Zinc-Oxide) TFT (thin film transistor) and an extended sensing pad. The TFT acts as the sensing and readout device, while the sensing pad ensures the isolation of biological solution from the transistor channel layer, and meanwhile increases the sensing area. The biosensor is functionalized by first applying ZnO nanorods to increase the surface area for attracting electrical charges of EGFR (epidermal growth factor receptor) antibodies. The device is able to selectively detect 36.2 fM of EGFR in the total protein solution of 0.1 ng/ml extracted from squamous cell carcinoma (SCC). Furthermore, the conjugation duration of the functionalized device with EGFR can be limited to 3 min, implying that the biosensor has the advantage for real-time detection.


Asunto(s)
Anticuerpos Inmovilizados/química , Técnicas Biosensibles/instrumentación , Nanotubos/química , Transistores Electrónicos , Óxido de Zinc/química , Línea Celular , Línea Celular Tumoral , Diseño de Equipo , Galio/química , Humanos , Indio/química , Límite de Detección
14.
Opt Lett ; 38(2): 184-6, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23454956

RESUMEN

In recent years, researchers have demonstrated negative refraction theoretically and experimentally by pumping optical power into photonic crystal (PhC) or waveguide structures. The concept of negative refraction can be used to create a perfect lens that focuses an object smaller than the wavelength. By inserting two-dimensional PhCs into the peripheral of a semiconductor light emitting structure, this study presents an electroluminescent device with negative refraction in the visible wavelength range. This approach produces polarization dependent collimation behavior in far-field radiation patterns. The modal dispersion of negative refraction results in strong group velocity modulation, and self-focusing and -defocusing behaviors are apparent from light extraction. This study further verifies experimental results by using theoretic calculations based on equifrequency contours.

15.
Nanotechnology ; 23(5): 055202, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22238275

RESUMEN

Traditional methods of detecting cancer cells, such as fluorescence, have their limits and can hardly be used for identification during tumor resection. Here we report an alternative tumor detection technology using ZnO nanorods bonded to antibodies as cancer cell probes. Our experiment shows that antibodies toward epidermal growth factor receptor (EGFR) can be connected to ZnO nanorods and to EGFR receptors of SCC (squamous cell carcinoma). The cancer cell can be recognized by the naked eye or an optical microscope with the help of purple light emission from ZnO/EGFR antibody probes. On the other hand, for cells with less EGFR expression, in our case Hs68, no purple light was observed as the probes were washed off. From the photoluminescent spectra, the peak intensity ratio between the purple light (from ZnO at the wavelength 377 nm) and the green band (from the autofluorescence of cells) is much higher with the presence in SCC, as compared with Hs68. The ZnO/EGFR antibody probes have the potential to be applied to surgery for real-time tumor cell identification. The cancer cells will be excised with the help of purple light emission.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Nanoconjugados/química , Óxido de Zinc/química , Óxido de Aluminio , Anticuerpos/química , Anticuerpos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Mediciones Luminiscentes/métodos , Nanotubos/química , Espectrometría de Fluorescencia , Óxido de Zinc/metabolismo
16.
J Mol Med (Berl) ; 90(3): 321-30, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22038170

RESUMEN

Solar lights encompass ultraviolet (UV), visible, and infrared spectrum. Most previous studies focused on the harmful UV effects, and the biologic effects of lights at other spectrums remained unclear. Recently, lights at visible region have been used for regenerative purposes. Using the process of vitiligo repigmentation as a research model, we focused on elucidating the pro-differentiation effects induced by visible light. We first showed that helium-neon (He-Ne) laser (632.8 nm) irradiation stimulated differentiation of primitive pigment cells, an effect not replicable by UVB treatment even at high and damaging doses. In addition, significant increases of mitochondrial DNA copy number and the regulatory genes for mitochondrial biogenesis were induced by He-Ne laser irradiation. Mechanistically, we demonstrated that He-Ne laser initiated mitochondrial retrograde signaling via a Ca(2+)-dependent cascade. The impact on cytochrome c oxidase within the mitochondria is responsible for the efficacy of He-Ne laser in promoting melanoblast differentiation. Taken together, we propose that visible lights from the sun provide important environmental cues for the relatively quiescent stem or primitive cells to differentiate. In addition, our results also indicate that visible light may be used for regenerative medical purposes involving stem cells.


Asunto(s)
Diferenciación Celular/efectos de la radiación , Láseres de Gas/uso terapéutico , Melanocitos/citología , Vitíligo/radioterapia , Animales , Señalización del Calcio , Línea Celular , Complejo IV de Transporte de Electrones/metabolismo , Humanos , Luz , Terapia por Luz de Baja Intensidad , Melaninas/metabolismo , Melanocitos/metabolismo , Melanocitos/efectos de la radiación , Ratones , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Neón , Pigmentación , Rayos Ultravioleta , Vitíligo/metabolismo
17.
Mol Imaging Biol ; 11(4): 253-62, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19225845

RESUMEN

PURPOSE: This study examined the feasibility of simultaneous dopamine and serotonin transporter imaging using [(123)I]ADAM and [(99m)Tc]TRODAT-1 single photon emission computed tomography (SPECT). PROCEDURES: Simultaneous [(123)I]ADAM (185 MBq) and [(99m)Tc]TRODAT-1 (740 MBq) SPECT was performed in three age-matched female Formosan rock monkeys. An asymmetric energy window was used for dual, and symmetric energy windows were used for single-isotope imaging. Oral fluoxetine (20 mg) and intravenous methylphenidate HCl (1 mg/kg) were given 24 h and 10 min, respectively, before dual-isotope SPECT to test imaging specificities of [(123)I]ADAM and [(99m)Tc]TRODAT-1. RESULTS: Comparable image quality and uptake ratios between dual- and single-isotope SPECT scans were found. Dual-isotope SPECT in fluoxetine-pretreated monkeys showed decreased uptake of [(123)I]-ADAM, but not of [(99m)Tc]TRODAT-1. Dual-isotope SPECT in methylphenidate-pretreated monkeys showed decreased [(99m)Tc]TRODAT-1 uptake without affecting [(123)I]-ADAM uptake. CONCLUSION: Simultaneous [(123)I]-ADAM and [(99m)Tc]TRODAT-1 SPECT appears promising in nonhuman primates and may provide a suitable preclinical model with further clinical implications.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Cinanserina/análogos & derivados , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Compuestos de Organotecnecio/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tropanos/metabolismo , Análisis de Varianza , Animales , Cinanserina/metabolismo , Dopamina/metabolismo , Femenino , Fluoxetina/farmacología , Haplorrinos , Procesamiento de Imagen Asistido por Computador , Metilfenidato/farmacología , Radiofármacos/metabolismo
18.
Neurotoxicology ; 29(4): 638-46, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18571730

RESUMEN

Effects of d-amphetamine on the renal outer medullary potassium (ROMK1) channels were tested in the Xenopus oocytes expression system. Xenopus oocytes were injected with mRNA coding for wild-type or mutant ROMK1 channels. Giant inside-out patch-clamp recordings were performed. d-Amphetamine inhibited the activity of ROMK1 channels in a manner that was concentration-dependent but voltage-independent. ROMK1 channels are regulated by intracellular pH (pH i) and protein kinase A (PKA). d-Amphetamine decreased the activity of wild-type and pH i gating residue mutant (K80M) channels over a range of pH i values. However, d-amphetamine failed to reduce channel activity in the presence of PKA inhibitors (H89 and KT 5720) and had no inhibitory effect on the mutants of PKA-phosphorylation sites (S44A, S219A, or S313A), mutants that mimicked the negative charge carried by a phosphate group bound to a serine (S44D, S219D, or S313D), or mutant channels with a positive charge (S219R). These findings suggest that d-amphetamine inhibits ROMK1 channels independently of the pH i. The effects of d-amphetamine on ROMK1 channels may be due to a conformational change induced by PKA-mediated phosphorylation, but not to charge-charge interactions.


Asunto(s)
Dextroanfetamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Potenciales de la Membrana/efectos de los fármacos , Canales de Potasio de Rectificación Interna/fisiología , Animales , Carbazoles/farmacología , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Inhibidores Enzimáticos , Femenino , Isoquinolinas/farmacología , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Microinyecciones , Mutagénesis Sitio-Dirigida/métodos , Oocitos , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/genética , Pirroles/farmacología , Serina/genética , Serina/metabolismo , Sulfonamidas/farmacología , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...