Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-36883859

RESUMEN

Lead is one of the most toxic substances. However, there are few ratiometric fluorescent probes for sensing Pb2+ in aqueous solution as well as living cells because specific ligands for Pb2+ ions have not been well characterized. Considering the interactions between Pb2+ and peptides, we developed ratiometric fluorescent probes for Pb2+ based on the peptide receptor in two steps. First, we synthesized fluorescent probes (1-3) based on the tetrapeptide receptor (ECEE-NH2) containing hard and soft ligands by conjugation with diverse fluorophores that showed excimer emission when they aggregated. After investigation of fluorescent responses to metal ions, benzothiazolyl-cyanovinylene was evaluated as an appropriate fluorophore for ratiometric detection of Pb2+. Next, we modified the peptide receptor to decrease the number of hard ligands and/or to replace Cys with disulfide bond and methylated Cys for improving selectivity and cell permeability. From this process, we developed two fluorescent probes (3 and 8) among the probes (1-8) that exhibited remarkable ratiometric sensing properties for Pb2+ including high water solubility (≤2% DMF), visible light excitation, high sensitivity, selectivity for Pb2+, low detection limits (<10 nM), and fast response (<6 min). The binding mode study revealed that specific Pb2+-peptide interactions of the probes caused nanosized aggregates in which the fluorophores of the probes came close each other, exhibiting excimer emission. In particular, 8 based on tetrapeptide bearing a disulfide bond and two carboxyl groups with a good permeability successfully quantified intracellular uptake of Pb2+ in live cells through ratiometric fluorescent signals. The ratiometric sensing system based on specific metal-peptide interactions and excimer emission process could provide a valuable tool to quantify Pb2+ in live cells and pure aqueous solutions.

2.
Theranostics ; 13(3): 873-895, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36793872

RESUMEN

Rationale: Overexpression of NAD(P)H:quinone oxidoreductase 1 (NQO1) is associated with tumor cell proliferation and growth in several human cancer types. However, the molecular mechanisms underlying the activity of NQO1 in cell cycle progression are currently unclear. Here, we report a novel function of NQO1 in modulation of the cell cycle regulator, cyclin-dependent kinase subunit-1 (CKS1), at the G2/M phase through effects on the stability of c­Fos. Methods: The roles of the NQO1/c-Fos/CKS1 signaling pathway in cell cycle progression were analyzed in cancer cells using synchronization of the cell cycle and flow cytometry. The mechanisms underlying NQO1/c-Fos/CKS1-mediated regulation of cell cycle progression in cancer cells were studied using siRNA approaches, overexpression systems, reporter assays, co-immunoprecipitation, pull-down assays, microarray analysis, and CDK1 kinase assays. In addition, publicly available data sets and immunohistochemistry were used to investigate the correlation between NQO1 expression levels and clinicopathological features in cancer patients. Results: Our results suggest that NQO1 directly interacts with the unstructured DNA-binding domain of c-Fos, which has been implicated in cancer proliferation, differentiation, and development as well as patient survival, and inhibits its proteasome-mediated degradation, thereby inducing CKS1 expression and regulation of cell cycle progression at the G2/M phase. Notably, a NQO1 deficiency in human cancer cell lines led to suppression of c-Fos-mediated CKS1 expression and cell cycle progression. Consistent with this, high NQO1 expression was correlated with increased CKS1 and poor prognosis in cancer patients. Conclusions: Collectively, our results support a novel regulatory role of NQO1 in the mechanism of cell cycle progression at the G2/M phase in cancer through effects on c­Fos/CKS1 signaling.


Asunto(s)
Ciclo Celular , NAD(P)H Deshidrogenasa (Quinona) , Neoplasias , Humanos , División Celular , Línea Celular Tumoral , Fase G2 , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Neoplasias/genética
3.
ACS Omega ; 7(34): 29684-29691, 2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-36061651

RESUMEN

Although diverse cell penetrating motifs not only from naturally occurring proteins but also from synthetic peptides have been discovered and developed, the selectivity of cargo delivery connected to these motifs into the desired target cells is generally low. Here, we demonstrate the selective cytotoxicity tuning of an anticancer KLA peptide with a cell penetrating motif activatable by matrix metalloproteinase-2 (MMP2). The anionic masking sequence introduced at the end of the KLA peptide through an MMP2-cleavable linker is selectively cleaved by MMP2 and the cationic cell penetrating motif is activated. Upon treatment of the peptide to H1299 cells (high MMP2 level), it is selectively internalized into the cells by MMP2, which consequently induces membrane disruption and cell death. In contrast, the peptide shows negligible cytotoxicity toward A549 cancer cells with low MMP2 levels. Furthermore, the selective therapeutic efficacy of the peptide induced by MMP2 is also corroborated using in vivo study.

4.
Cancers (Basel) ; 13(14)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34298763

RESUMEN

Three-dimensional (3D) cancer cell culture systems have been developed to aid the study of molecular mechanisms in cancer development, identify therapeutic targets, and test drug candidates. In this study, we developed a strategy for mimicking the hypoxic tumor microenvironment in a 3D cancer cell culture system using multi-layer, nanofibrous poly(ε-caprolactone) (PCL) scaffold (pNFS)-based cancer cell cultures. We found that human colon cancer cells infiltrated pNFS within 3 days and could be cultured three-dimensionally within the NFS. When incubated in four stacks of 30 µm-thick pNFS for 3 days, colon cancer cells in layer three showed partially reduced entry into the S phase, whereas those in layer four, located farthest from the media, showed a marked reduction in S-phase entry. As a consequence, cells in layer four exhibited hypoxia-induced disorganization of F-actin on day 3, and those in layers three and four showed an increase in the expression of the hypoxia-induced transcription factor HIF-1α and its target genes, Glut1, CA9, VEGF, and LDHA. Consistent with these results, doxorubicin- and ionizing radiation-induced cell death was reduced in colon cancer cells cultured in layers three and four. These results suggest that pNFS-based multi-layer colon cancer cell cultures mimic the hypoxic tumor microenvironment and are useful for bioassays.

5.
FASEB J ; 35(3): e21391, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33565155

RESUMEN

Atherosclerosis is a chronic inflammatory disease of the arterial wall. It has been known that development of atherosclerosis is closely related to activation of tumor necrosis factor α (TNF-α). The objective of this study was to elucidate the effects of TNF-α blockade with brusatol on endothelial activation under pro-atherosclerotic conditions. To this end, we examined the effects of brusatol on TNF-α-induced intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) expression in human aortic endothelial cells (HAECs) using western blot and THP-1 adhesion assays. Brusatol induced a decrease in TNF-α-induced ICAM-1 and VCAM-1 expression through inhibiting TNFR1 expression, leading to suppression of endothelial inflammation independently of the NRF2 (nuclear factor erythroid 2-related factor 2) pathway. The mechanism underlying brusatol-induced TNF receptor 1 (TNFR1) inhibition was investigated with the aid of protein synthesis, co-immunoprecipitation, and cytokine arrays. Notably, brusatol inhibited TNFR1 protein synthesis and suppressed both the canonical nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) signaling pathway and TNF-α-induced cytokine secretion. We further tested the functional effect of brusatol on atherosclerosis development in vivo using two different atherosclerosis mouse models, specifically, acute partial carotid ligation and conventional chronic high-fat diet-fed mouse models. Administration of brusatol led to significant suppression of atherosclerosis development in both mouse models. Our finding that brusatol prevents atherosclerosis via inhibition of TNFR1 protein synthesis supports the potential of downregulation of cell surface TNFR1 as an effective therapeutic approach to inhibit development of atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Cuassinas/uso terapéutico , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Animales , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Complejo de la Endopetidasa Proteasomal/fisiología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Molécula 1 de Adhesión Celular Vascular/genética
6.
Anal Chem ; 92(7): 4917-4925, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32153189

RESUMEN

It is highly challenging to develop fast and sensitive fluorescent methods for monitoring organic mercury in purely aqueous solutions as well as live cells. Especially, selective fluorescent detection of methylmercury over inorganic mercury ions has not been reported. We developed a fast and sensitive fluorescent detection method for Hg2+ ions as well as methylmercury using an amino acid-based fluorescent probe (1) and SDS micelles. The fluorescent probe in SDS micelles detected sensitively and selectively Hg2+ ions and methylmercury among 16 metal ions in purely aqueous solution by the enhancement of the red emission at 575 nm, and the detection of methylmercury was completed within 1 min. The probe in SDS micelles with EDTA showed highly sensitive and selective turn on detection for methylmercury over Hg2+. The limit of detection was 9.1 nM for Hg2+ (1.8 ppb, R2 = 0.989) and 206 nM for CH3Hg+ (R2 = 0.997). 1 rapidly penetrated live cells and detected intracellular Hg2+ ions as well as CH3Hg+ by the enhancement of both red emissions and green emissions. Subsequent treatment of EDTA into the cell confirmed the selective detection of methylmercury in the cells. The present work indicated that the fluorescent probe with micelle systems provided a fast, sensitive, and selective detection method for monitoring inorganic mercury as well as methyl mercury.


Asunto(s)
Colorantes Fluorescentes/química , Compuestos de Metilmercurio/análisis , Contaminantes Químicos del Agua/química , Células A549 , Humanos , Micelas , Conformación Molecular , Imagen Óptica , Soluciones , Espectrometría de Fluorescencia
7.
Chem Commun (Camb) ; 56(19): 2941-2944, 2020 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-32040107

RESUMEN

We present a reaction-based fluorescent probe (1) for Hg2+ and CH3Hg+, based on the displacement reaction of the arylboronic acid with the mercury species. 1 showed promising sensing properties for Hg2+ and CH3Hg+, such as high selectivity and sensitivity, turn-on response, fast response to Hg2+ (<2 min) and CH3Hg+ (<5 min), low detection limits and operation in purely aqueous solutions.


Asunto(s)
Ácidos Borónicos/análisis , Mercurio/análisis , Compuestos de Metilmercurio/análisis , Espectrometría de Fluorescencia/métodos , Límite de Detección
8.
Bioconjug Chem ; 31(1): 43-50, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31841634

RESUMEN

The stimuli-responsive conformational transformation of peptides possessing a constrained form triggered by specific biological microenvironment would provide an effective strategy for the development of highly specific peptide therapeutics. Here, we developed a peptide containing a cytotoxic helical KLA sequence with therapeutic specificity through the use of stimuli-responsive conformational transformation. The KLA peptide is modified to form a cyclic structure to allow for constrained helicity that confers low cytotoxicity. The modified KLA peptide is electrostatically complexed to hyaluronic acid to facilitate enhanced endocytosis into the cancer cells. After endocytosis, the peptide is released from the complex into the cellular cytoplasm by hyaluronidases on the surface of the cellular membrane. Specific intracellular stimuli then trigger the release of the strain that suppresses peptide helicity, and the inherent helical conformation of the KLA peptide is restored. Therefore, the stimuli-responsive conformational transformation of a peptide from low to high helicity selectively induces cell death by disruption of the plasma and mitochondrial membrane.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Descubrimiento de Drogas , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Membranas Mitocondriales/efectos de los fármacos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Conformación Proteica en Hélice alfa
9.
Int J Mol Sci ; 20(18)2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491980

RESUMEN

5-Fluorouracil (5-FU) is an important chemotherapeutic agent for the systemic treatment of colorectal cancer (CRC), but its effectiveness against CRC is limited by increased 5-FU resistance caused by the hypoxic tumor microenvironment. The purpose of our study was to assess the feasibility of using quinacrine (QC) to increase the efficacy of 5-FU against CRC cells under hypoxic conditions. QC reversed the resistance to 5-FU induced by hypoxia in CRC cell lines, as determined using ATP-Glo cell viability assays and clonogenic survival assays. Treatment of cells with 5-FU under hypoxic conditions had no effect on the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a regulator of cellular resistance to oxidative stress, whereas treatment with QC alone or in combination with 5-FU reduced Nrf2 expression in all CRC cell lines tested. Overexpression of Nrf2 effectively prevented the increase in the number of DNA double-strand breaks induced by QC alone or in combination with 5-FU. siRNA-mediated c-Jun N-terminal kinase-1 (JNK1) knockdown inhibited the QC-mediated Nrf2 degradation in CRC cells under hypoxic conditions. The treatment of CRC xenografts in mice with the combination of QC and 5-FU was more effective in suppressing tumor growth than QC or 5-FU alone. QC increases the susceptibility of CRC cells to 5-FU under hypoxic conditions by enhancing JNK1-dependent Nrf2 degradation.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos/genética , Fluorouracilo/farmacología , Hipoxia/genética , Hipoxia/metabolismo , Factor 2 Relacionado con NF-E2/genética , Quinacrina/farmacología , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Modelos Animales de Enfermedad , Fluorouracilo/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Modelos Biológicos , Factor 2 Relacionado con NF-E2/metabolismo , Estadificación de Neoplasias , Proteolisis , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Lett ; 416: 31-41, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29247825

RESUMEN

Hypoxia leads to cancer progression and promotes the metastatic potential of cancer cells. MicroRNAs (miRNAs) are small non-coding RNA that have emerged as key players involved in cancer development and progression. Hypoxia alters a set of hypoxia-mediated miRNAs expression during tumor development and it may function as oncogenes or tumor-suppressors. However, the roles and molecular mechanisms of hypoxia-regulatory miRNAs in colorectal cancer (CRC) progression remain poorly understood. Here we firstly identified miR-590-5p as hypoxia-sensitive miRNAs which was upregulated in colon cancer cells under hypoxia. Hypoxia-induced miR-590-5p suppressed the expression of RECK, in turn, promoting cell invasiveness and migratory abilities via activation of matrix metalloproteinases (MMPs) and filopodia protrusion in vitro. Inhibition of miR-590-5p suppressed tumor growth and metastasis in mouse xenograft and CRC liver metastasis models via inhibition of MMPs activity. Clinical analysis revealed higher miR-590-5p expression in CRC, compared to normal specimens. Furthermore, miR-590-5p expression was significantly increased in liver metastasis as compared to their matched primary CRC. Taken together, our findings provide the first evidence that miR-590-5p may have potential as a therapeutic target for CRC patients with metastasis.


Asunto(s)
Neoplasias Colorrectales/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Metaloproteinasas de la Matriz/genética , MicroARNs/genética , Regiones no Traducidas 3'/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células HCT116 , Humanos , Hipoxia , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Trasplante Heterólogo
13.
Chemistry ; 23(67): 16966-16971, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29077239

RESUMEN

Utilizing stimuli-responsive conformational transformation of a cyclic peptide as a gatekeeper for mesoporous nanocarriers has several advantages such as facile introduction of targeting capabilities, low enzymatic degradation during blood circulation and enhanced specific binding to selected cells. In this report, a Asn-Gly-Arg (NGR)-containing dual-functional cyclic peptide gatekeeper on the surface of mesoporous nanocarrier is prepared not only for active targeting of the aminopeptidase N (APN) expressed on cancer cells but also stimuli-responsive intracellular drug release triggered by a glutathione (GSH)-induced conformational transformation of the peptide gatekeeper. The peptide gatekeeper on the surface of nanocarriers exhibits on-off gatekeeping by conformational transformation triggered by intracellular glutathione of the cancer cells. H1299 cells (high APN expression) showed greater uptake of the nanocarrier by endocytosis and higher apoptosis than A549 cells (low APN expression).


Asunto(s)
Antineoplásicos/farmacología , Antígenos CD13/metabolismo , Doxorrubicina/farmacología , Nanocápsulas/química , Péptidos Cíclicos/química , Dióxido de Silicio/química , Antineoplásicos/química , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Preparaciones de Acción Retardada , Doxorrubicina/química , Liberación de Fármacos , Glutatión/química , Humanos , Conformación Molecular , Oligopéptidos/química , Tamaño de la Partícula , Porosidad , Propiedades de Superficie
14.
Chem Asian J ; 12(21): 2813-2818, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28921899

RESUMEN

αv ß3 Integrin is upregulated on many cancer cells. We designed a dual functional cyclic peptide gatekeeper with a capability of stimuli-responsive conformational transformation which could serve as a selective cell-targeting on-off gatekeeper for mesoporous nanocarriers. The advantage of employing the motif of stimuli-induced conformational transformation of cyclic peptides is that they could be utilized not only as an on-off gatekeeper for the triggered release of cargo drugs but also as a targeting ligand of the carriers to desired cells with their respective binding receptors. The peptide gatekeepers on the surface of nanocarriers exhibited on-off gatekeeping via conformational transformation triggered by intracellular glutathione levels of the cancer cells. The cyclic RGD sequence of the peptide gatekeepers enhanced the intracellular uptake into tumor cells (A549) and the therapeutic efficacy of the nanocarrier.


Asunto(s)
Nanopartículas/química , Péptidos Cíclicos/química , Línea Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Humanos , Tamaño de la Partícula , Porosidad , Conformación Proteica , Propiedades de Superficie
15.
Theranostics ; 7(14): 3415-3431, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28912885

RESUMEN

HIF-1 (hypoxia-inducible factor-1) regulates the expression of ~100 genes involved in angiogenesis, metastasis, tumor growth, chemoresistance and radioresistance, underscoring the growing interest in targeting HIF-1 for cancer control. In the present study, we investigated the molecular mechanisms underlying brusatol-induced HIF-1α degradation and cell death in colorectal cancer under hypoxia (0.5% O2). Under hypoxia, pretreatment of cancer cells with brusatol increased HIF-1α degradation and cancer cell death in a dose-dependent manner. This effect was mediated by activation of prolyl hydroxylases (PHDs), as evidenced by the block of brusatol-induced HIF-1α degradation and cancer cell death by both pharmacological inhibition and siRNA-mediated knockdown of PHDs. In addition, a ferrous iron chelator (2,2'-bypyridyl) blocked brusatol-induced degradation of HIF-1α and cancer cell death in hypoxia by inhibiting PHD activation. We further found that brusatol inhibited c-Myc expression, and showed that overexpression of c-Myc prevented brusatol-induced degradation of HIF-1α and cancer cell death by increasing mitochondrial ROS production and subsequent ROS-mediated transition of ferrous iron to ferric iron. Consistent with these results, treatment of tumor-bearing mice with brusatol significantly suppressed tumor growth by promoting PHD-mediated HIF-1α degradation. Collectively, our results suggest that brusatol-mediated inhibition of c-Myc/ROS signaling pathway increases HIF-1α degradation by promoting PHD activity and induces cell death in colorectal cancer under hypoxia.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Neoplasias Colorrectales/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-myc/metabolismo , Cuassinas/farmacología , Animales , Antineoplásicos/uso terapéutico , Hipoxia de la Célula , Neoplasias Colorrectales/tratamiento farmacológico , Células HCT116 , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Prolil Hidroxilasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Cuassinas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
16.
Nanoscale ; 9(20): 6901-6909, 2017 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-28503686

RESUMEN

Tissue hypoxia developed in most malignant tumors makes a significant difference to normal tissues in the reduction potential and the activity of various bioreductive enzymes. Given the superior enzymatic activity of NAD(P)H:quinone oxidoreductase 1 (NQO1, a cytosolic reductase up-regulated in many human cancers) in hypoxia relative to that in normoxia, NQO1 has great potential for targeting hypoxic tumor cells. In the present report, the core concept of hypoxic NQO1-responsive mesoporous silica nanoparticles (MSNs) is based on the reasoning that the superior enzymatic activity of NQO1 within hypoxic cancer cells can be utilized as a key stimulus for the selective cleavage of an azobenzene stalk triggering the on-off gatekeeping for controlled release of guest drugs. We corroborate that the NQO1 specifically triggers to release the entrapped drug in the nanochannel of MSNs by reductive cleavage of the azobenzene linker only under hypoxic conditions in a controlled manner not only in vitro but also in vivo. Therefore, our results indicate that Si-Azo-CD-PEG could be utilized as a hypoxic cancer-targeting drug delivery carrier, and further suggest that the azobenzene linker could generally be useful for the construction of hypoxic NQO1-responsive nanomaterials.


Asunto(s)
Ciclodextrinas/química , Sistemas de Liberación de Medicamentos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Nanopartículas , Hipoxia Tumoral/efectos de los fármacos , Humanos , Dióxido de Silicio
17.
Nat Commun ; 7: 13593, 2016 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-27966538

RESUMEN

Overexpression of NQO1 is associated with poor prognosis in human cancers including breast, colon, cervix, lung and pancreas. Yet, the molecular mechanisms underlying the pro-tumorigenic capacities of NQO1 have not been fully elucidated. Here we show a previously undescribed function for NQO1 in stabilizing HIF-1α, a master transcription factor of oxygen homeostasis that has been implicated in the survival, proliferation and malignant progression of cancers. We demonstrate that NQO1 directly binds to the oxygen-dependent domain of HIF-1α and inhibits the proteasome-mediated degradation of HIF-1α by preventing PHDs from interacting with HIF-1α. NQO1 knockdown in human colorectal and breast cancer cell lines suppresses HIF-1 signalling and tumour growth. Consistent with this pro-tumorigenic function for NQO1, high NQO1 expression levels correlate with increased HIF-1α expression and poor colorectal cancer patient survival. These results collectively reveal a function of NQO1 in the oxygen-sensing mechanism that regulates HIF-1α stability in cancers.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Colorrectales/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Técnicas de Silenciamiento del Gen , Homeostasis , Humanos , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Oxígeno/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica
18.
Sci Rep ; 6: 27382, 2016 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-27263528

RESUMEN

HIF-1 (hypoxia-inducible factor-1) regulates the expression of more than 70 genes involved in angiogenesis, tumor growth, metastasis, chemoresistance, and radioresistance. Thus, there is growing interest in using HIF-1 inhibitors as anticancer drugs. Docetaxel, a Food and Drug Administration-approved anticancer drug, is reported to enhance HIF-1α degradation. Here, we investigated the molecular mechanism underlying docetaxel-induced HIF-1α degradation and cancer cell death under hypoxic conditions. Docetaxel pretreatment enhanced the polyubiquitination and proteasome-mediated degradation of HIF-1α, and increased cancer cell death under hypoxic conditions. Docetaxel also activated the prolyl hydroxylase, PHD1, in hypoxia, and pharmacological inhibition or siRNA-mediated knockdown of PHD1 prevented docetaxel-induced HIF-1α degradation and cancer cell death. Additionally, siRNA-mediated JNK2 knockdown blocked docetaxel-induced HIF-1α degradation and cancer cell death by inhibiting PHD1 activation. A luciferase reporter assay revealed that inhibition of the JNK2/PHD1 signaling pathway significantly increased the transcriptional activity of HIF-1 in docetaxel-treated cancer cells under hypoxia. Consistent with these results, docetaxel-treated JNK2-knockdown tumors grew much faster than control tumors through inhibition of docetaxel-induced PHD1 activation and degradation of HIF-1α. Our results collectively show that, under hypoxic conditions, docetaxel induces apoptotic cell death through JNK2/PHD1 signaling-mediated HIF-1α degradation.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Taxoides/farmacología , Secuencia de Aminoácidos , Línea Celular Tumoral , Docetaxel , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/química , Proteína Quinasa 9 Activada por Mitógenos/genética , Neoplasias/enzimología , Neoplasias/metabolismo , Proteolisis , Ubiquitinación
19.
Biosens Bioelectron ; 85: 437-444, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27208475

RESUMEN

Developing fluorescent probes for monitoring intracellular Cu(+) is important for human health and disease, whereas a few types of their receptors showing a limited range of binding affinities for Cu(+) have been reported. In the present study, we first report a novel peptide receptor of a fluorescent probe for the detection of Cu(+). Dansyl-labeled tripeptide probe (Dns-LLC) formed a 1:1 complex with Cu(+) and showed a turn-on fluorescent response to Cu(+) in aqueous buffered solutions. The dissociation constant of Dns-LLC for Cu(+) was determined to be 12 fM, showing that Dns-LLC had more potent binding affinity for Cu(+) than those of previously reported chemical probes for Cu(+). The binding mode study showed that the thiol group of the peptide receptor plays a critical role in potent binding with Cu(+) and the sulfonamide and amide groups of the probe might cooperate to form a complex with Cu(+). Dns-LLC detected Cu(+) selectively by a turn-on response among various biologically relevant metal ions, including Cu(2+) and Zn(2+). The selectivity of the peptide-based probe for Cu(+) was strongly dependent on the position of the cysteine residue in the peptide receptor part. The fluorescent peptide-based probe penetrated the living RKO cells and successfully detected Cu(+) in the Golgi apparatus in live cells by a turn-on response. Given the growing interest in imaging Cu(+) in live cells, a novel peptide receptor of Cu(+) will offer the potential for developing a variety of fluorescent probes for Cu(+) in the field of copper biochemistry.


Asunto(s)
Cobre/análisis , Colorantes Fluorescentes/química , Aparato de Golgi/química , Imagen Óptica/métodos , Péptidos/química , Técnicas Biosensibles/métodos , Cationes/análisis , Línea Celular , Línea Celular Tumoral , Humanos , Espectrometría de Fluorescencia/métodos
20.
Nanoscale ; 8(15): 8070-7, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27021628

RESUMEN

Mesoporous silica nanocontainers (MSNs) with biologically responsive gatekeepers have great potential for effective delivery of cargo molecules to the desired sites. For that purpose, peptides could be effective candidates as gatekeepers because of their bioresponsiveness and targeting capability. Taking advantage of the zinc finger domain peptide (CXXC), we designed a biocompatible all-peptide gatekeeper (WCGKC) with on-off gatekeeping capability through stimulus-responsive conformational conversion and the steric bulkiness of the tryptophan unit. The turn structure induced by an intramolecular disulfide bond of the peptide gatekeeper (WCGKC-SS) completely inhibited the release of the entrapped doxorubicin (DOX). However, upon reduction of the disulfide bond by glutathione (GSH), the peptide conformation was converted to a random structure, which opened the orifice of the mesopore leading to the release of DOX. The amine moiety of the lysine of the peptide gatekeeper was PEGylated to enhance dispersion stability and biocompatibility of the nanocontainer. Furthermore, the MSNs with the peptide gatekeeper (PEG-WCGKC-SS-Si) selectively released the entrapped DOX in A549 human lung cancer cells in a controlled manner triggered by intracellular GSH, but not in CCD normal lung cells containing a low intracellular GSH level. In A549 cells, DOX-loaded PEG-WCGKC-SS-Si exhibited about 10-times higher cytotoxicity induced by apoptosis than that in CCD cells.


Asunto(s)
Portadores de Fármacos/química , Nanocápsulas/química , Células A549 , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Línea Celular , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Humanos , Oligopéptidos/química , Conformación Proteica , Dióxido de Silicio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...