Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Analyst ; 144(17): 5136-5142, 2019 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-31338492

RESUMEN

γ-Glutamyl transpeptidase (GGT) has been reported as a biomarker of hepatocellular carcinoma (HCC), and its imaging is of great benefit for early detection in precise medicine as well as intraoperative navigation. Herein, we have designed and synthesized a novel near-infrared fluorescent probe coupled aggregation-induced emission (AIE) and excited-state intramolecular proton transfer (ESIPT) effect for the detection of GGT. Thanks to conjugated glutamate acid, this probe could be dispersed in aqueous solution and showed barely any fluorescence emission. Through a GGT-mediated enzymatic reaction, the aggregation state of the probe in aqueous solution was changed and an intramolecular hydrogen bond was formed, resulting in an enhanced fluorescence emission. An excellent linear relationship was observed and the concentration of GGT measured was in the range of 10-90 U L-1 with a limit of detection calculated at 2.9 U L-1. Its feasibility has been confirmed by detecting GGT in HepG2 cells with high specificity and long-term sustainability, satisfying clinical need. Moreover, this nanoprobe showed great potential for precise medicine guided surgery by realizing fluorescence imaging in human liver tumour tissue and distinguishing it from normal tissue. Thus, we supposed that our AIE coupled ESIPT fluorescent nanoprobe has great potential in the early detection of HCC, the selective fluorescence imaging of GGT positive cells during surgery and application in precision medicine.


Asunto(s)
Biomarcadores de Tumor/análisis , Colorantes Fluorescentes/química , gamma-Glutamiltransferasa/análisis , Benzotiazoles/síntesis química , Benzotiazoles/química , Colorantes Fluorescentes/síntesis química , Glutamatos/síntesis química , Glutamatos/química , Células Hep G2 , Humanos , Límite de Detección , Neoplasias Hepáticas/diagnóstico , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Medicina de Precisión/métodos , Sensibilidad y Especificidad
2.
Mikrochim Acta ; 186(6): 353, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31098674

RESUMEN

The authors describe the syntheses and application of glutathione-capped gold nanoclusters (AuNCs) with thermoresponsive properties. The AuNCs have excitation/emission maxima at 430/610 nm and the bright redfluorescence changes along with the temperature in the range from 0 to 90 °C which covers the normal temperature range of living cells. In the range of physiological temperatures (35-42 °C), the temperature resolution is 0.73 °C. The AuNCs display excellent colloidal stability and biocompatibility. They were used for fluorometric temperature detection and imaging of hepatic stellate cells. With such attractive features, the AuNCs are quite promising luminescence nanothermometers. Graphical abstract Schematic presentation of the fluorescence of glutathione-capped gold nanoclusters (AuNCs) as nanothermometers in living cells. The AuNCs have excitation/emission maxima at 430/610 nm and the red fluorescence changes with temperature in a wide range of 0 to 90 °C which covers the normal temperature of living cells.


Asunto(s)
Fluorometría , Oro/química , Nanopartículas del Metal/química , Nanotecnología , Imagen Óptica , Temperatura , Supervivencia Celular , Células Cultivadas , Fluorometría/instrumentación , Humanos , Rayos Infrarrojos , Nanotecnología/instrumentación
3.
Mikrochim Acta ; 185(4): 218, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29594600

RESUMEN

A nanoprobe is described for the ratiometric fluorometric determination of sulfite ions. Upconversion nanoparticles (UCNPs) of the type ß-NaYF4:Yb(III),Er(III),Tm(III) were covalently modified with the molecular probe HIAN which is a hydroxynaphthalimide fluorophore modified with a (cationic) indolinium moiety. Under excitation at 980 nm, the green emission of the UCNPs (peaking at 543 nm) is almost totally quenched, while the NIR emission (peaking at 802 nm) remains unaffected. In the presence of sulfite or bisulfite (hydrogen sulfite), the green fluorescence is restored and can be visually observed. A ratiometric method was worked out by measurement of the ratio of the green and NIR emissions. The analytical range extends from 10 to 250 µM, the limit of detection is 0.14 µM, and the assay can be performed within 40 s. Graphical abstract Based on the use of a molecular probe for sulfite and hydrogen sulfite, and by exploiting an inner filter effect (IFE), an assay for sulfite/hydrogen sulfite was developed by using upconversion nanoparticles (UCNPs). Addition reaction of sulfite/bisulfite with the material results in weakened IFE and enhanced green fluorescence of the UCNPs at excitation/emission wavelengths of 980/543 nm.

4.
Anal Chem ; 89(18): 10085-10093, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28828856

RESUMEN

Specific detection or imaging protein has high potential to contribute greatly to medical diagnosis, biological research, and therapeutic applications. The level of human serum albumin (HSA) in blood is related to a variety of diseases and thus serves as an important biomarker for fast clinical diagnosis. Here we report the use of aggregation-induced emission (AIE) based supramolecular assembly to design biomolecular responsive smart organic nanomaterials for detection protein HSA. The designed nanoprobes were aggregates of small molecules and silent in fluorescence, but in the presence of HSA they disassembled and produced a clear turn-on fluorescent signal. Of a small library of nanoprobes constructed for HSA detection, structure-optical signaling and screening studies revealed that nanoprobe 7 is the most efficient one. Mechanism studies showed that nanoprobe 7 was bonded with Site I of HSA through the multiple noncovalent interactions. The resultant restriction of intramolecular rotation of nanoprobe 7 in the hydrophobic cavity of HSA induced fluorescent emission, which was validated by competitive binding assays and molecular docking. More importantly, nanoprobe 7 was successfully applied to recognize and quantify HSA in human serum samples. This study demonstrates nanoprobe 7 is a promising tool for clinical real and fast detection of HSA and thus may find many applications, and the molecular assembly based on AIE also opens a new avenue for designing smart nanomaterials for the sensitive and selective detection for varied analytes.


Asunto(s)
Colorantes Fluorescentes/química , Nanoestructuras/química , Albúmina Sérica Humana/análisis , Fluorescencia , Colorantes Fluorescentes/síntesis química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas
5.
Org Biomol Chem ; 15(20): 4375-4382, 2017 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-28474717

RESUMEN

Sulfur dioxide (SO2) is a widely distributed air pollutant, and humans can easily be exposed to sulfite by inhaling SO2, thus inducing respiratory responses and diseases. Hence, to develop a rapid, sensitive and selective method for detection of sulfites is of great importance. Herein, we designed and synthesized a novel tetraphenyl imidazole compound TIBM with aggregation-induced emission enhancement (AIEE). TIBM can self-assemble into well-organized nanoparticles and is reported as an excellent probe for detection of sulfite with high selectivity and sensitivity. The nanoprobe performed very well for the detection of sulfite with an ultrafast detection time (15 s) and an ultralow detection limit (7.4 nM), which is superior to most of the reported probes. Moreover, the nanoprobe was successfully used to detect sulfite in food samples with a favorable accuracy. In addition, we developed paper-based devices for point-of-care detection of sulfite with naked eyes. Furthermore, due to its high water solubility, cell membrane permeability and good biocompatibility, the nanoproboe was further applied to detect sulfite in living systems. This study may offer some helpful insights for designing other AIE-based fluorescent nanosensors for various analytes.


Asunto(s)
Colorantes Fluorescentes/química , Análisis de los Alimentos/métodos , Contaminación de Alimentos/análisis , Nanopartículas/química , Imagen Óptica/métodos , Sulfitos/análisis , Compuestos de Bencilideno , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Fluorescencia , Colorantes Fluorescentes/síntesis química , Humanos , Concentración de Iones de Hidrógeno , Imidazoles , Estructura Molecular , Nitrilos , Tamaño de la Partícula , Teoría Cuántica
6.
Acta Biochim Biophys Sin (Shanghai) ; 46(1): 15-21, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24247269

RESUMEN

A subpopulation of cancer stem cells is recognized as the cause of tumorigenesis and spreading. To investigate the effects of casticin (5,3'-dihydroxy-3,6,7,4'-tetramethoxyflavone), derived from Fructus Viticis Simplicifoliae, on lung cancer stem cells, we isolated and identified a subpopulation of lung cancer stem-like cells (LCSLCs) from non-small-cell lung carcinoma A549 cells with the features including self-renewal capacity and high invasiveness in vitro, elevated tumorigenic activity in vivo, and high expression of stemness markers CD133, CD44, and aldehyde dehydrogenase 1 (ALDH1), using serum-free suspension sphere-forming culture method. We then found that casticin could suppress the proliferation of LCSLCs in a concentration-dependent manner with an IC50 value of 0.4 µmol/L, being much stronger than that in parental A549 cells. In addition, casticin could suppress the self-renewal and invasion of LCSLCs concomitant with decreased CD133, CD44, and ALDH1 protein expression and reduced MMP-9 activity. Further experiments showed that casticin suppressed self-renewal and invasion at least partly through down-regulation of Akt phosphorylation. In conclusion, casticin suppressed the characteristics of LCSLCs, suggesting that casticin may be a candidate compound for curing lung cancer via eliminating cancer stem cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Flavonoides/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Invasividad Neoplásica/prevención & control , Fosforilación
7.
Phytomedicine ; 135: 156036, 2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39277988

RESUMEN

BACKGROUND: Casticin (CAS), a natural flavonoid found in Viticis Fructus, Viticis Cannabifoliae Fructus, and Semen Euphorbiae, shows anti-inflammatory activity and efficacy against various cancers. However, its effect on stemness associated with self-renewal in cervical cancer (CC) cells remains unclear, as well as the underlying mechanism. PURPOSE: The primary objective of this study was to examine the effect of CAS on CC stemness and to explore the underpinning regulatory mechanism. METHODS: HeLa cells underwent treatment with varying concentrations of CAS (0, 10, 30, 100 nM). To evaluate the impacts of CAS on CC stemness and tumorigenicity, sphere- and colony-formation assays and a xenograft model were employed. The study involved screening for changes in miRNAs and their target genes. The miRNA array identified an upregulation in miRNAs, whereas the mRNA array detected a downregulation of specific target genes. The latter genes were found to regulate stem cell-related genes through miR-342-3p in HeLa cells administered CAS. Next, whether miR-342-3p directly targets FOXM1 when upregulated by CAS was assessed by the luciferase reporter assay. qRT-PCR was performed to analyze miR-342-3p expression. Additionally, immunoblotting was conducted to assess the protein amounts of FoxM1 and stemness-related factors (CD133, CD49f, Nanog, and Sox2). Function rescue experiments were conducted to determine the mechanism of CAS in stemness regulation. These experiments involved utilizing a miR-342-3p inhibitor and overexpressing FOXM1 in HeLa cells. RESULTS: CAS decreased in vitro stemness, suppressing sphere- and colony-formation capabilities of CC. It also dose-dependently downregulated the expression of stemness-associated proteins, including CD133, CD49f, Nanog, and Sox2. Moreover, CAS inhibited in vivo carcinogenesis, remarkably reducing tumor growth in mice bearing HeLa cell xenografts. Analysis revealed downregulated FOXM1 expression in HeLa cells treated with CAS. In the luciferase reporter assay, miR-342-3p was found to directly target FOXM1 in CAS-treated HeLa cells. Additionally, miR-342-3p inhibitor transfection successfully rescued CAS' suppressive impact on stemness. Furthermore, overexpression of FOXM1 did not induce changes in miR-342-3p expression. However, it effectively rescued CAS' suppressive effects on stemness. Moreover, CAS also inhibited stemness, upregulated miR-342-3p, and lowered FOXM1 expression in the SiHa cell line. CONCLUSION: CAS suppresses self-renewal-associated stemness by targeting FOXM1 via miR-342-3p upregulation. These findings suggest CAS is promising as a novel therapeutic candidate in CC.

8.
Int J Surg ; 109(12): 3974-3982, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37755372

RESUMEN

OBJECTIVE: To compare robot-assisted thyroidectomy (RT) and open thyroidectomy (OT) through a prospective cohort study focusing on the rate of postoperative hypoparathyroidism, efficacy, and quality of life (QoL). SUMMARY BACKGROUND DATA: Hypoparathyroidism is a frequent complication after thyroidectomy. Reducing the risk of hypoparathyroidism after total thyroidectomy is a crucial and difficult task for thyroid surgeons. METHODS: We prospectively enroled 306 patients with papillary thyroid carcinoma into an RT group and OT group. The former used "super-meticulous" capsular dissection) and the latter used traditional meticulous capsular dissection. Patients were evaluated by scales [Short Form (SF)-36, Visual Impairment Scale (VIS), Swallowing Impairment Scale (SIS), Neck Impairment Scale (NIS), Scar questionnaire (SCAR-Q)]. RESULTS: The rates of transient hypoparathyroidism, permanent hypoparathyroidism, and transient hypocalcemia after surgery in the OT group and RT group were significantly different ( P <0.001). SIS and VIS scores in the two groups were significantly different ( P <0.001). SF-36 showed significant differences ( P <0.001) in the subsections of "physiological function", "body pain", "general health", "vitality", "social function", "role emotional", and "mental health" between the two groups. SCAR-Q showed that the length and appearance of scars showed significant differences between the two groups. CONCLUSIONS: RT with Super-meticulous capsular dissection can protect parathyroid function and improve postoperative QoL, and could be a new option for robot-assisted surgery against thyroid cancer.


Asunto(s)
Carcinoma Papilar , Hipoparatiroidismo , Procedimientos Quirúrgicos Robotizados , Neoplasias de la Tiroides , Humanos , Tiroidectomía/efectos adversos , Calidad de Vida , Procedimientos Quirúrgicos Robotizados/efectos adversos , Estudios Prospectivos , Carcinoma Papilar/cirugía , Neoplasias de la Tiroides/cirugía , Neoplasias de la Tiroides/patología , Hipoparatiroidismo/epidemiología , Hipoparatiroidismo/etiología , Hipoparatiroidismo/prevención & control , Disección del Cuello/efectos adversos , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/cirugía , Estudios Retrospectivos
9.
Chin J Integr Med ; 29(3): 224-232, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35809177

RESUMEN

OBJECTIVE: To explore whether casticin (CAS) suppresses stemness in cancer stem-like cells (CSLCs) obtained from human cervical cancer (CCSLCs) and the underlying mechanism. METHODS: Spheres from HeLa and CaSki cells were used as CCSLCs. DNA methyltransferase 1 (DNMT1) activity and mRNA levels, self-renewal capability (Nanog and Sox2), and cancer stem cell markers (CD133 and CD44), were detected by a colorimetric DNMT activity/inhibition assay kit, quantitative real-time reverse transcription-polymerase chain reaction, sphere and colony formation assays, and immunoblot, respectively. Knockdown and overexpression of DNMT1 by transfection with shRNA and cDNA, respectively, were performed to explore the mechanism for action of CAS (0, 10, 30, and 100 nmol/L). RESULTS: DNMT1 activity was increased in CCSLCs compared with HeLa and CaSki cells (P<0.05). In addition, HeLa-derived CCSLCs transfected with DNMT1 shRNA showed reduced sphere and colony formation abilities, and lower CD133, CD44, Nanog and Sox2 protein expressions (P<0.05). Conversely, overexpression of DNMT1 in HeLa cells exhibited the oppositive effects. Furthermore, CAS significantly reduced DNMT1 activity and transcription levels as well as stemness in HeLa-derived CCSLCs (P<0.05). Interestingly, DNMT1 knockdown enhanced the inhibitory effect of CAS on stemness. As expected, DNMT1 overexpression reversed the inhibitory effect of CAS on stemness in HeLa cells. CONCLUSION: CAS effectively inhibits stemness in CCSLCs through suppression of DNMT1 activation, suggesting that CAS acts as a promising preventive and therapeutic candidate in cervical cancer.


Asunto(s)
Neoplasias del Cuello Uterino , Femenino , Humanos , Línea Celular Tumoral , Células HeLa , Células Madre Neoplásicas/metabolismo , ARN Interferente Pequeño/metabolismo , Neoplasias del Cuello Uterino/metabolismo
10.
Oncol Rep ; 48(4)2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36004463

RESUMEN

Subsequently to the publication of the above article, an interested reader drew to the authors' attention that there appeared to be two pairs of images in Fig. 2A and B on p. 1159 and Fig. 4 on p. 1161 that contained overlapping sections, such that these figures, which were intending to show the results from differently performed experiments, may have been derived from the same original sources. The authors have examined their original data, and realize that, although Fig. 2 was correct as presented in the article, these data were erroneously and inadvertently included in Fig. 4. The revised version of Fig. 4, which shows the inhibition of sphere­forming ability by 7­difluoromethoxyl­5,4'­di­n­octyl genistein (DOFG) in gastric cancer stem­like cells derived from SGC­7901 cells, is shown below, now including the correct data for the panels showing treatment with 0 and 1.0 µmol/l DOFG, and with re­quantification of these data. The authors are grateful to the Editor of Oncology Reports for allowing them the opportunity to publish a Corrigendum, and all the authors agree with its publication. Furthermore, they apologize to the readership for any inconvenience caused. [Oncology Reports 36: 1157­1165, 2016; DOI: 10.3892/or.2016.4848].

11.
Talanta ; 225: 121950, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33592705

RESUMEN

Abnormal level of alkaline phosphatase (ALP) activity has been linked to many diseases in human. The development of fluorescent molecular probes that can report the expression and activity of ALP in various biological systems will be extremely valuable. However, the in vivo monitoring for ALP in living cells and more complex biological systems remains a great challenge. The excited-state intramolecular proton transfer (ESIPT) probe with proportional fluorescence has low background noise, while the aggregation induced emission (AIE) probe has the advantages of signal amplification and good light stability. Herein, an "AIE + ESIPT" fluorescent probe 2-(benzo[d]thiazol-2-yl)-4-(1,4,5-triphenyl-1H-imidazole-2-yl)phenyl dihydrogen phosphate (THP) was constructed for the highly selective and sensitive detection of ALP. By introducing a phosphate ester at the hydroxyl position of the solid fluorophore 2-(benzo[d]thiazol-2-yl)-4-(1,4,5-triphenyl-1H-imidazole-2-yl)phenol, ESIPT was hindered and the probe present a faint blue fluorescence in DMSO solution. While ALP was introduced, causing the phosphate in THP hydrolyzed, and the ESIPT process was restored to yield a yellow fluorescence at 550 nm, thereby achieving proportionality detection. THP exhibited high selectivity and sensitively to ALP with low limit of detection (1.21228 U/L), and the reaction completed within 20 min. In addition, with its outstanding advantages of low biological toxicity and enzyme conversion characteristics, THP has been successfully applied to ALP imaging in living cells (Hela cells, A549 cells and Hek293 cells), and can provide in situ information on the reaction site. Therefore, THP has the potential for detecting ALP activity in biomedical application.


Asunto(s)
Fosfatasa Alcalina , Colorantes Fluorescentes , Células HEK293 , Células HeLa , Humanos , Espectrometría de Fluorescencia
12.
Anticancer Agents Med Chem ; 20(14): 1654-1663, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32329692

RESUMEN

BACKGROUND: We previously demonstrated that isovitexin (apigenin-6-C-glucoside, ISOV) suppressed the stemness of human Hepatocellular Carcinoma (HCC) cells. However, the mechanism of its action remains to be deciphered. OBJECTIVE: The current study was to examine whether ISOV regulates the miR-34a expression and hence suppresses the stemness of HCC SK-Hep-1 cells. METHODS: After identification of the stemness, apoptosis resistance and decreased miR-34a expression of spheres from SK-Hep-1 cells (SK-SC), we utilized transfection of a miR-34a mimic or inhibitor to investigate the effects of ISOV on miR-34a, Bcl-2, Bax and Mcl-1 expression in order to understand the mechanism underlying ISOV-mediated repression of stemness and promotion of apoptosis. RESULTS: Our results demonstrated that SK-SC displayed higher stemness and resistance to apoptosis, as well as reduced miR-34a levels compared to SK-Hep-1 cells. ISOV suppressed sphere and colony formation, and decreased CD44+ cell populations. In addition, ABCG2, ALDH1, and NANOG mRNA levels were decreased, while there was a concomitant increase in miR-34a levels. With regards to apoptosis-related proteins, ISOV increased Bax protein levels, and reduced Bcl-2 and Mcl-1 protein levels in SK-SC. Importantly, there was a cooperative effect when miR-34a was overexpressed in the presence of ISOV in SK-SC, and down-regulation of miR-34a attenuated the effects of ISOV in SK-Hep-1 cells. CONCLUSION: We suggest that ISOV-mediated miR-34a upregulation induces apoptosis and suppresses the stemness of SK-SC. Our data indicate that ISOV exhibits therapeutic potential for the treatment of HCC.


Asunto(s)
Antineoplásicos/farmacología , Apigenina/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , MicroARNs/genética , Regulación hacia Arriba/efectos de los fármacos , Antineoplásicos/química , Apigenina/química , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Estructura Molecular , Relación Estructura-Actividad , Células Tumorales Cultivadas , Regulación hacia Arriba/genética
13.
J Oncol ; 2020: 8978930, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32308683

RESUMEN

BACKGROUND: Whether DNA methyltransferase 1 (DNMT1)/miR-34a/FoxM1 signaling promotes the stemness of liver cancer stem cells (LCSCs) remains unclear. This study aimed to assess whether methylation-based silencing of miR-34a by DNMT1 contributes to stemness features via FoxM1 upregulation in LCSCs. METHODS: The CD133+ subgroup of MHCC97H cells sorted by MACS was used as LCSCs. DNMT1, BMI1, SOX2, and OCT4 mRNA levels, and miR-34a amounts were determined by qRT-PCR. DNMT1, CD44, and FoxM1 proteins were analyzed by immunoblot. Sphere and colony formation abilities were detected by respective assays. CD133+ cell percentages were assessed by flow cytometry. In vivo oncogenicity was evaluated using a tumor xenograft model in mice. The effects of DNMT1/miR-34a signaling on the stemness of LCSCs were examined by knockdown or overexpression of DNMT1 and/or transfection of miR-34a mimic or inhibitor using lentivirus-delivery systems. FoxM1 association with miR-34a was detected by a reporter assay. RESULTS: We here showed that LCSCs exhibited elevated DNMT1 activity and expression, lower miR-34a expression with higher promoter methylation, and stronger stemness, compared with the parental liver cancer cells. DNMT1 knockdown repressed DNMT1, increased miR-34a amounts by promoter demethylation, and reduced stemness in LCSCs, whereas DNMT1 overexpression had the opposite effects in liver cancer cells. Transfection with miR-34a mimic repressed the stemness of LCSCs, while miR-34a inhibitor significantly downregulated miR-34a and enhanced stemness, without affecting DNMT1 in liver cancer cells. MiR-34a mimic rescued the effects of DNMT1 overexpression on the stemness of LCSCs, without affecting DNMT1 expression. Finally, FOXM1 was identified as a direct target by miR-34a in LCSCs. CONCLUSIONS: We revealed that aberrant activation of DNMT1 causes miR-34a promoter methylation and suppression, leading to FoxM1 upregulation by disinhibition and promotion of LCSC stemness. These findings suggest that blockage of DNMT1/miR-34a-mediated FOXM1 upregulation might suppress liver cancer by targeting LCSCs.

14.
Cancer Manag Res ; 12: 5759-5771, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32765079

RESUMEN

BACKGROUND: Manganese superoxide dismutase (MnSOD) induces FoxM1 expression, subsequently contributing to migration in several cancer cells. Isovitexin (ISOV) was recently found to downregulate MnSOD and FoxM1, decreasing stemness in hepatocellular carcinoma (HCC) stem-like cells (HCSLCs). The current study aimed to determine whether inhibition of migration, invasion and EMT in HCSLCs by ISOV results from MnSOD/FoxM1 signaling blockade and subsequent Twist1, Slug, ZEB1 and MMP-2 downregulation. MATERIALS AND METHODS: We examined the migratory and invasive capabilities and EMT phenotype in HCC cells and their HCSLCs, respectively, by wound-healing assay, transwell invasion assay and Western blot after treatment with non-cytotoxic concentrations of ISOV, and explored the mechanism by which ISOV affects migration, invasion and EMT by MnSOD or FoxM1 knockdown and/or overexpression in HCSLCs or HCC cells. RESULTS: The results showed that ISOV not only downregulated MnSOD and FoxM1 but also suppressed the migratory and invasive capabilities and reversed the EMT phenotype in HCSLCs, which was reflected by elevated E-cadherin protein amounts, and reduced N-cadherin, Twist1, Slug, ZEB1 and MMP-2 protein levels. The suppressive effects of ISOV on the migratory and invasive capabilities and EMT phenotype could be potentiated by MnSOD or FoxM1 knockdown in HCSLCs, and attenuated by MnSOD or FoxM1 overexpression in HCC cells. Importantly, FoxM1 overexpression reversed MnSOD knockdown combined with ISOV suppression on the migratory and invasive capabilities and EMT phenotype in HCSLCs, while having little effects on MnSOD expression. CONCLUSION: Collectively, the above findings demonstrated that ISOV suppresses migration, invasion and EMT in HCSLCs by blocking MnSOD/FoxM1 signaling subsequently inhibiting the expression of EMT-related transcription factors and MMP-2.

15.
Oncol Lett ; 20(3): 2506-2515, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32782570

RESUMEN

Manganese superoxide dismutase (MnSOD) promotes invasive and migratory activities by upregulating Forkhead box protein M1 (FoxM1) expression. The present study investigated whether modulation of MnSOD and FoxM1 expression was responsible for the antitumor effects of genistein on cancer stem-like cells (CSLCs) derived from non-small cell lung cancer cells (NSCLCs). Spheroids prepared from H460 or A549 cells were defined as lung cancer stem-like cells (LCSLCs) and were treated with genistein. The Cell Counting Kit-8 assay was performed to assess human lung fibroblast IMR-90 cell proliferation, as well as NSCLC H460 and A549 cell proliferation following treatment with genistein. MnSOD, FoxM1, cluster of differentiation (CD)133, CD44, BMI1 proto-oncogene, polycomb ring finger (Bmi1) and Nanog homeobox (Nanog) protein expression levels were examined via western blotting. The sphere formation assay was conducted to evaluate LCSLC self-renewal potential, and LSCLC migratory and invasive activities were analyzed using the wound healing and Transwell invasion assays, respectively. Knockdown and overexpression of MnSOD and FOXM1 via short hairpin-RNA or cDNA transfection were performed. The results indicated that genistein (80 and 100 µM) suppressed H460 and A549 cell viability compared with IMR-90 cells. Sub-cytotoxic concentrations of genistein (20 and 40 µM) inhibited sphere formation activity and decreased the protein expression levels of CD133, CD44, Bmi1 and Nanog in LCSLCs compared with the control group. Genistein also suppressed the migratory and invasive activities of LCSLCs compared with the control group. MnSOD and FoxM1 overexpression antagonized the effects of genistein (40 µM), whereas MnSOD and FoxM1 knockdown enhanced the inhibitory effects of genistein (20 µM) on CSLC characteristics of LCSLCs. Overall, the results suggested that genistein suppressed lung cancer cell CSLC characteristics by modulating MnSOD and FoxM1 expression levels.

16.
Biomed Pharmacother ; 109: 1960-1966, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30551451

RESUMEN

Accumulating evidences have emphasized the essential roles of differentially expressed miRNAs in papillary thyroid cancer (PTC) and follicular thyroid carcinoma (FTC) progression. MiR-128 has been reported to be down-regulated in multiple cancers to restrain tumor growth. However, the role of miR-128 in the development of PTC and FTC and the underlying mechanism remain to be unclear. In this present study, the results indicated that miR-128 expression was markedly down-regulated in PTC and FTC tissues and various thyroid carcinoma cell lines. Functional analysis indicated that over-expression of miR-128 suppressed PTC and FTC cancer cell growth, induced apoptosis and cell cycle arrest in G0/G1 phase. In addition, miR-128 over-expression markedly inhibited cancer cell migration and invasion. However, the processes above were reversed by silencing miR-128 expressions in thyroid tumor cells. Following, we characterized sphingosine kinase-1 (SPHK1) as a direct target of miR-128 that interacted with the 3'-untranslated region (UTR) of SPHK1, and the results were confirmed by using luciferase-reporter assay. We also observed that SPHK1 expression was decreased and negatively correlated with miR-128 expression in PTC and FTC tissues clinically. Importantly, ectopic expression of SPHK1 significantly abrogated the tumor-suppressive effect induced by miR-128, as supported by the reduced apoptosis, while the enhanced proliferation and metastasis. Finally, over-expressing miR-128 apparently reduced the tumor growth rate and tumor weight in vivo using xenograft tumor model, accompanied with a remarkable decrease of SPHK1. Thus, our study illustrated that miR-128 might be a tumor suppressor microRNA that played an essential role in thyroid carcinoma progression.


Asunto(s)
Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Neoplasias de la Tiroides/genética , Regiones no Traducidas 3'/genética , Animales , Apoptosis/genética , Carcinoma Papilar/genética , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Regulación hacia Abajo/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Glándula Tiroides/patología
17.
Talanta ; 194: 38-45, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30609547

RESUMEN

Aluminum ions, as a kind of non-essential element, show a negative impact on plants growth and contribute to diseases caused by nervous breakdown like Alzheimer's disease and Parkinson's disease. Hence, the detection of Al3+ in environment and human body is of great significance and valuableness. Herein, a N-H-type excited-state intramolecular proton transfer (ESIPT) based self-assemble nanoprobe has been designed and synthesized for the detection of Al3+. Owing to phthalandione and tosyl group, probe ABTTA could self-assemble into nanoparticles with nearly no fluorescence. However, ABTTA would be further aggregated by Al3+ ions in aqueous solution which results in enhanced fluorescence (peaking at 555 nm). Based on this principle, a novel analytical method for detection of Al3+ was established with a limit of detection of 50 nM, which was much better than most of reported probes. Moreover, the nanoprobe with excellent water solubility was successfully applied for the detection of Al3+ in real water sample with satisfactory measurement accuracy. Benefited from the favorable water solubility, the nanoprobe was further utilized to the detection of Al3+ in human cancer samples, demonstrating its potential application for biological imaging.


Asunto(s)
Aluminio/análisis , Aluminio/química , Ácido Benzoico/química , Sondas Moleculares/química , Nanopartículas/química , Agua/química , Supervivencia Celular , Células HeLa , Humanos , Enlace de Hidrógeno , Concentración de Iones de Hidrógeno , Modelos Moleculares , Conformación Molecular , Protones
18.
Oncol Rep ; 40(3): 1401-1410, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30015975

RESUMEN

Casticin, a natural polymethoxyflavone isolated from A. annua, V. trifolia, and V. agnus­castus induces apoptosis in cancer cells by activating FoxO3a. However, whether casticin inhibits in vitro carcinogenesis and cancer stem cell (CSC) characteristics, and whether casticin activates FoxO3a in small cell lung cancer (SCLC) cells remain unclear. We here demonstrated that casticin decreased sphere­ and colony­formation capabilities, and downregulated uPAR and CD133 in second­generation spheres, which were considered as lung cancer stem­like cells (LCSLCs), from SCLC H446 cells, in a concentration­dependent manner. In addition, casticin dose­dependently elevated the phosphorylation levels of AMPK and ACC, and reduced p­FoxO3a expression. The above effects were attenuated by AMPK knockdown with small interfering RNAs (siRNAs). FoxO3a silencing resulted in decreased protein expression of FoxO3a, increased in vitro carcinogenesis and CSC characteristics, with no appreciable effects on AMPK and ACC phosphorylation, and displayed similar activities to those neutralizing the effects of casticin on in vitro carcinogenesis and CSC characteristics. These findings reveal a novel mechanism for regulating AMPK/FoxO3a signaling in response to casticin, suggesting a new strategy for SCLC therapy by targeting cancer stem­like cells.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Flavonoides/farmacología , Proteína Forkhead Box O3/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Quinasas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Apoptosis , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proliferación Celular , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosforilación , Transducción de Señal/efectos de los fármacos , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Protein Pept Lett ; 25(6): 548-559, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29848256

RESUMEN

BACKGROUND: Protein is a macromolecule, consisting of one or more long chains of amino residues. And it plays a vital role in the life system, especially for the evaluation of many diseases. Thus, the quantitative measurement of protein is benefit for the diagnosis and monitoring of diseases. Among the sensing methods currently developed, fluorescence detection is of particular concern as a technique with the potential to provide accurate and quantitative analyses. A wide variety of molecular probes for protein detection have been designed and synthesized for sensing, imaging, and biomedical applications. CONCLUSION: In this report, we provide an overview of the varied probes from small molecular to nanoparticle-based sensors which have been applied to detect a various proteins from bench to bed. Furthermore, some emerging trends, future directions as well as challenges in this rapidly growing field are discussed.


Asunto(s)
Colorantes Fluorescentes/química , Proteínas/análisis , Técnicas Biosensibles/métodos , Fluorescencia , Humanos , Imagen Molecular/métodos , Nanopartículas/química , Investigación Biomédica Traslacional
20.
Oncol Rep ; 36(3): 1731-8, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27461522

RESUMEN

Sorafenib, a multi-kinase inhibitor, has shown its promising antitumor effect in a series of clinical trials, and has been approved as the current standard treatment for advanced hepatocellular carcinoma (HCC). 8-Bromo­7-methoxychrysin (BrMC) is a novel chrysin synthetic analogue that has been reported to inhibit the growth of various tumor cells and possess properties for targeting liver cancer stem cells (LCSCs) . The present study investigated the synergistic targeting effects on the properties of liver cancer stem-like cells (LCSLCs) by a combination of sorafenib and BrMC in SMMC-7721 cell line. We also investigated whether this effect involves regulation of HIF-1α, Twist and NF-κB protein. We found that the sphere-forming cells (SFCs) from the SMMC­7721 cells possessed the properties of LCSLCs. Sorafenib diminished the self-renewal capacity and downregulated the expression of stem cell biomarkers (CD133, CD44 and ALDH1) in a dose-dependent manner, while BrMC cooperated with sorafenib to strengthen this inhibition. Moreover, the combination of sorafenib and BrMC led to a remarkable decrease in the cellular migration and invasion, the downregulation of N-cadherin protein and upregulation of E-cadherin protein, and increase of cell apoptosis in LCSLCs. BrMC has a remarkable antagonistic effect on the upregulation of protein expression and DNA binding activity of NF-κB (p65) induced by sorafenib. In addition, our results indicated that the synergistic inhibition of sorafenib and BrMC on the characteristics of LCSLCs involves the downregulated expression of HIF-1α and EMT regulator Twist1. Collectively, the combination therapy of sorafenib and BrMC could be a new and promising therapeutic approach in the treatment of HCC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Flavonoides/administración & dosificación , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/administración & dosificación , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Niacinamida/administración & dosificación , Sorafenib
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA