Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 663
Filtrar
1.
Bull Exp Biol Med ; 176(1): 82-86, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38091144

RESUMEN

We studied the effects of single and combined action of protons and carbon ions 12C6+ on the pool of MCF-7 human breast cancer stem cells. Single irradiation with a beam of protons or carbon ions had no significant effects on the relative number of cancer stem cells (CSC). The effects of combined irradiation in a total equieffective dose of 4 Gy depended on the sequence of exposure to ionizing radiations: the relative number of CSC did not change after irradiation with carbon ions and then with protons, but increased in the case of the reverse sequence. The most favorable result, i.e. a decrease in the CSC pool, was observed in the case of sequential irradiation with carbon ions and protons and their equal contribution to total equieffective dose. In this case, the absolute number of CSC decreased by on average 2.1 times in comparison with the control (p<0.05). The revealed regularities are of interest for the further development of new methods of radiation therapy.


Asunto(s)
Neoplasias de la Mama , Protones , Humanos , Femenino , Células MCF-7 , Neoplasias de la Mama/radioterapia , Iones , Carbono , Células Madre Neoplásicas/efectos de la radiación
2.
Life Sci ; 320: 121541, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-36870386

RESUMEN

Modern cancer chemotherapy originated in the 1940s, and since then, many chemotherapeutic agents have been developed. However, most of these agents show limited response in patients due to innate and acquired resistance to therapy, which leads to the development of multi-drug resistance to different treatment modalities, leading to cancer recurrence and, eventually, patient death. One of the crucial players in inducing chemotherapy resistance is the aldehyde dehydrogenase (ALDH) enzyme. ALDH is overexpressed in chemotherapy-resistant cancer cells, which detoxifies the generated toxic aldehydes from chemotherapy, preventing the formation of reactive oxygen species and, thus, inhibiting the induction of oxidative stress and the stimulation of DNA damage and cell death. This review discusses the mechanisms of chemotherapy resistance in cancer cells promoted by ALDH. In addition, we provide detailed insight into the role of ALDH in cancer stemness, metastasis, metabolism, and cell death. Several studies investigated targeting ALDH in combination with other treatments as a potential therapeutic regimen to overcome resistance. We also highlight novel approaches in ALDH inhibition, including the potential synergistic employment of ALDH inhibitors in combination with chemotherapy or immunotherapy against different cancers, including head and neck, colorectal, breast, lung, and liver.


Asunto(s)
Aldehído Deshidrogenasa , Resistencia a Antineoplásicos , Inmunoterapia , Neoplasias , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/radioterapia , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Animales , Metástasis de la Neoplasia , Muerte Celular , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/efectos de la radiación
3.
Cells ; 11(14)2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35883571

RESUMEN

Systematic recurrence of glioblastoma (GB) despite surgery and chemo-radiotherapy is due to GB stem cells (GBSC), which are particularly invasive and radioresistant. Therefore, there is a need to identify new factors that might be targeted to decrease GBSC invasive capabilities as well as radioresistance. Patient-derived GBSC were used in this study to demonstrate a higher expression of the glycoprotein M6a (GPM6A) in invasive GBSC compared to non-invasive cells. In 3D invasion assays performed on primary neurospheres of GBSC, we showed that blocking GPM6A expression by siRNA significantly reduced cell invasion. We also demonstrated a high correlation of GPM6A with the oncogenic protein tyrosine phosphatase, PTPRZ1, which regulates GPM6A expression and cell invasion. The results of our study also show that GPM6A and PTPRZ1 are crucial for GBSC sphere formation. Finally, we demonstrated that targeting GPM6A or PTPRZ1 in GBSC increases the radiosensitivity of GBSC. Our results suggest that blocking GPM6A or PTPRZ1 could represent an interesting approach in the treatment of glioblastoma since it would simultaneously target proliferation, invasion, and radioresistance.


Asunto(s)
Glioblastoma , Glicoproteínas de Membrana , Proteínas del Tejido Nervioso , Glioblastoma/genética , Glioblastoma/radioterapia , Humanos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/efectos de la radiación , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/efectos de la radiación , Tolerancia a Radiación , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores
4.
Asian Pac J Cancer Prev ; 23(2): 419-427, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35225452

RESUMEN

BACKGROUND: The presence of cancer stem-like cells within tumor microenvironment distinctly governs response to chemo-radiotherapy. The ALDH1 (Aldehyde dehydrogenase 1) has emerged as a cancer stem cell (CSC) marker in various tumors. The aim of the study was to examine the expression of ALDH1 in HNSCC patients undergoing radiotherapy to evaluate its correlation with clinicopathological parameter, treatment response and survival. METHODS: Expression of ALDH1 was evaluated by immunohistochemistry in 90 histopathologically confirmed HNSCC patients and 90 matched controls. The association between ALDH1 expression, clinicopathological parameters and treatment response was determined. RESULTS: The immunohistochemistry results showed that ALDH1 was consistently expressed in all the HNSCC specimens although at different intensities. On the other hand, control specimens did not show similar expression of ALDH1. ALDH1 expression demonstrated statistically significant association with tumor size (p<0.001), lymph node status (p<0.001), stage (p<0.001), grade (p<0.001) and treatment response (p<0.001). Multivariate ordinal logistic regression analysis indicated alcohol and ALDH1 as an independent predictor of responsiveness to radiotherapy in HNSCC patients. Multivariate Cox regression analysis indicated that lymph node status (p=0.020), grade (p=0.006) and recurrence (p=0.002) were potential independent predictors of overall survival. CONCLUSION: From previous studies, ALDH1 has been contemplated not only as a promising prognostic and diagnostic marker but also as a likely drug target. Our study gives new understanding regarding the association between ALDH1, cancer prognosis and radioresistance. Our findings suggest that ALDH1, lymph node status, grade and alcohol could be the viable targets for HNSCC and it also provides new prospects for radiotherapy sensitivity in HNSCC.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/efectos de la radiación , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de Cabeza y Cuello/radioterapia , Tolerancia a Radiación/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/enzimología , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Adulto , Biomarcadores de Tumor/efectos de la radiación , Estudios de Casos y Controles , Femenino , Neoplasias de Cabeza y Cuello/mortalidad , Humanos , Inmunohistoquímica , Ganglios Linfáticos/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Células Madre Neoplásicas/efectos de la radiación , Pronóstico , Modelos de Riesgos Proporcionales , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Resultado del Tratamiento , Microambiente Tumoral/efectos de la radiación
5.
Sci Rep ; 12(1): 1056, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35058559

RESUMEN

Cancer stem-like cells (CSCs) within solid tumors exhibit radioresistance, leading to recurrence and distant metastasis after radiotherapy. To experimentally study the characteristics of CSCs, radioresistant cell lines were successfully established using fractionated X-ray irradiation. The fundamental characteristics of CSCs in vitro have been previously reported; however, the relationship between CSC and acquired radioresistance remains uncertain. To efficiently study this relationship, we performed both in vitro experiments and theoretical analysis using a cell-killing model. Four types of human oral squamous carcinoma cell lines, non-radioresistant cell lines (SAS and HSC2), and radioresistant cell lines (SAS-R and HSC2-R), were used to measure the surviving fraction after single-dose irradiation, split-dose irradiation, and multi-fractionated irradiation. The SAS-R and HSC2-R cell lines were more positive for one of the CSC marker aldehyde dehydrogenase activity than the corresponding non-radioresistant cell lines. The theoretical model analysis showed that changes in both the experimental-based ALDH (+) fractions and DNA repair efficiency of ALDH (-) fractions (i.e., sub-lethal damage repair) are required to reproduce the measured cell survival data of non-radioresistant and radioresistant cell lines. These results suggest that the enhanced cell recovery in SAS-R and HSC2-R is important when predicting tumor control probability in radiotherapy to require a long dose-delivery time; in other words, intensity-modulated radiation therapy is ideal. This work provides a precise understanding of the mechanism of radioresistance, which is induced after irradiation of cancer cells.


Asunto(s)
Reparación del ADN , Células Madre Neoplásicas/efectos de la radiación , Tolerancia a Radiación , Aldehído Deshidrogenasa/metabolismo , Línea Celular Tumoral/efectos de la radiación , Supervivencia Celular , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Rayos X
6.
Life Sci ; 288: 120163, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34822797

RESUMEN

AIMS: To investigate the role of tumor acidification in cell behavior, migration, and treatment resistance of oral squamous cell carcinoma (OSCC). MAIN METHODS: The SCC4 and SCC25 cell lines were exposed to acidified (pH 6.8) cell culture medium for 7 days. Alternatively, a long-term acidosis was induced for 21 days. In addition, to mimic dynamic pH fluctuation of the tumor microenvironment, cells were reconditioned to neutral pH after experimental acidosis. This study assessed cell proliferation and viability by sulforhodamine B and flow cytometry. Individual and collective cell migration was analyzed by wound healing, time lapse, and transwell assays. Modifications of cell phenotype, EMT induction and stemness potential were investigated by qRT-PCR, western blot, and immunofluorescence. Finally, resistance to chemo- and radiotherapy of OSCC when exposed to acidified environmental conditions (pH 6.8) was determined. KEY FINDINGS: The exposure to an acidic microenvironment caused an initial reduction of OSCC cells viability, followed by an adaptation process. Acidic adapted cells acquired a mesenchymal-like phenotype along with increased migration and motility indexes. Moreover, tumoral extracellular acidity was capable to induce cellular stemness and to increase chemo- and radioresistance of oral cancer cells. SIGNIFICANCE: In summary, the results showed that the acidic microenvironment leads to a more aggressive and treatment resistant OSCC cell population.


Asunto(s)
Ácidos/efectos adversos , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Neoplasias de la Boca/patología , Células Madre Neoplásicas/patología , Tolerancia a Radiación , Microambiente Tumoral , Antineoplásicos/farmacología , Apoptosis , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Movimiento Celular , Proliferación Celular , Cisplatino/efectos adversos , Rayos gamma/efectos adversos , Humanos , Neoplasias de la Boca/etiología , Neoplasias de la Boca/terapia , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación , Células Tumorales Cultivadas
7.
Int J Mol Sci ; 22(23)2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34884511

RESUMEN

Glioblastoma multiforme is a malignant primary brain tumor with a poor prognosis and high rates of chemo-radiotherapy failure, mainly due to a small cell fraction with stem-like properties (GSCs). The mechanisms underlying GSC response to radiation need to be elucidated to enhance sensitivity to treatments and to develop new therapeutic strategies. In a previous study, two GSC lines, named line #1 and line #83, responded differently to carbon ions and photon beams, with the differences likely attributable to their own different metabolic fingerprint rather than to radiation type. Data from the literature showed the capability of RHPS4, a G-quadruplex stabilizing ligand, to sensitize the glioblastoma radioresistant U251MG cells to X-rays. The combined metabolic effect of ligand #190, a new RHPS4-derivative showing reduced cardiotoxicity, and a photon beam has been monitored by magnetic resonance (MR) spectroscopy for the two GSC lines, #1 and #83, to reveal whether a synergistic response occurs. MR spectra from both lines were affected by single and combined treatments, but the variations of the analysed metabolites were statistically significant mainly in line #1, without synergistic effects due to combination. The multivariate analysis of ten metabolites shows a separation between control and treated samples in line #1 regardless of treatment type, while separation was not detected in line #83.


Asunto(s)
Acridinas/farmacología , G-Cuádruplex , Glioblastoma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Fotones , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Supervivencia Celular , Glioblastoma/patología , Glioblastoma/radioterapia , Humanos , Ligandos , Espectroscopía de Resonancia Magnética/métodos , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/efectos de la radiación
8.
PLoS One ; 16(12): e0260864, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34852024

RESUMEN

Secreted frizzled-related protein 2 (SFRP2) is a glycoprotein with frizzled-like cysteine-rich domain that binds with Wnt ligands or frizzled receptors to regulate Wnt signaling. SFRP2 is frequently hypermethylated in glioma patients, and analysis of TCGA data indicates that SFRP2 is one of the most downregulated genes in radiotherapy treated glioma patients. In the present study, we aimed to explore the potential function of SFRP2 in tumorigenesis and radioresistance of glioma. The RNA sequencing data of TCGA glioma samples were downloaded and analyzed. SFRP2 expression in 166 glioma patients was evaluated by qRT-PCR. The potential functions of SFRP2 in glioma were evaluated by loss-of-function assays and gain-of-function assays in glioma cell lines. We found that SFRP2 was downregulated in radiotherapy-treated glioma patients, and low SFRP2 expression was correlated with advanced tumor stage and poor prognosis. CRISP/Cas9-meidated SFRP2 knockdown promoted soft agar colony formation, cancer stemness and radioresistance of glioma cells, while enforced SFRP2 expression exhibited opposite effects. Moreover, Wnt/ß-catenin signaling was activated in radiotherapy treated glioma patients. SFRP2 knockdown activated Wnt/ß-catenin signaling in glioma cell lines, while overexpression of SFRP2 inhibited Wnt/ß-catenin activation. Besides, pharmacological inhibition of Wnt/ß-catenin signaling by XAV-939 abrogated the effects of SFRP2 knockdown on cancer stemness and radioresistance of glioma cells. Our data for the first time demonstrated a role of SFRP2 in radioresistance of glioma cells, and suggested that inhibition of Wnt/ß-catenin signaling might be a potential strategy for increasing radiosensitivity of glioma patients.


Asunto(s)
Neoplasias Encefálicas/patología , Regulación hacia Abajo , Perfilación de la Expresión Génica/métodos , Glioma/patología , Proteínas de la Membrana/genética , Células Madre Neoplásicas/metabolismo , Tolerancia a Radiación , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Regulación hacia Abajo/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Glioma/genética , Glioma/radioterapia , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Masculino , Ratones , Clasificación del Tumor , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación , Pronóstico , Tolerancia a Radiación/efectos de los fármacos , Análisis de Secuencia de ARN , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/efectos de la radiación
9.
Bull Exp Biol Med ; 172(2): 245-249, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34853973

RESUMEN

We compared the formation of γH2AX foci (marker of DNA double-strand breaks) in human lung fibroblasts (MRC-5 line) during their 24-h incubation in a medium containing 3H-labeled thymidine or amino acids (glycine, alanine, and proline) with specific radioactivity from 100 to 400 MBq/liter. A linear dependence of changes in the number of γH2AX foci on the specific radioactivity of the medium was revealed. The quantitative yield of DNA double-strand breaks under the influence of 3H-thymidine was more than 2-fold higher than under the influence of 3H-labeled amino acids. Comparative analysis of the yields of DNA double-strand breaks during cell incubation with 3H-labeled amino acids showed that 3H-alanine produced more pronounced effect that 3H-proline, which is consistent with the data on the content of their non-radioactive analogs in chromatin proteins.


Asunto(s)
Roturas del ADN de Doble Cadena , Fibroblastos , Histonas/genética , Pulmón , Tritio/farmacología , Aminoácidos/química , Aminoácidos/farmacología , Células Cultivadas , Medios de Cultivo/química , Medios de Cultivo/farmacología , Roturas del ADN de Doble Cadena/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Histonas/metabolismo , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Pulmón/efectos de la radiación , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación , Timidina/química , Timidina/farmacología , Tritio/química
10.
J Exp Clin Cancer Res ; 40(1): 282, 2021 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-34488821

RESUMEN

BACKGROUND: Glioblastoma Multiforme (GBM) is a malignant primary brain tumor in which the standard treatment, ionizing radiation (IR), achieves a median survival of about 15 months. GBM harbors glioblastoma stem-like cells (GSCs), which play a crucial role in therapeutic resistance and recurrence. METHODS: Patient-derived GSCs, GBM cell lines, intracranial GBM xenografts, and GBM sections were used to measure mRNA and protein expression and determine the related molecular mechanisms by qRT-PCR, immunoblot, immunoprecipitation, immunofluorescence, OCR, ECAR, live-cell imaging, and immunohistochemistry. Orthotopic GBM xenograft models were applied to investigate tumor inhibitory effects of glimepiride combined with radiotherapy. RESULTS: We report that GSCs that survive standard treatment radiation upregulate Speedy/RINGO cell cycle regulator family member A (Spy1) and downregulate CAP-Gly domain containing linker protein 3 (CLIP3, also known as CLIPR-59). We discovered that Spy1 activation and CLIP3 inhibition coordinately shift GBM cell glucose metabolism to favor glycolysis via two cellular processes: transcriptional regulation of CLIP3 and facilitating Glucose transporter 3 (GLUT3) trafficking to cellular membranes in GBM cells. Importantly, in combination with IR, glimepiride, an FDA-approved medication used to treat type 2 diabetes mellitus, disrupts GSCs maintenance and suppresses glycolytic activity by restoring CLIP3 function. In addition, combining radiotherapy and glimepiride significantly reduced GBM growth and improved survival in a GBM orthotopic xenograft mouse model. CONCLUSIONS: Our data suggest that radioresistant GBM cells exhibit enhanced stemness and glycolytic activity mediated by the Spy1-CLIP3 axis. Thus, glimepiride could be an attractive strategy for overcoming radioresistance and recurrence by rescuing CLIP3 expression.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/radioterapia , Glucólisis , Humanos , Masculino , Ratones , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/genética , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/efectos de la radiación , Tolerancia a Radiación , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34502158

RESUMEN

Cancer stem cells (CSCs) play a critical role in radiation resistance and recurrence. Thus, drugs targeting CSCs can be combined with radiotherapy to improve its antitumor efficacy. Here, we investigated whether a gallotannin extract from Bouea macrophylla seed (MPSE) and its main bioactive compound, pentagalloyl glucose (PGG), could suppress the stemness trait and further confer the radiosensitivity of head and neck squamous cell carcinoma (HNSCC) cell lines. In this study, we evaluate the effect of MPSE or PGG to suppress CSC-like phenotypes and radiosensitization of HNSCC cell lines using a series of in vitro experiments, tumorsphere formation assay, colony formation assay, apoptosis assay, and Western blotting analysis. We demonstrate that MPSE or PGG is able to suppress tumorsphere formation and decrease protein expression of cancer stem cell markers. MPSE or PGG also enhanced the radiosensitivity in HNSCC cells. Pretreatment of cells with MPSE or PGG increased IR-induced DNA damage (γ-H2Ax) and enhanced radiation-induced cell death. Notably, we observed that pretreatment with MPSE or PGG attenuated the IR-induced stemness-like properties characterized by tumorsphere formation and the CD44 CSC marker. Our findings describe a novel strategy for increasing therapeutic efficacy for head and neck cancer patients using the natural products MPSE and PGG.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Taninos Hidrolizables/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación , Extractos Vegetales/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Semillas/química , Anacardiaceae/química , Animales , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello , Humanos , Taninos Hidrolizables/química , Ratones , Estructura Molecular , Células Madre Neoplásicas/metabolismo , Extractos Vegetales/química , Fármacos Sensibilizantes a Radiaciones/química , Semillas/anatomía & histología
12.
Int J Mol Sci ; 22(17)2021 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-34502547

RESUMEN

Cancer stem cells (CSCs) can be induced from differentiated cancer cells in the tumor microenvironment or in response to treatments and exhibit chemo- and radioresistance, leading to tumor recurrence and metastasis. We previously reported that triple negative breast cancer (TNBC) cells with acquired radioresistance exhibited more aggressive features due to an increased CSC population. Therefore, here, we isolated CSCs from radiotherapy-resistant (RT-R)-TNBC cells and investigated the effects of these CSCs on tumor progression and NK cell-mediated cytotoxicity. Compared to MDA-MB-231 and RT-R-MDA-MB-231 cells, CD24-/low/CD44+ cells isolated from RT-R-MDA-MB-231 cells showed increased proliferation, migration and invasion abilities, and induced expression of tumor progression-related molecules. Moreover, similar to MDA-MB-231 cells, CD24-/low/CD44+ cells recruited NK cells but suppressed NK cell cytotoxicity by regulating ligands for NK cell activation. In an in vivo model, CD24-/low/CD44+ cell-injected mice showed enhanced tumor progression and lung metastasis via upregulation of tumor progression-related molecules and altered host immune responses. Specifically, NK cells were recruited into the peritumoral area tumor but lost their cytotoxicity due to the altered expression of activating and inhibitory ligands on tumors. These results suggest that CSCs may cause tumor evasion of immune cells, resulting in tumor progression.


Asunto(s)
Neoplasias de la Mama/inmunología , Citotoxicidad Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Células Madre Neoplásicas/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Antígeno CD24/inmunología , Antígeno CD24/metabolismo , Línea Celular , Línea Celular Tumoral , Movimiento Celular/inmunología , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/efectos de la radiación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Receptores de Hialuranos/inmunología , Receptores de Hialuranos/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Radioterapia/métodos
13.
Cell Rep ; 36(9): 109647, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34469737

RESUMEN

Brain tumor stem cells (BTSCs) and intratumoral heterogeneity represent major challenges in glioblastoma therapy. Here, we report that the LGALS1 gene, encoding the carbohydrate binding protein, galectin1, is a key regulator of BTSCs and glioblastoma resistance to therapy. Genetic deletion of LGALS1 alters BTSC gene expression profiles and results in downregulation of gene sets associated with the mesenchymal subtype of glioblastoma. Using a combination of pharmacological and genetic approaches, we establish that inhibition of LGALS1 signaling in BTSCs impairs self-renewal, suppresses tumorigenesis, prolongs lifespan, and improves glioblastoma response to ionizing radiation in preclinical animal models. Mechanistically, we show that LGALS1 is a direct transcriptional target of STAT3 with its expression robustly regulated by the ligand OSM. Importantly, we establish that galectin1 forms a complex with the transcription factor HOXA5 to reprogram the BTSC transcriptional landscape. Our data unravel an oncogenic signaling pathway by which the galectin1/HOXA5 complex maintains BTSCs and promotes glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Galectina 1/metabolismo , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Transcripción Genética , Anciano , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Calixarenos/farmacología , Línea Celular Tumoral , Proliferación Celular , Autorrenovación de las Células , Receptores ErbB/genética , Receptores ErbB/metabolismo , Galectina 1/antagonistas & inhibidores , Galectina 1/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/radioterapia , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones SCID , Persona de Mediana Edad , Mutación , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/efectos de la radiación , Tolerancia a Radiación , Fármacos Sensibilizantes a Radiaciones/farmacología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Neurochem Res ; 46(12): 3222-3246, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34420141

RESUMEN

Since few reports have mentioned the role of FOSL1 in the radiotherapy sensitivity of glioma, this study would dig deep into this aspect. Cancer stem cells (CSCs) isolated by magnetic bead assay were identified by microscopy, qRT-PCR and western blot. The number of apoptotic cells was counted 72 h after X-ray irradiation to evaluate the sensitivity of cancer cells to radiotherapy. The effects of radiotherapy, FOSL1 and miR-27a-5p on basic cell functions were detected by functional experiments. The expressions of FOSL1, apoptosis-related genes and miR-27a-5p were detected by qRT-PCR and western blot as needed. The differential expression of FOSL1 and the effect of miR-27a-5p on survival rate were analyzed using GEPIA and UALCAN, respectively. FOSL1 was found highly expressed in glioma cells and patients. The appearance of spherical cells and high expressions of CSC-related markers indicated the successful isolation of CSC-like cells. The increment of X-ray dose enhanced the sensitivity of cancer cells to radiotherapy. Radiotherapy down-regulated cell viability and the expressions of FOSL1 and Bcl-2, but up-regulated cell apoptosis and the expressions of cleaved caspase-3 and Bax, which could be partially reversed by overexpressed FOSL1 or further enhanced by shFOSL1. MiR-27a-5p was highly expressed in in patients with glioma, which was associated with poor prognosis, while shFOSL1-inhibited miR-27a-5p expression enhanced the sensitivity of glioma stem cells to radiotherapy. In vivo experiments further verified the results obtained from in vitro experiments. Silent FOSL1 strengthened the radiosensitivity of glioma by down-regulating miR-27a-5p.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Glioma/radioterapia , MicroARNs/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de la radiación , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Tolerancia a Radiación , Animales , Apoptosis , Proliferación Celular , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-fos/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Ann Clin Lab Sci ; 51(4): 521-528, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34452890

RESUMEN

OBJECTIVE: Radioresistance-induced locoregional recurrence remains a major cause of low survival rates. However, the mechanism of treatment failure in these lung cancer patients has not been determined. In the current study, we tried to explore the potential molecular mechanism. METHODS: The fractionated irradiations were continued until the total concentration reached 80 Gy, and we established radioresistant subclones derived from A549 lines (designated as A549/R). The MTT assay, wound healing assay, transwell assay, and soft agar colony formation assay were employed to detect the proliferation, migration, invasion, and clonogenicity of the cells, respectively. Western blot and Fluorescence Activating Cell Sorter (FACS) indicated the expression of the markers. RESULTS: A549/R cells proliferated more slowly than the parental A549 cells. A significant acceleration in cell migration and invasion was revealed in A549/R cells compared with A549 cells. The expression levels of mesenchymal markers (N-cadherin, vimentin, claudin-1, and Snail) increased, while epithelial markers (E-cadherin and ß-catenin) decreased in A549/R cells. Meanwhile, the expression levels of stemness markers (Oct4, Notch1, and CD133) increased in A549/R cells, and A549/R cells showed more sphere-forming activity compared with A549 cells. CONCLUSION: Fractionated irradiation could promote epithelial-mesenchymal transition and enhance the migration, invasion, and stemness-like properties in A549 cells, elucidating the possible radioresistance mechanisms of the cancer cells.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Rayos gamma , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , Células A549 , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Movimiento Celular , Proliferación Celular , Fraccionamiento de la Dosis de Radiación , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación
16.
BMC Complement Med Ther ; 21(1): 189, 2021 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-34217266

RESUMEN

BACKGROUND: Radioresistance can pose a significant obstacle to the effective treatment of breast cancers. Epithelial-mesenchymal transition (EMT) is a critical step in the acquisition of stem cell traits and radioresistance. Here, we investigated whether Maprang seed extract (MPSE), a gallotannin-rich extract of seed from Bouea macrophylla Griffith, could inhibit the radiation-induced EMT process and enhance the radiosensitivity of breast cancer cells. METHODS: Breast cancer cells were pre-treated with MPSE before irradiation (IR), the radiosensitizing activity of MPSE was assessed using the colony formation assay. Radiation-induced EMT and stemness phenotype were identified using breast cancer stem cells (CSCs) marker (CD24-/low/CD44+) and mammosphere formation assay. Cell motility was determined via the wound healing assay and transwell migration. Radiation-induced cell death was assessed via the apoptosis assay and SA-ß-galactosidase staining for cellular senescence. CSCs- and EMT-related genes were confirmed by real-time PCR (qPCR) and Western blotting. RESULTS: Pre-treated with MPSE before irradiation could reduce the clonogenic activity and enhance radiosensitivity of breast cancer cell lines with sensitization enhancement ratios (SERs) of 2.33 and 1.35 for MCF7 and MDA-MB231cells, respectively. Pretreatment of breast cancer cells followed by IR resulted in an increased level of DNA damage maker (γ-H2A histone family member) and enhanced radiation-induced cell death. Irradiation induced EMT process, which displayed a significant EMT phenotype with a down-regulated epithelial marker E-cadherin and up-regulated mesenchymal marker vimentin in comparison with untreated breast cancer cells. Notably, we observed that pretreatment with MPSE attenuated the radiation-induced EMT process and decrease some stemness-like properties characterized by mammosphere formation and the CSC marker. Furthermore, pretreatment with MPSE attenuated the radiation-induced activation of the pro-survival pathway by decrease the expression of phosphorylation of ERK and AKT and sensitized breast cancer cells to radiation. CONCLUSION: MPSE enhanced the radiosensitivity of breast cancer cells by enhancing IR-induced DNA damage and cell death, and attenuating the IR-induced EMT process and stemness phenotype via targeting survival pathways PI3K/AKT and MAPK in irradiated breast cancer cells. Our findings describe a novel strategy for increasing the efficacy of radiotherapy for breast cancer patients using a safer and low-cost natural product, MPSE.


Asunto(s)
Transición Epitelial-Mesenquimal , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación , Extractos Vegetales/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Anacardiaceae/química , Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama , Línea Celular Tumoral , Femenino , Humanos , Taninos Hidrolizables/farmacología , Semillas/química
17.
J Cancer Res Clin Oncol ; 147(11): 3299-3312, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34291358

RESUMEN

PURPOSE: While the stem cell marker Musashi-1 (MSI-1) has been identified as a key player in a wide array of malignancies, few findings exist on its prognostic relevance and relevance for cancer cell death and therapy resistance in breast cancer. METHODS: First, we determined prognostic relevance of MSI-1 in database analyses regarding multiple survival outcomes. To substantiate findings, MSI-1 was artificially downregulated in MCF-7 breast cancer cells and implications for cancer stem cell markers, cell apoptosis and apoptosis regulator p21, proliferation and radiation response were analyzed via flow cytometry and colony formation. Radiation-induced p21 expression changes were investigated using a dataset containing patient samples obtained before and after irradiation and own in vitro experiments. RESULTS: MSI-1 is a negative prognostic marker for disease-free and distant metastasis-free survival in breast cancer and tends to negatively influence overall survival. MSI-1 knockdown downregulated stem cell gene expression and proliferation, but increased p21 levels and apoptosis. Similar to the MSI-1 knockdown effect, p21 expression was strongly increased after irradiation and was expressed at even higher levels in MSI-1 knockdown cells after irradiation. Finally, combined use of MSI-1 silencing and irradiation reduced cancer cell survival. CONCLUSION: MSI-1 is a prognostic marker in breast cancer. MSI-1 silencing downregulates proliferation while increasing apoptosis. The anti-proliferation mediator p21 was upregulated independently after both MSI-1 knockdown and irradiation and even more after both treatments combined, suggesting synergistic potential. Radio-sensitization effects after combining radiation and MSI-1 knockdown underline the potential of MSI-1 as a therapeutic target.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/radioterapia , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Anciano , Apoptosis/fisiología , Neoplasias de la Mama/patología , Proliferación Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Regulación hacia Abajo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Pronóstico , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Tolerancia a Radiación
18.
Mol Cancer Ther ; 20(9): 1672-1679, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34158343

RESUMEN

AZD0530, a potent small-molecule inhibitor of the Src kinase family, is an anticancer drug used in the treatment of various cancers. In the case of glioblastoma (GBM), where resistance to radiotherapy frequently occurs, Src kinase is known as one of the molecules responsible for imparting radioresistance to GBM. Thus, we evaluated the effect of AZD0530 on the radiosensitivity of human GBM cells and human glioblastoma stem-like cells (GSCs). We show that Src activity of GBM and GSC is increased by radiation and inhibited by AZD0530, and using clonogenic assays, AZD0530 enhances the radiosensitivity of GBM and GSCs. Also, AZD0530 induced a prolongation of radiation-induced γH2AX without specific cell cycle and mitotic index changes, suggesting that AZD0530-induced radiosensitization in GBM cells and GSCs results from the inhibition of DNA repair. In addition, AZD0530 was shown to inhibit the radiation-induced EGFR/PI3K/AKT pathway, which is known to promote and regulate radioresistance and survival of GBM cells by radiation. Finally, mice bearing orthotopic xenografts initiated from GBM cells were then used to evaluate the in vivo response to AZD0530 and radiation. The combination of AZD0530 and radiation showed the longest median survival compared with any single modality. Thus, these results show that AZD0530 enhances the radiosensitivity of GBM cells and GSCs and suggest the possibility of AZD0530 as a clinical radiosensitizer for treatment of GBM.


Asunto(s)
Benzodioxoles/farmacología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/radioterapia , Células Madre Neoplásicas/efectos de la radiación , Quinazolinas/farmacología , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Antineoplásicos/farmacología , Apoptosis , Ciclo Celular , Proliferación Celular , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Mol Sci ; 22(8)2021 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33924205

RESUMEN

Cancer stem cells (CSCs) play an important role in cancer recurrence and metastasis. It is suggested that the CSC properties in heterogeneous cancer cells can be induced by ionizing radiation (IR). This study investigated the role of DLX2 in the radioresistance and CSC properties induced by IR in NSCLC cancer cells. Here, A549 cells were exposed to fractionated irradiation at a cumulative dose of 52 Gy (4 Gy × 13 times) for a generation of radioresistant cells. After fractionated irradiation, surviving A549 cells exhibited resistance to IR and enhanced expression of various cancer stem cell markers. They also showed upregulation of mesenchymal molecular markers and downregulation of epithelial molecular markers, correlating with an increase in the migration and invasion. Fractionated irradiation triggered the secretion of TGF-ß1 and DLX2 expression. Interestingly, the increased DLX2 following fractionated irradiation seemed to induce the expression of the gene for the EGFR-ligand betacellulin via Smad2/3 signaling. To contrast, DLX2 knockdown dramatically decreased the expression of CSC markers, migration, and proliferation. Moreover, A549 cells expressing DLX2 shRNA formed tumors with a significantly smaller volume compared to those expressing control shDNA in a mouse xenograft assay. These results suggest that DLX2 overexpression in surviving NSCLC cancer cells after fractionated IR exposure is involved in the cancer stemness, radioresistance, EMT, tumor survival, and tumorigenic capability.


Asunto(s)
Autorrenovación de las Células/efectos de la radiación , Rayos gamma , Proteínas de Homeodominio/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Transducción de Señal/efectos de la radiación , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células A549 , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de la radiación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Técnicas de Inactivación de Genes , Humanos , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Tolerancia a Radiación , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Int J Mol Sci ; 22(8)2021 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-33921230

RESUMEN

Hepatocellular carcinoma (HCC) is a major histological subtype of primary liver cancer. Ample evidence suggests that the pathological properties of HCC originate from hepatic cancer stem cells (CSCs), which are responsible for carcinogenesis, recurrence, and drug resistance. Cold atmospheric-pressure plasma (CAP) and plasma-activated medium (PAM) induce apoptosis in cancer cells and represent novel and powerful anti-cancer agents. This study aimed to determine the anti-cancer effect of CAP and PAM in HCC cell lines with CSC characteristics. We showed that the air-based CAP and PAM selectively induced cell death in Hep3B and Huh7 cells with CSC characteristics, but not in the normal liver cell line, MIHA. We observed both caspase-dependent and -independent cell death in the PAM-treated HCC cell lines. Moreover, we determined whether combinatorial PAM therapy with various anti-cancer agents have an additive effect on cell death in Huh7. We found that PAM highly increased the efficacy of the chemotherapeutic agent, cisplatin, while enhanced the anti-cancer effect of doxorubicin and the targeted-therapy drugs, trametinib and sorafenib to a lesser extent. These findings support the application of CAP and PAM as anti-cancer agents to induce selective cell death in cancers containing CSCs, suggesting that the combinatorial use of PAM and some specific anti-cancer agents is complemented mechanistically.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Medios de Cultivo/efectos de la radiación , Neoplasias Hepáticas/tratamiento farmacológico , Gases em Plasma , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Línea Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Medios de Cultivo/farmacología , Doxorrubicina/farmacología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...