Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Mol Carcinog ; 61(8): 812-824, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35652616

RESUMEN

Radiotherapy (RT) is a conventional cancer therapeutic modality. However, cancer cells tend to develop radioresistance after a period of treatment. Diagnostic markers and therapeutic targets for radiosensitivity are severely lacking. Our recently published studies demonstrated that the cell division cycle (CDC6) is a critical molecule contributing to radioresistance, and maybe a potential therapeutic target to overcome radioresistance. In the present study, we for the first time reported that Norcantharidin (NCTD), a demethylated form of cantharidin, re-sensitized radioresistant cancer cells to overcome radioresistance, and synergistically promoted irradiation (IR)-induced cell killing and apoptosis by inducing CDC6 protein degradation. Mechanistically, NCTD induced CDC6 protein degradation through the ubiquitin-proteasome pathways. By using small interfering RNA (siRNA) interference or small compound inhibitors, we further determined that NCTD induced CDC6 protein degradation through a neddylation-dependent pathway, but not through Huwe1, Cyclin F, and APC/C-mediated ubiquitin-proteasome pathways. We screened the six most relevant Cullin subunits (CUL1, 2, 3, 4A, 4B, and 5) using siRNAs. The knockdown of Cullin1 but not the other five cullins remarkably elevated CDC6 protein levels. NCTD promoted the binding of Cullin1 to CDC6, thereby promoting CDC6 protein degradation through a Cullin1 neddylation-mediated ubiquitin-proteasome pathway. NCTD can be used in combination with radiotherapy to achieve better anticancer efficacy, or work as a radiosensitizer to overcome cancer radioresistance.


Asunto(s)
Proteínas de Ciclo Celular , Neoplasias , Apoptosis , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteínas de Ciclo Celular/metabolismo , Proteínas Cullin , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , ARN Interferente Pequeño/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/metabolismo
2.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(7): 721-8, 2016 Jul.
Artículo en Zh | MEDLINE | ID: mdl-27592578

RESUMEN

OBJECTIVE: To determine the vaginal flora distribution in cervical cancer patients and the common pathogenic bacteria as well as drug resistance, and to explore the correlation of vaginal bacterial infection and high-risk human papillomavirus (HR-HPV) infection with cervical cancer.
 METHODS: A total of 216 patients with cervical cancer served as an experimental group, and 53 patients with chronic cervicitis served as a control group. The patients' vaginal fluid in two groups was collected before the treatment for regular bacterial culture and HPV testing. The distribution and drug resistance of bacteria in two groups of vaginal secretion were observed, and the correlation of the bacterial infection and HPV infection with the cervical cancer was analyzed.
 RESULTS: The gram-negative and gram-positive bacteria accounted for 74.38% and 21.49% in the experimental group, respectively. They were mainly resistant to ampicillin and piperacillin or penicillin and erythromycin. The gram-negative and gram-positive bacteria accounted for 42.31% and 23.08% in the control group, respectively. They were mainly resistant to ampicillin and piperacillin or penicillin. HPV-positive rates in the experiment group and the control group were 60.65% and 41.51%, respectively. There were 70 patients (32.41%) and 12 patients (22.64%) with both bacterial infection and HPV-positive infection in the experiment group and the control group, respectively. However, there was no statistical difference between the 2 groups (P>0.05). 
 CONCLUSION: Escherichia coli are the main pathogen in cervical cancer and they are highly resistant to antibiotics. Bacterial infection in genital tract is not an efficient cofactor for HPV to cause the cervical cancer.


Asunto(s)
Farmacorresistencia Bacteriana , Infecciones del Sistema Genital , Neoplasias del Cuello Uterino , Femenino , Infecciones por Bacterias Gramnegativas , Bacterias Grampositivas , Infecciones por Bacterias Grampositivas , Humanos
3.
Heliyon ; 10(17): e37066, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39296204

RESUMEN

Objective: This study explores the correlation between Forkhead box M1 (FOXM1) and ATP-binding cassette subfamily C member 5 (ABCC5) in relation to paclitaxel resistance in cervical cancer. It aims to identify potential cervical cancer stem cell markers, offering fresh perspectives for developing therapeutic strategies to overcome paclitaxel chemoresistance in cervical cancer. Methods: Paclitaxel-resistant Hela cells (Hela/Taxol) were developed by intermittently exposing Hela cells to progressively increasing concentrations of paclitaxel. We assessed the biological properties of both Hela and Hela/Taxol cells using various assays: cell proliferation, clonogenic, cell cycle, apoptosis, scratch, and transwell. To determine which markers better represent tumor stem cells, we analyzed various known and potential stem cell markers in combination. Flow cytometry was employed to measure the proportion of positive markers in both parental and drug-resistant cell lines. Following statistical analysis to establish relative stability, CD133+ABCC5+ cells were sorted for further examination. Subsequent tests included sphere-forming assays and Western blot analysis to detect the presence of the stem cell-specific protein Sox2, aiding in the identification of viable cervical cancer stem cell markers. Results: The Hela/Taxol cell line exhibited significantly enhanced proliferation, migration, and invasion capabilities compared to the Hela cell line, alongside a marked reduction in apoptosis rates (P < 0.01). Notably, proportions of CD44+, CD24+CD44+, ABCC5+, CD24+CD44+ABCC5+, CD44+ABCC5+, CD24+CD44+FOXM1+, CD44+FOXM1+, CD133+ABCC5+, and CD133+FOXM1+ were significantly higher (P < 0.05). Furthermore, the size and number of spheres formed byCD133+ABCC5+ cells were greater in the sorted Hela/Taxol line (P < 0.01), with increased expression of the stem cell marker Sox2 (P < 0.001). Conclusion: The Hela/Taxol cells demonstrate increased tumoral stemness, suggesting that CD133+ABCC5+ may serve as a novel marker for cervical cancer stem cells.

4.
Bioengineered ; 13(3): 6100-6112, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35192432

RESUMEN

Previous studies suggest that SRPK1 (serine/arginine-rich protein-specific kinase 1) is involved in tumorigenesis and closely related to unfavorable outcomes. However, its expression pattern in cervical squamous cell carcinoma (CESC) remains uncovered. In this study, we initially investigated the clinical significance and function of SRPK1 in human CESC. Data mining and analysis on SRPK1 mRNA expression in CESC samples were conducted using TCGA database, which indicated that SRPK1 mRNA was significantly upregulated in CESC samples. Protein expression of SRPK1 was tested by immunohistochemistry in a retrospective cohort (n = 122), revealing a higher SRPK1 protein abundance in CESC specimens whose aberrant up-regulation was obviously related to worse survival. Cox proportional hazards regression analysis further confirmed the role of SRPK1 as an independent prognostic factor of CESC. Cellular experiments validated that SRPK1 may function through enhancing CESC proliferation, migration, and invasion. In conclusion, aberrant up-regulation of SRPK1 is remarkably related to progression and unfavorable prognosis of CESC, which can serve as a novel prognostic biomarker and therapeutic target for CESC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias del Cuello Uterino , Femenino , Humanos , Arginina , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Pronóstico , Proteínas Serina-Treonina Quinasas/genética , Estudios Retrospectivos , ARN Mensajero/genética , Serina , Neoplasias del Cuello Uterino/metabolismo
5.
Biomed Res Int ; 2022: 3032590, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35141332

RESUMEN

OBJECTIVE: The aim of the present study was to investigate the effect of forkhead box M1 (FOXM1) to paclitaxel resistance in cervical cancer cells, to determine the underlying mechanism, and to identify novel targets for the treatment of paclitaxel-resistant cervical cancer. METHODS: Paclitaxel-resistant Caski cells (Caski/Taxol cells) were established by intermittently exposing the Caski cells to gradually increasing concentrations of paclitaxel. The association between FOXM1, ATP-binding cassette subfamily C member 5 (ABCC5), and cervical cancer cell drug resistance was assessed by overexpressing or knocking down the expression of FOXM1 in Caski or Caski/Taxol cells. The protein and mRNA expression levels, the ratio of cellular apoptosis, and cell migration as well as intracellular drug concentrations were measured in cells following the different treatments. RESULTS: After the successful establishment of resistant Caski/Taxol cells, cell cycle distribution analysis showed that a significantly larger percentage of Caski/Taxol cells was in the G0/G1 stage compared with the Caski cells (P < 0.01), whereas a significantly larger percentage of Caski cells was in the S and G2/M stage compared with the Caski/Taxol cells following treatment with paclitaxel (P < 0.01). Both the protein and mRNA expression levels of FOXM1 and ABCC5 transporters were significantly higher in the paclitaxel-resistant Caski/Taxol cells compared with Caski cells (P < 0.05). Knockdown of FOXM1 significantly lowered the protein expression levels of FOXM1 and ABCC5. Intracellular paclitaxel concentrations were significantly higher amongst the Caski/Taxol cells following the knockdown of FOXM1 by shRNA or Siomycin A (P < 0.05). CONCLUSION: FOXM1 promotes drug resistance in cervical cancer cells by regulating ABCC5 gene transcription. The knockdown of FOXM1 with shRNA or Siomycin A promotes paclitaxel-induced cell death by regulating ABCC5 gene transcription.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Resistencia a Antineoplásicos/genética , Proteína Forkhead Box M1/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Neoplasias del Cuello Uterino/genética , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Paclitaxel/farmacología , Transcripción Genética/efectos de los fármacos
6.
Cell Death Dis ; 8(3): e2659, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28277541

RESUMEN

Paclitaxel is clinically used as a first-line chemotherapeutic regimen for several cancer types, including head and neck cancers. However, acquired drug resistance results in the failure of therapy, metastasis and relapse. The drug efflux mediated by ATP-binding cassette (ABC) transporters and the survival signals activated by forkhead box (FOX) molecules are critical in the development of paclitaxel drug resistance. Whether FOX molecules promote paclitaxel resistance through drug efflux remains unknown. In this study, we developed several types of paclitaxel-resistant (TR) nasopharyngeal carcinoma (NPC) cells. These TR NPC cells acquired cancer stem cell (CSC) phenotypes and underwent epithelial to mesenchymal transition (EMT), and developed multidrug resistance. TR cells exhibited stronger drug efflux than parental NPC cells, leading to the reduction of intracellular drug concentrations and drug insensitivity. After screening the gene expression of ABC transporters and FOX molecules, we found that FOXM1 and ABCC5 were consistently overexpressed in the TR NPC cells and in patient tumor tissues. Further studies demonstrated that FOXM1 regulated abcc5 gene transcription by binding to the FHK consensus motifs at the promoter. The depletion of FOXM1 or ABCC5 with siRNA significantly blocked drug efflux and increased the intracellular concentrations of paclitaxel, thereby promoting paclitaxel-induced cell death. Siomycin A, a FOXM1 inhibitor, significantly enhanced in vitro cell killing by paclitaxel in drug-resistant NPC cells. This study is the first to identify the roles of FOXM1 in drug efflux and paclitaxel resistance by regulating the gene transcription of abcc5, one of the ABC transporters. Small molecular inhibitors of FOXM1 or ABCC5 have the potential to overcome paclitaxel chemoresistance in NPC patients.


Asunto(s)
Carcinoma/tratamiento farmacológico , Proteína Forkhead Box M1/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Neoplasias Nasofaríngeas/tratamiento farmacológico , Paclitaxel/administración & dosificación , Carcinoma/genética , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA