Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres











Gamme d'année
1.
Cell Commun Signal ; 22(1): 145, 2024 02 22.
Article de Anglais | MEDLINE | ID: mdl-38388432

RÉSUMÉ

BACKGROUND: ZEB1, a core transcription factor involved in epithelial-mesenchymal transition (EMT), is associated with aggressive cancer cell behavior, treatment resistance, and poor prognosis across various tumor types. Similarly, the expression and activity of CD73, an ectonucleotidase implicated in adenosine generation, is an important marker of tumor malignancy. Growing evidence suggests that EMT and the adenosinergic pathway are intricately linked and play a pivotal role in cancer development. Therefore, this study focuses on exploring the correlations between CD73 and ZEB1, considering their impact on tumor progression. METHODS: We employed CRISPR/Cas9 technology to silence CD73 expression in cell lines derived from papillary thyroid carcinoma. These same cells underwent lentiviral transduction of a reporter of ZEB1 non-coding RNA regulation. We conducted studies on cell migration using scratch assays and analyses of cellular speed and polarity. Additionally, we examined ZEB1 reporter expression through flow cytometry and immunocytochemistry, complemented by Western blot analysis for protein quantification. For further insights, we applied gene signatures representing different EMT states in an RNA-seq expression analysis of papillary thyroid carcinoma samples from The Cancer Genome Atlas. RESULTS: Silencing CD73 expression led to a reduction in ZEB1 non-coding RNA regulation reporter expression in a papillary thyroid carcinoma-derived cell line. Additionally, it also mitigated ZEB1 protein expression. Moreover, the expression of CD73 and ZEB1 was correlated with alterations in cell morphology characteristics crucial for cell migration, promoting an increase in cell polarity index and cell migration speed. RNA-seq analysis revealed higher expression of NT5E (CD73) in samples with BRAF mutations, accompanied by a prevalence of partial-EMT/hybrid state signature expression. CONCLUSIONS: Collectively, our findings suggest an association between CD73 expression and/or activity and the post-transcriptional regulation of ZEB1 by non-coding RNA, indicating a reduction in its absence. Further investigations are warranted to elucidate the relationship between CD73 and ZEB1, with the potential for targeting them as therapeutic alternatives for cancer treatment in the near future.


Sujet(s)
Tumeurs de la thyroïde , Facteurs de transcription , Humains , Cancer papillaire de la thyroïde , Lignée cellulaire tumorale , Facteurs de transcription/génétique , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/anatomopathologie , ARN non traduit , Facteur de transcription Zeb1/génétique
2.
Braz J Otorhinolaryngol ; 90(1): 101358, 2024.
Article de Anglais | MEDLINE | ID: mdl-37989078

RÉSUMÉ

OBJECTIVES: Nasopharyngeal carcinoma (NPC) is an aggressive epithelial cancer. The expression of miR-186 is decreased in a variety of malignancies and can promote the invasion and metastasis of cancer cells. This study aimed to explore the role and possible mechanism of miR-186 in the metastasis and epithelial-mesenchymal transformation (EMT) of NPC. METHODS: The expression of miR-186 in NPC tissues and cells was detected by RT-PCR. Then, miR-186 mimic was used to transfect NPC cell lines C666-1 and CNE-2, and cell activity, invasion and migration were detected by CCK8, transwell and scratch assay, respectively. The expression of EMT-related proteins was analyzed by western blotting analysis. The binding relationship between miR-186 and target gene Zinc Finger E-Box Binding Homeobox 1 (ZEB1) was confirmed by double luciferase assay. RESULTS: The expression of miR-186 in NPC was significantly decreased, and transfection of miR-186 mimic could significantly inhibit the cell activity, invasion, and migration, and regulate the protein expressions of E-cadherin, N-cadherin and vimentin in C666-1 and CNE-2 cells. Further experiments confirmed that miR-186 could directly target ZEB1 and negatively regulate its expression. In addition, ZEB1 has been confirmed to be highly expressed in NPC, and inhibition of ZEB1 could inhibit the activity, invasion, metastasis and EMT of NPC cells. And co-transfection of miR-186 mimic and si-ZEB1 could further inhibit the proliferation and metastasis of NPC. CONCLUSION: miR-186 may inhibit the proliferation, metastasis and EMT of NPC by targeting ZEB1, and the miR-186/ZEB1 axis plays an important role in NPC.


Sujet(s)
Carcinomes , microARN , Tumeurs du rhinopharynx , Humains , Cancer du nasopharynx/anatomopathologie , microARN/génétique , microARN/métabolisme , Lignée cellulaire tumorale , Carcinomes/génétique , Carcinomes/anatomopathologie , Transition épithélio-mésenchymateuse/génétique , Tumeurs du rhinopharynx/génétique , Tumeurs du rhinopharynx/anatomopathologie , Régulation de l'expression des gènes tumoraux/génétique , Prolifération cellulaire , Invasion tumorale/génétique , Facteur de transcription Zeb1/génétique , Facteur de transcription Zeb1/métabolisme
3.
Braz. j. otorhinolaryngol. (Impr.) ; Braz. j. otorhinolaryngol. (Impr.);90(1): 101358, 2024. graf
Article de Anglais | LILACS-Express | LILACS | ID: biblio-1534080

RÉSUMÉ

Abstract Objectives Nasopharyngeal carcinoma (NPC) is an aggressive epithelial cancer. The expression of miR-186 is decreased in a variety of malignancies and can promote the invasion and metastasis of cancer cells. This study aimed to explore the role and possible mechanism of miR-186 in the metastasis and epithelial-mesenchymal transformation (EMT) of NPC. Methods The expression of miR-186 in NPC tissues and cells was detected by RT-PCR. Then, miR-186 mimic was used to transfect NPC cell lines C666-1 and CNE-2, and cell activity, invasion and migration were detected by CCK8, transwell and scratch assay, respectively. The expression of EMT-related proteins was analyzed by western blotting analysis. The binding relationship between miR-186 and target gene Zinc Finger E-Box Binding Homeobox 1 (ZEB1) was confirmed by double luciferase assay. Results The expression of miR-186 in NPC was significantly decreased, and transfection of miR-186 mimic could significantly inhibit the cell activity, invasion, and migration, and regulate the protein expressions of E-cadherin, N-cadherin and vimentin in C666-1 and CNE-2 cells. Further experiments confirmed that miR-186 could directly target ZEB1 and negatively regulate its expression. In addition, ZEB1 has been confirmed to be highly expressed in NPC, and inhibition of ZEB1 could inhibit the activity, invasion, metastasis and EMT of NPC cells. And co-transfection of miR-186 mimic and si-ZEB1 could further inhibit the proliferation and metastasis of NPC. Conclusion miR-186 may inhibit the proliferation, metastasis and EMT of NPC by targeting ZEB1, and the miR-186/ZEB1 axis plays an important role in NPC.

4.
Int J Mol Sci ; 24(6)2023 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-36982993

RÉSUMÉ

Cancer biologists have focused on studying cancer stem cells (CSCs) because of their ability to self-renew and recapitulate tumor heterogeneity, which increases their resistance to chemotherapy and is associated with cancer relapse. Here, we used two approaches to isolate CSCs: the first involved the metabolic enzyme aldehyde dehydrogenase ALDH, and the second involved the three cell surface markers CD44, CD117, and CD133. ALDH cells showed a higher zinc finger E-box binding homeobox 1 (ZEB1) microRNA (miRNA) expression than CD44/CD117/133 triple-positive cells, which overexpressed miRNA 200c-3p: a well-known microRNA ZEB1 inhibitor. We found that ZEB1 inhibition was driven by miR-101-3p, miR-139-5p, miR-144-3p, miR-199b-5p, and miR-200c-3p and that the FaDu Cell Line inhibition occurred at the mRNA level, whereas HN13 did not affect mRNA expression but decreased protein levels. Furthermore, we demonstrated the ability of the ZEB1 inhibitor miRNAs to modulate CSC-related genes, such as TrkB, ALDH, NANOG, and HIF1A, using transfection technology. We showed that ALDH was upregulated upon ZEB1-suppressed miRNA transfection (Mann-Whitney ** p101 = 0.009, t-test ** p139 = 0.009, t-test ** p144 = 0.002, and t-test *** p199 = 0.0006). Overall, our study enabled an improved understanding of the role of ZEB1-suppressed miRNAs in CSC biology.


Sujet(s)
Tumeurs de la tête et du cou , microARN , Humains , microARN/métabolisme , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux , Récidive tumorale locale/génétique , Facteur de transcription Zeb1/génétique , Facteur de transcription Zeb1/métabolisme , Tumeurs de la tête et du cou/génétique , Cellules souches tumorales/métabolisme , ARN messager/génétique , Mouvement cellulaire/génétique , Prolifération cellulaire
5.
Oral Dis ; 29(5): 1991-2005, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-35403781

RÉSUMÉ

Chemoresistance is associated with tumor recurrence, metastases, and short survival. Cisplatin is one of the most used chemotherapies in cancer treatment, including head and neck squamous cell carcinoma (HNSCC), and many patients develop resistance. Here, we established cell lines resistant to cisplatin to better understand epigenetics and biological differences driving the progression of HNSCC after treatment. Cisplatin resistance was established in CAL-27 and SCC-9 cell lines. Gene expression of HDAC1, HDAC2, SIRT1, MTA1, KAT2B, KAT6A, KAT6B, and BRD4 indicated the cisplatin activates the epigenetic machinery. Increases in tumor aggressiveness were detected by BMI-1 and KI-67 in more resistant cell lines. Changes in cellular shape and epithelial-mesenchymal transition (EMT) activation were also observed. HDAC1 and ZEB1 presented an opposite distribution with down-regulation of HDAC1 and up-regulation of ZEB1 in the course of chemoresistance. Up-regulation of ZEB1 and BMI-1 in patients with HNSCC is also associated with a poor response to therapy. Cancer stem cells (CSC) population increased significantly with chemoresistance. Down-regulation of HDAC1, HDAC2, and SIRT1 and accumulation of Vimentin and ZEB1 were observed in the CSC population. Our results suggest that in the route to cisplatin chemoresistance, epigenetic modifications can be associated with EMT activation and CSC accumulation which originate more aggressive tumors.


Sujet(s)
Carcinome épidermoïde , Tumeurs de la tête et du cou , Humains , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Cisplatine/pharmacologie , Sirtuine-1/génétique , Sirtuine-1/métabolisme , Sirtuine-1/usage thérapeutique , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/génétique , Carcinome épidermoïde/métabolisme , Transition épithélio-mésenchymateuse/génétique , Protéines nucléaires/métabolisme , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Récidive tumorale locale/anatomopathologie , Cellules souches tumorales/métabolisme , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Régulation de l'expression des gènes tumoraux , Histone acetyltransferases/métabolisme
6.
Neuromolecular Med ; 25(1): 64-74, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-35716340

RÉSUMÉ

Medulloblastoma (MB) is a malignant brain tumor that afflicts mostly children and adolescents and presents four distinct molecular subgroups, known as WNT, SHH, Group 3, and Group 4. ZEB1 is a transcription factor that promotes the expression of mesenchymal markers while restraining expression of epithelial and polarity genes. Because of ZEB1 involvement in cerebellum development, here we investigated the role of ZEB1 in MB. We found increased expression of ZEB1 in MB tumor samples compared to normal cerebellar tissue. Expression was higher in the SHH subgroup when compared to all other MB molecular subgroups. High ZEB1 expression was associated with poor prognosis in Group 3 and Group 4, whereas in patients with WNT tumors poorer prognosis were related to lower ZEB1 expression. There was a moderate correlation between ZEB1 and MYC expression in Group 3 and Group 4 MB. Treatment with the immunomodulator and histone deacetylase (HDAC) inhibitor fingolimod (FTY720) reduced ZEB1 expression specifically in D283 cells, which are representative of Group 3 and Group 4 MB. These findings reveal novel subgroup-specific associations of ZEB1 expression with survival in patients with MB and suggest that ZEB1 expression can be reduced by pharmacological agents that target HDAC activity.


Sujet(s)
Tumeurs du cerveau , Tumeurs du cervelet , Médulloblastome , Enfant , Adolescent , Humains , Médulloblastome/traitement médicamenteux , Médulloblastome/génétique , Cervelet , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Chlorhydrate de fingolimod/usage thérapeutique , Tumeurs du cervelet/traitement médicamenteux , Tumeurs du cervelet/génétique , Facteur de transcription Zeb1/génétique , Facteur de transcription Zeb1/métabolisme
7.
Clin Transl Oncol ; 25(3): 817-829, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36418641

RÉSUMÉ

BACKGROUND: Lung squamous cell carcinoma (LUSC) is recognized as the major subtypes of non-small cell lung cancer (NSCLC). Circulating tumor cells (CTCs) are critical players in tumor metastasis. A molecular profiling of CTCs has previously identified notch receptor 1 (Notch1) as an important mediator in NSCLC. Therefore, we investigate Notch1 roles in LUSC and its related mechanisms. METHODS: The serum levels of Notch1 were measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The CTCs isolated from blood samples were characterized via an immunofluorescence method. Cell motion was determined using Transwell chambers. The regulatory relationship between Notch1 and zinc finger E-box-binding homeobox 1 (ZEB1) was verified by chromatin immunoprecipitation (ChIP) and luciferase reporter assays. The protein levels were detected by western blotting. RESULTS: Higher Notch1 expression in patients with LUSC than that in normal controls was observed. Notch1 knockdown inhibited cell motion and epithelial-mesenchymal transition (EMT). ZEB1 transcriptionally activated Notch1. ZEB1 upregulation exacerbated the malignant phenotypes of CTCs. CONCLUSION: ZEB1-activated Notch1 promotes malignant phenotypes of CTCs in LUSC and indicates poor prognosis.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Carcinome épidermoïde , Tumeurs du poumon , microARN , Cellules tumorales circulantes , Humains , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/génétique , Carcinome épidermoïde/anatomopathologie , Poumon , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/génétique , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Invasion tumorale/génétique , microARN/métabolisme , Facteur de transcription Zeb1/génétique , Récepteur Notch1
8.
Clinics (Sao Paulo) ; 77: 100089, 2022.
Article de Anglais | MEDLINE | ID: mdl-35905576

RÉSUMÉ

OBJECTIVE: This study aimed to investigate the effect of Zinc Finger E-box Binding Homeobox 1 (ZEB1) regulation by Micro Ribonucleic acid (miR)-448 on Breast Cancer (BC) cells and their sensitivity to chemotherapy. METHODS: miR-448 and ZEB1 mRNA levels in BC and normal tissues were detected by qPCR, and ZEB1 protein was detected by Western Blotting (WB). The correlation between miR-448 and tumor metastasis, clinical staging, and ZEB1 expression was analyzed. MCF-7 cells were transfected or co-transfected with the miR-448 mimic, oe-ZEB1, or their negative controls. Changes in miR-448 and ZEB1 expression were detected by qPCR and WB. Cell proliferation was determined by CCK-8 assays, invasion changes were analyzed by Transwell assays, and apoptosis was detected by flow cytometry. RESULTS: miR-448 expression in BC tissues was lower than that in normal tissues, while ZEB1 expression was increased in the former. ZEB1 expression was lower in BC patients with lymph node metastasis than in those without. In patients with clinical stage I-III BC, miR-448 expression decreased with an increase in tumor stage, which was negatively correlated with ZEB1 expression. Upregulation of miR-448 expression can suppress MCF-7 cell proliferation and invasion and promote apoptosis. Upregulation of ZEB1 expression in cells overexpressing miR-448 can partially reverse the inhibition of BC cell growth induced by miR-448. miR-448 can enhance the sensitivity of cells toward paclitaxel and 5-fluorouracil. CONCLUSIONS: miR-448 suppresses cell proliferation and invasion and promotes apoptosis by targeting ZEB1. Moreover, it can increase the sensitivity of cells toward paclitaxel and 5-fluorouracil.


Sujet(s)
Tumeurs du sein , microARN , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Femelle , Fluorouracil , Régulation de l'expression des gènes tumoraux , Gènes homéotiques , Humains , Invasion tumorale , Paclitaxel , Facteur de transcription Zeb1 , Doigts de zinc
9.
Adv Exp Med Biol ; 1393: 51-64, 2022.
Article de Anglais | MEDLINE | ID: mdl-36587301

RÉSUMÉ

Prostate cancer (PCa) incidence has increased during the last decades, becoming one of the leading causes of death by cancer in men worldwide. During an extended period of prostate cancer, malignant cells are androgen-sensitive being testosterone the main responsible for tumor growth. Accordingly, treatments blocking production and action of testosterone are mostly used. However, during disease progression, PCa cells become androgen insensitive producing a castration-resistant stage with a worse prognosis. Overcoming castration-resistant prostate cancer (CRPC) has become a great challenge in the management of this disease. In the search for molecular pathways leading to therapy resistance, the epithelial-mesenchymal transition (EMT), and particularly the transcription factors zinc finger E-box-binding homeobox 1 (Zeb1) and zinc finger protein SNAI1 (Snail), master genes of the EMT, have shown to have pivotal roles. Also, the discovery that cancer stem cells (CSCs) can be generated de novo from their non-CSCs counterpart has led to the question whereas these EMT transcription factors could be implicated in this dynamic conversion between non-CSC and CSC. In this review, we analyze evidence supporting the idea that Zeb1 and Snail induce cell malignancy and cancer stem cell phenotype in prostate cells, increasing androgen synthesis capacity and therapy resistance.


Sujet(s)
Tumeurs de la prostate , Facteurs de transcription de la famille Snail , Facteur de transcription Zeb1 , Humains , Mâle , Androgènes/métabolisme , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse , Cellules souches tumorales/anatomopathologie , Phénotype , Prostate/métabolisme , Prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Testostérone/métabolisme , Facteur de transcription Zeb1/génétique , Facteur de transcription Zeb1/métabolisme , Facteurs de transcription de la famille Snail/métabolisme
10.
Clinics ; Clinics;77: 100089, 2022. graf
Article de Anglais | LILACS-Express | LILACS | ID: biblio-1404328

RÉSUMÉ

Abstract Objective: This study aimed to investigate the effect of Zinc Finger E-box Binding Homeobox 1 (ZEB1) regulation by Micro Ribonucleic acid (miR)-448 on Breast Cancer (BC) cells and their sensitivity to chemotherapy. Methods: miR-448 and ZEB1 mRNA levels in BC and normal tissues were detected by qPCR, and ZEB1 protein was detected by Western Blotting (WB). The correlation between miR-448 and tumor metastasis, clinical staging, and ZEB1 expression was analyzed. MCF-7 cells were transfected or co-transfected with the miR-448 mimic, oe-ZEB1, or their negative controls. Changes in miR-448 and ZEB1 expression were detected by qPCR and WB. Cell proliferation was determined by CCK-8 assays, invasion changes were analyzed by Transwell assays, and apoptosis was detected by flow cytometry. Results: miR-448 expression in BC tissues was lower than that in normal tissues, while ZEB1 expression was increased in the former. ZEB1 expression was lower in BC patients with lymph node metastasis than in those without. In patients with clinical stage I-III BC, miR-448 expression decreased with an increase in tumor stage, which was negatively correlated with ZEB1 expression. Upregulation of miR-448 expression can suppress MCF-7 cell proliferation and invasion and promote apoptosis. Upregulation of ZEB1 expression in cells overexpressing miR-448 can partially reverse the inhibition of BC cell growth induced by miR-448. miR-448 can enhance the sensitivity of cells toward paclitaxel and 5-fluorouracil. Conclusions: miR-448 suppresses cell proliferation and invasion and promotes apoptosis by targeting ZEB1. Moreover, it can increase the sensitivity of cells toward paclitaxel and 5-fluorouracil.

11.
Mol Cell Biochem ; 476(11): 4107-4116, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34292482

RÉSUMÉ

The transcription factor Zinc finger E-box binding 1 (ZEB1) displays a range of regulatory activities in cell function and embryonic development, including driving epithelial-mesenchymal transition. Several aspects of ZEB1 function can be regulated by its functional interactions with noncoding RNA types, namely microRNAs (miRNAs) and long noncoding RNAs (lncRNAs). Increasing evidence indicates that ZEB1 importantly influences cancer initiation, tumor progression, metastasis, and resistance to treatment. Cancer is the main disease-related cause of death in children and adolescents. Although the role of ZEB1 in pediatric cancer is still poorly understood, emerging findings have shown that it is expressed and regulates childhood solid tumors including osteosarcoma, retinoblastoma, neuroblastoma, and central nervous system tumors. Here, we review the evidence supporting a role for ZEB1, and its interplays with miRNAs and lncRNAs, in pediatric cancers.


Sujet(s)
microARN/génétique , Tumeurs/génétique , Tumeurs/métabolisme , ARN long non codant/génétique , Facteur de transcription Zeb1/métabolisme , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Carcinogenèse , Enfant , Transition épithélio-mésenchymateuse , Humains , Tumeurs/anatomopathologie , Neuroblastome/génétique , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Ostéosarcome/anatomopathologie , Tumeurs de la rétine/génétique , Tumeurs de la rétine/métabolisme , Tumeurs de la rétine/anatomopathologie , Rétinoblastome/génétique , Rétinoblastome/métabolisme , Rétinoblastome/anatomopathologie , Facteur de transcription Zeb1/génétique
12.
Clin Transl Oncol ; 23(3): 491-500, 2021 Mar.
Article de Anglais | MEDLINE | ID: mdl-32613412

RÉSUMÉ

PURPOSE: This study set out to probe into the effect and mechanism of miR-144-3p on radiosensitivity of gastric cancer (GC) cells. METHODS: Cancer tissue and paracancerous tissue of GC patients admitted to our hospital were collected, their miR-144-3p expression was tested, GC cells were transfected, and survival and biological behavior of those cells under radiation were detected. RESULTS: After detection, miR-144-3p expression was down-regulated in GC tissue, while ZEB1 was up-regulated. There was no remarkable difference in the survival fraction of cells in each group before receiving radiation, but that of tumor cells decreased obviously (p < 0.05) after radiation exposure. Survival fraction of cells overexpressing miR-144-3p or silencing ZEB1 decreased more obviously, while the inhibition of miR-144-3p or overexpressing ZEB1 was weaker. Biological behavior of cells under 6 Gy radiation was detected. It was found that miR-144-3p overexpression or silencing ZEB1 dramatically inhibited the proliferation activity of GC cells under 6 Gy radiation, increased the levels of pro-apoptotic Bax and caspase-3 proteins (p < 0.05) and decreased the anti-apoptotic protein Bcl-2 level (p < 0.05), resulting in an increase in the apoptosis rate of cells. miR-144-3p was confirmed to be ZEB1 targeting site by dual luciferase report. Moreover, rescue experiments prove that it can increase the radiosensitivity of GC cells by regulating ZEB1 expression. CONCLUSION: miR-144-3p expression was down-regulated in GC, and it can increase the radiosensitivity of those cells by inhibiting ZEB1 expression.


Sujet(s)
microARN/métabolisme , Radiotolérance , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/radiothérapie , Facteur de transcription Zeb1/métabolisme , Apoptose , Caspase-3/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des radiations , Survie cellulaire , Régulation négative , Femelle , Muqueuse gastrique/métabolisme , Extinction de l'expression des gènes , Humains , Mâle , Adulte d'âge moyen , Protéines proto-oncogènes c-bcl-2/métabolisme , Dose de rayonnement , Transfection/méthodes , Régulation positive , Facteur de transcription Zeb1/génétique , Protéine Bax/métabolisme
13.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;54(5): e9700, 2021. tab, graf
Article de Anglais | LILACS | ID: biblio-1180737

RÉSUMÉ

Lung adenocarcinomas are usually sensitive to radiation therapy, but some develop resistance. Radiation resistance can lead to poor patient prognosis. Studies have shown that lung adenocarcinoma cells (H1299 cells) can develop radioresistance through epithelial-mesenchymal transition (EMT), and this process is regulated by miRNAs. However, it is unclear which miRNAs are involved in the process of EMT. In our present study, we found that miR-183 expression was increased in a radioresistant lung adenocarcinoma cell line (H1299R cells). We then explored the regulatory mechanism of miR-183 and found that it may be involved in the regulation of zinc finger E-box-binding homeobox 1 (ZEB1) expression and mediate EMT in lung adenocarcinoma cells. qPCR results showed that miR-183, ZEB1, and vimentin were highly expressed in H1299R cells, whereas no difference was observed in E-cadherin expression. Western blot results showed that ZEB1 and vimentin were highly expressed in H1299R cells, while E-cadherin expression was decreased. When miR-183 expression was inhibited in H1299R cells, radiation resistance, proliferation, and cell migration were decreased. The expression of ZEB1 and vimentin in H1299R cells was decreased, while the expression of E-cadherin was increased. Moreover, miR-183 overexpression in H1299 cells enhanced radiation resistance, proliferative capacity, and cell migration ability. The expression of ZEB1 and vimentin in H1299 cells was increased, while that of E-cadherin was decreased. In conclusion, miR-183 may promote EMT and radioresistance in H1299 cells, and targeting the miR-183-ZEB1 signaling pathway may be a promising approach for lung cancer treatment.


Sujet(s)
Humains , microARN/génétique , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/radiothérapie , Tumeurs du poumon/génétique , Tumeurs du poumon/radiothérapie , Régulation de l'expression des gènes tumoraux , Mouvement cellulaire , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse
14.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;53(4): e9288, 2020.
Article de Anglais | LILACS-Express | LILACS | ID: biblio-1439698

RÉSUMÉ

Diabetic nephropathy (DN) is one of the leading causes of mortality in diabetic patients. Long non-coding RNA zinc finger E-box binding homeobox 1 antisense 1 (ZEB1-AS1) plays a crucial role in the development of various diseases, including DN. However, the molecular mechanism of ZEB1-AS1 in DN pathogenesis remains elusive. An in vitro DN model was established by treating HK-2 cells with high glucose (HG). Quantitative polymerase chain reaction (qRT-PCR) was utilized to detect the expression levels of ZEB1-AS1, microRNA-216a-5p (miR-216a-5p), and bone morphogenetic protein 7 (BMP7). Western blot assay was used to evaluate the protein levels of BMP7, epithelial-to-mesenchymal transition (EMT)-related proteins, and fibrosis markers. Additionally, the interaction among ZEB1-AS1, miR-216a-5p, and BMP7 was predicted by MiRcode (http://www.mircode.org) and starBase 2.0 (omics_06102, omicX), and confirmed by luciferase reporter assay. ZEB1-AS1 and BMP7 were down-regulated, while miR-216a-5p was highly expressed in kidney tissues of DN patients. Consistently, HG treatment decreased the levels of ZEB1-AS1 and BMP7, whereas HG increased miR-216a-5p expression in HK-2 cells in a time-dependent manner. ZEB1-AS1 upregulation inhibited HG-induced EMT and fibrogenesis. Furthermore, ZEB1-AS1 directly targeted miR-216a-5p, and overexpression of miR-216a-5p restored the inhibitory effects of ZEB1-AS1 overexpression on EMT and fibrogenesis. BMP7 was negatively targeted by miR-216a-5p. In addition, ZEB1-AS1 suppressed HG-induced EMT and fibrogenesis by regulating miR-216a-5p and BMP-7. lncRNA ZEB1-AS1 inhibited high glucose-induced EMT and fibrogenesis via regulating miR-216a-5p/BMP7 axis in diabetic nephropathy, providing a potential target for DN therapy.

15.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;53(4): e9288, 2020. graf
Article de Anglais | LILACS | ID: biblio-1089349

RÉSUMÉ

Diabetic nephropathy (DN) is one of the leading causes of mortality in diabetic patients. Long non-coding RNA zinc finger E-box binding homeobox 1 antisense 1 (ZEB1-AS1) plays a crucial role in the development of various diseases, including DN. However, the molecular mechanism of ZEB1-AS1 in DN pathogenesis remains elusive. An in vitro DN model was established by treating HK-2 cells with high glucose (HG). Quantitative polymerase chain reaction (qRT-PCR) was utilized to detect the expression levels of ZEB1-AS1, microRNA-216a-5p (miR-216a-5p), and bone morphogenetic protein 7 (BMP7). Western blot assay was used to evaluate the protein levels of BMP7, epithelial-to-mesenchymal transition (EMT)-related proteins, and fibrosis markers. Additionally, the interaction among ZEB1-AS1, miR-216a-5p, and BMP7 was predicted by MiRcode (http://www.mircode.org) and starBase 2.0 (omics_06102, omicX), and confirmed by luciferase reporter assay. ZEB1-AS1 and BMP7 were down-regulated, while miR-216a-5p was highly expressed in kidney tissues of DN patients. Consistently, HG treatment decreased the levels of ZEB1-AS1 and BMP7, whereas HG increased miR-216a-5p expression in HK-2 cells in a time-dependent manner. ZEB1-AS1 upregulation inhibited HG-induced EMT and fibrogenesis. Furthermore, ZEB1-AS1 directly targeted miR-216a-5p, and overexpression of miR-216a-5p restored the inhibitory effects of ZEB1-AS1 overexpression on EMT and fibrogenesis. BMP7 was negatively targeted by miR-216a-5p. In addition, ZEB1-AS1 suppressed HG-induced EMT and fibrogenesis by regulating miR-216a-5p and BMP-7. lncRNA ZEB1-AS1 inhibited high glucose-induced EMT and fibrogenesis via regulating miR-216a-5p/BMP7 axis in diabetic nephropathy, providing a potential target for DN therapy.


Sujet(s)
Humains , Néphropathies diabétiques/métabolisme , Protéine morphogénétique osseuse de type 7/métabolisme , Transition épithélio-mésenchymateuse/physiologie , ARN long non codant/physiologie , Facteur de transcription Zeb1/métabolisme , Régulation négative , Régulation positive , Cellules cultivées , microARN/métabolisme , Néphropathies diabétiques/génétique , Réaction de polymérisation en chaine en temps réel
16.
Front Oncol ; 9: 1323, 2019.
Article de Anglais | MEDLINE | ID: mdl-31828042

RÉSUMÉ

ZEB1 is a master regulator of the Epithelial-to-Mesenchymal Transition (EMT) program. While extensive evidence confirmed the importance of ZEB1 as an EMT transcription factor that promotes tumor invasiveness and metastasis, little is known about its regulation. In this work, we screened for potential regulatory links between ZEB1 and multiple cellular kinases. Exploratory in silico analysis aided by phospho-substrate antibodies and ZEB1 deletion mutants led us to identify several potential phospho-sites for the family of PKC kinases in the N-terminus of ZEB1. The analysis of breast cancer cell lines panels with different degrees of aggressiveness, together with the evaluation of a battery of kinase inhibitors, allowed us to expose a robust correlation between ZEB1 and PKCα both at mRNA and protein levels. Subsequent validation experiments using siRNAs against PKCα revealed that its knockdown leads to a concomitant decrease in ZEB1 levels, while ZEB1 knockdown had no impact on PKCα levels. Remarkably, PKCα-mediated downregulation of ZEB1 recapitulates the inhibition of mesenchymal phenotypes, including inhibition in cell migration and invasiveness. These findings were extended to an in vivo model, by demonstrating that the stable knockdown of PKCα using lentiviral shRNAs markedly impaired the metastatic potential of MDA-MB-231 breast cancer cells. Taken together, our findings unveil an unforeseen regulatory pathway comprising PKCα and ZEB1 that promotes the activation of the EMT in breast cancer cells.

17.
Asian J Androl ; 21(5): 460-467, 2019.
Article de Anglais | MEDLINE | ID: mdl-30880686

RÉSUMÉ

One of the factors promoting tumoral progress is the abnormal activation of the epithelial-mesenchymal transition (EMT) program which has been associated with chemoresistance in tumoral cells. The transcription factor zinc finger E-box-binding homeobox 1 (ZEB1), a key EMT activator, has recently been related to docetaxel resistance, the main chemotherapeutic used in advanced prostate cancer treatment. The mechanisms involved in this protective effect are still unclear. In a previous work, we demonstrated that ZEB1 expression induced an EMT-like phenotype in prostate cancer cell lines. In this work, we used prostate cancer cell lines 22Rv1 and DU145 to study the effect of ZEB1 modulation on docetaxel resistance and its possible mechanisms. The results showed that ZEB1 overexpression conferred to 22Rv1 cell resistance to docetaxel while its silencing made DU145 cells more sensitive to it. Analysis of resistance markers showed no presence of ATP-binding cassette subfamily B member 1 (MDR1) and no changes in breast cancer resistance protein (BCRP) or ATP-binding cassette subfamily C member 10 (MRP7). However, a correlation between ZEB1, multidrug resistance-associated protein 1 (MRP1), and ATP-binding cassette subfamily C member 4 (MRP4) expression was observed. MRP4 inhibition, using MK571, resensitized cells with ZEB1 overexpression to docetaxel treatment. In addition, modulation of ZEB1 and subsequent change in MRP4 expression correlated with a lower apoptotic response to docetaxel, characterized by lower B-cell lymphoma 2 (Bcl2), high BCL2-associated X protein (Bax), and high active caspase 3 expression. The response to docetaxel in our model seems to be mediated mainly by activation of the apoptotic death program. Our results showed that modulation of MRP4 could be a mediator of ZEB1-related resistance to docetaxel in prostate cancer, making it a possible marker for chemotherapy response in patients who do not express MDR1.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Docetaxel/usage thérapeutique , Tumeurs de la prostate/traitement médicamenteux , Facteur de transcription Zeb1/métabolisme , Technique de Western , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Extinction de l'expression des gènes , Humains , Mâle , Tumeurs de la prostate/métabolisme
18.
Asian J Androl ; 20(3): 294-299, 2018.
Article de Anglais | MEDLINE | ID: mdl-29271397

RÉSUMÉ

It has been reported that one of the factors that promotes tumoral progression is the abnormal activation of the epithelial-mesenchymal transition program. This process is associated with tumoral cells acquiring invasive and malignant properties and has the transcription factor zinc finger E-box-binding homeobox 1 (ZEB1) as one of its main activators. However, the role of ZEB1 in promoting malignancy in prostate cancer (PCa) is still unclear. Here, we report that ZEB1 expression correlates with Gleason score in PCa samples and that expression of ZEB1 regulates epithelial-mesenchymal transition and malignant characteristics in PCa cell lines. The results showed that ZEB1 expression is higher in samples of higher malignancy and that overexpression of ZEB1 was able to induce epithelial-mesenchymal transition by upregulating the mesenchymal marker Vimentin and downregulating the epithelial marker E-Cadherin. On the contrary, ZEB1 silencing repressed Vimentin expression and upregulated E-Cadherin. ZEB1 expression conferred enhanced motility and invasiveness and a higher colony formation capacity to 22Rv1 cells whereas DU145 cells with ZEB1 silencing showed a decrease in those same properties. The results showed that ZEB1 could be a key promoter of tumoral progression toward advanced stages of PCa.


Sujet(s)
Transition épithélio-mésenchymateuse/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Facteur de transcription Zeb1/génétique , Facteur de transcription Zeb1/métabolisme , Cadhérines/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Extinction de l'expression des gènes , Humains , Mâle , Grading des tumeurs , Invasion tumorale/génétique , Tumeurs de la prostate/anatomopathologie , Vimentine/métabolisme
19.
Mol Immunol ; 66(2): 290-8, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-25910959

RÉSUMÉ

T cell activation leads to the induction of genes that are required for appropriate immune responses. This includes CRTAM (Class-I MHC-restricted T cell associated molecule), a protein that plays a key role in T cell development, proliferation, and generating cell polarity during activation. We previously characterized the CRTAM promoter and described how AP-1 family members are important for inducing CRTAM expression upon antigenic activation. Here, we show that CRTAM is a molecular target for ZEB1 (zinc finger E-box-binding protein), a homeodomain/Zn finger transcription factor. Overexpression of ZEB1 repressed CRTAM promoter activity, as well as endogenous CRTAM levels in human T cells. ZEB1-mediated transcriptional repression was abolished when E-box-like elements in the CRTAM promoter are mutated. In summary, ZEB1 functions as a transcriptional repressor for the CRTAM gene in both non-stimulated and stimulated T cells, thereby modulating adaptive immune responses.


Sujet(s)
Régulation de l'expression des gènes/immunologie , Protéines à homéodomaine/génétique , Immunoglobulines/génétique , Facteurs de transcription/génétique , Immunité acquise , Sites de fixation , Gènes rapporteurs , Protéines à homéodomaine/immunologie , Humains , Immunoglobulines/immunologie , Cellules Jurkat , Luciferases/génétique , Luciferases/métabolisme , Activation des lymphocytes , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/immunologie , Régions promotrices (génétique) , Liaison aux protéines , Transduction du signal , Facteur de transcription AP-1/génétique , Facteur de transcription AP-1/immunologie , Facteurs de transcription/immunologie , Transcription génétique , Facteur de transcription Zeb1
20.
Res Vet Sci ; 97(3): 554-9, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25447746

RÉSUMÉ

ZEB1 and ZEB2 have been recently related to cancer prognosis. We investigated their expression and its association with clinicopathological parameters and overall survival in invasive micropapillary carcinoma (IMPC), which is a metastasising neoplasm of the canine mammary gland. Immunohistochemical evaluation showed nuclear and cytoplasmic staining for ZEB2 and nuclear staining for ZEB1. 'In situ' areas presented higher positivity for cytoplasmic ZEB2 than invasive areas of IMPC did (p = 0.03). ZEB1 positivity was associated with a low histological grade (p = 0.01). A shorter overall survival rate was observed in IMPCs that were positive for cytoplasmic ZEB2 (p = 0.04). Antibodies specificity in canine species was confirmed by western blot. Our results indicated that cytoplasmic ZEB2 appears to be an important factor in the early stages of malignancy and predicts a poor overall survival rate for IMPC in this canine mammary cancer model. ZEB1 downregulation appears to be associated with the dedifferentiation process of IMPC.


Sujet(s)
Carcinome papillaire/métabolisme , Protéines à homéodomaine/métabolisme , Tumeurs mammaires de l'animal/métabolisme , Protéines de répression/métabolisme , Animaux , Technique de Western , Noyau de la cellule/métabolisme , Survie cellulaire/physiologie , Cytoplasme/métabolisme , Chiens , Femelle , Immunohistochimie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE