Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 23(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35008872

ABSTRACT

The Wnt/ß-catenin pathway plays an important role in tumor progression and chemotherapy resistance and seems to be essential for the maintenance of cancer stem cells (CSC) in several tumor types. However, the interplay of these factors has not been fully addressed in bladder cancer. Here, our goal was to analyze the role of the Wnt/ß-catenin pathway in paclitaxel resistance and to study the therapeutic efficacy of its inhibition in bladder cancer cells, as well as to determine its influence in the maintenance of the CSC-like phenotype in bladder cancer. Our results show that paclitaxel-resistant HT1197 cells have hyperactivation of the Wnt/ß-catenin pathway and increased CSC-like properties compared with paclitaxel-sensitive 5637 cells. Paclitaxel sensitivity diminishes in 5637 cells after ß-catenin overexpression or when they are grown as tumorspheres, enriched for the CSC-like phenotype. Additionally, downregulation of ß-catenin or inhibition with XAV939 sensitizes HT1197 cells to paclitaxel. Moreover, a subset of muscle-invasive bladder carcinomas shows aberrant expression of ß-catenin that associates with positive expression of the CSC marker ALDH1A1. In conclusion, we demonstrate that Wnt/ß-catenin signaling contributes to paclitaxel resistance in bladder cancer cells with CSC-like properties.


Subject(s)
Drug Resistance, Neoplasm , Neoplastic Stem Cells/metabolism , Paclitaxel/therapeutic use , Urinary Bladder Neoplasms/metabolism , Wnt Signaling Pathway , Cell Line, Tumor , Humans , Neoplastic Stem Cells/pathology , Urinary Bladder Neoplasms/pathology
2.
Cancers (Basel) ; 10(12)2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30563080

ABSTRACT

Paclitaxel is a treatment option for advanced or metastatic bladder cancer after the failure of first-line cisplatin and gemcitabine, although resistance limits its clinical benefits. Mcl-1 is an anti-apoptotic protein that promotes resistance to paclitaxel in different tumors. Obatoclax, a BH3 mimetic of the Bcl-2 family of proteins, antagonizes Mcl-1 and hence may reverse paclitaxel resistance in Mcl-1-overexpressing tumors. In this study, paclitaxel-sensitive 5637 and -resistant HT1197 bladder cancer cells were treated with paclitaxel, obatoclax, or combinations of both. Apoptosis, cell cycle, and autophagy were measured by Western blot, flow cytometry, and fluorescence microscopy. Moreover, Mcl-1 expression was analyzed by immunohistochemistry in bladder carcinoma tissues. Our results confirmed that paclitaxel alone induced Mcl-1 downregulation and apoptosis in 5637, but not in HT1197 cells; however, combinations of obatoclax and paclitaxel sensitized HT1197 cells to the treatment. In obatoclax-treated 5637 and obatoclax + paclitaxel-treated HT1197 cells, the blockade of the autophagic flux correlated with apoptosis and was associated with caspase-dependent cleavage of beclin-1. Obatoclax alone delayed the cell cycle in 5637, but not in HT1197 cells, whereas combinations of both retarded the cell cycle and reduced mitotic slippage. In conclusion, obatoclax sensitizes HT1197 cells to paclitaxel-induced apoptosis through the blockade of the autophagic flux and effects on the cell cycle. Furthermore, Mcl-1 is overexpressed in many invasive bladder carcinomas, and it is related to tumor progression, so Mcl-1 expression may be of predictive value in bladder cancer.

3.
Mol Cancer Ther ; 15(7): 1713-25, 2016 07.
Article in English | MEDLINE | ID: mdl-27196755

ABSTRACT

Prostate cancer is the leading cause of cancer-related death among men in developed countries. Although castration therapy is initially effective, prostate cancers progress to hormone-refractory disease and in this case taxane-based chemotherapy is widely used. Castration-resistant prostate cancer cells often develop resistance to chemotherapy agents and the search for new therapeutic strategies is necessary. In this article, we demonstrate that PKCδ silencing favors mitotic arrest after paclitaxel treatment in PC3 and LNCaP cells; however, this is associated with resistance to paclitaxel-induced apoptosis. In prostate cancer cells, PKCδ seems to exert a proapoptotic role, acting as a negative regulator of the canonical Wnt/ß-catenin pathway. PKCδ silencing induces activation of Wnt/ß-catenin pathway and the expression of its target genes, including Aurora kinase A, which is involved in activation of Akt and both factors play a key role in GSK3ß inactivation and consequently in the stabilization of ß-catenin and antiapoptotic protein Mcl-1. We also show that combined treatments with paclitaxel and Wnt/ß-catenin or Akt inhibitors improve the apoptotic response to paclitaxel, even in the absence of PKCδ. Finally, we observe that high Gleason score prostate tumors lose PKCδ expression and this correlates with higher activation of ß-catenin, inactivation of GSK3ß, and higher levels of Aurora kinase A and Mcl-1 proteins. These findings suggest that targeting Wnt/ß-catenin or Akt pathways may increase the efficacy of taxane chemotherapy in advanced human prostate cancers that have lost PKCδ expression. Mol Cancer Ther; 15(7); 1713-25. ©2016 AACR.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Paclitaxel/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Kinase C-delta/deficiency , Wnt Signaling Pathway/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Chromones/pharmacology , Gene Expression , Gene Silencing , Humans , Male , Mitosis/drug effects , Mitosis/genetics , Models, Biological , Morpholines/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , beta Catenin/metabolism
4.
Mol Cancer Ther ; 13(10): 2372-83, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25122070

ABSTRACT

PTTG1 protein, the human securin, has a central role in sister chromatid separation during mitosis, and its altered expression has been reported in many tumor types. Paclitaxel is a widely used chemotherapeutic drug, whose mechanism of action is related to its ability to arrest cells in mitosis and the subsequent induction of the intrinsic apoptotic pathway. By using two prostate cancer cell lines with different responses to paclitaxel treatment, we have identified two situations in which PTTG1 influences cell fate differentially. In slippage-prone PC3 cells, both PTTG1 downregulation and overexpression induce an increase in mitotic cells that is associated with diminished apoptosis after paclitaxel treatment. In LNCaP cells, however, PTTG1 downregulation prevents mitotic entry and, subsequently, inhibits mitosis-associated, paclitaxel-induced apoptosis. In contrast, PTTG1 overexpression induces an increase in mitotic cells and apoptosis after paclitaxel treatment. We have also identified a role for Mcl-1 protein in preventing apoptosis during mitosis in PC3 cells, as simultaneous PTTG1 and Mcl-1 silencing enhances mitosis-associated apoptosis after paclitaxel treatment. The finding that a more efficient mitotic arrest alone in PC3 cells is not enough to increase apoptosis was also confirmed with the observation that a selected paclitaxel-resistant PC3 cell line showed an apoptosis-resistant phenotype associated with increased mitosis upon paclitaxel treatment. These findings could contribute to identify putative responsive and nonresponsive cells and help us to approach incomplete responses to paclitaxel in the clinical setting.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Securin/biosynthesis , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Down-Regulation , Gene Silencing , Humans , Male , Mitosis/drug effects , Mitosis/genetics , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Securin/genetics , Transfection
5.
Clin Exp Metastasis ; 29(4): 349-58, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22274591

ABSTRACT

PTPL1, a non-receptor type protein tyrosine phosphatase, has been involved in the regulation of apoptosis and invasiveness of various tumour cell types, but its role in prostate cancer remained to be investigated. We report here that downregulation of PTPL1 by small interfering RNA in PC3 cells decreases cell proliferation and concomitantly reduces the expression of cell cycle-related proteins such as cyclins E and B1, PCNA, PTTG1 and phospho-histone H3. PTPL1 downregulation also increases the invasion ability of PC3 cells through Matrigel coated membranes. cDNA array of PTPL1-silenced PC3 cells versus control cells showed an upregulation of invasion-related genes such as uPA, uPAR, tPA, PAI-1, integrin α6 and osteopontin. This increased expression was also confirmed in PTPL1-silenced DU145 prostate cancer cells by quantitative real time PCR and western blot. These findings suggest that PTPL1 is an important mediator of central cellular processes such as proliferation and invasion.


Subject(s)
Cell Cycle , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , Blotting, Western , Cell Cycle/drug effects , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Enzymologic , Humans , Integrin alpha6/genetics , Integrin alpha6/metabolism , Male , Neoplasm Invasiveness/genetics , Osteopontin/genetics , Osteopontin/metabolism , Phenotype , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Prostatic Neoplasms/enzymology , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
6.
Breast Cancer Res Treat ; 133(3): 917-28, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22076480

ABSTRACT

Taxanes are being used for the treatment of breast cancer. However, cancer cells frequently develop resistance to these drugs with the subsequent recurrence of the tumor. MDA-MB-231 and T-47D breast cancer cell lines were used to assess the effect of paclitaxel treatment on apoptosis and cell cycle, the possible mechanisms of paclitaxel resistance as well as the enhancement of paclitaxel-induced apoptosis based on its combination with phenylethyl isothiocyanate (PEITC). T-47D cells undergo apoptosis in response to paclitaxel treatment. The induction of apoptosis was associated with a robust mitotic arrest and the disruption of Bcl-xL/Bak interaction. By contrary, MDA-MB-231 cells were insensitive to paclitaxel-induced apoptosis and this was associated with a high percentage of cells that slip out of paclitaxel-imposed mitotic arrest and also with the maintenance of Bcl-xL/Bak interaction. The sequential treatment of MDA-MB-231 cells with PEITC followed by paclitaxel inhibited the slippage induced by paclitaxel and increased the apoptosis induction achieved with any of the drugs alone. In breast cancer tissues, high Bcl-xL expression was correlated with a shorter time of disease-free survival in patients treated with a chemotherapeutic regimen that contains paclitaxel, in a statistically significant way. Thus, resistance to paclitaxel in MDA-MB-231 cells is related to the inability to disrupt the Bcl-xL/Bak interaction and increased slippage. In this context, the combination of a drug that induces a strong mitotic arrest, such as paclitaxel, with another that inhibits slippage, such as PEITC, translates into increased apoptotic induction.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , M Phase Cell Cycle Checkpoints/drug effects , Paclitaxel/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Line, Tumor , Female , Humans , Isothiocyanates/pharmacology , Paclitaxel/therapeutic use , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...