Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 13(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670655

ABSTRACT

BACKGROUND: Fascin1 is the key actin-bundling protein involved in cancer invasion and metastasis whose expression is associated with bad prognosis in tumor from different origins. METHODS: In the present study, virtual screening (VS) was performed for the search of Fascin1 inhibitors and RAL, an FDA-approved inhibitor of human immunodeficiency virus-1 (HIV-1) integrase, was identified as a potential Fascin1 inhibitor. Biophysical techniques including nuclear magnetic resonance (NMR) and differential scanning fluorimetry (DSF) were carried out in order to confirm RAL as a Fascin1 blocker. The effect of RAL on actin-bundling activity Fascin1 was assessed by transmission electron microscopy (TEM), immunofluorescence, migration, and invasion assays on two human colorectal adenocarcinoma cell lines: HCT-116 and DLD-1. In addition, the anti-metastatic potential of RAL was in vivo evaluated by using the zebrafish animal model. RESULTS: NMR and DSF confirmed in silico predictions and TEM demonstrated the RAL-induced disorganization of the actin structure compared to control conditions. The protrusion of lamellipodia in cancer cell line overexpressing Fascin1 (HCT-116) was abolished in the presence of this drug. By following the addition of RAL, migration of HCT-116 and DLD-1 cell lines was significantly inhibited. Finally, using endogenous and exogenous models of Fascin1 expression, the invasive capacity of colorectal tumor cells was notably impaired in the presence of RAL in vivo assays; without undesirable cytotoxic effects. CONCLUSION: The current data show the in vitro and in vivo efficacy of the antiretroviral drug RAL in inhibiting human colorectal cancer cells invasion and metastasis in a Fascin1-dependent manner.

2.
Int J Mol Sci ; 21(6)2020 Mar 14.
Article in English | MEDLINE | ID: mdl-32183342

ABSTRACT

Serrated adenocarcinoma (SAC) is a tumor recognized by the WHO as a histological subtype accounting for around 9% of colorectal carcinomas. Compared to conventional carcinomas, SACs are characterized by a worse prognosis, weak development of the immune response, an active invasive front and a frequent resistance to targeted therapy due to a high occurrence of KRAS or BRAF mutation. Nonetheless, several high-throughput studies have recently been carried out unveiling the biology of this cancer and identifying potential molecular targets, favoring a future histologically based treatment. This review revises the current evidence, aiming to propose potential molecular targets and specific treatments for this aggressive tumor.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Microsatellite Instability , Neovascularization, Pathologic/pathology , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Escape/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
3.
Exp Mol Med ; 52(2): 281-292, 2020 02.
Article in English | MEDLINE | ID: mdl-32080340

ABSTRACT

Serrated adenocarcinoma (SAC) is more invasive, has worse outcomes than conventional colorectal carcinoma (CRC), and is characterized by frequent resistance to anti-epidermal growth factor receptor (EGFR) and overexpression of fascin1, a key protein in actin bundling that plays a causative role in tumor invasion and is overexpressed in different cancer types with poor prognosis. In silico screening of 9591 compounds, including 2037 approved by the Food and Drug Administration (FDA), was performed, and selected compounds were analyzed for their fascin1 binding affinity by differential scanning fluorescence. The results were compared with migrastatin as a typical fascin1 inhibitor. In silico screening and differential scanning fluorescence yielded the FDA-approved antidepressant imipramine as the most evident potential fascin1 blocker. Biophysical and different in vitro actin-bundling assays confirm this activity. Subsequent assays investigating lamellipodia formation and migration and invasion of colorectal cancer cells in vitro using 3D human tissue demonstrated anti-fascin1 and anti-invasive activities of imipramine. Furthermore, expression profiling suggests the activity of imipramine on the actin cytoskeleton. Moreover, in vivo studies using a zebrafish invasion model showed that imipramine is tolerated, its anti-invasive and antimetastatic activities are dose-dependent, and it is associated with both constitutive and induced fascin1 expression. This is the first study that demonstrates an antitumoral role of imipramine as a fascin1 inhibitor and constitutes a foundation for a molecular targeted therapy for SAC and other fascin1-overexpressing tumors.


Subject(s)
Antidepressive Agents/pharmacology , Carrier Proteins/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Imipramine/pharmacology , Microfilament Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Macrolides/pharmacology , Neoplasm Invasiveness/pathology , Piperidones/pharmacology , Zebrafish
4.
Nat Med ; 25(7): 1073-1081, 2019 07.
Article in English | MEDLINE | ID: mdl-31270502

ABSTRACT

Bladder cancer is lethal in its advanced, muscle-invasive phase with very limited therapeutic advances1,2. Recent molecular characterization has defined new (epi)genetic drivers and potential targets for bladder cancer3,4. The immune checkpoint inhibitors have shown remarkable efficacy but only in a limited fraction of bladder cancer patients5-8. Here, we show that high G9a (EHMT2) expression is associated with poor clinical outcome in bladder cancer and that targeting G9a/DNMT methyltransferase activity with a novel inhibitor (CM-272) induces apoptosis and immunogenic cell death. Using an immunocompetent quadruple-knockout (PtenloxP/loxP; Trp53loxP/loxP; Rb1loxP/loxP; Rbl1-/-) transgenic mouse model of aggressive metastatic, muscle-invasive bladder cancer, we demonstrate that CM-272 + cisplatin treatment results in statistically significant regression of established tumors and metastases. The antitumor effect is significantly improved when CM-272 is combined with anti-programmed cell death ligand 1, even in the absence of cisplatin. These effects are associated with an endogenous antitumor immune response and immunogenic cell death with the conversion of a cold immune tumor into a hot tumor. Finally, increased G9a expression was associated with resistance to programmed cell death protein 1 inhibition in a cohort of patients with bladder cancer. In summary, these findings support new and promising opportunities for the treatment of bladder cancer using a combination of epigenetic inhibitors and immune checkpoint blockade.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cisplatin/therapeutic use , Enhancer of Zeste Homolog 2 Protein/physiology , Female , Histocompatibility Antigens , Humans , Mice , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
5.
Clin Cancer Res ; 25(1): 390-402, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30242024

ABSTRACT

PURPOSE: Bladder cancer is a clinical and social problem due to its high incidence and recurrence rates. It frequently appears in elderly patients showing other medical comorbidities that hamper the use of standard chemotherapy. We evaluated the activity of CDK4/6 inhibitor as a new therapy for patients unfit for cisplatin (CDDP). EXPERIMENTAL DESIGN: Bladder cancer cell lines were tested for in vitro sensitivity to CDK4/6 inhibition. A novel metastatic bladder cancer mouse model was developed and used to test its in vivo activity. RESULTS: Cell lines tested were sensitive to CDK4/6 inhibition, independent on RB1 gene status. Transcriptome analyses and knockdown experiments revealed a major role for FOXM1 in this response. CDK4/6 inhibition resulted in reduced FOXM1 phosphorylation in vitro and in vivo and showed synergy with CDDP, allowing a significant tumor regression. FOXM1 exerted important oncogenic roles in bladder cancer. CONCLUSIONS: CDK4/6 inhibitors, alone or in combination, are a novel therapeutic strategy for patients with advanced bladder cancer previously classified as unfit for current treatment options.


Subject(s)
Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Forkhead Box Protein M1/genetics , Urinary Bladder Neoplasms/drug therapy , Aged , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Female , Heterografts , Humans , Male , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Phosphorylation/drug effects , Progression-Free Survival , Protein Kinase Inhibitors/pharmacology , Retinoblastoma Binding Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
6.
Clin Cancer Res ; 23(23): 7388-7399, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28928159

ABSTRACT

Purpose: Bladder cancer is a current clinical and social problem. At diagnosis, most patients present with nonmuscle-invasive tumors, characterized by a high recurrence rate, which could progress to muscle-invasive disease and metastasis. Bone morphogenetic protein (BMP)-dependent signaling arising from stromal bladder tissue mediates urothelial homeostasis by promoting urothelial cell differentiation. However, the possible role of BMP ligands in bladder cancer is still unclear.Experimental Design: Tumor and normal tissue from 68 patients with urothelial cancer were prospectively collected and analyzed for expression of BMP and macrophage markers. The mechanism of action was assessed in vitro by experiments with bladder cancer cell lines and peripheral blood monocyte-derived macrophages.Results: We observed BMP4 expression is associated and favored type II macrophage differentiation. In vitro experiments showed that both recombinant BMP4 and BMP4-containing conditioned media from bladder cancer cell lines favored monocyte/macrophage polarization toward M2 phenotype macrophages, as shown by the expression and secretion of IL10. Using a series of human bladder cancer patient samples, we also observed increased expression of BMP4 in advanced and undifferentiated tumors in close correlation with epithelial-mesenchymal transition (EMT). However, the p-Smad 1,5,8 staining in tumors showing EMT signs was reduced, due to the increased miR-21 expression leading to reduced BMPR2 expression.Conclusions: These findings suggest that BMP4 secretion by bladder cancer cells provides the M2 signal necessary for a protumoral immune environment. In addition, the repression of BMPR2 by miR-21 makes the tumor cells refractory to the prodifferentiating actions mediated by BMP ligands, favoring tumor growth. Clin Cancer Res; 23(23); 7388-99. ©2017 AACR.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Gene Expression Regulation, Neoplastic , Macrophage Activation/genetics , Macrophages/metabolism , Urinary Bladder Neoplasms/genetics , Aged , Aged, 80 and over , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Disease Progression , Disease-Free Survival , Epithelial-Mesenchymal Transition/genetics , Female , Humans , K562 Cells , Macrophages/classification , Male , MicroRNAs/genetics , Middle Aged , Prospective Studies , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
7.
Oncotarget ; 8(6): 10531-10542, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28060766

ABSTRACT

The high rates of tumor recurrence and progression represent a major clinical problem in non-muscle invasive bladder cancer. Previous data showed that EZH2-dependent signaling mediates these processes, whereas the frequent alterations of PIK3CA gene (copy gains and mutations) are predictive of reduced recurrence. Here we show, using clinical samples and bladder cancer cell lines, a functional interaction between EZH2- and PIK3CA-dependent signaling pathways. PIK3CA alterations mediated, on the one hand, the increased expression of two miRNAs, miR-101 and miR-138, which posttranscriptionally downregulate EZH2 expression. On the other hand, PIK3CA alterations facilitate the activation of Akt which phosphorylates EZH2 on Ser21, precluding the trimethylation of histone H3 in K27. Remarkably the increased expression of miR101 or miR138 and the expression of Ser21-phosphorylated EZH2 are good prognostic factors regarding non-muscle invasive bladder cancer recurrence and progression. Collectively, this study provides molecular evidences indicating that the gene expression rewiring occurring in primary bladder tumors, associated with increased EZH2 expression and activity and mediating the increased recurrence and progression risk, are prevented by PIK3CA-dependent signaling. This molecular process may have deep implications in the management of bladder cancer patients and in the design of novel molecularly targeted therapeutic approaches.


Subject(s)
Biomarkers, Tumor/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Mutation , Signal Transduction , Urinary Bladder Neoplasms/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/metabolism , Disease Progression , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Kaplan-Meier Estimate , Methylation , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Time Factors , Transcription, Genetic , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy
8.
Int J Mol Sci ; 16(11): 27107-32, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26580594

ABSTRACT

Bladder Cancer (BC) represents a current clinical and social challenge. The recent studies aimed to describe the genomic landscape of BC have underscored the relevance of epigenetic alterations in the pathogenesis of these tumors. Among the epigenetic alterations, histone modifications occupied a central role not only in cancer, but also in normal organism homeostasis and development. EZH2 (Enhancer of Zeste Homolog 2) belongs to the Polycomb repressive complex 2 as its catalytic subunit, which through the trimethylation of H3 (Histone 3) on K27 (Lysine 27), produces gene silencing. EZH2 is frequently overexpressed in multiple tumor types, including BC, and plays multiple roles besides the well-recognized histone mark generation. In this review, we summarize the present knowledge on the oncogenic roles of EZH2 and its potential use as a therapeutic target, with special emphasis on BC pathogenesis and management.


Subject(s)
Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , DNA Methylation , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Histones/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Targeted Therapy , Polycomb Repressive Complex 2/antagonists & inhibitors , Protein Binding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Urinary Bladder Neoplasms/drug therapy
9.
Oncotarget ; 6(39): 42258-75, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26517683

ABSTRACT

Bladder cancer (BC) is a highly prevalent disease, ranking fifth in the most common cancers worldwide. Various miRNAs have recently emerged as potential prognostic biomarkers in cancer. The miR-200 family, which repressed the epithelial-to-mesenchymal transition (EMT), is repressed in multiple advanced cancers. However, its expression and function in BC is still poorly understood. Here we show that miR-200 family displays increased expression, probably due to the activation of specific oncogenic signaling pathways, and reduced promoter methylation, in BC compared to normal bladder samples. Furthermore, we show that the expression of these miRNAs is decreased in high grade and stage tumors, and the down-regulation is associated with patient's poor clinical outcome. Our data indicate that the miR-200 family plays distinct roles in Non-Muscle (NMIBC) and Muscle-Invasive BC (MIBC). In MIBC, miR-200 expression post transcriptionally regulates EMT-promoting transcription factors ZEB1 and ZEB2, whereas suppresses BMI1 expression in NMIBC. Interestingly, we show that increased EZH2 and/or BMI1 expression repress the expression of miR-200 family members. Collectively, these findings support a model of BC progression through a coordinated action between the Polycomb Repression Complex (PRC) members repressing the miR-200 expression, which ultimately favors invasive BC development. Since pharmacological inhibition of EZH2 in BC cell lines lead to increased miR-200 expression, our findings may support new therapeutic strategies for BC clinical management.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Polycomb-Group Proteins/genetics , Urinary Bladder Neoplasms/genetics , Aged , Aged, 80 and over , Blotting, Western , Cell Line, Tumor , DNA Methylation , Enhancer of Zeste Homolog 2 Protein , Epithelial-Mesenchymal Transition/genetics , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins/metabolism , Prognosis , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
10.
Clin Epigenetics ; 7: 109, 2015.
Article in English | MEDLINE | ID: mdl-26457124

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) have been claimed as key molecular players in gene expression regulation, being involved in diverse epigenetic processes. They are aberrantly expressed in various tumors, but their exact role in bladder cancer is still obscure. We have recently found a major role of the Polycomb repression complex in recurrence of non-muscle-invasive bladder cancer. Here, we report the xpression of Polycomb-related lncRNAs:antisense noncoding RNA in the INK4 locus (ANRIL) and HOX antisense intergenic RNA (HOTAIR) in these tumors. FINDINGS: We studied a dataset of non-invasive bladder cancer samples by quantitative reverse transcription PCR (RT-qPCR) and analyzed also invasive bladder cancer samples using TCGA data. Our results showed that, while ANRIL seemed not to have a determining role, an increased HOTAIR expression appeared in recurrent and high-graded tumors associated with poor prognosis. In addition, through genome-wide transcriptome analyses, we observed that HOTAIR-EZH2-complex-regulated genes can efficiently discriminate between non-tumoral, recurrent, and non-recurrent bladder cancer samples. We also observed a significant correlation between EZH2 and HOTAIR expression levels. Using overexpression, knockdown, and pharmacological approaches in bladder cancer cell lines, we also observed that EZH2 regulates HOTAIR expression. CONCLUSIONS: Our findings indicate that HOTAIR expression has prognostic value for bladder cancer progression, recurrence, and survival and suggest that HOTAIR plays active roles in modulating the cancer epigenome, becoming an interesting candidate as a target for cancer diagnosis and therapy. The observed HOTAIR regulation by EZH2 and the possibility of modulating EZH2 activity with specific inhibitors open new possible paths to be explored in bladder cancer therapy.

11.
Oncotarget ; 6(27): 24230-45, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26203771

ABSTRACT

The TP63 gene codes for two major isoform types, TAp63 and ΔNp63, with probable opposite roles in tumorigenesis. The ΔNp63α protein is frequently amplified and overexpressed in different epithelial tumors. Accordingly, it has been considered a potential oncogene. Nonetheless, a possible metastatic suppressor activity has also been suggested based on the experimental observation that its expression is reduced or even absent in advanced invasive tumors. Such metastatic suppressor activities are often related to tumors bearing point mutated TP53 gene. However, its potential roles in TP53-deficient tumors are poorly characterized. Here we show that in spontaneous tumors, induced by the epidermal-specific Trp53 ablation, the reduction of ΔNp63 expression is an early event, whereas it is re-expressed in the lung metastatic lesions. Using knock down and ectopic expression approaches, we show that ΔNp63 expression opposes the epithelial-mesenchymal transition and reduces the metastatic potential of the cells. This process occurs through the modulation of ΔNp63-dependent downstream targets (including transcription factors and microRNAs) likely to play metastatic roles. Further, ΔNp63 also favors the expression of factors involved in iPS reprogramming, thus suggesting that it can also modulate specific stem cell traits in mouse epidermal tumor cells. Overall, our data assign antimetastatic roles to ΔNp63 in the context of p53 deficiency and epidermis.


Subject(s)
Down-Regulation , Epidermis/metabolism , Skin Neoplasms/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Line , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Keratinocytes/metabolism , Mice , Microscopy, Fluorescence , Mutation , Neoplasm Metastasis , Phosphoproteins/genetics , Point Mutation , Skin/pathology , Skin Neoplasms/metabolism , Stem Cells/cytology , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...