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










Publication year range
1.
Cell Death Dis ; 15(5): 370, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806454

ABSTRACT

In ovarian tumors, the omental microenvironment profoundly influences the behavior of cancer cells and sustains the acquisition of stem-like traits, with major impacts on tumor aggressiveness and relapse. Here, we leverage a patient-derived platform of organotypic cultures to study the crosstalk between the tumor microenvironment and ovarian cancer stem cells. We discovered that the pro-tumorigenic transcription factor FOXM1 is specifically induced by the microenvironment in ovarian cancer stem cells, through activation of FAK/YAP signaling. The microenvironment-induced FOXM1 sustains stemness, and its inactivation reduces cancer stem cells survival in the omental niche and enhances their response to the PARP inhibitor Olaparib. By unveiling the novel role of FOXM1 in ovarian cancer stemness, our findings highlight patient-derived organotypic co-cultures as a powerful tool to capture clinically relevant mechanisms of the microenvironment/cancer stem cells crosstalk, contributing to the identification of tumor vulnerabilities.


Subject(s)
Forkhead Box Protein M1 , Neoplastic Stem Cells , Ovarian Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/drug effects , Forkhead Box Protein M1/metabolism , Forkhead Box Protein M1/genetics , Female , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/drug effects , Cell Line, Tumor , Signal Transduction/drug effects , YAP-Signaling Proteins/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Mice , Gene Expression Regulation, Neoplastic/drug effects , Animals , Phthalazines/pharmacology , Piperazines/pharmacology
2.
Eur J Med Chem ; 246: 114961, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36495629

ABSTRACT

Biomedical applications of molecules that are able to modulate ß-adrenergic signaling have become increasingly attractive over the last decade, revealing that ß-adrenergic receptors (ß-ARs) are key targets for a plethora of therapeutic interventions, including cancer. Despite successes in ß-AR drug discovery, identification of ß-AR ligands that are useful as selective chemical tools in pharmacological studies of the three ß-AR subtypes, or lead compounds for drug development is still a highly challenging task. This is mainly due to the intrinsic plasticity of ß-ARs as G protein-coupled receptors in conjunction with the requirement for functional receptor subtype selectivity, tissue specificity and minimal off-target effects. With the aim to provide insight into structure-activity relationships for the three ß-AR subtypes, we have synthesized and obtained the pharmacological profile of a series of structurally diverse compounds (named MC) that were designed based on the aryloxy-propanolamine scaffold of SR59230A. Comparative analysis of their predicted binding mode within the active and inactive states of the receptors in combination with their pharmacological profile revealed key structural elements that control their activity as agonists or antagonists, in addition to clues about substituents that mediate selectivity for one receptor subtype over the others. We anticipate that these results will facilitate selective ß-AR drug development efforts.


Subject(s)
Receptors, Adrenergic, beta , Receptors, G-Protein-Coupled , Humans , Receptors, Adrenergic, beta/chemistry , Receptors, Adrenergic, beta/metabolism , Ligands , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Structure-Activity Relationship
3.
Oncogenesis ; 9(1): 5, 2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32001675

ABSTRACT

Adenoid cystic carcinoma (ACC) is a rare cancer that preferentially occurs in the head and neck, breast, as well as in other sites. It is an aggressive cancer with high rates of recurrence and distant metastasis. Patients with advanced disease are generally incurable due to the lack of effective systemic therapies. Activation of the master transcriptional regulator MYB is the genomic hallmark of ACC. MYB activation occurs through chromosomal translocation, copy number gain or enhancer hijacking, and is the key driving event in the pathogenesis of ACC. However, the functional consequences of alternative mechanisms of MYB activation are still uncertain. Here, we show that overexpression of MYB or MYB-NFIB fusions leads to transformation of human glandular epithelial cells in vitro and results in analogous cellular and molecular consequences. MYB and MYB-NFIB expression led to increased cell proliferation and upregulation of genes involved in cell cycle control, DNA replication, and DNA repair. Notably, we identified the DNA-damage sensor kinase ATR, as a MYB downstream therapeutic target that is overexpressed in primary ACCs and ACC patient-derived xenografts (PDXs). Treatment with the clinical ATR kinase inhibitor VX-970 induced apoptosis in MYB-positive ACC cells and growth inhibition in ACC PDXs. To our knowledge, ATR is the first example of an actionable target downstream of MYB that could be further exploited for therapeutic opportunities in ACC patients. Our findings may also have implications for other types of neoplasms with activation of the MYB oncogene.

4.
Oncogenesis ; 9(1): 3, 2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31900399

ABSTRACT

The MYC family of transcription factors is a major driver of human cancer and potential therapeutic target. However, no clinically viable drugs have been yet developed that are able to directly tackle MYC oncoproteins. In our laboratory, we are exploring alternative approaches aiming to disturb signalling downstream of MYC. MYCN is frequently activated in neuroblastoma, a paediatric solid malignancy that, in its metastatic form, has a very poor prognosis. An important pathway regulated by MYC is the CKS1/SKP2/p27kip1 axis. In this study, we have repurposed the anti-psychotic drug Prozac to disrupt CKS1/SKP2/p27Kip1 signalling and assess its potential as an anti-neuroblastoma agent in vitro and in vivo. Using DNA editing technology, we show that stabilisation of p27Kip1 operated by Prozac in MYC-activated cells is essential for the anti-neuroblastoma activity of the drug. Furthermore, dosing mice with a concentration of Prozac equivalent to that used in long-term clinical trials in children with psychiatric disorders caused a significant reduction of metastatic disease in two models of high-risk neuroblastoma. The favourable toxicity profile of Prozac suggests that long-term treatments might be implemented in children with MYC/CKS1high neuroblastomas.

5.
Oncol Rep ; 42(1): 35-42, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31115546

ABSTRACT

Drug-resistant neuroblastoma remains a major challenge in paediatric oncology and novel and less toxic therapeutic approaches are urgently needed to improve survival and reduce the side effects of traditional therapeutic interventions. Mesenchymal stem cells (MSCs) are an attractive candidate for cell and gene therapy since they are recruited by and able to infiltrate tumours. This feature has been exploited by creating genetically modified MSCs that are able to combat cancer by delivering therapeutic molecules. Whether neuroblastomas attract systemically delivered MSCs is still controversial. We investigated whether MSCs engineered to express tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) could: i) cause death of classic and primary neuroblastoma cell lines in vitro; ii) migrate to tumour sites in vivo; and iii) reduce neuroblastoma growth in xenotransplantation experiments. We observed that classic and primary neuroblastoma cell lines expressing death receptors could be killed by TRAIL-loaded MSCs in vitro. When injected in the peritoneum of neuroblastoma-bearing mice, TRAIL-MSCs migrated to tumour sites, but were unable to change the course of cancer development. These results indicated that MSCs have the potential to be used to deliver drugs in neuroblastoma patients, but more effective biopharmaceuticals should be used instead of TRAIL.


Subject(s)
Genetic Engineering/methods , Mesenchymal Stem Cells/cytology , Neuroblastoma/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Female , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Neuroblastoma/metabolism , Neuroblastoma/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Xenograft Model Antitumor Assays
7.
J Med Chem ; 59(23): 10451-10469, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27801583

ABSTRACT

A series of novel 1,3,4-oxadiazoles was synthesized and evaluated for their cytotoxic activity in in vitro tumor models. Four of the new compounds (2d, 2j, 2k, and 2n) showed growth inhibition in the XTT dye assay. The most active agent, 2j, showed high potency against human cancer cells with IC50s ranging from 0.05 to 1.7 µM. Preliminary SAR correlations suggested that the nature of chains on the oxadiazole is important for antitumor potency in vitro. Compound 2j determined a G2/M arrest of the cell cycle and also activated a strong apoptotic response. The ß-tubulin immunofluorescence analysis indicated that compound 2j effectively inhibited the microtubule organization in all cancer cell lines, causing the formation of abnormal spindle, which did not affect the normal human fibroblast cells NB1, Mrc-5 and IBR3. For all these reasons, compound 2j could be a good candidate in chemopreventive or chemotherapeutic strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Oxadiazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Structure-Activity Relationship
8.
Clin Cancer Res ; 22(13): 3398-409, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27076624

ABSTRACT

PURPOSE: Neuroblastoma is a childhood malignancy originating from the sympathetic nervous system with a complex biology, prone to metastasize and relapse. High-risk, metastatic cases are explained in part by amplification or mutation of oncogenes, such as MYCN and ALK, and loss of tumor suppressor genes in chromosome band 1p. However, it is fundamental to identify other pathways responsible for the large portion of neuroblastomas with no obvious molecular alterations. EXPERIMENTAL DESIGN: Neuroblastoma cell lines were used for the assessment of tumor growth in vivo and in vitro Protein expression in tissues and cells was assessed using immunofluorescence and IHC. The association of promyelocytic leukemia (PML) expression with neuroblastoma outcome and relapse was calculated using log-rank and Mann-Whitney tests, respectively. Gene expression was assessed using chip microarrays. RESULTS: PML is detected in the developing and adult sympathetic nervous system, whereas it is not expressed or is low in metastatic neuroblastoma tumors. Reduced PML expression in patients with low-risk cancers, that is, localized and negative for the MYCN proto-oncogene, is strongly associated with tumor recurrence. PML-I, but not PML-IV, isoform suppresses angiogenesis via upregulation of thrombospondin-2 (TSP2), a key inhibitor of angiogenesis. Finally, PML-I and TSP2 expression inversely correlates with tumor angiogenesis and recurrence in localized neuroblastomas. CONCLUSIONS: Our work reveals a novel PML-I-TSP2 axis for the regulation of angiogenesis and cancer relapse, which could be used to identify patients with low-risk, localized tumors that might benefit from chemotherapy. Clin Cancer Res; 22(13); 3398-409. ©2016 AACR.


Subject(s)
Neoplasm Recurrence, Local/pathology , Neovascularization, Pathologic/pathology , Neuroblastoma/pathology , Promyelocytic Leukemia Protein/metabolism , Thrombospondins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neural Crest/embryology , Neuroblastoma/genetics , Promyelocytic Leukemia Protein/genetics , Protein Isoforms/genetics , Proto-Oncogene Mas , Risk Factors , Stem Cells/cytology , Sympathetic Nervous System/embryology , Thrombospondins/genetics , Tumor Suppressor Proteins/genetics
9.
Mol Cancer ; 12: 37, 2013 May 04.
Article in English | MEDLINE | ID: mdl-23642048

ABSTRACT

BACKGROUND: In a previous report, we described the in vitro and in vivo antiproliferative and proapoptotic activity of a hydroxylated biphenyl (D6), a structural analogue of curcumin, on malignant melanoma and neuroblastoma tumours. In this paper, we investigated the molecular changes induced by such a compound, underlying cell growth arrest and apoptosis in melanoma cells. RESULTS: To shed light on the mechanisms of action of D6, we firstly demonstrated its quick cellular uptake and subsequent block of cell cycle in G2/M phase transition. A gene expression profile analysis of D6-treated melanoma cells and fibroblasts was then carried out on high density microarrays, to assess gene expression changes induced by this compound. The expression profile study evidenced both an induction of stress response pathways and a modulation of cell growth regulation mechanisms. In particular, our data suggest that the antiproliferative and proapoptotic activities of D6 in melanoma could be partially driven by up-regulation of the p53 signalling pathways as well as by down-regulation of the PI3K/Akt and NF-kB pathways. Modulation of gene expression due to D6 treatment was verified by western blot analysis for single proteins of interest, confirming the results from the gene expression profile analysis. CONCLUSIONS: Our findings contribute to the understanding of the mechanisms of action of D6, through a comprehensive description of the molecular changes induced by this compound at the gene expression level, in agreement with the previously reported anti-tumour effects on melanoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/genetics , Melanoma/metabolism , Signal Transduction/drug effects , Antineoplastic Agents/metabolism , Cell Cycle/drug effects , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/analogs & derivatives , Curcumin/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Stress, Physiological/drug effects , Transcription, Genetic
10.
Anticancer Res ; 33(3): 871-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23482756

ABSTRACT

BACKGROUND: In malignant melanoma (MM), overexpression of αvß3 integrin is linked to a more metastatic phenotype. Development of anti-αvß3 agents able to counteract melanoma progression would be helpful for disease treatment. A new selective ligand of αvß3, RGDechi-hCit, has anti-angiogenic properties against endothelial cells in animal angiogenesis models. The aim of this study was to evaluate the in vitro effects of the RGDechi-hCit peptide on MM cell lines. MATERIALS AND METHODS: Cytofluorimetric analysis characterized the cell surface expression of αvß3 integrin on seven MM cell lines: A375, WM266-4, SK-Mel-28, Sbcl2, LB24Dagi, PR-Mel and PNP-Mel. Cell proliferation, adhesion, and migration assays were carried out using the αvß3-antagonist RGDechi-hCit. RESULTS: Proliferation was not significantly inhibited by RGDechi-hCit, although striking morphological changes were detected in MM cell lines highly expressing αvß3. Conversely, assays on fibronectin-coated plates showed a significant RGDechi-hCit dose-dependent inhibitory effect on both adhesion and migration. CONCLUSION: The data demonstrate anti-adhesion and anti-migration, but not antiproliferative, activities of RGDechi-hCit against MM cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Integrin alphaVbeta3/antagonists & inhibitors , Melanoma/drug therapy , Peptides/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Humans , Integrin alphaVbeta3/analysis , Melanoma/pathology , Melanoma/secondary
11.
J Cell Biochem ; 114(3): 728-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23060074

ABSTRACT

Cyclin-dependent kinase 9 (Cdk9) is a serine-threonine kinase, involved in many cellular processes. The regulatory units of Cdk9 are the T family Cyclins (T1, T2) and Cyclin K. Cyclin T2 has two forms termed Cyclin T2a and Cyclin T2b that arise by an alternative splicing of the primary transcript. Upon induction of muscle differentiation, MyoD recruits Cdk9/Cyclin T2 on muscle-specific gene promoter sequences. This complex is able to phosphorylate the C-terminal domain of RNA polymerase II, enhancing MyoD function and promoting myogenic differentiation. This work focuses on the characterization of two murine Cyclin T2 isoforms and the evaluation of the role of Cdk9/Cyclin T2 complexes during the skeletal muscle differentiation. This study demonstrated a predominant expression of isoform b in all stages of differentiation. Moreover, both isoforms of Cyclin T2 are able to activate the myogenic program but Cyclin T2b has a predominant role, in particular during the latest stages.


Subject(s)
Cell Differentiation/genetics , Cyclin T/genetics , Cyclin T/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Alternative Splicing , Animals , Base Sequence , Cell Division , Cells, Cultured , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation, Developmental , Mice , Muscle, Skeletal/embryology , MyoD Protein/metabolism , Myoblasts , Phosphorylation , Promoter Regions, Genetic , Protein Isoforms/genetics , RNA Polymerase II/metabolism , Sequence Analysis, DNA , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...