Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Cell ; 142(2): 230-42, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20655466

ABSTRACT

Human telomeres are protected from DNA damage by a nucleoprotein complex that includes the repeat-binding factor TRF2. Here, we report that TRF2 regulates the 5' exonuclease activity of its binding partner, Apollo, a member of the metallo-beta-lactamase family that is required for telomere integrity during S phase. TRF2 and Apollo also suppress damage to engineered interstitial telomere repeat tracts that were inserted far away from chromosome ends. Genetic data indicate that DNA topoisomerase 2alpha acts in the same pathway of telomere protection as TRF2 and Apollo. Moreover, TRF2, which binds preferentially to positively supercoiled DNA substrates, together with Apollo, negatively regulates the amount of TOP1, TOP2alpha, and TOP2beta at telomeres. Our data are consistent with a model in which TRF2 and Apollo relieve topological stress during telomere replication. Our work also suggests that cellular senescence may be caused by topological problems that occur during the replication of the inner portion of telomeres.


Subject(s)
Antigens, Neoplasm/metabolism , DNA Repair Enzymes/metabolism , DNA Replication , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Cellular Senescence , DNA Damage , Exodeoxyribonucleases , Humans , Protein Structure, Tertiary
2.
Int J Mol Sci ; 23(7)2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35409143

ABSTRACT

Telomeres are crucial structures that preserve genome stability. Their progressive erosion over numerous DNA duplications determines the senescence of cells and organisms. As telomere length homeostasis is critical for cancer development, nowadays, telomere maintenance mechanisms are established targets in cancer treatment. Besides telomere elongation, telomere dysfunction impinges on intracellular signaling pathways, in particular DNA damage signaling and repair, affecting cancer cell survival and proliferation. This review summarizes and discusses recent findings in anticancer drug development targeting different "telosome" components.


Subject(s)
Neoplasms , Telomerase , Cellular Senescence , DNA/metabolism , DNA Damage , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis
3.
Nucleic Acids Res ; 47(7): 3365-3382, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30698737

ABSTRACT

The telomeric protein TRF2 is overexpressed in several human malignancies and contributes to tumorigenesis even though the molecular mechanism is not completely understood. By using a high-throughput approach based on the multiplexed Luminex X-MAP technology, we demonstrated that TRF2 dramatically affects VEGF-A level in the secretome of cancer cells, promoting endothelial cell-differentiation and angiogenesis. The pro-angiogenic effect of TRF2 is independent from its role in telomere capping. Instead, TRF2 binding to a distal regulatory element promotes the expression of SULF2, an endoglucosamine-6-sulfatase that impairs the VEGF-A association to the plasma membrane by inducing post-synthetic modification of heparan sulfate proteoglycans (HSPGs). Finally, we addressed the clinical relevance of our findings showing that TRF2/SULF2 expression is a worse prognostic biomarker in colorectal cancer (CRC) patients.


Subject(s)
Colonic Neoplasms/metabolism , Sulfotransferases/genetics , Telomeric Repeat Binding Protein 2/metabolism , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/metabolism , Heparin/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic , Sulfatases , Sulfotransferases/biosynthesis , Telomeric Repeat Binding Protein 2/deficiency , Xenograft Model Antitumor Assays
4.
Int J Mol Sci ; 22(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34638655

ABSTRACT

DNA G-quadruplex (G4) structures, either within gene promoter sequences or at telomeres, have been extensively investigated as potential small-molecule therapeutic targets. However, although G4s forming at the telomeric DNA have been extensively investigated as anticancer targets, few studies focus on the telomeric repeat-containing RNA (TERRA), transcribed from telomeres, as potential pharmacological targets. Here, a virtual screening approach to identify a library of drug-like putative TERRA G4 binders, in tandem with circular dichroism melting assay to study their TERRA G4-stabilizing properties, led to the identification of a new hit compound. The affinity of this compound for TERRA RNA and some DNA G4s was analyzed through several biophysical techniques and its biological activity investigated in terms of antiproliferative effect, DNA damage response (DDR) activation, and TERRA RNA expression in high vs. low TERRA-expressing human cancer cells. The selected hit showed good affinity for TERRA G4 and no binding to double-stranded DNA. In addition, biological assays showed that this compound is endowed with a preferential cytotoxic effect on high TERRA-expressing cells, where it induces a DDR at telomeres, probably by displacing TERRA from telomeres. Our studies demonstrate that the identification of TERRA G4-targeting drugs with potential pharmacological effects is achievable, shedding light on new perspectives aimed at discovering new anticancer agents targeting these G4 structures.


Subject(s)
RNA/genetics , Telomere/genetics , Antineoplastic Agents/pharmacology , Binding Sites/drug effects , Binding Sites/genetics , DNA/genetics , DNA Damage/drug effects , DNA Damage/genetics , G-Quadruplexes/drug effects , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/genetics , Structure-Activity Relationship , Telomere/drug effects
5.
Chemistry ; 25(47): 11085-11097, 2019 Aug 22.
Article in English | MEDLINE | ID: mdl-31219221

ABSTRACT

Naphthalene diimide (NDI) dyads exhibiting a different substitution pattern and linker length have been synthesised and evaluated as G-quadruplex (G4) ligands, by investigating their cytotoxicity in selected cell lines. The dyads with the long C7 linker exhibit extremely low IC50 values, below 10 nm, on different cancer cell lines. Contrary, the dyads with the shorter C4 linker were much less effective, with IC values increasing up to 1 µm. Among the three dyads with the longest linker, small differences in the IC50 values emerge, suggesting that the linker length plays a more important role than the substitution pattern. We have further shown that the dyads are able to induce cellular DNA damage response, which is not limited to the telomeric regions and is likely the origin of their cytotoxicity. Both absorption titration and dynamic light scattering of the most cytotoxic dyads in the presence of hTel22 highlight their ability to induce effective G4 aggregation, acting as non-covalent cross-linking agents.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , G-Quadruplexes , Imides/pharmacology , Naphthalenes/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Imides/chemical synthesis , Imides/chemistry , Ligands , Metaphase/drug effects , Microscopy, Fluorescence , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Telomere/drug effects , Telomere/metabolism
6.
Nucleic Acids Res ; 45(4): 1820-1834, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27923994

ABSTRACT

Telomere repeat binding factor 2 (TRF2) has been increasingly recognized to be involved in telomere maintenance and DNA damage response. Here, we show that TRF2 directly binds SIRT6 in a DNA independent manner and that this interaction is increased upon replication stress. Knockdown of SIRT6 up-regulates TRF2 protein levels and counteracts its down-regulation during DNA damage response, leading to cell survival. Moreover, we report that SIRT6 deactetylates in vivo the TRFH domain of TRF2, which in turn, is ubiquitylated in vivo activating the ubiquitin-dependent proteolysis. Notably, overexpression of the TRF2cT mutant failed to be stabilized by SIRT6 depletion, demonstrating that the TRFH domain is required for its post-transcriptional modification. Finally, we report an inverse correlation between SIRT6 and TRF2 protein expression levels in a cohort of colon rectal cancer patients. Taken together our findings describe TRF2 as a novel SIRT6 substrate and demonstrate that acetylation of TRF2 plays a crucial role in the regulation of TRF2 protein stability, thus providing a new route for modulating its expression level during oncogenesis and damage response.


Subject(s)
DNA Damage , Sirtuins/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Acetylation , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Cell Line , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans , Immunohistochemistry , Models, Molecular , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Conformation , Protein Stability , Proteolysis/drug effects , Recombinant Fusion Proteins/metabolism , Sirtuins/chemistry , Substrate Specificity , Telomeric Repeat Binding Protein 2/chemistry , Telomeric Repeat Binding Protein 2/genetics , Ubiquitination
7.
Nucleic Acids Res ; 44(4): 1579-90, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26511095

ABSTRACT

Cancer stem cells (CSCs) have been identified in several solid malignancies and are now emerging as a plausible target for drug discovery. Beside the questionable existence of CSCs specific markers, the expression of CD133 was reported to be responsible for conferring CSC aggressiveness. Here, we identified two G-rich sequences localized within the introns 3 and 7 of the CD133 gene able to form G-quadruplex (G4) structures, bound and stabilized by small molecules. We further showed that treatment of patient-derived colon CSCs with G4-interacting agents triggers alternative splicing that dramatically impairs the expression of CD133. Interestingly, this is strongly associated with a loss of CSC properties, including self-renewing, motility, tumor initiation and metastases dissemination. Notably, the effects of G4 stabilization on some of these CSC properties are uncoupled from DNA damage response and are fully recapitulated by the selective interference of the CD133 expression.In conclusion, we provided the first proof of the existence of G4 structures within the CD133 gene that can be pharmacologically targeted to impair CSC aggressiveness. This discloses a class of potential antitumoral agents capable of targeting the CSC subpopulation within the tumoral bulk.


Subject(s)
Antigens, CD/genetics , G-Quadruplexes , Glycoproteins/genetics , Neoplasm Invasiveness/genetics , Neoplastic Stem Cells/metabolism , Peptides/genetics , AC133 Antigen , Antigens, CD/chemistry , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glycoproteins/chemistry , Humans , Neoplastic Stem Cells/pathology , Peptides/chemistry , Protein Biosynthesis
8.
Nucleic Acids Res ; 43(3): 1759-69, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25618850

ABSTRACT

Here, with the aim of obtaining insight into the intriguing selectivity of G-quadruplex (G4) ligands toward cancer compared to normal cells, a genetically controlled system of progressive transformation in human BJ fibroblasts was analyzed. Among the different comparative evaluations, we found a progressive increase of DNA damage response (DDR) markers throughout the genome from normal toward immortalized and transformed cells. More interestingly, sensitivity to G4 ligands strongly correlated with the presence of a basal level of DNA damage, including at the telomeres, where the chromosome ends were exposed to the DDR without concurrent induction of DNA repair activity, as revealed by the lack of 53BP1 recruitment and telomere aberrations. The link between telomere uncapping and the response to G4 stabilization was directly assessed by showing that a partial TRF2 depletion, causing a basal level of telomere localized DDR, rendered telomerized fibroblasts prone to G4-induced telomere damage and anti-proliferative defects. Taken together these data strongly indicate that the presence of a basal level of telomere-associated DDR is a determinant of susceptibility to G4 stabilization.


Subject(s)
DNA Damage , G-Quadruplexes/drug effects , Neoplasms/genetics , Telomere , Blotting, Western , Chromatin Immunoprecipitation , Humans , In Situ Hybridization, Fluorescence , Tumor Cells, Cultured
9.
Org Biomol Chem ; 14(30): 7238-49, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27383473

ABSTRACT

We report on the potential of a water-soluble tetracationic quaternary ammonium naphthalene diimide (NDI) as multifunctional agent of interest for theranostic applications. The DNA binding ability of this NDI has been investigated. NDI exhibits high binding constants for G-quadruplex DNA but it is not selective for this type of DNA. Taking advantage of its intrinsic fluorescence and singlet oxygen sensitizing ability, cellular uptake, cytotoxicity and photocytotoxicity have been investigated. The intense emission in the red/NIR allows monitoring of the cell permeability of this charged tetracationic NDI, accumulating into the cell nuclei. No dark cytotoxicity has been observed on selected tumor cell lines. Irradiation of the NDI loaded cells with red light reduces cell viability up to 40% and causes a significant increase of the percentage of cells expressing γH2AX foci indicating DNA damage. The presence of distinct DNA damage foci inside the nucleus suggests that the NDI molecule might induce DNA damage in specific sites. To the best of our knowledge this is the first NDI exhibiting PDT activity at µM concentration combined with low dark cytotoxicity.


Subject(s)
Fluorescent Dyes/chemistry , Fluorescent Dyes/toxicity , Imides/chemistry , Imides/toxicity , Naphthalenes/chemistry , Naphthalenes/toxicity , Theranostic Nanomedicine/methods , Biological Transport , Cell Line, Tumor , Cell Survival , Differential Thermal Analysis/methods , Fluorescent Dyes/metabolism , G-Quadruplexes , Humans , Imides/metabolism , Molecular Structure , Naphthalenes/metabolism , Optical Imaging , Photochemical Processes , Singlet Oxygen/chemistry , Structure-Activity Relationship , Thermodynamics
10.
Nucleic Acids Res ; 42(5): 2945-57, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24335081

ABSTRACT

Tumor angiogenesis is mainly mediated by vascular endothelial growth factor (VEGF), a pro-angiogenic factor produced by cancer cells and active on the endothelium through the VEGF receptor 2 (VEGFR-2). Here we identify a G-rich sequence within the proximal promoter region of vegfr-2, able to form an antiparallel G-quadruplex (G4) structure. This G4 structure can be efficiently stabilized by small molecules with the consequent inhibition of vegfr-2 expression. Functionally, the G4-mediated reduction of VEGFR-2 protein causes a switching off of signaling components that, converging on actin cytoskeleton, regulate the cellular events leading to endothelial cell proliferation, migration and differentiation. As a result of endothelial cell function impairment, angiogenic process is strongly inhibited by G4 ligands both in vitro and in vivo. Interestingly, the G4-mediated antiangiogenic effect seems to recapitulate that observed by using a specific interference RNA against vegfr-2, and it is strongly antagonized by overexpressing the vegfr-2 gene. In conclusion, we describe the evidence for the existence of G4 in the promoter of vegfr-2, whose expression and function can be markedly inhibited by G4 ligands, thereby revealing a new, and so far undescribed, way to block VEGFR-2 as target for anticancer therapy.


Subject(s)
G-Quadruplexes , Neoplasms/blood supply , Neovascularization, Pathologic , Promoter Regions, Genetic , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Human Umbilical Vein Endothelial Cells/physiology , Humans , Ligands , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
J Am Chem Soc ; 136(48): 16708-11, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25393214

ABSTRACT

The shelterin protein TRF2 has come to the limelight for its role in telomere maintenance and tumorigenesis. Herein, the application of rational design and synthesis allowed identifying the first TRF2TRFH binder able to elicit a marked DNA damage response in cancer cells. This work paves the way for the unprecedented employment of a chemical tool to finely tune specific mechanisms underlying telomere maintenance.


Subject(s)
Drug Design , Peptides, Cyclic/pharmacology , Telomeric Repeat Binding Protein 2/antagonists & inhibitors , Humans , Models, Molecular , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Telomeric Repeat Binding Protein 2/metabolism
12.
Cell Death Dis ; 15(5): 358, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777849

ABSTRACT

Recruitment of fibroblasts to tumors and their activation into cancer-associated fibroblasts (CAFs) is a strategy used by tumor cells to direct extracellular matrix (ECM) remodeling, invasion, and metastasis, highlighting the need to investigate the molecular mechanisms driving CAF function. Endothelin-1 (ET-1) regulates the communication between cancer and stroma and facilitates the progression of serous ovarian cancer (SOC). By binding to Endothelin A (ETA) and B (ETB) receptors, ET-1 enables the recruitment of ß-arrestin1 (ß-arr1) and the formation of signaling complexes that coordinate tumor progression. However, how ET-1 receptors might "educate" human ovarian fibroblasts (HOFs) to produce altered ECM and promote metastasis remains to be elucidated. This study identifies ET-1 as a pivotal factor in the activation of CAFs capable of proteolytic ECM remodeling and the generation of heterotypic spheroids containing cancer cells with a propensity to metastasize. An autocrine/paracrine ET-1/ETA/BR/ß-arr1 loop enhances HOF proliferation, upregulates CAF marker expression, secretes pro-inflammatory cytokines, and increases collagen contractility, and cell motility. Furthermore, ET-1 facilitates ECM remodeling by promoting the lytic activity of invadosome and activation of integrin ß1. In addition, ET-1 signaling supports the formation of heterotypic HOF/SOC spheroids with enhanced ability to migrate through the mesothelial monolayer, and invade, representing metastatic units. The blockade of ETA/BR or ß-arr1 silencing prevents CAF activation, invadosome function, mesothelial clearance, and the invasive ability of heterotypic spheroids. In vivo, therapeutic inhibition of ETA/BR using bosentan (BOS) significantly reduces the metastatic potential of combined HOFs/SOC cells, associated with enhanced apoptotic effects on tumor cells and stromal components. These findings support a model in which ET-1/ß-arr1 reinforces tumor/stroma interaction through CAF activation and fosters the survival and metastatic properties of SOC cells, which could be counteracted by ETA/BR antagonists.


Subject(s)
Cancer-Associated Fibroblasts , Ovarian Neoplasms , Podosomes , beta-Arrestin 1 , Humans , Female , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , beta-Arrestin 1/metabolism , beta-Arrestin 1/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Podosomes/metabolism , Endothelin-1/metabolism , Neoplasm Metastasis , Receptor, Endothelin A/metabolism , Signal Transduction , Extracellular Matrix/metabolism , Cell Movement , Cell Proliferation , Animals , Fibroblasts/metabolism , Neoplasm Invasiveness
13.
Methods ; 57(1): 93-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22410593

ABSTRACT

The burgeoning knowledge about the structure of telomeres and the roles of various factors involved in telomere maintenance provides several possible targets for pharmacological intervention. To date the area that has received major attention regarding drug discovery is the targeting the telomeric G-quadruplex (G4) structure. G4 ligands were initially designed to counteract telomerase action at telomeres. Surprisingly, their antiproliferative effects can occur in telomerase negative cells and follow kinetics, which cannot be merely explained by telomere shortening, suggesting that these compounds affect other pathways, not necessarily related to telomere biology. Impressively, it has been shown that polyaromatic compounds featuring end-stacking binding properties trigger a strong DNA damage response at telomeres. This is typical of the telomere deprotection occurring during cellular senescence or upon telomere injury. It emerged that the G4-interacting agents are more than simple telomerase inhibitors and that their direct target is rather telomere than telomerase. This review summarizes the most valid experimental approaches for studying the pharmacological telomere damage induced by G4-ligand complexes.


Subject(s)
DNA , G-Quadruplexes , Ligands , Telomerase , Telomere , Cellular Senescence/drug effects , DNA/chemistry , DNA/pharmacology , DNA Damage/drug effects , Humans , Telomerase/antagonists & inhibitors , Telomerase/chemistry , Telomere/chemistry , Telomere/drug effects
14.
Aging Cell ; 22(11): e13944, 2023 11.
Article in English | MEDLINE | ID: mdl-37858982

ABSTRACT

Drug repositioning strategy represents a valid tool to accelerate the pharmacological development through the identification of new applications for already existing compounds. In this view, we aimed at discovering molecules able to trigger telomere-localized DNA damage and tumor cell death. By applying an automated high-content spinning-disk microscopy, we performed a screening aimed at identifying, on a library of 527 drugs, molecules able to negatively affect the expression of TRF2, a key protein in telomere maintenance. FK866, resulting from the screening as the best candidate hit, was then validated at biochemical and molecular levels and the mechanism underlying its activity in telomere deprotection was elucidated both in vitro and in vivo. The results of this study allow us to discover a novel role of FK866 in promoting, through the production of reactive oxygen species, telomere loss and deprotection, two events leading to an accumulation of DNA damage and tumor cell death. The ability of FK866 to induce telomere damage and apoptosis was also demonstrated in advanced preclinical models evidencing the antitumoral activity of FK866 in triple-negative breast cancer-a particularly aggressive breast cancer subtype still orphan of targeted therapies and characterized by high expression levels of both NAMPT and TRF2. Overall, our findings pave the way to the development of novel anticancer strategies to counteract triple-negative breast cancer, based on the use of telomere deprotecting agents, including NAMPT inhibitors, that would rapidly progress from bench to bedside.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Drug Repositioning , Cell Death , Apoptosis , Telomere , Telomeric Repeat Binding Protein 2/genetics , Cell Line, Tumor
15.
Proc Natl Acad Sci U S A ; 106(8): 2806-11, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19202075

ABSTRACT

The activation of endothelin-A receptor (ET(A)R) by endothelin-1 (ET-1) has a critical role in ovarian tumorigenesis and progression. To define the molecular mechanism in ET-1-induced tumor invasion and metastasis, we focused on beta-arrestins as scaffold and signaling proteins of G protein-coupled receptors. Here, we demonstrate that, in ovarian cancer cells, beta-arrestin is recruited to ET(A)R to form two trimeric complexes: one through the interaction with Src leading to epithelial growth factor receptor (EGFR) transactivation and beta-catenin Tyr phosphorylation, and the second through the physical association with axin, contributing to release and inactivation of glycogen synthase kinase (GSK)-3beta and beta-catenin stabilization. The engagement of beta-arrestin in these two signaling complexes concurs to activate beta-catenin signaling pathways. We then demonstrate that silencing of both beta-arrestin-1 and beta-arrestin-2 inhibits ET(A)R-driven signaling, causing suppression of Src, mitogen-activated protein kinase (MAPK), AKT activation, as well as EGFR transactivation and a complete inhibition of ET-1-induced beta-catenin/TCF transcriptional activity and cell invasion. ET(A)R blockade with the specific ET(A)R antagonist ZD4054 abrogates the engagement of beta-arrestin in the interplay between ET(A)R and the beta-catenin pathway in the invasive program. Finally, ET(A)R is expressed in 85% of human ovarian cancers and is preferentially co-expressed with beta-arrestin-1 in the advanced tumors. In a xenograft model of ovarian metastasis, HEY cancer cells expressing beta-arrestin-1 mutant metastasize at a reduced rate, highlighting the importance of this molecule in promoting metastases. ZD4054 treatment significantly inhibits metastases, suggesting that specific ET(A)R antagonists, by disabling multiple signaling activated by ET(A)R/beta-arrestin, may represent new therapeutic opportunities for ovarian cancer.


Subject(s)
Arrestins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Ovarian Neoplasms/pathology , Receptor, Endothelin A/metabolism , Signal Transduction , beta Catenin/metabolism , Blotting, Western , Cell Line, Tumor , ErbB Receptors/genetics , Female , Humans , Microscopy, Fluorescence , Ovarian Neoplasms/metabolism , Phosphorylation , Transcriptional Activation , Transplantation, Heterologous , Tyrosine/metabolism , beta Catenin/chemistry , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
16.
Front Oncol ; 12: 862806, 2022.
Article in English | MEDLINE | ID: mdl-35719951

ABSTRACT

Purpose: Tumor-microenvironment interactions are important determinants of drug resistance in colorectal cancer (CRC). We, therefore, set out to understand how interactions between genetically characterized CRC cells and stromal fibroblasts might influence response to molecularly targeted inhibitors. Techniques: Sensitivity to PI3K/AKT/mTOR pathway inhibitors of CRC cell lines, with known genetic background, was investigated under different culture conditions [serum-free medium, fibroblasts' conditioned medium (CM), direct co-culture]. Molecular pathway activation was monitored using Western Blot analysis. Immunoprecipitation was used to detect specific mTOR complex activation. Immunofluorescence was used to analyze cellular PTEN distribution, while different mutant PTEN plasmids were used to map the observed function to specific PTEN protein domains. Results: Exposure to fibroblast-CM resulted in increased growth-inhibitory response to double PI3K/mTOR inhibitors in PTEN-competent CRC cell lines harboring KRAS and PI3K mutations. Such functional effect was attributable to fibroblast-CM induced paradoxical PI3K/mTORC1 pathway activation, occurring in the presence of a functional PTEN protein. At a molecular level, fibroblast-CM induced C-tail phosphorylation and cytoplasmic redistribution of the PTEN protein, thereby impairing its lipid phosphatase function and favored the formation of active, RAPTOR-containing, mTORC1 complexes. However, PTEN's lipid phosphatase function appeared to be dispensable, while complex protein-protein interactions, also involving PTEN/mTOR co-localization and subcellular distribution, were crucial for both mTORC1 activation and sensitivity to double PI3K/mTOR inhibitors. Data Interpretation: Microenvironmental cues, in particular soluble factors produced by stromal fibroblasts, profoundly influence PI3K pathway signaling and functional response to specific inhibitors in CRC cells, depending on their mutational background and PTEN status.

17.
Chem Commun (Camb) ; 58(85): 11913-11916, 2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36196950

ABSTRACT

The single-stranded RNA genome of SARS-CoV-2 contains some G-quadruplex-forming G-rich elements which are putative drug targets. Here, we performed a ligand-based pharmacophore virtual screening of FDA approved drugs to find candidates targeting such RNA structures. Further in silico and in vitro assays identified three drugs as emerging SARS-CoV-2 RNA G-quadruplex binders.


Subject(s)
COVID-19 Drug Treatment , Drug Repositioning , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Ligands , Molecular Docking Simulation , RNA, Viral/genetics , SARS-CoV-2 , G-Quadruplexes
18.
Nucleic Acids Res ; 37(16): 5353-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19596811

ABSTRACT

Functional telomeres are required to maintain the replicative ability of cancer cells and represent putative targets for G-quadruplex (G4) ligands. Here, we show that the pentacyclic acridinium salt RHPS4, one of the most effective and selective G4 ligands, triggers damages in cells traversing S phase by interfering with telomere replication. Indeed, we found that RHPS4 markedly reduced BrdU incorporation at telomeres and altered the dynamic association of the telomeric proteins TRF1, TRF2 and POT1, leading to chromosome aberrations such as telomere fusions and telomere doublets. Analysis of the molecular damage pathway revealed that RHPS4 induced an ATR-dependent ATM signaling that plays a functional role in the cellular response to RHPS4 treatment. We propose that RHPS4, by stabilizing G4 DNA at telomeres, impairs fork progression and/or telomere processing resulting in telomere dysfunction and activation of a replication stress response pathway. The detailed understanding of the molecular mode of action of this class of compounds makes them attractive tools to understand telomere biology and provides the basis for a rational use of G4 ligands for the therapy of cancer.


Subject(s)
Acridines/pharmacology , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , G-Quadruplexes/drug effects , Protein Serine-Threonine Kinases/metabolism , Telomere/drug effects , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Line , DNA Damage , DNA Replication/drug effects , Humans , Ligands , Signal Transduction , Telomere/chemistry , Telomere/metabolism
19.
J Clin Invest ; 117(11): 3236-47, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17932567

ABSTRACT

Functional telomeres are required for the replicability of cancer cells. The G-rich strand of telomeric DNA can fold into a 4-stranded structure known as the G-quadruplex (G4), whose stabilization alters telomere function limiting cancer cell growth. Therefore, the G4 ligand RHPS4 may possess antitumor activity. Here, we show that RHPS4 triggers a rapid and potent DNA damage response at telomeres in human transformed fibroblasts and melanoma cells, characterized by the formation of several telomeric foci containing phosphorylated DNA damage response factors gamma-H2AX, RAD17, and 53BP1. This was dependent on DNA repair enzyme ATR, correlated with delocalization of the protective telomeric DNA-binding protein POT1, and was antagonized by overexpression of POT1 or TRF2. In mice, RHPS4 exerted its antitumor effect on xenografts of human tumor cells of different histotype by telomere injury and tumor cell apoptosis. Tumor inhibition was accompanied by a strong DNA damage response, and tumors overexpressing POT1 or TRF2 were resistant to RHPS4 treatment. These data provide evidence that RHPS4 is a telomere damage inducer and that telomere disruption selectively triggered in malignant cells results in a high therapeutic index in mice. They also define a functional link between telomere damage and antitumor activity and reveal the key role of telomere-protective factors TRF2 and POT1 in response to this anti-telomere strategy.


Subject(s)
Acridines/metabolism , Antineoplastic Agents/metabolism , DNA Damage , G-Quadruplexes , Telomere/pathology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Repair , Histones/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neoplasm Transplantation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Shelterin Complex , Telomere/genetics , Telomere/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Transplantation, Heterologous , Tumor Suppressor p53-Binding Protein 1
20.
Genes (Basel) ; 11(3)2020 03 13.
Article in English | MEDLINE | ID: mdl-32183119

ABSTRACT

Most human tumors maintain telomere lengths by telomerase, whereas a portion of them (10%-15%) uses a mechanism named alternative lengthening of telomeres (ALT). The telomeric G-quadruplex (G4) ligand RHPS4 is known for its potent antiproliferative effect, as shown in telomerase-positive cancer models. Moreover, RHPS4 is also able to reduce cell proliferation in ALT cells, although the influence of G4 stabilization on the ALT mechanism has so far been poorly investigated. Here we show that sensitivity to RHPS4 is comparable in ALT-positive (U2OS; SAOS-2) and telomerase-positive (HOS) osteosarcoma cell lines, unlinking the telomere maintenance mechanism and RHPS4 responsiveness. To investigate the impact of G4 stabilization on ALT, the cardinal ALT hallmarks were analyzed. A significant induction of telomeric doublets, telomeric clusterized DNA damage, ALT-associated Promyelocytic Leukaemia-bodies (APBs), telomere sister chromatid exchanges (T-SCE) and c-circles was found exclusively in RHPS4-treated ALT cells. We surmise that RHPS4 affects ALT mechanisms through the induction of replicative stress that in turn is converted in DNA damage at telomeres, fueling recombination. In conclusion, our work indicates that RHPS4-induced telomeric DNA damage promotes overactivation of telomeric recombination in ALT cells, opening new questions on the therapeutic employment of G4 ligands in the treatment of ALT positive tumors.


Subject(s)
G-Quadruplexes , Osteosarcoma/genetics , Telomere Homeostasis/genetics , Telomere/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DNA Damage/genetics , DNA Replication/genetics , Humans , Osteosarcoma/pathology , Signal Transduction/genetics , Telomerase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL