Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(4)2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36835480

RESUMEN

Aiming to identify highly effective and selective G-quadruplex ligands as anticancer candidates, five natural compounds were investigated here, i.e., the alkaloids Canadine, D-Glaucine and Dicentrine, as well as the flavonoids Deguelin and Millettone, selected as analogs of compounds previously identified as promising G-quadruplex-targeting ligands. A preliminary screening with the G-quadruplex on the Controlled Pore Glass assay proved that, among the investigated compounds, Dicentrine is the most effective ligand of telomeric and oncogenic G-quadruplexes, also showing good G-quadruplex vs. duplex selectivity. In-depth studies in solution demonstrated the ability of Dicentrine to thermally stabilize telomeric and oncogenic G-quadruplexes without affecting the control duplex. Interestingly, it showed higher affinity for the investigated G-quadruplex structures over the control duplex (Kb~106 vs. 105 M-1), with some preference for the telomeric over the oncogenic G-quadruplex model. Molecular dynamics simulations indicated that Dicentrine preferentially binds the G-quadruplex groove or the outer G-tetrad for the telomeric and oncogenic G-quadruplexes, respectively. Finally, biological assays proved that Dicentrine is highly effective in promoting potent and selective anticancer activity by inducing cell cycle arrest through apoptosis, preferentially targeting G-quadruplex structures localized at telomeres. Taken together, these data validate Dicentrine as a putative anticancer candidate drug selectively targeting cancer-related G-quadruplex structures.


Asunto(s)
Antineoplásicos , G-Cuádruplex , Neoplasias , Humanos , Ligandos , Simulación de Dinámica Molecular , Antineoplásicos/farmacología , Telómero/metabolismo
2.
Immunity ; 39(2): 372-85, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23973224

RESUMEN

Endogenous tryptophan (Trp) metabolites have an important role in mammalian gut immune homeostasis, yet the potential contribution of Trp metabolites from resident microbiota has never been addressed experimentally. Here, we describe a metabolic pathway whereby Trp metabolites from the microbiota balance mucosal reactivity in mice. Switching from sugar to Trp as an energy source (e.g., under conditions of unrestricted Trp availability), highly adaptive lactobacilli are expanded and produce an aryl hydrocarbon receptor (AhR) ligand-indole-3-aldehyde-that contributes to AhR-dependent Il22 transcription. The resulting IL-22-dependent balanced mucosal response allows for survival of mixed microbial communities yet provides colonization resistance to the fungus Candida albicans and mucosal protection from inflammation. Thus, the microbiota-AhR axis might represent an important strategy pursued by coevolutive commensalism for fine tuning host mucosal reactivity contingent on Trp catabolism.


Asunto(s)
Candida albicans/inmunología , Interleucinas/metabolismo , Limosilactobacillus reuteri/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Triptófano/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Candidiasis/inmunología , Metabolismo Energético , Femenino , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Indolamina-Pirrol 2,3,-Dioxigenasa/deficiencia , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indoles/metabolismo , Interleucina-17/deficiencia , Interleucina-17/genética , Limosilactobacillus reuteri/crecimiento & desarrollo , Limosilactobacillus reuteri/inmunología , Metagenoma , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Probióticos , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/genética , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Triptófano/química , Interleucina-22
3.
Nucleic Acids Res ; 47(7): 3365-3382, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30698737

RESUMEN

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.


Asunto(s)
Neoplasias del Colon/metabolismo , Sulfotransferasas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Proteoglicanos de Heparán Sulfato/química , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Neovascularización Patológica , Sulfatasas , Sulfotransferasas/biosíntesis , Proteína 2 de Unión a Repeticiones Teloméricas/deficiencia , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Chemistry ; 25(47): 11085-11097, 2019 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-31219221

RESUMEN

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.


Asunto(s)
Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , G-Cuádruplex , Imidas/farmacología , Naftalenos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Imidas/síntesis química , Imidas/química , Ligandos , Metafase/efectos de los fármacos , Microscopía Fluorescente , Naftalenos/síntesis química , Naftalenos/química , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Telómero/efectos de los fármacos , Telómero/metabolismo
5.
Nucleic Acids Res ; 45(4): 1820-1834, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-27923994

RESUMEN

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.


Asunto(s)
Daño del ADN , Sirtuinas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Acetilación , Antineoplásicos Fitogénicos/farmacología , Camptotecina/farmacología , Línea Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Humanos , Inmunohistoquímica , Modelos Moleculares , Poli(ADP-Ribosa) Polimerasas/metabolismo , Unión Proteica , Conformación Proteica , Estabilidad Proteica , Proteolisis/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Sirtuinas/química , Especificidad por Sustrato , Proteína 2 de Unión a Repeticiones Teloméricas/química , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Ubiquitinación
6.
Nucleic Acids Res ; 44(4): 1579-90, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26511095

RESUMEN

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.


Asunto(s)
Antígenos CD/genética , G-Cuádruplex , Glicoproteínas/genética , Invasividad Neoplásica/genética , Células Madre Neoplásicas/metabolismo , Péptidos/genética , Antígeno AC133 , Antígenos CD/química , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/química , Humanos , Células Madre Neoplásicas/patología , Péptidos/química , Biosíntesis de Proteínas
7.
Nucleic Acids Res ; 43(3): 1759-69, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25618850

RESUMEN

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.


Asunto(s)
Daño del ADN , G-Cuádruplex/efectos de los fármacos , Neoplasias/genética , Telómero , Western Blotting , Inmunoprecipitación de Cromatina , Humanos , Hibridación Fluorescente in Situ , Células Tumorales Cultivadas
8.
Org Biomol Chem ; 14(30): 7238-49, 2016 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-27383473

RESUMEN

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.


Asunto(s)
Colorantes Fluorescentes/química , Colorantes Fluorescentes/toxicidad , Imidas/química , Imidas/toxicidad , Naftalenos/química , Naftalenos/toxicidad , Nanomedicina Teranóstica/métodos , Transporte Biológico , Línea Celular Tumoral , Supervivencia Celular , Análisis Diferencial Térmico/métodos , Colorantes Fluorescentes/metabolismo , G-Cuádruplex , Humanos , Imidas/metabolismo , Estructura Molecular , Naftalenos/metabolismo , Imagen Óptica , Procesos Fotoquímicos , Oxígeno Singlete/química , Relación Estructura-Actividad , Termodinámica
9.
Nucleic Acids Res ; 42(5): 2945-57, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24335081

RESUMEN

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.


Asunto(s)
G-Cuádruplex , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Regiones Promotoras Genéticas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neovascularización Fisiológica , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Blood ; 119(4): 967-77, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22147891

RESUMEN

Aspergillus fumigatus is a model fungal pathogen and a common cause of severe infections and diseases. CD8⁺ T cells are present in the human and murine T-cell repertoire to the fungus. However, CD8⁺ T-cell function in infection and the molecular mechanisms that control their priming and differentiation into effector and memory cells in vivo remain elusive. In the present study, we report that both CD4⁺ and CD8⁺ T cells mediate protective memory responses to the fungus contingent on the nature of the fungal vaccine. Mechanistically, class I MHC-restricted, CD8⁺ memory T cells were activated through TLR3 sensing of fungal RNA by cross-presenting dendritic cells. Genetic deficiency of TLR3 was associated with susceptibility to aspergillosis and concomitant failure to activate memory-protective CD8⁺ T cells both in mice and in patients receiving stem-cell transplantations. Therefore, TLR3 essentially promotes antifungal memory CD8⁺ T-cell responses and its deficiency is a novel susceptibility factor for aspergillosis in high-risk patients.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Linfocitos T CD8-positivos/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Huésped Inmunocomprometido , Memoria Inmunológica , Receptor Toll-Like 3/metabolismo , Animales , Presentación de Antígeno , Antígenos Fúngicos/uso terapéutico , Aspergilosis/genética , Aspergilosis/prevención & control , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Estudios de Cohortes , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Susceptibilidad a Enfermedades , Femenino , Vacunas Fúngicas/uso terapéutico , Humanos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Organismos Libres de Patógenos Específicos , Receptor Toll-Like 3/genética
12.
Cancer Cell ; 10(6): 473-86, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17157788

RESUMEN

Che-1 is a RNA polymerase II-binding protein involved in the transcription of E2F target genes and induction of cell proliferation. Here we show that Che-1 contributes to DNA damage response and that its depletion sensitizes cells to anticancer agents. The checkpoint kinases ATM/ATR and Chk2 interact with Che-1 and promote its phosphorylation and accumulation in response to DNA damage. These Che-1 modifications induce a specific recruitment of Che-1 on the TP53 and p21 promoters. Interestingly, it has a profound effect on the basal expression of p53, which is preserved following DNA damage. Notably, Che-1 contributes to the maintenance of the G2/M checkpoint induced by DNA damage. These findings identify a mechanism by which checkpoint kinases regulate responses to DNA damage.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/fisiología , Genes p53 , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/fisiología , Animales , Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , División Celular , Quinasa de Punto de Control 2 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Fase G2 , Humanos , Ratones , Células 3T3 NIH , Fosforilación , Regiones Promotoras Genéticas , Transcripción Genética
13.
Nature ; 451(7175): 211-5, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-18185592

RESUMEN

Half a century ago, chronic granulomatous disease (CGD) was first described as a disease fatally affecting the ability of children to survive infections. Various milestone discoveries have since been made, from an insufficient ability of patients' leucocytes to kill microbes to the underlying genetic abnormalities. In this inherited disorder, phagocytes lack NADPH oxidase activity and do not generate reactive oxygen species, most notably superoxide anion, causing recurrent bacterial and fungal infections. Patients with CGD also suffer from chronic inflammatory conditions, most prominently granuloma formation in hollow viscera. The precise mechanisms of the increased microbial pathogenicity have been unclear, and more so the reasons for the exaggerated inflammatory response. Here we show that a superoxide-dependent step in tryptophan metabolism along the kynurenine pathway is blocked in CGD mice with lethal pulmonary aspergillosis, leading to unrestrained Vgamma1(+) gammadelta T-cell reactivity, dominant production of interleukin (IL)-17, defective regulatory T-cell activity and acute inflammatory lung injury. Although beneficial effects are induced by IL-17 neutralization or gammadelta T-cell contraction, complete cure and reversal of the hyperinflammatory phenotype are achieved by replacement therapy with a natural kynurenine distal to the blockade in the pathway. Effective therapy, which includes co-administration of recombinant interferon-gamma (IFN-gamma), restores production of downstream immunoactive metabolites and enables the emergence of regulatory Vgamma4(+) gammadelta and Foxp3(+) alphabeta T cells. Therefore, paradoxically, the lack of reactive oxygen species contributes to the hyperinflammatory phenotype associated with NADPH oxidase deficiencies, through a dysfunctional kynurenine pathway of tryptophan catabolism. Yet, this condition can be reverted by reactivating the pathway downstream of the superoxide-dependent step.


Asunto(s)
Enfermedad Granulomatosa Crónica/metabolismo , Enfermedad Granulomatosa Crónica/patología , Inflamación/metabolismo , Quinurenina/metabolismo , Triptófano/metabolismo , Animales , Aspergilosis/complicaciones , Aspergilosis/inmunología , Aspergilosis/microbiología , Aspergilosis/patología , Aspergillus fumigatus/fisiología , Enfermedad Crónica , Modelos Animales de Enfermedad , Enfermedad Granulomatosa Crónica/complicaciones , Enfermedad Granulomatosa Crónica/tratamiento farmacológico , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/patología , Interferón gamma/inmunología , Interferón gamma/uso terapéutico , Interleucina-17/deficiencia , Interleucina-17/metabolismo , Quinurenina/uso terapéutico , Pulmón/inmunología , Pulmón/patología , Enfermedades Pulmonares Fúngicas/complicaciones , Enfermedades Pulmonares Fúngicas/inmunología , Enfermedades Pulmonares Fúngicas/microbiología , Enfermedades Pulmonares Fúngicas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/deficiencia , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Superóxidos/metabolismo , Linfocitos T/enzimología , Linfocitos T/inmunología , Linfocitos T/patología
14.
J Exp Clin Cancer Res ; 43(1): 75, 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38459559

RESUMEN

BACKGROUND: Breast Cancer (BC) can be classified, due to its heterogeneity, into multiple subtypes that differ for prognosis and clinical management. Notably, triple negative breast cancer (TNBC) - the most aggressive BC form - is refractory to endocrine and most of the target therapies. In this view, taxane-based therapy still represents the elective strategy for the treatment of this tumor. However, due variability in patients' response, management of TNBC still represents an unmet medical need. Telomeric Binding Factor 2 (TRF2), a key regulator of telomere integrity that is over-expressed in several tumors, including TNBC, has been recently found to plays a role in regulating autophagy, a degradative process that is involved in drug detoxification. Based on these considerations, we pointed, here, at investigating if TRF2, regulating autophagy, can affect tumor sensitivity to therapy. METHODS: Human TNBC cell lines, over-expressing or not TRF2, were subjected to treatment with different taxanes and drug efficacy was tested in terms of autophagic response and cell proliferation. Autophagy was evaluated first biochemically, by measuring the levels of LC3, and then by immunofluorescence analysis of LC3-puncta positive cells. Concerning the proliferation, cells were subjected to colony formation assays associated with western blot and FACS analyses. The obtained results were then confirmed also in mouse models. Finally, the clinical relevance of our findings was established by retrospective analysis on a cohort of TNBC patients subjected to taxane-based neoadjuvant chemotherapy. RESULTS: This study demonstrated that TRF2, inhibiting autophagy, is able to increase the sensitivity of TNBC cells to taxanes. The data, first obtained in in vitro models, were then recapitulated in preclinical mouse models and in a cohort of TNBC patients, definitively demonstrating that TRF2 over-expression enhances the efficacy of taxane-based neoadjuvant therapy in reducing tumor growth and its recurrence upon surgical intervention. CONCLUSIONS: Based on our finding it is possible to conclude that TRF2, already known for its role in promoting tumor formation and progression, might represents an Achilles' heel for cancer. In this view, TRF2 might be exploited as a putative biomarker to predict the response of TNBC patients to taxane-based neoadjuvant chemotherapy.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Estudios Retrospectivos , Taxoides/farmacología , Taxoides/uso terapéutico , Hidrocarburos Aromáticos con Puentes/farmacología , Hidrocarburos Aromáticos con Puentes/uso terapéutico , Línea Celular Tumoral
15.
Aging Cell ; 22(11): e13944, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37858982

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Reposicionamiento de Medicamentos , Muerte Celular , Apoptosis , Telómero , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Línea Celular Tumoral
16.
EMBO Mol Med ; 15(1): e16033, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36426578

RESUMEN

The telomeric repeat-binding factor 2 (TRF2) is a telomere-capping protein that plays a key role in the maintenance of telomere structure and function. It is highly expressed in different cancer types, and it contributes to cancer progression. To date, anti-cancer strategies to target TRF2 remain a challenge. Here, we developed a miRNA-based approach to reduce TRF2 expression. By performing a high-throughput luciferase screening of 54 candidate miRNAs, we identified miR-182-3p as a specific and efficient post-transcriptional regulator of TRF2. Ectopic expression of miR-182-3p drastically reduced TRF2 protein levels in a panel of telomerase- or alternative lengthening of telomeres (ALT)-positive cancer cell lines. Moreover, miR-182-3p induced DNA damage at telomeric and pericentromeric sites, eventually leading to strong apoptosis activation. We also observed that treatment with lipid nanoparticles (LNPs) containing miR-182-3p impaired tumor growth in triple-negative breast cancer (TNBC) models, including patient-derived tumor xenografts (PDTXs), without affecting mouse survival or tissue function. Finally, LNPs-miR-182-3p were able to cross the blood-brain barrier and reduce intracranial tumors representing a possible therapeutic option for metastatic brain lesions.


Asunto(s)
MicroARNs , Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , Apoptosis , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Telómero/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
17.
Cell Death Dis ; 14(7): 441, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460459

RESUMEN

BH3 mimetics, targeting the Bcl-2 family anti-apoptotic proteins, represent a promising therapeutic opportunity in cancers. ABT-199, the first specific Bcl-2 inhibitor, was approved by FDA for the treatment of several hematological malignancies. We have recently discovered IS21, a novel pan BH3 mimetic with preclinical antitumor activity in several tumor types. Here, we evaluated the efficacy of IS21 and other BH3 mimetics, both as single agents and combined with the currently used antineoplastic agents in T-cell acute lymphoblastic leukemia, ovarian cancer, and melanoma. IS21 was found to be active in T-cell acute lymphoblastic leukemia, melanoma, lung, pancreatic, and ovarian cancer cell lines. Bcl-xL and Mcl-1 protein levels predicted IS21 sensitivity in melanoma and ovarian cancer, respectively. Exploring IS21 mechanism of action, we found that IS21 activity depends on the presence of BAX and BAK proteins: complexes between Bcl-2 and Bcl-xL proteins and their main binding partners were reduced after IS21 treatment. In combination experiments, BH3 mimetics sensitized leukemia cells to chemotherapy, ovarian cancer cells and melanoma models to PARP and MAPK inhibitors, respectively. We showed that this enhancing effect was related to the potentiation of the apoptotic pathway, both in hematologic and solid tumors. In conclusion, our data suggest the use of inhibitors of anti-apoptotic proteins as a therapeutic strategy to enhance the efficacy of anticancer treatment.


Asunto(s)
Antineoplásicos , Melanoma , Neoplasias Ováricas , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Femenino , Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Proteína bcl-X/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Línea Celular Tumoral
18.
Autophagy ; 19(5): 1479-1490, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36310382

RESUMEN

TERF2/TRF2 is a pleiotropic telomeric protein that plays a crucial role in tumor formation and progression through several telomere-dependent and -independent mechanisms. Here, we uncovered a novel function for this protein in regulating the macroautophagic/autophagic process upon different stimuli. By using both biochemical and cell biology approaches, we found that TERF2 binds to the non-histone chromatin-associated protein HMGB1, and this interaction is functional to the nuclear/cytoplasmic protein localization. Specifically, silencing of TERF2 alters the redox status of the cells, further exacerbated upon EBSS nutrient starvation, promoting the cytosolic translocation and the autophagic activity of HMGB1. Conversely, overexpression of wild-type TERF2, but not the mutant unable to bind HMGB1, negatively affects the cytosolic translocation of HMGB1, counteracting the stimulatory effect of EBSS starvation. Moreover, genetic depletion of HMGB1 or treatment with inflachromene, a specific inhibitor of its cytosolic translocation, completely abolished the pro-autophagic activity of TERF2 silencing. In conclusion, our data highlighted a novel mechanism through which TERF2 modulates the autophagic process, thus demonstrating the key role of the telomeric protein in regulating a process that is fundamental, under both physiological and pathological conditions, in defining the fate of the cells.Abbreviations: ALs: autolysosomes; ALT: alternative lengthening of telomeres; ATG: autophagy related; ATM: ATM serine/threonine kinase; CQ: Chloroquine; DCFDA: 2',7'-dichlorofluorescein diacetate; DDR: DNA damage response; DHE: dihydroethidium; EBSS: Earle's balanced salt solution; FACS: fluorescence-activated cell sorting; GFP: green fluorescent protein; EGFP: enhanced green fluorescent protein; GSH: reduced glutathione; GSSG: oxidized glutathione; HMGB1: high mobility group box 1; ICM: inflachromene; IF: immunofluorescence; IP: immunoprecipitation; NAC: N-acetyl-L-cysteine; NHEJ: non-homologous end joining; PLA: proximity ligation assay; RFP: red fluorescent protein; ROS: reactive oxygen species; TIF: telomere-induced foci; TERF2/TRF2: telomeric repeat binding factor 2.


Asunto(s)
Proteína HMGB1 , Proteína HMGB1/genética , Daño del ADN , Autofagia/genética , Telómero/metabolismo , Proteínas Nucleares/metabolismo
19.
Antimicrob Agents Chemother ; 56(8): 4268-76, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22644025

RESUMEN

This study investigated the possible mechanisms underlying the paradoxical caspofungin activity in vivo in preclinical aspergillosis. We evaluated the activity of escalating doses of caspofungin in vivo in different preclinical models of invasive aspergillosis, including mice deficient for selected innate immune receptors. The therapeutic efficacy of caspofungin in experimental invasive aspergillosis was strictly dose dependent, being observed at doses of 0.1 and 1 mg/kg of body weight depending on the experimental models. Paradoxical increase in pulmonary fungal burden as well as inflammatory pathology was observed at the highest dose of caspofungin (5 mg/kg), occurred independently of the so-called Eagle effect and susceptibility to caspofungin in vitro, and was contingent upon the presence of TLR2, Dectin-1, and TLR9. Increased expression of Dectin-1 and TLR9 were observed upon exposure to caspofungin in vitro and in vivo. Together, these findings suggest that the net activity of caspofungin in vivo is orchestrated by the activation, directly or indirectly, of multiple innate immune receptors.


Asunto(s)
Antifúngicos/farmacología , Aspergilosis/tratamiento farmacológico , Equinocandinas/farmacología , Lectinas Tipo C/metabolismo , Enfermedades Pulmonares Fúngicas/tratamiento farmacológico , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Antifúngicos/administración & dosificación , Antifúngicos/uso terapéutico , Aspergilosis/inmunología , Aspergilosis/microbiología , Aspergillus fumigatus/efectos de los fármacos , Caspofungina , Equinocandinas/administración & dosificación , Equinocandinas/uso terapéutico , Femenino , Humanos , Lipopéptidos , Pulmón/microbiología , Enfermedades Pulmonares Fúngicas/inmunología , Enfermedades Pulmonares Fúngicas/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Receptores Inmunológicos/metabolismo
20.
Blood ; 116(24): 5394-402, 2010 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-20807886

RESUMEN

The C-type lectin receptor Dectin-1 plays a pivotal role in antifungal immunity. In this study, the recently characterized human DECTIN1 Y238X early stop codon polymorphism leading to diminished Dectin-1 receptor activity was studied in relation to invasive aspergillosis susceptibility and severity in patients receiving hematopoietic stem cell transplantation. We found that the presence of the DECTIN1 Y238X polymorphism in either donors or recipients of hematopoietic stem cell transplantation increased susceptibility to aspergillosis, with the risk being highest when the polymorphism was present simultaneously in both donors and recipients (adjusted hazard ratio = 3.9; P = .005). Functionally, the Y238X polymorphism impaired the production of interferon-γ and interleukin-10 (IL-10), in addition to IL-1ß, IL-6, and IL-17A, by human peripheral mononuclear cells and Dectin-1 on human epithelial cells contributed to fungal recognition. Mechanistically, studies on preclinical models of infection in intact or bone marrow-transplanted Dectin-1 knockout mice revealed that protection from infection requires a distinct, yet complementary, role of both donor and recipient Dectin-1. This study discloses Dectin-1 deficiency as a novel susceptibility factor for aspergillosis in high-risk patients and identifies a previously unsuspected role for Dectin-1 in antifungal immunity that is the ability to control both resistance and tolerance to the fungus contingent on hematopoietic/nonhematopoietic compartmentalization.


Asunto(s)
Aspergilosis/etiología , Susceptibilidad a Enfermedades/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Polimorfismo Genético/inmunología , Adolescente , Adulto , Anciano , Animales , Aspergilosis/genética , Aspergilosis/inmunología , Niño , Citocinas/biosíntesis , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Hongos/inmunología , Humanos , Lectinas Tipo C , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas del Tejido Nervioso/deficiencia , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA