Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biomater Adv ; 161: 213881, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38749213

RESUMEN

Nanoparticle's success as drug delivery systems for cancer treatment has been achieved through passive targeting mechanisms. However, tumor heterogeneity and rapid drug clearance limit the treatment efficacy. Improved outcomes and selective drug release can be achieved by grafting ligands at the surface of nanocarriers that bind molecules overexpressed in the tumor microenvironment (TME). In this work, we developed a docetaxel-loaded nanoemulsions (NEs) binding an anti-netrin-1 monoclonal antibody (NP137) to selectively target the netrin-1 protein overexpressed in many different tumors. The goal is to refine a combined approach utilizing NP137 and docetaxel as an improved tumor-targeting chemotherapeutic agent for addressing triple-negative breast cancer (TNBC). Several factors have been considered for the optimization of the active targeted drug delivery system via the click-chemistry conjugation, as the impact of PEGylated surfactant that stabilize the NEs shell on conjugation efficiency, cytocompatibility with EMT6 cell line and colloidal stability over time of NEs. Results showed that a 660 Da PEG chain length contributed to NEs colloidal stability and had no impact on cell viability or on the antibody binding ability for its ligand after surface conjugation. Moreover, docetaxel was encapsulated into the oily core of NEs, with an encapsulation efficiency of 70 %. To validate our treatment strategy in vivo, the 4T1 murine breast cancer model was used. As a result, the comparison of active-targeted and non-targeted NEs revealed that only active-targeted NE could decrease the tumor growth rate.


Asunto(s)
Docetaxel , Nanopartículas , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Animales , Femenino , Nanopartículas/química , Línea Celular Tumoral , Ratones , Docetaxel/farmacología , Docetaxel/uso terapéutico , Docetaxel/administración & dosificación , Humanos , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico
2.
iScience ; 27(2): 108903, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38318383

RESUMEN

Although the involvement of protein kinase CK2 in cancer is well-documented, there is a need for selective CK2 inhibitors suitable for investigating CK2 specific roles in cancer-related biological pathways and further exploring its therapeutic potential. Here, we report the discovery of AB668, an outstanding selective inhibitor that binds CK2 through a bivalent mode, interacting both at the ATP site and an allosteric αD pocket unique to CK2. Using caspase activation assay, live-cell imaging, and transcriptomic analysis, we have compared the effects of this bivalent inhibitor to representative ATP-competitive inhibitors, CX-4945, and SGC-CK2-1. Our results show that in contrast to CX-4945 or SGC-CK2-1, AB668, by targeting the CK2 αD pocket, has a distinct mechanism of action regarding its anti-cancer activity, inducing apoptotic cell death in several cancer cell lines and stimulating distinct biological pathways in renal cell carcinoma.

3.
EMBO Mol Med ; 15(4): e16732, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36876343

RESUMEN

Targeted radionuclide therapy is a revolutionary tool for the treatment of highly spread metastatic cancers. Most current approaches rely on the use of vectors to deliver radionuclides to tumor cells, targeting membrane-bound cancer-specific moieties. Here, we report the embryonic navigation cue netrin-1 as an unanticipated target for vectorized radiotherapy. While netrin-1, known to be re-expressed in tumoral cells to promote cancer progression, is usually characterized as a diffusible ligand, we demonstrate here that netrin-1 is actually poorly diffusible and bound to the extracellular matrix. A therapeutic anti-netrin-1 monoclonal antibody (NP137) has been preclinically developed and was tested in various clinical trials showing an excellent safety profile. In order to provide a companion test detecting netrin-1 in solid tumors and allowing the selection of therapy-eligible patients, we used the clinical-grade NP137 agent and developed an indium-111-NODAGA-NP137 single photon emission computed tomography (SPECT) contrast agent. NP137-111 In provided specific detection of netrin-1-positive tumors with an excellent signal-to-noise ratio using SPECT/CT imaging in different mouse models. The high specificity and strong affinity of NP137 paved the way for the generation of lutetium-177-DOTA-NP137, a novel vectorized radiotherapy, which specifically accumulated in netrin-1-positive tumors. We demonstrate here, using tumor cell-engrafted mouse models and a genetically engineered mouse model, that a single systemic injection of NP137-177 Lu provides important antitumor effects and prolonged mouse survival. Together, these data support the view that NP137-111 In and NP137-177 Lu may represent original and unexplored imaging and therapeutic tools against advanced solid cancers.


Asunto(s)
Neoplasias , Radioinmunoterapia , Animales , Ratones , Línea Celular Tumoral , Neoplasias/diagnóstico por imagen , Neoplasias/radioterapia , Radioinmunoterapia/métodos , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Netrina-1/metabolismo
4.
J Enzyme Inhib Med Chem ; 38(1): 2121821, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36650907

RESUMEN

The mitochondrial voltage-dependent anion channel 1 (VDAC1) plays a central role in metabolism and apoptosis, which makes it a promising therapeutic target. Nevertheless, molecular mechanisms governing VDAC1 functioning remain unclear. Small-molecule ligands specifically interacting with the channel provide an attractive way of exploring its structure-function relationships and can possibly be used as founding stones for future drug-candidates. While around 30 VDAC1 ligands have been identified over the years, various techniques have been used by research teams, making a fair and direct comparison between compounds impossible. To tackle this issue, we performed ligand-binding assays on a representative set of seventeen known VDAC1 ligands using nano-differential scanning fluorimetry and microscale thermophoresis. While all the compounds have been confirmed as VDAC1 ligands by at least one method, combining both technologies lead to the selection of four molecules (cannabidiol, curcumin, DIDS and VBIT4) as chemical starting points for future design of VDAC1 selective ligands.


Asunto(s)
Cannabidiol , Canal Aniónico 1 Dependiente del Voltaje , Canal Aniónico 1 Dependiente del Voltaje/química , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Mitocondrias/metabolismo , Apoptosis , Cannabidiol/metabolismo
5.
ACS Med Chem Lett ; 13(2): 319-326, 2022 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-35178189

RESUMEN

Nine- and twelve-membered triaza-macrocycles were appended to one end of homospermidine to make polyamine lassos. These compounds were shown to be potent polyamine transport inhibitors (PTIs) using pancreatic ductal adenocarcinoma L3.6pl cells, which have high polyamine transport activity. The smaller triazacyclononane-based lasso significantly reduced the uptake of a fluorescent polyamine probe and inhibited spermidine uptake and reduced intracellular polyamine levels in difluoromethylornithine (DFMO)-treated L3.6pl cells. Both designs were shown to be effective inhibitors of 3H-spermidine uptake, with the smaller lasso outperforming the larger lasso. When the smaller lasso was challenged to inhibit each of the three radiolabeled native polyamines, it had similar K i values as those of the known PTIs, Trimer44NMe and AMXT1501. Because of these promising properties, these materials may have future anticancer applications in polyamine blocking therapy, an approach that couples a polyamine biosynthesis inhibitor (DFMO) with a PTI to lower intracellular polyamines and suppress cell growth.

6.
EMBO Mol Med ; 13(4): e12878, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33719214

RESUMEN

The navigation cue netrin-1 is well-documented for its key role in cancer development and represents a promising therapeutic target currently under clinical investigation. Phase 1 and 2 clinical trials are ongoing with NP137, a humanized monoclonal antibody against netrin-1. Interestingly, the epitope recognized by NP137 in netrin-1 shares 90% homology with its counterpart in netrin-3, the closest member to netrin-1 in humans, for which little is known in the field of cancer. Here, we unveiled that netrin-3 appears to be expressed specifically in human neuroblastoma (NB) and small cell lung cancer (SCLC), two subtypes of neuroectodermal/neuroendocrine lineages. Netrin-3 and netrin-1 expression are mutually exclusive, and the former is driven by the MYCN oncogene in NB, and the ASCL-1 or NeuroD1 transcription factors in SCLC. Netrin-3 expression is correlated with disease stage, aggressiveness, and overall survival in NB. Mechanistically, we confirmed the high affinity of netrin-3 for netrin-1 receptors and we demonstrated that netrin-3 genetic silencing or interference using NP137, delayed tumor engraftment, and reduced tumor growth in animal models. Altogether, these data support the targeting of netrin-3 in NB and SCLC.


Asunto(s)
Neoplasias Pulmonares , Neuroblastoma , Carcinoma Pulmonar de Células Pequeñas , Animales , Humanos , Netrina-1 , Netrinas
7.
Cancer Res ; 80(10): 1970-1980, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32060146

RESUMEN

The Sonic Hedgehog (SHH) pathway plays a key role in cancer. Alterations of SHH canonical signaling, causally linked to tumor progression, have become rational targets for cancer therapy. However, Smoothened (SMO) inhibitors have failed to show clinical benefit in patients with cancers displaying SHH autocrine/paracrine expression. We reported earlier that the SHH receptor Patched (PTCH) is a dependence receptor that triggers apoptosis in the absence of SHH through a pathway that differs from the canonical one, thus generating a state of dependence on SHH for survival. Here, we propose a dual function for SHH: its binding to PTCH not only activates the SHH canonical pathway but also blocks PTCH-induced apoptosis. Eighty percent, 64%, and 8% of human colon, pancreatic, and lung cancer cells, respectively, overexpressed SHH at transcriptional and protein levels. In addition, SHH-overexpressing cells expressed all the effectors of the PTCH-induced apoptotic pathway. Although the canonical pathway remained unchanged, autocrine SHH interference in colon, pancreatic, and lung cell lines triggered cell death through PTCH proapoptotic signaling. In vivo, SHH interference in colon cancer cell lines decreased primary tumor growth and metastasis. Therefore, the antitumor effect associated to SHH deprivation, usually thought to be a consequence of the inactivation of the canonical SHH pathway, is, at least in part, because of the engagement of PTCH proapoptotic activity. Together, these data strongly suggest that therapeutic strategies based on the disruption of SHH/PTCH interaction in SHH-overexpressing cancers should be explored. SIGNIFICANCE: Sonic Hedgehog-overexpressing tumors express PTCH-induced cell death effectors, suggesting that this death signaling could be activated as an antitumor strategy.


Asunto(s)
Apoptosis/fisiología , Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Receptores Patched/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/fisiología , Embrión de Pollo , Xenoinjertos , Humanos , Ratones , Transducción de Señal/fisiología , Pez Cebra
8.
Theranostics ; 8(18): 5126-5142, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30429890

RESUMEN

In ultrasound molecular imaging (USMI), ligand-functionalized microbubbles (MBs) are used to visualize vascular endothelial targets. Netrin-1 is upregulated in 60% of metastatic breast cancers and promotes tumor progression. A novel netrin-1 interference therapy requires the assessment of netrin-1 expression prior to treatment. In this study, we studied netrin-1 as a target for USMI and its potential as a companion diagnostic in breast cancer models. Methods: To verify netrin-1 expression and localization, an in vivo immuno-localization approach was applied, in which anti-netrin-1 antibody was injected into living mice 24 h before tumor collection, and revealed with secondary fluorescent antibody for immunofluorescence analysis. Netrin-1 interactions with the cell surface were studied by flow cytometry. Netrin-1-targeted MBs were prepared using MicroMarker Target-Ready (VisualSonics), and validated in in vitro binding assays in static conditions or in a flow chamber using purified netrin-1 protein or netrin-1-expressing cancer cells. In vivo USMI of netrin-1 was validated in nude mice bearing human netrin-1-positive SKBR7 tumors or weakly netrin-1-expressing MDA-MB-231 tumors using the Vevo 2100 small animal imaging device (VisualSonics). USMI feasibility was further tested in transgenic murine FVB/N Tg(MMTV/PyMT634Mul) (MMTV-PyMT) mammary tumors. Results: Netrin-1 co-localized with endothelial CD31 in netrin-1-positive breast tumors. Netrin-1 binding to the surface of endothelial HUVEC and cancer cells was partially mediated by heparan sulfate proteoglycans. MBs targeted with humanized monoclonal anti-netrin-1 antibody bound to netrin-1-expressing cancer cells in static and dynamic conditions. USMI signal was significantly increased with anti-netrin-1 MBs in human SKBR7 breast tumors and transgenic murine MMTV-PyMT mammary tumors compared to signals recorded with either isotype control MBs or after blocking of netrin-1 with humanized monoclonal anti-netrin-1 antibody. In weakly netrin-1-expressing human tumors and normal mammary glands, no difference in imaging signal was observed with anti-netrin-1- and isotype control MBs. Ex vivo analysis confirmed netrin-1 expression in MMTV-PyMT tumors. Conclusions: These results show that USMI allowed reliable detection of netrin-1 on the endothelium of netrin-1-positive human and murine tumors. Significant differences in USMI signal for netrin-1 reflected the significant differences in netrin-1 mRNA & protein expression observed between different breast tumor models. The imaging approach was non-invasive and safe, and provided the netrin-1 expression status in near real-time. Thus, USMI of netrin-1 has the potential to become a companion diagnostic for the stratification of patients for netrin-1 interference therapy in future clinical trials.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Inmunoterapia/métodos , Imagen Molecular/métodos , Terapia Molecular Dirigida/métodos , Netrina-1/análisis , Ultrasonografía/métodos , Animales , Anticuerpos/administración & dosificación , Neoplasias de la Mama/patología , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Xenoinjertos , Humanos , Ratones Desnudos , Ratones Transgénicos , Microburbujas , Trasplante de Neoplasias , Netrina-1/antagonistas & inhibidores , Resultado del Tratamiento
9.
Nat Commun ; 8: 16074, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28719575

RESUMEN

Notch signalling is a causal determinant of cancer and efforts have been made to develop targeted therapies to inhibit the so-called canonical pathway. Here we describe an unexpected pro-apoptotic role of Notch3 in regulating tumour angiogenesis independently of the Notch canonical pathway. The Notch3 ligand Jagged-1 is upregulated in a fraction of human cancer and our data support the view that Jagged-1, produced by cancer cells, is inhibiting the apoptosis induced by the aberrant Notch3 expression in tumour vasculature. We thus present Notch3 as a dependence receptor inducing endothelial cell death while this pro-apoptotic activity is blocked by Jagged-1. Along this line, using Notch3 mutant mice, we demonstrate that tumour growth and angiogenesis are increased when Notch3 is silenced in the stroma. Consequently, we show that the well-documented anti-tumour effect mediated by γ-secretase inhibition is at least in part dependent on the apoptosis triggered by Notch3 in endothelial cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Células Endoteliales/metabolismo , Neoplasias Pulmonares/metabolismo , Neovascularización Patológica , Receptor Notch3/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Muerte Celular , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteína Jagged-1/metabolismo , Ratones Endogámicos C57BL
10.
Biochem Biophys Res Commun ; 488(3): 466-470, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28501620

RESUMEN

Alzheimer's disease is a growing concern in the context of the increasing lifespan of the populations. The work presented here is part of the fight against this threat. It supports a therapeutic approach to reduce the incidence of Alzheimer's disease, taking advantage of the specific binding of several domains of Netrin-1 to the ß-amyloid precursor protein. This basic knowledge shall then be used to predict, design or characterize lead compounds that may in turn inhibit/delay Alzheimer's disease's progression, extending the therapeutic offer of the other leads already being investigated in this line. The present work is focused on the interaction of the various portions of APP with the three domains of Netrin-1, the so-called LamNT, EGF-like and NTR domains respectively. It reveals in detail which portions of APP and Netrin-1 are specifically involved in these interactions, using ELISA technique in combination with protein-protein binding simulations. So far unsuspected interaction sites located in Netrin-1 EGF-like and NTR domains open possibilities for new therapeutic approaches in which these sites will be specifically targeted.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Placa Amiloide/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Precursor de Proteína beta-Amiloide/química , Humanos , Modelos Moleculares , Factores de Crecimiento Nervioso/química , Netrina-1 , Unión Proteica , Proteínas Supresoras de Tumor/química
11.
Nat Commun ; 7: 13515, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27901020

RESUMEN

Netrins, a family of laminin-related molecules, have been proposed to act as guidance cues either during nervous system development or the establishment of the vascular system. This was clearly demonstrated for netrin-1 via its interaction with the receptors DCC and UNC5s. However, mainly based on shared homologies with netrin-1, netrin-4 was also proposed to play a role in neuronal outgrowth and developmental/pathological angiogenesis via interactions with netrin-1 receptors. Here, we present the high-resolution structure of netrin-4, which shows unique features in comparison with netrin-1, and show that it does not bind directly to any of the known netrin-1 receptors. We show that netrin-4 disrupts laminin networks and basement membranes (BMs) through high-affinity binding to the laminin γ1 chain. We hypothesize that this laminin-related function is essential for the previously described effects on axon growth promotion and angiogenesis. Our study unveils netrin-4 as a non-enzymatic extracellular matrix protein actively disrupting pre-existing BMs.


Asunto(s)
Orientación del Axón/fisiología , Membrana Basal/metabolismo , Laminina/fisiología , Neovascularización Fisiológica/fisiología , Netrinas/fisiología , Animales , Axones/fisiología , Pollos , Membrana Corioalantoides/fisiología , Cristalografía por Rayos X , Femenino , Células HEK293 , Humanos , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Netrinas/ultraestructura , Unión Proteica , Multimerización de Proteína , Ratas , Ratas Sprague-Dawley , Células de Schwann , Ensayos Antitumor por Modelo de Xenoinjerto
12.
EMBO Mol Med ; 8(8): 863-77, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27378792

RESUMEN

In a number of human cancers, NTN1 upregulation inhibits apoptosis induced by its so-called dependence receptors DCC and UNC5H, thus promoting tumor progression. In other cancers however, the selective inhibition of this dependence receptor death pathway relies on the silencing of pro-apoptotic effector proteins. We show here that a substantial fraction of human breast tumors exhibits simultaneous DNA methylation-dependent loss of expression of NTN1 and of DAPK1, a serine threonine kinase known to transduce the netrin-1 dependence receptor pro-apoptotic pathway. The inhibition of DNA methylation by drugs such as decitabine restores the expression of both NTN1 and DAPK1 in netrin-1-low cancer cells. Furthermore, a combination of decitabine with NTN1 silencing strategies or with an anti-netrin-1 neutralizing antibody potentiates tumor cell death and efficiently blocks tumor growth in different animal models. Thus, combining DNA methylation inhibitors with netrin-1 neutralizing agents may be a valuable strategy for combating cancer.


Asunto(s)
Neoplasias de la Mama/patología , Metilación de ADN , Regulación hacia Abajo , Factores de Crecimiento Nervioso/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proteínas Quinasas Asociadas a Muerte Celular/biosíntesis , Humanos , Netrina-1
13.
Cancer Cell ; 29(2): 173-85, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26859457

RESUMEN

Netrin-1 has been shown to be up-regulated in a fraction of human cancers as a mechanism to allow these tumors to escape the pro-apoptotic activity of some of its main dependence receptors, the UNC5 homologs (UNC5H). Here we identify the V-2 domain of netrin-1 to be important for its interaction with the Ig1/Ig2 domains of UNC5H2. We generate a humanized anti-netrin-1 antibody that disrupts the interaction between netrin-1 and UNC5H2 and triggers death of netrin-1-expressing tumor cells in vitro. We also present evidence that combining the anti-netrin-1 antibody with epidrugs such as decitabine could be effective in treating tumors showing no or modest netrin-1 expression. These results support that this antibody is a promising drug candidate.


Asunto(s)
Neoplasias/terapia , Factores de Crecimiento Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Neoplasias/metabolismo , Neoplasias/patología , Factores de Crecimiento Nervioso/inmunología , Receptores de Netrina , Netrina-1 , Unión Proteica , Proteínas Supresoras de Tumor/inmunología
14.
EMBO Mol Med ; 8(2): 96-104, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26882243

RESUMEN

DCC (Deleted in Colorectal Carcinoma) has been demonstrated to constrain tumor progression by inducing apoptosis unless engaged by its ligand netrin-1. This has been shown in breast and colorectal cancers; however, this tumor suppressive function in other cancers is not established. Using a transgenic mouse model, we report here that inhibition of DCC-induced apoptosis is associated with lymphomagenesis. In human diffuse large B-cell lymphoma (DLBCL), an imbalance of the netrin-1/DCC ratio suggests a loss of DCC-induced apoptosis, either via a decrease in DCC expression in germinal center subtype or by up-regulation of netrin-1 in activated B-cell (ABC) one. Such imbalance is also observed in mantle cell lymphoma (MCL). Using a netrin-1 interfering antibody, we demonstrate both in vitro and in vivo that netrin-1 acts as a survival factor for ABC-DLBCL and MCL tumor cells. Together, these data suggest that interference with the netrin-1/DCC interaction could represent a promising therapeutic strategy in netrin-1-positive DLBCL and MCL.


Asunto(s)
Linfoma de Células B Grandes Difuso/patología , Linfoma de Células B Grandes Difuso/terapia , Linfoma de Células del Manto/patología , Linfoma de Células del Manto/terapia , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Animales , Anticuerpos/administración & dosificación , Anticuerpos/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Línea Celular Tumoral , Receptor DCC , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones , Ratones Transgénicos , Netrina-1 , Unión Proteica
15.
Pharmaceuticals (Basel) ; 8(2): 279-302, 2015 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-26061121

RESUMEN

Due to their system of annulated 6-5-5-6-membered rings, indenoindoles have sparked great interest for the design of ATP-competitive inhibitors of human CK2. In the present study, we prepared twenty-one indeno[1,2-b]indole derivatives, all of which were tested in vitro on human CK2. The indenoindolones 5a and 5b inhibited human CK2 with an IC50 of 0.17 and 0.61 µM, respectively. The indeno[1,2-b]indoloquinone 7a also showed inhibitory activity on CK2 at a submicromolar range (IC50 = 0.43 µM). Additionally, a large number of indenoindole derivatives was evaluated for their cytotoxic activities against the cell lines 3T3, WI-38, HEK293T and MEF.

16.
Nucleic Acids Res ; 43(12): 5838-54, 2015 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-26007656

RESUMEN

DNA methylation is thought to induce transcriptional silencing through the combination of two mechanisms: the repulsion of transcriptional activators unable to bind their target sites when methylated, and the recruitment of transcriptional repressors with specific affinity for methylated DNA. The Methyl CpG Binding Domain proteins MeCP2, MBD1 and MBD2 belong to the latter category. Here, we present MBD2 ChIPseq data obtained from the endogenous MBD2 in an isogenic cellular model of oncogenic transformation of human mammary cells. In immortalized (HMEC-hTERT) or transformed (HMLER) cells, MBD2 was found in a large proportion of methylated regions and associated with transcriptional silencing. A redistribution of MBD2 on methylated DNA occurred during oncogenic transformation, frequently independently of local DNA methylation changes. Genes downregulated during HMEC-hTERT transformation preferentially gained MBD2 on their promoter. Furthermore, depletion of MBD2 induced an upregulation of MBD2-bound genes methylated at their promoter regions, in HMLER cells. Among the 3,160 genes downregulated in transformed cells, 380 genes were methylated at their promoter regions in both cell lines, specifically associated by MBD2 in HMLER cells, and upregulated upon MBD2 depletion in HMLER. The transcriptional MBD2-dependent downregulation occurring during oncogenic transformation was also observed in two additional models of mammary cell transformation. Thus, the dynamics of MBD2 deposition across methylated DNA regions was associated with the oncogenic transformation of human mammary cells.


Asunto(s)
Transformación Celular Neoplásica/genética , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Proteínas Represoras/metabolismo , Sitios de Unión , Mama/citología , Línea Celular , Línea Celular Transformada , Regulación hacia Abajo , Células Epiteliales/metabolismo , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Fenotipo , Telomerasa/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
17.
Bull Cancer ; 100(12): 1261-74, 2013 Dec.
Artículo en Francés | MEDLINE | ID: mdl-24240434

RESUMEN

Whereas the classic dogma postulates that transmembrane receptors remain inactive at the plasma membrane unless bound by their specific ligand, it was suggested that some receptors may actually be active not only in the presence of their ligand, but also in their absence. In this latter case, the signaling downstream of these unbound receptors leads to apoptosis. These receptors were consequently named dependence receptors, as their cell expression renders the survival of the cell dependent on the presence in the cell environment of its respective ligand. This dual function - positive in the presence of ligand, negative in the absence of ligand - is hypothesized to lead these receptors to have key roles both during embryonic development and in the regulation of tumorigenesis. In the context of cancer, the hypothesis is that these receptors are tumor suppressors that would limit tumor progression by inducing apoptosis of tumor cells outside of settings of ligand accessibility/availability. This was recently formally demonstrated for the prototypical dependence receptors that bind netrin-1- i.e., DCC and UNC5H. Because expression of DCC and UNC5H is a constraint for tumor progression, their expression is often lost in many aggressive cancers. However, a loss of dependence receptors is not always the selective advantage used by tumor cells to escape this survival dependence on the presence of the ligand. Indeed, it was shown that in many cancers, tumor cells acquire the preferred autocrine expression of ligands of dependence receptor. This selective advantage for the tumor is much more appealing in terms of therapeutic opportunities. Drugs based on the interference on the interaction between dependence receptors and their ligands allow tumor cell death in vitro and trigger tumor growth and metastases inhibition in mice. This review describes how a basic cell biology concept has provided in a near future new tools to fight cancer.


Asunto(s)
Apoptosis/fisiología , Ligandos , Neoplasias , Receptores de Superficie Celular/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Comunicación Autocrina/fisiología , Supervivencia Celular/fisiología , Progresión de la Enfermedad , Humanos , Ratones , Terapia Molecular Dirigida/métodos , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/fisiopatología , Neoplasias/terapia , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo
18.
PLoS Biol ; 11(8): e1001623, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23940460

RESUMEN

The Hedgehog signaling is a determinant pathway for tumor progression. However, while inhibition of the Hedgehog canonical pathway-Patched-Smoothened-Gli-has proved efficient in human tumors with activating mutations in this pathway, recent clinical data have failed to show any benefit in other cancers, even though Sonic Hedgehog (SHH) expression is detected in these cancers. Cell-adhesion molecule-related/down-regulated by Oncogenes (CDON), a positive regulator of skeletal muscle development, was recently identified as a receptor for SHH. We show here that CDON behaves as a SHH dependence receptor: it actively triggers apoptosis in the absence of SHH. The pro-apoptotic activity of unbound CDON requires a proteolytic cleavage in its intracellular domain, allowing the recruitment and activation of caspase-9. We show that by inducing apoptosis in settings of SHH limitation, CDON expression constrains tumor progression, and as such, decreased CDON expression observed in a large fraction of human colorectal cancer is associated in mice with intestinal tumor progression. Reciprocally, we propose that the SHH expression, detected in human cancers and previously considered as a mechanism for activation of the canonical pathway in an autocrine or paracrine manner, actually provides a selective tumor growth advantage by blocking CDON-induced apoptosis. In support of this notion, we present the preclinical demonstration that interference with the SHH-CDON interaction triggers a CDON-dependent apoptosis in vitro and tumor growth inhibition in vivo. The latter observation qualifies CDON as a relevant alternative target for anticancer therapy in SHH-expressing tumors.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Moléculas de Adhesión Celular/genética , Línea Celular , Línea Celular Tumoral , Femenino , Proteínas Hedgehog/genética , Humanos , Masculino , Ratones , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/genética
19.
PLoS One ; 8(6): e67071, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785518

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype of kidney cancer and is often characterized by mutations or deletions of the Von Hippel Lindau (VHL) tumour suppressor gene. Aurora gene family members are implicated in proper mitotic progression and spindle checkpoint function and play a crucial role in cancer progression. In the present study, we assessed the expression of Aurora-A in a cohort of 30 ccRCC with fully characterized VHL status (wt/wt or mut/del) and Fuhrman grade. Aurora-A transcript and protein levels were significantly increased in high Fuhrman grade tumours and in VHLwt/wt tumours. These results suggest that Aurora-A and VHL interact in the ccRCC. We demonstrated that the two proteins interact in vivo and identified the Ser72 on the sequence of VHL as the unique site phosphorylated by Aurora-A.


Asunto(s)
Aurora Quinasa A/metabolismo , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Aurora Quinasa A/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Modelos Biológicos , Mutación , Clasificación del Tumor , Fosforilación , Unión Proteica , Transcripción Genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
20.
J Med Chem ; 53(21): 7647-63, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-20925391

RESUMEN

The polyamine transport system (PTS) whose activity is up-regulated in cancer cells is an attractive target for drug design. Two heterocyclic (azepine and benzazepine) systems were conjugated to various polyamine moieties through an amidine bound to afford 18 compounds which were evaluated for their affinity for the PTS and their ability to use the PTS for cell delivery. Structure-activity relationship studies and lead optimization afforded two attractive PTS targeting compounds. The azepine-spermidine conjugate 14 is a very selective substrate of the PTS that may serve as a vector for radioelements used for diagnoses or therapeutics in nuclear medicine. The nitrobenzazepine-spermine conjugate 28 is a very powerful PTS inhibitor with very low intrinsic cytotoxicity, able to prevent the growth of polyamine depleted cells in presence of exogenous polyamines.


Asunto(s)
Antineoplásicos/síntesis química , Azepinas/síntesis química , Benzazepinas/síntesis química , Poliaminas/síntesis química , Poliaminas/metabolismo , Espermidina/análogos & derivados , Espermina/análogos & derivados , Animales , Antineoplásicos/farmacología , Azepinas/farmacología , Benzazepinas/farmacología , Transporte Biológico/efectos de los fármacos , Células CHO , Proliferación Celular/efectos de los fármacos , Cricetinae , Cricetulus , Ensayos de Selección de Medicamentos Antitumorales , Leucemia L1210 , Poliaminas/farmacología , Espermidina/síntesis química , Espermidina/farmacología , Espermina/síntesis química , Espermina/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...