Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell Mol Life Sci ; 77(21): 4209-4222, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32270227

RESUMEN

Inherited or acquired mutations can lead to pathological outcomes. However, in a process defined as synthetic rescue, phenotypic outcome created by primary mutation is alleviated by suppressor mutations. An exhaustive characterization of these mutations in humans is extremely valuable to better comprehend why patients carrying the same detrimental mutation exhibit different pathological outcomes or different responses to treatment. Here, we first review all known suppressor mutations' mechanisms characterized by genetic screens on model species like yeast or flies. However, human suppressor mutations are scarce, despite some being discovered based on orthologue genes. Because of recent advances in high-throughput screening, developing an inventory of human suppressor mutations for pathological processes seems achievable. In addition, we review several screening methods for suppressor mutations in cultured human cells through knock-out, knock-down or random mutagenesis screens on large scale. We provide examples of studies published over the past years that opened new therapeutic avenues, particularly in oncology.


Asunto(s)
Mutagénesis , Supresión Genética , Animales , Sistemas CRISPR-Cas , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Humanos , Neoplasias/genética , Interferencia de ARN
2.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34360928

RESUMEN

Xeroderma Pigmentosum protein C (XPC) is involved in recognition and repair of bulky DNA damage such as lesions induced by Ultra Violet (UV) radiation. XPC-mutated cells are, therefore, photosensitive and accumulate UVB-induced pyrimidine dimers leading to increased cancer incidence. Here, we performed a high-throughput screen to identify chemicals capable of normalizing the XP-C phenotype (hyper-photosensitivity and accumulation of photoproducts). Fibroblasts from XP-C patients were treated with a library of approved chemical drugs. Out of 1280 tested chemicals, 16 showed ≥25% photo-resistance with RZscore above 2.6 and two drugs were able to favor repair of 6-4 pyrimidine pyrimidone photoproducts (6-4PP). Among these two compounds, Isoconazole could partially inhibit apoptosis of the irradiated cells especially when cells were post-treated directly after UV irradiation while Clemizole Hydrochloride-mediated increase in viability was dependent on both pre and post treatment. No synergistic effect was recorded following combined drug treatment and the compounds exerted no effect on the proliferative capacity of the cells post UV exposure. Amelioration of XP-C phenotype is a pave way towards understanding the accelerated skin cancer initiation in XP-C patients. Further examination is required to decipher the molecular mechanisms targeted by these two chemicals.


Asunto(s)
Bencimidazoles/farmacología , Miconazol/análogos & derivados , Enfermedades de la Piel/tratamiento farmacológico , Rayos Ultravioleta/efectos adversos , Xerodermia Pigmentosa/tratamiento farmacológico , Línea Celular , Supervivencia Celular/efectos de los fármacos , Reposicionamiento de Medicamentos , Humanos , Miconazol/farmacología
3.
J Neurooncol ; 144(3): 489-499, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31482266

RESUMEN

PURPOSE: Glioblastoma is the most aggressive malignant brain tumor. Despite multimodal treatments, median survival is only 15 months for glioblastoma patients, with tumor recurring in the resection margins after surgical removal. Hypothermia is emerging as an interesting and safe treatment for several conditions. In the context of glioblastoma, we propose that moderate hypothermia could inhibit both cell proliferation and migration, and thus help prevent secondary tumor growth. METHODS: In vitro experiments on A172, U251, U87 and T98G human glioblastoma cell lines explored the effects of severe (23 °C), moderate (28 °C), and mild (33 °C) hypothermia. We further investigated the effects of moderate hypothermia on cell proliferation, migration, morphology, and cell cycle distribution. RESULTS: Similar results were obtained with all four cell lines, indicating a consistent and broad effect of moderate hypothermia. Hypothermia inhibited both cell proliferation and non-oriented migration in a dose-dependent manner, with a significant reduction at 33 °C and almost total arrest at 28 °C. Cell proliferation arrest was long-lasting and oriented cell migration was also reduced at 28 °C. Moreover, moderate hypothermia significantly altered cell cycle distribution, with cells accumulating in the G2/M phase, leading to cell cycle arrest. Lastly, hypothermia at 28 °C also affected cell morphology by deteriorating cell membranes and altering cell shape. CONCLUSIONS: The presented results demonstrate that moderate hypothermia could be a promising adjuvant therapy for glioblastoma treatment as it strongly inhibits both cell proliferation and migration. If in vivo preclinical results corroborate our findings, therapeutic hypothermia applied at the resection margins could probably delay tumor recurrence, combined with current treatments.


Asunto(s)
Puntos de Control del Ciclo Celular , Movimiento Celular , Proliferación Celular , Glioblastoma/prevención & control , Hipotermia , Apoptosis , Glioblastoma/patología , Humanos , Células Tumorales Cultivadas , Cicatrización de Heridas
4.
Biochem Biophys Res Commun ; 507(1-4): 348-354, 2018 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-30446228

RESUMEN

The miR-143/145 cluster is down-regulated in cervical tumor cells suggesting a role in tumorigenesis including cytoskeleton remodeling, a key event for tumor progression. The aim of the present work was to determine the role of miR-143/145 in the modulation of the myosin regulator phospho-myosin light chain (pMLC). HeLa monolayer and tridimensional cultures were transfected with miR-143 or miR-145 mimics inhibiting cell viability, proliferation, migration and invasion, mainly through miR-145. MiR-145 transfection increased pMLC levels by targeting the MYPT1 subunit of the regulatory myosin phosphatase. MYPT1 knockdown by siRNAs reproduced miR-145 effects suggesting miR-145 as a tumor suppressor through MYPT1 targeting, leading to a subsequent increase of pMLC levels with implications for cervical cell viability, migration and invasion.


Asunto(s)
Técnicas de Cultivo de Célula , Movimiento Celular , MicroARNs/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Secuencia de Bases , Movimiento Celular/genética , Supervivencia Celular/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Queratinocitos/metabolismo , MicroARNs/genética , Cadenas Ligeras de Miosina/metabolismo , Invasividad Neoplásica , Fosforilación , ARN Interferente Pequeño/metabolismo , Esferoides Celulares/metabolismo , Ensayo de Tumor de Célula Madre
5.
IEEE Trans Nanobioscience ; 22(3): 455-466, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36215360

RESUMEN

Unlike Quality by Testing approach, where products were tested only after drug manufacturing, Quality by Design (QbD) is a proactive control quality paradigm, which handles risks from the early development steps. In QbD, regression models built from experimental data are used to predict a risk mapping called Design Space in which the developers can identify values of critical input factors leading to acceptable probabilities to meet the efficacy and safety specifications for the expected product. These empirical models are often limited to quantitative responses. Moreover, in practice the smallness and incompleteness of datasets degrade the quality of predictions. In this study, a Bayesian approach including variable selection, parameter estimation and model quality assessment is proposed and assessed using a real case study devoted to the development of a Cationic Nano-Lipid Structures for siRNA Transfection. Two original model structures are also included to describe both binary and percentage response variables. The results confirm the practical relevance and applicability of the Bayesian implementation of the QbD analysis.


Asunto(s)
Teorema de Bayes , ARN Interferente Pequeño/genética , Control de Calidad
6.
Angiogenesis ; 15(4): 609-22, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22869002

RESUMEN

Inflammatory neovascularization, such as choroidal neovascularization (CNV), occur in the presence of Notch expressing macrophages. DLL4s anti-angiogenic effect on endothelial cells (EC) has been widely recognized, but its influence on Notch signaling on macrophages and its overall effect in inflammatory neovascularization is not well understood. We identified macrophages and ECs as the main Notch 1 and Notch 4 expressing cells in CNV. A soluble fraction spanning Ser28-Pro525 of the murine extracellular DLL4 domain (sDLL4/28-525) activated the Notch pathway, as it induces Notch target genes in macrophages and ECs and inhibited EC proliferation and vascular sprouting in aortic rings. In contrast, sDLL4/28-525 increased pro-angiogenic VEGF, and IL-1ß expression in macrophages responsible for increased vascular sprouting observed in aortic rings incubated in conditioned media from sDLL4/28-525 stimulated macrophages. In vivo, Dll4(+/-) mice developed significantly more CNV and sDLL4/28-525 injections inhibited CNV in Dll4(+/-) CD1 mice. Similarly, sDLL4/28-525 inhibited CNV in C57Bl6 and its effect was reversed by a γ-secretase inhibitor that blocks Notch signaling. The inhibition occurred despite increased VEGF, IL-1ß expression in infiltrating inflammatory macrophages in sDLL4/28-525 treated mice and might be due to direct inhibition of EC proliferation in laser-induced CNV as demonstrated by EdU labelling in vivo. In conclusion, Notch activation on macrophages and ECs leads to opposing effects in inflammatory neovascularization in situations such as CNV.


Asunto(s)
Neovascularización Coroidal/prevención & control , Endotelio Vascular/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Macrófagos Peritoneales/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Secuencia de Bases , Western Blotting , Proteínas de Unión al Calcio , Cartilla de ADN , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Blood ; 115(20): 4083-92, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20231429

RESUMEN

The role of c-Jun NH(2)-terminal kinase 1 (JNK1) in hemostasis and thrombosis remains unclear. We show here, with JNK1-deficient (JNK1(-/-)) mice, that JNK1 plays an important role in platelet biology and thrombus formation. In tail-bleeding assays, JNK1(-/-) mice exhibited longer bleeding times than wild-type mice (396 +/- 39 seconds vs 245 +/- 32 seconds). We also carried out in vitro whole-blood perfusion assays on a collagen matrix under arterial shear conditions. Thrombus formation was significantly reduced for JNK1(-/-) platelets (51%). In an in vivo model of thrombosis induced by photochemical injury to cecum vessels, occlusion times were 4.3 times longer in JNK1(-/-) arterioles than in wild-type arterioles. Moreover, in vitro studies carried out in platelet aggregation conditions demonstrated that, at low doses of agonists, platelet secretion was impaired in JNK1(-/-) platelets, leading to altered integrin alphaIIbbeta3 activation and reduced platelet aggregation, via a mechanism involving protein kinase C. JNK1 thus appears to be essential for platelet secretion in vitro, consistent with its role in thrombus growth in vivo. Finally, we showed that ERK2 and another isoform of JNK affect platelet aggregation through 2 pathways, one dependent and another independent of JNK1.


Asunto(s)
Plaquetas/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Agregación Plaquetaria , Trombosis/metabolismo , Animales , Coagulación Sanguínea , Western Blotting , Citometría de Flujo , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Recuento de Plaquetas , Pruebas de Función Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteína Quinasa C/metabolismo
8.
Cells ; 11(22)2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36429042

RESUMEN

This is a spectacular moment for genetics to evolve in genome editing, which encompasses the precise alteration of the cellular DNA sequences within various species. One of the most fascinating genome-editing technologies currently available is Clustered Regularly Interspaced Palindromic Repeats (CRISPR) and its associated protein 9 (CRISPR-Cas9), which have integrated deeply into the research field within a short period due to its effectiveness. It became a standard tool utilized in a broad spectrum of biological and therapeutic applications. Furthermore, reliable disease models are required to improve the quality of healthcare. CRISPR-Cas9 has the potential to diversify our knowledge in genetics by generating cellular models, which can mimic various human diseases to better understand the disease consequences and develop new treatments. Precision in genome editing offered by CRISPR-Cas9 is now paving the way for gene therapy to expand in clinical trials to treat several genetic diseases in a wide range of species. This review article will discuss genome-editing tools: CRISPR-Cas9, Zinc Finger Nucleases (ZFNs), and Transcription Activator-Like Effector Nucleases (TALENs). It will also encompass the importance of CRISPR-Cas9 technology in generating cellular disease models for novel therapeutics, its applications in gene therapy, and challenges with novel strategies to enhance its specificity.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Humanos , Sistemas CRISPR-Cas/genética , Nucleasas con Dedos de Zinc , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Tecnología
9.
J Control Release ; 345: 646-660, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35339579

RESUMEN

Complex in vitro models of human immune cells and intestinal mucosa may have a translation-assisting role in the assessment of anti-inflammatory compounds. Chronic inflammation of the gastrointestinal tract is a hallmark of inflammatory bowel diseases (IBD). In both IBD entities, Crohn's disease and ulcerative colitis, impaired immune cell activation and dysfunctional epithelial barrier are the common pathophysiology. Current therapeutic approaches are targeting single immune modulator molecules to stop disease progression and reduce adverse effects. Such molecular targets can be difficult to assess in experimental animal models of colitis, due to the disease complexity and species differences. Previously, a co-culture model based on human epithelial cells and monocytes arranged in a physiological microenvironment was used to mimic inflamed mucosa for toxicological and permeability studies. The leaky gut model described here, a co-culture of Caco-2, THP-1 and MUTZ-3 cells, was used to mimic IBD-related pathophysiology and for combined investigations of permeability and target engagement of two Janus kinase (JAK) inhibitors, tofacitinib (TOFA) and a JAK1-targeting siRNA nanomedicine. The co-culture just before reaching confluency of the epithelium was used to mimic the compromised intestinal barrier. Delivery efficacy and target engagement against JAK1 was quantified via downstream analysis of STAT1 protein phosphorylation after IFN-γ stimulation. Compared to a tight barrier, the leaky gut model showed 92 ± 5% confluence, a barrier function below 200 Ω*cm2, and enhanced immune response to bacteria-derived lipopolysaccharides. By confocal microscopy we observed an increased accumulation of siJAK1-nanoparticles within the sub-confluent regions leading to uptake into immune cells near the epithelium. A concentration-dependent downregulation of JAK/STAT pathway was observed for siJAK1-nanoparticles (10 ± 12% to 16 ± 12%), whereas TOFA inhibition was 86 ± 2%, compared to untreated cells. By mimicking the status of severely damaged epithelium, like in IBD, the leaky gut model holds promise as a human in vitro system to evaluate the efficacy of anti-inflammatory drugs and nanomedicines.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Inhibidores de las Cinasas Janus , Animales , Células CACO-2 , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Janus Quinasa 1/metabolismo , Inhibidores de las Cinasas Janus/metabolismo , Inhibidores de las Cinasas Janus/farmacología , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus/metabolismo , Nanomedicina , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal
10.
J Crohns Colitis ; 16(2): 286-300, 2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-34286840

RESUMEN

BACKGROUND AND AIMS: Inflammatory bowel diseases are highly debilitating conditions that require constant monitoring and life-long medication. Current treatments are focused on systemic administration of immunomodulatory drugs, but they have a broad range of undesirable side-effects. RNA interference is a highly specific endogenous mechanism that regulates the expression of the gene at the transcript level, which can be repurposed using exogenous short interfering RNA [siRNA] to repress expression of the target gene. While siRNA therapeutics can offer an alternative to existing therapies, with a high specificity critical for chronically administrated drugs, evidence of their potency compared to chemical kinase inhibitors used in clinics is still lacking in alleviating an adverse inflammatory response. METHODS: We provide a framework to select highly specific siRNA, with a focus on two kinases strongly involved in pro-inflammatory diseases, namely JAK1 and JAK3. Using western-blot, real-time quantitative PCR and large-scale analysis, we assessed the specificity profile of these siRNA drugs and compared their efficacy to the most recent and promising kinase inhibitors for Janus kinases [Jakinibs], tofacitinib and filgotinib. RESULTS: siRNA drugs can reach higher efficiency and selectivity at lower doses [5 pM vs 1 µM] than Jakinibs. Moreover, JAK silencing lasted up to 11 days, even with 6 h pulse transfection. CONCLUSIONS: The siRNA-based drugs developed hold the potential to develop more potent therapeutics for chronic inflammatory diseases.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Quinasas Janus , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/genética , Quinasas Janus/genética , Quinasas Janus/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal
11.
J Hepatol ; 54(6): 1185-94, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21145827

RESUMEN

BACKGROUND & AIMS: C/EBPbeta is an important mediator of several cellular processes, such as differentiation, proliferation, and survival of hepatic cells. However, a complete catalog of the targets of C/EBPbeta or the mechanism by which this transcription factor regulates certain liver-dependent pathways has not been clearly determined. Two major natural isoforms of this transcription factor exist: the liver-enriched activating protein (LAP) and the liver-enriched inhibitory protein (LIP), a functional LAP antagonist. In this study, we used the opposing transcriptional effects driven by LAP and LIP to determine the genuine C/EBPbeta molecular signature in the Hep3B human hepatoma cell line. We subsequently investigated the role of each of the LAP and LIP isoforms in drug-induced Hep3B cell death. METHODS: We engineered Hep3B cells with regulated LAP or LIP expression using the Tet-off expression system. The genes that showed inverse regulation by LAP and LIP were identified by cDNA array analysis. The cohort of direct-C/EBPbeta-targets was distinguished from indirect-targets by ChIP-on-chip analysis. RESULTS: We characterized 676 genes by this approach. Among these genes, 39 are novel direct targets of C/EBPbeta. Eleven of these new direct targets are involved in cell survival, suggesting critical roles for LAP/LIP isoforms in this cellular process. Therefore, we examined the effects of LAP and LIP over-expression on cell survival. We show that LIP promotes survival in staurosporine- or taxol-induced Hep3B cell death. CONCLUSIONS: Our study provides new molecular and cellular insights into the role of C/EBPbeta in cells of hepatic origin.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Carcinoma Hepatocelular/patología , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Muerte Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Perfilación de la Expresión Génica , Ingeniería Genética , Humanos , Neoplasias Hepáticas/patología , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Paclitaxel/farmacología , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estaurosporina/farmacología , Transcripción Genética
12.
Am J Pathol ; 177(6): 3192-201, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20971743

RESUMEN

RNA interference mediated by small interfering RNAs (siRNAs) has emerged as a potential therapeutic approach to treat various diseases, including cancer. Recent studies with several animal models of posttraumatic revascularization demonstrated that synthetic siRNAs may produce therapeutic effects in a target-independent manner through the stimulation of the toll-like receptor-3 (TLR3)/interferon pathway and suppression of angiogenesis. To analyze the impact of siRNAs on tumor angiogenesis, we injected transgenic mice developing hepatocellular carcinoma (HCC) with either control siRNAs or siRNA targeting neuropilin-1. We found that treatment with these siRNAs led to a comparable reduction in tumor liver volume and to inhibition of tumor vasculature remodeling. We further determined that TLR3, which recognizes double-stranded siRNA, was up-regulated in mouse HCC. Treatment of HCC mice with polyinosinic-polycytidylic acid [poly(I:C)], a TLR3 agonist, led to both a reduction of tumor liver enlargement and a decrease in hepatic arterial blood flow, indicating that TLR3 is functional and may mediate both anti-angiogenic and anti-tumor responses. We also demonstrated that siRNAs increased interferon-γ levels in the liver. In vitro, interferon-γ inhibited proliferation of endothelial cells. In addition, we found that siRNAs inhibited endothelial cell proliferation and morphogenesis in an interferon-γ-independent manner. Our results suggest that synthetic siRNAs inhibit target-independently HCC growth and angiogenesis through the activation of the innate interferon response and by directly inhibiting endothelial cell function.


Asunto(s)
Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Neoplasias Hepáticas/patología , Neovascularización Patológica/prevención & control , ARN Interferente Pequeño/farmacología , Animales , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/genética , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Neovascularización Patológica/genética , Interferencia de ARN/fisiología , Carga Tumoral/efectos de los fármacos
13.
J Clin Invest ; 117(6): 1527-37, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17510705

RESUMEN

Endothelial progenitor cell (EPC) transplantation has beneficial effects for therapeutic neovascularization; however, only a small proportion of injected cells home to the lesion and incorporate into the neocapillaries. Consequently, this type of cell therapy requires substantial improvement to be of clinical value. Erythropoietin-producing human hepatocellular carcinoma (Eph) receptors and their ephrin ligands are key regulators of vascular development. We postulated that activation of the EphB4/ephrin-B2 system may enhance EPC proangiogenic potential. In this report, we demonstrate in a nude mouse model of hind limb ischemia that EphB4 activation with an ephrin-B2-Fc chimeric protein increases the angiogenic potential of human EPCs. This effect was abolished by EphB4 siRNA, confirming that it is mediated by EphB4. EphB4 activation enhanced P selectin glycoprotein ligand-1 (PSGL-1) expression and EPC adhesion. Inhibition of PSGL-1 by siRNA reversed the proangiogenic and adhesive effects of EphB4 activation. Moreover, neutralizing antibodies to E selectin and P selectin blocked ephrin-B2-Fc-stimulated EPC adhesion properties. Thus, activation of EphB4 enhances EPC proangiogenic capacity through induction of PSGL-1 expression and adhesion to E selectin and P selectin. Therefore, activation of EphB4 is an innovative and potentially valuable therapeutic strategy for improving the recruitment of EPCs to sites of neovascularization and thereby the efficiency of cell-based proangiogenic therapy.


Asunto(s)
Células Endoteliales/metabolismo , Células Madre Fetales/metabolismo , Glicoproteínas de Membrana/metabolismo , Neovascularización Fisiológica , Receptor EphB4/metabolismo , Animales , Secuencia de Bases , Adhesión Celular , Células Cultivadas , Cartilla de ADN/genética , Selectina E/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Efrina-B2/metabolismo , Efrina-B2/farmacología , Sangre Fetal/citología , Células Madre Fetales/citología , Células Madre Fetales/efectos de los fármacos , Miembro Posterior/irrigación sanguínea , Humanos , Técnicas In Vitro , Isquemia/metabolismo , Isquemia/patología , Isquemia/terapia , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Selectina-P/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor EphB4/antagonistas & inhibidores , Receptor EphB4/genética
14.
Biol Cell ; 101(9): 525-39, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19281453

RESUMEN

BACKGROUND INFORMATION: Endothelial cells play a major role in angiogenesis, the process by which new blood vessels arise from a pre-existing vascular bed. VEGF-A (vascular endothelial growth factor-A) is a key regulator of angiogenesis during both development and in adults. HGF (hepatocyte growth factor) is a pleiotropic cytokine that may promote VEGF-A-driven angiogenesis, although the signalling mechanisms underlying this co-operation are not completely understood. RESULTS: We analysed the effects of the combination of VEGF-A and HGF on the activation of VEGFR-2 (VEGF receptor-2) and c-met receptors, and on the stimulation of downstream signalling pathways in endothelial cells. We found that VEGFR-2 and c-met do not physically associate and do not transphosphorylate each other, suggesting that co-operation involves signalling events more distal from receptor activation. We demonstrate that the VEGF isoform VEGF-A(165) and HGF stimulate a similar set of MAPKs (mitogen-activated protein kinases), although the kinetics and strengths of the activation differ depending on the growth factor and pathway. An enhanced activation of the signalling was observed when endothelial cells were stimulated by the combination of VEGF-A(165) and HGF. Moreover, the combination of VEGF-A and HGF results in a statistically significant synergistic activation of ERK1/2 (extracellular-signal-regulated kinase 1/2) and p38 kinases. We demonstrated that VEGF-A(165) and HGF activate FAK (focal adhesion kinase) with different kinetics and stimulate the recruitment of phosphorylated FAK to different subsets of focal adhesions. VEGF-A(165) and HGF regulate distinct morphogenic aspects of the cytoskeletal remodelling that are associated with the preferential activation of Rho or Rac respectively, and induce structurally distinct vascular-like patterns in vitro in a Rho- or Rac-dependent manner. CONCLUSIONS: Under angiogenic conditions, combining VEGF-A with HGF can promote neovascularization by enhancing intracellular signalling and allowing more finely regulated control of the signalling molecules involved in the regulation of the cytoskeleton and cellular migration and morphogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Receptor Cross-Talk , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Cultivadas , Factor de Crecimiento de Hepatocito/genética , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Oncogene ; 38(28): 5700-5724, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31043708

RESUMEN

Androgen receptor (AR) signalling is a key prostate cancer (PC) driver, even in advanced 'castrate-resistant' disease (CRPC). To systematically identify microRNAs (miRs) modulating AR activity in lethal disease, hormone-responsive and -resistant PC cells expressing a luciferase-based AR reporter were transfected with a miR inhibitor library; 78 inhibitors significantly altered AR activity. Upon validation, miR-346, miR-361-3p and miR-197 inhibitors markedly reduced AR transcriptional activity, mRNA and protein levels, increased apoptosis, reduced proliferation, repressed EMT, and inhibited PC migration and invasion, demonstrating additive effects with AR inhibition. Corresponding miRs increased AR activity through a novel and anti-dogmatic mechanism of direct association with AR 6.9 kb 3'UTR and transcript stabilisation. In addition, miR-346 and miR-361-3p modulation altered levels of constitutively active AR variants, and inhibited variant-driven PC cell proliferation, so may contribute to persistent AR signalling in CRPC in the absence of circulating androgens. Pathway analysis of AGO-PAR-CLIP-identified miR targets revealed roles in DNA replication and repair, cell cycle, signal transduction and immune function. Silencing these targets, including tumour suppressors ARHGDIA and TAGLN2, phenocopied miR effects, demonstrating physiological relevance. MiR-346 additionally upregulated the oncogene, YWHAZ, which correlated with grade, biochemical relapse and metastasis in patients. These AR-modulatory miRs and targets correlated with AR activity in patient biopsies, and were elevated in response to long-term enzalutamide treatment of patient-derived CRPC xenografts. In summary, we identified miRs that modulate AR activity in PC and CRPC, via novel mechanisms, and may represent novel PC therapeutic targets.


Asunto(s)
MicroARNs/fisiología , Neoplasias de la Próstata/tratamiento farmacológico , Receptores Androgénicos/fisiología , Regiones no Traducidas 3' , Elementos sin Sentido (Genética) , Benzamidas , Línea Celular Tumoral , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Humanos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Nitrilos , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/patología , Transducción de Señal
16.
Cell Death Dis ; 7(12): e2505, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27906189

RESUMEN

Inhibition of protein degradation by blocking Cullin-RING E3 ligases (CRLs) is a new approach in cancer therapy though of unknown risk because CRL inhibition may stabilize both oncoproteins and tumor suppressors. Probing CRLs in prostate cancer cells revealed a remarkable plasticity of cells with TMPRSS2-ERG translocation. CRL suppression by chemical inhibition or knockdown of RING component RBX1 led to reversible G0/G1 cell cycle arrest that prevented cell apoptosis. Conversely, complete blocking of CRLs at a higher inhibitor dose-induced cytotoxicity that was amplified by knockdown of CRL regulator Cand1. We analyzed cell signaling to understand how varying degrees of CRL inhibition translated to distinct cell fates. Both tumor suppressor and oncogenic cell signaling pathways and transcriptional activities were affected, with pro-metastatic Wnt/ß-catenin as the most upregulated. Suppression of the NF-κB pathway contributed to anti-apoptotic effect, and androgen receptor (AR) and ERG played decisive, though opposite, roles: AR was involved in protective quiescence, whereas ERG promoted apoptosis. These data define AR-ERG interaction as a key plasticity and survival determinant in prostate cancer and suggest supplementary treatments that may overcome drug resistance mechanisms regulated by AR-ERG interaction.


Asunto(s)
Plasticidad de la Célula , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/metabolismo , Línea Celular Tumoral , Linaje de la Célula/efectos de los fármacos , Plasticidad de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclopentanos/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Modelos Biológicos , Proteína NEDD8 , Pirimidinas/farmacología , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Transcripción Genética/efectos de los fármacos , Regulador Transcripcional ERG/metabolismo
17.
Elife ; 42015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26349035

RESUMEN

Sumoylation during genotoxic stress regulates the composition of DNA repair complexes. The yeast metalloprotease Wss1 clears chromatin-bound sumoylated proteins. Wss1 and its mammalian analog, DVC1/Spartan, belong to minigluzincins family of proteases. Wss1 proteolytic activity is regulated by a cysteine switch mechanism activated by chemical stress and/or DNA binding. Wss1 is required for cell survival following UV irradiation, the smt3-331 mutation and Camptothecin-induced formation of covalent topoisomerase 1 complexes (Top1cc). Wss1 forms a SUMO-specific ternary complex with the AAA ATPase Cdc48 and an adaptor, Doa1. Upon DNA damage Wss1/Cdc48/Doa1 is recruited to sumoylated targets and catalyzes SUMO chain extension through a newly recognized SUMO ligase activity. Activation of Wss1 results in metalloprotease self-cleavage and proteolysis of associated proteins. In cells lacking Tdp1, clearance of topoisomerase covalent complexes becomes SUMO and Wss1-dependent. Upon genotoxic stress, Wss1 is vacuolar, suggesting a link between genotoxic stress and autophagy involving the Doa1 adapter.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Mutágenos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Proteolisis , Sumoilación , Proteína que Contiene Valosina
18.
Sci Rep ; 5: 8336, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25673565

RESUMEN

MiRNAs are key regulators of gene expression. By binding to many genes, they create a complex network of gene co-regulation. Here, using a network-based approach, we identified miRNA hub groups by their close connections and common targets. In one cluster containing three miRNAs, miR-612, miR-661 and miR-940, the annotated functions of the co-regulated genes suggested a role in small GTPase signalling. Although the three members of this cluster targeted the same subset of predicted genes, we showed that their overexpression impacted cell fates differently. miR-661 demonstrated enhanced phosphorylation of myosin II and an increase in cell invasion, indicating a possible oncogenic miRNA. On the contrary, miR-612 and miR-940 inhibit phosphorylation of myosin II and cell invasion. Finally, expression profiling in human breast tissues showed that miR-940 was consistently downregulated in breast cancer tissues.


Asunto(s)
Citoesqueleto de Actina/genética , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Citoesqueleto de Actina/metabolismo , Neoplasias de la Mama/metabolismo , Movimiento Celular/genética , Proliferación Celular , Biología Computacional/métodos , Bases de Datos de Ácidos Nucleicos , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Anotación de Secuencia Molecular , Proteínas de Unión al GTP Monoméricas/metabolismo , Transducción de Señal
19.
Sci Rep ; 5: 14221, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26382112

RESUMEN

Phenotypic screening monitors phenotypic changes induced by perturbations, including those generated by drugs or RNA interference. Currently-used methods for scoring screen hits have proven to be problematic, particularly when applied to physiologically relevant conditions such as low cell numbers or inefficient transfection. Here, we describe the Φ-score, which is a novel scoring method for the identification of phenotypic modifiers or hits in cell-based screens. Φ-score performance was assessed with simulations, a validation experiment and its application to gene identification in a large-scale RNAi screen. Using robust statistics and a variance model, we demonstrated that the Φ-score showed better sensitivity, selectivity and reproducibility compared to classical approaches. The improved performance of the Φ-score paves the way for cell-based screening of primary cells, which are often difficult to obtain from patients in sufficient numbers. We also describe a dedicated merging procedure to pool scores from small interfering RNAs targeting the same gene so as to provide improved visualization and hit selection.

20.
Biomaterials ; 34(38): 10099-108, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24060421

RESUMEN

The behaviour of cancerous epithelial prostatic cells (PC3) growing on polyelectrolytes (PE) coatings was compared to the behaviour of immortalized normal prostatic cells (PNT-2). The cell behaviour was evaluated and quantified in terms of initial cell attachment, growth, metabolic activity, morphometry, adhesion, apoptosis and stress related gene expression. Both the anionic PSS (poly(sodium 4-styrenesulphonate))-terminated surface and cationic PAH (poly(allylamine hydrochloride))-terminated surfaces were not cytotoxic. The initial attachment of cells was better on the PAH-terminated surface compared to fibronectin. However, the proliferation rate of PC3 cells was reduced on the PAH-terminated surface and slightly increased on the PSS coatings. Only PAH prevented the clustering phenotype of PC3 and reduced the number of focal adhesion points as compared to fibronectin or PSS coatings. In contrast, none of the PE surfaces significantly affected the biological responses of PNT-2 cells. PAH-terminating films provide a tool to preferentially modulate the growth of some cancerous phenotypes, in this case as a micro-environment that reduces the growth of metastatic PC3 cells.


Asunto(s)
Polímeros/química , Polímeros/farmacología , Próstata/patología , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Masculino , Modelos Teóricos , Polímeros/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA