Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 24(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36675105

RESUMEN

Neuroblastoma is a neural crest cell-derived pediatric tumor characterized by high inter- and intra-tumor heterogeneity, and by a poor outcome in advanced stages. Patient-derived xenografts (PDXs) have been shown to be useful models for preserving and expanding original patient biopsies in vivo, and for studying neuroblastoma biology in a more physiological setting. The maintenance of genetic, histologic, and phenotypic characteristics of the original biopsy along serial PDX passages in mice is a major concern regarding this model. Here we analyze consecutive PDX passages in mice, at both transcriptomic and histological levels, in order to identify potential changes or highlight similarities to the primary sample. We studied temporal changes using mRNA and miRNA expression and correlate those with neuroblastoma aggressiveness using patient-derived databases. We observed a shortening of tumor onset and an increase in proliferative potential in the PDXs along serial passages. This behavior correlates with changes in the expression of genes related to cell proliferation and neuronal differentiation, including signaling pathways described as relevant for neuroblastoma malignancy. We also identified new genes and miRNAs that can be used to stratify patients according to survival, and which could be potential new players in neuroblastoma aggressiveness. Our results highlight the usefulness of the PDX neuroblastoma model and reflect phenotypic changes that might be occurring in the mouse environment. These findings could be useful for understanding the progression of tumor aggressiveness in this pathology.


Asunto(s)
MicroARNs , Neuroblastoma , Humanos , Animales , Ratones , Pase Seriado , Neuroblastoma/metabolismo , Transcriptoma , Perfilación de la Expresión Génica , Proliferación Celular , MicroARNs/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Neuroinflammation ; 13(1): 276, 2016 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-27769255

RESUMEN

BACKGROUND: The CCAAT/enhancer-binding protein ß (C/EBPß) is a transcription factor implicated in the control of proliferation, differentiation, and inflammatory processes mainly in adipose tissue and liver; although more recent results have revealed an important role for this transcription factor in the brain. Previous studies from our laboratory indicated that CCAAT/enhancer-binding protein ß is implicated in inflammatory process and brain injury, since mice lacking this gene were less susceptible to kainic acid-induced injury. More recently, we have shown that the complement component 3 gene (C3) is a downstream target of CCAAT/enhancer-binding protein ß and it could be a mediator of the proinflammatory effects of this transcription factor in neural cells. METHODS: Adult male Wistar rats (8-12 weeks old) were used throughout the study. C/EBPß+/+ and C/EBPß-/- mice were generated from heterozygous breeding pairs. Animals were injected or not with kainic acid, brains removed, and brain slices containing the hippocampus analyzed for the expression of both CCAAT/enhancer-binding protein ß and C3. RESULTS: In the present work, we have further extended these studies and show that CCAAT/enhancer-binding protein ß and C3 co-express in the CA1 and CA3 regions of the hippocampus after an excitotoxic injury. Studies using CCAAT/enhancer-binding protein ß knockout mice demonstrate a marked reduction in C3 expression after kainic acid injection in these animals, suggesting that indeed this protein is regulated by C/EBPß in the hippocampus in vivo. CONCLUSIONS: Altogether these results suggest that CCAAT/enhancer-binding protein ß could regulate brain disorders, in which excitotoxic and inflammatory processes are involved, at least in part through the direct regulation of C3.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Complemento C3/genética , Agonistas de Aminoácidos Excitadores/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo , Ácido Kaínico/toxicidad , Degeneración Nerviosa/inducido químicamente , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Antígeno CD11b/metabolismo , Complemento C3/metabolismo , Modelos Animales de Enfermedad , Fluoresceínas/metabolismo , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Transgénicos , Degeneración Nerviosa/patología , Neuroglía/metabolismo , Neuroglía/patología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
3.
J Neuroinflammation ; 12: 14, 2015 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-25617152

RESUMEN

BACKGROUND: The CCAAT/enhancer-binding protein ß (C/EBPß) is a transcription factor, which was first identified as a regulator of differentiation and inflammatory processes mainly in adipose tissue and liver; however, its function in the brain was largely unknown for many years. Previous studies from our laboratory indicated that C/EBPß is implicated in inflammatory process and brain injury, since mice lacking this gene were less susceptible to kainic acid-induced injury. METHODS: We first performed cDNA microarrays analysis using hippocampal RNA isolated from C/EBPß (+/+) and C/EBPß (-/-) mice. Immunocytochemical and immunohistochemical studies were done to evaluate C/EBPß and C3 levels. Transient transfection experiments were made to analyze transcriptional regulation of C3 by C/EBPß. To knockdown C/EBPß and C3 expression, mouse astrocytes were infected with lentiviral particles expressing an shRNA specific for C/EBPß or an siRNA specific for C3. RESULTS: Among the genes displaying significant changes in expression was complement component 3 (C3), which showed a dramatic decrease in mRNA content in the hippocampus of C/EBPß (-/-) mice. C3 is the central component of the complement and is implicated in different brain disorders. In this work we have found that C/EBPß regulates C3 levels in rodents glial in vitro and in the rat Substantia nigra pars compacta (SNpc) in vivo following an inflammatory insult. Analysis of the mouse C3 promoter showed that it is directly regulated by C/EBPß through a C/EBPß consensus site located at position -616/-599 of the gene. In addition, we show that depletion of C/EBPß by a specific shRNA results in a significant decrease in the levels of C3 together with a reduction in the increased levels of pro-inflammatory agents elicited by lipopolysaccharide treatment. CONCLUSIONS: Altogether, these results indicate that C3 is a downstream target of C/EBPß, and it could be a mediator of the pro-inflammatory effects of this transcription factor in neural cells.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Complemento C3/genética , Regulación de la Expresión Génica/genética , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular Tumoral , Células Cultivadas , Complemento C3/metabolismo , Ciclooxigenasa 2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/citología , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Neuroblastoma , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Fragmentos de Péptidos/metabolismo , Polisacáridos/farmacología , Ratas , Ratas Wistar
4.
Neuro Oncol ; 25(2): 248-260, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35608632

RESUMEN

BACKGROUND: Glioblastoma (GBM) is a highly lethal malignancy for which neoangiogenesis serves as a defining hallmark. The anti-VEGF antibody, bevacizumab, has been approved for the treatment of recurrent GBM, but resistance is universal. METHODS: We analyzed expression data of GBM patients treated with bevacizumab to discover potential resistance mechanisms. Patient-derived xenografts (PDXs) and cultures were interrogated for effects of phosphofructokinase-1, muscle isoform (PFKM) loss on tumor cell motility, migration, and invasion through genetic and pharmacologic targeting. RESULTS: We identified PFKM as a driver of bevacizumab resistance. PFKM functions dichotomize based on subcellular location: cytosolic PFKM interacted with KIF11, a tubular motor protein, to promote tumor invasion, whereas nuclear PFKM safeguarded genomic stability of tumor cells through interaction with NBS1. Leveraging differential transcriptional profiling, bupivacaine phenocopied genetic targeting of PFKM, and enhanced efficacy of bevacizumab in preclinical GBM models in vivo. CONCLUSION: PFKM drives novel molecular pathways in GBM, offering a translational path to a novel therapeutic paradigm.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Bevacizumab/farmacología , Bevacizumab/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Fosfofructoquinasa-1 , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo
5.
J Neurochem ; 122(6): 1193-202, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22774807

RESUMEN

Increased levels of glutamate causing excitotoxic damage accompany many neurological disorders. A well-characterized model of excitotoxic damage involves administration of kainic acid (KA), which causes limbic seizure activity and subsequent neuronal death, particularly in the CA1 and CA3 areas of the hippocampus. Inhibition of the enzyme glycogen synthase kinase-3 (GSK-3) and cAMP levels might play an important role in neuroprotection. As intracellular cAMP levels depend, in part, on the activity of the phosphodiesterase enzymes (PDEs), these enzymes have recently emerged as potential therapeutic targets for the treatment of several diseases. In previous works, we have shown a potent anti-inflammatory and neuroprotective effect of GSK-3 inhibition in a model of excitotoxicity, as well as a reduction of nigrostriatal dopaminergic neuronal cell death after phosphodiesterase 7 inhibition, which leads to an increase in cAMP levels. This study was undertaken to determine whether simultaneous inhibition of GSK-3 and PDE-7 by a novel 5-imino-1,2,4-thiadiazole compound, named VP1.14, could prevent the massive neuronal loss in the hippocampus evoked by intrahippocampal injection of KA. Here, we show that rats treated with VP1.14 showed a reduced inflammatory response after KA injection, and exhibited a significant reduction in pyramidal cell loss in the CA1 and CA3 areas of the hippocampus. Studies with hippocampal HT22 cells in vitro also showed a clear neuroprotective effect of VP1.14 and an anti-inflammatory effect shown by a decrease in the nitrite liberation and in the expression of pro-inflammatory cytokines by primary cultures of astrocytes treated with lipopolysaccharide.


Asunto(s)
Agonistas de Aminoácidos Excitadores/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/patología , Fármacos Neuroprotectores/farmacología , Tiadiazoles/farmacología , Factores de Edad , Animales , Animales Recién Nacidos , Línea Celular , Hipocampo/metabolismo , Inyecciones Intralesiones , Masculino , Ratones , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Ratas , Ratas Wistar
6.
Biomedicines ; 10(2)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35203541

RESUMEN

Adult diffuse glioma, particularly glioblastoma (GBM), is a devastating tumor of the central nervous system. The existential threat of this disease requires on-going treatment to counteract tumor progression. The present outcome is discouraging as most patients will succumb to this disease. The low cure rate is consistent with the failure of first-line therapy, radiation and temozolomide (TMZ). Even with their therapeutic mechanism of action to incur lethal DNA lesions, tumor growth remains undeterred. Delivering additional treatments only delays the inescapable development of therapeutic tolerance and disease recurrence. The urgency of establishing lifelong tumor control needs to be re-examined with a greater focus on eliminating resistance. Early genomic and transcriptome studies suggest each tumor subtype possesses a unique molecular network to safeguard genome integrity. Subsequent seminal work on post-therapy tumor progression sheds light on the involvement of DNA repair as the causative contributor for hypermutation and therapeutic failure. In this review, we will provide an overview of known molecular factors that influence the engagement of different DNA repair pathways, including targetable vulnerabilities, which can be exploited for clinical benefit with the use of specific inhibitors.

7.
Nat Commun ; 13(1): 5303, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36085345

RESUMEN

The RNA world is changing our views about sensing and resolution of DNA damage. Here, we develop single-molecule DNA/RNA analysis approaches to visualize how nascent RNA facilitates the repair of DNA double-strand breaks (DSBs). RNA polymerase II (RNAPII) is crucial for DSB resolution in human cells. DSB-flanking, RNAPII-generated nascent RNA forms RNA:DNA hybrids, guiding the upstream DNA repair steps towards favouring the error-free Homologous Recombination (HR) pathway over Non-Homologous End Joining. Specific RNAPII inhibitor, THZ1, impairs recruitment of essential HR proteins to DSBs, implicating nascent RNA in DNA end resection, initiation and execution of HR repair. We further propose that resection factor CtIP interacts with and helps re-activate RNAPII when paused by the RNA:DNA hybrids, collectively promoting faithful repair of chromosome breaks to maintain genomic integrity.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Reparación del ADN por Unión de Extremidades , Humanos , ARN/genética , ARN Polimerasa II , Reparación del ADN por Recombinación
8.
J Med Chem ; 64(3): 1423-1434, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33502198

RESUMEN

Despite the recent advances in cancer therapeutics, highly aggressive cancer forms, such as glioblastoma (GBM), still have very low survival rates. The intracellular scaffold protein syntenin, comprising two postsynaptic density protein-95/discs-large/zona occludens-1 (PDZ) domains, has emerged as a novel therapeutic target in highly malignant phenotypes including GBM. Here, we report the development of a novel, highly potent, and metabolically stable peptide inhibitor of syntenin, KSL-128114, which binds the PDZ1 domain of syntenin with nanomolar affinity. KSL-128114 is resistant toward degradation in human plasma and mouse hepatic microsomes and displays a global PDZ domain selectivity for syntenin. An X-ray crystal structure reveals that KSL-128114 interacts with syntenin PDZ1 in an extended noncanonical binding mode. Treatment with KSL-128114 shows an inhibitory effect on primary GBM cell viability and significantly extends survival time in a patient-derived xenograft mouse model. Thus, KSL-128114 is a novel promising candidate with therapeutic potential for highly aggressive tumors, such as GBM.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Péptidos/química , Péptidos/farmacología , Sinteninas/efectos de los fármacos , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Ratones , Microsomas/metabolismo , Modelos Moleculares , Mutación , Unión Proteica , Difracción de Rayos X , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Nat Commun ; 11(1): 4709, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32948765

RESUMEN

Glioblastoma cancer-stem like cells (GSCs) display marked resistance to ionizing radiation (IR), a standard of care for glioblastoma patients. Mechanisms underpinning radio-resistance of GSCs remain largely unknown. Chromatin state and the accessibility of DNA lesions to DNA repair machineries are crucial for the maintenance of genomic stability. Understanding the functional impact of chromatin remodeling on DNA repair in GSCs may lay the foundation for advancing the efficacy of radio-sensitizing therapies. Here, we present the results of a high-content siRNA microscopy screen, revealing the transcriptional elongation factor SPT6 to be critical for the genomic stability and self-renewal of GSCs. Mechanistically, SPT6 transcriptionally up-regulates BRCA1 and thereby drives an error-free DNA repair in GSCs. SPT6 loss impairs the self-renewal, genomic stability and tumor initiating capacity of GSCs. Collectively, our results provide mechanistic insights into how SPT6 regulates DNA repair and identify SPT6 as a putative therapeutic target in glioblastoma.


Asunto(s)
Reparación del ADN , Inestabilidad Genómica , Glioblastoma/genética , Células Madre Neoplásicas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Apoptosis , Proteína BRCA1 , Neoplasias Encefálicas/genética , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Glioblastoma/patología , Células HEK293 , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Células Madre Neoplásicas/patología , ARN Interferente Pequeño/genética , Tolerancia a Radiación , Radiación Ionizante , Transcriptoma
10.
EBioMedicine ; 49: 82-95, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31685444

RESUMEN

BACKGROUND: Neuroblastoma is a paediatric tumour originated from sympathoadrenal precursors and characterized by its heterogeneity and poor outcome in advanced stages. Intra-tumoral cellular heterogeneity has emerged as an important feature in neuroblastoma, with a potential major impact on tumour aggressiveness and response to therapy. CD44 is an adhesion protein involved in tumour progression, metastasis and stemness in different cancers; however, there has been controversies about the significance of CD44 expression in neuroblastoma and its relationship with tumour progression. METHODS: We have performed transcriptomic analysis on patient tumour samples studying the outcome of patients with high CD44 expression. Adhesion, invasion and proliferation assays were performed in sorted CD44high neuroblastoma cells. Tumoursphere cultures have been used to enrich in undifferentiated stem-like cells and to asses self-renewal and differentiation potential. We have finally performed in vivo tumorigenic assays on cell line-derived or Patient-derived xenografts. FINDINGS: We show that high CD44 expression is associated with low survival in high-grade human neuroblastoma, independently of MYCN amplification. CD44 is expressed in a cell population with neural crest stem-like features, and with the capacity to generate multipotent, undifferentiated tumourspheres in culture. These cells are more invasive and proliferative in vitro. CD44 positive cells obtained from tumours are more tumorigenic and metastatic, giving rise to aggressive neuroblastic tumours at high frequency upon transplantation. INTERPRETATION: We describe an unexpected intra-tumoural heterogeneity within cellular entities expressing CD44 in neuroblastoma, and propose that CD44 has a role in neural crest stem-like undifferentiated cells, which can contribute to tumorigenesis and malignancy in this type of cancer. FUNDING: Research supported by grants from the "Asociación Española contra el Cáncer" (AECC), the Spanish Ministry of Science and Innovation SAF program (SAF2016-80412-P), and the European Research Council (ERC Starting Grant to RP).


Asunto(s)
Receptores de Hialuranos/metabolismo , Cresta Neural/patología , Células-Madre Neurales/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Ratones SCID , Células Madre Multipotentes/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Esferoides Celulares/patología , Análisis de Supervivencia
11.
Sci Rep ; 7(1): 13526, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29051532

RESUMEN

The CCAAT/Enhancer binding protein ß (C/EBPß) is a transcription factor involved in numerous physiological as well as pathological conditions in the brain. However, little is known regarding its possible role in neurodegenerative disorders. We have previously shown that C/EBPß regulates the expression of genes involved in inflammatory processes and brain injury. Here, we have analyzed the effects of C/EBPß interference in dopaminergic cell death and glial activation in the 6-hydroxydopamine model of Parkinson's disease. Our results showed that lentivirus-mediated C/EBPß deprivation conferred marked in vitro and in vivo neuroprotection of dopaminergic cells concomitant with a significant attenuation of the level of the inflammatory response and glial activation. Additionally, C/EBPß interference diminished the induction of α-synuclein in the substantia nigra pars compacta of animals injected with 6-hydroxydopamine. Taking together, these results reveal an essential function for C/EBPß in the pathways leading to inflammatory-mediated brain damage and suggest novel roles for C/EBPß in neurodegenerative diseases, specifically in Parkinson's disease, opening the door for new therapeutic interventions.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Enfermedad de Parkinson/patología , Animales , Apoptosis/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/genética , Células Cultivadas , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Humanos , Masculino , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Oxidopamina/farmacología , Enfermedad de Parkinson/metabolismo , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Porción Compacta de la Sustancia Negra/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , alfa-Sinucleína/metabolismo
12.
Oncotarget ; 8(52): 89775-89792, 2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-29163787

RESUMEN

Pediatric tumors arise upon oncogenic transformation of stem/progenitor cells during embryonic development. Given this scenario, the existence of non-tumorigenic stem cells included within the aberrant tumoral niche, with a potential role in tumor biology, is an intriguing and unstudied possibility. Here, we describe the presence and function of non-tumorigenic neural crest-derived progenitor cells in aggressive neuroblastoma (NB) tumors. These cells differentiate into neural crest typical mesectodermal derivatives, giving rise to tumor stroma and promoting proliferation and tumor aggressiveness. Furthermore, an analysis of gene expression profiles in stage 4/M NB revealed a neural crest stem cell (NCSC) gene signature that was associated to stromal phenotype and high probability of relapse. Thus, this NCSC gene expression signature could be used in prognosis to improve stratification of stage 4/M NB tumors. Our results might facilitate the design of new therapies by targeting NCSCs and their contribution to tumor stroma.

13.
Oncotarget ; 6(6): 4369-84, 2015 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25738360

RESUMEN

We have previously shown that decreased expression of CCAAT/enhancer binding protein ß (C/EBPß) inhibits the growth of glioblastoma cells and diminishes their transformation capacity and migration. In agreement with this, we showed that C/EBPß depletion decreases the mRNA levels of different genes involved in metastasis and invasion. Among these, we found S100 calcium binding protein A4 (S100A4) to be almost undetectable in glioblastoma cells deficient in C/EBPß. Here, we have evaluated the possible role of S100A4 in the observed effects of C/EBPß in glioblastoma cells and the mechanism through which S100A4 levels are controlled by C/EBPß. Our results show that C/EBPß suppression significantly reduced the levels of S100A4 in murine GL261 and human T98G glioblastoma cells. By employing an S100A4-promoter reporter, we observed a significant induction in the transcriptional activation of the S100A4 gene by C/EBPß. Furthermore, overexpression of S100A4 in C/EBPß-depleted glioblastoma cells reverses the enhanced migration and motility induced by this transcription factor. Our data also point to a role of S100A4 in glioblastoma cell invasion and suggest that the C/EBPß gene controls the invasive potential of GL261 and T98G cells through direct regulation of S100A4. Finally, this study indicates a role of C/EBPß on the maintenance of the stem cell population present in GL261 glioblastoma cells.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Glioblastoma/patología , Proteínas S100/biosíntesis , Animales , Western Blotting , Línea Celular Tumoral , Movimiento Celular/fisiología , Inmunoprecipitación de Cromatina , Humanos , Ratones , Invasividad Neoplásica/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína de Unión al Calcio S100A4 , Activación Transcripcional/fisiología , Transfección
14.
Neurobiol Aging ; 36(2): 1160-73, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25457552

RESUMEN

Different studies have suggested that the nucleotide cyclic adenosine 3', 5'-monophosphate can actively play an important role as a neuroprotective and anti-inflammatory agent after a brain injury. The phosphodiesterase 7 (PDE7) enzyme is one of the enzymes responsible for controlling specifically the intracellular levels of cyclic adenosine 3', 5'-monophosphate in the immune and central nervous systems. Therefore, this enzyme could play an important role in brain inflammation and neurodegeneration. In this regard, using different chemical inhibitors of PDE7 we have demonstrated their neuroprotective and anti-inflammatory activity in different models of neurodegenerative disorders, including Parkinson's disease (PD). In the present study, we have used the toxin 6-hydroxydopamine and lipopolysaccharide to model PD and explore the protective effects of PDE7B deficiency in dopaminergic neurons cell death. Lentivirus-mediated PDE7B deprivation conferred marked in vitro and in vivo neuroprotection against 6-hydroxydopamine and lipopolysaccharide toxicity in dopaminergic neurons and preserved motor function involving the dopamine system in mouse. Our results substantiate previous data and provide a validation of PDE7B enzyme as a valuable new target for therapeutic development in the treatment of PD.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/antagonistas & inhibidores , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/genética , Silenciador del Gen , Lentivirus/genética , Actividad Motora/genética , Degeneración Nerviosa/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/terapia , Interferencia de ARN , ARN Interferente Pequeño/genética , Animales , Células Cultivadas , Sistema Nervioso Central/metabolismo , AMP Cíclico/metabolismo , AMP Cíclico/fisiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/deficiencia , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/fisiología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/fisiología , Encefalitis/genética , Humanos , Sistema Inmunológico/metabolismo , Masculino , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología
17.
PLoS One ; 6(10): e24842, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22003384

RESUMEN

BACKGROUND: The dentate gyrus of the hippocampus is one of the regions in which neurogenesis takes place in the adult brain. We have previously demonstrated that CCAAT/enhancer binding protein ß (C/EBPß) is expressed in the granular layer of the dentate gyrus of the adult mouse hippocampus. Taking into account the important role of C/EBPß in the consolidation of long term memory, the fact that newborn neurons in the hippocampus contribute to learning and memory processes, and the role of this transcription factor, previously demonstrated by our group, in regulating neuronal differentiation, we speculated that this transcription factor could regulate stem/progenitor cells in this region of the brain. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show, using C/EBPß knockout mice, that C/EBPß expression is observed in the subset of newborn cells that proliferate in the hippocampus of the adult brain. Mice lacking C/EBPß present reduced survival of newborn cells in the hippocampus, a decrease in the number of these cells that differentiate into neurons and a diminished number of cells that are proliferating in the subgranular zone of the dentate gyrus. These results were further confirmed in vitro. Neurosphere cultures from adult mice deficient in C/EBPß present less proliferation and neuronal differentiation than neurospheres derived from wild type mice. CONCLUSIONS/SIGNIFICANCE: In summary, using in vivo and in vitro strategies, we have identified C/EBPß as a key player in the proliferation and survival of the new neurons produced in the adult mouse hippocampus. Our results support a novel role of C/EBPß in the processes of adult hippocampal neurogenesis, providing new insights into the mechanisms that control neurogenesis in this region of the brain.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Giro Dentado/citología , Giro Dentado/metabolismo , Neurogénesis , Animales , Animales Recién Nacidos , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Giro Dentado/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
18.
PLoS One ; 5(11): e13879, 2010 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-21079728

RESUMEN

BACKGROUND: Thiadiazolidinones (TDZD) are small heterocyclic compounds first described as non-ATP competitive inhibitors of glycogen synthase kinase 3ß (GSK-3ß). In this study, we analyzed the effects of 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), on murine GL261 cells growth in vitro and on the growth of established intracerebral murine gliomas in vivo. METHODOLOGY/PRINCIPAL FINDINGS: Our data show that TDZD-8 decreased proliferation and induced apoptosis of GL261 glioblastoma cells in vitro, delayed tumor growth in vivo, and augmented animal survival. These effects were associated with an early activation of extracellular signal-regulated kinase (ERK) pathway and increased expression of EGR-1 and p21 genes. Also, we observed a sustained activation of the ERK pathway, a concomitant phosphorylation and activation of ribosomal S6 kinase (p90RSK) and an inactivation of GSK-3ß by phosphorylation at Ser 9. Finally, treatment of glioblastoma stem cells with TDZD-8 resulted in an inhibition of proliferation and self-renewal of these cells. CONCLUSIONS/SIGNIFICANCE: Our results suggest that TDZD-8 uses a novel mechanism to target glioblastoma cells, and that malignant progenitor population could be a target of this compound.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glioblastoma/prevención & control , Tiadiazoles/farmacología , Carga Tumoral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Caspasa 2/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Glioblastoma/metabolismo , Glioblastoma/patología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Immunoblotting , Inmunohistoquímica , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/metabolismo , Trasplante de Neoplasias , Fosforilación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA