Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neural Regen Res ; 19(9): 1929-1939, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38227517

RESUMEN

Neurogenesis is a tightly regulated process in time and space both in the developing embryo and in adult neurogenic niches. A drastic change in the transcriptome and proteome of radial glial cells or neural stem cells towards the neuronal state is achieved due to sophisticated mechanisms of epigenetic, transcriptional, and post-transcriptional regulation. Understanding these neurogenic mechanisms is of major importance, not only for shedding light on very complex and crucial developmental processes, but also for the identification of putative reprogramming factors, that harbor hierarchically central regulatory roles in the course of neurogenesis and bare thus the capacity to drive direct reprogramming towards the neuronal fate. The major transcriptional programs that orchestrate the neurogenic process have been the focus of research for many years and key neurogenic transcription factors, as well as repressor complexes, have been identified and employed in direct reprogramming protocols to convert non-neuronal cells, into functional neurons. The post-transcriptional regulation of gene expression during nervous system development has emerged as another important and intricate regulatory layer, strongly contributing to the complexity of the mechanisms controlling neurogenesis and neuronal function. In particular, recent advances are highlighting the importance of specific RNA binding proteins that control major steps of mRNA life cycle during neurogenesis, such as alternative splicing, polyadenylation, stability, and translation. Apart from the RNA binding proteins, microRNAs, a class of small non-coding RNAs that block the translation of their target mRNAs, have also been shown to play crucial roles in all the stages of the neurogenic process, from neural stem/progenitor cell proliferation, neuronal differentiation and migration, to functional maturation. Here, we provide an overview of the most prominent post-transcriptional mechanisms mediated by RNA binding proteins and microRNAs during the neurogenic process, giving particular emphasis on the interplay of specific RNA binding proteins with neurogenic microRNAs. Taking under consideration that the molecular mechanisms of neurogenesis exert high similarity to the ones driving direct neuronal reprogramming, we also discuss the current advances in in vitro and in vivo direct neuronal reprogramming approaches that have employed microRNAs or RNA binding proteins as reprogramming factors, highlighting the so far known mechanisms of their reprogramming action.

2.
Cells ; 12(10)2023 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-37408252

RESUMEN

The Neurovascular Unit (NVU), composed of glia (astrocytes, oligodendrocytes, microglia), neurons, pericytes and endothelial cells, is a dynamic interface ensuring the physiological functioning of the central nervous system (CNS), which gets affected and contributes to the pathology of several neurodegenerative diseases. Neuroinflammation is a common feature of neurodegenerative diseases and is primarily related to the activation state of perivascular microglia and astrocytes, which constitute two of its major cellular components. Our studies focus on monitoring in real time the morphological changes of perivascular astrocytes and microglia, as well as their dynamic interactions with the brain vasculature, under physiological conditions and following systemic neuroinflammation triggering both microgliosis and astrogliosis. To this end, we performed 2-photon laser scanning microscopy (2P-LSM) for intravital imaging of the cortex of transgenic mice visualizing the dynamics of microglia and astroglia following neuroinflammation induced by systemic administration of the endotoxin lipopolysaccharide (LPS). Our results indicate that following neuroinflammation the endfeet of activated perivascular astrocytes lose their close proximity and physiological cross-talk with vasculature, an event that most possibly contributes to a loss of blood-brain barrier (BBB) integrity. At the same time, microglial cells become activated and exhibit a higher extent of physical contact with the blood vessels. These dynamic responses of perivascular astrocytes and microglia are peaking at 4 days following LPS administration; however, they still persist at a lower level at 8 days after LPS injection, revealing incomplete reversal of inflammation affecting the glial properties and interactions within the NVU.


Asunto(s)
Astrocitos , Microglía , Animales , Ratones , Astrocitos/patología , Microglía/patología , Lipopolisacáridos/efectos adversos , Enfermedades Neuroinflamatorias , Células Endoteliales/patología , Encéfalo/patología , Inflamación/patología , Ratones Transgénicos
3.
Stem Cell Reports ; 18(4): 915-935, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36963393

RESUMEN

The microRNA (miRNA) miR-124 has been employed supplementary to neurogenic transcription factors (TFs) and other miRNAs to enhance direct neurogenic conversion. The aim of this study was to investigate whether miR-124 is sufficient to drive direct reprogramming of astrocytes to induced neurons (iNs) on its own and elucidate its independent mechanism of reprogramming action. Our data show that miR-124 is a potent driver of the reprogramming switch of astrocytes toward an immature neuronal fate by directly targeting the RNA-binding protein Zfp36L1 implicated in ARE-mediated mRNA decay and subsequently derepressing Zfp36L1 neurogenic interactome. To this end, miR-124 contribution in iNs' production largely recapitulates endogenous neurogenesis pathways, being further enhanced upon addition of the neurogenic compound ISX9, which greatly improves iNs' differentiation and functional maturation. Importantly, miR-124 is potent in guiding direct conversion of reactive astrocytes to immature iNs in vivo following cortical trauma, while ISX9 supplementation confers a survival advantage to newly produced iNs.


Asunto(s)
MicroARNs , Células-Madre Neurales , Astrocitos/metabolismo , Neuronas/metabolismo , Diferenciación Celular/genética , Células-Madre Neurales/metabolismo , MicroARNs/genética , MicroARNs/metabolismo
4.
Int J Dev Biol ; 66(1-2-3): 199-209, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34549796

RESUMEN

Direct reprogramming of glial cells into induced-neurons is a promising strategy for CNS repair after acute injury or neurodegenerative diseases. Grey matter astrocytes, which exhibit features of neural stem cells when activated, are an ideal cell source for direct neuronal conversion. The aim of the study is the investigation of the neuronal reprogramming capacity of CEND1 and/or Neurogenin-2 (NEUROG2) upon their overexpression on primary human adult cortical astrocytes. Our data indicate that adult human cortical astrocytes can be directly reprogrammed by either CEND1 or NEUROG2 to cells with differentiated neuronal morphology, exhibiting long neurites and branched processes. Exploration of gene expression dynamics along the conversion process revealed that neuronal genes are significantly up-regulated while astrocytic genes are down-regulated. Differentiated induced-neurons (iNs) exhibit either GABAergic or glutamatergic/dopaminergic identity upon CEND1 and NEUROG2 overexpression respectively. Co-expression of CEND1 and NEUROG2 in double-transduced cultures induced elevated expression levels of neural progenitor/stem genes and appearance of highly proliferative spheres with neural progenitor cell (NPC) properties in culture.


Asunto(s)
Astrocitos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas de la Membrana , Proteínas del Tejido Nervioso , Células-Madre Neurales , Adulto , Astrocitos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas
5.
Neurobiol Dis ; 157: 105449, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34274460

RESUMEN

Microglia are key players in Multiple Sclerosis (MS), expressing many susceptibility genes for this disease. They constantly survey the brain microenvironment, but the precise functional relationships between microglia and pathological processes remain unknown. We performed a detailed assessment of microglial dynamics in three distinct grey matter regions in a cuprizone-induced demyelination model. We found that microglial activation preceded detectable demyelination and showed regional specificities, such as prominent phagocytic activity in cortical layer 5 and early hypertrophic morphology in hippocampal CA1. Demyelination happened earliest in cortical layer 5, although was more complete in CA1. In cortical layer 2/3, microglial activation and demyelination were less pronounced but microglia became hyper-ramified with slower process movement during remyelination, thereby maintaining local brain surveillance. Profiling of microglia using specific morphological and motility parameters revealed region-specific heterogeneity of microglial responses in the grey matter that might serve as sensitive indicators of progression in CNS demyelinating diseases.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Corteza Cerebral/metabolismo , Enfermedades Desmielinizantes/metabolismo , Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Remielinización , Animales , Región CA1 Hipocampal/patología , Aumento de la Célula , Proliferación Celular , Corteza Cerebral/patología , Quelantes/toxicidad , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Sustancia Gris , Hipocampo/metabolismo , Hipocampo/patología , Imagenología Tridimensional , Ratones , Ratones Noqueados , Microglía/patología , Microscopía Confocal , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/patología , Imagen Óptica , Fagocitosis , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo
6.
Methods Mol Biol ; 2155: 41-61, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32474866

RESUMEN

Direct neuronal reprogramming, rewiring the epigenetic and transcriptional network of a differentiated cell type to neuron, apart from being a very promising approach for the treatment of brain injury and neurodegeneration, also offers a prime opportunity to investigate the molecular underpinnings of neuronal cell fate determination, as the precise molecular mechanisms that establish neuronal fate and diversity at the transcriptional and epigenetic level are incompletely understood. Recent studies from a number of groups, including ours, have shown that astrocytes can be directly reprogrammed into functional neurons in vitro and in vivo following ectopic overexpression of combinations of transcription factors, neurogenic proteins, miRNAs, and small chemical molecules.In this chapter we describe the protocols for in vitro converting primary cortical astrocytes of mouse and human origin to induced neurons, through forced expression of two neurogenic molecules, either each one alone or in combination: the master regulatory bHLH proneural transcription factor NEUROGENIN-2 (NEUROG2) and the neurogenic protein CEND1. Forced expression of each one of the two neurogenic proteins in primary astrocytes via retroviral gene transfer results in their direct conversion to subtype-specific induced neurons, while simultaneous coexpression of both molecules drives them predominantly toward acquisition of a neural precursor cell (NPC) state. Although mouse and human astrocytes exhibit differences in their reprogramming rate and particular characteristics, they can both get efficiently in vitro transdifferentiated to NPCs and induced neurons upon NEUROG2 or/and CEND1 forced expression using the reprogramming protocols described in the chapter, presenting valuable cellular platforms for mechanistic studies and in vivo applications to restore neurodegeneration.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Transdiferenciación Celular , Técnicas de Reprogramación Celular , Reprogramación Celular , Neuronas/citología , Neuronas/metabolismo , Animales , Biomarcadores , Técnicas de Cultivo de Célula , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Marcación de Gen , Vectores Genéticos/genética , Humanos , Ratones , Microscopía Confocal , Medicina Regenerativa , Imagen de Lapso de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Breast Cancer Res ; 21(1): 132, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796128

RESUMEN

BACKGROUND: ERBB-2 is overexpressed in about 20% of breast cancers (BCs), indicating poor prognosis. The receptor activator of nuclear factor-κB (RANK) pathway is implicated in ERBB-2 (+) BC. The purpose of this study was to elucidate the underlying molecular mechanism of this interaction and the beneficial impact of dual targeting of RANK and ERBB-2 pathways. METHODS: We used SKBR3, MCF7, MDA-MB-453, and BT-474 human BC cell lines. We examined RANK and RANKL expression using RT-PCR, Western blot, and immunofluorescence. The evaluation of RANK expression in a cohort of BC patients was performed using immunohistochemistry. The interaction between RANK and ERBB family members was detected using proximity ligation assay (PLA), which enables the visualization of interacting proteins. We used inhibitors of both pathways [trastuzumab (T), pertuzumab (P), denosumab (D)]. NF-κB pathway activation was studied using Western blot. Cell growth and viability was evaluated using XTT, flow cytometry, and clonogenic assay. For cell migration evaluation, scratch assay was performed. Data were analyzed by one-way ANOVA. RESULTS: Cell lines express RANK and RANKL. RANK immunostaining was also detected in human BC tissue samples. RANK receptor dimerizes with ERBB family members. RANK/ERBB-2 dimer number seems to be associated with ERBB-2 expression (SKBR3, 5.4; BT-474, 8.2; MCF7, 0.7; MDA-MB-453, 0.3). RANK/ERBB-2 dimers were decreased in the presence of the inhibitors D, T, and P, while they were increased after RANKL (R) treatment in SKBR3 (m, 5.4; D, 1.2; T, 1.9; DT, 0.6; TP, 1; DTP, 0.4; R, 11.8) and BT-474 (m, 8.2; D, 3.1; T, 4.3; DT, 0.7; TP, 3.4; DTP, 3.2; R, 11.6). Combination targeting of SKBR3 further decreased NF-κB pathway activation compared to single targeting. In SKBR3, RANKL and ERBB-2 blockage resulted in reduced cell proliferation, increased apoptosis, and lower metastatic potential compared to mock cells (m) and reversed values in RANKL presence. The combination treatment of SKBR3 with D, T, and P had an advantage in functional traits compared to single targeting. Denosumab suppressed NF-κB signaling and diminished proliferation rate in MDA-MB-453 cells. MCF7 did not correspond to inhibitors. CONCLUSIONS: The results indicate a novel physical and molecular association between ERBB-2 and RANK pathways that affects ERBB-2 (+) BC growth. We also present data suggesting that the combination of anti-ERBB-2 agents and RANKL inhibitors have a potential direct anti-tumor effect and should be further tested in certain BC patients.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/metabolismo , Receptores ErbB/antagonistas & inhibidores , Terapia Molecular Dirigida , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Cultivadas , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Cell Rep ; 29(4): 932-945.e7, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31644914

RESUMEN

Local cues in the adult neurogenic niches dynamically regulate homeostasis in neural stem cells, whereas their identity and associated molecular mechanisms remain poorly understood. Here, we show that corticotropin-releasing hormone (CRH), the major mediator of mammalian stress response and a key neuromodulator in the adult brain, is necessary for hippocampal neural stem cell (hiNSC) activity under physiological conditions. In particular, we demonstrate functionality of the CRH/CRH receptor (CRHR) system in mouse hiNSCs and conserved expression in humans. Most important, we show that genetic deficiency of CRH impairs hippocampal neurogenesis, affects spatial memory, and compromises hiNSCs' responsiveness to environmental stimuli. These deficits have been partially restored by virus-mediated CRH expression. Additionally, we provide evidence that local disruption of the CRH/CRHR system reduces neurogenesis, while exposure of adult hiNSCs to CRH promotes neurogenic activity via BMP4 suppression. Our findings suggest a critical role of CRH in adult neurogenesis, independently of its stress-related systemic function.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Hipocampo/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Animales , Línea Celular , Células Cultivadas , Hormona Liberadora de Corticotropina/genética , Hipocampo/citología , Hipocampo/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Memoria Espacial
9.
Neurosci Bull ; 35(6): 979-995, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31079319

RESUMEN

We used Drosophila melanogaster as an experimental model to express mouse and pig BM88/CEND1 (cell cycle exit and neuronal differentiation 1) in order to investigate its potential functional effects on Drosophila neurogenesis. BM88/CEND1 is a neuron-specific protein whose function is implicated in triggering cells to exit from the cell cycle and differentiate towards a neuronal phenotype. Transgenic flies expressing either mouse or pig BM88/CEND1 in the nervous system had severe neuronal phenotypes with variable expressivity at various stages of embryonic development. In early embryonic stage 10, BM88/CEND1 expression led to an increase in the neural-specific antigenicity of neuroectoderm at the expense of precursor cells [neuroblasts (Nbs) and ganglion mother cells (GMCs)] including the defective formation and differentiation of the MP2 precursors, whereas at later stages (12-15), protein accumulation induced gross morphological defects primarily in the CNS accompanied by a reduction of Nb and GMC markers. Furthermore, the neuronal precursor cells of embryos expressing BM88/CEND1 failed to carry out proper cell-cycle progression as revealed by the disorganized expression patterns of specific cell-cycle markers. BM88/CEND1 accumulation in the Drosophila eye affected normal eye disc development by disrupting the ommatidia. Finally, we demonstrated that expression of BM88/CEND1 modified/reduced the levels of activated MAP kinase indicating a functional effect of BM88/CEND1 on the MAPK signaling pathway. Our findings suggest that the expression of mammalian BM88/CEND1 in Drosophila exerts specific functional effects associated with neuronal precursor cell formation during embryonic neurogenesis and proper eye disc development. This study also validates the use of Drosophila as a powerful model system in which to investigate gene function and the underlying molecular mechanisms.


Asunto(s)
Diferenciación Celular/fisiología , Drosophila melanogaster/embriología , Desarrollo Embrionario/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Fenómenos Fisiológicos del Sistema Nervioso , Sistema Nervioso/patología , Animales , Proliferación Celular , Embrión no Mamífero , Ojo/patología , Femenino , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Sistema Nervioso/metabolismo , Neurogénesis , Neuronas/metabolismo , Transducción de Señal , Células Madre/fisiología , Porcinos
10.
J Cancer Res Clin Oncol ; 145(5): 1221-1233, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30805773

RESUMEN

PURPOSE: Androgen receptor (AR) is playing an important role in the progression of a subset of TNBC. We evaluated the impact of ERß expression along with anti-AR drugs in AR-positive TNBC. METHODS: ERß expression was examined in AR-positive TNBC cell line using MTT assay, scratch and Annexin V-FITC assay in the presence or absence of anti-androgens. Protein levels of involved molecules were assessed using Western blot. Receptors' localization was detected by immunofluorescence and their physical association was examined using proximity ligation assay (PLA), which enables the visualization of interacting proteins in fixed cells and tissues. RESULTS: Transient transfection of ERß in MDA-MB 453 AR-positive TNBC cell line significantly inhibited cell proliferation, metastatic potential and induced apoptosis. ERß expression reversed the aggravating role of AR in both indirect and direct ways. Indirectly, ERß decreased AR activation through the inhibition of PI3K/AKT signaling pathway. Directly, ERß formed heterodimers with AR in MDA-MB 453 cells and in human tissue samples impeding AR from forming homodimers. Enzalutamide is a more potent anti-androgen in AR + TNBC compared to bicalutamide. ERß expression increased the sensitivity of MDA-MB 453 cells to anti-androgens and especially to enzalutamide. The administration of enzalutamide enhanced AR:ERß heterodimers formation increasing the anti-tumor capacity of ERß. CONCLUSIONS: Collectively, our results provide evidence for a novel mechanism by which ERß exerts oncosuppressive effect in AR-positive TBNC through direct and indirect interactions with AR. Moreover, ERß expression may identify a new subset of TNBC that would respond more favorable to anti-androgens.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Receptor beta de Estrógeno/metabolismo , Feniltiohidantoína/análogos & derivados , Receptores Androgénicos/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Apoptosis , Benzamidas , Biomarcadores , Línea Celular Tumoral , Proliferación Celular , Receptor beta de Estrógeno/química , Receptor beta de Estrógeno/genética , Expresión Génica , Humanos , Inmunohistoquímica , Nitrilos , Feniltiohidantoína/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Androgénicos/química , Receptores Androgénicos/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
11.
Front Neurosci ; 12: 800, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30450032

RESUMEN

Huntington's Disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the exon-1 of the IT15 gene encoding the protein Huntingtin. Expression of mutated Huntingtin in humans leads to dysfunction and ultimately degeneration of selected neuronal populations of the striatum and cerebral cortex. Current available HD therapy relies on drugs to treat chorea and control psychiatric symptoms, however, no therapy has been proven to slow down disease progression or prevent disease onset. Thus, although 24 years have passed since HD gene identification, HD remains a relentless progressive disease characterized by cognitive dysfunction and motor disability that leads to death of the majority of patients, on average 10-20 years after its onset. Up to now several molecular pathways have been implicated in the process of neurodegeneration involved in HD and have provided potential therapeutic targets. Based on these data, approaches currently under investigation for HD therapy aim on the one hand at getting insight into the mechanisms of disease progression in a human-based context and on the other hand at silencing mHTT expression by using antisense oligonucleotides. An innovative and still poorly investigated approach is to identify new factors that increase neurogenesis and/or induce reprogramming of endogenous neuroblasts and parenchymal astrocytes to generate new healthy neurons to replace lost ones and/or enforce neuroprotection of pre-existent striatal and cortical neurons. Here, we review studies that use human disease-in-a-dish models to recapitulate HD pathogenesis or are focused on promoting in vivo neurogenesis of endogenous striatal neuroblasts and direct neuronal reprogramming of parenchymal astrocytes, which combined with neuroprotective protocols bear the potential to re-establish brain homeostasis lost in HD.

12.
Neuropharmacology ; 117: 408-421, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28219718

RESUMEN

The Regulator of G protein Signalling 4 (RGS4) is a multitask protein that interacts with and negatively modulates opioid receptor signalling. Previously, we showed that the δ-opioid receptor (δ-OR) forms a multiprotein signalling complex consisting of Gi/Go proteins and the Signal Transducer and Activator of Transcription 5B (STAT5B) that leads to neuronal differentiation and neurite outgrowth upon δ-ΟR activation. Here, we investigated whether RGS4 could participate in signalling pathways to regulate neurotropic events. We demonstrate that RGS4 interacts directly with STAT5B independently of δ-ΟR presence both in vitro and in living cells. This interaction involves the N-terminal portion of RGS4 and the DNA-binding SH3 domain of STAT5B. Expression of RGS4 in HEK293 cells expressing δ-OR and/or erythropoietin receptor results in inhibition of [D-Ser2, Leu5, Thr6]-enkephalin (DSLET)-and erythropoietin-dependent STAT5B phosphorylation and subsequent transcriptional activation. DSLET-dependent neurite outgrowth of neuroblastoma cells is also blocked by RGS4 expression, whereas primary cortical cultures of RGS4 knockout mice (RGS4-/-) exhibit enhanced neuronal sprouting after δ-OR activation. Additional studies in adult brain extracts from RGS4-/- mice revealed increased levels of p-STAT5B. Finally, neuronal progenitor cultures from RGS4-/- mice exhibit enhanced proliferation with concomitant increases in the mRNA levels of the anti-apoptotic STAT5B target genes bcl2 and bcl-xl. These observations suggest that RGS4 is implicated in opioid dependent neuronal differentiation and neurite outgrowth via a "non-canonical" signaling pathway regulating STAT5B-directed responses.


Asunto(s)
Neurogénesis/fisiología , Proyección Neuronal/fisiología , Neuronas/metabolismo , Proteínas RGS/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/fisiología , Corteza Cerebral/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/metabolismo , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas RGS/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Eritropoyetina/metabolismo , Receptores Opioides delta/metabolismo , Proteína bcl-X/metabolismo
13.
Cancer Biol Ther ; 17(8): 799-812, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259689

RESUMEN

Breast cancer stem cells (BCSC) have been identified in breast carcinoma as CD44(+)/CD24(-/low) cells, which display tumorigenic activity and have the ability to self-renew, differentiate and metastasize. Previous studies showed that extracellular HSP90 (eHSP90) participates in the invasion and metastatic processes of various cancers including breast cancer. Here, we show for the first time that eHSP90 is over-expressed in mammosphere cultures that are derived from the MDA-MB-231, MDA-MB-453 and MCF-7 breast cancer cell lines. These mammospheres are highly enriched in cells of the CD44(+)/CD24(-/low) BCSC phenotype and additionally show high expression of the BCSC markers CD49f and Sox2. Thus our results indicate that eHSP90 represents a potential novel BCSC marker. Moreover, we present evidence that eHSP90 is functionally involved in BCSC activity in vitro and in vivo. Selective neutralization of eHSP90, using the monoclonal antibody mAb 4C5, has the capacity to inhibit stem cell activity in vitro because the formation of mammosphere-derived colonies is dramatically reduced in its presence. In vivo, the treatment of mice with mAb4C5 using a prophylactic protocol, significantly inhibited the primary growth of MDA-MB-231 and mammosphere-derived tumors. More importantly, administration of this antibody in a therapeutic protocol caused a statistically significant regression of established tumors derived from MDA-MB-231 originating mammospheres. Tumor regression was even greater when mAb 4C5 was administered in combination with paclitaxel. Overall, our findings implicate eHSP90 as a potential novel BCSC biomarker. Moreover they show that eHSP90 participates in BCSC-derived primary tumor growth. Finally, we provide additional support for the possible therapeutic value of mAb4C5 in the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas HSP90 de Choque Térmico/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones
14.
Stem Cell Reports ; 5(3): 405-18, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26321141

RESUMEN

Recent studies demonstrate that astroglia from non-neurogenic brain regions can be reprogrammed into functional neurons through forced expression of neurogenic factors. Here we explored the effect of CEND1 and NEUROG2 on reprogramming of mouse cortical astrocytes and embryonic fibroblasts. Forced expression of CEND1, NEUROG2, or both resulted in acquisition of induced neuronal cells expressing subtype-specific markers, while long-term live-cell imaging highlighted the existence of two different modes of neuronal trans-differentiation. Of note, a subpopulation of CEND1 and NEUROG2 double-transduced astrocytes formed spheres exhibiting neural stem cell properties. mRNA and protein expression studies revealed a reciprocal feedback loop existing between the two molecules, while knockdown of endogenous CEND1 demonstrated that it is a key mediator of NEUROG2-driven neuronal reprogramming. Our data suggest that common reprogramming mechanisms exist driving the conversion of lineage-distant somatic cell types to neurons and reveal a critical role for CEND1 in NEUROG2-driven astrocytic reprogramming.


Asunto(s)
Astrocitos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Reprogramación Celular , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Animales , Astrocitos/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Embrión de Mamíferos/citología , Fibroblastos/citología , Proteínas de la Membrana/genética , Ratones , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Neuronas/citología
15.
Methods Mol Biol ; 1210: 9-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25173157

RESUMEN

The central nervous system (CNS) can be damaged by a wide range of conditions resulting in loss of specific populations of neurons and/or glial cells and in the development of defined psychiatric or neurological symptoms of varying severity. As the CNS has limited inherent capacity to regenerate lost tissue and self-repair, the development of therapeutic strategies for the treatment of CNS insults remains a serious scientific challenge with potential important clinical applications. In this context, strategies involving transplantation of specific cell populations, such as stem cells and neural stem cells (NSCs), to replace damaged cells offers an opportunity for the development of cell-based therapies. Along these lines, in this review we describe a protocol which involves transplantation of NPCs, genetically engineered to overexpress the neurogenic molecule Cend1 and have thus the potency to differentiate with higher frequency towards the neuronal lineage in a rodent model of stab wound cortical injury.


Asunto(s)
Lesiones Encefálicas/terapia , Células-Madre Neurales/citología , Trasplante de Células Madre , Animales , Conducta Animal , Técnicas de Cultivo de Célula , Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Ratones , Ratones Transgénicos , Transducción Genética
16.
CNS Neurol Disord Drug Targets ; 11(7): 829-35, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23131156

RESUMEN

The most prominent pathological feature in Parkinson's disease (PD) is the progressive and selective loss of mesencephalic dopaminergic neurons of the nigrostriatal tract. The present study was conducted in order to investigate whether naive and or genetically modified neural stem/precursor cells (NPCs) can survive, differentiate and functionally integrate in the lesioned striatum. To this end, stereotaxic injections of 6-OHDA in the right ascending nigrostriatal dopaminergic pathway of mice and subsequent NPC transplantations were performed, followed by apomorphine-induced rotations and double-immunofluorescence experiments. Our results demonstrate that transplanted embryonic NPCs derived from the cortical ventricular zone of E14.5 transgenic mouse embryos expressing the green fluorescent protein (GFP) under control of the beta-actin promoter and cultured as neurospheres can survive in the host striatum for at least three weeks after transplantation. The percentage of surviving GFP-positive cells in the host striatum ranges from 0.2% to 0.6% of the total transplanted NPCs. Grafted cells functionally integrate in the striatum, as indicated by the statistically significant decrease of contralateral rotations after apomorphine treatment. Furthermore, we show that within the striatal environment GFP-positive cells differentiate into beta-III tubulin-expressing neurons, but not glial cells. Most importantly, GFP-positive cells further differentiate to dopaminergic (TH-positive) and medium size spiny (DARPP-32- positive) neuronal phenotypes. Over-expression of the cell cycle exit and neuronal differentiation protein Cend1 in NPCs enhances the generation of GABAergic, but not dopaminergic, neuronal phenotypes after grafting in the lesioned striatum. Our results encourage the development of strategies involving NPC transplantation for the treatment of neurodegenerative diseases.


Asunto(s)
Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Células-Madre Neurales/trasplante , Neuronas/patología , Enfermedad de Parkinson/terapia , Sustancia Negra/patología , Animales , Antiparkinsonianos/uso terapéutico , Apomorfina/uso terapéutico , Conducta Animal/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cuerpo Estriado/efectos de los fármacos , Embrión de Mamíferos/citología , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/patología , Supervivencia de Injerto , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidopamina , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Sustancia Negra/efectos de los fármacos , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
17.
Stem Cells ; 29(1): 89-98, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21280160

RESUMEN

Neural stem/progenitor cells maintain their identity via continuous self-renewal and suppression of differentiation. Gain-of-function experiments in the chick revealed an involvement for Sox1-3 transcription factors in the maintenance of the undifferentiated neural progenitor (NP) identity. However, the mechanism(s) employed by each factor has not been resolved. Here, we derived cortical neural/stem progenitor cells from wild-type and Sox1-null mouse embryos and found that Sox1 plays a key role in the suppression of neurogenic cell divisions. Loss of Sox1 leads to progressive depletion of self-renewing cells, elongation of the cell cycle of proliferating cells, and significant increase in the number of cells exiting the cell cycle. In proliferating NP cells, Sox1 acts via a prospero-related homeobox 1 (Prox1)-mediated pathway to block cell cycle exit that leads to neuronal differentiation in vivo and in vitro. Thus, our results demonstrate that Sox1 regulates the size of the cortical NP pool via suppression of Prox1-mediated neurogenic cell divisions.


Asunto(s)
Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Factores de Transcripción SOXB1/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Bromodesoxiuridina/análisis , Ciclo Celular/genética , Ciclo Celular/fisiología , División Celular/genética , División Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Técnicas de Sustitución del Gen , Proteínas de Homeodominio/genética , Inmunohistoquímica , Ratones , Ratones Mutantes , Células-Madre Neurales/citología , Neurogénesis/genética , Factores de Transcripción SOXB1/genética , Proteínas Supresoras de Tumor/genética
18.
Curr Gene Ther ; 11(2): 90-100, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21291359

RESUMEN

The inability of the central nervous system (CNS) to efficiently repair damages results in severe functional impairment after trauma or neurodegenerative/demyelinating diseases. Regeneration failure is attributed to inhibitory molecules creating a nonpermissive environment for axonal regrowth, and dictates the necessity for the development of novel therapeutic strategies. An emerging approach for improving regeneration is the use of gene therapy to manipulate cell adhesion molecule expression in experimental animal models of degeneration. Alternatively, cell transplantation to replace lost neurons and the grafting of myelinating cells to repair demyelinating lesions are promising approaches for treating CNS injuries and demyelination. Schwann cells (SCs), oligodendrocyte progenitors, olfactory ensheathing cells and embryonic and neural stem cells have been shown to form myelin after transplantation into the demyelinated CNS. The repair capacity of the peripheral nervous system (PNS) is much higher, but there is still a limit to the amount of nerve loss that can be bridged after injury, and longer nerve gaps call for the use of conduits populated with living cells. In both cases, the interaction of grafted cells with the host environment is of paramount importance for the incorporation and functional integration of these cells and the manipulation of cell adhesion molecules is an attractive approach towards achieving this goal. In this review we summarize data from the recent literature regarding the manipulation of cell adhesion molecule expression towards CNS and PNS repair and discuss the prospects for future therapeutic applications.


Asunto(s)
Moléculas de Adhesión Celular/genética , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Terapia Genética/métodos , Enfermedades del Sistema Nervioso/terapia , Animales , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/metabolismo , Trasplante de Células , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo
19.
J Neurochem ; 115(5): 1137-49, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20846298

RESUMEN

For biotechnological applications, insect cell lines are primarily known as hosts for the baculovirus expression system that is capable to direct synthesis of high levels of recombinant proteins through use of powerful viral promoters. Here, we demonstrate the implementation of two alternative approaches based on the baculovirus system for production of a mammalian recombinant glycoprotein, comprising the extracellular part of the cell adhesion molecule L1, with potential important therapeutic applications in nervous system repair. In the first approach, the extracellular part of L1 bearing a myc tag is produced in permanently transformed insect cell lines and purified by affinity chromatography. In the second approach, recombinant baculoviruses that express L1-Fc chimeric protein, derived from fusion of the extracellular part of L1 with the Fc part of human IgG1, under the control of a mammalian promoter are used to infect mammalian HEK293 and primary Schwann cells. Both the extracellular part of L1 bearing a myc tag accumulating in the supernatants of insect cultures as well as L1-Fc secreted by transduced HEK293 or Schwann cells are capable of increasing the motility of Schwann cells with similar efficiency in a gap bridging bioassay. In addition, baculovirus-transduced Schwann cells show enhanced motility when grafted on organotypic cultures of neonatal brain slices while they retain their ability to myelinate CNS axons. This proof-of-concept that the migratory properties of myelin-forming cells can be modulated by recombinant protein produced in insect culture as well as by means of baculovirus-mediated adhesion molecule expression in mammalian cells may have beneficial applications in the field of CNS therapies.


Asunto(s)
Baculoviridae/metabolismo , Movimiento Celular/fisiología , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Células de Schwann/fisiología , Animales , Animales Recién Nacidos , Antígeno CD146/química , Antígeno CD146/metabolismo , Movimiento Celular/genética , Células Cultivadas , Cromatografía de Afinidad/métodos , Técnicas de Cocultivo/métodos , Regulación de la Expresión Génica/genética , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Insectos , Ratones , Vaina de Mielina/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/química , Molécula L1 de Adhesión de Célula Nerviosa/genética , Prosencéfalo/citología , Prosencéfalo/fisiología , Ratas , Ratas Wistar , Nervio Ciático/citología , Transducción Genética/métodos
20.
J Neurochem ; 115(2): 460-74, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20681949

RESUMEN

Strategies to enhance neural stem/precursor cell (NPC) capacity to yield multipotential, proliferative, and migrating pools of cells that can efficiently differentiate into neurons could be crucial for structural repair after neurodegenerative damage. Here, we have generated a lentiviral vector for expression of insulin-like growth factor-I (IGF-1) and investigated the impact of IGF-1 transduction on the properties of cultured NPCs (IGF-1-NPCs). Under proliferative conditions, IGF-1 transduction promoted cell cycle progression via cyclin D1 up-regulation and Akt phosphorylation. Remarkably upon differentiation-inducing conditions, IGF-1-NPCs cease to proliferate and differentiate to a greater extent into neurons with significantly longer neurites, at the expense of astrocytes. Moreover, using live imaging we provide evidence that IGF-1 transduction enhances the motility and tissue penetration of grafted NPCs in cultured cortical slices. These results illustrate the important consequence of IGF-1 transduction in regulating NPC functions and offer a potential strategy to enhance the prospective repair potential of NPCs.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neuronas/fisiología , Células Madre/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/citología , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Ventrículos Cerebrales/citología , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Citometría de Flujo/métodos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Hidroxiurea/farmacología , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/farmacología , Lentivirus/fisiología , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Transducción Genética/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...