Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Acta Biomater ; 58: 12-25, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28576716

RESUMEN

Cancer stem cells (CSCs) are responsible for drug resistance, tumor recurrence, and metastasis in several cancer types, making their eradication a primary objective in cancer therapy. Glioblastoma Multiforme (GBM) tumors are usually composed of a highly infiltrating CSC subpopulation, which has Nestin as a putative marker. Since the majority of these infiltrating cells are able to elude conventional therapies, we have developed gold nanorods (AuNRs) functionalized with an engineered peptide capable of specific recognition and selective eradication of Nestin positive infiltrating GBM-CSCs. These AuNRs generate heat when irradiated by a near-infrared laser, and cause localized cell damage. Nanoparticle internalization assays performed with GBM-CSCs or Nestin negative cells cultured as two-dimensional (2D) monolayers or embedded in three-dimensional (3D) biodegradable-hydrogels of tunable mechanical properties, revealed that the AuNRs were mainly internalized by GBM-CSCs, and not by Nestin negative cells. The AuNRs were taken up via energy-dependent and caveolae-mediated endocytic mechanisms, and were localized inside endosomes. Photothermal treatments resulted in the selective elimination of GBM-CSCs through cell apoptosis, while Nestin negative cells remained viable. Results also indicated that GBM-CSCs embedded in hydrogels were more resistant to AuNR photothermal treatments than when cultured as 2D monolayers. In summary, the combination of our engineered AuNRs with our tunable hydrogel system has shown the potential to provide an in vitro platform for the evaluation and screening of AuNR-based cancer therapeutics, leading to a substantial advancement in the application of AuNRs for targeted GBM-CSC therapy. STATEMENT OF SIGNIFICANCE: There is an urgent need for reliable and efficient therapies for the treatment of Glioblastoma Multiforme (GBM), which is currently an untreatable brain tumor form with a very poor patient survival rate. GBM tumors are mostly comprised of cancer stem cells (CSCs), which are responsible for tumor reoccurrence and therapy resistance. We have developed gold nanorods functionalized with an engineered peptide capable of selective recognition and eradication of GBM-CSCs via heat generation by nanorods upon NIR irradiation. An in vitro evaluation of nanorod therapeutic activities was performed in 3D synthetic-biodegradable hydrogel models with distinct biomechanical cues, and compared to 2D cultures. Results indicated that cells cultured in 3D were more resistant to photothermolysis than in 2D systems.


Asunto(s)
Doxorrubicina , Sistemas de Liberación de Medicamentos , Glioblastoma , Oro , Hidrogeles/química , Nanotubos/química , Péptidos , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Oro/química , Oro/farmacología , Humanos , Péptidos/química , Péptidos/farmacología
2.
Stem Cells Transl Med ; 3(7): 801-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24855275

RESUMEN

We present a method to efficiently culture primary chromaffin progenitors from the adult bovine adrenal medulla in a defined, serum-free monolayer system. Tissue is dissociated and plated for expansion under support by the mitogen basic fibroblast growth factor (bFGF). The cultures, although not homogenous, contain a subpopulation of cells expressing the neural stem cell marker Hes3 that also propagate. In addition, Hes3 is also expressed in the adult adrenal medulla from where the tissue is taken. Differentiation is induced by bFGF withdrawal and switching to Neurobasal medium containing B27. Following differentiation, Hes3 expression is lost, and cells acquire morphologies and biomarker expression patterns of chromaffin cells and dopaminergic neurons. We tested the effect of different treatments that we previously showed regulate Hes3 expression and cell number in cultures of fetal and adult rodent neural stem cells. Treatment of the cultures with a combination of Delta4, Angiopoietin2, and a Janus kinase inhibitor increases cell number during the expansion phase without significantly affecting catecholamine content levels. Treatment with cholera toxin does not significantly affect cell number but reduces the ratio of epinephrine to norepinephrine content and increases the dopamine content relative to total catecholamines. These data suggest that this defined culture system can be used for target identification in drug discovery programs and that the transcription factor Hes3 may serve as a new biomarker of putative adrenomedullary chromaffin progenitor cells.


Asunto(s)
Médula Suprarrenal/metabolismo , Técnicas de Cultivo de Célula , Células Cromafines/metabolismo , Células-Madre Neurales/metabolismo , Factores de Transcripción/metabolismo , Médula Suprarrenal/citología , Médula Suprarrenal/efectos de los fármacos , Angiopoyetina 2/farmacología , Animales , Biomarcadores/metabolismo , Catecolaminas/metabolismo , Bovinos , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Toxina del Cólera/farmacología , Células Cromafines/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/farmacología , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Proteínas de la Membrana/farmacología , Células-Madre Neurales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética
3.
Sci Rep ; 3: 1095, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23393614

RESUMEN

Tumors exhibit complex organization and contain a variety of cell populations. The realization that the regenerative properties of a tumor may be largely confined to a cell subpopulation (cancer stem cell) is driving a new era of anti-cancer research. Cancer stem cells from Glioblastoma Multiforme tumors express markers that are also expressed in non-cancerous neural stem cells, including nestin and Sox2. We previously showed that the transcription factor Hes3 is a marker of neural stem cells, and that its expression is inhibited by JAK activity. Here we show that Hes3 is also expressed in cultures from glioblastoma multiforme which express neural stem cell markers, can differentiate into neurons and glia, and can recapitulate the tumor of origin when transplanted into immunocompromised mice. Similar to observations in neural stem cells, JAK inhibits Hes3 expression. Hes3 RNA interference reduces the number of cultured glioblastoma cells suggesting a novel therapeutic strategy.


Asunto(s)
Neoplasias del Sistema Nervioso Central/patología , Proteínas de Unión al ADN/metabolismo , Glioblastoma/patología , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/metabolismo , Angiopoyetina 2/metabolismo , Animales , Biomarcadores/metabolismo , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/metabolismo , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Janus Quinasa 1/metabolismo , Janus Quinasa 1/farmacología , Ratones , Células Madre Neoplásicas/patología , Fosforilación , ARN Interferente Pequeño , Proteínas Represoras , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/genética , Células Tumorales Cultivadas
4.
Eur J Neurosci ; 34(3): 374-81, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21714817

RESUMEN

It is important to determine the mechanisms controlling the number of neurons in the nervous system. Previously, we reported that neuronal activity plays a central role in controlling neuron number in the neonatal hippocampus of rodents. Neuronal survival requires sustained activation of the serine-threonine kinase Akt, which is initiated by neurotrophins and continued for several hours by neuronal activity and integrin signaling. Here, we focus on the CA3 region to show that neuronal apoptosis requires p53. As in wild-type animals, neuronal death occurs in the first postnatal week and ends by postnatal day (P)10 in p53(-/-) mice. During this period, the CA3 region of p53(-/-) mice contains significantly lower numbers of apoptotic cells, and at the end of the death period, it contains more neurons than the wild type. At P10, the p53(-/-) CA3 region contains a novel subpopulation of neurons with small soma size. These neurons show normal levels of tropomyosin receptor kinase receptor activation, but lower levels of activated Akt than the neurons with somata of normal size. These results suggest that p53 is the key downstream regulator of the novel survival-signaling pathway that regulates the number of CA3 neurons in the first 10 days of postnatal life.


Asunto(s)
Muerte Celular/fisiología , Hipocampo/citología , Neuronas/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Animales Recién Nacidos , Hipocampo/patología , Integrinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Crecimiento Nervioso/metabolismo , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/metabolismo
5.
Proc Natl Acad Sci U S A ; 106(32): 13570-5, 2009 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-19628689

RESUMEN

In Parkinson's disease, multiple cell types in many brain regions are afflicted. As a consequence, a therapeutic strategy that activates a general neuroprotective response may be valuable. We have previously shown that Notch ligands support neural precursor cells in vitro and in vivo. Here we show that neural precursors express the angiopoietin receptor Tie2 and that injections of angiopoietin2 activate precursors in the adult brain. Signaling downstream of Tie2 and the Notch receptor regulate blood vessel formation. In the adult brain, angiopoietin2 and the Notch ligand Dll4 activate neural precursors with opposing effects on the density of blood vessels. A model of Parkinson's disease was used to show that angiopoietin2 and Dll4 rescue injured dopamine neurons with motor behavioral improvement. A combination of growth factors with little impact on the vasculature retains the ability to stimulate neural precursors and protect dopamine neurons. The cellular and pharmacological basis of the neuroprotective effects achieved by these single treatments merits further analysis.


Asunto(s)
Encéfalo/patología , Dopamina/metabolismo , Neuronas/patología , Células Madre/citología , Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Muerte Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor TIE-2/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo
6.
Nature ; 442(7104): 823-6, 2006 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-16799564

RESUMEN

The hope of developing new transplantation therapies for degenerative diseases is limited by inefficient stem cell growth and immunological incompatibility with the host. Here we show that Notch receptor activation induces the expression of the specific target genes hairy and enhancer of split 3 (Hes3) and Sonic hedgehog (Shh) through rapid activation of cytoplasmic signals, including the serine/threonine kinase Akt, the transcription factor STAT3 and mammalian target of rapamycin, and thereby promotes the survival of neural stem cells. In both murine somatic and human embryonic stem cells, these positive signals are opposed by a control mechanism that involves the p38 mitogen-activated protein kinase. Transient administration of Notch ligands to the brain of adult rats increases the numbers of newly generated precursor cells and improves motor skills after ischaemic injury. These data indicate that stem cell expansion in vitro and in vivo, two central goals of regenerative medicine, may be achieved by Notch ligands through a pathway that is fundamental to development and cancer.


Asunto(s)
Receptores Notch/metabolismo , Sistemas de Mensajero Secundario , Células Madre/citología , Células Madre/metabolismo , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Recuento de Células , Diferenciación Celular , División Celular , Supervivencia Celular , Células Cultivadas , Embrión de Mamíferos/citología , Humanos , Ligandos , Ratones , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Ratas , Medicina Regenerativa , Factor de Transcripción STAT3/metabolismo , Serina-Treonina Quinasas TOR , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
J Neurosci ; 24(9): 2277-85, 2004 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-14999078

RESUMEN

Recent studies indicate that neuroprotection afforded by brain-derived neurotrophic factor (BDNF) is mediated by extracellular signal-regulated kinase (ERK) and phosphatidylinositol-3 kinase (PI3K). However, the mechanisms by which ERK and PI3K exert neuroprotection are not completely understood. Because ERK1/2 and PI3K both stimulate serum response element (SRE)-mediated gene expression, and serum response factor (SRF) is indispensable for SRE-mediated transcription, we investigated whether SRF contributes to ERK1/2 and PI3K neuroprotection. To accomplish this goal, we used an established experimental paradigm in which BDNF protects postnatal cortical neurons against both trophic deprivation and camptothecin-induced DNA damage. BDNF protection against camptothecin is mediated primarily by ERK1/2 activation, whereas its protection against trophic deprivation is mainly through stimulation of the PI3K pathway (Hetman et al., 1999). Here we demonstrate that expression of a wild-type SRF is sufficient to protect postnatal cortical neurons against camptothecin or trophic deprivation. Expression of a dominant-negative SRF partially reversed BDNF neuroprotection against both apoptotic insults. Moreover, the dominant-negative SRF inhibited neuroprotection against trophic withdrawal afforded by expression of a constitutive active PI3K. In addition, protection against camptothecin by expression of constitutive active mitogen-activated protein kinase kinase 1, an upstream kinase that activates ERK1/2, was also blocked by expression of the dominant-negative SRF. These data suggest that SRF is both necessary and sufficient for BDNF neuroprotection of cortical neurons against trophic deprivation and DNA damage. Our data provide a direct demonstration of a biological function of SRF in neurons and a novel downstream neuroprotective mechanism common to both ERK1/2 and PI3K pathways.


Asunto(s)
Neuronas/metabolismo , Factor de Respuesta Sérica/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Camptotecina/antagonistas & inhibidores , Camptotecina/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Daño del ADN/fisiología , Activación Enzimática/efectos de los fármacos , Expresión Génica , Genes Dominantes , MAP Quinasa Quinasa 1 , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/biosíntesis , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores de Crecimiento Nervioso/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Respuesta Sérica/biosíntesis , Factor de Respuesta Sérica/genética , Transducción de Señal/fisiología , Transfección
8.
Brain Res Dev Brain Res ; 136(1): 35-42, 2002 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-12036515

RESUMEN

Recently, human protein associated with MYC, PAM, has been cloned and characterized as a large protein that interacts with the transcriptional-activating domain of Myc. The regional expression pattern of PAM in brains has not been yet been defined. Expression patterns of PAM in both rat and mouse brains were examined by using in situ hybridization. Here, we demonstrate that PAM mRNA is highly expressed in specific anatomical regions including hippocampus, dentate gyrus and cerebellum. In these areas, PAM mRNA is restricted to pyramidal cells of hippocampus and granule cells of dentate gyrus and cerebellum. During development, PAM mRNA expression is differentially regulated. It is turned on after birth and up-regulated during the first postnatal 2 weeks. Thereafter, PAM mRNA expression remains elevated into adulthood. The regional distribution of PAM in brain is similar to that observed for several adenylyl cyclase isoforms such as type I isoform. However, no obvious alterations of PAM mRNA expression are detected in brains of mice deficient in type I or type 8 or type 1 and type 8 isoforms of adenylyl cyclase. Thus, adenylyl cyclase does not appear to alter the expression of PAM.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/genética , Cerebelo/crecimiento & desarrollo , Giro Dentado/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Oxigenasas de Función Mixta , Adenilil Ciclasas/metabolismo , Animales , Cerebelo/citología , Cerebelo/fisiología , Giro Dentado/citología , Giro Dentado/fisiología , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Mitosis/fisiología , Neuronas/citología , ARN Mensajero/análisis , Ratas , Ratas Long-Evans , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA