Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Intervalo de año de publicación
1.
Cell Transplant ; 26(4): 669-677, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28403913

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder, characterized by the loss of dopaminergic neurons in the substantia nigra and their projections to the striatum. Several processes have been described as potential inducers of the dopaminergic neuron death, such as inflammation, oxidative stress, and mitochondrial dysfunction. However, the death of dopaminergic neurons seems to be multifactorial, and its cause remains unclear. ATP-activating purinergic receptors influence various physiological functions in the CNS, including neurotransmission. Purinergic signaling is also involved in pathological scenarios, where ATP is extensively released and promotes sustained purinergic P2X7 receptor (P2X7R) activation and consequent induction of cell death. This effect occurs, among other factors, by oxidative stress and during the inflammatory response. On the other hand, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) is involved in energy metabolism and mitochondrial biogenesis. Expression and activity upregulation of this protein has been related with reduction of oxidative stress and neuroprotection. Therefore, P2X7R and PGC-1α are potential targets in the treatment of PD. Here hemiparkinsonism was induced by unilateral stereotactic injection of 6-OHDA in a rat model. After 7 days, the establishment of PD was confirmed and followed by treatment with the P2X7R antagonist Brilliant Blue G (BBG) or PGC-1α agonist fenofibrate. BBG, but not fenofibrate, reverted hemiparkinsonian behavior accompanied by an increase in tyrosine hydroxylase immunoreactivity in the substantia nigra. Our results suggest that the P2X7R may be a therapeutic target in Parkinson's disease.


Asunto(s)
Dopamina/metabolismo , Fenofibrato/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Colorantes de Rosanilina/uso terapéutico , Animales , Western Blotting , Modelos Animales de Enfermedad , Fenofibrato/farmacología , Masculino , Enfermedad de Parkinson/patología , Ratas Sprague-Dawley , Colorantes de Rosanilina/farmacología , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/metabolismo
2.
J Cell Sci ; 129(18): 3437-48, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528403

RESUMEN

During brain development, cells proliferate, migrate and differentiate in highly accurate patterns. In this context, published results indicate that bradykinin functions in neural fate determination, favoring neurogenesis and migration. However, mechanisms underlying bradykinin function are yet to be explored. Our findings indicate a previously unidentified role for bradykinin action in inducing neuron-generating division in vitro and in vivo, given that bradykinin lengthened the G1-phase of the neural progenitor cells (NPC) cycle and increased TIS21 (also known as PC3 and BTG2) expression in hippocampus from newborn mice. This role, triggered by activation of the kinin-B2 receptor, was conditioned by ERK1/2 activation. Moreover, immunohistochemistry analysis of hippocampal dentate gyrus showed that the percentage of Ki67(+) cells markedly increased in bradykinin-treated mice, and ERK1/2 inhibition affected this neurogenic response. The progress of neurogenesis depended on sustained ERK phosphorylation and resulted in ERK1/2 translocation to the nucleus in NPCs and PC12 cells, changing expression of genes such as Hes1 and Ngn2 (also known as Neurog2). In agreement with the function of ERK in integrating signaling pathways, effects of bradykinin in stimulating neurogenesis were reversed following removal of protein kinase C (PKC)-mediated sustained phosphorylation.


Asunto(s)
Bradiquinina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/enzimología , Neuronas/metabolismo , Animales , Calcio/metabolismo , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones Endogámicos C57BL , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Células PC12 , Fenotipo , Fosforilación/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos
3.
Neurochem Res ; 40(10): 2091-101, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25758980

RESUMEN

Acetylcholinesterase (AChE) inhibition has been described as the main mechanism of organophosphate (OP)-evoked toxicity. OPs represent a human health threat, because chronic exposure to low doses can damage the developing brain, and acute exposure can produce long-lasting damage to adult brains, despite post-exposure medical countermeasures. Although the main mechanism of OP toxicity is AChE inhibition, several lines of evidence suggest that OPs also act by other mechanisms. We hypothesized that rat neural progenitor cells extracted on embryonic day 14.5 would be affected by constant inhibition of AChE from chronic exposure to OP or pyridostigmine (a reversible AChE blocker) during differentiation. In this work, the OP paraoxon decreased cell viability in concentrations >50 µM, as measured with the MTT assay; however, this effect was not dose-dependent. Reduced viability could not be attributed to blockade of AChE activity, since treatment with 200 µM pyridostigmine did not affect cell viability, even after 6 days. Although changes in protein expression patterns were noted in both treatments, the distribution of differentiated phenotypes, such as the percentages of neurons and glial cells, was not altered, as determined by flow cytometry. Since paraoxon and pyridostigmine each decreased neurite outgrowth (but did not prevent differentiation), we infer that developmental patterns may have been affected.


Asunto(s)
Acetilcolinesterasa/metabolismo , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Bromuro de Piridostigmina/farmacología , Animales , Encéfalo/efectos de los fármacos , Células Cultivadas , Inhibidores de la Colinesterasa/farmacología , Células-Madre Neurales/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/metabolismo , Paraoxon
4.
Cytometry A ; 83(1): 76-89, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23044513

RESUMEN

The identification and isolation of multipotent neural stem and progenitor cells in the brain, giving rise to neurons, astrocytes, and oligodendrocytes initiated many studies in order to understand basic mechanisms of endogenous neurogenesis and repair mechanisms of the nervous system and to develop novel therapeutic strategies for cellular regeneration therapies in brain disease. A previous review (Trujillo et al., Cytometry A 2009;75:38-53) focused on the importance of extrinsic factors, especially neurotransmitters, for directing migration and neurogenesis in the developing and adult brain. Here, we extend our review discussing the effects of the principal growth and neurotrophic factors as well as their intracellular signal transduction on neurogenesis, fate determination and neuroprotective mechanisms. Many of these mechanisms have been elucidated by in vitro studies for which neural stem cells were isolated, grown as neurospheres, induced to neural differentiation under desired experimental conditions, and analyzed for embryonic, progenitor, and neural marker expression by flow and imaging cytometry techniques. The better understanding of neural stem cells proliferation and differentiation is crucial for any therapeutic intervention aiming at neural stem cell transplantation and recruitment of endogenous repair mechanisms.


Asunto(s)
Encéfalo/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Factores de Crecimiento Nervioso/fisiología , Neurogénesis/fisiología , Animales , Encefalopatías/fisiopatología , Diferenciación Celular/fisiología , Proliferación Celular , Humanos , Ratones , Células-Madre Neurales/fisiología , Ratas , Transducción de Señal/fisiología
5.
J Biol Chem ; 287(53): 44046-61, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23132855

RESUMEN

Bradykinin is not only important for inflammation and blood pressure regulation, but also involved in neuromodulation and neuroprotection. Here we describe novel functions for bradykinin and the kinin-B2 receptor (B2BkR) in differentiation of neural stem cells. In the presence of the B2BkR antagonist HOE-140 during rat neurosphere differentiation, neuron-specific ß3-tubulin and enolase expression was reduced together with an increase in glial protein expression, indicating that bradykinin-induced receptor activity contributes to neurogenesis. In agreement, HOE-140 affected in the same way expression levels of neural markers during neural differentiation of murine P19 and human iPS cells. Kinin-B1 receptor agonists and antagonists did not affect expression levels of neural markers, suggesting that bradykinin-mediated effects are exclusively mediated via B2BkR. Neurogenesis was augmented by bradykinin in the middle and late stages of the differentiation process. Chronic treatment with HOE-140 diminished eNOS and nNOS as well as M1-M4 muscarinic receptor expression and also affected purinergic receptor expression and activity. Neurogenesis, gliogenesis, and neural migration were altered during differentiation of neurospheres isolated from B2BkR knock-out mice. Whole mount in situ hybridization revealed the presence of B2BkR mRNA throughout the nervous system in mouse embryos, and less ß3-tubulin and more glial proteins were expressed in developing and adult B2BkR knock-out mice brains. As a underlying transcriptional mechanism for neural fate determination, HOE-140 induced up-regulation of Notch1 and Stat3 gene expression. Because pharmacological treatments did not affect cell viability and proliferation, we conclude that bradykinin-induced signaling provides a switch for neural fate determination and specification of neurotransmitter receptor expression.


Asunto(s)
Diferenciación Celular , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Receptor de Bradiquinina B2/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Wistar , Receptor de Bradiquinina B2/genética , Transducción de Señal
6.
J Biol Chem ; 287(35): 29690-701, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22730318

RESUMEN

The diffusible messenger NO plays multiple roles in neuroprotection, neurodegeneration, and brain plasticity. Argininosuccinate synthase (AS) is a ubiquitous enzyme in mammals and the key enzyme of the NO-citrulline cycle, because it provides the substrate L-arginine for subsequent NO synthesis by inducible, endothelial, and neuronal NO synthase (NOS). Here, we provide evidence for the participation of AS and of the NO-citrulline cycle in the progress of differentiation of neural stem cells (NSC) into neurons, astrocytes, and oligodendrocytes. AS expression and activity and neuronal NOS expression, as well as l-arginine and NO(x) production, increased along neural differentiation, whereas endothelial NOS expression was augmented in conditions of chronic NOS inhibition during differentiation, indicating that this NOS isoform is amenable to modulation by extracellular cues. AS and NOS inhibition caused a delay in the progress of neural differentiation, as suggested by the decreased percentage of terminally differentiated cells. On the other hand, BDNF reversed the delay of neural differentiation of NSC caused by inhibition of NO(x) production. A likely cause is the lack of NO, which up-regulated p75 neurotrophin receptor expression, a receptor required for BDNF-induced differentiation of NSC. We conclude that the NO-citrulline cycle acts together with BDNF for maintaining the progress of neural differentiation.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diferenciación Celular/fisiología , Citrulina/metabolismo , Células-Madre Neurales/metabolismo , Óxido Nítrico/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Células Cultivadas , Regulación Enzimológica de la Expresión Génica/fisiología , Proteínas del Tejido Nervioso , Células-Madre Neurales/citología , Neuronas/citología , Neuronas/metabolismo , Óxido Nítrico Sintasa/biosíntesis , Oligodendroglía/citología , Oligodendroglía/metabolismo , Ratas , Ratas Wistar , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/metabolismo
7.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2D.9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22415841

RESUMEN

This unit describes the culture and induction of in vitro models of neural differentiation and strategies to evaluate the participation of extrinsic and intrinsic factors in modulation of this process. Protocols focus on large-scale expansion of pluripotent P19 murine embryonic carcinoma cells and their induction to neural differentiation in the presence of retinoic acid, closely resembling conditions of early neuroectodermal differentiation. Procedures are also described for obtaining rat neural precursor cells (NPCs) or neurospheres and for differentiating them in the absence of growth factors. Experimental strategies are reported using P19 cells and NPCs as in vitro models for studying the actions of extrinsic and intrinsic factors on morphology, proliferation, viability, neural phenotype determination, and progress of differentiation, as well as the functionality of ion channels and metabotropic receptors in inducing calcium fluxes at different developmental stages. The methods described here may be useful for optimizing in vitro protocols for stem cell differentiation into defined neural populations, as well as for studying mechanisms that underlie neurogenesis and gliogenesis.


Asunto(s)
Diferenciación Celular , Forma de la Célula , Células Madre de Carcinoma Embrionario/patología , Neuronas/citología , Esferoides Celulares/citología , Animales , Apoptosis , Bromodesoxiuridina/metabolismo , Calcio/metabolismo , Proliferación Celular , Supervivencia Celular , Corteza Cerebral/citología , Criopreservación , Cuerpos Embrioides/citología , Inmunohistoquímica , Mesencéfalo/citología , Ratones , Microscopía Confocal , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Esferoides Celulares/metabolismo
8.
Int J Dev Neurosci ; 30(2): 91-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22248690

RESUMEN

Embryonic carcinoma cells are widely used models for studying the mechanisms of proliferation and differentiation occurring during early embryogenesis. We have now investigated how down-regulation of P2X2 and P2X7 receptor expression by RNA interference (RNAi) affects neural differentiation and phenotype specification of P19 embryonal carcinoma cells. Wild-type P19 embryonal carcinoma cells or cells stably expressing shRNAs targeting P2X2 or P2X7 receptor expression were induced to differentiate into neurons and glial cells in the presence of retinoic acid. Silencing of P2X2 receptor expression along differentiation promoted cell proliferation and an increase in the percentage of cells expressing glial-specific GFAP, while the presence of beta-3 tubulin-positive cells diminished at the same time. Proliferation induction in the presence of stable anti-P2X2 receptor RNAi points at a mechanism where glial proliferation is favored over growth arrest of progenitor cells which would allow neuronal maturation. Differently from the P2X2 receptor, inhibition of P2X7 receptor expression during neural differentiation of P19 cells resulted in a decrease in cell proliferation and GFAP expression, suggesting the need of functional P2X7 receptors for the progress of gliogenesis. The results obtained in this study indicate the importance of purinergic signaling for cell fate determination during neural differentiation, with P2X2 and P2X7 receptors promoting neurogenesis and gliogenesis, respectively. The shRNAs down-regulating P2X2 or P2X7 receptor gene expression, developed during this work, present useful tools for studying mechanisms of neural differentiation in other stem cell models.


Asunto(s)
Células Madre de Carcinoma Embrionario/citología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuroglía/citología , Neuronas/citología , Receptores Purinérgicos P2X2/fisiología , Receptores Purinérgicos P2X7/fisiología , Tretinoina/fisiología , Animales , Diferenciación Celular/genética , Células Madre de Carcinoma Embrionario/metabolismo , Células Madre de Carcinoma Embrionario/fisiología , Ratones , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Neurogénesis/genética , Neuroglía/metabolismo , Neuroglía/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Interferencia de ARN/fisiología , Receptores Purinérgicos P2X2/genética , Receptores Purinérgicos P2X7/genética , Transducción de Señal/genética
9.
J Mol Neurosci ; 44(3): 141-6, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20617399

RESUMEN

Neural differentiation has been extensively studied in vitro in a model termed neurospheres, which consists of aggregates of neural progenitor cells. Previous studies suggest that they have a great potential for the treatment of neurological disorders. One of the major challenges for scientists is to control cell fate and develop ideal culture conditions for neurosphere expansion in vitro, without altering their features. Similar to human neural progenitors, rat neurospheres cultured in the absence of epidermal and fibroblast growth factors for a short period increased the levels of ß-3 tubulin and decreased the expression of glial fibrillary acidic protein and nestin, compared to neurospheres cultured in the presence of these factors. In this work, we show that rat neurospheres cultured in suspension under mitogen-free condition presented significant higher expression of P2X2 and P2X6 receptor subunits, when compared to cells cultured in the presence of growth factors, suggesting a direct relationship between P2X2/6 receptor expression and induction of neuronal differentiation in mitogen-free cultured rat neurospheres.


Asunto(s)
Células-Madre Neurales/fisiología , Neurogénesis , Neuronas/fisiología , Receptores Purinérgicos P2X2/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Células-Madre Neurales/citología , Neuronas/citología , Ratas , Ratas Wistar , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2X2/genética
10.
An Acad Bras Cienc ; 81(3): 443-52, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19722014

RESUMEN

Cell therapy for neurological disorders has advanced, and neural precursor cells (NPC) may become the ideal candidates for neural transplantation in a wide range of diseases. However, additional work has to be done to determine either the ideal culture environment for NPC expansion in vitro, without altering their plasticity, or the FGF-2 and EGF mechanisms of cell signaling in neurospheres growth, survival and differentiation. In this work we evaluated mouse neurospheres cultured with and without FGF-2 and EGF containing medium and showed that those growth factors are responsible for NPC proliferation. It is also demonstrated that endogenous production of growth factors shifts from FGF-2 to IGF-1/PDGFb upon EGF and FGF-2 withdrawal. Mouse NPC cultured in suspension showed different patterns of neuronal localization (core versus shell) for both EGF and FGF-2 withdrawal and control groups. Taken together, these results show that EGF and FGF-2 removal play an important role in NPC differentiation and may contribute to a better understanding of mechanisms of NPC differentiation. Our findings suggest that depriving NPC of growth factors prior to grafting might enhance their chance to effectively integrate into the host.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/fisiología , Células Madre/citología , Células Madre/fisiología
11.
An. acad. bras. ciênc ; 81(3): 443-452, Sept. 2009. ilus, graf
Artículo en Inglés | LILACS | ID: lil-523972

RESUMEN

Cell therapy for neurological disorders has advanced, and neural precursor cells (NPC) may become the ideal candidates for neural transplantation in a wide range of diseases. However, additional work has to be done to determine either the ideal culture environment for NPC expansion in vitro, without altering their plasticity, or the FGF-2 and EGF mechanisms of cell signaling in neurospheres growth, survival and differentiation. In this work we evaluated mouse neurospheres cultured with and without FGF-2 and EGF containing medium and showed that those growth factors are responsible for NPC proliferation. It is also demonstrated that endogenous production of growth factors shifts from FGF-2 to IGF-1/PDGFb upon EGF and FGF-2 withdrawal. Mouse NPC cultured in suspension showed different patterns of neuronal localization (core versus shell) for both EGF and FGF-2 withdrawal and control groups. Taken together, these results show that EGF and FGF-2 removal play an important role in NPC differentiation and may contribute to a better understanding of mechanisms of NPC differentiation. Our findings suggest that depriving NPC of growth factors prior to grafting might enhance their chance to effectively integrate into the host.


As terapias celulares para doenças neurológicas têm avançado e células precursoras neurais (NPC) surgem como candidatas ideais para o transplante de células neurais em muitas doenças. No entanto, trabalhos adicionais devem ser feitos para determinar o ambiente de cultivo ideal para a expansão in vitro das NPC, sem alterar sua plasticidade, e os mecanismos de sinalização celular do fator de crescimento epidérmico (EGF) e fator de crescimento de fibroblasto 2 (FGF-2) no crescimento, sobrevivência e diferenciação da neuroesfera. Nesse trabalho avaliamosNPCcultivadas na presença e na ausência de FGF-2 e EGF e mostramos que esses fatores de crescimento são responsáveis pela proliferação das NPC. Também foi demonstrado que a produção endógena de fatores de crescimento alterna de FGF-2 a fator de crescimento de insulina 1 (IGF-1) e fator de crescimento derivado de plaquetas b (PDGFb) após remoção de EGF e FGF-2. NPC de camundongo cultivadas em suspensão mostraram padrões de localização neuronal distintos (centro versus borda) tanto no grupo controle como no grupo sem EGF e FGF-2. Juntos, esses resultados mostram que a remoção de EGF e FGF-2 exerce importante ação na diferenciação de NPC e possivelmente contribui para melhor compreensão dos mecanismos envolvidos na diferenciação. Nossos achados sugerem que, privando as NPC de fatores de crescimento antes do transplante, talvez aumente as chances de que as células efetivamente se integrem ao hospedeiro.


Asunto(s)
Animales , Ratones , Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , /farmacología , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Células Madre/efectos de los fármacos , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Neuronas/citología , Neuronas/fisiología , Células Madre/citología , Células Madre/fisiología
12.
Nephron Exp Nephrol ; 112(1): e20-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19342870

RESUMEN

In this study we evaluated whether administration of stem cells of neural origin (neural precursor cells, NPCs) could be protective against renal ischemia-reperfusion injury (IRI). We hypothesized that stem cell outcomes are not tissue-specific and that NPCs can improve tissue damage through paracrine mechanisms, especially due to immunomodulation. To this end, Wistar rats (200-250 g) were submitted to 1-hour ischemia and treated with NPCs (4 x 10(6) cells/animal) at 4 h of reperfusion. To serve as controls, ischemic animals were treated with cerebellum homogenate harvested from adult rat brain. All groups were sacrificed at 24 h of reperfusion. NPCs were isolated from rat fetus telencephalon and cultured until neurosphere formation (7 days). Before administration, NPCs were labeled with carboxyfluorescein diacetate succinimydylester (CFSE). Kidneys were harvested for analysis of cytokine profile and macrophage infiltration. At 24 h, NPC treatment resulted in a significant reduction in serum creatinine (IRI + NPC 1.21 + 0.18 vs. IRI 3.33 + 0.14 and IRI + cerebellum 2.95 + 0.78 mg/dl, p < 0.05) and acute tubular necrosis (IRI + NPC 46.0 + 2.4% vs. IRI 79.7 + 14.2%, p < 0.05). NPC-CFSE and glial fibrillary acidic protein (GFAP)-positive cells (astrocyte marker) were found exclusively in renal parenchyma, which also presented GFAP and SOX-2 (an embryonic neural stem cell marker) mRNA expression. NPC treatment resulted in lower renal proinflammatory IL1-beta and TNF-alpha expression and higher anti-inflammatory IL-4 and IL-10 transcription. NPC-treated animals also had less macrophage infiltration and decreased serum proinflammatory cytokines (IL-1beta, TNF-alpha and INF-gamma). Our data suggested that NPC therapy improved renal function by influencing immunological responses.


Asunto(s)
Riñón/irrigación sanguínea , Neuronas , Daño por Reperfusión/terapia , Trasplante de Células Madre , Animales , Masculino , Ratas , Ratas Wistar
13.
PLoS One ; 4(4): e5222, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19370156

RESUMEN

Induction of adult rat bone marrow mesenchymal stem cells (MSC) by means of chemical compounds (beta-mercaptoethanol, dimethyl sulfoxide and butylated hydroxyanizole) has been proposed to lead to neuronal transdifferentiation, and this protocol has been broadly used by several laboratories worldwide. Only a few hours of MSC chemical induction using this protocol is sufficient for the acquisition of neuronal-like morphology and neuronal protein expression. However, given that cell death is abundant, we hypothesize that, rather than true neuronal differentiation, this particular protocol leads to cellular toxic effects. We confirm that the induced cells with neuronal-like morphology positively stained for NF-200, S100, beta-tubulin III, NSE and MAP-2 proteins. However, the morphological and molecular changes after chemical induction are also associated with an increase in the apoptosis of over 50% of the plated cells after 24 h. Moreover, increased intracellular cysteine after treatment indicates an impairment of redox circuitry during chemical induction, and in vitro electrophysiological recordings (patch-clamp) of the chemically induced MSC did not indicate neuronal properties as these cells do not exhibit Na(+) or K(+) currents and do not fire action potentials. Our findings suggest that a disruption of redox circuitry plays an important role in this specific chemical induction protocol, which might result in cytoskeletal alterations and loss of functional ion-gated channels followed by cell death. Despite the neuronal-like morphology and neural protein expression, induced rat bone marrow MSC do not have basic functional neuronal properties, although it is still plausible that other methods of induction and/or sources of MSC can achieve a successful neuronal differentiation in vitro.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Neuronas/efectos de los fármacos , Compuestos Orgánicos/farmacología , Oxidación-Reducción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Apoptosis , Células de la Médula Ósea/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Cisteína/metabolismo , Células Madre Mesenquimatosas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/fisiología , Ratas
14.
Cytometry A ; 75(1): 38-53, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18988295

RESUMEN

In the past years, many reports have described the existence of neural progenitor and stem cells in the adult central nervous system capable of generating new neurons, astrocytes, and oligodendrocytes. This discovery has overturned the central assumption in the neuroscience field, of no new neurons being originated in the brain after birth and provided the fundaments to understand the molecular basis of neural differentiation and to develop new therapies for neural tissue repair. Although the mechanisms underlying cell fate during neural development are not yet understood, the importance of intrinsic and extrinsic factors and of an appropriate microenvironment is well known. In this context, emerging evidence strongly suggests that glial cells play a key role in controlling multiple steps of neurogenesis. Those cells, of particular radial glia, are important for migration, cell specification, and integration of neurons into a functional neural network. This review aims to present an update in the neurogenesis area and highlight the modulation of neural stem cell differentiation by neurotransmitters, growth factors, and their receptors, with possible applications for cell therapy strategies of neurological disorders.


Asunto(s)
Diferenciación Celular , Sistema Nervioso Central/citología , Enfermedades del Sistema Nervioso/terapia , Neuronas/citología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Sistema Nervioso Central/efectos de los fármacos , Humanos , Calicreínas/metabolismo , Cininas/metabolismo , Ratones , Enfermedades del Sistema Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neurotransmisores/farmacología , Receptores Colinérgicos/metabolismo , Receptores Purinérgicos/metabolismo , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Células Madre/fisiología
15.
Cytometry A ; 73(4): 361-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18302192

RESUMEN

Neural progenitor cells were isolated from rat fetal telencephalon and proliferate as neurospheres in the presence of EGF, FGF-2, and heparin. In the absence of these growth factors, neurospheres differentiate into neurons, astrocytes, and oligodendrocytes. Using an embryonal carcinoma cell line as in vitro differentiation model, we have already demonstrated the presence of an autocrine loop system between kinin-B2 receptor activity and secretion of its ligand bradykinin (BK) as prerequisites for final neuronal differentiation (Martins et al., J Biol Chem 2005; 280: 19576-19586). The aim of this study was to verify the activity of the kallikrein-kinin system (KKS) during neural progenitor cell differentiation. Immunofluorescence studies and flow cytometry analysis revealed increases in glial fibrillary acidic protein and beta-3 tubulin expression and decrease in the number of nestin-positive cells along neurospheres differentiation, indicating the transition of neural progenitor cells to astrocytes and neurons. Kinin-B2 receptor expression and activity, secretion of BK into the medium, and presence of high-molecular weight kininogen suggest the participation of the KKS in neurosphere differentiation. Functional kinin-B2 receptors and BK secretion indicate an autocrine loop during neurosphere differentiation to neurons, astrocytes, and oligodendrocytes, reflecting events occurring during early brain development.


Asunto(s)
Neuronas/citología , Neuronas/metabolismo , Receptor de Bradiquinina B2/biosíntesis , Animales , Carcinoma Embrionario/metabolismo , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Calicreínas/química , Masculino , Modelos Biológicos , Neuronas/patología , Ratas , Ratas Wistar , Células Madre/citología
16.
J Mol Endocrinol ; 33(3): 623-38, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15591023

RESUMEN

ACTH is the hormone known to control adrenal cortex function and maintenance in the intact animal but, in culture, it inhibits proliferation of adrenocortical cells from different mammalian species, a puzzle that has remained unsolved for nearly 30 years. In this paper we compare ACTH and fibroblast growth factor 2 (FGF2) antagonistic effects on the cell cycle in the Y1 cell line, a functional lineage of mouse adreno-cortical tumor cells. This cell line displays chronic high levels of c-Ki-Ras-GTP, high active constitutive levels of phosphatidylinositol 3-OH kinase/Protein Kinase B (PI3K/AKT) and low constitutive basal expression of c-Myc, which accounts for a minor deregulation of the cell cycle. In G0/G1-arrested Y1 cells, over-expression of the dominant negative mutant HaRasN17 drastically reduces c-Ki-Ras-GTP levels, eliminating basal c-Myc expression and basal S phase entry. PI3K/Akt seems to be the downstream pathway from c-Ki-ras for deregulation of c-Myc basal expression, since wortmannin abolishes c-Myc expression in serum-starved, G0/G1-arrested Y1 cells. FGF2 is a strong mitogen for Y1 cells, promoting -- in a manner dependent on the MEK/ERK pathway -- c-myc transcription induction, c-Myc protein stabilization and S phase entry in G0/G1-arrested Y1 cells. On the other hand, ACTH causes c-Myc protein destabilization, partially blocking S phase entry induced by FGF2, by a process dependent on the cAMP/protein kinase A (PKA) pathway. The whole pathway activated by ACTH to destabilize c-Myc protein in Y1 cells might comprise the following steps: ACTH receptor -->cAMP/PKA --> Akt deactivation -->GSK3 activity liberation --> c-Myc Thr58 phosphorylation. We demonstrate that c-Myc regulation is a central key in the cell cycle control by these factors, since enforced expression of c-Myc through the MycER chimera abrogates the ACTH inhibitory effect over FGF2-induced S phase entry.


Asunto(s)
Corteza Suprarrenal/efectos de los fármacos , Corteza Suprarrenal/metabolismo , Hormona Adrenocorticotrópica/farmacología , Ciclo Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Corteza Suprarrenal/citología , Animales , Bovinos , Línea Celular Tumoral , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Ratones , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal
17.
Biochemistry ; 42(7): 2116-21, 2003 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-12590600

RESUMEN

Arginine vasopressin (AVP) is a nonapeptide long known as an endocrine and paracrine regulator of important systemic functions, namely, vasoconstriction, gluconeogenesis, corticosteroidogenesis, and excretion of water and urea. Here we report, for the first time, that AVP specifically inhibits expression of the cyclin D1 gene, leading to cell cycle blockage and halting cell proliferation. In G0/G1-arrested mouse Y1 adrenocortical tumor cells, maintained in serum-free medium (SFM), AVP mimics FGF2, promoting rapid ERK1/2 activation (5 min) followed by c-Fos protein induction (2 h). PKC inhibitor Go6983 and PI3K inhibitors wortmannin and LY294002 all inhibit ERK1/2 activation by AVP, but not by FGF2. Thus, AVP and FGF2 concur to activate ERK1/2 by different regulatory pathways. However, AVP is not a mitogenic factor for Y1 cells. On the contrary, AVP strongly antagonizes FGF2 late induction (2-5 h) of the cyclin D1 gene, down-regulating both cyclin D1 mRNA and protein. AVP inhibition of cyclin D1 expression is sufficient to block G1 phase progression and cell entry into the S phase, monitored by BrdU nuclear labeling. In addition, AVP completely inhibits proliferation of Y1 cells in 10% fetal calf serum (10% FCS) medium. On the other hand, ectopic expression of the cyclin D1 protein renders Y1 cells resistant to AVP for both entry into the S phase in SFM and continuous proliferation in 10% FCS medium. In conclusion, inhibition of cyclin D1 expression by AVP is an efficient mechanism of cell cycle blockage and consequent proliferation inhibition in Y1 adrenocortical cells.


Asunto(s)
Arginina Vasopresina/fisiología , Ciclo Celular/fisiología , Ciclina D1/antagonistas & inhibidores , Ciclina D1/biosíntesis , Regulación de la Expresión Génica/fisiología , Inhibidores de Crecimiento/fisiología , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/patología , Neoplasias de la Corteza Suprarrenal/enzimología , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Animales , Arginina Vasopresina/farmacología , Ciclo Celular/efectos de los fármacos , Células Clonales , Medios de Cultivo Condicionados , Ciclina D1/genética , Resistencia a Antineoplásicos , Activadores de Enzimas/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fase G1/efectos de los fármacos , Fase G1/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Imitación Molecular , Fosfatidilinositol 3-Quinasas/fisiología , Proteína Quinasa C/fisiología , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase de Descanso del Ciclo Celular/fisiología , Transfección , Células Tumorales Cultivadas/enzimología
18.
Biochemistry ; 41(31): 10133-40, 2002 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-12146978

RESUMEN

Here we report antimitogenic mechanisms activated by the adrenocorticotropic hormone (ACTH) in the mouse Y1 adrenocortical tumor cell line. ACTH receptors activate the Galphas/adenylate cyclase cAMP/PKA pathway to promote dephosphorylation of Akt/PKB enzymes, leading to induction of the cyclin-dependent kinases' (CDKs) inhibitor p27(Kip1). Y1 cells display high constitutive levels of phosphorylated Akt/PKB dependent on chronically elevated c-Ki-Ras.GTP and PI3K activity. Expression of the dominant negative mutant RasN17 in Y1 cells results in strong reduction of both c-Ki-Ras.GTP and phosphorylated Akt/PKB, which are restored by FGF2 treatments. Inhibitors of PI3K lead to rapid dephosphorylation of Akt/PKB and block phosphorylation of Akt/PKB promoted by FGF2. ACTH rapidly promotes dephosphorylation of Akt/PKB in Y1 adrenal cells, while constitutively high levels of c-Ki-Ras.GTP remain unchanged. ACTH and cAMP elevating agents fail to cause Akt/PKB dephosphorylation in PKA-deficient clonal mutants of Y1 cells. In addition, cholera toxin, forskolin, and 8BrcAMP all mimic ACTH, causing dephosphorylation of Akt/PKB in wild-type Y1 cells. ACTH is unable to prevent Akt/PKB phosphorylation, promoted by FGF2 in clonal lines of RasN17-Y1 transfectants displaying negligible levels of c-Ki-Ras.GTP. ACTH promotes strong p27(Kip1) protein induction in wild-type Y1 adrenocortical cells but not in PKA-deficient Y1-clonal mutants nor in RasN17-Y1 transfectants. PI3K inhibitors induce p27(Kip1) protein in all cells studied, i.e., wild type and transfectants. The inverse correlation between levels of phosphorylated Akt/PKB and of p27(Kip1) protein caused by ACTH suggests a novel antimitogenic pathway activated by ACTH and mediated by cAMP/PKA in the mouse Y1 adrenocortical tumor cell line.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/metabolismo , Hormona Adrenocorticotrópica/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ciclinas/biosíntesis , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Neoplasias de la Corteza Suprarrenal/enzimología , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...