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












Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-37835152

RESUMEN

This study aimed to analyze and gain an in-depth understanding of the experiences pertaining to successful aging in middle-aged women in South Korea. A sample of 12 middle-aged women, capable of sharing their lived experiences, was divided into three age-based groups: those in their 40s, those in their 50s, and those aged 60-65 years. The collected data were analyzed using Colaizzi's phenomenological method. Five theme clusters and ten themes emerged. The experiences of successful aging among middle-aged women were categorized as: "Coping with changes in the mind and body", "Financially stable life", "Undergoing the aging process with a healthy family", "Preparations for dying well", and "Pursuing a meaningful, harmonious life". These findings highlight the need for programs that prepare middle-aged women to positively accept and enjoy older adulthood by identifying and addressing the factors essential for successful aging and reducing any negative emotions attached to aging and older adulthood.


Asunto(s)
Adaptación Psicológica , Envejecimiento , Persona de Mediana Edad , Humanos , Femenino , Anciano , Envejecimiento/psicología , República de Corea , Recolección de Datos , Investigación Cualitativa
2.
J Ginseng Res ; 44(3): 435-441, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32372865

RESUMEN

BACKGROUND: As a process of aging, skeletal muscle mass and function gradually decrease. It is reported that ginsenoside Rb1 and Rb2 play a role as AMP-activated protein kinase activator, resulting in regulating glucose homeostasis, and Rb1 reduces oxidative stress in aged skeletal muscles through activating the phosphatidylinositol 3-kinase/Akt/Nrf2 pathway. We examined the effects of Rb1 and Rb2 on differentiation of the muscle stem cells and myotube formation. METHODS: C2C12 myoblasts treated with Rb1 and/or Rb2 were differentiated and induced to myotube formation, followed by immunoblotting for myogenic marker proteins, such as myosin heavy chain, MyoD, and myogenin, or immunostaining for myosin heavy chain or immunoprecipitation analysis for heterodimerization of MyoD/E-proteins. RESULTS: Rb1 and Rb2 enhanced myoblast differentiation through accelerating MyoD/E-protein heterodimerization and increased myotube hypertrophy, accompanied by activation of Akt/mammalian target of rapamycin signaling. In addition, Rb1 and Rb2 induced the MyoD-mediated transdifferentiation of the rhabdomyosarcoma cells into myoblasts. Furthermore, co-treatment with Rb1 and Rb2 had synergistically enhanced myoblast differentiation through Akt activation. CONCLUSION: Rb1 and Rb2 upregulate myotube growth and myogenic differentiation through activating Akt/mammalian target of rapamycin signaling and inducing myogenic conversion of fibroblasts. Thus, our first finding indicates that Rb1 and Rb2 have strong potential as a helpful remedy to prevent and treat muscle atrophy, such as age-related muscular dystrophy.

3.
EMBO J ; 38(24): e101196, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31750563

RESUMEN

Parkinson's disease (PD) is neurodegenerative movement disorder characterized by degeneration of midbrain-type dopamine (mDA) neurons in the substantia nigra (SN). The RNA-binding protein Lin28 plays a role in neuronal stem cell development and neuronal differentiation. In this study, we reveal that Lin28 conditional knockout (cKO) mice show degeneration of mDA neurons in the SN, as well as PD-related behavioral deficits. We identify a loss-of-function variant of LIN28A (R192G substitution) in two early-onset PD patients. Using an isogenic human embryonic stem cell (hESC)/human induced pluripotent stem cell (hiPSC)-based disease model, we find that the Lin28 R192G variant leads to developmental defects and PD-related phenotypes in mDA neuronal cells that can be rescued by expression of wild-type Lin28A. Cell transplantation experiments in PD model rats show that correction of the LIN28A variant in the donor patient (pt)-hiPSCs leads to improved behavioral phenotypes. Our data link LIN28A to PD pathogenesis and suggest future personalized medicine targeting this variant in patients.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Sustancia Negra/metabolismo , Animales , Conducta Animal , Trasplante de Células , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Células Madre Embrionarias/fisiología , Edición Génica , Predisposición Genética a la Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Ratones , Ratones Noqueados , Mutación , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Enfermedad de Parkinson/genética , Ratas , Trasplante de Células Madre
4.
J Ginseng Res ; 43(3): 475-481, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31308819

RESUMEN

BACKGROUND: The ginsenoside Rg1 has been shown to exert various pharmacological activities with health benefits. Previously, we have reported that Rg1 promoted myogenic differentiation and myotube growth in C2C12 myoblasts. In this study, the in vivo effect of Rg1 on fiber-type composition and oxidative metabolism in skeletal muscle was examined. METHODS: To examine the effect of Rg1 on skeletal muscle, 3-month-old mice were treated with Rg1 for 5 weeks. To assess muscle strength, grip strength tests were performed, and the lower hind limb muscles were harvested, followed by various detailed analysis, such as histological staining, immunoblotting, immunostaining, and real-time quantitative reverse transcription polymerase chain reaction. In addition, to verify the in vivo data, primary myoblasts isolated from mice were treated with Rg1, and the Rg1 effect on myotube growth was examined by immunoblotting and immunostaining analysis. RESULTS: Rg1 treatment increased the expression of myosin heavy chain isoforms characteristic for both oxidative and glycolytic muscle fibers; increased myofiber sizes were accompanied by enhanced muscle strength. Rg1 treatment also enhanced oxidative muscle metabolism with elevated oxidative phosphorylation proteins. Furthermore, Rg1-treated muscles exhibited increased levels of anabolic S6 kinase signaling. CONCLUSION: Rg1 improves muscle functionality via enhancing muscle gene expression and oxidative muscle metabolism in mice.

5.
Toxicol Lett ; 292: 12-19, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29680376

RESUMEN

Bisphenol A (BPA), one of the most widespread endocrine disrupting chemicals, is known as an artificial estrogen, which interacts with estrogen receptor (ER). In this study, we investigated the effects of BPA and estradiol on myoblast differentiation and the underlying signaling mechanism. Exposure to BPA (0.01-1 µM) in mouse myoblast C2C12 cells attenuated myogenic differentiation via the reduced expression of muscle-specific genes, such as myosin heavy chain (MHC), MyoD, and Myogenin, without the alteration of cell proliferation and viability. BPA-exposed C2C12 myoblasts also showed a reduction of Akt phosphorylation ((37-61) %, p < 0.001), a key event for myogenesis. Similarly to BPA, estradiol (0.01-1 µM) reduced the expression of muscle-specific proteins and the formation of multinucleated myotubes, and attenuated the muscle differentiation-specific phosphorylation of Akt ((42-59) %, p < 0.001). We conclude that BPA and estradiol suppress myogenic differentiation through the inhibition of Akt signaling.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Diferenciación Celular/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Estradiol/toxicidad , Desarrollo de Músculos/efectos de los fármacos , Mioblastos/efectos de los fármacos , Fenoles/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Ratones , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mioblastos/enzimología , Mioblastos/patología , Fosforilación
6.
J Ginseng Res ; 42(1): 116-121, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29348730

RESUMEN

BACKGROUND: Black ginseng (BG) has greatly enhanced pharmacological activities relative to white or red ginseng. However, the effect and molecular mechanism of BG on muscle growth has not yet been examined. In this study, we investigated whether BG could regulate myoblast differentiation and myotube hypertrophy. METHODS: BG-treated C2C12 myoblasts were differentiated, followed by immunoblotting for myogenic regulators, immunostaining for a muscle marker, myosin heavy chain or immunoprecipitation analysis for myogenic transcription factors. RESULTS: BG treatment of C2C12 cells resulted in the activation of Akt, thereby enhancing heterodimerization of MyoD and E proteins, which in turn promoted muscle-specific gene expression and myoblast differentiation. BG-treated myoblasts formed larger multinucleated myotubes with increased diameter and thickness, accompanied by enhanced Akt/mTOR/p70S6K activation. Furthermore, the BG treatment of human rhabdomyosarcoma cells restored myogenic differentiation. CONCLUSION: BG enhances myoblast differentiation and myotube hypertrophy by activating Akt/mTOR/p70S6k axis. Thus, our study demonstrates that BG has promising potential to treat or prevent muscle loss related to aging or other pathological conditions, such as diabetes.

7.
J Ginseng Res ; 41(4): 608-614, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29021711

RESUMEN

BACKGROUND: Ginsenoside Rg1 belongs to protopanaxatriol-type ginsenosides and has diverse pharmacological activities. In this report, we investigated whether Rg1 could upregulate muscular stem cell differentiation and muscle growth. METHODS: C2C12 myoblasts, MyoD-transfected 10T1/2 embryonic fibroblasts, and HEK293T cells were treated with Rg1 and differentiated for 2 d, subjected to immunoblotting, immunocytochemistry, or immunoprecipitation. RESULTS: Rg1 activated promyogenic kinases, p38MAPK (mitogen-activated protein kinase) and Akt signaling, that in turn promote the heterodimerization with MyoD and E proteins, resulting in enhancing myogenic differentiation. Through the activation of Akt/mammalian target of rapamycin pathway, Rg1 induced myotube growth and prevented dexamethasone-induced myotube atrophy. Furthermore, Rg1 increased MyoD-dependent myogenic conversion of fibroblast. CONCLUSION: Rg1 upregulates promyogenic kinases, especially Akt, resulting in improvement of myoblast differentiation and myotube growth.

8.
Nat Commun ; 8: 15926, 2017 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-28649987

RESUMEN

Adipose progenitor cells (APCs) reside in a vascular niche, located within the perivascular compartment of adipose tissue blood vessels. Yet, the signals and mechanisms that govern adipose vascular niche formation and APC niche interaction are unknown. Here we show that the assembly and maintenance of the adipose vascular niche is controlled by PPARγ acting within APCs. PPARγ triggers a molecular hierarchy that induces vascular sprouting, APC vessel niche affinity and APC vessel occupancy. Mechanistically, PPARγ transcriptionally activates PDGFRß and VEGF. APC expression and activation of PDGFRß promotes the recruitment and retention of APCs to the niche. Pharmacologically, targeting PDGFRß disrupts APC niche contact thus blocking adipose tissue expansion. Moreover, enhanced APC expression of VEGF stimulates endothelial cell proliferation and expands the adipose niche. Consequently, APC niche communication and retention are boosted by VEGF thereby impairing adipogenesis. Our data indicate that APCs direct adipose tissue niche expansion via a PPARγ-initiated PDGFRß and VEGF transcriptional axis.


Asunto(s)
Adipocitos/metabolismo , PPAR gamma/metabolismo , Nicho de Células Madre , Células Madre/metabolismo , Adipocitos/citología , Adipogénesis , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Proliferación Celular , Femenino , Masculino , Ratones , PPAR gamma/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/citología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Brain ; 139(Pt 10): 2722-2739, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27538419

RESUMEN

The original properties of tissue-specific stem cells, regardless of their tissue origins, are inevitably altered during in vitro culturing, lessening the clinical and research utility of stem cell cultures. Specifically, neural stem cells derived from the ventral midbrain lose their dopamine neurogenic potential, ventral midbrain-specific phenotypes, and repair capacity during in vitro cell expansion, all of which are critical concerns in using the cultured neural stem cells in therapeutic approaches for Parkinson's disease. In this study, we observed that the culture-dependent changes of neural stem cells derived from the ventral midbrain coincided with loss of RNA-binding protein LIN28A expression. When LIN28A expression was forced and sustained during neural stem cell expansion using an inducible expression-vector system, loss of dopamine neurogenic potential and midbrain phenotypes after long-term culturing was blocked. Furthermore, dopamine neurons that differentiated from neural stem cells exhibited remarkable survival and resistance against toxic insults. The observed effects were not due to a direct action of LIN28A on the differentiated dopamine neurons, but rather its action on precursor neural stem cells as exogene expression was switched off in the differentiating/differentiated cultures. Remarkable and reproducible behavioural recovery was shown in all Parkinson's disease rats grafted with neural stem cells expanded with LIN28A expression, along with extensive engraftment of dopamine neurons expressing mature neuronal and midbrain-specific markers. These findings suggest that LIN28A expression during stem cell expansion could be used to prepare therapeutically competent donor cells.

10.
Sci Rep ; 6: 32025, 2016 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-27554447

RESUMEN

The intracellular Raf-Erk signaling pathway is activated during neural stem cell (NSC) proliferation, and neuronal and astrocytic differentiation. A key question is how this signal can evoke multiple and even opposing NSC behaviors. We show here, using a constitutively active Raf (ca-Raf), that Raf-Erk activation in NSCs induces neuronal differentiation in a cell-autonomous manner. By contrast, it causes NSC proliferation and the formation of astrocytes in an extrinsic autocrine/paracrine manner. Thus, treatment of NSCs with medium (CM) conditioned in ca-Raf-transduced NSCs (Raf-CM; RCM) became activated to form proliferating astrocytes resembling radial glial cells (RGCs) or adult-type NSCs. Infusion of Raf-CM into injured mouse brains caused expansion of the NSC population in the subventricular zone, followed by the formation of new neurons that migrated to the damaged site. Our study shows an example how molecular mechanisms dissecting NSC behaviors can be utilized to develop regenerative therapies in brain disorders.


Asunto(s)
Encéfalo/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células-Madre Neurales/metabolismo , Regeneración/fisiología , Quinasas raf/metabolismo , Animales , Astrocitos/citología , Encéfalo/embriología , Recuento de Células , Diferenciación Celular , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/citología , Neuronas/citología , Neuronas/metabolismo , Ratas Sprague-Dawley
12.
Cell Metab ; 14(1): 116-22, 2011 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-21723509

RESUMEN

White adipose tissue regulates metabolism; the importance of this control is highlighted by the ongoing pandemic of obesity and associated complications such as diabetes, atherosclerosis, and cancer. White adipose tissue maintenance is a dynamic process, yet very little is known about how pharmacologic stimuli affect such plasticity. Combining in vivo lineage marking and BrdU labeling strategies, we found that rosiglitazone, a member of the thiazolidinedione class of glucose-lowering medicines, markedly increases the evolution of adipose progenitors into adipocytes. Notably, chronic rosiglitazone administration disrupts the adipogenic and self-renewal capacities of the stem cell compartment and alters its molecular characteristics. These data unravel unknown aspects of adipose dynamics and provide a basis to manipulate the adipose lineage for therapeutic ends.


Asunto(s)
Tejido Adiposo Blanco/citología , Tiazolidinedionas/farmacología , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Ratones , Células Madre/citología , Células Madre/metabolismo
13.
J Clin Invest ; 121(6): 2326-35, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21576821

RESUMEN

Parkinson disease (PD) involves the selective loss of midbrain dopamine (mDA) neurons and is a possible target disease for stem cell-based therapy. Human induced pluripotent stem cells (hiPSCs) are a potentially unlimited source of patient-specific cells for transplantation. However, it is critical to evaluate the safety of hiPSCs generated by different reprogramming methods. Here, we compared multiple hiPSC lines derived by virus- and protein-based reprogramming to human ES cells (hESCs). Neuronal precursor cells (NPCs) and dopamine (DA) neurons delivered from lentivirus-based hiPSCs exhibited residual expression of exogenous reprogramming genes, but those cells derived from retrovirus- and protein-based hiPSCs did not. Furthermore, NPCs derived from virus-based hiPSCs exhibited early senescence and apoptotic cell death during passaging, which was preceded by abrupt induction of p53. In contrast, NPCs derived from hESCs and protein-based hiPSCs were highly expandable without senescence. DA neurons derived from protein-based hiPSCs exhibited gene expression, physiological, and electrophysiological properties similar to those of mDA neurons. Transplantation of these cells into rats with striatal lesions, a model of PD, significantly rescued motor deficits. These data support the clinical potential of protein-based hiPSCs for personalized cell therapy of PD.


Asunto(s)
Reprogramación Celular , Dopamina/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Factores de Transcripción de Tipo Kruppel/fisiología , Neuronas/citología , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Trastornos Parkinsonianos/cirugía , Proteínas Proto-Oncogénicas c-myc/fisiología , Factores de Transcripción SOXB1/fisiología , Animales , Apoptosis , Arginina , Diferenciación Celular , Línea Celular/trasplante , Linaje de la Célula , Senescencia Celular , Regulación del Desarrollo de la Expresión Génica , Genes p53 , Vectores Genéticos/farmacología , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Lentivirus/fisiología , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Proteínas Proto-Oncogénicas c-myc/genética , Ratas , Retroviridae/fisiología , Factores de Transcripción SOXB1/genética , Proteína p53 Supresora de Tumor/biosíntesis
14.
Stem Cells ; 28(3): 501-12, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20049900

RESUMEN

Effective dopamine (DA) neuron differentiation from neural precursor cells (NPCs) is prerequisite for precursor/stem cell-based therapy of Parkinson's disease (PD). Nurr1, an orphan nuclear receptor, has been reported as a transcription factor that can drive DA neuron differentiation from non-dopaminergic NPCs in vitro. However, Nurr1 alone neither induces full neuronal maturation nor expression of proteins found specifically in midbrain DA neurons. In addition, Nurr1 expression is inefficient in inducing DA phenotype expression in NPCs derived from certain species such as mouse and human. We show here that Foxa2, a forkhead transcription factor whose role in midbrain DA neuron development was recently revealed, synergistically cooperates with Nurr1 to induce DA phenotype acquisition, midbrain-specific gene expression, and neuronal maturation. Thus, the combinatorial expression of Nurr1 and Foxa2 in NPCs efficiently yielded fully differentiated nigral (A9)-type midbrain neurons with clearly detectable DA neuronal activities. The effects of Foxa2 in DA neuron generation were observed regardless of the brain regions or species from which NPCs were derived. Furthermore, DA neurons generated by ectopic Foxa2 expression were more resistant to toxins. Importantly, Foxa2 expression resulted in a rapid cell cycle exit and reduced cell proliferation. Consistently, transplantation of NPCs transduced with Nurr1 and Foxa2 generated grafts enriched with midbrain-type DA neurons but reduced number of proliferating cells, and significantly reversed motor deficits in a rat PD model. Our findings can be applied to ongoing attempts to develop an efficient and safe precursor/stem cell-based therapy for PD.


Asunto(s)
Diferenciación Celular/genética , Factor Nuclear 3-beta del Hepatocito/genética , Neuronas/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Trasplante de Células Madre/métodos , Células Madre/metabolismo , Animales , Proliferación Celular , Supervivencia Celular/genética , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Dopamina/metabolismo , Humanos , Ratones , Neurogénesis/genética , Neuronas/citología , Neuronas/trasplante , Enfermedad de Parkinson/cirugía , Fenotipo , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Sustancia Negra/citología , Sustancia Negra/metabolismo , Transfección/métodos , Resultado del Tratamiento
15.
Stem Cells ; 27(9): 2238-46, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19522012

RESUMEN

Nurr1 is a transcription factor specific for the development and maintenance of the midbrain dopamine (DA) neurons. Exogenous Nurr1 in neural precursor (NP) cells induces the differentiation of DA neurons in vitro that are capable of reversing motor dysfunctions in a rodent model for Parkinson disease. The promise of this therapeutic approach, however, is unclear due to poor cell survival and phenotype loss of DA cells after transplantation. We herein demonstrate that Nurr1 proteins undergo ubiquitin-proteasome-system-mediated degradation in differentiating NP cells. The degradation process is activated by a direct Akt-mediated phosphorylation of Nurr1 proteins and can be prevented by abolishing the Akt-target sequence in Nurr1 (Nurr1(Akt)). Overexpression of Nurr1(Akt) in NP cells yielded DA neurons in which Nurr1 protein levels were maintained for prolonged periods. The sustained Nurr1 expression endowed the Nurr1(Akt)-induced DA neurons with resistance to toxic stimuli, enhanced survival, and sustained DA phenotypes in vitro and in vivo after transplantation.


Asunto(s)
Dopamina/metabolismo , Neuronas/citología , Neuronas/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/farmacología , Western Blotting , Butadienos/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Inmunoprecipitación , Mesencéfalo/citología , Morfolinas/farmacología , Nitrilos/farmacología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Estabilidad Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Mol Ther ; 16(11): 1873-82, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18781144

RESUMEN

Neural precursor cells (NPCs) are regarded as a promising source of donor cells in transplantation-based therapies for neurodegenerative disorders. However, poor survival and limited neuronal differentiation of the transplanted NPCs remain critical limitations for developing therapeutic strategies. In this study, we investigated the effects of the proneural basic helix-loop-helix (bHLH) transcription factors Mash1 and Neurogenin 2 (Ngn2) in neuronal differentiation and survival of NPCs. Induction of Mash1 or Ngn2 expression strikingly enhanced neuronal differentiation of cultured NPCs in vitro. Ngn2-transduced NPCs underwent a rapid cell cycle arrest, which often accompanies differentiation. In contrast, cells continuously expanded upon Mash1 expression during NPC differentiation. Notably, sonic hedgehog (SHH) was upregulated by Mash1 and mediated the proliferative and survival effects of Mash1 on NPCs. Upon transplantation into adult rat brains, Mash1-expressing NPCs yielded large grafts enriched with neurons compared to control LacZ-transduced NPCs. Interestingly, enhancements in neuronal yield, as well as in donor cell survival, were also achieved by transplanting Ngn2-transduced NPCs. We show that a differentiation stage- and cell density-dependent survival effect of Ngn2 involves neurotrophin3 (NT3)/TrkC-mediated signaling. Together, these findings suggest potential benefits of bHLH gene manipulation to develop successful transplantation strategies for brain disorders.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Células Madre Embrionarias/trasplante , Proteínas del Tejido Nervioso/genética , Neuronas/trasplante , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Encéfalo/citología , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Proteínas Hedgehog/fisiología , Técnicas In Vitro , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/citología , Neurotrofina 3/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Transducción Genética
17.
Exp Cell Res ; 313(19): 4066-81, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17936272

RESUMEN

We have cultivated highly uniform populations of neural precursor cells, which retain their region-specific identities, from various rat embryonic brain regions. The roles of the proneural basic-helix-loop-helix (bHLH) factors neurogenin2 (Ngn2) and Mash1 in gamma-aminobutyric acid (GABA) neuron differentiation were explored in the region-specific cultures. Consistent with previous in vivo studies, forced expression of Mash1 promoted GABA neuron formation from the precursors derived from the developing forebrains, whereas Ngn2 displayed an inhibitory role in forebrain GABA neuron differentiation. Functional analyses of mutant bHLH proteins indicated that the helix-loop-helix domains of Mash1 and Ngn2, known as the structures for protein-protein interactions, impart the distinct activities. Intriguingly, the regulatory activities of Mash1 and Ngn2 in GABA neuron differentiation from the hindbrain- and spinal cord-derived precursor cells were completely opposite of those observed in the forebrain-derived cultures: increased GABA neuron yield by Ngn2 and decreased yield by Mash1 were shown in the precursors of those posterior brain regions. No clear difference that depended on dorsal-ventral brain regions was observed in the bHLH-mediated activities. Finally, we demonstrated that Otx2, the expression of which is developmentally confined to the regions anterior to the isthmus, is a factor responsible for the anterior-posterior region-dependent opposite effects of the bHLH proteins.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Química Encefálica , Diferenciación Celular , Proteínas del Tejido Nervioso/fisiología , Neuronas/citología , Factores de Transcripción Otx/fisiología , Animales , Células Cultivadas , Prosencéfalo/química , Ratas , Rombencéfalo/química , Ácido gamma-Aminobutírico
18.
Mol Cell Biol ; 27(12): 4293-305, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17438128

RESUMEN

In the developing mouse brain, the highest Bcl-X(L) expression is seen at the peak of neurogenesis, whereas the peak of Bax expression coincides with the astrogenic period. While such observations suggest an active role of the Bcl-2 family proteins in the generation of neurons and astrocytes, no definitive demonstration has been provided to date. Using combinations of gain- and loss-of-function assays in vivo and in vitro, we provide evidence for instructive roles of these proteins in neuronal and astrocytic fate specification. Specifically, in Bax knockout mice, astrocyte formation was decreased in the developing cortices. Overexpression of Bcl-X(L) and Bax in embryonic cortical precursors induced neural and astrocytic differentiation, respectively, while inhibitory RNAs led to the opposite results. Importantly, inhibition of caspase activity, dimerization, or mitochondrial localization of Bcl-X(L)/Bax proteins indicated that the differentiation effects of Bcl-X(L)/Bax are separable from their roles in cell survival and apoptosis. Lastly, we describe activation of intracellular signaling pathways and expression of basic helix-loop-helix transcriptional factors specific for the Bcl-2 protein-mediated differentiation.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo , Animales , Astrocitos/citología , Astrocitos/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Inhibidores de Caspasas , Diferenciación Celular , Células Cultivadas , Materiales Biocompatibles Revestidos/metabolismo , Cruzamientos Genéticos , Dimerización , Activación Enzimática , Fibronectinas/metabolismo , Homocigoto , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , Neuronas/citología , Neuronas/fisiología , ARN Interferente Pequeño/genética , Retroviridae/genética , Transducción de Señal/fisiología , Transducción Genética , Proteína X Asociada a bcl-2/química , Proteína X Asociada a bcl-2/genética , Proteína bcl-X/química , Proteína bcl-X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...