Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Stem Cells ; 39(7): 929-944, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33609411

RESUMEN

Lysosomes have recently been implicated in regulation of quiescence in adult neural stem cells (NSCs). Whether lysosomes regulate the differentiation of neural stem-progenitor cells (NPCs) in the embryonic brain has remained unknown, however. We here show that lysosomes are more abundant in rapidly dividing NPCs than in differentiating neurons in the embryonic mouse neocortex and ganglionic eminence. The genes for TFEB and TFE3, master regulators of lysosomal biosynthesis, as well as other lysosome-related genes were also expressed at higher levels in NPCs than in differentiating neurons. Anatomic analysis revealed accumulation of lysosomes at the apical and basal endfeet of NPCs. Knockdown of TFEB and TFE3, or that of the lysosomal transporter Slc15a4, resulted in premature differentiation of neocortical NPCs. Conversely, forced expression of an active form of TFEB (TFEB-AA) suppressed neuronal differentiation of NPCs in association with upregulation of NPC-related genes. These results together point to a previously unappreciated role for TFEB and TFE3, and possibly for lysosomes, in maintenance of the undifferentiated state of embryonic NPCs. We further found that lysosomes are even more abundant in an NPC subpopulation that rarely divides and includes the embryonic origin of adult NSCs than in the majority of NPCs that divide frequently for construction of the embryonic brain, and that overexpression of TFEB-AA also suppressed the cell cycle of neocortical NPCs. Our results thus also implicate lysosomes in establishment of the slowly dividing, embryonic origin of adult NSCs.


Asunto(s)
Neocórtex , Células-Madre Neurales , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Diferenciación Celular/fisiología , Lisosomas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Células-Madre Neurales/metabolismo
2.
Int J Mol Sci ; 20(7)2019 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-30959855

RESUMEN

Cathepsin D is one of the major lysosomal aspartic proteases that is essential for the normal functioning of the autophagy-lysosomal system. In the kidney, cathepsin D is enriched in renal proximal tubular epithelial cells, and its levels increase during acute kidney injury. To investigate how cathepsin D-deficiency impacts renal proximal tubular cells, we employed a conditional knockout CtsDflox/-; Spink3Cre mouse. Immunohistochemical analyses using anti-cathepsin D antibody revealed that cathepsin D was significantly decreased in tubular epithelial cells of the cortico-medullary region, mainly in renal proximal tubular cells of this mouse. Cathepsin D-deficient renal proximal tubular cells showed an increase of microtubule-associated protein light chain 3 (LC3; a marker for autophagosome/autolysosome)-signals and an accumulation of abnormal autophagic structures. Renal ischemia/reperfusion injury resulted in an increase of early kidney injury marker, Kidney injury molecule 1 (Kim-1), in the cathepsin D-deficient renal tubular epithelial cells of the CtsDflox/-; Spink3Cre mouse. Inflammation marker was also increased in the cortico-medullary region of the CtsDflox/-; Spink3Cre mouse. Our results indicated that lack of cathepsin D in the renal tubular epithelial cells led to an increase of sensitivity against ischemia/reperfusion injury.


Asunto(s)
Catepsina D/deficiencia , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Daño por Reperfusión/enzimología , Daño por Reperfusión/patología , Animales , Autofagia , Catepsina D/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Integrasas/metabolismo , Ratones
3.
Am J Pathol ; 187(7): 1586-1600, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28502476

RESUMEN

Neurologic phenotypes of cathepsin D (CTSD)-deficient mice, a murine model of neuronal ceroid lipofuscinoses, indicate the importance of CTSD for the maintenance of metabolism in central nervous system neurons. To further understand the role of CTSD in central nervous system neurons, we generated mice with a CTSD deficiency specifically in the Purkinje cells (PCs) (CTSDFlox/Flox;GRID2-Cre) and compared their phenotypes with those of PC-selective Atg7-deficient (Atg7Flox/Flox;GRID2-Cre) mice. In both strains of mice, PCs underwent degeneration, but the CTSD-deficient PCs disappeared more rapidly than their Atg7-deficient counterparts. When CTSD-deficient PCs died, the neuronal cell bodies became shrunken, filled with autophagosomes and autolysosomes, and had nuclei with dispersed small chromatin fragments. The dying Atg7-deficient PCs also showed similar ultrastructures, indicating that the neuronal cell death of CTSD- and Atg7-deficient PCs was distinct from apoptosis. Immunohistochemical observations showed the formation of calbindin-positive axonal spheroids and the swelling of vesicular GABA transporter-positive presynaptic terminals that were more pronounced in Atg7-deficient PCs than in CTSD-deficient PCs. An accumulation of tubular vesicles may have derived from the smooth endoplasmic reticulum; nascent autophagosome-like structures with double membranes was a common feature in the swollen axons of these PCs. These results suggested that PCs were more vulnerable to CTSD deficiency in lysosomes than to autophagy impairment, and this vulnerability does not depend on the severity of axonal swelling.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/genética , Catepsina D/genética , Lipofuscinosis Ceroideas Neuronales/genética , Animales , Autofagia , Proteína 7 Relacionada con la Autofagia/metabolismo , Axones/metabolismo , Axones/ultraestructura , Catepsina D/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/ultraestructura , Inmunohistoquímica , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Ratones , Lipofuscinosis Ceroideas Neuronales/metabolismo , Lipofuscinosis Ceroideas Neuronales/patología , Neuronas/metabolismo , Neuronas/ultraestructura , Fagosomas/metabolismo , Fagosomas/ultraestructura , Células de Purkinje/metabolismo , Células de Purkinje/patología , Células de Purkinje/ultraestructura
4.
Am J Pathol ; 186(8): 2143-2151, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27301359

RESUMEN

Toll-like receptors (TLRs) are one of the main contributors that induce inflammation under tissue injury and infection. Because excessive inflammation can aggravate disease states, it is important to control inflammation at a moderate level. Herein, we show that hyaluronan (HA) oligomer, HA tetrasaccharide (HA4), could suppress the expression of proinflammatory cytokine IL-1ß when stimulated with both TLR2- and TLR4-specific agonists in primary hippocampal neurons. To understand the effect of HA4 against ischemic insult, we performed hypoxic-ischemic (H/I) brain injury against neonatal mice. HA4 treatment significantly prevented hippocampal pyramidal cell death even 7 days after H/I injury, compared with the control mice. Although TLR2 and TLR4 are known as receptors for HA and also act as a receptor for inducing inflammation, only TLR2-deficient mice showed tolerance against H/I injury. Moreover, HA4 administration suppressed gliosis by inhibiting the activation of NF-κB, the downstream target of TLR2, which led to the suppression of IL-1ß expression. Taken together, our data suggest that the neuroprotective effect of HA4 relies on antagonizing the TLR2/NF-κB pathway to reduce inflammation through suppressing the expression of proinflammatory cytokines after neonatal H/I brain injury.


Asunto(s)
Ácido Hialurónico/farmacología , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Células Piramidales/patología , Receptor Toll-Like 2/metabolismo , Animales , Muerte Celular , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Ácido Hialurónico/química , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Fármacos Neuroprotectores , Reacción en Cadena de la Polimerasa , Células Piramidales/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Biochem Biophys Res Commun ; 469(3): 405-11, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26682926

RESUMEN

Cathepsin D (CD) is the major lysosomal aspartic protease and is widely distributed in the cells of various mammalian tissues. CD participates in various physiological events such as regulation of programmed cell death, activation of enzymatic precursors, and metabolic degradation of intracellular proteins through macroautophagy. To investigate the role of CD in pancreatic acinar cells, which constitute the exocrine pancreas, we generated and examined mice specifically deficient for CD in pancreatic acinar cells. CD deficient mice showed normal pancreatic development and autophagic activity, although LC3-II, which is a marker of the autophagosome, accumulates in both physiological and pancreatitis conditions. Moreover, CD deficiency leads to accumulation of matured cathepsin B (CB) and cathepsin L (CL) which are members of the cysteine protease family. We therefore conclude that CD in pancreatic acinar cells is implicated in CB and CL degradation but not in autophagic activity.


Asunto(s)
Células Acinares/metabolismo , Células Acinares/patología , Catepsina B/metabolismo , Catepsina D/metabolismo , Catepsina L/metabolismo , Pancreatitis/metabolismo , Animales , Autofagia , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Pancreatitis/patología
6.
Mol Ther Methods Clin Dev ; 32(1): 101176, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38225934

RESUMEN

Thirty genes are involved in the biosynthesis and modification of glycosylphosphatidylinositol (GPI)-anchored proteins, and defects in these genes cause inherited GPI deficiency (IGD). PIGA is X-linked and involved in the first step of GPI biosynthesis, and only males are affected by variations in this gene. The main symptoms of IGD are neurological abnormalities, such as developmental delay and seizures. There is no effective treatment at present. We crossed Nestin-Cre mice with Piga-floxed mice to generate CNS-specific Piga knockout (KO) mice. Hemizygous KO male mice died by P10 with severely defective growth. Heterozygous Piga KO female mice are mosaic for Piga expression and showed severe defects in growth and myelination and died by P25. Using these mouse models, we evaluated the effect of gene replacement therapy with adeno-associated virus (AAV). It expressed efficacy within 6 days, and the survival of male mice was extended to up to 3 weeks, whereas 40% of female mice survived for approximately 1 year and the growth defect was improved. However, liver cancer developed in all three treated female mice at 1 year of age, which was probably caused by the AAV vector bearing a strong CAG promoter.

7.
J Neurosci ; 32(27): 9238-47, 2012 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-22764232

RESUMEN

In adult mammalian brains, neural stem cells (NSCs) exist in the subventricular zone (SVZ), where persistent neurogenesis continues throughout life. Those NSCs produce neuroblasts that migrate into the olfactory bulb via formation of transit-amplifying cells, which are committed precursor cells of the neuronal lineage. In this SVZ niche, cell-cell communications conducted by diffusible factors as well as physical cell-cell contacts are important for the regulation of the proliferation and fate determination of NSCs. Previous studies have suggested that extracellular purinergic signaling, which is mediated by purine compounds such as ATP, plays important roles in cell-cell communication in the CNS. Purinergic signaling also promotes the proliferation of adult NSCs in vitro. However, the in vivo roles of purinergic signaling in the neurogenic niche still remain unknown. In this study, ATP infusion into the lateral ventricle of the mouse brain resulted in an increase in the numbers of rapidly dividing cells and Mash1-positive transit-amplifying cells (Type C cells) in the SVZ. Mash1-positive cells express the P2Y1 purinergic signaling receptor and infusion of the P2Y1 receptor-specific antagonist MRS2179 decreased the number of rapidly dividing bromodeoxyuridine (BrdU)-positive cells and Type C cells. Moreover, a 17% reduction of rapidly dividing BrdU-positive cells and a 19% reduction of Mash1-positive cells were observed in P2Y1 knock-out mice. Together, these results suggest that purinergic signaling promotes the proliferation of rapidly dividing cells and transit-amplifying cells, in the SVZ niche through the P2Y1 receptor.


Asunto(s)
Envejecimiento/fisiología , Proliferación Celular , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/fisiología , Receptores Purinérgicos P2Y1/fisiología , Transducción de Señal/fisiología , Envejecimiento/genética , Animales , División Celular/genética , División Celular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Receptores Purinérgicos P2Y1/deficiencia , Receptores Purinérgicos P2Y1/genética , Transducción de Señal/genética , Nicho de Células Madre/fisiología
8.
Development ; 137(18): 3037-46, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20685736

RESUMEN

Motile cilia generate constant fluid flow over epithelial tissue, and thereby influence diverse physiological processes. Such functions of ciliated cells depend on the planar polarity of the cilia and on their basal bodies being oriented in the downstream direction of fluid flow. Recently, another type of basal body planar polarity, characterized by the anterior localization of the basal bodies in individual cells, was reported in the multiciliated ependymal cells that line the surface of brain ventricles. However, little is known about the cellular and molecular mechanisms by which this polarity is established. Here, we report in mice that basal bodies move in the apical cell membrane during differentiation to accumulate in the anterior region of ependymal cells. The planar cell polarity signaling pathway influences basal body orientation, but not their anterior migration, in the neonatal brain. Moreover, we show by pharmacological and genetic studies that non-muscle myosin II is a key regulator of this distribution of basal bodies. This study demonstrates that the orientation and distribution of basal bodies occur by distinct mechanisms.


Asunto(s)
Movimiento Celular , Polaridad Celular , Epéndimo/crecimiento & desarrollo , Epéndimo/metabolismo , Miosina Tipo II/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Cilios/metabolismo , Epéndimo/citología , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos ICR , Microscopía Electrónica de Rastreo , Miosina Tipo II/genética , Biosíntesis de Proteínas
9.
J Biol Chem ; 286(15): 13754-64, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21349854

RESUMEN

Control of the growth and differentiation of neural stem cells is fundamental to brain development and is largely dependent on the Notch signaling pathway. The mechanism by which the activity of Notch is regulated during brain development has remained unclear, however. Fbxw7 (also known as Fbw7, SEL-10, hCdc4, or hAgo) is the F-box protein subunit of an Skp1-Cul1-F-box protein (SCF)-type ubiquitin ligase complex that plays a central role in the degradation of Notch family members. We now show that mice with brain-specific deletion of Fbxw7 (Nestin-Cre/Fbxw7(F/F) mice) die shortly after birth with morphological abnormalities of the brain and the absence of suckling behavior. The maintenance of neural stem cells was sustained in association with the accumulation of Notch1 and Notch3, as well as up-regulation of Notch target genes in the mutant mice. Astrogenesis was also enhanced in the mutant mice in vivo, and the differentiation of neural progenitor cells was skewed toward astrocytes rather than neurons in vitro, with the latter effect being reversed by treatment of the cells with a pharmacological inhibitor of the Notch signaling pathway. Our results thus implicate Fbxw7 as a key regulator of the maintenance and differentiation of neural stem cells in the brain.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas F-Box/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Células Madre/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Ratones , Ratones Noqueados , Neuroglía/citología , Neuronas/citología , Receptor Notch1/genética , Receptor Notch2/genética , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Células Madre/citología , Ubiquitina-Proteína Ligasas/genética
10.
PLoS One ; 16(8): e0256693, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34437622

RESUMEN

Induction and augmentation of labor is one of the most common obstetrical interventions. However, this intervention is not free of risks and could cause adverse events, such as hyperactive uterine contraction, uterine rupture, and amniotic-fluid embolism. Our previous study using a new animal model showed that labor induced with high-dose oxytocin (OXT) in pregnant mice resulted in massive cell death in selective brain regions, specifically in male offspring. The affected brain regions included the prefrontal cortex (PFC), but a detailed study in the PFC subregions has not been performed. In this study, we induced labor in mice using high-dose OXT and investigated neonatal brain damage in detail in the PFC using light and electron microscopy. We found that TUNEL-positive or pyknotic nuclei and Iba-1-positive microglial cells were detected more abundantly in infralimbic (IL) and prelimbic (PL) cortex of the ventromedial PFC (vmPFC) in male pups delivered by OXT-induced labor than in the control male pups. These Iba-1-positive microglial cells were engulfing dying cells. Additionally, we also noticed that in the forceps minor (FMI) of the corpus callosum (CC), the number of TUNEL-positive or pyknotic nuclei and Iba-1-positive microglial cells were largely increased and Iba-1-positive microglial cells phagocytosed massive dying cells in male pups delivered by high-dose OXT-induced labor. In conclusion, IL and PL of the vmPFC and FMI of the CC, were susceptible to brain damage in male neonates after high-dose OXT-induced labor.


Asunto(s)
Cuerpo Calloso/patología , Trabajo de Parto Inducido , Oxitocina/toxicidad , Corteza Prefrontal/patología , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio/metabolismo , Muerte Celular , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/ultraestructura , Modelos Animales de Enfermedad , Femenino , Sistema Límbico/patología , Masculino , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/patología , Fagocitosis/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/ultraestructura , Embarazo , Reproducibilidad de los Resultados
11.
Biochem Biophys Res Commun ; 379(4): 1114-9, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19161980

RESUMEN

Mesenchymal stem cells (MSCs) are a heterogeneous subset of stromal stem cells isolated from many adult tissues. Previous studies reported that MSCs can differentiate to both mesodermal and neural lineages by a phenomenon referred to as ''dedifferentiation'' or ''transdifferentiation''. However, since MSCs have only been defined in vitro, much of their development in vivo is still unknown. Here, we prospectively identified MSCs in the bone marrow from adult transgenic mice encoding neural crest-specific P0-Cre/Floxed-EGFP and Wnt1-Cre/Floxed-EGFP. EGFP-positive MSCs formed spheres that expressed neural crest stem cell genes and differentiated into neurons, glial cells, and myofibroblasts. Interestingly, we observed MSCs both in the GFP(+) and GFP(-) fraction and found that there were no significant differences in the in vitro characteristics between these two populations. Our results suggest that MSCs in adult bone marrow have at least two developmental origins, one of which is the neural crest.


Asunto(s)
Linaje de la Célula , Células Madre Mesenquimatosas/citología , Cresta Neural/citología , Animales , Ataxina-1 , Ataxinas , Antígenos de Grupos Sanguíneos/análisis , Diferenciación Celular , Células Clonales , Proteínas Fluorescentes Verdes/genética , Antígenos Comunes de Leucocito/análisis , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/análisis , Cresta Neural/química , Proteínas Nucleares/análisis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis
12.
Autophagy ; 14(5): 764-777, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28513333

RESUMEN

Conditional knockout mice for Atg9a, specifically in brain tissue, were generated to understand the roles of ATG9A in the neural tissue cells. The mice were born normally, but half of them died within one wk, and none lived beyond 4 wk of age. SQSTM1/p62 and NBR1, receptor proteins for selective autophagy, together with ubiquitin, accumulated in Atg9a-deficient neurosoma at postnatal d 15 (P15), indicating an inhibition of autophagy, whereas these proteins were significantly decreased at P28, as evidenced by immunohistochemistry, electron microscopy and western blot. Conversely, degenerative changes such as spongiosis of nerve fiber tracts proceeded in axons and their terminals that were occupied with aberrant membrane structures and amorphous materials at P28, although no clear-cut degenerative change was detected in neuronal cell bodies. Different from autophagy, diffusion tensor magnetic resonance imaging and histological observations revealed Atg9a-deficiency-induced dysgenesis of the corpus callosum and anterior commissure. As for the neurite extensions of primary cultured neurons, the neurite outgrowth after 3 d culturing was significantly impaired in primary neurons from atg9a-KO mouse brains, but not in those from atg7-KO and atg16l1-KO brains. Moreover, this tendency was also confirmed in Atg9a-knockdown neurons under an atg7-KO background, indicating the role of ATG9A in the regulation of neurite outgrowth that is independent of autophagy. These results suggest that Atg9a deficiency causes progressive degeneration in the axons and their terminals, but not in neuronal cell bodies, where the degradations of SQSTM1/p62 and NBR1 were insufficiently suppressed. Moreover, the deletion of Atg9a impaired nerve fiber tract formation.


Asunto(s)
Proteínas Relacionadas con la Autofagia/deficiencia , Axones/metabolismo , Proteínas de la Membrana/deficiencia , Red Nerviosa/metabolismo , Proteínas de Transporte Vesicular/deficiencia , Animales , Proteínas Relacionadas con la Autofagia/metabolismo , Axones/ultraestructura , Células Cultivadas , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Integrasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Neuritas/metabolismo , Neuritas/ultraestructura , Fenotipo , Proteínas/metabolismo , Células de Purkinje/metabolismo , Células de Purkinje/ultraestructura , Proteína Sequestosoma-1/metabolismo , Proteínas de Transporte Vesicular/metabolismo
13.
J Neurosci ; 26(24): 6627-36, 2006 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-16775151

RESUMEN

Recent studies have revealed that the adult mammalian brain has the capacity to regenerate some neurons after various insults. However, the precise mechanism of insult-induced neurogenesis has not been demonstrated. In the normal brain, GFAP-expressing cells in the subventricular zone (SVZ) of the lateral ventricles include a neurogenic cell population that gives rise to olfactory bulb neurons only. Herein, we report evidence that, after a stroke, these cells are capable of producing new neurons outside the olfactory bulbs. SVZ GFAP-expressing cells labeled by a cell-type-specific viral infection method were found to generate neuroblasts that migrated toward the injured striatum after middle cerebral artery occlusion. These neuroblasts in the striatum formed elongated chain-like cell aggregates similar to those in the normal SVZ, and these chains were observed to be closely associated with thin astrocytic processes and blood vessels. Finally, long-term tracing of the green fluorescent-labeled cells with a Cre-loxP system revealed that the SVZ-derived neuroblasts differentiated into mature neurons in the striatum, in which they expressed neuronal-specific nuclear protein and formed synapses with neighboring striatal cells. These results highlight the role of the SVZ in neuronal regeneration after a stroke and its potential as an important therapeutic target for various neurological disorders.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Cuerpo Estriado/patología , Ventrículos Laterales/citología , Neuronas/fisiología , Células Madre/fisiología , Accidente Cerebrovascular/metabolismo , Animales , Animales Modificados Genéticamente , Recuento de Células/métodos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Técnica del Anticuerpo Fluorescente/métodos , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Infarto de la Arteria Cerebral Media , Ratones , Ratones Endogámicos ICR , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/ultraestructura , Neuropéptidos/metabolismo , Accidente Cerebrovascular/fisiopatología , Factores de Tiempo
14.
PLoS One ; 6(3): e17610, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21412425

RESUMEN

BACKGROUND: Induced pluripotent stem (iPS) cells are generated from mouse and human somatic cells by the forced expression of defined transcription factors. Although most somatic cells are capable of acquiring pluripotency with minimal gene transduction, the poor efficiency of cell reprogramming and the uneven quality of iPS cells are still important problems. In particular, the choice of cell type most suitable for inducing high-quality iPS cells remains unclear. METHODOLOGY/PRINCIPAL FINDINGS: Here, we generated iPS cells from PDGFRα+ Sca-1+ (PαS) adult mouse mesenchymal stem cells (MSCs) and PDGFRα⁻ Sca-1⁻ osteo-progenitors (OP cells), and compared the induction efficiency and quality of individual iPS clones. MSCs had a higher reprogramming efficiency compared with OP cells and Tail Tip Fibroblasts (TTFs). The iPS cells induced from MSCs by Oct3/4, Sox2, and Klf4 appeared to be the closest equivalent to ES cells by DNA microarray gene profile and germline-transmission efficiency. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that a purified source of undifferentiated cells from adult tissue can produce high-quality iPS cells. In this context, prospectively enriched MSCs are a promising candidate for the efficient generation of high-quality iPS cells.


Asunto(s)
Separación Celular/métodos , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Quimera , Células Clonales , Regulación de la Expresión Génica/efectos de los fármacos , Células Germinativas , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Proteína Homeótica Nanog , Puromicina/farmacología
15.
J Exp Med ; 206(11): 2483-96, 2009 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-19841085

RESUMEN

Mesenchymal stem cells (MSCs) are defined as cells that undergo sustained in vitro growth and can give rise to multiple mesenchymal lineages. Because MSCs have only been isolated from tissue in culture, the equivalent cells have not been identified in vivo and little is known about their physiological roles or even their exact tissue location. In this study, we used phenotypic, morphological, and functional criteria to identify and prospectively isolate a subset of MSCs (PDGFRalpha+Sca-1+CD45-TER119-) from adult mouse bone marrow. Individual MSCs generated colonies at a high frequency and could differentiate into hematopoietic niche cells, osteoblasts, and adipocytes after in vivo transplantation. Naive MSCs resided in the perivascular region in a quiescent state. This study provides the useful method needed to identify MSCs as defined in vivo entities.


Asunto(s)
Células de la Médula Ósea/citología , Separación Celular/métodos , Trasplante de Células Madre Mesenquimatosas , Células Madre Multipotentes/citología , Tejido Adiposo/citología , Tejido Adiposo/efectos de la radiación , Animales , Células de la Médula Ósea/efectos de la radiación , Diferenciación Celular/efectos de la radiación , Linaje de la Célula/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Clonales , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/citología , Células Endoteliales/efectos de la radiación , Hematopoyesis , Mesodermo/citología , Mesodermo/efectos de la radiación , Ratones , Células Madre Multipotentes/efectos de la radiación , Fenotipo , Tolerancia a Radiación , Irradiación Corporal Total
16.
J Cell Sci ; 121(Pt 8): 1204-12, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18349072

RESUMEN

During brain development, neural progenitor cells extend across the thickening brain wall and undergo mitosis. To understand how these two completely different cellular events are coordinated, we focused on the transcription pattern of the nestin gene (Nes), which encodes an intermediate filament protein strongly expressed in neural progenitor cells. To visualize nestin expression in vivo, we generated transgenic mice that expressed a destabilized fluorescent protein under the control of Nes second intronic enhancer (E/nestin:dVenus). During the neurogenic stage, when the brain wall thickens markedly, we found that nestin was regulated in a cell-cycle-dependent manner. Time-lapse imaging showed that nestin gene expression was upregulated during G1-S phase, when the neural progenitor cells elongate their fibers. However, nestin expression dramatically declined in G2-M phase, when progenitor cells round up to undergo mitosis. The cell-cycle-dependent phosphorylation of an upstream regulator class III POU transcription factor (Pou3f2 or Brn2) reduced its binding activity to the nestin core enhancer element and was therefore responsible for the decreased Nes transcription in G2-M phase. Collectively, these findings demonstrate precisely orchestrated gene regulation that correlates with the 3D morphological changes in neural progenitor cells in vivo.


Asunto(s)
Ciclo Celular , Células Madre Embrionarias/citología , Proteínas de Filamentos Intermediarios/genética , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/citología , Animales , Ensayo de Cambio de Movilidad Electroforética , Femenino , Ratones , Ratones Transgénicos , Nestina , Embarazo
17.
Genesis ; 45(2): 90-100, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17245775

RESUMEN

In the past decades, the function of the Wnt canonical pathway during embryogenesis has been intensively investigated; however, little survey of neonatal and adult tissues has been made, and the role of this pathway remains largely unknown. To investigate its role in mature tissues, we generated two new reporter transgenic mouse lines, ins-TOPEGFP and ins-TOPGAL, that drive EGFP and beta-galactosidase expression under TCF/beta-catenin, respectively. To obtain the accurate expression pattern, we flanked these transgenes with the HS4 insulator to reduce chromosomal positional effects. Analysis of embryos showed that the reporter genes were activated in regions where canonical Wnt activity has been implicated. Furthermore, their expression patterns were consistent in both lines, indicating the accuracy of the reporter signal. In the neonatal brain, the reporter signal was detected in the mesencephalon and hippocampus. In the adult mice, the reporter signal was found in the mature pericenteral hepatocytes in the normal liver. Furthermore, during inflammation the number of T cells expressing the reporter gene increased in the adult spleen. Thus, in this research, we identified two organs, i.e., the liver and spleen, as novel organs in which the Wnt canonical signal is in motion in the adult. These transgenic lines will provide us broader opportunities to investigate the function of the Wnt canonical pathway in vivo.


Asunto(s)
Encéfalo/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Hígado/metabolismo , Transducción de Señal/fisiología , Bazo/metabolismo , Proteínas Wnt/metabolismo , Animales , Animales Recién Nacidos , Células COS , Células Cultivadas , Chlorocebus aethiops , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/genética , Proteínas Wnt/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA