Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Sci Rep ; 10(1): 10659, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32606354

RESUMEN

In human blood, oxygen is mainly transported by red blood cells. Accordingly, the dissolved oxygen level in plasma is expected to be limited, although it has not been quantified yet. Here, by developing dedicated methods and tools, we determined that human plasma pO2 = 8.4 mmHg (1.1% O2). Oxygen solubility in plasma was believed to be similar to water. Here we reveal that plasma has an additional ascorbate-dependent oxygen-reduction activity. Plasma experimental oxygenation oxidizes ascorbate (49.5 µM in fresh plasma vs < 2 µM in oxidized plasma) and abolishes this capacity, which is restored by ascorbate supplementation. We confirmed these results in vivo, showing that the plasma pO2 is significantly higher in ascorbate-deficient guinea pigs (Ascorbateplasma < 2 µM), compared to control (Ascorbateplasma > 15 µM). Plasma low oxygen level preserves the integrity of oxidation-sensitive components such as ubiquinol. Circulating leucocytes are well adapted to these conditions, since the abundance of their mitochondrial network is limited. These results shed a new light on the importance of oxygen exposure on leucocyte biological study, in regards with the reducing conditions they encounter in vivo; but also, on the manipulation of blood products to improve their integrity and potentially improve transfusions' efficacy.


Asunto(s)
Ácido Ascórbico/sangre , Ácido Ascórbico/metabolismo , Oxígeno/sangre , Plasma/metabolismo , Animales , Línea Celular , Linaje de la Célula/fisiología , Eritrocitos/metabolismo , Cobayas , Células HEK293 , Células Hep G2 , Humanos , Hipoxia/sangre , Hipoxia/metabolismo , Oxidación-Reducción , Solubilidad , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
2.
Gastroenterology ; 158(6): 1650-1666.e15, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32032583

RESUMEN

BACKGROUND & AIMS: Gastric chief cells, a mature cell type that secretes digestive enzymes, have been proposed to be the origin of metaplasia and cancer through dedifferentiation or transdifferentiation. However, studies supporting this claim have had technical limitations, including issues with the specificity of chief cell markers and the toxicity of drugs used. We therefore sought to identify genes expressed specifically in chief cells and establish a model to trace these cells. METHODS: We performed transcriptome analysis of Mist1-CreERT-traced cells, with or without chief cell depletion. Gpr30-rtTA mice were generated and crossed to TetO-Cre mice, and lineage tracing was performed after crosses to R26-TdTomato mice. Additional lineage tracing experiments were performed using Mist1-CreERT, Kitl-CreERT, Tff1-Cre, and Tff2-Cre mice crossed to reporter mice. Mice were given high-dose tamoxifen or DMP-777 or were infected with Helicobacter pylori to induce gastric metaplasia. We studied mice that expressed mutant forms of Ras in gastric cells, using TetO-KrasG12D, LSL-KrasG12D, and LSL-HrasG12V mice. We analyzed stomach tissues from GPR30-knockout mice. Mice were given dichloroacetate to inhibit pyruvate dehydrogenase kinase (PDK)-dependent cell competition. RESULTS: We identified GPR30, the G-protein-coupled form of the estrogen receptor, as a cell-specific marker of chief cells in gastric epithelium of mice. Gpr30-rtTA mice crossed to TetO-Cre;R26-TdTomato mice had specific expression of GPR30 in chief cells, with no expression noted in isthmus stem cells or lineage tracing of glands. Expression of mutant Kras in GPR30+ chief cells did not lead to the development of metaplasia or dysplasia but, instead, led to a reduction in labeled numbers of chief cells and a compensatory expansion of neck lineage, which was derived from upper Kitl+ clones. Administration of high-dose tamoxifen, DMP-777, or H pylori decreased the number of labeled chief cells. Chief cells were eliminated from epithelia via GPR30- and PDK-dependent cell competition after metaplastic stimuli, whereas loss of GRP30 or inhibition of PDK activity preserved chief cell numbers and attenuated neck lineage cell expansion. CONCLUSIONS: In tracing studies of mice, we found that most chief cells are lost during metaplasia and therefore are unlikely to contribute to gastric carcinogenesis. Expansion of cells that coexpress neck and chief lineage markers, known as spasmolytic polypeptide-expressing metaplasia, does not occur via dedifferentiation from chief cells but, rather, through a compensatory response from neck progenitors to replace the eliminated chief cells.


Asunto(s)
Células Principales Gástricas/fisiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Azetidinas/toxicidad , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Ácido Dicloroacético/administración & dosificación , Modelos Animales de Enfermedad , Mucosa Gástrica/citología , Mucosa Gástrica/efectos de los fármacos , Infecciones por Helicobacter/microbiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metaplasia/inducido químicamente , Metaplasia/microbiología , Metaplasia/patología , Ratones , Ratones Noqueados , Piperazinas/toxicidad , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/antagonistas & inhibidores , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Células Madre/fisiología , Tamoxifeno/toxicidad
3.
Endocrinology ; 160(7): 1701-1718, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31135891

RESUMEN

Tanycytes have recently been accepted as neural stem/progenitor cells in the postnatal hypothalamus. Persistent retina and anterior neural fold homeobox (Rax) expression is characteristic of tanycytes in contrast to its transient expression of whole hypothalamic precursors. In this study, we found that Rax+ residual cells in the maturation phase of hypothalamic differentiation in mouse embryonic stem cell (mESC) cultures had similar characteristics to ventral tanycytes. They expressed typical neural stem/progenitor cell markers, including Sox2, vimentin, and nestin, and differentiated into mature neurons and glial cells. Quantitative RT-PCR analysis showed that Rax+ residual cells expressed Fgf-10, Fgf-18, and Lhx2, which are expressed by ventral tanycytes. They highly expressed tanycyte-specific genes Dio2 and Gpr50 compared with Rax+ early hypothalamic progenitor cells. Therefore, Rax+ residual cells in the maturation phase of hypothalamic differentiation were considered to be more differentiated and similar to late progenitor cells and tanycytes. They self-renewed and formed neurospheres when cultured with exogenous FGF-2. Additionally, these Rax+ neurospheres differentiated into three neuronal lineages (neurons, astrocytes, and oligodendrocytes), including neuropeptide Y+ neuron, that are reported to be differentiated from ventral tanycytes toward the arcuate nuclei. Thus, Rax+ residual cells were multipotent neural stem/progenitor cells. Rax+ neurospheres were stably passaged and retained high Sox2 expression even after multiple passages. These results suggest the successful induction of Rax+ tanycyte-like cells from mESCs [induced tanycyte-like (iTan) cells]. These hypothalamic neural stem/progenitor cells may have potential in regenerative medicine and as a research tool.


Asunto(s)
Linaje de la Célula/fisiología , Células Madre Embrionarias/metabolismo , Células Ependimogliales/metabolismo , Hipotálamo/metabolismo , Células-Madre Neurales/metabolismo , Animales , Células Cultivadas , Células Madre Embrionarias/citología , Células Ependimogliales/citología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hipotálamo/citología , Proteínas con Homeodominio LIM/metabolismo , Ratones , Células-Madre Neurales/citología , Factores de Transcripción/metabolismo
4.
PLoS One ; 13(7): e0200210, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29979748

RESUMEN

Hearing loss is the most common sensorineural disorder, affecting over 5% of the population worldwide. Its most frequent cause is the loss of hair cells (HCs), the mechanosensory receptors of the cochlea. HCs transduce incoming sounds into electrical signals that activate auditory neurons, which in turn send this information to the brain. Although some spontaneous HC regeneration has been observed in neonatal mammals, the very small pool of putative progenitor cells that have been identified in the adult mammalian cochlea is not able to replace the damaged HCs, making any hearing impairment permanent. To date, guided differentiation of human cells to HC-like cells has only been achieved using either embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs). However, use of such cell types suffers from a number of important disadvantages, such as the risk of tumourigenicity if transplanted into the host´s tissue. We have obtained cells expressing hair cell markers from cultures of human fibroblasts by overexpression of GFI1, Pou4f3 and ATOH1 (GPA), three genes that are known to play a critical role in the development of HCs. Immunocytochemical, qPCR and RNAseq analyses demonstrate the expression of genes typically expressed by HCs in the transdifferentiated cells. Our protocol represents a much faster approach than the methods applied to ESCs and iPSCs and validates the combination of GPA as a set of genes whose activation leads to the direct conversion of human somatic cells towards the hair cell lineage. Our observations are expected to contribute to the development of future therapies aimed at the regeneration of the auditory organ and the restoration of hearing.


Asunto(s)
Transdiferenciación Celular/fisiología , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores/metabolismo , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos , Células Ciliadas Auditivas/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Miosina VIIa , Miosinas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factor de Transcripción Brn-3C/genética , Factor de Transcripción Brn-3C/metabolismo , Factores de Transcripción/genética , Tretinoina/farmacología
5.
Dev Dyn ; 245(3): 388-401, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26442704

RESUMEN

BACKGROUND: Retinoic acid (RA), the bioactive derivative of vitamin A, is essential for vertebrate heart development. Both excess and reduced RA signaling lead to cardiovascular malformations affecting the outflow tract (OFT). To address the cellular mechanisms underlying the effects of RA signaling during OFT morphogenesis, we used transient maternal RA supplementation to rescue the early lethality resulting from inactivation of the murine retinaldehyde dehydrogenase 2 (Raldh2) gene. RESULTS: By embryonic day 13.5, all rescued Raldh2(-/-) hearts exhibit severe, reproducible OFT septation defects, although wild-type and Raldh2(+/-) littermates have normal hearts. Cardiac neural crest cells (cNCC) were present in OFT cushions of Raldh2(-/-) mutant embryos but ectopically located in the periphery of the endocardial cushions, rather than immediately underlying the endocardium. Excess mesenchyme was generated by Raldh2(-/-) mutant endocardium, which displaced cNCC derivatives from their subendocardial, medial position. CONCLUSIONS: RA signaling affects not only cNCC numbers but also their position relative to endocardial mesenchyme during the septation process. Our study shows that inappropriate coordination between the different cell types of the OFT perturbs its morphogenesis and leads to a severe congenital heart defect, persistent truncus arteriosus.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Linaje de la Célula/fisiología , Corazón/embriología , Organogénesis/fisiología , Transducción de Señal/fisiología , Tretinoina/metabolismo , Aldehído Oxidorreductasas/genética , Animales , Ratones , Ratones Noqueados
6.
Diabetes ; 63(5): 1572-82, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24487022

RESUMEN

Transcription factor forkhead box O1 (FoxO1) regulates energy expenditure (EE), food intake, and hepatic glucose production. These activities have been mapped to specific hypothalamic neuronal populations using cell type-specific knockout experiments in mice. To parse out the integrated output of FoxO1-dependent transcription from different neuronal populations and multiple hypothalamic regions, we used transgenic mice expressing Cre recombinase from the Nkx2.1 promoter to ablate loxP-flanked Foxo1 alleles from a majority of hypothalamic neurons (Foxo1KO(Nkx2.1) mice). This strategy resulted in the expected inhibition of FoxO1 expression, but only produced a transient reduction of body weight as well as a decreased body length. The transient decrease of body weight in male mice was accompanied by decreased fat mass. Male Foxo1KO(Nkx2.1) mice show food intake similar to that in wild-type controls, and, although female knockout mice eat less, they do so in proportion to a reduced body size. EE is unaffected in Foxo1KO(Nkx2.1) mice, although small increases in body temperature are present. Unlike other neuron-specific Foxo1 knockout mice, Foxo1KO(Nkx2.1) mice are not protected from diet-induced obesity. These studies indicate that, unlike the metabolic effects of highly restricted neuronal subsets (proopiomelanocortin, neuropeptide Y/agouti-related peptide, and steroidogenic factor 1), those of neurons derived from the Nkx2.1 lineage either occur in a FoxO1-independent fashion or are compensated for through developmental plasticity.


Asunto(s)
Metabolismo Energético/fisiología , Factores de Transcripción Forkhead/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Peso Corporal/fisiología , Linaje de la Célula/fisiología , Ingestión de Alimentos/fisiología , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Masculino , Ratones , Ratones Transgénicos , Neuropéptido Y/metabolismo , Proteínas Nucleares/genética , Obesidad/genética , Obesidad/metabolismo , Proopiomelanocortina/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal/fisiología , Factor Nuclear Tiroideo 1 , Factores de Transcripción/genética
7.
Cell Reprogram ; 14(6): 530-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23194456

RESUMEN

In this study, we describe the production of buffalo parthenogenetic blastocysts and subsequent isolation of parthenogenetic embryonic stem cell (PGESC)-like cells. PGESC colonies exhibited dome-shaped morphology and were clearly distinguishable from the feeder layer cells. Different stages of development of parthenogenetic embryos and derived embryonic stem cell (ESC)-like cells expressed key ESC-specific markers, including OCT-4, NANOG, SOX-2, FOXD3, REX-1, STAT-3, TELOMERASE, NUCLEOSTEMIN, and cMYC. Immunofluorescence-based studies revealed that the PGESCs were positive for surface-based pluripotent markers, viz., SSEA-3, SSEA-4, TRA 1-80, TRA 1-60, CD-9, and CD-90 and exhibited high alkaline phosphatase (ALP) activity. PGEC cell-like cells formed embryoid body (EB)-like structures in hanging drop cultures and when cultured for extended period of time spontaneously differentiated into derivatives of three embryonic germ layers as confirmed by RT-PCR for ectodermal (CYTOKERATIN8, NF-68), mesodermal (MSX1, BMP-4, ASA), and endodermal markers (AFP, HNF-4, GATA-4). Differentiation of PGESCs toward the neuronal lineage was successfully directed by supplementation of serum-containing media with retinoic acid. Our results indicate that the isolated ESC-like cells from parthenogenetic blastocyst hold properties of ESCs and express markers of pluripotency. The pluripotency markers were also expressed by early cleavage-stage of buffalo embryos.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Búfalos/embriología , Embrión de Mamíferos/embriología , Células Madre Embrionarias/metabolismo , Partenogénesis , Células Madre Pluripotentes/metabolismo , Animales , Antineoplásicos/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Estratos Germinativos/citología , Estratos Germinativos/embriología , Células Madre Pluripotentes/citología , Tretinoina/farmacología
8.
J Ethnopharmacol ; 127(2): 242-50, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19922785

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Astragalus polysaccharide (APS), obtained from Astragalus membranaceus, displays a range of activities in many systems, including the promotion of immune responses, anti-inflammation, and the protection of vessels. It possesses potent pharmacological activity on differentiation to the erythroid lineage. AIM OF THE STUDY: To investigate the effects of APS on the erythroid differentiation and the mechanism of action by microarray analysis in K562 cells. MATERIALS AND METHODS: Benzidine staining, semi-quantitative RT-PCR, Western blot and microarray methods were used to survey the effects of APS on inducing erythroid differentiation and the changes of gene expression profile in K562 cells. RESULTS: Of the 13.2% positive cells detected by benzidine staining, the induction was the highest with 200 microg/ml APS on 72h. Ggamma-mRNA expression and fetal hemoglobin synthesis were significantly up-regulated. Microarray analysis showed that 31 genes were up-regulated and 108 genes were down-regulated. These differential expression genes generally regulate protein binding, cellular metabolic process, the cell proliferation, and transcriptional activator activity. The gamma-globin gene was up-regulated, the genes related with erythroid differentiation such as LMO2, Runx1 and GTF2I were up-regulated, while Bklf, Eklf, EPHB4 and Sp1 were down-regulated. CONCLUSIONS: Our studies indicate that APS indicate potent activities on the erythroid differentiation by modulating genes of LMO2, Klf1, Klf3, Runx1, EphB4 and Sp1, increasing gamma-globin mRNA expression and fetal hemoglobin synthesis in K562 cells.


Asunto(s)
Astragalus propinquus , Linaje de la Célula/efectos de los fármacos , Células Eritroides/efectos de los fármacos , Polisacáridos/farmacología , Análisis por Matrices de Proteínas/métodos , Linaje de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Células Eritroides/citología , Células Eritroides/metabolismo , Humanos , Células K562 , Polisacáridos/aislamiento & purificación
9.
Lancet Neurol ; 8(9): 819-29, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19679274

RESUMEN

The increasing availability of human pluripotent stem cells provides new prospects for neural-replacement strategies and disease-related basic research. With almost unlimited potential for self-renewal, the use of human embryonic stem cells (ESCs) bypasses the restricted supply and expandability of primary cells that has been a major bottleneck in previous neural transplantation approaches. Translation of developmental patterning and cell-type specification techniques to human ESC cultures enables in vitro generation of various neuronal and glial cell types. The derivation of stably proliferating neural stem cells from human ESCs further facilitates standardisation and circumvents the problem of batch-to-batch variations commonly encountered in "run-through" protocols, which promote terminal differentiation of pluripotent stem cells into somatic cell types without defined intermediate precursor stages. The advent of cell reprogramming offers an opportunity to translate these advances to induced pluripotent stem cells, thereby enabling the generation of neurons and glia from individual patients. Eventually, reprogramming could provide a supply of autologous neural cells for transplantation, and could lead to the establishment of cellular model systems of neurological diseases.


Asunto(s)
Encefalopatías/terapia , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Células Madre Embrionarias/trasplante , Trasplante de Células Madre/métodos , Trasplante de Células Madre/tendencias , Animales , Linaje de la Célula/fisiología , Evaluación Preclínica de Medicamentos , Células Madre Embrionarias/citología , Humanos , Modelos Biológicos , Neurogénesis/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Células Madre Pluripotentes/trasplante , Trasplante Autólogo/métodos , Trasplante Autólogo/tendencias
10.
J Neurosci Res ; 87(15): 3277-87, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19437544

RESUMEN

The general view of development consists of the acquisition of committed/differentiated phenotypes following a period of self-renewal and progenitor expansion. Lineage specification and progression are phenomena of antagonistic events, silencing tissue-specific gene expression in precursors to allow self-renewal and multipotentiality, and subsequently suppressing proliferation and embryonic gene expression to promote the restricted expression of tissue-specific genes during maturation. The high mobility group-containing Sox family of transcription factors constitutes one of the earliest classes of genes to be expressed during embryonic development. These proteins not only are indispensable for progenitor cell specification but also are critical for terminal differentiation of multiple cell types in a wide variety of lineages. Sox transcription factors are now known to induce or repress progenitor cell characteristics and cell proliferation or to activate the expression of tissue-specific genes. Sox proteins fulfill their diverse functions in developmental regulation by distinct molecular mechanisms. Not surprisingly, in addition to DNA binding and bending, Sox transcription factors also interact with different protein partners to function as coactivators or corepressors of downstream target genes. Here we seek to provide an overview of the current knowledge of Sox gene functional mechanisms, in an effort to understand their roles in both development and pathology.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Transcripción SOX/metabolismo , Células Madre/metabolismo , Animales , Proliferación Celular , Humanos , Organogénesis/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción SOX/genética , Transactivadores/genética , Transactivadores/metabolismo
11.
J Comp Neurol ; 515(1): 56-71, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19399893

RESUMEN

Numerous central nervous system (CNS) disorders share a common pathology in dysregulation of gamma-aminobutyric acid (GABA) inhibitory signaling. Transplantation of GABA-releasing cells at the site of disinhibition holds promise for alleviating disease symptoms with fewer side effects than traditional drug therapies. We manipulated fibroblast growth factor (FGF)-2 deprivation and mammalian achaete-scute homolog (MASH)1 transcription factor levels in an attempt to amplify the default GABAergic neuronal fate in cultured rat embryonic neural precursor cells (NPCs) for use in transplantation studies. Naïve and MASH1 lentivirus-transduced NPCs were maintained in FGF-2 or deprived of FGF-2 for varying lengths of time. Immunostaining and quantitative analysis showed that GABA- and beta-III-tubulin-immunoreactive cells generally decreased through successive passages, suggesting a loss of neurogenic potential in rat neurospheres expanded in vitro. However, FGF-2 deprivation resulted in a small, but significantly increased population of GABAergic cells derived from passaged neurospheres. In contrast to naïve and GFP lentivirus-transduced clones, MASH1 transduction resulted in increased bromodeoxyuridine (BrdU) incorporation and clonal colony size. Western blotting showed that MASH1 overexpression and FGF-2 deprivation additively increased beta-III-tubulin and decreased cyclic nucleotide phosphodiesterase (CNPase) expression, whereas FGF-2 deprivation alone attenuated glial fibrillary acidic protein (GFAP) expression. These results suggest that low FGF-2 signaling and MASH1 activity can operate in concert to enrich NPC cultures for a GABA neuronal phenotype. When transplanted into the adult rat spinal cord, this combination also yielded GABAergic neurons. These findings indicate that, even for successful utilization of the default GABAergic neuronal precursor fate, a combination of both extrinsic and intrinsic manipulations will likely be necessary to realize the full potential of NSC grafts in restoring function.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Trasplante de Tejido Encefálico/métodos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Interneuronas/trasplante , Trasplante de Células Madre/métodos , Ácido gamma-Aminobutírico/metabolismo , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Interneuronas/citología , Interneuronas/metabolismo , Masculino , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Médula Espinal/metabolismo , Médula Espinal/cirugía , Transducción Genética/métodos , Tubulina (Proteína)/metabolismo
12.
BMC Biotechnol ; 8: 56, 2008 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-18638414

RESUMEN

BACKGROUND: Although extensive research has been performed to control differentiation of neural stem cells - still, the response of those cells to diverse cell culture conditions often appears to be random and difficult to predict. To this end, we strived to obtain stabilized protocol of NHA cells differentiation - allowing for an increase in percentage yield of neuronal cells. RESULTS: Uncommitted GFAP and SOX2 positive neural progenitors - so-called, Normal Human Astrocytes (NHA) were differentiated in different environmental conditions to: only neural cells consisted of neuronal [MAP2+, GFAP-] and glial [GFAP+, MAP2-] population, non-neural cells [CD44+, VIMENTIN+, FIBRONECTIN+, MAP2-, GFAP-, S100beta-, SOX2-], or mixture of neural and non-neural cells.In spite of successfully increasing the percentage yield of glial and neuronal vs. non-neural cells by means of environmental changes, we were not able to increase significantly the percentage of neuronal (GABA-ergic and catecholaminergic) over glial cells under several different cell culture testing conditions. Supplementing serum-free medium with several growth factors (SHH, bFGF, GDNF) did not radically change the ratio between neuronal and glial cells--i.e., 1,1:1 in medium without growth factors and 1,4:1 in medium with GDNF, respectively. CONCLUSION: We suggest that biotechnologists attempting to enrich in vitro neural cell cultures in one type of cells - such as that required for transplantology purposes, should consider the strong limiting influence of intrinsic factors upon extracellular factors commonly tested in cell culture conditions.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Neuroglía/citología , Neuroglía/fisiología , Neuronas/citología , Neuronas/fisiología , Células Madre/efectos de los fármacos , Células Madre/fisiología , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Biomarcadores/análisis , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula/métodos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Células Cultivadas , Medio de Cultivo Libre de Suero/farmacología , Proteínas de Unión al ADN/análisis , Factores de Crecimiento de Fibroblastos/farmacología , Fibronectinas/análisis , Proteína Ácida Fibrilar de la Glía/análisis , Proteínas HMGB/análisis , Humanos , Receptores de Hialuranos/análisis , Proteínas Asociadas a Microtúbulos/análisis , Factores de Crecimiento Nervioso/análisis , Factores de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/efectos de los fármacos , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/análisis , Factores de Transcripción SOXB1 , Células Madre/citología , Factores de Transcripción/análisis , Vimentina/análisis
13.
Cytotherapy ; 9(5): 427-38, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17786604

RESUMEN

BACKGROUND: Periodontal disease is a degenerative illness that leads to resorption of the alveolar bone. Mesenchymal stromal cells (MSC) represent a novel tool for the production of biologic constructs for the treatment of degenerative bone diseases. The preparation of MSC differentiated into osteogenic lineage for clinical use requires the fulfillment of strict good manufacturing practice (GMP) procedures. METHODS: MSC were isolated from BM samples and then cultured under GMP conditions. MSC were characterized phenotypically and for their differentiative potential. Cells were seeded onto collagen scaffolds (Gingistat) and induced to differentiate into osteogenic lineages using clinical grade drugs compared with standard osteogenic supplements. Alizarin Red S stain was used to test the deposition of the mineral matrix. Standard microbiologic analysis was performed to verify the product sterility. RESULTS: The resulting MSC were negative for CD33, CD34 and HLA-DR but showed high expression of CD90, CD105 and HLA-ABC (average expressions of 94.3%, 75.8% and 94.2%, respectively). Chondrogenic, osteogenic and adipogenic differentiation potential was demonstrated. The MSC retained their ability to differentiate into osteogenic lineage when seeded onto collagen scaffolds after exposure to a clinical grade medium. Cell numbers and cell viability were adequate for clinical use, and microbiologic assays demonstrated the absence of any contamination. DISCUSSION: In the specific context of a degenerative bone disease with limited involvement of skeletal tissue, the combined use of MSC, exposed to an osteogenic clinical grade medium, and biomimetic biodegradable scaffolds offers the possibility of producing adequate numbers of biologic tissue-engineered cell-based constructs for use in clinical trials.


Asunto(s)
Materiales Biocompatibles/farmacología , Regeneración Ósea/fisiología , Resorción Ósea/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Enfermedades Periodontales/terapia , Células del Estroma/fisiología , Implantes Absorbibles , Conservadores de la Densidad Ósea/farmacología , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Regeneración Ósea/efectos de los fármacos , Resorción Ósea/etiología , Resorción Ósea/fisiopatología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Células Cultivadas , Colágeno/farmacología , Regeneración Tisular Dirigida/métodos , Humanos , Maxilares/patología , Maxilares/fisiopatología , Osteoblastos/citología , Osteoblastos/fisiología , Enfermedades Periodontales/fisiopatología , Células del Estroma/efectos de los fármacos
14.
Neuron ; 55(2): 217-30, 2007 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-17640524

RESUMEN

The progression of progenitors to oligodendrocytes requires proliferative arrest and the activation of a transcriptional program of differentiation. While regulation of cell cycle exit has been extensively characterized, the molecular mechanisms responsible for the initiation of differentiation remain ill-defined. Here, we identify the transcription factor Yin Yang 1 (YY1) as a critical regulator of oligodendrocyte progenitor differentiation. Conditional ablation of yy1 in the oligodendrocyte lineage in vivo induces a phenotype characterized by defective myelination, ataxia, and tremor. At the cellular level, lack of yy1 arrests differentiation of oligodendrocyte progenitors after they exit from the cell cycle. At the molecular level, YY1 acts as a lineage-specific repressor of transcriptional inhibitors of myelin gene expression (Tcf4 and Id4), by recruiting histone deacetylase-1 to their promoters during oligodendrocyte differentiation. Thus, we identify YY1 as an essential component of the transcriptional network regulating the transition of oligodendrocyte progenitors from cell cycle exit to differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Oligodendroglía/metabolismo , Células Madre/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Linaje de la Célula/fisiología , Ratones , Ratones Noqueados , Ratones Mutantes , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Oligodendroglía/citología , Células Madre/citología , Transcripción Genética/fisiología
15.
BMC Dev Biol ; 7: 82, 2007 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-17612403

RESUMEN

BACKGROUND: Programmed cell death is used to remove excess cells between ommatidia in the Drosophila pupal retina. This death is required to establish the crystalline, hexagonal packing of ommatidia that characterizes the adult fly eye. In previously described echinus mutants, interommatidial cell sorting, which precedes cell death, occurred relatively normally. Interommatidial cell death was partially suppressed, resulting in adult eyes that contained excess pigment cells, and in which ommatidia were mildly disordered. These results have suggested that echinus functions in the pupal retina primarily to promote interommatidial cell death. RESULTS: We generated a number of new echinus alleles, some likely null mutants. Analysis of these alleles provides evidence that echinus has roles in cell sorting as well as cell death. echinus encodes a protein with homology to ubiquitin-specific proteases. These proteins cleave ubiquitin-conjugated proteins at the ubiquitin C-terminus. The echinus locus encodes multiple splice forms, including two proteins that lack residues thought to be critical for deubiquitination activity. Surprisingly, ubiquitous expression in the eye of versions of Echinus that lack residues critical for ubiquitin specific protease activity, as well as a version predicted to be functional, rescue the echinus loss-of-function phenotype. Finally, genetic interactions were not detected between echinus loss and gain-of-function and a number of known apoptotic regulators. These include Notch, EGFR, the caspases Dronc, Drice, Dcp-1, Dream, the caspase activators, Rpr, Hid, and Grim, the caspase inhibitor DIAP1, and Lozenge or Klumpfuss. CONCLUSION: The echinus locus encodes multiple splice forms of a protein with homology to ubiquitin-specific proteases, but protease activity is unlikely to be required for echinus function, at least when echinus is overexpressed. Characterization of likely echinus null alleles and genetic interactions suggests that echinus acts at a novel point(s) to regulate interommatidial cell sorting and/or cell death in the fly eye.


Asunto(s)
Apoptosis/genética , Cisteína Endopeptidasas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Retina/metabolismo , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Apoptosis/fisiología , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Cisteína Endopeptidasas/metabolismo , ADN Complementario/química , ADN Complementario/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestructura , Endopeptidasas/genética , Endopeptidasas/metabolismo , Ojo/citología , Ojo/metabolismo , Ojo/ultraestructura , Femenino , Expresión Génica , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Microscopía Electrónica de Rastreo , Datos de Secuencia Molecular , Mutación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , Retina/citología , Retina/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Ubiquitina/metabolismo , Proteasas Ubiquitina-Específicas
16.
Hepatology ; 45(6): 1462-70, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17538966

RESUMEN

UNLABELLED: The experimental protocols used in the investigation of stem cell-mediated liver regeneration in rodents are characterized by activation of the hepatic stem cell compartment in the canals of Hering followed by transit amplification of oval cells and their subsequent differentiation along hepatic lineages. Although the protocols are numerous and often used interchangeably across species, a thorough comparative phenotypic analysis of oval cells in rats and mice using well-established and generally acknowledged molecular markers has not been provided. In the present study, we evaluated and compared the molecular phenotypes of oval cells in several of the most commonly used protocols of stem cell-mediated liver regeneration-namely, treatment with 2-acetylaminofluorene and partial (70%) hepatectomy (AAF/PHx); a choline-deficient, ethionine-supplemented (CDE) diet; a 3,5-diethoxycarbonyl-1,4-dihydro-collidin (DDC) diet; and N-acetyl-paraaminophen (APAP). Reproducibly, oval cells showing reactivity for cytokeratins (CKs), muscle pyruvate kinase (MPK), the adenosine triphosphate-binding cassette transporter ABCG2/BCRP1 (ABCG2), alpha-fetoprotein (AFP), and delta-like protein 1/preadipocyte factor 1 (Dlk/Pref-1) were induced in rat liver treated according to the AAF/PHx and CDE but not the DDC protocol. In mouse liver, the CDE, DDC, and APAP protocols all induced CKs and ABCG2-positive oval cells. However, AFP and Dlk/Pref-1 expression was rarely detected in oval cells. CONCLUSION: Our results delineate remarkable phenotypic discrepancies exhibited by oval cells in stem cell-mediated liver regeneration between rats and mice and underline the importance of careful extrapolation between individual species.


Asunto(s)
Biomarcadores/metabolismo , Regeneración Hepática/fisiología , Hígado/citología , Hígado/fisiología , Células Madre/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteínas de Unión al Calcio , Linaje de la Célula/fisiología , Femenino , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Queratinas/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Fenotipo , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Células Madre/citología , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo
17.
Mol Reprod Dev ; 74(10): 1295-302, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17290421

RESUMEN

Embryonic stem (ES) cells derived from mammalian embryos have the ability to form any terminally differentiated cell of the body. We herein describe production of parthenogenetic buffalo (Bubalus Bubalis) blastocysts and subsequent isolation of an ES cell line. Established parthenogenetic ES (PGES) cells exhibited diploid karyotype and high telomerase activity. PGES cells showed remarkable long-term proliferative capacity providing the possibility for unlimited expansion in culture. Furthermore, these cells expressed key ES cell-specific markers defined for primate species including stage-specific embryonic antigen-4 (SSEA-4), tumor rejection antigen-1-81 (TRA-1-81), and octamer-binding transcription factor 4 (Oct-4). In vitro, in the absence of a feeder layer, cells readily formed embryoid bodies (EBs). When cultured for an extended period of time, EBs spontaneously differentiated into derivatives of three embryonic germ layers as detected by PCR for ectodermal (nestin, oligodendrocytes, and tubulin), mesodermal (scleraxis, alpha-skeletal actin, collagen II, and osteocalcin) and endodermal markers (insulin and alpha-fetoprotein). Differentiation of PGES cells toward chondrocyte lineage was directed by supplementing serum-containing media with ascorbic acid, beta-glycerophosphate, and dexamethasone. Moreover, when PGES cells were injected into nude mice, teratomas with derivatives representing all three embryonic germ layers were produced. Our results suggest that the cell line isolated from a parthenogenetic blastocyst holds properties of ES cells, and can be used as an in vitro model to study the effects of imprinting on cell differentiation and as an a invaluable material for extensive molecular studies on imprinted genes.


Asunto(s)
Búfalos/embriología , Diferenciación Celular , Células Madre Embrionarias/citología , Partenogénesis/fisiología , Animales , Biomarcadores/análisis , Linaje de la Célula/fisiología , Células Cultivadas , Células Madre Embrionarias/trasplante , Ratones , Ratones Desnudos
18.
Ment Retard Dev Disabil Res Rev ; 12(2): 85-96, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16807909

RESUMEN

In white matter disorders such as leukodystrophies (LD), periventricular leucomalacia (PVL), or multiple sclerosis (MS), the hypomyelination or the remyelination failure by oligodendrocyte progenitor cells involves errors in the sequence of events that normally occur during development when progenitors proliferate, migrate through the white matter, contact the axon, and differentiate into myelin-forming oligodendrocytes. Multiple mechanisms underlie the eventual progressive deterioration that typifies the natural history of developmental demyelination in LD and PVL and of adult-onset demyelination in MS. Over the past few years, pathophysiological studies have mostly focused on seeking abnormalities that impede oligodendroglial maturation at the level of migration, myelination, and survival. In contrast, there has been a strikingly lower interest for early proliferative and differentiation events that are likely to be equally critical for white matter development and myelin repair. This review highlights the Yin and Yang principles of interactions between intrinsic factors that coordinately regulate progenitor cell division and the onset of differentiation, i.e. the initial steps of oligodendrocyte lineage progression that are obviously crucial in health and diseases.


Asunto(s)
Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Oligodendroglía/patología , Adulto , Ciclina E/fisiología , Quinasa 2 Dependiente de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Humanos , Recién Nacido , Leucomalacia Periventricular/genética , Leucomalacia Periventricular/patología , Esclerosis Múltiple/patología , Vaina de Mielina/genética , Vaina de Mielina/patología , Fosforilación , Células Madre/patología
19.
J Cell Physiol ; 203(3): 573-82, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15573398

RESUMEN

While it has been assumed that osteoblasts in the human support osteoclast formation, in vitro evidence of this is currently lacking. We tested the ability of normal human trabecular bone-derived osteoblasts (NHBCs) to support osteoclast formation from human peripheral blood mononuclear cells (PBMC) in response to treatment with either 1alpha,25-dihydroxyvitamin D3 (1,25D) or parathyroid hormone (PTH), using a serum-replete medium previously used to support human osteoclast formation on a stroma of murine ST-2 cells. Under these conditions, NHBC did not support osteoclast formation, as assessed by morphological, histochemical, and functional criteria, despite our previous results demonstrating a link between induction of RANKL mRNA expression and NHBC phenotype in these media. We next tested a defined, serum-free medium (SDM) on NHBC phenotype, their expression of RANKL and OPG, and their ability to support osteoclast formation. SDM, containing dexamethasone (DEX) and 1,25D, induced phenotypic maturation of NHBC, based on the expression of STRO-1 and the bone/liver/kidney isoform of alkaline phosphatase (AP). PTH as a single factor did not induce phenotypic change. 1,25D and DEX induced the greatest ratio of RANKL:OPG mRNA, predictive of supporting osteoclast formation. Consistent with this, co-culture of NHBC with CD14+ PBMC, or bone marrow mononuclear cell (BMMC), or CD34+ BMMC precursors in SDM + 1,25D + DEX, resulted in functional osteoclast formation. Osteoclast formation also occurred in PTH + DEX stimulated co-cultures. Interestingly, SDM supplemented with recombinant RANKL (25-100 ng/ml) and M-CSF (25 ng/ml), did not induce osteoclast formation from any of the osteoclast precursor populations in stromal-free cultures, unlike serum-replete medium. This study demonstrates that under the appropriate conditions, adult human primary osteoblasts can support de novo osteoclast formation, and this model will enable the detailed study of the role of both cell types in this process.


Asunto(s)
Remodelación Ósea/fisiología , Huesos/metabolismo , Diferenciación Celular/fisiología , Osteoblastos/metabolismo , Osteoclastos/metabolismo , ADP-Ribosil Ciclasa/metabolismo , Fosfatasa Alcalina/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Animales , Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Huesos/citología , Huesos/efectos de los fármacos , Calcitriol/farmacología , Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Células Cultivadas , Técnicas de Cocultivo , Medio de Cultivo Libre de Suero/farmacología , Dexametasona/farmacología , Proteínas Ligadas a GPI , Glicoproteínas/genética , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacología , Ratones , Osteoblastos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoprotegerina , Hormona Paratiroidea/farmacología , Ligando RANK , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/genética , Receptores del Factor de Necrosis Tumoral/genética , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células del Estroma/metabolismo
20.
Glia ; 49(4): 480-91, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15578660

RESUMEN

Oligodendrocytes (OL), cells that myelinate axons in the CNS, differentiate from early to late oligodendrocyte progenitor cells (OPC) to become mature OL. Unlike the case in the rodent brain, myelin formation starts prenatally in the human brain, but the sequence of OL development and the onset of myelination are not well understood. We studied the human fetal forebrain at midgestation (17-23 gestational weeks, g.w.) using OL lineage-specific antibodies and mRNA probes. Early OPC were present in a gradient from the subventricular zone to the cortical plate. Their close apposition to radial glia fibers suggests a possible role of these fibers in OPC migration. Late OPC reached peak density in the subplate layer, whereas multipolar cells with the morphology of mature OL were restricted to the emerging white matter. At 20 g.w., myelinated axons were observed in the diencephalon, but not in the telencephalon, consistent with caudal-to-rostral progression of myelination. Interestingly, in organotypic slice cultures of the same gestational ages, the subventricular zone contained a considerably greater number of the mature OL cells, suggesting the presence of inhibitory signals in vivo. Overall, in addition to considerable similarities with rodents, important differences in temporal and spatial distribution and regulatory signals for OL differentiation exist in the human brain.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Oligodendroglía/metabolismo , Células Madre/metabolismo , Telencéfalo/embriología , Antígenos/genética , Antígenos/metabolismo , Biomarcadores , Comunicación Celular/fisiología , Linaje de la Célula/fisiología , Forma de la Célula/fisiología , Desarrollo Fetal , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/fisiología , Fibras Nerviosas Mielínicas/ultraestructura , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/citología , Técnicas de Cultivo de Órganos , Fenotipo , Proteoglicanos/genética , Proteoglicanos/metabolismo , ARN Mensajero/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Células Madre/citología , Telencéfalo/citología , Telencéfalo/metabolismo , Tálamo/citología , Tálamo/embriología , Tálamo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA