Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Lasers Med Sci ; 36(1): 139-146, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32607713

RESUMEN

Phototherapy is an effective therapeutic option in the treatment of vitiligo; however, responses varied among the different types. The underlying mechanism has scarcely been investigated. To investigate and compare the effects of phototherapy on the mutation of melanocyte lineage differentiated from human scalp-derived neural crest stem cells (HS-NCSCs) with p75 neurotrophin receptor expression positive and p75 neurotrophin receptor expression negative group in vitro, the HS-NCSCs were isolated from fetal scalp tissue, which is identified by immunofluorescent staining. The p75(+) and p75(-) cells from HS-NCSCs were isolated by magnetic cell sorting, respectively. The embryonic neural crest stem cell biomarkers were detected by RT-PCR. Narrow-band UVB (NB-UVB) was used to irradiate the cells. Cell proliferation was evaluated by cell count. Tyrosinase, Tyrp1, and Tyrp2 gene expression were measured by quantitative RT-PCR. Tyrosinase and GRCR protein levels were investigated by Western blot analysis. The electrophoretic strip showed that Sox2, Oct4, Sox10, and Nestin of p75(+) HS-NCSCs were brighter than the p75(-) HS-NCSCs. After the same dose radiation with NB-UVB, the cell proliferation of p75(+) group showed less inhibitory rate compared with the p75(-) HS-NCSCs. The tyrosinase mRNA and protein expression of differentiated melanocytes increased significantly in the group of p75(+) HS-NCSCs compared with the p75(-) group. The melanocytic mutation of p75(+) HS-NCSCs increased significantly compared with the p75(-) HS-NCSCs under NB-UVB, which indicated there were more melanocyte precursors in the differentiated cells from p75(+) HS-NCSCs. This may provide new insights for the different repigmentation efficacy of segmental and non-segmental vitiligo.


Asunto(s)
Linaje de la Célula/efectos de la radiación , Melanocitos/citología , Melanocitos/efectos de la radiación , Cresta Neural/citología , Fototerapia , Receptor de Factor de Crecimiento Nervioso/metabolismo , Cuero Cabelludo/citología , Células Madre/citología , Biomarcadores/metabolismo , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Humanos , Melanocitos/metabolismo , Mutación/genética , Células Madre/efectos de la radiación , Terapia Ultravioleta
2.
Development ; 146(16)2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31399472

RESUMEN

WNT/ß-catenin signaling is crucial for neural crest (NC) formation, yet the effects of the magnitude of the WNT signal remain ill-defined. Using a robust model of human NC formation based on human pluripotent stem cells (hPSCs), we expose that the WNT signal modulates the axial identity of NCs in a dose-dependent manner, with low WNT leading to anterior OTX+ HOX- NC and high WNT leading to posterior OTX- HOX+ NC. Differentiation tests of posterior NC confirm expected derivatives, including posterior-specific adrenal derivatives, and display partial capacity to generate anterior ectomesenchymal derivatives. Furthermore, unlike anterior NC, posterior NC exhibits a transient TBXT+/SOX2+ neuromesodermal precursor-like intermediate. Finally, we analyze the contributions of other signaling pathways in posterior NC formation, which suggest a crucial role for FGF in survival/proliferation, and a requirement of BMP for NC maturation. As expected retinoic acid (RA) and FGF are able to modulate HOX expression in the posterior NC. Surprisingly, early RA supplementation prohibits NC formation. This work reveals for the first time that the amplitude of WNT signaling can modulate the axial identity of NC cells in humans.


Asunto(s)
Cresta Neural/embriología , Vía de Señalización Wnt , beta Catenina/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Línea Celular , Polaridad Celular , Factores de Crecimiento de Fibroblastos/fisiología , Células Madre Embrionarias Humanas , Humanos , Cresta Neural/citología , Neurogénesis , Células Madre Pluripotentes , Tretinoina/metabolismo
3.
Cell Physiol Biochem ; 52(6): 1361-1380, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31075188

RESUMEN

BACKGROUND/AIMS: Human Dental Pulp Stem Cells (hDPSCs) are one of the most promising types of cells to regenerate nerve tissues. Standard DMEM+10% fetal bovine serum (FBS) culture medium allows a fast expansion of hDPSC as a surface-adherent cell monolayer. However, the use of FBS also compromises the clinical use of these protocols, and its longterm presence favors hDPSCs differentiation toward mesenchymal cell-derived lineages, at the expense of a reduced capability to generate neural cells. The objective of this work was to characterize the role of neurotrophin signaling on hDPSCs using a serum-free culture protocol, and to assess the neurogenic and gliogenic capacity of hDPSCs for future nerve tissue bioengineering and regeneration. METHODS: We compared the different expression of neurotrophin receptors by RT-PCR, Q-PCR, and IF of hDPSCs cultured with different growth media in the presence or absence of serum. Moreover, we assessed the response of hDPSCs to stimulation of neurotransmitter receptors by live cell calcium imaging under these different media. Finally, we compared the osteogenic potential of hDPSCs by Alizarin red staining, and the differentiation to gliogenic/neurogenic fates by immunostaining for Schwann lineage and neuronal lineage markers. We tested a commercial serum-free medium designed for the growth of mesenchymal stem cells: StemPro MSCTM (STP). RESULTS: hDPSCs cultured in STP generated small non-adherent floating dentospheres that showed very low proliferation rates, in contrast to standard FBS-containing medium. We found that hDPSCs grown in STP conditions overexpressed neurotrophin receptor genes NTRK2 (TrkB) and NTRK3 (TrkC). Interestingly, the stimulation of these receptors by adding their respective ligands BDNF and NT-3 to STP medium enhanced the neural crest (NC) progenitor features of cultured hDPSCs. We observed a 10 to 100-fold increase of migratory NC cell markers HNK1 and P75NTR, and a significant overexpression of pluripotency core factors SOX2, OCT4 and NANOG. Moreover, hDPSCs cultured in BDNF/NT-3 supplemented STP showed a largely increased potential to differentiate towards neuronal and Schwann glial lineage cells, assessed by positive immunostaining for DCX, NeuN and S100ß, p75NTR markers, respectively. CONCLUSION: Our results demonstrate that the use of BDNF and NT-3 combined with STP induced the partial reprogramming of ectomesenchymal hDPSCs to generate early NC progenitor cells, which are far more competent for neuronal and glial differentiation than hDPSCs grown in the presence of FBS.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Reprogramación Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero/farmacología , Factores de Crecimiento Nervioso/farmacología , Adolescente , Adulto , Antígenos CD57/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Pulpa Dental/citología , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/citología , Neurogénesis/efectos de los fármacos , Neurotrofina 3 , Receptor trkA/genética , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Células Madre/citología , Células Madre/metabolismo , Adulto Joven
4.
Cell Stem Cell ; 24(4): 637-653.e9, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30951662

RESUMEN

Increasing evidence suggests that cancer cells highjack developmental programs for disease initiation and progression. Melanoma arises from melanocytes that originate during development from neural crest stem cells (NCSCs). Here, we identified the transcription factor Yin Yang 1 (Yy1) as an NCSCs regulator. Conditional deletion of Yy1 in NCSCs resulted in stage-dependent hypoplasia of all major neural crest derivatives due to decreased proliferation and increased cell death. Moreover, conditional ablation of one Yy1 allele in a melanoma mouse model prevented tumorigenesis, indicating a particular susceptibility of melanoma cells to reduced Yy1 levels. Combined RNA sequencing (RNA-seq), chromatin immunoprecipitation (ChIP)-seq, and untargeted metabolomics demonstrated that YY1 governs multiple metabolic pathways and protein synthesis in both NCSCs and melanoma. In addition to directly regulating a metabolic gene set, YY1 can act upstream of MITF/c-MYC as part of a gene regulatory network controlling metabolism. Thus, both NCSC development and melanoma formation depend on an intricate YY1-controlled metabolic program.


Asunto(s)
Melanoma/metabolismo , Melanoma/patología , Cresta Neural/citología , Cresta Neural/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factor de Transcripción YY1/deficiencia
5.
Mol Med Rep ; 17(4): 5423-5427, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29393463

RESUMEN

Transplacental bone morphogenetic protein (BMP)4 RNA interference (RNAi) is a technique used to knockdown genes in embryos. BMP4 are essential for the development of nervous system in the differentiation of neural crest stem cells (NCSCs). The failure of differentiation and migration of NCSCs may lead to aganglionosis. In the present study, pregnant mice were divided into three groups: Ringer's group, pSES group and RNAi­BMP4 group. In order to silence the BMP4 gene in the first generation (F1), 11.5 day pregnant mice were injected with the small interfering RNA BMP4 plasmid, pSES or Ringer's solution via the tail vein. Semi­quantitative reverse transcriptase­polymerase chain reaction (RT­PCR)and western blotting were employed to ensure the downregulation of BMP4. Finally, X­rays were performed following a barium enema. Aganglionosis was diagnosed by general anatomy and immunohistochemistry. Compared with the control group, transplacental RNAi was able to downregulate the BMP4­Smad4 of 11.5 day embryos, as determined by semi­quantitative RT­PCR and western blotting. The megacolons of the mice were demonstrated by X­ray and confirmed by general anatomy. Aganglionosis of colonic mucosa and submucosa were diagnosed by pathology, and immunohistochemistry. Knockdown of BMP4 in pregnant mice at the middle embryonic stage led to aganglionosis. It was therefore demonstrated that BMP­Smad was essential to the NCSCs of middle stage embryos. BMP­Smad served important roles in the generation of aganglionosis. This technique of knockdown BMP4 gene may be used to establish an aganglionosis mouse model.


Asunto(s)
Proteína Morfogenética Ósea 4/deficiencia , Diferenciación Celular , Técnicas de Silenciamiento del Gen , Enfermedad de Hirschsprung/genética , Cresta Neural/citología , Células-Madre Neurales/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Embrión de Mamíferos , Femenino , Silenciador del Gen , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Enfermedad de Hirschsprung/metabolismo , Masculino , Ratones , Embarazo , ARN Interferente Pequeño/genética
6.
Arch Toxicol ; 91(11): 3613-3632, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28477266

RESUMEN

Many in vitro tests have been developed to screen for potential neurotoxicity. However, only few cell function-based tests have been used for comparative screening, and thus experience is scarce on how to confirm and evaluate screening hits. We addressed these questions for the neural crest cell migration test (cMINC). After an initial screen, a hit follow-up strategy was devised. A library of 75 compounds plus internal controls (NTP80-list), assembled by the National Toxicology Program of the USA (NTP) was used. It contained some known classes of (developmental) neurotoxic compounds. The primary screen yielded 23 confirmed hits, which comprised ten flame retardants, seven pesticides and six drug-like compounds. Comparison of concentration-response curves for migration and viability showed that all hits were specific. The extent to which migration was inhibited was 25-90%, and two organochlorine pesticides (DDT, heptachlor) were most efficient. In the second part of this study, (1) the cMINC assay was repeated under conditions that prevent proliferation; (2) a transwell migration assay was used as a different type of migration assay; (3) cells were traced to assess cell speed. Some toxicants had largely varying effects between assays, but each hit was confirmed in at least one additional test. This comparative study allows an estimate on how confidently the primary hits from a cell function-based screen can be considered as toxicants disturbing a key neurodevelopmental process. Testing of the NTP80-list in more assays will be highly interesting to assemble a test battery and to build prediction models for developmental toxicity.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Cresta Neural/citología , Pruebas de Toxicidad/métodos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , DDT/toxicidad , Evaluación Preclínica de Medicamentos/métodos , Heptacloro/toxicidad , Humanos , Cresta Neural/efectos de los fármacos , Imagen de Lapso de Tiempo
7.
ALTEX ; 34(1): 75-94, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27463612

RESUMEN

Migration of neural crest cells (NCCs) is one of the pivotal processes of human fetal development. Malformations arise if NCC migration and differentiation are impaired genetically or by toxicants. In the currently available test systems for migration inhibition of NCC (MINC), the manual generation of a cell-free space results in extreme operator dependencies, and limits throughput. Here a new test format was established. The assay avoids scratching by plating cells around a commercially available circular stopper. Removal of the stopper barrier after cell attachment initiates migration. This microwell-based circular migration zone NCC function assay (cMINC) was further optimized for toxicological testing of human pluripotent stem cell (hPSC)-derived NCCs. The challenge of obtaining data on viability and migration by automated image processing was addressed by developing a freeware. Data on cell proliferation were obtained by labelling replicating cells, and by careful assessment of cell viability for each experimental sample. The role of cell proliferation as an experimental confounder was tested experimentally by performing the cMINC in the presence of the proliferation-inhibiting drug cytosine arabinoside (AraC), and by a careful evaluation of mitotic events over time. Data from these studies led to an adaptation of the test protocol, so that toxicant exposure was limited to 24 h. Under these conditions, a prediction model was developed that allows classification of toxicants as either inactive, leading to unspecific cytotoxicity, or specifically inhibiting NC migration at non-cytotoxic concentrations.


Asunto(s)
Ensayos de Migración Celular/métodos , Movimiento Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Cresta Neural/efectos de los fármacos , Proliferación Celular , Supervivencia Celular , Humanos , Cresta Neural/citología , Células Madre , Teratógenos/toxicidad
8.
Stem Cells ; 34(11): 2721-2732, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27300003

RESUMEN

Prenatal folic acid (FA) supplementation prevents neural tube defects. Folate receptor alpha (FRα) is critical for embryonic development, including neural crest (NC) development. Previously we showed that FRα translocates to the nucleus in response to FA, where it acts as a transcription factor. In this study, we examined if FA through interaction with FRα regulates stem cell characteristics of cranial neural crest cells (CNCCs)-critical for normal development. We hypothesized that FRα upregulates coding genes and simultaneously downregulates non-coding miRNA which targets coding genes in CNCCs. Quantitative RT-PCR and chromatin immunoprecipitation showed that FRα upregulates Oct4, Sox2, and Klf4 by binding to their cis-regulator elements-5' enhancer/promoters defined by H3K27Ac and p300 occupancy. FA via FRα downregulates miRNAs, miR-138 and miR-let-7, which target Oct4 and Trim71 (an Oct4 downstream effector), respectively. Co-immunoprecipitation data suggests that FRα interacts with the Drosha-DGCR8 complex to affect pre-miRNA processing. Transfecting anti-miR-138 or anti-miR-let-7 into non-proliferating neural crest cells (NCCs) derived from Splotch (Sp-/- ), restored their proliferation potential. In summary, these results suggest a novel pleiotropic role of FRα: (a) direct activation of Oct4, Sox2, and Klf4 genes; and (b) repression of biogenesis of miRNAs that target these genes or their effector molecules. Stem Cells 2016;34:2721-2732.


Asunto(s)
Receptor 1 de Folato/genética , Factores de Transcripción de Tipo Kruppel/genética , MicroARNs/genética , Células-Madre Neurales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción SOXB1/genética , Animales , Antagomirs/genética , Antagomirs/metabolismo , Femenino , Receptor 1 de Folato/antagonistas & inhibidores , Receptor 1 de Folato/metabolismo , Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/agonistas , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Cresta Neural/citología , Cresta Neural/efectos de los fármacos , Cresta Neural/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/agonistas , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factor de Transcripción PAX3/deficiencia , Factor de Transcripción PAX3/genética , Regiones Promotoras Genéticas , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Factores de Transcripción SOXB1/agonistas , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
9.
Stem Cells Dev ; 25(13): 1020-32, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26956615

RESUMEN

The specification of pluripotent stem cells into the bone-forming osteoblasts has been explored in a number of studies. However, the current body of literature has yet to adequately address the role of Wnt glycoproteins in the differentiation of pluripotent stem cells along the osteogenic lineage. During mouse embryonic stem cell (ESC) in vitro osteogenesis, the noncanonical WNT5a is expressed early on. Cells either sorted by their positive WNT5a expression or when supplemented with recombinant WNT5a (rWNT5a) during a 2-day window showed significantly enhanced osteogenic yield. Mechanistically, rWNT5a supplementation upregulated protein kinase C (PKC), calcium/calmodulin-dependent kinase II (CamKII) and c-Jun N-terminal kinase (JNK) activity while antagonizing the key effector of canonical Wnt signaling: ß-catenin. Conversely, when recombinant WNT3a (rWNT3a) or other positive regulators of ß-catenin were employed during this same time window there was a decrease in osteogenic marker expression. However, if rWNT3a was supplemented during a time window following rWNT5a treatment, osteogenic differentiation was enhanced both in murine and human ESCs. Elucidating the role of these WNT ligands in directing the early stages of osteogenesis has the potential to considerably improve tissue engineering protocols and applications for regenerative medicine.


Asunto(s)
Linaje de la Célula , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias de Ratones/citología , Osteogénesis , Proteína Wnt-5a/metabolismo , Animales , Calcificación Fisiológica/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Colecalciferol/farmacología , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Cresta Neural/citología , Osteogénesis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Proteína Wnt3A/farmacología , beta Catenina/metabolismo
10.
Stem Cell Reports ; 4(4): 712-26, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25818812

RESUMEN

Here we report the successful generation and long-term expansion of SOX9-expressing CD271(+)PDGFRα(+)CD73(+) chondrogenic ectomesenchymal cells from the PAX3/SOX10/FOXD3-expressing MIXL1(-)CD271(hi)PDGFRα(lo)CD73(-) neural crest-like progeny of human pluripotent stem cells in a chemically defined medium supplemented with Nodal/Activin/transforming growth factorß (TGFß) inhibitor and fibroblast growth factor (FGF). When "primed" with TGFß, such cells efficiently formed translucent cartilage particles, which were completely mineralized in 12 weeks in immunocompromized mice. The ectomesenchymal cells were expandable without loss of chondrogenic potential for at least 16 passages. They maintained normal karyotype for at least 10 passages and expressed genes representing embryonic progenitors (SOX4/12, LIN28A/B), cranial mesenchyme (ALX1/3/4), and chondroprogenitors (SOX9, COL2A1) of neural crest origin (SOX8/9, NGFR, NES). Ectomesenchyme is a source of many craniofacial bone and cartilage structures. The method we describe for obtaining a large quantity of human ectomesenchymal cells will help to model craniofacial disorders in vitro and potentially provide cells for the repair of craniofacial damage.


Asunto(s)
Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Pluripotentes/citología , Factor de Transcripción SOX9/metabolismo , Antígenos de Superficie/metabolismo , Biomarcadores , Cartílago , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Autorrenovación de las Células/genética , Condrogénesis/efectos de los fármacos , Condrogénesis/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunofenotipificación , Cresta Neural/citología , Células Madre Pluripotentes/efectos de los fármacos , Factor de Transcripción SOX9/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología
11.
Birth Defects Res B Dev Reprod Toxicol ; 104(1): 11-22, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25689142

RESUMEN

BACKGROUND: Developmental zinc (Zn) deficiency increases the incidence of heart anomalies in rat fetuses, in regions and structures derived from the outflow tract. Given that the development of the outflow tract requires the presence of cardiac neural crest cells (cNCC), we speculated that Zn deficiency selectively kills cNCC and could lead to heart malformations. METHODS: Cardiac NCC were isolated from E10.5 rat embryos and cultured in control media (CTRL), media containing 3 µM of the cell permeable metal chelator N, N, N', N'-tetrakis (2-pyridylmethyl) ethylene diamine (TPEN), or in TPEN-treated media supplemented with 3 µM Zn (TPEN + Zn). Cardiac NCC were collected after 6, 8, and 24 h of treatment to assess cell viability, proliferation, and apoptosis. RESULTS: The addition of TPEN to the culture media reduced free intracellular Zn pools and cell viability as assessed by low ATP production, compared to cells grown in control or Zn-supplemented media. There was an accumulation of reactive oxygen species, a release of mitochondrial cytochrome c into the cytoplasm, and an increased cellular expression of active caspase-3 in TPEN-treated cNCC compared to cNCC cultured in CTRL or TPEN + Zn media. CONCLUSION: Zn deficiency can result in oxidative stress in cNCC, and subsequent decreases in their population and metabolic activity. These data support the concept that Zn deficiency associated developmental heart defects may arise in part as a consequence of altered cNCC metabolism.


Asunto(s)
Espacio Intracelular/metabolismo , Miocardio/citología , Cresta Neural/citología , Zinc/deficiencia , Zinc/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cobre/metabolismo , Citocromos c/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Etilenodiaminas/farmacología , Femenino , Hierro/metabolismo , Cresta Neural/efectos de los fármacos , Cresta Neural/metabolismo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Zinc/farmacología
12.
Dev Biol ; 396(1): 107-20, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25281935

RESUMEN

In this study, we investigated the gene regulatory network that governs formation of the Zona limitans intrathalamica (ZLI), a signaling center that secretes Sonic Hedgehog (Shh) to control the growth and regionalization of the caudal forebrain. Using loss- and gain-of-function, explants and grafting experiments in amphibians, we demonstrate that barhl2 acts downstream of otx2 and together with the iroquois (irx)-3 gene in establishment of the ZLI compartment initiated by Shh influence. We find that the presumptive (pre)-ZLI domain expresses barhl2, otx2 and irx3, whereas the thalamus territory caudally bordering the pre-ZLI expresses barhl2, otx2 and irx1/2 and early on irx3. We demonstrate that Barhl2 activity is required for determination of the ZLI and thalamus fates and that within the p2 alar plate the ratio of Irx3 to Irx1/2 contributes to ZLI specification and size determination. We show that when continuously exposed to Shh, neuroepithelial cells coexpressing barhl2, otx2 and irx3 acquire two characteristics of the ZLI compartment-the competence to express shh and the ability to segregate from anterior neural plate cells. In contrast, neuroepithelial cells expressing barhl2, otx2 and irx1/2, are not competent to express shh. Noteworthy in explants, under Shh influence, ZLI-like cells segregate from thalamic-like cells. Our study establishes that Barhl2 activity plays a key role in p2 alar plate patterning, specifically ZLI formation, and provides new insights on establishment of the signaling center of the caudal forebrain.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/fisiología , Proteínas de Homeodominio/fisiología , Proteínas del Tejido Nervioso/fisiología , Factores de Transcripción Otx/fisiología , Prosencéfalo/embriología , Tálamo/embriología , Factores de Transcripción/fisiología , Proteínas de Xenopus/fisiología , Animales , Blastómeros/ultraestructura , Tipificación del Cuerpo , Perfilación de la Expresión Génica , Genes Homeobox , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Cresta Neural/citología , Células Neuroepiteliales/citología , Oligonucleótidos Antisentido/química , Ratas , Transducción de Señal , Factores de Tiempo , Xenopus laevis
13.
Stem Cells Dev ; 23(1): 44-55, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23952781

RESUMEN

Human skin-derived precursors (hSKP) are postnatal stem cells with neural crest properties that reside in the dermis of human skin. These cells can be easily isolated from small (fore) skin segments and have the capacity to differentiate into multiple cell types. In this study, we show that upon exposure to hepatogenic growth factors and cytokines, hSKP acquire sufficient hepatic features that could make these cells suitable in vitro tools for hepatotoxicity screening of new chemical entities and already existing pharmaceutical compounds. Indeed, hepatic differentiated hSKP [hSKP-derived hepatic progenitor cells (hSKP-HPC)] express hepatic progenitor cell markers (EPCAM, NCAM2, PROM1) and adult hepatocyte markers (ALB), as well as key biotransformation enzymes (CYP1B1, FMO1, GSTA4, GSTM3) and influx and efflux drug transporters (ABCC4, ABCA1, SLC2A5). Using a toxicogenomics approach, we could demonstrate that hSKP-HPC respond to acetaminophen exposure in a comparable way to primary human hepatocytes in culture. The toxicological responses "liver damage", "liver proliferation", "liver necrosis" and "liver steatosis" were found to be significantly enriched in both in vitro models. Also genes associated with either cytotoxic responses or induction of apoptosis (BCL2L11, FOS, HMOX1, TIMP3, and AHR) were commonly upregulated and might represent future molecular biomarkers for hepatotoxicity. In conclusion, our data gives a first indication that hSKP-HPC might represent a suitable preclinical model for in vitro screening of hepatotoxicity. To the best of our knowledge, this is the first report in which human postnatal stem cells derived from skin are described as a potentially relevant cell source for in vitro hepatotoxicity testing of pharmaceutical compounds.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Hepatocitos/efectos de los fármacos , Piel/citología , Células Madre/efectos de los fármacos , Apoptosis/efectos de los fármacos , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Hepatocitos/citología , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Hígado/lesiones , Cresta Neural/citología , Células Madre/citología
14.
Vitam Horm ; 87: 143-73, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22127242

RESUMEN

Maternal folic acid (FA) intake has beneficial effects in preventing neural tube defects and may also play a role in the prevention of adult onset diseases such as Alzheimer's disease, dementia, neuropsychiatric disorders, cardiovascular diseases, and cerebral ischemia. This review will focus on the effects of maternal FA intake on neural crest stem cell proliferation and differentiation. Although FA is generally considered beneficial, it has the potential of promoting cell proliferation at the expense of differentiation. In some situations, this may lead to miscarriage or postnatal developmental abnormalities. Therefore, a blind approach such as "FA for everyone" is not necessarily the best course of action. Ultimately, the best approach for FA supplementation, and potentially other nutritional supplements, will include customized patient genomic profiles for determining dose and duration.


Asunto(s)
Células Madre Embrionarias/citología , Ácido Fólico/administración & dosificación , Fenómenos Fisiologicos Nutricionales Maternos , Cresta Neural/citología , Animales , Diferenciación Celular , Proliferación Celular , Anomalías Congénitas/metabolismo , Anomalías Congénitas/patología , Anomalías Congénitas/prevención & control , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Femenino , Desarrollo Fetal , Ácido Fólico/uso terapéutico , Humanos , Cresta Neural/metabolismo , Cresta Neural/patología , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Defectos del Tubo Neural/prevención & control , Embarazo
15.
Dev Dyn ; 238(10): 2522-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19718754

RESUMEN

Myosin-X (MyoX) belongs to a large family of unconventional, nonmuscle, actin-dependent motor proteins. We show that MyoX is predominantly expressed in cranial neural crest (CNC) cells in embryos of Xenopus laevis and is required for head and jaw cartilage development. Knockdown of MyoX expression using antisense morpholino oligonucleotides resulted in retarded migration of CNC cells into the pharyngeal arches, leading to subsequent hypoplasia of cartilage and inhibited outgrowth of the CNC-derived trigeminal nerve. In vitro migration assays on fibronectin using explanted CNC cells showed significant inhibition of filopodia formation, cell attachment, spreading and migration, accompanied by disruption of the actin cytoskeleton. These data support the conclusion that MyoX has an essential function in CNC migration in the vertebrate embryo.


Asunto(s)
Movimiento Celular/fisiología , Miosinas/metabolismo , Cresta Neural/citología , Proteínas de Xenopus/metabolismo , Xenopus laevis , Animales , Adhesión Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Morfogénesis/fisiología , Miosinas/genética , Cresta Neural/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Fenotipo , Proteínas de Xenopus/genética , Xenopus laevis/anatomía & histología , Xenopus laevis/embriología , Xenopus laevis/metabolismo
16.
Stem Cells ; 25(12): 3133-42, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17761753

RESUMEN

The activation of Notch signaling in neural crest stem cells (NCSCs) results in the rapid loss of neurogenic potential and differentiation into glia. We now show that the attenuation of endogenous Notch signaling within expanding NCSC clones by the Notch ligand soluble Jagged1 (sJ1), maintains NCSCs in a clonal self-renewing state in vitro without affecting their sensitivity to instructive differentiation signals observed previously during NCSC self-renewal. sJ1 functions as a competitive inhibitor of Notch signaling to modulate endogenous cell-cell communication to levels sufficient to inhibit neural differentiation but insufficient to instruct gliogenic differentiation. Attenuated Notch signaling promotes the induction and nonclassic release of fibroblast growth factor 1 (FGF1). The functions of sJ1 and FGF1 signaling are complementary, as abrogation of FGF signaling diminishes the ability of sJ1 to promote NCSC expansion, yet the secondary NCSCs maintain the dosage sensitivity of the founder. These results validate and build upon previous studies on the role of Notch signaling in stem cell self-renewal and suggest that the differentiation bias or self-renewal potential of NCSCs is intrinsically linked to the level of endogenous Notch signaling. This should provide a unique opportunity for the expansion of NCSCs ex vivo without altering their differentiation bias for clinical cell replacement or transplant strategies in tissue repair. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Diferenciación Celular/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de la Membrana/fisiología , Cresta Neural/citología , Cresta Neural/fisiología , Células Madre/citología , Células Madre/fisiología , Animales , Proteínas de Unión al Calcio/genética , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Células Clonales/citología , Células Clonales/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína Jagged-1 , Proteínas de la Membrana/genética , Inhibición Neural/genética , Inhibición Neural/fisiología , Neuronas/citología , Neuronas/fisiología , Ratas , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Receptor Notch1/fisiología , Proteínas Serrate-Jagged , Transducción de Señal/genética , Transducción de Señal/fisiología , Solubilidad , Células Madre/metabolismo
17.
Birth Defects Res A Clin Mol Teratol ; 79(3): 231-5, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17183584

RESUMEN

BACKGROUND: Diabetes mellitus during pregnancy increases the risk for congenital heart disease in the offspring. The majority of the cardiovascular malformations occur in the outflow tract and pharyngeal arch arteries, where neural crest cells are essential for normal development. We studied the effects of specific exposure of neural crest cells to elevated glucose on heart development. Antioxidants reduce the damaging effect of glucose on neural crest cells in vitro; therefore, we investigated the effect of supplementing N-acetylcysteine in vivo. METHODS: Cardiac neural crest of HH 8-12 chicken embryos was directly exposed by a single injection in the neural tube with 30 mM D-glucose (or 30 mM L-glucose as a control). To examine the effect of a reduction in oxidative stress, we added 2 mM N-acetylcysteine to the injected D-glucose. RESULTS: Exposure of neural crest cells to elevated D-glucose-induced congenital heart malformations in 82% of the embryos. In the embryos injected with L-glucose, only 9% developed a heart malformation. As expected, all malformations were located in the outflow tract and pharyngeal arch arteries. The frequency of heart malformations decreased from 82% to 27% when 2 mM N-acetylcysteine was added to the injected D-glucose. CONCLUSIONS: These data are the first to confirm that the vulnerability of neural crest cells to elevated glucose induces congenital heart malformations. The fact that N-acetylcysteine limits the teratogenicity of glucose implies that its damaging effect is mediated by an increase of oxidative stress in the neural crest cells.


Asunto(s)
Acetilcisteína/uso terapéutico , Glucosa/toxicidad , Cardiopatías Congénitas/inducido químicamente , Corazón/embriología , Cresta Neural/efectos de los fármacos , Animales , Embrión de Pollo , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/prevención & control , Cresta Neural/citología , Cresta Neural/embriología
18.
Development ; 132(24): 5491-502, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16291787

RESUMEN

Gonadotropin-releasing hormone (GnRH) is found in a wide range of vertebrate tissues, including the nervous system. In general, GnRH has two functions: endocrine, acting as a releasing hormone; and neuromodulatory, affecting neural activity in the peripheral and central nervous system. The best understood population of GnRH cells is that of the hypothalamus, which is essential for reproduction. Less well understood are the populations of GnRH cells found in the terminal nerve and midbrain, which appear to be neuromodulatory in function. The GnRH-containing cells of the midbrain are proposed to arise from the mesencephalic region of the neural tube. Previously, we showed that neuromodulatory GnRH cells of the terminal nerve arise from cranial neural crest. To test the hypothesis that neuromodulatory GnRH cells of the midbrain also arise from neural crest, we used gene knockdown experiments in zebrafish to disrupt neural crest development. We demonstrate that decrement of the function of foxd3 and/or sox10, two genes important for the development and specification of neural crest, resulted in a reduction and/or loss of GnRH cells of the midbrain, as well as a reduction in the number of terminal nerve GnRH cells. Therefore, our data support a neural crest origin for midbrain GnRH cells. Additionally, we demonstrate that knockdown of kallmann gene function resulted in the loss of endocrine GnRH cells of the hypothalamus, but not of neuromodulatory GnRH cells of the midbrain and terminal nerve, thus providing additional evidence for separate pathways controlling the development of neuromodulatory and endocrine GnRH cells.


Asunto(s)
Proteínas Portadoras/fisiología , Diferenciación Celular/fisiología , Factores de Transcripción Forkhead/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Proteínas del Grupo de Alta Movilidad/fisiología , Mesencéfalo/citología , Cresta Neural/citología , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Proteínas Portadoras/genética , Nervios Craneales/citología , Nervios Craneales/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/fisiología , Femenino , Factores de Transcripción Forkhead/genética , Proteínas del Grupo de Alta Movilidad/genética , Hipotálamo/citología , Hipotálamo/embriología , Masculino , Mesencéfalo/embriología , Mutación , Cresta Neural/embriología , Factores de Transcripción SOXE , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
19.
Mol Cell Biol ; 23(20): 7122-33, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14517283

RESUMEN

The serine/threonine kinase PAK4 is a target for the Rho GTPase Cdc42 and has been shown to regulate cell morphology and cytoskeletal organization in mammalian cells. To examine the physiological and developmental functions of PAK4, we have disrupted the PAK4 gene in mice. The absence of PAK4 led to lethality by embryonic day 11.5, a result most likely due to a defect in the fetal heart. Striking abnormalities were also evident in the nervous systems of PAK4-deficient embryos. These embryos had dramatic defects in neuronal development and axonal outgrowth. In particular, spinal cord motor neurons and interneurons failed to differentiate and migrate to their proper positions. This is probably related to the role for PAK4 in the regulation of cytoskeletal organization and cell and/or extracellular matrix adhesion. PAK4-null embryos also had defects in proper folding of the caudal portion of the neural tube, suggesting an important role for PAK4 in neural tube development.


Asunto(s)
Neuronas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteína de Unión al GTP cdc42/metabolismo , Animales , Axones/metabolismo , Northern Blotting , Bromodesoxiuridina/farmacología , Adhesión Celular , Diferenciación Celular , Línea Celular , Movimiento Celular , Clonación Molecular , Citoesqueleto/metabolismo , ADN Complementario/metabolismo , Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Vectores Genéticos , Genotipo , Corazón/embriología , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Cresta Neural/anomalías , Cresta Neural/citología , Cresta Neural/embriología , Neuronas/metabolismo , Neuronas/patología , Plásmidos/metabolismo , Reacción en Cadena de la Polimerasa , Médula Espinal/embriología , Factores de Tiempo , Distribución Tisular , Quinasas p21 Activadas
20.
Development ; 130(11): 2525-34, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12702665

RESUMEN

Targeted inactivation of the mouse retinaldehyde dehydrogenase 2 (RALDH2/ALDH1a2), the enzyme responsible for early embryonic retinoic acid synthesis, is embryonic lethal because of defects in early heart morphogenesis. Transient maternal RA supplementation from E7.5 to (at least) E8.5 rescues most of these defects, but the supplemented Raldh2(-/-) mutants die prenatally, from a lack of septation of the heart outflow tract (Niederreither, K., Vermot, J., Messaddeq, N., Schuhbaur, B., Chambon, P. and Dollé, P. (2001). Development 128, 1019-1031). We have investigated the developmental basis for this defect, and found that the RA-supplemented Raldh2(-/-) embryos exhibit impaired development of their posterior (3rd-6th) branchial arch region. While the development of the first and second arches and their derivatives, as well as the formation of the first branchial pouch, appear to proceed normally, more posterior pharyngeal pouches fail to form and the pharyngeal endoderm develops a rudimentary, pouch-like structure. All derivatives of the posterior branchial arches are affected. These include the aortic arches, pouch-derived organs (thymus, parathyroid gland) and post-otic neural crest cells, which fail to establish segmental migratory pathways and are misrouted caudally. Patterning and axonal outgrowth of the posterior (9th-12th) cranial nerves is also altered. Vagal crest deficiency in Raldh2(-/-) mutants leads to agenesis of the enteric ganglia, a condition reminiscent of human Hirschprung's disease. In addition, we provide evidence that: (i) wildtype Raldh2 expression is restricted to the posteriormost pharyngeal mesoderm; (ii) endogenous RA response occurs in both the pharyngeal endoderm and mesoderm, and extends more rostrally than Raldh2 expression up to the 2nd arch; (iii) RA target genes (Hoxa1, Hoxb1) are downregulated in both the pharyngeal endoderm and mesoderm of mutant embryos. Thus, RALDH2 plays a crucial role in producing RA required for pharyngeal development, and RA is one of the diffusible mesodermal signals that pattern the pharyngeal endoderm.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Región Branquial/embriología , Sistema Nervioso Entérico/embriología , Tretinoina/metabolismo , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Animales , Región Branquial/efectos de los fármacos , Región Branquial/metabolismo , Movimiento Celular , Nervios Craneales/anomalías , Nervios Craneales/embriología , Síndrome de DiGeorge/etiología , Endodermo/metabolismo , Sistema Nervioso Entérico/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Enfermedad de Hirschsprung/etiología , Humanos , Intercambio Materno-Fetal , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Cresta Neural/citología , Fenotipo , Embarazo , Rombencéfalo/embriología , Transducción de Señal , Tretinoina/administración & dosificación , Nervio Vago/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA