Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Perinatol ; 37(1): 77-80, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27684424

RESUMEN

OBJECTIVE: Fetal echocardiography, physical examination and pulse oximetry detect only half of coarctation of aorta (CoA) cases. We aimed to quantify delayed arrival and diminished amplitude of lower extremity photoplethysmographic (PPG) pulses relative to the right hand in affected patients. STUDY DESIGN: We studied 8 CoA infants and 32 healthy controls. The pulse arrival time difference between foot and hand (f-hTD) and pulse amplitude ratio (F/H) were measured on PPG signal waveforms by digitally-determining maxima and minima of systolic decrease of light transmission. Mann-Whitney test was used for group comparisons. RESULTS: In comparison to healthy newborns, CoA infants' PPG waveforms demonstrated prolonged f-hTD (mean±s.d. of 73.2±26.6 versus 35.2±8.3 ms, P<0.001) and lower F/H (0.57±0.26 versus 0.99±0.58, P=0.014). CONCLUSIONS: F-hTD and F/H are quantifiable from hand- and foot-derived PPG waveforms and are significantly different in CoA versus healthy newborns. Larger studies are needed to validate PPG for improved critical congenital heart disease screening.


Asunto(s)
Coartación Aórtica/diagnóstico , Coartación Aórtica/fisiopatología , Fotopletismografía , Presión Sanguínea , Estudios de Casos y Controles , Femenino , Frecuencia Cardíaca , Humanos , Recién Nacido , Masculino , Tamizaje Neonatal , Oximetría , Prueba de Estudio Conceptual
2.
Cell Death Dis ; 7(6): e2272, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27336716

RESUMEN

Partial pancreatic duct ligation (PDL) of mouse pancreas induces a doubling of the ß-cell mass mainly through proliferation of pre-existing and newly formed ß-cells. The molecular mechanism governing this process is still largely unknown. Given the inflammatory nature of PDL and inflammation-induced signaling via the signal transducer and activator of transcription 3 (STAT3), the activation and the role of STAT3 in PDL-induced ß-cell proliferation were investigated. Duct ligation stimulates the expression of several cytokines that can act as ligands inducing STAT3 signaling and phosphorylation in ß-cells. ß-Cell cycling increased by conditional ß-cell-specific Stat3 knockout and decreased by STAT3 activation through administration of interleukin-6. In addition, the level of DNA damage in ß-cells of PDL pancreas increased after deletion of Stat3. These data indicate a role for STAT3 in maintaining a steady state in the ß-cell, by modulating its cell cycle and protection from DNA damage.


Asunto(s)
Ciclo Celular , Citoprotección , Daño del ADN , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Factor de Transcripción STAT3/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Citocinas/metabolismo , Citoprotección/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/farmacología , Antígeno Ki-67/metabolismo , Ligadura , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Conductos Pancreáticos/efectos de los fármacos , Conductos Pancreáticos/patología , Proteínas Recombinantes/farmacología
3.
Neural Comput ; 25(1): 75-100, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22970870

RESUMEN

We simulate the inhibition of Ia-glutamatergic excitatory postsynaptic potential (EPSP) by preceding it with glycinergic recurrent (REN) and reciprocal (REC) inhibitory postsynaptic potentials (IPSPs). The inhibition is evaluated in the presence of voltage-dependent conductances of sodium, delayed rectifier potassium, and slow potassium in five α-motoneurons (MNs). We distribute the channels along the neuronal dendrites using, alternatively, a density function of exponential rise (ER), exponential decay (ED), or a step function (ST). We examine the change in EPSP amplitude, the rate of rise (RR), and the time integral (TI) due to inhibition. The results yield six major conclusions. First, the EPSP peak and the kinetics depending on the time interval are either amplified or depressed by the REC and REN shunting inhibitions. Second, the mean EPSP peak, its TI, and RR inhibition of ST, ER, and ED distributions turn out to be similar for analogous ranges of G. Third, for identical G, the large variations in the parameters' values can be attributed to the sodium conductance step (g(Na_step)) and the active dendritic area. We find that small g(Na_step) on a few dendrites maintains the EPSP peak, its TI, and RR inhibition similar to the passive state, but high g(Na_step) on many dendrites decrease the inhibition and sometimes generates even an excitatory effect. Fourth, the MN's input resistance does not alter the efficacy of EPSP inhibition. Fifth, the REC and REN inhibitions slightly change the EPSP peak and its RR. However, EPSP TI is depressed by the REN inhibition more than the REC inhibition. Finally, only an inhibitory effect shows up during the EPSP TI inhibition, while there are both inhibitory and excitatory impacts on the EPSP peak and its RR.


Asunto(s)
Simulación por Computador , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Inhibidores/fisiología , Modelos Neurológicos , Neuronas Motoras/fisiología , Inhibición Neural/fisiología , Animales , Gatos , Dendritas/fisiología , Ácido Glutámico/fisiología , Glicina/fisiología , Canales de Potasio/fisiología , Ratas , Canales de Sodio/fisiología , Sinapsis/fisiología
4.
Diabetologia ; 55(1): 154-65, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21947380

RESUMEN

AIMS/HYPOTHESIS: The aim of the study was to identify surface bio-markers and corresponding antibody tools that can be used for the imaging and immunoisolation of the pancreatic beta cell and its progenitors. This may prove essential to obtain therapeutic grade human beta cells via stem cell differentiation. METHODS: Using bioinformatics-driven data mining, we generated a gene list encoding putative plasma membrane proteins specifically expressed at distinct stages of the developing pancreas and islet beta cells. In situ hybridisation and immunohistochemistry were used to further prioritise and identify candidates. RESULTS: In the developing pancreas seizure related 6 homologue like (SEZ6L2), low density lipoprotein receptor-related protein 11 (LRP11), dispatched homologue 2 (Drosophila) (DISP2) and solute carrier family 30 (zinc transporter), member 8 (SLC30A8) were found to be expressed in early islet cells, whereas discoidin domain receptor tyrosine kinase 1 (DDR1) and delta/notch-like EGF repeat containing (DNER) were expressed in early pancreatic progenitors. The expression pattern of DDR1 overlaps with the early pancreatic and duodenal homeobox 1 (PDX1)⁺/NK6 homeobox 1 (NKX6-1)⁺ multipotent progenitor cells from embryonic day 11, whereas DNER expression in part overlaps with neurogenin 3 (NEUROG3)⁺ cells. In the adult pancreas SEZ6L2, LRP11, DISP2 and SLC30A8, but also FXYD domain containing ion transport regulator 2 (FXYD2), tetraspanin 7 (TSPAN7) and transmembrane protein 27 (TMEM27), retain an islet-specific expression, whereas DDR1 is undetectable. In contrast, DNER is expressed at low levels in peripheral mouse and human islet cells. Re-expression of DDR1 and upregulation of DNER is observed in duct-ligated pancreas. Antibodies to DNER and DISP2 have been successfully used in cell sorting. CONCLUSIONS/INTERPRETATION: Extracellular epitopes of SEZ6L2, LRP11, DISP2, DDR1 and DNER have been identified as useful tags by applying specific antibodies to visualise pancreatic cell types at specific stages of development. Furthermore, antibodies recognising DISP2 and DNER are suitable for FACS-mediated cell purification.


Asunto(s)
Antígenos de Superficie/metabolismo , Separación Celular/métodos , Islotes Pancreáticos/metabolismo , Células Madre/metabolismo , Adulto , Animales , Biomarcadores/metabolismo , Línea Celular , Biología Computacional/métodos , Minería de Datos , Citometría de Flujo , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Islotes Pancreáticos/citología , Islotes Pancreáticos/embriología , Ratones , Ratones Endogámicos BALB C , Técnicas de Cultivo de Órganos , Células Madre/citología
5.
Neural Comput ; 22(7): 1764-85, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20235819

RESUMEN

We simulate reconstructed alpha-motoneurons (MNs) under physiological and morphological realistic parameters and compare the modeled reciprocal (REC) and recurrent (REN) inhibitory postsynaptic potentials (IPSPs) containing voltage-dependent channels on the dendrites with the IPSPs of a passive MN model. Three distribution functions of the voltage-dependent channels on the dendrites are applied: a step function (ST) with uniform spatial dispersion; an exponential decay (ED) function, with channels with high density located proximal to the soma; and an exponential rise (ER) with a higher density of channels located distally. The excitatory and REN inhibitory inputs are located as a gaussian function on the dendrites, while the REC inhibitory synapses are located proximal to the soma. Our simulations generate four key results. (1) The distribution pattern of the voltage-dependent channels does not affect the IPSP peak, its time integral (TI), or its rate of rise (RR). However, the IPSP peak decreased in the presence of the active dendrites, while the EPSP peak increased. (2) Proximally located IPSP conductance produces greater IPSP peak, RR, and TI. (3) Increased duration of the IPSP produces greater RR and moderately increased TI and has a small effect on the peak amplitude. (4) The IPSP of both REC and REN models is specific to each MN: its amplitude is proportional to the MNs' input resistance, R(N); the increase of IPSP at the proximal location of the IPSP synapses is inversely related to R(N); and the effect of the IPSP conductance duration is insensitive to R(N).


Asunto(s)
Simulación por Computador , Potenciales Postsinápticos Inhibidores/fisiología , Canales Iónicos/fisiología , Neuronas Motoras/fisiología , Inhibición Neural/fisiología , Médula Espinal/fisiología , Animales , Compartimento Celular/fisiología , Membrana Celular/fisiología , Dendritas/fisiología , Humanos , Neuronas Motoras/clasificación , Conducción Nerviosa/fisiología , Vías Nerviosas/fisiología , Transmisión Sináptica/fisiología
6.
Cell Biol Toxicol ; 23(4): 241-56, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17171431

RESUMEN

The intestinal epithelium is a particularly interesting tissue as (1) it is in a constant cell renewal from a stem cell pool located in the crypts which form, with the underlying fibroblasts, a stem cell niche and (2) the pluripotent stem cells give rise to four main cell types: enterocytes, mucus, endocrine, and Paneth cells. The mechanisms leading to the determination of phenotype commitment and cell-specific expressions are still poorly understood. Although transgenic mouse models are powerful tools for elucidating the molecular cascades implicated in these processes, cell culture approaches bring easy and elegant ways to study cellular behavior, cell interactions, and cell signaling pathways for example. In the present review, we will describe the major tissue culture technologies that allow differentiation of epithelial cells from undifferentiated embryonic or crypt cells. We will point to the necessity of the re-creation of a complex microenvironment that allows full differentiation process to occur. We will also summarize the characteristics and interesting properties of the cell lines established from human colorectal tumors.


Asunto(s)
Alternativas a las Pruebas en Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Mucosa Intestinal/citología , Modelos Biológicos , Células Madre/citología , Animales , Línea Celular Transformada , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Humanos , Mucosa Intestinal/fisiología , Células Madre/fisiología
7.
Diabetes Metab ; 32(5 Pt 2): 532-3, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17130813

RESUMEN

This short review presents the recent breakthroughts in our understanding of the important steps controlling pancreas morphogenesis and differentiation, and on the transcription factors regulating pancreas organogenesis and islet cell differentiation and involved in the specification of the beta and alpha cell lineages. All these studies should permit a comprehensive view of the full genetic program necessary to produce mature and functional beta cells and thus, should be instrumental to guide future strategies for cell replacement therapies in type 1 diabetes.


Asunto(s)
Islotes Pancreáticos/crecimiento & desarrollo , Humanos , Factores de Transcripción/metabolismo
8.
Proc Natl Acad Sci U S A ; 102(5): 1490-5, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15668393

RESUMEN

Heterozygous mutations in the human POU-homeobox TCF2 (vHNF1, HNF1beta) gene are associated with maturity-onset diabetes of the young, type 5, and abnormal urogenital tract development. Recently, pancreas atrophies have been reported in several maturity-onset diabetes of the young type 5 patients, suggesting that TCF2 is required not only for adult pancreas function but also for its normal development. Tcf2-deficient mice die before gastrulation because of defective visceral endoderm formation. To investigate the role of this factor in pancreas development, we rescued this early lethality by tetraploid aggregation. We show that TCF2 has an essential function in the first steps of pancreas development, correlated with its expression domain that demarcates the entire pancreatic buds from the earliest stages. Lack of TCF2 results in pancreas agenesis by embryonic day 13.5. At earlier stages, only a dorsal bud rudiment forms transiently and expresses the transcription factors Ipf1 and Hlxb9 but lacks the key transcription factor involved in the acquisition of a pancreatic fate, Ptf1a, as well as all endocrine precursor cells. Regional specification of the gut also is perturbed in Tcf2-/- embryos as manifested by ectopic expression of Shh and lack of Ihh and Ipf1 in the posterior stomach and duodenum. Our results highlight the requirement of Tcf2 for ensuring both accurate expression of key regulator molecules in the stomach-duodenal epithelium and proper acquisition of the pancreatic fate. This study provides further insights into early molecular events controlling pancreas development and may contribute to the development of cell-replacement strategies for diabetes.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Páncreas/anomalías , Páncreas/embriología , Factores de Transcripción/fisiología , Animales , Apoptosis , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito , Heterocigoto , Hibridación in Situ , Ratones , Ratones Noqueados , Morfogénesis , Poliploidía , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transcripción Genética
9.
Neuron ; 31(2): 203-17, 2001 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-11502253

RESUMEN

We have examined how genetic pathways that specify neuronal identity and regulate neurogenesis interface in the vertebrate neural tube. Here, we demonstrate that expression of the proneural gene Neurogenin2 (Ngn2) in the ventral spinal cord results from the modular activity of three enhancers active in distinct progenitor domains, suggesting that Ngn2 expression is controlled by dorsoventral patterning signals. Consistent with this hypothesis, Ngn2 enhancer activity is dependent on the function of Pax6, a homeodomain factor involved in specifying the identity of ventral spinal cord progenitors. Moreover, we show that Ngn2 is required for the correct expression of Pax6 and several homeodomain proteins expressed in defined neuronal populations. Thus, neuronal differentiation involves crossregulatory interactions between a bHLH-driven program of neurogenesis and genetic pathways specifying progenitor and neuronal identity in the spinal cord.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Médula Espinal/citología , Médula Espinal/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Elementos de Facilitación Genéticos , Proteínas del Ojo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Mutantes , Ratones Transgénicos , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box , Proteínas Recombinantes de Fusión , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras , Células Madre/citología , beta-Galactosidasa/genética
10.
Development ; 127(11): 2323-32, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10804175

RESUMEN

We have characterised the functions of the bHLH transcriptional repressors HES1 and HES5 in neurogenesis, using the development of the olfactory placodes in mouse embryos as a model. Hes1 and Hes5 are expressed with distinct patterns in the olfactory placodes and are subject to different regulatory mechanisms. Hes1 is expressed in a broad placodal domain, which is maintained in absence of the neural determination gene Mash1. In contrast, expression of Hes5 is restricted to clusters of neural progenitor cells and requires Mash1 function. Mutations in Hes1 and Hes5 also have distinct consequences on olfactory placode neurogenesis. Loss of Hes1 function leads both to expression of Mash1 outside of the normal domain of neurogenesis and to increased density of MASH1-positive progenitors within this domain, and results in an excess of neurons after a delay. A mutation in Hes5 does not produce any apparent defect. However, olfactory placodes that are double mutant for Hes1 and Hes5 upregulate Ngn1, a neural bHLH gene activated downstream of Mash1, and show a strong and rapid increase in neuronal density. Together, our results suggest that Hes1 regulates Mash1 transcription in the olfactory placode in two different contexts, initially as a prepattern gene defining the placodal domain undergoing neurogenesis and, subsequently, as a neurogenic gene controlling the density of neural progenitors in this domain. Hes5 synergizes with Hes1 and regulates neurogenesis at the level of Ngn1 expression. Therefore, the olfactory sensory neuron lineage is regulated at several steps by negative signals acting through different Hes genes and targeting the expression of different proneural gene homologs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Secuencias Hélice-Asa-Hélice , Proteínas de Homeodominio/metabolismo , Neuronas Aferentes/citología , Mucosa Olfatoria/embriología , Proteínas Represoras/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Ratones , Mutagénesis , Mucosa Olfatoria/citología , Mucosa Olfatoria/metabolismo , Fenotipo , Proteínas Represoras/genética , Factor de Transcripción HES-1 , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
11.
Mol Cell Biol ; 20(12): 4445-54, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10825208

RESUMEN

Hepatocyte nuclear factor 6 (HNF-6) is the prototype of a new class of cut homeodomain transcription factors. During mouse development, HNF-6 is expressed in the epithelial cells that are precursors of the exocrine and endocrine pancreatic cells. We have investigated the role of HNF-6 in pancreas differentiation by inactivating its gene in the mouse. In hnf6(-/-) embryos, the exocrine pancreas appeared to be normal but endocrine cell differentiation was impaired. The expression of neurogenin 3 (Ngn-3), a transcription factor that is essential for determination of endocrine cell precursors, was almost abolished. Consistent with this, we demonstrated that HNF-6 binds to and stimulates the ngn3 gene promoter. At birth, only a few endocrine cells were found and the islets of Langerhans were missing. Later, the number of endocrine cells increased and islets appeared. However, the architecture of the islets was perturbed, and their beta cells were deficient in glucose transporter 2 expression. Adult hnf6(-/-) mice were diabetic. Taken together, our data demonstrate that HNF-6 controls pancreatic endocrine differentiation at the precursor stage and identify HNF-6 as the first positive regulator of the proendocrine gene ngn3 in the pancreas. They also suggest that HNF-6 is a candidate gene for diabetes mellitus in humans.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio/fisiología , Proteínas del Tejido Nervioso/fisiología , Páncreas/citología , Páncreas/fisiología , Transactivadores/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Factor Nuclear 6 del Hepatocito , Ratones , Ratones Noqueados
12.
Development ; 127(12): 2515-22, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10821751

RESUMEN

Neurons and glial cells differentiate from common precursors. Whereas the gene glial cells missing (gcm) determines the glial fate in Drosophila, current data about the expression patterns suggest that, in mammals, gcm homologues are unlikely to regulate gliogenesis. Here, we found that, in mouse retina, the bHLH gene Hes5 was specifically expressed by differentiating Müller glial cells and that misexpression of Hes5 with recombinant retrovirus significantly increased the population of glial cells at the expense of neurons. Conversely, Hes5-deficient retina showed 30-40% decrease of Müller glial cell number without affecting cell survival. These results indicate that Hes5 modulates glial cell fate specification in mouse retina.


Asunto(s)
Proteínas de Unión al ADN/genética , Neuroglía/citología , Neuronas/citología , Retina/citología , Factores de Transcripción/genética , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Genes Reporteros , Proteínas Fluorescentes Verdes , Secuencias Hélice-Asa-Hélice , Etiquetado Corte-Fin in Situ , Proteínas Luminiscentes/análisis , Ratones , Neuroglía/fisiología , Técnicas de Cultivo de Órganos , Proteínas Recombinantes/metabolismo , Retina/fisiología , Factores de Transcripción/fisiología , Transfección
13.
Proc Natl Acad Sci U S A ; 97(4): 1607-11, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10677506

RESUMEN

In the mammalian pancreas, the endocrine cell types of the islets of Langerhans, including the alpha-, beta-, delta-, and pancreatic polypeptide cells as well as the exocrine cells, derive from foregut endodermal progenitors. Recent genetic studies have identified a network of transcription factors, including Pdx1, Isl1, Pax4, Pax6, NeuroD, Nkx2.2, and Hlxb9, regulating the development of islet cells at different stages, but the molecular mechanisms controlling the specification of pancreatic endocrine precursors remain unknown. neurogenin3 (ngn3) is a member of a family of basic helix-loop-helix transcription factors that is involved in the determination of neural precursor cells in the neuroectoderm. ngn3 is expressed in discrete regions of the nervous system and in scattered cells in the embryonic pancreas. We show herein that ngn3-positive cells coexpress neither insulin nor glucagon, suggesting that ngn3 marks early precursors of pancreatic endocrine cells. Mice lacking ngn3 function fail to generate any pancreatic endocrine cells and die postnatally from diabetes. Expression of Isl1, Pax4, Pax6, and NeuroD is lost, and endocrine precursors are lacking in the mutant pancreatic epithelium. Thus, ngn3 is required for the specification of a common precursor for the four pancreatic endocrine cell types.


Asunto(s)
Islotes Pancreáticos/embriología , Proteínas del Tejido Nervioso/genética , Páncreas/embriología , Factores de Transcripción/metabolismo , Proteínas de Xenopus , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario y Fetal , Proteínas del Ojo , Glucagón/metabolismo , Secuencias Hélice-Asa-Hélice , Histocitoquímica , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Insulina/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box , ARN Mensajero/metabolismo , Proteínas Represoras , Somatostatina/metabolismo , Transactivadores/metabolismo
14.
EMBO J ; 18(8): 2196-207, 1999 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-10205173

RESUMEN

While the transmembrane protein Notch plays an important role in various aspects of development, and diseases including tumors and neurological disorders, the intracellular pathway of mammalian Notch remains very elusive. To understand the intracellular pathway of mammalian Notch, the role of the bHLH genes Hes1 and Hes5 (mammalian hairy and Enhancer-of-split homologues) was examined by retrovirally misexpressing the constitutively active form of Notch (caNotch) in neural precursor cells prepared from wild-type, Hes1-null, Hes5-null and Hes1-Hes5 double-null mouse embryos. We found that caNotch, which induced the endogenous Hes1 and Hes5 expression, inhibited neuronal differentiation in the wild-type, Hes1-null and Hes5-null background, but not in the Hes1-Hes5 double-null background. These results demonstrate that Hes1 and Hes5 are essential Notch effectors in regulation of mammalian neuronal differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Homeodominio , Proteínas de la Membrana/genética , Proteínas Musculares/fisiología , Neuronas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células Cultivadas , Regulación de la Expresión Génica/genética , Inmunohistoquímica , Ratones , Receptores Notch , Retroviridae/genética , Factor de Transcripción HES-1
15.
Neuron ; 20(3): 483-94, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9539123

RESUMEN

neurogenin2 encodes a neural-specific basic helix-loop-helix (bHLH) transcription factor related to the Drosophila proneural factor atonal. We show here that the murine ngn2 gene is essential for development of the epibranchial placode-derived cranial sensory ganglia. An ngn2 null mutation blocks the delamination of neuronal precursors from the placodes, the first morphological sign of differentiation in these lineages. Mutant placodal cells fail to express downstream bHLH differentiation factors and the Notch ligand Delta-like 1. These data suggest that ngn2 functions like the Drosophila proneural genes in the determination of neuronal fate in distal cranial ganglia. Interestingly, the homeobox gene Phox2a is activated independently of ngn2 in epibranchial placodes, suggesting that neuronal fate and neuronal subtype identity may be specified independently in cranial sensory ganglia.


Asunto(s)
Ganglios Sensoriales/embriología , Secuencias Hélice-Asa-Hélice/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas Aferentes/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular/fisiología , Linaje de la Célula/genética , Nervios Craneales/anomalías , Nervios Craneales/citología , Nervios Craneales/embriología , Femenino , Ganglios Sensoriales/anomalías , Ganglios Sensoriales/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Ratones , Ratones Mutantes , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Mutagénesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas Aferentes/química , Embarazo , Somitos/citología , Células Madre/química , Células Madre/fisiología , Factores de Transcripción/genética
16.
Development ; 124(22): 4557-69, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9409673

RESUMEN

Zebrafish neurogenin1 encodes a basic helix-loop-helix protein which shares structural and functional characteristics with proneural genes of Drosophila melanogaster. neurogenin1 is expressed in the early neural plate in domains comprising more cells than the primary neurons known to develop from these regions and its expression is modulated by Delta/Notch signalling, suggesting that it is a target of lateral inhibition. Misexpression of neurogenin1 in the embryo results in development of ectopic neurons. Markers for different neuronal subtypes are not ectopically expressed in the same patterns in neurogenin1-injected embryos suggesting that the final identity of the ectopically induced neurons is modulated by local cues. Induction of ectopic motor neurons by neurogeninl requires coexpression of a dominant negative regulatory subunit of protein kinase A, an intracellular transducer of hedgehog signals. Moreover, the pattern of endogenous neurogenin1 expression in the neural plate is expanded in response to elevated levels of Hedgehog (Hh) signalling or abolished as a result of inhibition of Hh signalling. Together these data suggest that Hh signals regulate neurogenin1 expression and subsequently modulate the type of neurons produced by Neurogenin1 activity.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Transactivadores , Factores de Transcripción , Proteínas de Pez Cebra , Pez Cebra/embriología , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Genes de Insecto , Proteínas Hedgehog , Secuencias Hélice-Asa-Hélice/genética , Hibridación in Situ , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/citología , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Xenopus/genética , Proteínas de Xenopus , Pez Cebra/genética
17.
Development ; 124(8): 1611-21, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9108377

RESUMEN

The lineage of olfactory neurons has been relatively well characterized at the cellular level, but the genes that regulate the proliferation and differentiation of their progenitors are currently unknown. In this study, we report the isolation of a novel murine gene, Math4C/neurogenin1, which is distantly related to the Drosophila proneural gene atonal. We show that Math4C/neurogenin1 and the basic helix-loop-helix gene Mash1 are expressed in the olfactory epithelium by different dividing progenitor populations, while another basic helix-loop-helix gene, NeuroD, is expressed at the onset of neuronal differentiation. These expression patterns suggest that each gene marks a distinct stage of olfactory neuron progenitor development, in the following sequence: Mash1>Math4C/neurogenin1>NeuroD. We have previously reported that inactivation of Mash1 function leads to a severe reduction in the number of olfactory neurons. We show here that most cells in the olfactory epithelium of Mash1 mutant embryos fail to express Math4C/neurogenin1 or NeuroD. Strikingly, a subset of progenitor cells in a ventrocaudal domain of Mash1 mutant olfactory epithelium still express Math4C/neurogenin1 and NeuroD and differentiate into neurons. Cells in this domain also express Math4A/neurogenin2, another member of the Math4/neurogenin gene family, and not Mash1. Our results demonstrate that Mash1 is required at an early stage in the olfactory neuron lineage to initiate a differentiation program involving Math4C/neurogenin1 and NeuroD. Another gene activates a similar program in a separate population of olfactory neuron progenitors.


Asunto(s)
Proteínas de Unión al ADN/genética , Secuencias Hélice-Asa-Hélice , Proteínas del Tejido Nervioso/genética , Mucosa Olfatoria/embriología , Factores de Transcripción/genética , Proteínas de Xenopus , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Encéfalo/embriología , Clonación Molecular , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Mucosa Olfatoria/inervación , ARN Mensajero/genética , Alineación de Secuencia
18.
Dev Biol ; 180(1): 227-41, 1996 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-8948587

RESUMEN

Tissue-specific bHLH proteins play important roles in the specification and differentiation of neural cell lineages in invertebrate and vertebrate organisms. Two groups of bHLH proteins, atonal and achaete-scute, have proneural activities in Drosophila, and the mouse achaete-scute homolog MASH1 is required for the differentiation of several neural lineages. In a screen for proteins interacting with MASH1, we have isolated a novel bHLH protein related to atonal, named MATH4A, which is broadly expressed in neural precursor cells in the mouse embryonic CNS and PNS. Interaction assays in yeast and in vitro demonstrate that MATH4A interacts efficiently with both MASH1 and the ubiquitous bHLH protein E12. MATH4A-E12 heterodimers, but not MATH4A-MASH1, bind to a consensus E-box sequence. Math4A expression is restricted to undifferentiated neural precursors and is complementary to that of Mash1 in most regions of the nervous system. In particular, Math4A is transcribed at high levels in the cerebral cortex, dorsal thalamus, and epibranchial placodes, which present little or no Mash1 expression. However, expression of the two genes shows limited overlap in certain CNS regions (retina, preoptic area of the hypothalamus, midbrain, hindbrain). Its structure and expression pattern suggest that MATH4A may regulate an early step of neural development, either as a partner of ubiquitous bHLH proteins or associated with other neural-specific bHLH proteins.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/metabolismo , Secuencias Hélice-Asa-Hélice , Proteínas del Tejido Nervioso/biosíntesis , Sistema Nervioso/embriología , Neuronas/fisiología , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Clonación Molecular , Proteínas de Unión al ADN/química , Drosophila/embriología , Proteínas de Drosophila , Embrión de Mamíferos , Embrión no Mamífero , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Fenómenos Fisiológicos del Sistema Nervioso , Biosíntesis de Proteínas , Proteínas Recombinantes/biosíntesis , Saccharomyces cerevisiae , Homología de Secuencia de Aminoácido , Transcripción Genética
19.
Dev Dyn ; 203(1): 80-92, 1995 May.
Artículo en Inglés | MEDLINE | ID: mdl-7647376

RESUMEN

We report the detailed developmental expression profiles of three endothelial specific receptor tyrosine kinases (RTKs) flk-1, tek, tie, as well as vascular endothelial growth factor (VEGF), the flk-1 ligand. We also examined the expression of the other VEGF receptor, flt-1, during placental development. flk-1, tek, and tie transcripts were detected sequentially at one-half day intervals starting at E7.0, suggesting that each of these RTKs play a unique role during vascularization of the mouse embryo. All three RTKs were expressed in the extraembryonic and embryonic mesoderm in regions that eventually give rise to the vasculature. Except for the expression of tek and flk-1 in the mesoderm of the amnion, the expression of these RTKs from E8.5 onwards was virtually indistinguishable. An abundant amount of flt-1 transcripts was found in the spongiotrophoblast cells of the developing placenta from E8.0 onwards. This cellular compartment is located between the maternal and labyrinthine layers of the placenta, which both express VEGF. VEGF transcripts were detected as early as E7.0 in the endoderm juxtaposed to the flk-1 positive mesoderm, and later in development VEGF expression displayed an expression profile both contiguous with that of flk-1, and also in tissues found some distance from the flk-1-expressing endothelium. These results suggest a possible dual role for VEGF which includes a chemotactic and/or a cellular maintenance role for VEGF during vascularization of the mouse embryo.


Asunto(s)
Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Factores de Crecimiento Endotelial/genética , Linfocinas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Animales , Secuencia de Bases , Sondas de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Hibridación in Situ , Masculino , Ratones , Datos de Secuencia Molecular , Placenta/metabolismo , Placentación , Embarazo , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores TIE , Receptores de Factores de Crecimiento Endotelial Vascular , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
Genes Dev ; 8(16): 1897-909, 1994 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-7958865

RESUMEN

The receptor tyrosine kinases (RTKs) expressed on the surface of endothelial cells are likely to play key roles in initiating the program of endothelial cell growth during development and subsequent vascularization during wound healing and tumorigenesis. Expression of the Tek RTK during mouse development is restricted primarily to endothelial cells and their progenitors, the angioblasts, suggesting that Tek is a key participant in vasculogenesis. To investigate the role that Tek plays within the endothelial cell lineage, we have disrupted the Tek signaling pathway using two different genetic approaches. First, we constructed transgenic mice expressing a dominant-negative form of the Tek receptor. Second, we created a null allele of the tek gene by homologous recombination in embryonic stem (ES) cells. Transgenic mice expressing dominant-negative alleles of Tek or homozygous for a null allele of the tek locus both died in utero with similar defects in the integrity of their endothelium. By crossing transgenic mice that express the lacZ reporter gene under the transcriptional control of the endothelial cell-specific tek promoter, we found that the extraembryonic and embryonic vasculature was patterned correctly. However, homozygous tek embryos had approximately 30% and 75% fewer endothelial cells at day 8.5 and 9.0, respectively. Homozygous null embryos also displayed abnormalities in heart development, consistent with the conclusion that Tek is necessary for endocardial/myocardial interactions during development. On the basis of the analysis of mice carrying either dominant-negative or null mutations of the tek gene, these observations demonstrate that the Tek signaling pathway plays a critical role in the differentiation, proliferation, and survival of endothelial cells in the mouse embryo.


Asunto(s)
Endotelio Vascular/embriología , Proteínas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Secuencia de Bases , Cartilla de ADN/genética , ADN Complementario/genética , Endotelio Vascular/anomalías , Endotelio Vascular/enzimología , Femenino , Genes Dominantes , Corazón/embriología , Hemorragia/genética , Homocigoto , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Embarazo , Proteínas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor TIE-2 , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...