Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
J Perinatol ; 37(1): 77-80, 2017 01.
Article En | MEDLINE | ID: mdl-27684424

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.


Aortic Coarctation/diagnosis , Aortic Coarctation/physiopathology , Photoplethysmography , Blood Pressure , Case-Control Studies , Female , Heart Rate , Humans , Infant, Newborn , Male , Neonatal Screening , Oximetry , Proof of Concept Study
2.
Cell Death Dis ; 7(6): e2272, 2016 06 23.
Article En | MEDLINE | ID: mdl-27336716

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.


Cell Cycle , Cytoprotection , DNA Damage , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , STAT3 Transcription Factor/metabolism , Animals , Cell Cycle/drug effects , Cytokines/metabolism , Cytoprotection/drug effects , Insulin-Secreting Cells/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-6/pharmacology , Ki-67 Antigen/metabolism , Ligation , Male , Mice, Inbred BALB C , Mice, Knockout , Pancreatic Ducts/drug effects , Pancreatic Ducts/pathology , Recombinant Proteins/pharmacology
3.
Neural Comput ; 25(1): 75-100, 2013 Jan.
Article En | MEDLINE | ID: mdl-22970870

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.


Computer Simulation , Excitatory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/physiology , Models, Neurological , Motor Neurons/physiology , Neural Inhibition/physiology , Animals , Cats , Dendrites/physiology , Glutamic Acid/physiology , Glycine/physiology , Potassium Channels/physiology , Rats , Sodium Channels/physiology , Synapses/physiology
4.
Diabetologia ; 55(1): 154-65, 2012 Jan.
Article En | MEDLINE | ID: mdl-21947380

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.


Antigens, Surface/metabolism , Cell Separation/methods , Islets of Langerhans/metabolism , Stem Cells/metabolism , Adult , Animals , Biomarkers/metabolism , Cell Line , Computational Biology/methods , Data Mining , Flow Cytometry , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Mice , Mice, Inbred BALB C , Organ Culture Techniques , Stem Cells/cytology
5.
Neural Comput ; 22(7): 1764-85, 2010 Jul.
Article En | MEDLINE | ID: mdl-20235819

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).


Computer Simulation , Inhibitory Postsynaptic Potentials/physiology , Ion Channels/physiology , Motor Neurons/physiology , Neural Inhibition/physiology , Spinal Cord/physiology , Animals , Cell Compartmentation/physiology , Cell Membrane/physiology , Dendrites/physiology , Humans , Motor Neurons/classification , Neural Conduction/physiology , Neural Pathways/physiology , Synaptic Transmission/physiology
6.
Cell Biol Toxicol ; 23(4): 241-56, 2007 Jul.
Article En | MEDLINE | ID: mdl-17171431

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.


Animal Testing Alternatives , Cell Culture Techniques/methods , Cell Differentiation/physiology , Intestinal Mucosa/cytology , Models, Biological , Stem Cells/cytology , Animals , Cell Line, Transformed , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Intestinal Mucosa/physiology , Stem Cells/physiology
7.
Diabetes Metab ; 32(5 Pt 2): 532-3, 2006 Dec.
Article En | MEDLINE | ID: mdl-17130813

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.


Islets of Langerhans/growth & development , Humans , Transcription Factors/metabolism
8.
Proc Natl Acad Sci U S A ; 102(5): 1490-5, 2005 Feb 01.
Article En | MEDLINE | ID: mdl-15668393

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.


DNA-Binding Proteins/physiology , Pancreas/abnormalities , Pancreas/embryology , Transcription Factors/physiology , Animals , Apoptosis , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryonic Development , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-beta , Heterozygote , In Situ Hybridization , Mice , Mice, Knockout , Morphogenesis , Polyploidy , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic
9.
Neuron ; 31(2): 203-17, 2001 Aug 02.
Article En | MEDLINE | ID: mdl-11502253

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.


Cell Differentiation , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Neurons/cytology , Spinal Cord/cytology , Spinal Cord/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors , Enhancer Elements, Genetic , Eye Proteins , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Mutant Strains , Mice, Transgenic , PAX6 Transcription Factor , Paired Box Transcription Factors , Recombinant Fusion Proteins , Regulatory Sequences, Nucleic Acid , Repressor Proteins , Stem Cells/cytology , beta-Galactosidase/genetics
10.
Mol Cell Biol ; 20(12): 4445-54, 2000 Jun.
Article En | MEDLINE | ID: mdl-10825208

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.


Gene Expression Regulation/physiology , Homeodomain Proteins/physiology , Nerve Tissue Proteins/physiology , Pancreas/cytology , Pancreas/physiology , Trans-Activators/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Hepatocyte Nuclear Factor 6 , Mice , Mice, Knockout
11.
Development ; 127(12): 2515-22, 2000 Jun.
Article En | MEDLINE | ID: mdl-10821751

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.


DNA-Binding Proteins/genetics , Neuroglia/cytology , Neurons/cytology , Retina/cytology , Transcription Factors/genetics , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/physiology , Genes, Reporter , Green Fluorescent Proteins , Helix-Loop-Helix Motifs , In Situ Nick-End Labeling , Luminescent Proteins/analysis , Mice , Neuroglia/physiology , Organ Culture Techniques , Recombinant Proteins/metabolism , Retina/physiology , Transcription Factors/physiology , Transfection
12.
Development ; 127(11): 2323-32, 2000 Jun.
Article En | MEDLINE | ID: mdl-10804175

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.


DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Helix-Loop-Helix Motifs , Homeodomain Proteins/metabolism , Neurons, Afferent/cytology , Olfactory Mucosa/embryology , Repressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Mice , Mutagenesis , Olfactory Mucosa/cytology , Olfactory Mucosa/metabolism , Phenotype , Repressor Proteins/genetics , Transcription Factor HES-1 , Transcription Factors/biosynthesis , Transcription Factors/genetics
13.
Proc Natl Acad Sci U S A ; 97(4): 1607-11, 2000 Feb 15.
Article En | MEDLINE | ID: mdl-10677506

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.


Islets of Langerhans/embryology , Nerve Tissue Proteins/genetics , Pancreas/embryology , Transcription Factors/metabolism , Xenopus Proteins , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , DNA-Binding Proteins/metabolism , Embryonic and Fetal Development , Eye Proteins , Glucagon/metabolism , Helix-Loop-Helix Motifs , Histocytochemistry , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , In Situ Hybridization , In Situ Nick-End Labeling , Insulin/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors , RNA, Messenger/metabolism , Repressor Proteins , Somatostatin/metabolism , Trans-Activators/metabolism
14.
EMBO J ; 18(8): 2196-207, 1999 Apr 15.
Article En | MEDLINE | ID: mdl-10205173

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.


Cell Differentiation/physiology , Homeodomain Proteins , Membrane Proteins/genetics , Muscle Proteins/physiology , Neurons/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cells, Cultured , Gene Expression Regulation/genetics , Immunohistochemistry , Mice , Receptors, Notch , Retroviridae/genetics , Transcription Factor HES-1
15.
Neuron ; 20(3): 483-94, 1998 Mar.
Article En | MEDLINE | ID: mdl-9539123

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.


Ganglia, Sensory/embryology , Helix-Loop-Helix Motifs/physiology , Nerve Tissue Proteins/genetics , Neurons, Afferent/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/physiology , Cell Lineage/genetics , Cranial Nerves/abnormalities , Cranial Nerves/cytology , Cranial Nerves/embryology , Female , Ganglia, Sensory/abnormalities , Ganglia, Sensory/cytology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Mice , Mice, Mutant Strains , Motor Neurons/cytology , Motor Neurons/physiology , Mutagenesis/physiology , Nerve Tissue Proteins/metabolism , Neurons, Afferent/chemistry , Pregnancy , Somites/cytology , Stem Cells/chemistry , Stem Cells/physiology , Transcription Factors/genetics
16.
Development ; 124(22): 4557-69, 1997 Nov.
Article En | MEDLINE | ID: mdl-9409673

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.


Nerve Tissue Proteins/metabolism , Trans-Activators , Transcription Factors , Zebrafish Proteins , Zebrafish/embryology , Zebrafish/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Genes, Insect , Hedgehog Proteins , Helix-Loop-Helix Motifs/genetics , In Situ Hybridization , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nervous System/cytology , Nervous System/embryology , Nervous System/metabolism , Neurons/cytology , Neurons/metabolism , Proteins/genetics , Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Xenopus/genetics , Xenopus Proteins , Zebrafish/genetics
17.
Development ; 124(8): 1611-21, 1997 Apr.
Article En | MEDLINE | ID: mdl-9108377

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.


DNA-Binding Proteins/genetics , Helix-Loop-Helix Motifs , Nerve Tissue Proteins/genetics , Olfactory Mucosa/embryology , Transcription Factors/genetics , Xenopus Proteins , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Brain/embryology , Cloning, Molecular , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , Mice, Mutant Strains , Molecular Sequence Data , Olfactory Mucosa/innervation , RNA, Messenger/genetics , Sequence Alignment
18.
Dev Biol ; 180(1): 227-41, 1996 Nov 25.
Article En | MEDLINE | ID: mdl-8948587

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.


DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Helix-Loop-Helix Motifs , Nerve Tissue Proteins/biosynthesis , Nervous System/embryology , Neurons/physiology , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Cloning, Molecular , DNA-Binding Proteins/chemistry , Drosophila/embryology , Drosophila Proteins , Embryo, Mammalian , Embryo, Nonmammalian , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nervous System Physiological Phenomena , Protein Biosynthesis , Recombinant Proteins/biosynthesis , Saccharomyces cerevisiae , Sequence Homology, Amino Acid , Transcription, Genetic
19.
Dev Dyn ; 203(1): 80-92, 1995 May.
Article En | MEDLINE | ID: mdl-7647376

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.


Blood Vessels/embryology , Blood Vessels/metabolism , Endothelial Growth Factors/genetics , Lymphokines/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Animals , Base Sequence , DNA Probes/genetics , Female , Gene Expression Regulation, Developmental , Gestational Age , In Situ Hybridization , Male , Mice , Molecular Sequence Data , Placenta/metabolism , Placentation , Pregnancy , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, TIE , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Genes Dev ; 8(16): 1897-909, 1994 Aug 15.
Article En | MEDLINE | ID: mdl-7958865

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.


Endothelium, Vascular/embryology , Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Base Sequence , DNA Primers/genetics , DNA, Complementary/genetics , Endothelium, Vascular/abnormalities , Endothelium, Vascular/enzymology , Female , Genes, Dominant , Heart/embryology , Hemorrhage/genetics , Homozygote , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Pregnancy , Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptor, TIE-2 , Signal Transduction/genetics
...