Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Cell Tissue Res ; 371(2): 383-384, 2018 02.
Article in English | MEDLINE | ID: mdl-29127517

ABSTRACT

The published online version contains mistake. We apologize for errors in the lettering of Fig. 3d and also would like to correct the legend of Fig. 2b.

2.
Cell Tissue Res ; 370(2): 227-241, 2017 11.
Article in English | MEDLINE | ID: mdl-28936781

ABSTRACT

Neuron subtypes of the mature nervous system differ in the expression of characteristic marker genes while they share the expression of generic neuronal genes. The regulatory logic that maintains subtype-specific and pan-neuronal genes is not well understood. To begin to address this issue, we analyze RNA sequencing results from whole sympathetic ganglia and single sympathetic neurons in the mouse. We focus on gene products involved in the neuronal cytoskeleton, neurotransmitter synthesis and storage, transmitter release and reception and electrical information processing. We find a particular high correlation in the expression of stathmin 2 and several members of the tubulin beta family, classical pan-neuronal markers. Noradrenergic transmitter-synthesizing enzymes and transporters are also well correlated in their cellular transcript levels. In addition, noradrenergic marker transcript levels correlate well with selected pan-neuronal markers. Such a correlation in transcript levels is also seen between a number of selected ion channel, receptor and synaptic protein genes. These results provide the foundation for the analyses of the coordinated expression of downstream target genes in nerve cells.


Subject(s)
Ganglia, Sympathetic/cytology , Neurons/metabolism , Sympathetic Nervous System/cytology , Transcriptome , Animals , Ganglia, Sympathetic/metabolism , Ion Channels/genetics , Mice , Neurons/cytology , SNARE Proteins/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Stathmin/genetics , Sympathetic Nervous System/metabolism , Synaptotagmins/genetics , Tubulin/genetics , rab3 GTP-Binding Proteins/genetics
3.
Exp Neurol ; 223(2): 516-22, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20144891

ABSTRACT

Sympathetic neurons, like sensory neurons, increase neurite outgrowth after a conditioning lesion. Studies in leukemia inhibitory factor (LIF) knockout animals showed that the conditioning lesion effect in sensory neurons is dependent in part on this cytokine; however, similar studies on sympathetic neurons revealed no such effect. Comparable studies with sensory neurons taken from mice lacking the related cytokine interleukin-6 (IL-6) have yielded conflicting results. LIF and IL-6 belong to a family of cytokines known as the gp130 family because they act on receptors containing the subunit gp130. In sympathetic ganglia, axotomy leads to increases in mRNA for four of these cytokines (LIF, IL-6, IL-11, and oncostatin M). To test the role of this family of cytokines as a whole in the conditioning lesion response in sympathetic neurons, mice in which gp130 was selectively eliminated in noradrenergic neurons were studied. The postganglionic axons of the SCG were transected, and 7days later the ganglia were removed and neurite outgrowth was measured in explant and dissociated cell cultures. In both systems, neurons from wild type animals showed enhanced growth after a conditioning lesion. In contrast, no enhancement occurred in neurons from mutant animals. This lack of stimulation of outgrowth occurred despite an increase in expression of activating transcription factor 3 (ATF3) in the mutant mice. These studies demonstrate that stimulation of enhanced growth of sympathetic neurons after a conditioning lesion is dependent on gp130 cytokine signaling and is blocked in the absence of signaling by these cytokines in spite of an increase in ATF3.


Subject(s)
Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Neurites/physiology , Signal Transduction/physiology , Superior Cervical Ganglion/physiology , Activating Transcription Factor 3/metabolism , Animals , Axotomy , Cells, Cultured , Female , Interleukin-6/metabolism , Leukemia Inhibitory Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/physiology , STAT3 Transcription Factor/metabolism , Superior Cervical Ganglion/cytology
4.
Development ; 128(19): 3773-82, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11585803

ABSTRACT

During development, parasympathetic ciliary ganglion neurons arise from the neural crest and establish synaptic contacts on smooth and striate muscle in the eye. The factors that promote the ciliary ganglion pioneer axons to grow toward their targets have yet to be determined. Here, we show that glial cell line-derived neurotrophic factor (GDNF) and neurturin (NRTN) constitute target-derived factors for developing ciliary ganglion neurons. Both GDNF and NRTN are secreted from eye muscle located in the target and trajectory pathway of ciliary ganglion pioneer axons during the period of target innervation. After this period, however, the synthesis of GDNF declines markedly, while that of NRTN is maintained throughout the cell death period. Furthermore, both in vitro and in vivo function-blocking of GDNF at early embryonic ages almost entirely suppresses ciliary axon outgrowth. These results demonstrate that target-derived GDNF is necessary for ciliary ganglion neurons to innervate ciliary muscle in the eye. Since the down-regulation of GDNF in the eye is accompanied by down-regulation of GFRalpha1 and Ret, but not of GFRalpha2, in innervating ciliary ganglion neurons, the results also suggest that target-derived GDNF regulates the expression of its high-affinity coreceptors.


Subject(s)
Drosophila Proteins , Ganglia, Parasympathetic/embryology , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Parasympathetic Nervous System/embryology , Animals , Chick Embryo , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Contactins , Down-Regulation , Eye/embryology , Eye/metabolism , Ganglia, Parasympathetic/metabolism , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/metabolism , Neurturin , Parasympathetic Nervous System/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Ciliary Neurotrophic Factor/metabolism
5.
Phys Rev Lett ; 85(14): 3053-6, 2000 Oct 02.
Article in English | MEDLINE | ID: mdl-11006001

ABSTRACT

We show that, in a nanometric size stable electrodeposited Ni contact, it is possible to modify the magnetoresistance by applying current pulses and external magnetic fields whereby the same current path is used for detection and modification. We can pass from positive to negative magnetoresistance with values as large as 25% at room temperature, all in the same contact. We propose that the effect may be due to switching and moving domain walls in the contact region under the combination of current effects and external fields.


Subject(s)
Magnetics , Nanostructures/chemistry , Nickel/chemistry , Electrochemistry
6.
Development ; 127(18): 4073-81, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10952904

ABSTRACT

The dHAND basic helix-loop-helix transcription factor is expressed in neurons of sympathetic ganglia and has previously been shown to induce the differentiation of catecholaminergic neurons in avian neural crest cultures. We now demonstrate that dHAND expression is sufficient to elicit the generation of ectopic sympathetic neurons in vivo. The expression of the dHAND gene is controlled by bone morphogenetic proteins (BMPs), as suggested by BMP4 overexpression in vivo and in vitro, and by noggin-mediated inhibition of BMP function in vivo. The timing of dHAND expression in sympathetic ganglion primordia, together with the induction of dHAND expression in response to Phox2b implicate a role for dHAND as transcriptional regulator downstream of Phox2b in BMP-induced sympathetic neuron differentiation.


Subject(s)
Avian Proteins , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , DNA-Binding Proteins/metabolism , Ganglia, Sympathetic/cytology , Gene Expression Regulation, Developmental , Neurons/cytology , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Carrier Proteins , Catecholamines/physiology , Cells, Cultured , Chick Embryo , DNA-Binding Proteins/genetics , Epistasis, Genetic , Ganglia, Sympathetic/embryology , Genetic Vectors , Helix-Loop-Helix Motifs , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Microscopy, Fluorescence , Nerve Tissue Proteins , Neurons/metabolism , Proteins/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Retroviridae/genetics , Time Factors , Transcription Factors/genetics , Zebrafish Proteins
7.
Mech Dev ; 94(1-2): 25-36, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10842056

ABSTRACT

Fibroblast growth factor-8 (FGF-8) is an important signaling molecule in the generation and patterning of the midbrain, tooth, and limb. In this study we show that it is also involved in eye development. In the chick, Fgf-8 transcripts first appear in the distal optic vesicle when it contacts the head ectoderm. Subsequently Fgf-8 expression increases and becomes localized to the central area of the presumptive neural retina (NR) only. Application of FGF-8 has two main effects on the eye. First, it converts presumptive retinal pigment epithelium (RPE) into NR. This is apparent by the failure to express Bmp-7 and Mitf (a marker gene for the RPE) in the outer layer of the optic cup, coupled with the induction of NR genes, such as Rx, Sgx-1 and Fgf-8 itself. The induced retina displays the typical multilayered cytoarchitecture and expresses late neuronal differentiation markers such as synaptotagmin and islet-1. The second effect of FGF-8 exposure is the induction of both lens formation and lens fiber differentiation. This is apparent by the expression of a lens specific marker, L-Maf, and by morphological changes of lens cells. These results suggest that FGF-8 plays a role in the initiation and differentiation of neural retina and lens.


Subject(s)
Avian Proteins , Eye Proteins , Eye/embryology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/genetics , Chick Embryo , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Induction/genetics , Fibroblast Growth Factor 8 , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins , Lens, Crystalline/embryology , Maf Transcription Factors , Mesoderm , Microphthalmia-Associated Transcription Factor , Nerve Tissue Proteins/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors , Pigment Epithelium of Eye/embryology , Repressor Proteins , Retina/embryology , Transcription Factors/metabolism
8.
Eur J Neurosci ; 12(3): 994-1001, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10762330

ABSTRACT

Inhibitory glycine receptors (GlyRs) are known to mediate postsynaptic inhibition in spinal cord, brain stem and some higher brain regions. Several developmentally and regionally regulated GlyR isoforms exist, which result from a differential expression of the GlyR alpha (alpha1-alpha4) and beta subunit genes. Currently, very little is known about GlyRs containing the alpha4 subunit, whose existence was predicted from a partial genomic sequence. Here, we describe the isolation of complementary DNA (cDNA) sequences for the mouse and chick GlyR alpha4 subunits. We show that a mouse GlyR alpha4 subunit full-length cDNA directs the formation of functional homo-oligomeric strychnine-sensitive GlyRs in Xenopus laevis oocytes and mammalian cells, and that these resemble GlyRs composed of the alpha1 subunit in pharmacological profile and single-channel properties. In situ hybridization reveals high levels of GlyR alpha4 subunit transcripts in the embryonic (E13) chick spinal cord, lumbosacral sympathetic ganglia and dorsal root ganglia. The avian GlyR alpha4 subunit gene also shows male-specific expression in the developing genital ridge. The pharmacological profile of alpha4 subunit-containing receptors and deduced location of the avian GlyR alpha4 subunit are consistent with it being a component of the embryonic excitatory GlyRs previously identified in sympathetic neurons. Our data also suggest a novel role for GlyRs in the maturation of reproductive organs.


Subject(s)
Genitalia, Male/embryology , Genitalia, Male/metabolism , Receptors, Glycine/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Sympathetic Nervous System/embryology , Sympathetic Nervous System/metabolism , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Chick Embryo , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophysiology , Female , In Situ Hybridization , Male , Mice , Molecular Sequence Data , Oocytes/metabolism , Patch-Clamp Techniques , Pregnancy , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Xenopus laevis
9.
Mech Dev ; 92(2): 169-77, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10727856

ABSTRACT

During differentiation of sympathetic neurons in chick embryos, tyrosine hydroxylase (TH) and dopamine beta-hydroxylase (DBH) mRNAs become detectable during the same developmental period and are both induced by BMP 4. Later during sympathetic ganglion development, DBH is detectable in TH-positive and -negative cells. Moreover, BMPs reduce DBH mRNA in cultures of sympathetic neurons while leaving TH unaffected. The data provide evidence for a common regulation of TH and DBH early during sympathetic neuron differentiation and indicate that BMPs promote their initial expression but not the maintenance during later development. The time course of Phox2a and 2b expression suggests an evolutionary conserved role in noradrenergic induction. In addition, Phox2a, Phox2b, and c-ret may be involved in the differentiation of cholinergic sympathetic neurons.


Subject(s)
Dopamine beta-Hydroxylase/genetics , Drosophila Proteins , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/physiology , Sympathetic Nervous System/physiology , Transcription Factors/genetics , Transforming Growth Factor beta , Tyrosine 3-Monooxygenase/genetics , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Chick Embryo , Dopamine beta-Hydroxylase/drug effects , Embryonic Induction/drug effects , Embryonic Induction/genetics , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/drug effects , Nerve Tissue Proteins/drug effects , Neural Crest/drug effects , Neurons/drug effects , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Adrenergic/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/embryology , Transcription Factors/drug effects , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/metabolism
10.
Mech Dev ; 91(1-2): 91-6, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10704834

ABSTRACT

Sympathetic ganglia consist of noradrenergic and cholinergic neurons. The cholinergic marker protein vesicular acetylcholine transporter (VAChT) and the neuropeptide vasoactive intestinal peptide (VIP), co-expressed in mature cholinergic sympathetic neurons, are first detectable during embryonic development of rat sympathetic ganglia. However, the subpopulation of cholinergic sympathetic neurons which innervates sweat glands in mammalian footpads starts to express VAChT and VIP during the first postnatal weeks, under the influence of sweat gland-derived signals. In vitro evidence suggests that the sweat gland-derived cholinergic differentiation factor belongs to a group of neuropoietic cytokines, including LIF, CNTF and CT-1, that act through a LIFRbeta-containing cytokine receptor. To investigate whether the embryonic expression of cholinergic properties is elicited by a related cytokine, the expression of VAChT and VIP was analyzed in stellate ganglia of mice deficient for the cytokine receptor subunits LIFRbeta or CNTFRalpha. The density of VAChT- and VIP-immunoreactive cells in stellate ganglia of new-born animals was not different in LIFRbeta(-/-) and CNTFRalpha(-/-) ganglia as compared to ganglia from wild-type mice. These results demonstrate that the early, embryonic expression of VAChT and VIP is not induced by cytokines acting through LIFRbeta- or CNTFRalpha-containing receptors.


Subject(s)
Carrier Proteins/metabolism , Ciliary Neurotrophic Factor/metabolism , Growth Inhibitors/metabolism , Interleukin-6 , Lymphokines/metabolism , Membrane Transport Proteins , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, Cytokine/metabolism , Stellate Ganglion/metabolism , Vasoactive Intestinal Peptide/metabolism , Vesicular Transport Proteins , Animals , Leukemia Inhibitory Factor , Leukemia Inhibitory Factor Receptor alpha Subunit , Mice , Rabbits , Rats , Receptors, OSM-LIF , Vesicular Acetylcholine Transport Proteins
11.
AJR Am J Roentgenol ; 172(4): 1081-6, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10587151

ABSTRACT

OBJECTIVE: The aim of this study was to per form a three-dimensional analysis of the width of the subacromial space during passive and active arm abduction in healthy volunteers and patients with impingement syndrome. SUBJECTS AND METHODS: The shoulders of 10 healthy subjects and 10 patients with impingement syndrome were imaged with an open MR system during abduction, with and without activation of the shoulder muscles. An apparatus was designed for applying an adduction force of 10 N to the distal humerus during image acquisition, and the minimal acromiohumeral distance was measured after three-dimensional reconstruction. RESULTS: In the 10 healthy volunteers, muscle activity led to a significant decrease (-32%; p < .05) of the acromiohumeral distance at 60 degrees of abduction, whereas at 120 degrees of abduction the distance was significantly increased (+44%; p < .05). In these volunteers, muscle activation caused no significant effect at 90 degrees of abduction. However, in the 10 patients with impingement syndrome, muscle activity led to a significant decrease in the width of the subacromial space compared with that of the healthy contralateral side (-68%; p < .05). CONCLUSION: Muscle activity and arm position were found to cause systematic changes in the width of the subacromial space. However, functional deficits of the supraspinous muscle in patients with early-stage impingement syndrome were not apparent during muscle relaxation.


Subject(s)
Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Shoulder Impingement Syndrome/pathology , Shoulder Joint/pathology , Acromion/pathology , Adult , Clavicle/pathology , Female , Humans , Humerus/pathology , Male , Middle Aged , Movement , Shoulder Impingement Syndrome/physiopathology , Shoulder Joint/physiopathology
12.
Cell Tissue Res ; 297(3): 339-61, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10460483

ABSTRACT

Sympathetic ganglia are composed of noradrenergic neurons and cholinergic neurons that differ in the expression of neurotransmitter-synthesizing enzymes, neurotransmitter transporters and neuropeptides. The analysis of the cholinergic differentiation during development revealed important principles involved in the generation of neuronal diversity, in particular the importance of signals from the innervated target. Some peripheral targets, such as the sweat glands in the mammalian footpads, are purely cholinergically innervated in the adult, whereas skeletal muscle arteries receive both noradrenergic and cholinergic innervation. For sympathetic neurons innervating sweat glands there is convincing evidence that these neurons are initially noradrenergic and that the interaction of innervating fibers and target tissue induces a shift in the neurotransmitter phenotype from noradrenergic to cholinergic. In addition to this target-dependent differentiation, an earlier expression of cholinergic characters was observed in sympathetic ganglia that occurs before target contact. These data raise the possibility that different subpopulations of cholinergic sympathetic neurons, innervating distinct peripheral targets, may develop along distinct schedules. In vitro studies suggest that growth factors of the family of neuropoietic cytokines are involved in the specification of the cholinergic sympathetic phenotype. Recent in vivo studies that interfered with cytokine receptor expression in developing avian sympathetic ganglia indicate that only the late, target-dependent differentiation depends on cytokine signaling. The signals involved in the early, target-independent expression of cholinergic properties remain to be determined, as well as the identity of the target-derived cytokine. Thus, cholinergic sympathetic differentiation seems to be more complex than expected, involving either both target-independent and target-dependent control or only target-induced differentiation, according to the specific neuronal subpopulation and target.


Subject(s)
Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/physiology , Neurons/cytology , Neurons/physiology , Synaptic Transmission/physiology , Animals , Cell Differentiation/physiology , Choline , Humans
13.
Development ; 126(18): 4087-94, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10457017

ABSTRACT

The development of sympathetic neurons is controlled by a network of transcriptional regulators, including the paired homeodomain proteins Phox2a and Phox2b. To understand the role of Phox2 proteins in more detail, the effect of Phox2 overexpression was analysed in the avian peripheral nervous system. Phox2a expression in neural crest cultures elicited a strong increase in the number of sympathoadrenergic cells. Expression of Phox2a in the chick embryo promoted the generation of additional neurons expressing the noradrenergic marker genes DBH and TH, pan-neuronal genes SCG10 and NF160 and cholinergic genes ChAT and VAChT. Phox2a-induced neurons were found in ectopic locations such as dorsal root ganglia and peripheral nerve. Sympathoadrenergic development could be elicited in cultures of E5 dorsal root ganglia, demonstrating the presence of Phox2a-responsive cells in non-autonomic peripheral ganglia. Phox2b induced ectopic neurons in the chick embryo in the same way as Phox2a. These results show that Phox2 proteins are sufficient to promote sympathetic neuron generation and control, directly or indirectly, the expression of a large number of genes characteristic for sympathetic neurons.


Subject(s)
Drosophila Proteins , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Membrane Transport Proteins , Neurons/metabolism , Sympathetic Nervous System/embryology , Transcription Factors/genetics , Transcription Factors/metabolism , Vesicular Transport Proteins , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , Chick Embryo , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Culture Techniques , Dopamine beta-Hydroxylase/genetics , Dopamine beta-Hydroxylase/metabolism , Embryo, Nonmammalian/virology , Embryonic Induction/genetics , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons, Afferent/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Quail/embryology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Adrenergic/metabolism , Retroviridae/genetics , Sympathetic Nervous System/cytology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Vesicular Acetylcholine Transport Proteins
14.
J Neurosci ; 19(3): 1062-71, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9920669

ABSTRACT

We have studied the effect of retinoic acid on the expression of the neurotrophin receptors trkA, trkC, and p75 by neuroblasts and neurons at different axial levels along the embryonic mouse paravertebral sympathetic chain. In dissociated cultures of sympathetic neuroblasts, retinoic acid inhibited the developmental increase in trkA mRNA expression and the developmental decrease in trkC mRNA expression that normally occurs in these cells but did not affect p75 mRNA expression. At higher concentrations, retinoic acid also increased the proliferation of sympathetic neuroblasts. After sympathetic neuroblasts became postmitotic, retinoic acid no longer affected receptor expression. Studies with retinoic acid receptor agonists and antagonists indicated that the effects of retinoic acid on neurotrophin receptor expression were mediated mainly by alpha retinoic acid receptors, not beta or gamma receptors. The observation that alpha-antagonists increased trkA mRNA expression in intact sympathetic ganglion explants suggests that endogenous retinoic acid is a physiological regulator of trkA receptor expression.


Subject(s)
Ganglia, Sympathetic/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism , Stem Cells/metabolism , Tretinoin/pharmacology , Animals , Cell Division/drug effects , Cellular Senescence/physiology , Ganglia, Sympathetic/cytology , Mice/embryology , Mice, Inbred Strains , Neurons/cytology , Neurons/drug effects , Neurons/physiology , RNA, Messenger/metabolism , Receptor, trkA/genetics , Receptors, Retinoic Acid/physiology , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology
15.
Neuron ; 24(4): 861-70, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10624949

ABSTRACT

Bone morphogenetic proteins (BMPs) induce autonomic neurogenesis in neural crest cultures and stimulate sympathetic neuron development when overexpressed in vivo. We demonstrate that inhibition of BMPs in the chick embryo bythe BMP antagonist Noggin prevents sympathetic neuron generation. In Noggin-treated embryos, the noradrenergic marker genes tyrosine hydroxylase (TH) and dopamine-beta-hydroxylase (DBH), panneuronal neurofilament 160 (NF160) and SCG10 genes, and the transcriptional regulators Phox2b and Phox2a are not expressed in sympathetic ganglia. Whereas initial ganglion development is not affected, the expression of the basic helix-loop-helix transcription factor Cash-1 is strongly reduced. These results demonstrate that BMPs are essential for sympathetic neuron development and establish Cash-1 and Phox2 genes as downstream effectors of BMPs in this lineage.


Subject(s)
Avian Proteins , Bone Morphogenetic Proteins/physiology , Neurons/physiology , Sympathetic Nervous System/physiology , Transforming Growth Factor beta , Animals , Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins , Cell Differentiation/physiology , Cells, Cultured , Chick Embryo , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Ganglia/cytology , Ganglia/physiology , High Mobility Group Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neural Crest/cytology , Neural Crest/physiology , Norepinephrine/physiology , Proteins/pharmacology , SOXE Transcription Factors , Sympathetic Nervous System/cytology , Transcription Factors/genetics , Transcription Factors/physiology , Transcription, Genetic
16.
J Neurobiol ; 37(4): 672-83, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9858267

ABSTRACT

Previous studies suggest that ciliary neurotrophic factor (CNTF) may represent one of the extrinsic signals controlling the development of vertebrate retinal photoreceptors. In dissociated cultures from embryonic chick retina, exogenously applied CNTF has been shown to act on postmitotic rod precursor cells, resulting in an two- to fourfold increase in the number of cells acquiring an opsin-positive phenotype. We now demonstrate that the responsiveness of photoreceptor precursors to CNTF is confined to a brief phase between their final mitosis and their terminal differentiation owing to the temporally restricted expression of the CNTF receptor (CNTFR alpha). As shown immunocytochemically, CNTFR alpha expression in the presumptive photoreceptor layer of the chick retina starts at embryonic day 8 (E8) and is rapidly down-regulated a few days later prior to the differentiation of opsin-positive photoreceptors, both in vivo and in dissociated cultures from E8. We further show that the CNTF-dependent in vitro differentiation of rods is followed by a phase of photoreceptor-specific apoptotic cell death. The loss of differentiated rods during this apoptotic phase can be prevented by micromolar concentrations of retinol. Our results provide evidence that photoreceptor development depends on the sequential action of different extrinsic signals. The time course of CNTFR alpha expression and the in vitro effects suggest that CNTF or a related molecule is required during early stages of rod differentiation, while differentiated rods depend on additional protective factors for survival.


Subject(s)
Nerve Tissue Proteins/pharmacology , Photoreceptor Cells, Vertebrate/cytology , Retinal Rod Photoreceptor Cells/cytology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Ascorbic Acid/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Chick Embryo , Ciliary Neurotrophic Factor , Immunohistochemistry , In Situ Nick-End Labeling , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Ciliary Neurotrophic Factor , Receptors, Nerve Growth Factor/metabolism , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/embryology , Retinal Rod Photoreceptor Cells/metabolism , Rod Opsins/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Time Factors , Vitamin A/pharmacology , Vitamin E/pharmacology
17.
Development ; 125(23): 4791-801, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9806927

ABSTRACT

Sympathetic ganglia are composed of noradrenergic and cholinergic neurons. The differentiation of cholinergic sympathetic neurons is characterized by the expression of choline acetyltransferase (ChAT) and vasoactive intestinal peptide (VIP), induced in vitro by a subfamily of cytokines, including LIF, CNTF, GPA, OSM and cardiotrophin-1 (CT-1). To interfere with the function of these neuropoietic cytokines in vivo, antisense RNA for gp130, the common signal-transducing receptor subunit for neuropoietic cytokines, was expressed in chick sympathetic neurons, using retroviral vectors. A strong reduction in the number of VIP-expressing cells, but not of cells expressing ChAT or the adrenergic marker tyrosine hydroxylase (TH), was observed. These results reveal a physiological role of neuropoietic cytokines for the control of VIP expression during the development of cholinergic sympathetic neurons.


Subject(s)
Antigens, CD/physiology , Cytokines/physiology , Ganglia, Sympathetic/embryology , Gene Expression Regulation, Developmental , Membrane Glycoproteins/physiology , Neurons/physiology , Receptors, Cytokine/physiology , Vasoactive Intestinal Peptide/genetics , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Cells, Cultured , Chick Embryo , Choline O-Acetyltransferase/genetics , Cytokine Receptor gp130 , Ganglia, Sympathetic/cytology , Gene Expression Regulation , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neurons/classification , Neurons/cytology , Phenotype , RNA, Antisense , Recombinant Proteins/biosynthesis , Retroviridae , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Transfection , Tyrosine 3-Monooxygenase/genetics
18.
J Comp Neurol ; 400(2): 244-54, 1998 Oct 19.
Article in English | MEDLINE | ID: mdl-9766402

ABSTRACT

Ciliary neurotrophic factor (CNTF) exerts a multiplicity of effects on a broad spectrum of target cells, including retinal neurons. To investigate how this functional complexity relates to the regulation of CNTF receptor alpha (CNTFR alpha) expression, we have studied the developmental expression of the receptor protein in chick retina by using immunocytochemistry. During the course of development, the receptor is expressed in all retinal layers, but three levels of specificity can be observed. First, the expression is regulated temporally with immunoreactivity observed in ganglion cells (embryonic day 8 [E8] to adult), photoreceptor precursors (E8-E12), amacrine cells (E10 to adult), bipolar cells (E12-E18), differentiated rods (E18 to adult), and horizontal cells (adult). Second, expression is restricted to distinct subpopulations of principal retinal neurons: preferentially, large ganglion cells; subpopulations of amacrine cells, including a particular type of cholinergic neuron; a distinctly located type of bipolar cell; and rod photoreceptors. Third, expression exhibits subcellular restriction: it is confined largely to dendrites in mature amacrine cells and is restricted entirely to outer segments in mature rods. These data correlate with CNTF effects on the survival of ganglion cells and mature photoreceptors, the in vitro differentiation of photoreceptor precursors and cholinergic amacrine cells, and the number of bipolar cells in culture described here or in previous studies. Thus, our results demonstrate an exceptional degree of complexity with respect to the regulation of neuronal CNTFR alpha expression in a defined model system. This suggests that the same signaling pathway is used to mediate a variety of regulatory influences, depending on the developmental stage and cell type.


Subject(s)
Chick Embryo/growth & development , Neurons/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Retina/metabolism , Animals , Cells, Cultured , Choline O-Acetyltransferase/analysis , Immunoblotting , Immunohistochemistry , Neurons/classification , Receptor, Ciliary Neurotrophic Factor , Retina/embryology
19.
J Neurobiol ; 36(3): 379-94, 1998 Sep 05.
Article in English | MEDLINE | ID: mdl-9733073

ABSTRACT

During development of the avian neuromuscular system, lumbar spinal motoneurons (MNs) innervate their muscle targets in the hindlimb coincident with the onset and progression of MN programmed cell death (PCD). Paralysis (activity blockade) of embryos during this period rescues large numbers of MNs from PCD. Because activity blockade also results in enhanced axonal branching and increased numbers of neuromuscular synapses, it has been postulated that following activity blockade, increased numbers of MNs can gain access to muscle-derived trophic agents that prevent PCD. An assumption of the access hypothesis of MN PCD is the presence of an activity-dependent, muscle-derived sprouting or branching agent. Several previous studies of sprouting in the rodent neuromuscular system indicate that insulin-like growth factors (IGFs) are candidates for such a sprouting factor. Accordingly, in the present study we have begun to test whether the IGFs may play a similar role in the developing avian neuromuscular system. Evidence in support of this idea includes the following: (a) IGFs promote MN survival in vivo but not in vitro; (b) neutralizing antibodies against IGFs reduce MN survival in vivo; (c) both in vitro and in vivo, IGFs increase neurite growth, branching, and synapse formation; (d) activity blockade increases the expression of IGF-1 and IGF-2 mRNA in skeletal muscles in vivo; (e) in vivo treatment of paralyzed embryos with IGF binding proteins (IGF-BPs) that interfere with the actions of endogenous IGFs reduce MN survival, axon branching, and synapse formation; (f) treatment of control embryos in vivo with IGF-BPs also reduces synapse formation; and (g) treatment with IGF-1 prior to the major period of cell death (i.e., on embryonic day 6) increases subsequent synapse formation and MN survival and potentiates the survival-promoting actions of brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) administered during the subsequent 4- to 5-day period of PCD. Collectively, these data provide new evidence consistent with the role of the IGFs as activity-dependent, muscle-derived agents that play a role in regulating MN survival in the avian embryo.


Subject(s)
Apoptosis/physiology , Motor Neurons/physiology , Muscle, Skeletal/embryology , Muscle, Skeletal/innervation , Somatomedins/physiology , Animals , Cell Count , Cells, Cultured , Chick Embryo , Hindlimb/innervation , Immunohistochemistry , Insulin-Like Growth Factor Binding Proteins/pharmacology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor II/genetics , Oligonucleotides/pharmacology , Presynaptic Terminals/physiology , Reverse Transcriptase Polymerase Chain Reaction , Somatomedins/genetics , Synapses/physiology
20.
Blood ; 92(1): 59-67, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9639500

ABSTRACT

The loss of body cell mass (bcm) in senescence and wasting is poorly understood. We now show that the plasma cystine/acid soluble thiol ratio, ie, an indicator of the redox state, is increased in old age and cancer patients and correlated with a decrease in bcm and plasma albumin. A cause/effect relationship was suggested by two independent studies with N-acetyl-cysteine (NAC). NAC caused an increase in the bcm of healthy persons with high plasma cystine/thiol ratios, and treatment of cancer patients with NAC plus interleukin-2 caused an increase in bcm, plasma albumin, and functional capacity. Albumin levels below 680 micromol/L were associated with an increase in body water. Our studies suggest that the shift in the redox state may contribute to the loss of bcm and may provide a quantitative guideline for therapeutic intervention. Treatment of cancer patients with thiol-containing antioxidants may improve the quality of life.


Subject(s)
Acetylcysteine/administration & dosage , Aging/metabolism , Free Radical Scavengers/administration & dosage , Neoplasms/metabolism , Oxidation-Reduction , Adolescent , Adult , Aged , Aged, 80 and over , Cell Death , Child , Child, Preschool , Cystine/blood , Female , Free Radical Scavengers/therapeutic use , Humans , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/physiopathology , Oxidation-Reduction/drug effects , Serum Albumin/metabolism , Sulfhydryl Compounds/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...