Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
Genesis ; 58(5): e23356, 2020 05.
Article in English | MEDLINE | ID: mdl-32049434

ABSTRACT

Vertebrates possess paired cranial sensory ganglia derived from two embryonic cell populations, neural crest and placodes. Cranial sensory ganglia arose prior to the divergence of jawed and jawless vertebrates, but the developmental mechanisms that facilitated their evolution are unknown. Using gene expression and cell lineage tracing experiments in embryos of the sea lamprey, Petromyzon marinus, we find that in the cranial ganglia we targeted, development consists of placode-derived neuron clusters in the core of ganglia, with neural crest cells mostly surrounding these neuronal clusters. To dissect functional roles of neural crest and placode cell associations in these developing cranial ganglia, we used CRISPR/Cas9 gene editing experiments to target genes critical for the development of each population. Genetic ablation of SoxE2 and FoxD-A in neural crest cells resulted in differentiated cranial sensory neurons with abnormal morphologies, whereas deletion of DlxB in cranial placodes resulted in near-total loss of cranial sensory neurons. Taken together, our cell-lineage, gene expression, and gene editing results suggest that cranial neural crest cells may not be required for cranial ganglia specification but are essential for shaping the morphology of these sensory structures. We propose that the association of neural crest and placodes in the head of early vertebrates was a key step in the organization of neurons and glia into paired sensory ganglia.


Subject(s)
Ganglia, Sensory/growth & development , Lampreys/growth & development , Neural Crest/growth & development , Neurogenesis , Animals , Fish Proteins/genetics , Fish Proteins/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Ganglia, Sensory/cytology , Ganglia, Sensory/metabolism , Lampreys/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , Skull/growth & development
2.
Exp Brain Res ; 238(1): 111-119, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31802149

ABSTRACT

After exiting the hindbrain, branchial motor axons reach their targets in association with sensory ganglia. The trigeminal ganglion has been shown to promote motor axon growth from rhombomeres 2/3 and 4/5, but it is unknown whether this effect is ganglion specific and through which signals it is mediated. Here, we addressed these questions by co-cultures of ventral rhombomere 8 explants with cranial and spinal sensory ganglia in a collagen gel matrix. Our results show that all cranial sensory ganglia and even a trunk dorsal root ganglion can promote motor axon growth and that ganglia isolated from older embryos had a stronger effect on the axonal growth than younger ones. We found that brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) are necessary and sufficient for this effect. Altogether, our results demonstrate that the promoting effect of sensory ganglia on cranial motor axon growth is stage dependent, but not ganglion specific and is mediated by BDNF and NGF signals.


Subject(s)
Axons/physiology , Brain-Derived Neurotrophic Factor/physiology , Cranial Nerves/growth & development , Ganglia, Sensory/growth & development , Motor Neurons/physiology , Nerve Growth Factor/physiology , Animals , Chick Embryo , Ganglia, Spinal/growth & development
3.
Neuroscience ; 284: 632-642, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25450961

ABSTRACT

The primary vestibular neurons convey afferent information from hair cells in the inner ear to the vestibular nuclei and the cerebellum. The intrinsic firing properties of vestibular ganglion cells (VGCs) are heterogeneous to sustained membrane depolarization, and undergo marked developmental changes from phasic to tonic types during the early postnatal period. Previous studies have shown that low-voltage-activated potassium channels, Kv1 and Kv7, play a critical role in determining the firing pattern of VGCs. In the present study, we explored the developmental changes in the properties of hyperpolarization-activated current (Ih) in rat VGCs and the role played by Ih in determining the firing properties of VGCs. Tonic firing VGCs showed a larger current density of Ih as compared to phasic firing VGCs, and tonic firing VGCs became phasic firing in the presence of ZD7288, an Ih channel blocker, indicating that Ih contributes to control the firing pattern of VGCs. The amplitude of Ih increased and the activation kinetics of Ih became faster during the developmental period. Analysis of developmental changes in the expression of hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels revealed that expression of HCN1 protein and its mRNA increased during the developmental period, whereas expression of HCN2-4 protein and its mRNA did not change. Our results suggest that HCN1 channels as well as Kv1 channels are critical in determining the firing pattern of rat VGCs and that developmental up-regulation of HCN1 transforms VGCs from phasic to tonic firing phenotypes.


Subject(s)
Action Potentials/physiology , Ganglia, Sensory/growth & development , Neurons/physiology , Vestibular Nerve/growth & development , Action Potentials/drug effects , Animals , Blotting, Western , Cells, Cultured , Ganglia, Sensory/drug effects , Ganglia, Sensory/physiology , Gene Expression Regulation, Developmental , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Potassium Channels/metabolism , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Rats, Wistar , Real-Time Polymerase Chain Reaction , Vestibular Nerve/drug effects , Vestibular Nerve/physiology
4.
Biomed Res Int ; 2014: 704259, 2014.
Article in English | MEDLINE | ID: mdl-24738070

ABSTRACT

Previous studies indicated that intracerebroventricular administration of nerve growth factor (NGF) leads to massive Schwann cell hyperplasia surrounding the medulla oblongata and spinal cord. This study was designed to characterize the proliferation of peripheral glial cells, that is, Schwann and satellite cells, in the trigeminal ganglia and dorsal root ganglia (DRG) of adult rats during two weeks of NGF infusion using bromodeoxyuridine (BrdU) to label dividing cells. The trigeminal ganglia as well as the cervical and lumbar DRG were analyzed. Along the entire neuraxis a small number of dividing cells were observed within these regions under physiological condition. NGF infusion has dramatically increased the generation of new cells in the neuronal soma and axonal compartments of sensory ganglia and along the dorsal root and the dorsal root entry zone. Quantification of BrdU positive cells within sensory ganglia revealed a 2.3- to 3-fold increase in glial cells compared to controls with a similar response to NGF for the different peripheral ganglia examined. Immunofluorescent labeling with S100ß revealed that Schwann and satellite cells underwent mitosis after NGF administration. These data indicate that intracerebroventricular NGF infusion significantly induces gliogenesis in trigeminal ganglia and the spinal sensory ganglia and along the dorsal root entry zone as well as the dorsal root.


Subject(s)
Ganglia, Sensory/growth & development , Nerve Growth Factor/administration & dosage , Neuroglia/physiology , Spinal Nerve Roots/growth & development , Animals , Axons/drug effects , Ganglia, Sensory/drug effects , Infusions, Intraventricular , Neuroglia/drug effects , Rats , Spinal Cord/drug effects , Spinal Cord/growth & development , Spinal Nerve Roots/drug effects
5.
Biochem Biophys Res Commun ; 447(2): 299-303, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24709080

ABSTRACT

The neural system appears before the vascular system in the phylogenetic tree. During evolution, vascular system generation takes advantage of the pre-existing vascular endothelial growth factor (VEGF) in order to form its networks. Nevertheless, the role of VEGF in neuronal and glial cells is not yet completely understood. In order to support the hypothesis of a neural role for VEGF, we searched for VEGF- and VEGF receptor (VEGFR)-like immunoreactivities (immunohisto/cytochemistry and Western blotting) in the eyestalk of the invertebrate Ucides cordatus (Crustacea, Brachyura, Ucididae). Our results showed that both neurons and glial cells expressed VEGF-immunoreactivity, and that VEGFR was evidenced in neural cells. This is the first report about the VEGF/VEGFR-like immunoreactivities in the nervous tissue of a crustacean, and enables U. cordatus to be included in the repertoire of animal models used for ascertaining the role of VEGF in the nervous system.


Subject(s)
Brachyura/growth & development , Ganglia, Sensory/growth & development , Neurogenesis , Neurons/physiology , Receptors, Vascular Endothelial Growth Factor/physiology , Vascular Endothelial Growth Factor A/physiology , Visual Pathways/growth & development , Animals , Brachyura/cytology , Male , Neurons/cytology , Visual Pathways/cytology
6.
Biochim Biophys Acta ; 1842(6): 848-59, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24269586

ABSTRACT

The dysfunction of the ubiquitin proteasome system has been related to a broad array of neurodegenerative disorders in which the accumulation of misfolded protein aggregates causes proteotoxicity. The ability of proteasome inhibitors to induce cell cycle arrest and apoptosis has emerged as a powerful strategy for cancer therapy. Bortezomib is a proteasome inhibitor used as an antineoplastic drug, although its neurotoxicity frequently causes a severe sensory peripheral neuropathy. In this study we used a rat model of bortezomib treatment to study the nucleolar and Cajal body responses to the proteasome inhibition in sensory ganglion neurons that are major targets of bortezomib-induced neurotoxicity. Treatment with bortezomib induced dose-dependent dissociation of protein synthesis machinery (chromatolysis) and nuclear retention of poly(A) RNA granules resulting in neuronal dysfunction. However, as a compensatory response to the proteotoxic stress, both nucleoli and Cajal bodies exhibited reactive changes. These include an increase in the number and size of nucleoli, strong nucleolar incorporation of the RNA precursor 5'-fluorouridine, and increased expression of both 45S rRNA and genes encoding nucleolar proteins UBF, fibrillarin and B23. Taken together, these findings appear to reflect the activation of the nucleolar transcription in response to proteotoxic stress Furthermore, the number of Cajal bodies, a parameter related to transcriptional activity, increases upon proteasome inhibition. We propose that nucleoli and Cajal bodies are important targets in the signaling pathways that are activated by the proteotoxic stress response to proteasome inhibition. The coordinating activity of these two organelles in the production of snRNA, snoRNA and rRNA may contribute to neuronal survival after proteasome inhibition. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.


Subject(s)
Coiled Bodies/metabolism , Ganglia, Sensory/metabolism , Proteasome Endopeptidase Complex/metabolism , Sensory Receptor Cells/metabolism , Animals , Boronic Acids/administration & dosage , Bortezomib , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Nucleolus/metabolism , Cell Nucleus , Cytoplasm/metabolism , Ganglia, Sensory/growth & development , Humans , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Pyrazines/administration & dosage , Rats , Signal Transduction/drug effects
7.
Neuroscience ; 256: 271-81, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24161722

ABSTRACT

Neurochemical features in sympathetic and afferent neurons are subject to change during development. Nitric oxide (NO) plays a developmental role in the nervous system. To better understand the neuroplasticity of sympathetic and afferent neurons during postnatal ontogenesis, the distribution of neuronal NO synthase (nNOS) immunoreactivity was studied in the sympathetic para- and prevertebral, nodose ganglion (NG) and Th2 and L4 dorsal root ganglia (DRG) from female Wistar rats of different ages (newborn, 10-day-old, 20-day-old, 30-day-old, 2-month-old, 6-month-old, 1-year-old, and 3-year-old). nNOS-positive neurons were revealed in all sensory ganglia but not in sympathetic ones from birth onward. The percentage of nNOS-immunoreactive (IR) neurons increased during first 10 days of life from 41.3 to 57.6 in Th2 DRG, from 40.9 to 59.1 in L4 DRG and from 31.6 to 38.5 in NG. The percentage of nNOS-IR neurons did not change in the NG later during development and senescence. However, in Th2 and L4 DRG the proportion of nNOS-IR neurons was high in animals between 10 and 30days of life and decreased up to the second month of life. In 2-month-old rats, the percentage of nNOS-IR neurons was 52.9 in Th2 DRG and 51.3 in L4 DRG. We did not find statistically significant differences in the percentage of nNOS-IR neurons between Th2 and L4 DRG and between young and aged rats. In NG and DRG of 10-day-old and older rats, a high proportion of nNOS-IR neurons binds isolectin B4. In newborn animals, only 41.3%, 45.3% and 28.4% of nNOS neuron profiles bind to IB4 in Th2, L4 DRG and NG, respectively. In 10-day-old and older rats, the number of sensory nNOS-IR neurons binding IB4 reached more than 90% in DRG and more than 80% in NG. Only a small number of nNOS-positive cells showed immunoreactivity to calcitonin gene-related peptide, neurofilament 200, calretinin. The information provided here will also serve as a basis for future studies investigating mechanisms of the development of sensory neurons.


Subject(s)
Ganglia, Sensory/cytology , Ganglia, Sensory/growth & development , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/growth & development , Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Age Factors , Animals , Animals, Newborn , Female , Lectins/metabolism , Nerve Tissue Proteins/metabolism , Rats , Rats, Wistar
8.
Morfologiia ; 141(1): 75-7, 2012.
Article in Russian | MEDLINE | ID: mdl-22724340

ABSTRACT

Afferent neurons containing substance P (SP) were studied immunohistochemically in the sensory ganglion of the spinal nerve in 30 rats aged 10-90 days. The results obtained indicated that SP-immunoreactive neurons are present in thesel ganglia from the moment of birth. During the development, the percentage of SP-containing neurons decreased till day 10. SP-immunoreactive neurons were represented by the cells of very small or small size.


Subject(s)
Neurons, Afferent/metabolism , Spinal Nerves/growth & development , Substance P/metabolism , Animals , Female , Ganglia, Sensory/growth & development , Ganglia, Sensory/metabolism , Neurons, Afferent/cytology , Rats , Rats, Wistar , Spinal Nerves/metabolism , Substance P/analysis
9.
Neuroreport ; 21(10): 731-5, 2010 Jul 14.
Article in English | MEDLINE | ID: mdl-20531234

ABSTRACT

Phosphatase and tensin homolog (PTEN) is a tumor suppressor gene that controls neural stem cell renewal and differentiation and is a potential target for regeneration in the optic nerve. Here we show that it has a critical pattern of expression in the mammalian developing auditory system. PTEN was expressed in the cochlear-vestibular ganglion at embryonic day 10.5 and then progressively in hair cells as they differentiated from the base to the apex of the cochlea. By postnatal day 7, PTEN was downregulated in hair cells and subsequently in the neurons. This very specific, transient expression pattern suggests that PTEN plays a crucial role in the differentiation of the sensory neurons and hair cells and that it is a potential therapeutic target for hearing regeneration.


Subject(s)
Cochlea/growth & development , Ganglia, Sensory/growth & development , Hair Cells, Auditory/metabolism , PTEN Phosphohydrolase/metabolism , Sensory Receptor Cells/metabolism , Spiral Ganglion/growth & development , Animals , Auditory Pathways/embryology , Auditory Pathways/growth & development , Auditory Pathways/metabolism , Cochlea/embryology , Cochlea/metabolism , Epithelium/embryology , Epithelium/growth & development , Epithelium/metabolism , Ganglia, Sensory/embryology , Ganglia, Sensory/metabolism , Mice , Mice, Inbred ICR , Spiral Ganglion/embryology , Spiral Ganglion/metabolism
10.
J Insect Physiol ; 55(7): 599-607, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19482133

ABSTRACT

Stress-induced arrest of ventilatory motor pattern generation is tightly correlated with an abrupt increase in extracellular potassium concentration ([K+]o) within the metathoracic neuropil of the locust, Locusta migratoria. Na+/K+-ATPase inhibition with ouabain elicits repetitive surges of [K+]o that coincide with arrest and recovery of motor activity. Here we show that ouabain induces repetitive [K+]o events in a concentration-dependent manner. 10(-5)M, 10(-4)M, and 10(-3)M ouabain was bath-applied in semi-intact locust preparations. 10(-4)M and 10(-3)M ouabain reliably induced repetitive [K+]o events whereas 10(-5)M ouabain had no significant effect. In comparison to 10(-4)M ouabain, 10(-3)M ouabain increased the number and hastened the time to onset of repetitive [K+]o waves, prolonged [K+]o event duration, increased resting [K+]o, and diminished the absolute value of [K+]o waves. Recovery of motor patterning following [K+]o events was less likely in 10(-3)M ouabain. In addition, we show that K+ channel inhibition using TEA suppressed the onset and decreased the amplitude of ouabain-induced repetitive [K+]o waves. Our results demonstrate that ventilatory circuit function in the locust CNS is dependent on the balance between mechanisms of [K+] accumulation and [K+] clearance. We suggest that with an imbalance in favour of accumulation the system tends towards a bistable state with transitions mediated by positive feedback involving voltage-dependent K+ channels.


Subject(s)
Locusta migratoria/growth & development , Locusta migratoria/metabolism , Potassium/metabolism , Animals , Ganglia, Sensory/drug effects , Ganglia, Sensory/growth & development , Ganglia, Sensory/metabolism , Insect Proteins/metabolism , Locusta migratoria/drug effects , Locusta migratoria/enzymology , Male , Neuropil/drug effects , Neuropil/metabolism , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism
11.
Eur J Neurosci ; 27(10): 2629-42, 2008 May.
Article in English | MEDLINE | ID: mdl-18513316

ABSTRACT

Recent evidence indicates that endomorphins, endogenous mu-opioid receptor (MOR) agonists, modulate synaptic transmission in both somatic and visceral sensory pathways. Here we show that endomorphin-2 (END-2) is expressed in newborn rat dorsal root ganglion (DRG) and nodose-petrosal ganglion complex (NPG) neurons, and rarely co-localizes with brain-derived neurotrophic factor (BDNF). In order to examine activity-dependent release of END-2 from neurons, we established a model using dispersed cultures of DRG and NPG cells activated by patterned electrical field stimulation. To detect release of END-2, we developed a novel rapid capture enzyme-linked immunosorbent assay (ELISA), in which END-2 capture antibody was added to neuronal cultures shortly before their electrical stimulation. The conventional assay was effective at reliably detecting END-2 only when the cells were stimulated in the presence of CTAP, a MOR-selective antagonist. This suggests that the strength of the novel assay is related primarily to rapid capture of released END-2 before it binds to endogenous MORs. Using the rapid capture ELISA, we found that stimulation protocols known to induce plastic changes at sensory synapses were highly effective at releasing END-2. Removal of extracellular calcium or blocking voltage-activated calcium channels significantly reduced the release. Together, our data provide the first evidence that END-2 is expressed by newborn DRG neurons of all sizes found in this age group, and can be released from these, as well as from NPG neurons, in an activity-dependent manner. These results point to END-2 as a likely mediator of activity-dependent plasticity in sensory pathways.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Ganglia, Sensory/growth & development , Ganglia, Sensory/metabolism , Neurons, Afferent/metabolism , Oligopeptides/metabolism , Action Potentials/drug effects , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Calcium Signaling/drug effects , Cell Differentiation/physiology , Cells, Cultured , Electric Stimulation , Enzyme-Linked Immunosorbent Assay , Ganglia, Sensory/cytology , Ganglia, Spinal/cytology , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Neuronal Plasticity/physiology , Neurons, Afferent/cytology , Nodose Ganglion/cytology , Nodose Ganglion/growth & development , Nodose Ganglion/metabolism , Oligopeptides/analysis , Rats , Rats, Sprague-Dawley , Synapses/metabolism , Synaptic Transmission/physiology
12.
J Struct Biol ; 159(3): 451-61, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17587596

ABSTRACT

This paper studies the molecular organization, neuronal distribution and cellular differentiation dynamics of the giant fibrillar centers (GFCs) of nucleoli in rat sensory ganglia neurons. The GFC appeared as a round nucleolar domain (1-2 microm in diameter) partially surrounded by the dense fibrillar component and accompanied by numerous small FCs. By immunocytochemistry, the GFC concentrated the upstream binding factor, which may serve as a marker of this structure, and also contain RNA polymerase I, DNA topoisomerase I, SUMO-1 and Ubc9. However, they lack ubiquitin-proteasome conjugates and 20S proteasome. Transcription assay with 5'-fluorouridine incorporation revealed the presence of nascent RNA on the dense fibrillar component of the neuronal nucleolus, but not within the low electron-density area of the GFC. The formation of GFCs is neuronal size dependent: they were found in 58%, 30% and 0% of the large, medium and small neurons, respectively. GFCs first appeared during the postnatal period, concomitantly with a stage of neuronal growth, myelination and bioelectrical maturation. GFCs were not observed in segregated nucleoli induced by severe inhibition of RNA synthesis. We suggest that the formation of GFCs is associated with a high rate of ribosome biogenesis of the transcriptionally more active large-size neurons.


Subject(s)
Cell Nucleolus/ultrastructure , Ganglia, Sensory/growth & development , Ganglia, Sensory/ultrastructure , Neurons, Afferent/ultrastructure , Pol1 Transcription Initiation Complex Proteins/analysis , Animals , Cell Differentiation , Cell Nucleolus/chemistry , Ganglia, Sensory/metabolism , Male , Microscopy, Immunoelectron , Neurons, Afferent/metabolism , Proteasome Endopeptidase Complex/analysis , Proteasome Endopeptidase Complex/metabolism , RNA, Ribosomal/analysis , RNA, Ribosomal/metabolism , Rats , Rats, Sprague-Dawley , SUMO-1 Protein/analysis , SUMO-1 Protein/metabolism , Transcription, Genetic , Ubiquitin/analysis , Ubiquitin/metabolism
13.
Eur J Neurosci ; 24(6): 1581-90, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17004922

ABSTRACT

To study the role of different members of the bHLH gene family for sensory organ development we have generated NSCL-1 and NeuroD compound-mutant mice. Double homozygous animals were characterized by a more severe reduction of the petrosal and vestibulocochlear ganglia than NeuroD-knockout mice. The more severe reduction of the petrosal and vestibulocochlear ganglia in double-knockout mice indicates overlapping functions of the two genes during neuronal development. Interestingly, we also found that the two genes are jointly regulated by thyroid hormone during sensory hair cell development. We further present a detailed expression analysis of NSCL-1 and NSCL-2 during sensory neuron development. NSCL-1 expression was detected in all developing cranial ganglia including the petrosal and vestibulocochlear ganglion, in inner and outer hair cells of the organ of Corti and in hair cells of the vestibular system. Expression domains in other sensory structures include the retina, Merkel cells of the developing skin and sensory cells of the tongue. The expression of NSCL-2 was restricted to developing cranial ganglia, the retina and the vestibular nerve. Both NSCL-1 and NSCL-2 genes are active only in postmitotic neurons, indicating a role for neuronal cell migration and/or differentiation within the sensory system.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Ganglia, Sensory , Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/deficiency , Bromodeoxyuridine/metabolism , Cerebellum/embryology , Cerebellum/growth & development , Cerebellum/metabolism , Ear, Inner/embryology , Ear, Inner/growth & development , Ear, Inner/metabolism , Embryo, Mammalian , Female , Ganglia, Sensory/embryology , Ganglia, Sensory/growth & development , Ganglia, Sensory/metabolism , Hypothyroidism/etiology , Immunohistochemistry/methods , Mice , Mice, Knockout , Molecular Motor Proteins , Nerve Tissue Proteins/deficiency , Pregnancy , Proteins/metabolism , Skin/embryology , Skin/growth & development , Skin/metabolism , Tongue/embryology , Tongue/growth & development , Tongue/metabolism
14.
Pediatr Res ; 60(4): 371-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16940233

ABSTRACT

Hyperoxia, during development in rats, results in hypoxic chemosensitivity ablation, carotid body hypoplasia, and reduced chemoafferents. We hypothesized that hyperoxia increases reactive oxygen species (ROS) in cell bodies of chemoafferents. Organotypic slices of petrosal-nodose ganglia from rats at day of life (DOL) 5-6 and 17-18 were exposed to 8%, 21%, or 95% O(2) for 4 h in the presence or absence of the ROS-sensitive fluorescent indicator, CM-H(2)DCFDA, and propidium iodide was used to determine the relationship between cell death and oxygen tension. In tissue slices from DOL 5-6 rats, fluorescence intensity was 182.5 +/- 2.9 for hypoxia, 217.5 +/- 3.3 for normoxia, and 336.6 +/- 3.8 for hyperoxia, (mean +/- SEM, p < 0.001, ANOVA). Normoxia increased ROS levels by 19.2% from hypoxia (p < 0.01) with a further increase of 54.8% from normoxia to hyperoxia (p < 0.001). In tissue slices from DOL 17-18 rats, ROS levels increased with increasing oxygen tension but were less than in younger animals (p < 0.01, ANOVA). The antioxidants, NAC and TEMPO-9-AC, attenuated ROS levels and cell death. Electron microscopy demonstrated that hyperoxia damages the ultrastructure within petrosal ganglion neurons. Hyperoxic-induced increased levels of ROS in petrosal ganglion neurons may contribute to loss of hypoxic chemosensitivity during early postnatal development.


Subject(s)
Ganglia, Sensory/growth & development , Ganglia, Sensory/metabolism , Hyperoxia/metabolism , Neurons, Afferent/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Cell Culture Techniques , Cyclic N-Oxides/pharmacology , Dissection , Female , Fluoresceins , Ganglia, Sensory/ultrastructure , Hyperoxia/pathology , Neurons, Afferent/ultrastructure , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/analysis
15.
J Comp Neurol ; 497(4): 658-69, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16739199

ABSTRACT

Early dietary sodium restriction has profound influences on the organization of the gustatory brainstem. However, the anatomical relationships among multiple gustatory nerve inputs have not been examined. Through the use of triple-fluorescence labeling and confocal laser microscopy, terminal fields of the greater superficial petrosal (GSP), chorda tympani (CT), and glossopharyngeal (IX) nerves were visualized concurrently in the nucleus of the solitary tract (NTS) of developmentally sodium-restricted and control rats. Dietary sodium restriction during pre- and postnatal development resulted in a twofold increase in the volume of both the CT and the IX nerve terminal fields but did not affect the volume of the GSP terminal field. In controls, these nerve terminal fields overlapped considerably. The dietary manipulation significantly increased the overlapping zones among terminal fields, resulting in an extension of CT and IX fields past their normal boundaries. The differences in terminal field volumes were exaggerated when expressed relative to the respective NTS volumes. Furthermore, increased terminal field volumes could not be attributed to an increase in the number of afferents because ganglion cell counts did not differ between groups. Taken together, selective increases in terminal field volume and ensuing overlap among terminal fields suggest an increased convergence of these gustatory nerve terminals onto neurons in the NTS. The genesis of such convergence is likely related to disruption of cellular and molecular mechanisms during the development of individual terminal fields, the consequences of which have implications for corresponding functional and behavioral alterations.


Subject(s)
Neuronal Plasticity/physiology , Neurons, Afferent/physiology , Presynaptic Terminals/physiology , Solitary Nucleus/growth & development , Taste Buds/growth & development , Visceral Afferents/growth & development , Animals , Animals, Newborn , Chorda Tympani Nerve/cytology , Chorda Tympani Nerve/growth & development , Facial Nerve/cytology , Facial Nerve/growth & development , Female , Food, Formulated , Ganglia, Sensory/cytology , Ganglia, Sensory/growth & development , Glossopharyngeal Nerve/cytology , Glossopharyngeal Nerve/growth & development , Neurons, Afferent/cytology , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Sodium/deficiency , Sodium, Dietary/metabolism , Solitary Nucleus/cytology , Taste/physiology , Taste Buds/cytology , Visceral Afferents/cytology
16.
J Neurochem ; 96(6): 1708-17, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16539686

ABSTRACT

Inhibition of the proteasome by lactacystin, a specific blocker of the catalytic beta-subunits, results in transient neurite outgrowth by neuronal cell lines. Vice versa, as demonstrated in this study, treatment of pheochromocytoma (PC12) cells with nerve growth factor (NGF) or other differentiating agents reduces proteasomal activity. This is accompanied by an increase in mRNA and protein levels of the catalytically active subunits beta1, beta2 and beta5, but not of their inducible counterparts, indicating changes in subunit composition of the proteasome during neuronal differentiation. In contrast to neuronal cell lines, however, pre-treatment of primary neurons with proteasome inhibitors completely prevents axon formation, and lower concentrations of lactacystin (0.5-5 microm) significantly reduce axonal elongation and branching in vitro. Furthermore, established axonal networks degenerate rapidly and long-term survival of peripheral neurons is impaired in the presence of proteasome inhibitors. Axonal pathology is reminiscent of the morphological changes observed in neurodegenerative disorders and supports a crucial role of the constitutive catalytic subunits in axon initiation, maintenance and regeneration.


Subject(s)
Catalytic Domain/physiology , Cell Differentiation/physiology , Growth Cones/metabolism , Nervous System/growth & development , Nervous System/metabolism , Proteasome Endopeptidase Complex/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Animals, Newborn , Catalytic Domain/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Ganglia, Autonomic/drug effects , Ganglia, Autonomic/growth & development , Ganglia, Autonomic/metabolism , Ganglia, Sensory/drug effects , Ganglia, Sensory/growth & development , Ganglia, Sensory/metabolism , Growth Cones/drug effects , Growth Cones/ultrastructure , Microscopy, Electron, Scanning , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Nerve Net/drug effects , Nerve Net/growth & development , Nerve Net/metabolism , Nervous System/drug effects , PC12 Cells , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Eur J Neurosci ; 22(10): 2649-54, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16307607

ABSTRACT

First described in 1973, the Grueneberg ganglion (GG) is an arrow-shaped neuronal structure at the anterior end of the nasal cavity. It lines both sides of the nasal septum, within the nasal vestibule, close to the opening of the naris. The functions of the GG and the pattern of projections to the brain are not known. Here, we report that neurons of the mouse GG express olfactory marker protein, which is normally expressed in mature olfactory or vomeronasal sensory neurons. The approx. 500 cells in each GG are arranged in several densely packed cell clusters. Individual cells give rise to single axons, which fasciculate to form a nerve bundle that projects caudally. The axons terminate in glomeruli of the olfactory bulb, one or two large glomeruli associated with a semicircle of up to 10 smaller, somewhat diffusely organized glomeruli that surround the most anterior part of the accessory olfactory bulb. Development of the GG starts around embryonic day 16 and appears to be completed at birth; cell numbers then undergo a minor decrease during postnatal development. The strategic location of the GG, expression of olfactory marker protein, axonal projections to glomeruli at particular locations in the olfactory bulb and early development suggest that this neuronal structure performs specific chemosensory functions at neonatal stages.


Subject(s)
Axons/physiology , Ganglia, Sensory/physiology , Olfactory Bulb/physiology , Olfactory Pathways/physiology , Animals , Carbocyanines , Cell Count , Chemoreceptor Cells/physiology , Female , Ganglia, Sensory/cytology , Ganglia, Sensory/growth & development , Genes, Reporter , Lac Operon , Mice , Olfactory Bulb/cytology , Olfactory Marker Protein/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/growth & development , Pregnancy
18.
Dev Dyn ; 234(3): 791-801, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16217737

ABSTRACT

The formation of the eight independent endorgan compartments (sacculus, utricle, horizontal canal, anterior canal, posterior canal, lagena, amphibian papilla, and basilar papilla) of the Xenopus laevis inner ear is illustrated as the otic vesicle develops into a complex labyrinthine structure. The morphology of transverse sections and whole-mounts of the inner ear was assessed in seven developmental stages (28, 31, 37, 42, 45, 47, 50) using brightfield and laser scanning confocal microscopy. The presence of mechanosensory hair cells in the sensory epithelia was determined by identification of stereociliary bundles in cryosectioned tissue and whole-mounts of the inner ear labeled with the fluorescent F-actin probe Alexa-488 phalloidin. Between stages 28 and 45, the otic vesicle grows in size, stereociliary bundles appear and increase in number, and the pars inferior and pars superior become visible. The initial formation of vestibular compartments with their nascent stereociliary bundles is seen by larval stage 47, and all eight vestibular and auditory compartments with their characteristic sensory fields are present by larval stage 50. Thus, in Xenopus, inner ear compartments are established between stages 45 and 50, a 2-week period during which the ear quadruples in length in the anteroposterior dimension. The anatomical images presented here demonstrate the morphological changes that occur as the otic vesicle forms the auditory and vestibular endorgans of the inner ear. These images provide a resource for investigations of gene expression patterns in Xenopus during inner ear compartmentalization and morphogenesis.


Subject(s)
Ear, Inner/growth & development , Larva/growth & development , Xenopus laevis/growth & development , Actins/metabolism , Animals , Ganglia, Sensory/growth & development , Ganglia, Sensory/metabolism , Gene Expression Regulation, Developmental , Larva/metabolism , Xenopus laevis/metabolism
19.
Int J Dev Neurosci ; 23(7): 575-86, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16143489

ABSTRACT

NGF, a neurotrophic factor best known for its role in promoting cell survival, regulates many neurodevelopmental processes, including synaptic plasticity, neurite outgrowth and programmed cell death. Although there is a large amount of data regarding NGF in the developing nervous system of many species, there is little known about its regulation and role in the frog, Xenopus laevis. In this report, immunocytochemistry was used to characterize NGF protein expression in developing tadpoles. Protein expression was analyzed in tadpoles from stage 44/45 through stage 50, a period of development characterized by extensive neurite outgrowth, neuronal differentiation and an initial period of programmed cell death. Similar to other species, NGF was expressed in sensory cells and tissues, including the inner ear, eye, olfactory system, lateral line organs, papillae in the oral cavity, and gills tufts. In addition, NGF was expressed in specific cells in the central nervous system, cranial and dorsal root ganglia, spinal sensory and motoneurons, and muscle tissues in the tail and body cavity. In the mammalian nervous system, the cytokine, interleukin-1beta (IL-1beta) induces expression of NGF. In this report, double-label immunocytochemistry was used to determine the relationship between NGF and IL-1beta. Results showed most cell types and/or tissues that expressed NGF also expressed IL-1beta. However, NGF was typically associated with cellular and nuclear membranes, whereas IL-1beta appeared in the cytoplasm and nucleolus. The nuclear localization of IL-1beta supports the idea that it regulates gene transcription in the frog. The appearance of NGF and IL-1beta in the same cells suggests they may interact to influence neural development.


Subject(s)
Interleukin-1/metabolism , Nerve Growth Factor/metabolism , Nervous System/growth & development , Nervous System/metabolism , Animals , Blotting, Western , Ganglia, Sensory/growth & development , Ganglia, Sensory/metabolism , Immunohistochemistry , Larva , Muscles/metabolism , Nerve Fibers/metabolism , Nervous System/anatomy & histology , Sense Organs/growth & development , Sense Organs/metabolism , Xenopus laevis , Zygote/metabolism
20.
Auton Neurosci ; 120(1-2): 68-72, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15935738

ABSTRACT

Retrograde axonal transport of horseradish peroxidase was used in this study to determine the location and basic morphological parameters of neurons innervating the trachea in newborn, 10-, 20-, 30-day-old and 2-month-old kittens. Labeled neurons were detected in all animals in the nodose ganglion of the vagus nerve and in the spinal ganglia (C1-C7 and T1-T6 after injection of tracer into the cervical trachea, C5-C7 and T1-T8 with injection into the thoracic part of the trachea) from both sides. The content of vagal and spinal afferent neurons innervating the cervical part of trachea declined during development. The number of spinal afferent neurons with connections to the thoracic trachea did not change but the quantity of cells in nodose ganglion supplying the thoracic trachea increased from the moment of birth till 10 and 20 days and decreased later in postnatal development. In newborn, 10-day-old and 20-day-old animals, the largest number of afferent cells was connected with the cervical part of the trachea in comparison with the thoracic one, whereas in 2-month-old kittens the relation was opposite. We suggest that afferent innervation of the trachea is not morphologically complete at the moment of birth and does not become mature until the second month of life.


Subject(s)
Ganglia, Sensory/cytology , Ganglia, Sensory/growth & development , Neurons, Afferent/physiology , Trachea/innervation , Animals , Animals, Newborn , Cats , Ganglia, Spinal/cytology , Ganglia, Spinal/growth & development , Nodose Ganglion/cytology , Nodose Ganglion/growth & development , Trachea/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL