Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 26(4): 661-673, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28007910

ABSTRACT

GM2 gangliosidoses are a group of lysosomal storage disorders which include Sandhoff disease and Tay-Sachs disease. Dysregulation of glutamate receptors has been recently postulated in the pathology of Sandhoff disease. Glutamate receptor association with neuronal pentraxins 1 and 2, and the neuronal pentraxin receptor facilitates receptor potentiation and synaptic shaping. In this study, we have observed an upregulation of a novel form of neuronal pentraxin 1 (NP1-38) in the brains of a mouse model of Sandhoff disease and Tay-Sachs disease. In order to determine the impact of NP1 on the pathophysiology of Sandhoff disease mouse models, we have generated an Np1-/-Hexb-/- double knockout mouse, and observed extended lifespan, improved righting reflex and enhanced body condition relative to Hexb-/- mice, with no effect on gliosis or apoptotic markers in the CNS. Sandhoff mouse brain slices reveals a reduction in AMPA receptor-mediated currents, and increased variability in total glutamate currents in the CA1 region of the hippocampus; Np1-/-Hexb-/- mice show a correction of this phenotype, suggesting NP1-38 may be interfering with glutamate receptor function. Indeed, some of the psychiatric aspects of Sandhoff and Tay-Sachs disease (particularly late onset) may be attributed to a dysfunctional hippocampal glutamatergic system. Our work highlights a potential role for synaptic proteins, such as NP1 and glutamate receptors in lysosomal storage diseases.


Subject(s)
C-Reactive Protein/biosynthesis , CA1 Region, Hippocampal/metabolism , Nerve Tissue Proteins/biosynthesis , Sandhoff Disease/metabolism , Up-Regulation , beta-Hexosaminidase beta Chain/biosynthesis , Animals , C-Reactive Protein/genetics , CA1 Region, Hippocampal/pathology , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Sandhoff Disease/pathology , beta-Hexosaminidase beta Chain/genetics
2.
Front Neural Circuits ; 8: 140, 2014.
Article in English | MEDLINE | ID: mdl-25505386

ABSTRACT

The vesicular glutamate transporter 3 (VGLUT3) is expressed at several locations not normally associated with glutamate release. Although the function of this protein has been generally elusive, when expressed in non-glutamatergic synaptic terminals, VGLUT3 can not only allow glutamate co-transmission but also synergize the action of non-glutamate vesicular transporters. Interestingly, in the immature glycinergic projection between the medial nucleus of the trapezoid body (MNTB) and the lateral superior olive (LSO) of auditory brainstem, the transient early expression of VGLUT3 is required for normal developmental refinement. It has however been unknown whether the primary function of VGLUT3 in development of these inhibitory synapses is to enable glutamate release or to promote loading of inhibitory neurotransmitter through vesicular synergy. Using tissue from young mice in which Vglut3 had been genetically deleted, we evaluated inhibitory neurotransmission in the MNTB-LSO pathway. Our results show, in contrast to what has been seen at adult synapses, that VGLUT3 expression has little or no effect on vesicular synergy at the immature glycinergic synapse of brainstem. This finding supports the model that the primary function of increased VGLUT3 expression in the immature auditory brainstem is to enable glutamate release in a developing inhibitory circuit.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Glycine/metabolism , Superior Olivary Complex/growth & development , Trapezoid Body/growth & development , gamma-Aminobutyric Acid/metabolism , Amino Acid Transport Systems, Acidic/genetics , Animals , Auditory Pathways/growth & development , Auditory Pathways/physiology , Electric Stimulation , Mice, Knockout , Miniature Postsynaptic Potentials/physiology , Neural Inhibition/physiology , Patch-Clamp Techniques , Superior Olivary Complex/physiology , Synapses/physiology , Tissue Culture Techniques , Trapezoid Body/physiology
3.
Stem Cells Dev ; 23(16): 1937-46, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24694094

ABSTRACT

Several transcription factors and methods have been used to convert fibroblasts directly to neural fate and have provided insights into molecular mechanisms as to how each of these required factors orchestrate neural fate conversion. Here, we provide evidence and detailed characterization of the direct conversion process of primary adult human fibroblasts (hFib) to neural progenitor cells (NPC) using OCT4 alone. Factors previously associated with neural cell fate conversion were induced during hFib-NPC(OCT-4) generation, where OCT-4 alone was sufficient to induce neural fate conversion without the use of promiscuous small-molecule manipulation. Human Fib-NPC(OCT-4) proliferate, express neural stem/progenitor markers, and possess developmental potential that gives rise to all three major subtypes of neural cells: astrocytes, oligodendrocytes, and neurons with functional capacity. We propose a de-convoluted reprogramming approach for neural fate conversion in which OCT4 is sufficient for inducing neural conversion from hFib for disease modeling as well as the fundamental study of early neural fate induction.


Subject(s)
Cell Differentiation , Fibroblasts/physiology , Neural Stem Cells/physiology , Octamer Transcription Factor-3/physiology , Action Potentials , Adult , Animals , Cells, Cultured , Humans , Mice, Inbred NOD , Mice, SCID , SOXB1 Transcription Factors/metabolism , Stem Cell Transplantation/adverse effects , Teratoma/etiology , Teratoma/pathology
4.
PLoS One ; 8(9): e75688, 2013.
Article in English | MEDLINE | ID: mdl-24069436

ABSTRACT

The medial nucleus of the trapezoid body (MNTB) is a key nucleus in high-fidelity temporal processing that underlies sound localization in the auditory brainstem. While the glycinergic principal cells of the MNTB project to all primary nuclei of the superior olive, during development the projection from MNTB to the lateral superior olive (LSO) is of interest because this immature inhibitory projection is known to undergo tonotopic refinement during an early postnatal period, and because during this period individual MNTB terminals in the LSO transiently release glycine GABA and glutamate. Developmental changes in calcium-dependent release are understood to be required to allow various auditory nuclei to follow high frequency activity; however, little is known about maturation of calcium-dependent release in the MNTB-LSO pathway, which has been presumed to have less stringent requirements for high-fidelity temporal following. In acute brainstem slices of rats age postnatal day 1 to 15 we recorded whole-cell responses in LSO principal neurons to electrical stimulation in the MNTB in order to measure sensitivity to external calcium, the contribution of different voltage-gated calcium channel subtypes to vesicular release, and the maturation of these measures for both GABA/glycine and glutamate transmission. Our results establish that release of glutamate at MNTB-LSO synapses is calcium-dependent. Whereas no significant developmental changes were evident for glutamate release, GABA/glycine release underwent substantial changes over the first two postnatal weeks: soon after birth L-type, N-type, and P/Q-type voltage-gated calcium channels (VGCCs) together mediated release, but after hearing onset P/Q-type VGCCs predominated. Blockade of P/Q-type VGCCs reduced the estimated quantal number for GABA/gly and glutamate transmission at P5-8 and the frequency of evoked miniature glycinergic events at P12-15, without apparent effects on spontaneous release of neurotransmitter, supporting a model in which P/Q-type VGCCs are required for mature synchronous synaptic transmission, but not for spontaneous vesicle release.


Subject(s)
Auditory Cortex/metabolism , Auditory Pathways/physiology , Calcium/metabolism , Glutamic Acid/metabolism , Glycine/metabolism , Olivary Nucleus/metabolism , gamma-Aminobutyric Acid/metabolism , Action Potentials , Animals , Evoked Potentials , Neuronal Plasticity/physiology , Rats , Synaptic Transmission
5.
J Neurophysiol ; 106(5): 2570-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21832038

ABSTRACT

The lateral superior olive (LSO) integrates excitatory inputs driven by sound arriving at the ipsilateral ear with inhibitory inputs driven by sound arriving at the contralateral ear in order to compute interaural intensity differences needed for localizing high-frequency sound sources. Specific mechanisms necessary for developmental refinement of the inhibitory projection, which arises from the medial nucleus of the trapezoid body (MNTB), have only been partially deciphered. The demonstration that immature MNTB-LSO synapses release glutamate has led to a model in which early glutamate neurotransmission plays a major role in inhibitory plasticity. We used whole cell electrophysiology in acute auditory brain stem slices of neonatal rats to examine glutamatergic transmission in the developing MNTB-LSO pathway. Unexpectedly, AMPA receptor (AMPAR)-mediated responses were prevalent at the earliest ages. We found a salient developmental profile for NMDA receptor (NMDAR) activation, described both by the proportion of total glutamate current and by current durations, and we found evidence for distinct release probabilities for GABA/glycine and glutamate in the MNTB-LSO pathway. The developmental profile of NMDAR is consistent with the possibility that the inhibitory MNTB-LSO pathway experiences a sensitive period, driven by cochlear activity and mediated by GluN2B-containing NMDARs, between postnatal days 3 and 9. Differing neurotransmitter release probabilities could allow the synapse to switch between GABA/glycinergic transmission and mixed glutamate/GABA/glycinergic transmission in response to changing patterns of spiking activity.


Subject(s)
Auditory Pathways/physiology , Glutamic Acid/physiology , Neural Inhibition/physiology , Olivary Nucleus/physiology , Pons/physiology , Synaptic Potentials/physiology , Action Potentials/physiology , Age Factors , Animals , Animals, Newborn , Auditory Pathways/growth & development , Glycine/physiology , Neuronal Plasticity/physiology , Olivary Nucleus/growth & development , Patch-Clamp Techniques , Pons/growth & development , Rats , Rats, Sprague-Dawley , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Presynaptic/physiology , Sound Localization/physiology , Vesicular Glutamate Transport Proteins/physiology , gamma-Aminobutyric Acid/physiology
6.
PLoS One ; 6(6): e20756, 2011.
Article in English | MEDLINE | ID: mdl-21694776

ABSTRACT

Principal neurons of the lateral superior olive (LSO) compute the interaural intensity differences necessary for localizing high-frequency sounds. To perform this computation, the LSO requires precisely tuned, converging excitatory and inhibitory inputs that are driven by the two ears and that are matched for stimulus frequency. In rodents, the inhibitory inputs, which arise from the medial nucleus of the trapezoid body (MNTB), undergo extensive functional refinement during the first postnatal week. Similar functional refinement of the ascending excitatory pathway, which arises in the anteroventral cochlear nucleus (AVCN), has been assumed but has not been well studied. Using whole-cell voltage clamp in acute brainstem slices of neonatal rats, we examined developmental changes in input strength and pre- and post-synaptic properties of the VCN-LSO pathway. A key question was whether functional refinement in one of the two major input pathways might precede and then guide refinement in the opposite pathway. We find that elimination and strengthening of VCN inputs to the LSO occurs over a similar period to that seen for the ascending inhibitory (MNTB-LSO) pathway. During this period, the fractional contribution provided by NMDA receptors (NMDARs) declines while the contribution from AMPA receptors (AMPARs) increases. In the NMDAR-mediated response, GluN2B-containing NMDARs predominate in the first postnatal week and decline sharply thereafter. Finally, the progressive decrease in paired-pulse depression between birth and hearing onset allows these synapses to follow progressively higher frequencies. Our data are consistent with a model in which the excitatory and inhibitory projections to LSO are functionally refined in parallel during the first postnatal week, and they further suggest that GluN2B-containing NMDARs may mediate early refinement in the VCN-LSO pathway.


Subject(s)
Cochlear Nucleus/physiology , Hearing/physiology , Olivary Nucleus/physiology , Aging/physiology , Animals , Animals, Newborn , Excitatory Postsynaptic Potentials/physiology , Ion Channel Gating , Kinetics , Parturition , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors
7.
J Comp Neurol ; 519(12): 2417-33, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21456023

ABSTRACT

The lateral superior olive (LSO), a nucleus in the auditory brainstem, computes interaural intensity differences for sound localization by comparing converging excitatory and inhibitory inputs that carry tonotopically matched information from the two ears. Tonotopic refinement in the inhibitory projection pathway from the medial nucleus of the trapezoid body (MNTB) is known to be established during the first postnatal week in rats. During this period, immature MNTB terminals in the LSO contain vesicular transporters for both inhibitory and excitatory amino acids and release glutamate. The primary Ca(2+) sensors for vesicular release in the CNS are understood to be synaptotagmins, and in adult auditory brainstem synaptotagmin 2 is the predominant synaptotagmin. We asked here whether a different Ca(2+) sensor might be expressed in the immature auditory brainstem. We have found that synaptotagmin 1 is indeed expressed transiently in the immature auditory brainstem, most highly in those areas that receive glutamate-releasing immature inhibitory inputs from the MNTB, and that during the first postnatal week synaptotagmin 1 co-localizes with the vesicular glutamate transporter VGLUT3, a marker of glutamate-releasing immature inhibitory terminals from the MNTB. We suggest that immature MNTB terminals may contain two populations of synaptic vesicles, one expressing the vesicular inhibitory amino acid transporter together with synaptotagmin 2 and another expressing VGLUT3 together with synaptotagmin 1. Because Ca(2+) sensing is an important determinant of release properties for the presynaptic terminal, differential expression of the synaptotagmins might allow the differential release of excitatory and inhibitory neurotransmitters in response to differing patterns of neural activity.


Subject(s)
Auditory Pathways/physiology , Glutamic Acid/metabolism , Olivary Nucleus/physiology , Presynaptic Terminals/metabolism , Synaptotagmin II/metabolism , Synaptotagmin I/metabolism , Animals , Auditory Pathways/anatomy & histology , Immunohistochemistry , Olivary Nucleus/cytology , Rats , Sound Localization/physiology , Vesicular Glutamate Transport Proteins/metabolism
8.
Trends Neurosci ; 28(6): 290-6, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15927684

ABSTRACT

The ability to localize sound rapidly and accurately depends on the precise organization of inhibitory neuronal circuits in the auditory brainstem. However, the rules and mechanisms by which this precision is established during development are still poorly understood. Although activity-dependent reorganization has been known for over a decade to have a central role in this process, more recent studies have revealed an unanticipated degree of reorganization that occurs on levels ranging from cellular phenotype to network connectivity. These results suggest novel mechanisms by which immature inhibitory sound-localization circuits become optimized. Lessons from auditory brainstem circuits thus could provide insight into inhibitory development in other brain areas, where inhibitory networks are less experimentally accessible.


Subject(s)
Auditory Cortex/growth & development , Auditory Pathways/growth & development , Neural Inhibition/physiology , Sound Localization/physiology , Synapses/physiology , Age Factors , Animals , Glycine/metabolism , Neuronal Plasticity/physiology , gamma-Aminobutyric Acid/metabolism
9.
Nat Neurosci ; 8(3): 332-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15746915

ABSTRACT

Activity-dependent synapse refinement is crucial for the formation of precise excitatory and inhibitory neuronal circuits. Whereas the mechanisms that guide refinement of excitatory circuits are becoming increasingly clear, the mechanisms guiding inhibitory circuits have remained obscure. In the lateral superior olive (LSO), a nucleus in the mammalian sound localization system that receives inhibitory input from the medial nucleus of the trapezoid body (MNTB), specific elimination and strengthening of synapses that are both GABAergic and glycinergic (GABA/glycinergic synapses) is essential for the formation of a precise tonotopic map. We provide evidence that immature GABA/glycinergic synapses in the rat LSO also release the excitatory neurotransmitter glutamate, which activates postsynaptic NMDA receptors (NMDARs). Immunohistochemical studies demonstrate synaptic colocalization of the vesicular glutamate transporter 3 with the vesicular GABA transporter, indicating that GABA, glycine and glutamate are released from single MNTB terminals. Glutamatergic transmission at MNTB-LSO synapses is most prominent during the period of synapse elimination. Synapse-specific activation of NMDARs by glutamate release at GABAergic and glycinergic synapses could be important in activity-dependent refinement of inhibitory circuits.


Subject(s)
Auditory Pathways/growth & development , Neural Inhibition/physiology , Neurons/physiology , Olivary Nucleus/cytology , Olivary Nucleus/growth & development , Synapses/physiology , gamma-Aminobutyric Acid/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems, Acidic/metabolism , Animals , Animals, Newborn , Auditory Pathways/drug effects , Bicuculline/pharmacology , Cell Count/methods , Dose-Response Relationship, Radiation , Drug Interactions , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , GABA Antagonists/pharmacology , Glycine/metabolism , Glycine Agents , Immunohistochemistry/methods , In Vitro Techniques , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Inhibition/drug effects , Neural Inhibition/radiation effects , Neurons/drug effects , Neurons/radiation effects , Patch-Clamp Techniques/methods , Rats , Receptors, N-Methyl-D-Aspartate/physiology , Strychnine/pharmacology , Synapses/drug effects , Synapses/radiation effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Vesicular Glutamate Transport Protein 1 , Vesicular Glutamate Transport Proteins , Vesicular Inhibitory Amino Acid Transport Proteins , Vesicular Transport Proteins/metabolism
10.
J Neurophysiol ; 90(4): 2581-91, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12853437

ABSTRACT

The lateral superior olive (LSO) is a binaural auditory brain stem nucleus that plays a central role in sound localization. Survival and maturation of developing LSO neurons critically depend on intracellular calcium signaling. Here we investigated the mechanisms by which glutamatergic afferents from the cochlear nucleus increase intracellular calcium concentration in LSO neurons. Using fura-2 calcium imaging in slices prepared from neonatal mice, we found that cochlear nucleus afferents can activate all major classes of ionotropic and metabotropic glutamate receptors, each of which contributes to an increase in intracellular calcium. The specific activation of different glutamate receptor classes was dependent on response amplitudes and afferent stimulus patterns. Low-amplitude responses elicited by single stimuli were entirely mediated by calcium-impermeable AMPA/kainate receptors that activated voltage-gated calcium channels. Larger-amplitude responses elicited by either single stimuli or stimulus trains resulted in additional calcium influx through N-methyl-d-aspartate receptors. Finally, high-frequency stimulation also recruited group I and group II metabotropic glutamate receptors, both of which mobilized intracellular calcium. This calcium release in turn activated a strong influx of extracellular calcium through a membrane calcium channel that is distinct from voltage-gated calcium channels. Together, these results indicate that before hearing onset, distinct patterns of afferent activity generate qualitatively distinct types of calcium responses, which likely serve in guiding different aspects of LSO development.


Subject(s)
Calcium/metabolism , Olivary Nucleus/metabolism , Receptors, Glutamate/metabolism , Synapses/metabolism , Afferent Pathways/metabolism , Animals , Animals, Newborn , Female , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Receptors, Glutamate/classification
SELECTION OF CITATIONS
SEARCH DETAIL
...