Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Brain ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38478593

ABSTRACT

Full-length RIM1 and 2 are key components of the presynaptic active zone that ubiquitously control excitatory and inhibitory neurotransmitter release. Here, we report that the function of the small RIM isoform RIM4, consisting of a single C2 domain, is strikingly different from that of the long isoforms. RIM4 is dispensable for neurotransmitter release but plays a postsynaptic, cell-type specific role in cerebellar Purkinje cells that is essential for normal motor function. In the absence of RIM4, Purkinje cell intrinsic firing is reduced and caffeine-sensitive, and dendritic integration of climbing fibre input is disturbed. Mice lacking RIM4, but not mice lacking RIM1/2, selectively in Purkinje cells exhibit a severe, hours-long paroxysmal dystonia. These episodes can also be induced by caffeine, ethanol or stress and closely resemble the deficits seen with mutations of the PNKD (paroxysmal non-kinesigenic dystonia) gene. Our data reveal essential postsynaptic functions of RIM proteins and show non-overlapping specialized functions of a small isoform despite high homology to a single domain in the full-length proteins.

2.
Science ; 370(6523)2020 12 18.
Article in English | MEDLINE | ID: mdl-33335032

ABSTRACT

Myelin plasticity is critical for neurological function, including learning and memory. However, it is unknown whether this plasticity reflects uniform changes across all neuronal subtypes, or whether myelin dynamics vary between neuronal classes to enable fine-tuning of adaptive circuit responses. We performed in vivo two-photon imaging of myelin sheaths along single axons of excitatory callosal neurons and inhibitory parvalbumin-expressing interneurons in adult mouse visual cortex. We found that both neuron types show homeostatic myelin remodeling under normal vision. However, monocular deprivation results in adaptive myelin remodeling only in parvalbumin-expressing interneurons. An initial increase in elongation of myelin segments is followed by contraction of a separate cohort of segments. This data indicates that distinct classes of neurons individualize remodeling of their myelination profiles to diversify circuit tuning in response to sensory experience.


Subject(s)
Myelin Sheath/metabolism , Neocortex/metabolism , Neurons/metabolism , Visual Cortex/metabolism , Animals , Corpus Callosum/cytology , Corpus Callosum/metabolism , Female , GABAergic Neurons/metabolism , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Imaging , Neocortex/cytology , Neuronal Plasticity , Neurons/classification , Parvalbumins/metabolism , Visual Cortex/cytology
3.
Cell Rep ; 28(6): 1584-1595.e5, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390571

ABSTRACT

A key feature of brain plasticity is the experience-dependent selection of optimal connections, implemented by a set of activity-regulated genes that dynamically adjust synapse strength and number. The activity-regulated gene cpg15/neuritin has been previously implicated in stabilization and maturation of excitatory synapses. Here, we combine two-photon microscopy with genetic and sensory manipulations to dissect excitatory synapse formation in vivo and examine the role of activity and CPG15 in dendritic spine formation, PSD95 recruitment, and synapse stabilization. We find that neither visual experience nor CPG15 is required for spine formation. However, PSD95 recruitment to nascent spines and their subsequent stabilization requires both. Further, cell-autonomous CPG15 expression is sufficient to replace experience in facilitating PSD95 recruitment and spine stabilization. CPG15 directly interacts with α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors on immature dendritic spines, suggesting a signaling mode for this small extracellular molecule acting as an experience-dependent "selector" for spine stabilization and synapse maturation.


Subject(s)
Dendritic Spines/metabolism , Disks Large Homolog 4 Protein/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials , Female , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Neuronal Plasticity , Receptors, AMPA/metabolism
4.
Front Cell Neurosci ; 11: 160, 2017.
Article in English | MEDLINE | ID: mdl-28642685

ABSTRACT

Rab interacting molecules (RIMs) are multi-domain proteins that positively regulate the number of Ca2+ channels at the presynaptic active zone (AZ). Several molecular mechanisms have been demonstrated for RIM-binding to components of the presynaptic Ca2+ channel complex, the key signaling element at the AZ. Here, we report an interaction of the C2B domain of RIM2α and RIM3γ with the C-terminus of the pore-forming α-subunit of CaV1.3 channels (CaV1.3α1), which mediate stimulus-secretion coupling at the ribbon synapses of cochlear inner hair cells (IHCs). Co-expressing full-length RIM2α with a Ca2+ channel complex closely resembling that of IHCs (CaV1.3α1-CaVß2a) in HEK293 cells doubled the Ca2+-current and shifted the voltage-dependence of Ca2+ channel activation by approximately +3 mV. Co-expression of the short RIM isoform RIM3γ increased the CaV1.3α1-CaVß2a-mediated Ca2+-influx in HEK293 cells, but disruption of RIM3γ in mice left Ca2+-influx in IHCs and hearing intact. In conclusion, we propose that RIM2α and RIM3γ directly interact with the C-terminus of the pore-forming subunit of CaV1.3 Ca2+ channels and positively regulate their plasma membrane expression in HEK293 cells.

5.
Proc Natl Acad Sci U S A ; 112(24): E3141-9, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26034270

ABSTRACT

Ca(2+) influx triggers the fusion of synaptic vesicles at the presynaptic active zone (AZ). Here we demonstrate a role of Ras-related in brain 3 (Rab3)-interacting molecules 2α and ß (RIM2α and RIM2ß) in clustering voltage-gated CaV1.3 Ca(2+) channels at the AZs of sensory inner hair cells (IHCs). We show that IHCs of hearing mice express mainly RIM2α, but also RIM2ß and RIM3γ, which all localize to the AZs, as shown by immunofluorescence microscopy. Immunohistochemistry, patch-clamp, fluctuation analysis, and confocal Ca(2+) imaging demonstrate that AZs of RIM2α-deficient IHCs cluster fewer synaptic CaV1.3 Ca(2+) channels, resulting in reduced synaptic Ca(2+) influx. Using superresolution microscopy, we found that Ca(2+) channels remained clustered in stripes underneath anchored ribbons. Electron tomography of high-pressure frozen synapses revealed a reduced fraction of membrane-tethered vesicles, whereas the total number of membrane-proximal vesicles was unaltered. Membrane capacitance measurements revealed a reduction of exocytosis largely in proportion with the Ca(2+) current, whereas the apparent Ca(2+) dependence of exocytosis was unchanged. Hair cell-specific deletion of all RIM2 isoforms caused a stronger reduction of Ca(2+) influx and exocytosis and significantly impaired the encoding of sound onset in the postsynaptic spiral ganglion neurons. Auditory brainstem responses indicated a mild hearing impairment on hair cell-specific deletion of all RIM2 isoforms or global inactivation of RIM2α. We conclude that RIM2α and RIM2ß promote a large complement of synaptic Ca(2+) channels at IHC AZs and are required for normal hearing.


Subject(s)
Calcium Channels, L-Type/metabolism , Hair Cells, Auditory, Inner/metabolism , rab3 GTP-Binding Proteins/metabolism , Animals , Calcium Signaling , Electron Microscope Tomography , Evoked Potentials, Auditory, Brain Stem , Exocytosis , Hair Cells, Auditory, Inner/ultrastructure , Hearing/physiology , Ion Channel Gating , Mice , Mice, Inbred C57BL , Mice, Knockout , Otoacoustic Emissions, Spontaneous , Patch-Clamp Techniques , Spiral Ganglion/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptic Vesicles/metabolism , rab3 GTP-Binding Proteins/deficiency , rab3 GTP-Binding Proteins/genetics
6.
Exp Cell Res ; 335(2): 157-64, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25720549

ABSTRACT

Before fusing with the presynaptic plasma membrane to release neurotransmitter into the synaptic cleft synaptic vesicles have to be recruited to and docked at a specialized area of the presynaptic nerve terminal, the active zone. Exocytosis of synaptic vesicles is restricted to the presynaptic active zone, which is characterized by a unique and highly interconnected set of proteins. The protein network at the active zone is integrally involved in this process and also mediates changes in release properties, for example in response to alterations in the level of neuronal network activity. In recent years the development of novel techniques has greatly advanced our understanding of the molecular identity of respective active zone components as well as of the ultrastructure of this membranous subcompartment and of the SV release machinery. Furthermore, active zones are now viewed as dynamic structures whose composition and size are correlated with synaptic efficacy. Therefore, the dynamic remodeling of the protein network at the active zone has emerged as one potential mechanism underlying acute and long-term synaptic plasticity. Here, we will discuss this recent progress and its implications for our view of the role of the AZ in synaptic function.


Subject(s)
Presynaptic Terminals/metabolism , Synaptic Vesicles/metabolism , Adaptor Proteins, Vesicular Transport/physiology , Animals , Humans , Neurotransmitter Agents/metabolism , Presynaptic Terminals/ultrastructure
7.
J Neurosci ; 33(2): 824-39, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23303958

ABSTRACT

The large isoforms of the Rab3 interacting molecule (RIM) family, RIM1α/ß and RIM2α/ß, have been shown to be centrally involved in mediating presynaptic active zone function. The RIM protein family contains two additional small isoforms, RIM3γ and RIM4γ, which are composed only of the RIM-specific C-terminal C2B domain and varying N-terminal sequences and whose function remains to be elucidated. Here, we report that both, RIM3γ and RIM4γ, play an essential role for the development of neuronal arborization and of dendritic spines independent of synaptic function. γ-RIM knock-down in rat primary neuronal cultures and in vivo resulted in a drastic reduction in the complexity of neuronal arborization, affecting both axonal and dendritic outgrowth, independent of the time point of γ-RIM downregulation during dendrite development. Rescue experiments revealed that the phenotype is caused by a function common to both γ-RIMs. These findings indicate that γ-RIMs are involved in cell biological functions distinct from the regulation of synaptic vesicle exocytosis and play a role in the molecular mechanisms controlling the establishment of dendritic complexity and axonal outgrowth.


Subject(s)
Dendrites/physiology , Membrane Transport Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Animals , Blotting, Western , Cell Survival/physiology , Cells, Cultured , DNA Primers/genetics , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Female , Gene Silencing , Genetic Vectors/genetics , Golgi Apparatus/physiology , Golgi Apparatus/ultrastructure , HEK293 Cells , Humans , Immunohistochemistry , In Situ Hybridization , Lentivirus/genetics , Male , Membrane Transport Proteins/genetics , Nerve Tissue Proteins/genetics , Rats , Rats, Wistar , Subcellular Fractions/metabolism , Synapses/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...