Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 3648, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684645

ABSTRACT

Neuronal network formation is facilitated by recognition between synaptic cell adhesion molecules at the cell surface. Alternative splicing of cell adhesion molecules provides additional specificity in forming neuronal connections. For the teneurin family of cell adhesion molecules, alternative splicing of the EGF-repeats and NHL domain controls synaptic protein-protein interactions. Here we present cryo-EM structures of the compact dimeric ectodomain of two teneurin-3 isoforms that harbour the splice insert in the EGF-repeats. This dimer is stabilised by an EGF8-ABD contact between subunits. Cryo-EM reconstructions of all four splice variants, together with SAXS and negative stain EM, reveal compacted dimers for each, with variant-specific dimeric arrangements. This results in specific trans-cellular interactions, as tested in cell clustering and stripe assays. The compact conformations provide a structural basis for teneurin homo- and heterophilic interactions. Altogether, our findings demonstrate how alternative splicing results in rearrangements of the dimeric subunits, influencing neuronal recognition and likely circuit wiring.


Subject(s)
Alternative Splicing , Cryoelectron Microscopy , Neurons , Neurons/metabolism , Animals , Humans , Protein Multimerization , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/chemistry , Protein Isoforms/metabolism , Protein Isoforms/genetics , Protein Isoforms/chemistry , Models, Molecular
2.
Nat Commun ; 13(1): 6607, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36329006

ABSTRACT

Cell-surface expressed contactin 1 and neurofascin 155 control wiring of the nervous system and interact across cells to form and maintain paranodal myelin-axon junctions. The molecular mechanism of contactin 1 - neurofascin 155 adhesion complex formation is unresolved. Crystallographic structures of complexed and individual contactin 1 and neurofascin 155 binding regions presented here, provide a rich picture of how competing and complementary interfaces, post-translational glycosylation, splice differences and structural plasticity enable formation of diverse adhesion sites. Structural, biophysical, and cell-clustering analysis reveal how conserved Ig1-2 interfaces form competing heterophilic contactin 1 - neurofascin 155 and homophilic neurofascin 155 complexes whereas contactin 1 forms low-affinity clusters through interfaces on Ig3-6. The structures explain how the heterophilic Ig1-Ig4 horseshoe's in the contactin 1 - neurofascin 155 complex define the 7.4 nm paranodal spacing and how the remaining six domains enable bridging of distinct intercellular distances.


Subject(s)
Cell Adhesion Molecules , Contactin 1 , Cell Adhesion Molecules/metabolism , Nerve Growth Factors/metabolism , Contactins , Axons/metabolism , Cell Adhesion Molecules, Neuronal/metabolism
3.
EMBO J ; 41(9): e107505, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35099835

ABSTRACT

Establishment of correct synaptic connections is a crucial step during neural circuitry formation. The Teneurin family of neuronal transmembrane proteins promotes cell-cell adhesion via homophilic and heterophilic interactions, and is required for synaptic partner matching in the visual and hippocampal systems in vertebrates. It remains unclear how individual Teneurins form macromolecular cis- and trans-synaptic protein complexes. Here, we present a 2.7 Å cryo-EM structure of the dimeric ectodomain of human Teneurin4. The structure reveals a compact conformation of the dimer, stabilized by interactions mediated by the C-rich, YD-shell, and ABD domains. A 1.5 Å crystal structure of the C-rich domain shows three conserved calcium binding sites, and thermal unfolding assays and SAXS-based rigid-body modeling demonstrate that the compactness and stability of Teneurin4 dimers are calcium-dependent. Teneurin4 dimers form a more extended conformation in conditions that lack calcium. Cellular assays reveal that the compact cis-dimer is compatible with homomeric trans-interactions. Together, these findings support a role for teneurins as a scaffold for macromolecular complex assembly and the establishment of cis- and trans-synaptic interactions to construct functional neuronal circuits.


Subject(s)
Calcium , Tenascin , Animals , Calcium/metabolism , Humans , Neurons/metabolism , Protein Conformation , Scattering, Small Angle , Tenascin/chemistry , Tenascin/metabolism , X-Ray Diffraction
4.
Cell Rep ; 27(3): 666-675.e5, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995465

ABSTRACT

Dendritic inhibitory synapses are most efficient in modulating excitatory inputs localized on the same dendrite, but it is unknown whether their location is random or regulated. Here, we show that the formation of inhibitory synapses can be directed by excitatory synaptic activity on the same dendrite. We stimulated dendritic spines close to a GABAergic axon crossing by pairing two-photon glutamate uncaging with postsynaptic depolarization in CA1 pyramidal cells. We found that repeated spine stimulation promoted growth of a GABAergic bouton onto the same dendrite. The dendritic feedback signal required postsynaptic activation of DAGL, which produces the endocannabinoid 2-AG, and was mediated by CB1 receptors. We could also induce inhibitory bouton growth by local, brief applications of 2-AG. Our findings reveal a dendritic signaling mechanism to trigger growth of an inhibitory bouton at dendritic locations with strong excitatory synaptic activity, and this mechanism may serve to ensure inhibitory control over clustered excitatory inputs.


Subject(s)
Dendrites/physiology , Endocannabinoids/metabolism , Synapses/physiology , Animals , Axons/metabolism , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Excitatory Postsynaptic Potentials/drug effects , Female , Glutamic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Pyramidal Cells/physiology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
5.
J Neurosci ; 39(22): 4221-4237, 2019 05 29.
Article in English | MEDLINE | ID: mdl-30914448

ABSTRACT

Changes in inhibitory connections are essential for experience-dependent circuit adaptations. Defects in inhibitory synapses are linked to neurodevelopmental disorders, but the molecular processes underlying inhibitory synapse formation are not well understood. Here we use high-resolution two-photon microscopy in organotypic hippocampal slices from GAD65-GFP mice of both sexes to examine the signaling pathways induced by the postsynaptic signaling molecule Semaphorin4D (Sema4D) during inhibitory synapse formation. By monitoring changes in individual GFP-labeled presynaptic boutons, we found that the primary action of Sema4D is to induce stabilization of presynaptic boutons within tens of minutes. Stabilized boutons rapidly recruited synaptic vesicles, followed by accumulation of postsynaptic gephyrin and were functional after 24 h, as determined by electrophysiology and immunohistochemistry. Inhibitory boutons are only sensitive to Sema4D at a specific stage during synapse formation and sensitivity to Sema4D is regulated by network activity. We further examined the intracellular signaling cascade triggered by Sema4D and found that bouton stabilization occurs through rapid remodeling of the actin cytoskeleton. This could be mimicked by the actin-depolymerizing drug latrunculin B or by reducing ROCK activity. We discovered that the intracellular signaling cascade requires activation of the receptor tyrosine kinase MET, which is a well known autism risk factor. By using a viral approach to reduce MET levels specifically in inhibitory neurons, we found that their axons are no longer sensitive to Sema4D signaling. Together, our data yield important insights into the molecular pathway underlying activity-dependent Sema4D-induced synapse formation and reveal a novel role for presynaptic MET at inhibitory synapses.SIGNIFICANCE STATEMENT GABAergic synapses provide the main inhibitory control of neuronal activity in the brain. We wanted to unravel the sequence of molecular events that take place when formation of inhibitory synapses is triggered by a specific signaling molecule, Sema4D. We find that this signaling pathway depends on network activity and involves specific remodeling of the intracellular actin cytoskeleton. We also reveal a previously unknown role for MET at inhibitory synapses. Our study provides novel insights into the dynamic process of inhibitory synapse formation. As defects in GABAergic synapses have been implied in many brain disorders, and mutations in MET are strong risk factors for autism, our findings urge for a further investigation of the role of MET at inhibitory synapses.


Subject(s)
Antigens, CD/metabolism , Neurogenesis/physiology , Presynaptic Terminals/metabolism , Proto-Oncogene Proteins c-met/metabolism , Semaphorins/metabolism , Synapses/metabolism , Animals , Female , Hippocampus/metabolism , Male , Mice , Organ Culture Techniques
6.
Curr Biol ; 26(13): 1705-1712, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27265394

ABSTRACT

Kinesin and dynein motors drive bidirectional cargo transport along microtubules and have a critical role in polarized cargo trafficking in neurons [1, 2]. The kinesin-2 family protein KIF17 is a dendrite-specific motor protein and has been shown to interact with several dendritic cargoes [3-7]. However, the mechanism underlying the dendritic targeting of KIF17 remains poorly understood [8-11]. Using live-cell imaging combined with inducible trafficking assays to directly probe KIF17 motor activity in living neurons, we found that the polarized sorting of KIF17 to dendrites is regulated in multiple steps. First, cargo binding of KIF17 relieves autoinhibition and initiates microtubule-based cargo transport. Second, KIF17 does not autonomously target dendrites, but enters the axon where the actin cytoskeleton at the axon initial segment (AIS) prevents KIF17 vesicles from moving further into the axon. Third, dynein-based motor activity is able to redirect KIF17-coupled cargoes into dendrites. We propose a three-step model for polarized targeting of KIF17, in which the collective function of multiple motor teams is required for proper dendritic sorting.


Subject(s)
Axons/metabolism , Dendrites/metabolism , Kinesins/metabolism , Animals , Cells, Cultured , Microtubules/metabolism , Protein Transport , Rats
7.
Curr Biol ; 26(7): 849-61, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-26948876

ABSTRACT

Kinesin motor proteins play a fundamental role for normal neuronal development by controlling intracellular cargo transport and microtubule (MT) cytoskeleton organization. Regulating kinesin activity is important to ensure their proper functioning, and their misregulation often leads to severe human neurological disorders. Homozygous nonsense mutations in kinesin-binding protein (KBP)/KIAA1279 cause the neurological disorder Goldberg-Shprintzen syndrome (GOSHS), which is characterized by intellectual disability, microcephaly, and axonal neuropathy. Here, we show that KBP regulates kinesin activity by interacting with the motor domains of a specific subset of kinesins to prevent their association with the MT cytoskeleton. The KBP-interacting kinesins include cargo-transporting motors such as kinesin-3/KIF1A and MT-depolymerizing motor kinesin-8/KIF18A. We found that KBP blocks KIF1A/UNC-104-mediated synaptic vesicle transport in cultured hippocampal neurons and in C. elegans PVD sensory neurons. In contrast, depletion of KBP results in the accumulation of KIF1A motors and synaptic vesicles in the axonal growth cone. We also show that KBP regulates neuronal MT dynamics by controlling KIF18A activity. Our data suggest that KBP functions as a kinesin inhibitor that modulates MT-based cargo motility and depolymerizing activity of a subset of kinesin motors. We propose that misregulation of KBP-controlled kinesin motors may represent the underlying molecular mechanism that contributes to the neuropathological defects observed in GOSHS patients.


Subject(s)
Craniofacial Abnormalities/metabolism , Hirschsprung Disease/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Animals , Caenorhabditis elegans/metabolism , Carrier Proteins/metabolism , Kinesins/chemistry , Kinesins/metabolism , Mice , Neurons/metabolism , Synaptic Vesicles/metabolism
8.
Neuron ; 88(6): 1208-1226, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26671463

ABSTRACT

Axon formation, the initial step in establishing neuronal polarity, critically depends on local microtubule reorganization and is characterized by the formation of parallel microtubule bundles. How uniform microtubule polarity is achieved during axonal development remains an outstanding question. Here, we show that the tripartite motif containing (TRIM) protein TRIM46 plays an instructive role in the initial polarization of neuronal cells. TRIM46 is specifically localized to the newly specified axon and, at later stages, partly overlaps with the axon initial segment (AIS). TRIM46 specifically forms closely spaced parallel microtubule bundles oriented with their plus-end out. Without TRIM46, all neurites have a dendrite-like mixed microtubule organization resulting in Tau missorting and altered cargo trafficking. By forming uniform microtubule bundles in the axon, TRIM46 is required for neuronal polarity and axon specification in vitro and in vivo. Thus, TRIM46 defines a unique axonal cytoskeletal compartment for regulating microtubule organization during neuronal development.


Subject(s)
Axons/physiology , Axons/ultrastructure , Cell Polarity/physiology , Microtubules/physiology , Microtubules/ultrastructure , Nerve Tissue Proteins/physiology , Nerve Tissue Proteins/ultrastructure , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Cerebral Cortex/ultrastructure , Chlorocebus aethiops , Female , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/physiology , Neurons/ultrastructure , Pregnancy , Rats , Repressor Proteins/physiology , Repressor Proteins/ultrastructure
9.
Front Cell Neurosci ; 7: 219, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24312009

ABSTRACT

Synaptic connections in our brains change continuously and throughout our lifetime. Despite ongoing synaptic changes, a healthy balance between excitation and inhibition is maintained by various forms of homeostatic and activity-dependent adaptations, ensuring stable functioning of neuronal networks. In this review we summarize experimental evidence for activity-dependent changes occurring in inhibitory axons, in cultures as well as in vivo. Axons form many presynaptic terminals, which are dynamic structures sharing presynaptic material along the axonal shaft. We discuss how internal (e.g., vesicle sharing) and external factors (e.g., binding of cell adhesion molecules or secreted factors) may affect the formation and plasticity of inhibitory synapses.

SELECTION OF CITATIONS
SEARCH DETAIL
...