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.
J Neurophysiol ; 129(4): 767-780, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36883742

ABSTRACT

Generalization in motor learning refers to the transfer of a learned compensation to other relevant contexts. The generalization function is typically assumed to be of Gaussian shape, centered on the planned motion, although more recent studies associate generalization with the actual motion. Because motor learning is thought to involve multiple adaptive processes with different time constants, we hypothesized that these processes have different time-dependent contributions to the generalization. Guided by a model-based approach, the objective of the present study was to experimentally examine these contributions. We first reformulated a validated two-state adaptation model as a combination of weighted motor primitives, each specified as a Gaussian-shaped tuning function. Adaptation in this model is achieved by updating individual weights of the primitives of the fast and slow adaptive process separately. Depending on whether updating occurred in a plan-referenced or a motion-referenced manner, the model predicted distinct contributions to the overall generalization by the slow and fast process. We tested 23 participants in a reach adaptation task, using a spontaneous recovery paradigm consisting of five successive blocks of a long adaptation phase to a viscous force field, a short adaptation phase with the opposite force, and an error-clamp phase. Generalization was assessed in 11 movement directions relative to the trained target direction. Results of our participant population fell within a continuum of evidence for plan-referenced to evidence for motion-referenced updating. This mixture may reflect the differential weighting of explicit and implicit compensation strategies among participants.NEW & NOTEWORTHY Error-based reach adaptation can be modeled by fast and slow adaptive processes. Using a spontaneous recovery paradigm and model-based analyses, we tested how these processes generalize during force-field reach adaptation. Depending on whether the fast and slow adaptive processes operate by crediting the planned or actual motion, the model predicts distinct contributions of them to the overall generalization function. We show that human participants fall within a continuum of evidence for plan-referenced to motion-referenced updating.


Subject(s)
Generalization, Psychological , Learning , Humans , Movement , Adaptation, Physiological , Time , Psychomotor Performance
2.
Materials (Basel) ; 15(3)2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35160701

ABSTRACT

OBJECTIVES: Zirconia (Y-TZP) ceramics are considered as posterior fixed partial denture (FPD) materials; however, their applications are limited due to chipping. The use of monolithic lithium disilicate (LiDi) glass ceramics in posterior FPDs can be advantageous. This in vitro study aims to compare the loads until failure of posterior Y-TZP-FPDs and LiDi-FPDs before and after aging.

3.
PLoS One ; 15(10): e0240666, 2020.
Article in English | MEDLINE | ID: mdl-33075104

ABSTRACT

When we reach for an object during a passive whole body rotation, a tangential Coriolis force is generated on the arm. Yet, within a few trials, the brain adapts to this force so it does not disrupt the reach. Is this adaptation governed by a single-rate or dual-rate learning process? Here, guided by state-space modeling, we studied human reach adaptation in a fully-enclosed rotating room. After 90 pre-rotation reaches (baseline), participants were trained to make 240 to-and-fro reaches while the room rotated at 10 rpm (block A), then performed 6 reaches under opposite room rotation (block B), and subsequently made 100 post-rotation reaches (washout). A control group performed the same paradigm, but without the reaches during rotation block B. Single-rate and dual-rate models can be best dissociated if there would be full un-learning of compensation A during block B, but minimal learning of B. From the perspective of a dual-rate model, the un-learning observed in block B would mainly be caused by the faster state, such that the washout reaches would show retention effects of the slower state, called spontaneous recovery. Alternatively, following a single-rate model, the same state would govern the learning in block A and un-learning in block B, such that the washout reaches mimic the baseline reaches. Our results do not provide clear signs of spontaneous recovery in the washout reaches. Model fits further show that a single-rate process outperformed a dual-rate process. We suggest that a single-rate process underlies Coriolis force reach adaptation, perhaps because these forces relate to familiar body dynamics and are assigned to an internal cause.


Subject(s)
Adaptation, Physiological , Coriolis Force , Learning , Psychomotor Performance , Rotation , Adult , Arm/physiology , Female , Healthy Volunteers , Humans , Male , Young Adult
4.
Front Cell Neurosci ; 8: 185, 2014.
Article in English | MEDLINE | ID: mdl-25100946

ABSTRACT

During embryonic development the preoptic area (POA) gives rise to two populations of neurons which are generated at the same time, cortical interneurons and striatal cells. POA-derived cortical interneurons take a superficial path and avoid the developing striatum (Str) when they migrate to their target region. We found that EphB1, which is expressed in the striatal anlage, prevents cortical interneurons from entering the Str via ephrin-B3 reverse signaling. In contrast, for striatal neurons which also express ephrin-B3, EphB1 acts as a stop signal. This dual role of EphB1 is due to differences in ephrin-B3 reverse signaling cascades. For striatal neurons, binding of EphB1 to ephrin-B3 reduces endogenously high levels of pSrc and pFAK, which then causes the cells to stop migration. In contrast, in cortical interneurons EphB1-ephrin-B3 reverse signaling leads to phosphorylation of Src and focal adhesion kinase (FAK) which then mediates repulsion. Consistent with these in vitro findings, in an ephrin-B3 knockout mouse line, we discovered misrouted cortical interneurons in the Str and an over-migration of striatal neurons in their target region. Thus, EphB1/ephrin-B3 reverse signaling has a different impact on two sets of neurons which are generated at the same time and place: it can act as a repulsive cue for migrating neurons or it can terminate neuronal migration, a novel role of the Eph/ephrin system.

5.
Development ; 141(2): 460-71, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24381199

ABSTRACT

Inhibitory interneurons control the flow of information and synchronization in the cerebral cortex at the circuit level. During embryonic development, multiple subtypes of cortical interneurons are generated in different regions of the ventral telencephalon, such as the medial and caudal ganglionic eminence (MGE and CGE), as well as the preoptic area (POA). These neurons then migrate over long distances towards their cortical target areas. Diverse families of diffusible and cell-bound signaling molecules, including the Eph/ephrin system, regulate and orchestrate interneuron migration. Ephrin A3 and A5, for instance, are expressed at the borders of the pathway of MGE-derived interneurons and prevent these cells from entering inappropriate regions via EphA4 forward signaling. We found that MGE-derived interneurons, in addition to EphA4, also express ephrin A and B ligands, suggesting Eph/ephrin forward and reverse signaling in the same cell. In vitro and in vivo approaches showed that EphA4-induced reverse signaling in MGE-derived interneurons promotes their migration and that this effect is mediated by ephrin A2 ligands. In EphA4 mutant mice, as well as after ephrin A2 knockdown using in utero electroporation, we found delayed interneuron migration at embryonic stages. Thus, besides functions in guiding MGE-derived interneurons to the cortex through forward signaling, here we describe a novel role of the ephrins in driving these neurons to their target via reverse signaling.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Ephrin-A2/metabolism , Interneurons/physiology , Receptor, EphA4/metabolism , Telencephalon/embryology , Telencephalon/metabolism , Animals , Cell Movement/physiology , Cerebral Cortex/cytology , Ephrin-A2/antagonists & inhibitors , Ephrin-A2/genetics , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, EphA4/genetics , Signal Transduction , Telencephalon/cytology
6.
J Neurosci ; 31(50): 18364-80, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22171039

ABSTRACT

The integration of interneuron subtypes into specific microcircuits is essential for proper cortical function. Understanding to what extent interneuron diversity is regulated and maintained during development might help to reveal the principles that govern their role as synchronizing elements as well as causes for dysfunction. Particular interneuron subtypes are generated in a temporally regulated manner in the medial ganglionic eminence (MGE), the caudal ganglionic eminence, and the preoptic area (POA) of the basal telencephalon. Long-range tangential migration from their site of origin to cortical targets is orchestrated by a variety of attractive, repulsive, membrane-bound, and secreted signaling molecules, to establish the critical balance of inhibition and excitation. It remains unknown whether interneurons deriving from distinct domains are predetermined to migrate in particular routes and whether this process underlies cell type-specific regulation. We found that POA- and MGE-derived cortical interneurons migrate within spatially segregated corridors. EphrinB3, expressed in POA-derived interneurons traversing the superficial route, acts as a repellent signal for deeply migrating interneurons born in the MGE, which is mediated by EphA4 forward signaling. In contrast, EphA4 induces repulsive ephrinB3 reverse signaling in interneurons generated in the POA, restricting this population to the superficial path. Perturbation of this bidirectional ephrinB3/EphA4 signaling in vitro and in vivo leads to a partial intermingling of cells in these segregated migratory pathways. Thus, we conclude that cell contact-mediated bidirectional ephrinB3/EphA4 signaling mediates the sorting of MGE- and POA-derived interneurons in the deep and superficial migratory stream.


Subject(s)
Cell Movement/physiology , Ephrin-B3/metabolism , Interneurons/metabolism , Preoptic Area/metabolism , Receptor, EphA4/metabolism , Signal Transduction/physiology , Telencephalon/metabolism , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Mice , Preoptic Area/embryology , Telencephalon/embryology
7.
Cell Adh Migr ; 4(3): 400-8, 2010.
Article in English | MEDLINE | ID: mdl-20473036

ABSTRACT

Cortical interneurons are born in the proliferative zones of the ganglionic eminences in the subpallium and migrate to the developing cortex along well-defined tangential routes. The mechanisms regulating interneuron migration are not completely understood. Here we examine the role of class-A members of the Eph/ephrin system in directing the migration of interneurons. In situ hybridizations demonstrated that ephrin-A3 is expressed in the developing striatum, an area that is strictly avoided by migrating cortical interneurons in vivo, which express the EphA4 receptor. We then examined interneuron migration in grafting experiments, where explants of the medial ganglionic eminence (MGE) from enhanced green fluorescent protein-expressing transgenic mice were homotopically grafted into host slices from wildtype littermate embryos. After blocking ephrin-A ligands, many interneurons invaded the striatal anlage. Moreover, stripe assay experiments revealed that ephrin-A3 acts as a repellent cue for neurons from the medial ganglionic eminence. Downregulation of the EphA4 receptor via siRNA transfection reduced the repulsive effect of ephrin-A3, indicating that EphA4 mediates at least in part the repulsive effect of ephrin-A3 on these cells. Together, these results suggest that ephrin-A3 acts as a repulsive cue that restricts cortical interneurons from entering inappropriate regions and thus contributes to define the migratory route of cortical interneurons.


Subject(s)
Cell Movement , Cerebral Cortex/cytology , Ephrin-A3/metabolism , Ephrin-A4/metabolism , Interneurons/cytology , Telencephalon/cytology , Animals , Biological Assay , Down-Regulation , Humans , Interneurons/transplantation , Ligands , Median Eminence/cytology , Mice , Models, Biological , Neostriatum/metabolism , Neostriatum/pathology , Telencephalon/metabolism
8.
J Comp Neurol ; 517(5): 645-54, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19827157

ABSTRACT

Previous studies demonstrated that in ephrin-A5-deficient mice corticothalamic arbors are reduced by more than 50% in layer 4 of the somatosensory cortex (S1), where ephrin-A5 is normally expressed. Here we examined possible consequences of the reduced thalamic input on spiny stellate cells, the target neurons of thalamocortical afferents. Using ballistic delivery of particles coated with lipophilic dyes in fixed slices and confocal laser-microscopy, we could quantitatively analyze the morphology of these neurons. Cells were examined in S1 at postnatal day 8 (P8), when thalamic afferents establish synaptic contacts and the dendrites of their target cells are covered with filopodia, and at P23, after synapse formation and replacement of filopodia by spines. Our results indicate that at P8 the dendrites of cells in mutant animals exhibit more filopodia and are more branched than dendrites of wildtype cells. In contrast, there is no difference in the extent of the dendritic tree between knockout and control animals. At P23, dendrites of neurons in ephrin-A5-deficient mice are still more branched, but possess fewer spines than wildtype cells. Thus, at early stages layer 4 neurons appear to compensate the reduced thalamic input by increasing dendritic branching and the density of filopodia. However, while at later stages the dendrites of layer 4 neurons in mutants are still more branched, their spine density is now lower than in wildtype cells. Taken together, these data demonstrate that the structure of spiny stellate cells is shaped by thalamic input and Eph receptor signaling.


Subject(s)
Dendritic Spines/classification , Ephrin-A5/metabolism , Interneurons/cytology , Somatosensory Cortex/cytology , Animals , Dendritic Spines/metabolism , Ephrin-A5/genetics , Female , Interneurons/classification , Interneurons/metabolism , Male , Mice , Mice, Knockout , Pseudopodia/classification , Pseudopodia/metabolism , Somatosensory Cortex/metabolism
9.
Eur J Neurosci ; 28(1): 62-73, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18662335

ABSTRACT

Cortical interneurons are born in the germinative zones of the ganglionic eminences in the subpallium, and migrate tangentially in spatially and temporally well-defined corridors into the neocortex. Because ephrin-A5 is expressed in the ventricular zone (VZ) of the ganglionic eminences at these developmental stages, we examined the possible effects of this molecule on interneuron migration. Double-immunocytochemistry of dissociated neurons from the medial ganglionic eminences (MGE) revealed that calbindin-positive cells express the EphA4-receptor. In situ, EphA4 is strongly expressed in the subventricular zone of the ganglionic eminences. Using different in vitro assays, we found that ephrin-A5 acts as a repellent cue for MGE neurons. We then examined interneuron migration in slice overlay experiments, where MGE-derived explants from enhanced green fluorescent protein-expressing transgenic mice were homotopically grafted into host slices from wild-type littermate embryos. In these in vitro preparations, interneurons recapitulated in vivo cell migration in several respects. However, interneurons in brain slices also migrated in the VZ of the ganglionic eminences, a region that is strictly avoided in vivo. In situ hybridizations revealed that ephrin-A5 became downregulated in the VZ in vitro. When recombinant ephrin-A5-Fc was added to the slices, it preferentially bound to the VZ, and migrating MGE neurons avoided the VZ as in vivo. The restoration of the normal migration pathway in slices required ephrin-A5 clustering and signalling of Src family kinases. Together, these experiments suggest that ephrin-A5 acts as an inhibitory flank that contributes to define the pathway of migrating interneurons.


Subject(s)
Cell Movement/physiology , Ephrin-A5/metabolism , Interneurons/physiology , Animals , Brain/anatomy & histology , Brain/metabolism , Coculture Techniques , Cues , Ephrin-A5/genetics , Female , Fibroblasts/cytology , Fibroblasts/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interneurons/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , Receptor, EphA4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...