Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Frontline Gastroenterol ; 15(3): 214-221, 2024 May.
Article in English | MEDLINE | ID: mdl-38665794

ABSTRACT

Objective: UK endoscopy training is delivered by trainers possessing well developed endoscopy and teaching skills to help learners perform high-quality endoscopy. Train The Trainer (TTT) courses are effective, but additional trainer support is variable with little formal quality assurance. We performed a survey to map UK endoscopy training, assess trainer perspectives on training delivery and identify factors that would enhance training. Design/Method: An online survey was designed by trainer representatives, in collaboration with the JAG training committee, and collected responses from trainers registered on JAG endoscopy training system e-portfolio from April to June 2022. Results: There were 1024 responses from all trainer disciplines, with 813 (79%) completing TTT courses and 584 (57%) having job planned dedicated training lists (DTLs). Clinical endoscopists most frequently had job-planned DTLs (71%), and DTLs occurring at least weekly (58%). 293 (29%) respondents participated as course faculty. Trainers reported high levels of pre-procedure preparation, effective dialogue and frequent feedback. The DOPS forms were 'always/often' completed by 81% of clinical endoscopists, 73% of gastroenterologist and 58% of surgeons. 435 (42%) trainers never had peer feedback. Responses suggested training could improve by protecting training time, attending courses, participating as faculty and receiving feedback from experienced trainers. Conclusion: This survey demonstrates substantial proportions of highly motivated UK trainers who value time spent teaching and learning how to teach. Skills taught on the TTT courses are often actively used in everyday training. Improved trainer course access, protected training time and formal use of existing feedback tools by peers were highlighted as measures that could support trainers' development.

2.
Frontline Gastroenterol ; 13(3): 193-205, 2022.
Article in English | MEDLINE | ID: mdl-35493618

ABSTRACT

Introduction: Training and quality assurance in oesophagogastroduodenoscopy (OGD) is important to ensure competent practice. A national evidence-based review was undertaken to update and develop standards and recommendations for OGD training and certification. Methods: Under the oversight of the Joint Advisory Group (JAG), a modified Delphi process was conducted with stakeholder representation from British Society of Gastroenterology, Association of Upper Gastrointestinal Surgeons, trainees and trainers. Recommendations on OGD training and certification were formulated following literature review and appraised using Grading of Recommendations Assessment, Development and Evaluation. These were subjected to electronic voting to achieve consensus. Accepted statements were incorporated into the updated certification pathway. Results: In total, 32 recommendation statements were generated for the following domains: definition of competence (4 statements), acquisition of competence (12 statements), assessment of competence (10 statements) and post-certification support (6 statements). The consensus process led to following certification criteria: (1) performing ≥250 hands-on procedures; (2) attending a JAG-accredited basic skills course; (3) attainment of relevant minimal performance standards defined by British Society of Gastroenterology/Association of Upper Gastrointestinal Surgeons of Great Britain and Ireland, (4) achieving physically unassisted D2 intubation and J-manoeuvre in ≥95% of recent procedures, (5) satisfactory performance in formative and summative direct observation of procedural skills assessments. Conclusion: The JAG standards for diagnostic OGD have been updated following evidence-based consensus. These standards are intended to support training, improve competency assessment to uphold standards of practice and provide support to the newly-independent practitioner.

3.
Frontline Gastroenterol ; 13(1): 39-44, 2022.
Article in English | MEDLINE | ID: mdl-34963796

ABSTRACT

OBJECTIVE: Training in gastrointestinal endoscopy in the UK occurs predominantly in a real world one-to-one trainer to trainee interaction. Previous surveys have shown surgical and gastroenterology trainees have had mixed experiences of supervision and training, and no surveys have explored specifically the role of trainee to trainer feedback. This study aimed to explore the experience of training and of providing trainer feedback for all disciplines of endoscopy trainees. DESIGN/METHOD: An online survey designed in collaboration with Joint Advisory Committee training committee and trainee representatives was distributed from January 2020 but was interrupted by the COVID-19 pandemic and hence terminated early. RESULTS: There were 129 responses, including trainees from all disciplines and regions, of which 86/129 (66.7%) rated the culture in their endoscopy units favourably-either good or excellent. 65/129 (50.4%) trainees reported having one or more training lists allocated per week, with 41/129 (31.8%) reporting only ad hoc lists. 100/129 (77.5%) respondents were given feedback and 97/129 (75.2%) were provided with learning points from the list. 65/129 (50.4%) respondents reported their trainer completed a direct observation of procedure or direct observation of polypectomies. 73/129 (56.6%) respondents reported that they felt able to give feedback to their trainer, with 88/129 (68.2%) feeling they could do this accurately. Barriers to trainer feedback cited included time constraints, lack of anonymity and concerns about affecting the trainer-trainee relationship. CONCLUSION: Overall, the training environment has improved since previous surveys. There are still issues around interdisciplinary differences with some surgical trainees finding the training environment less welcoming, and trainee perceptions of hierarchical barriers and trainer responsiveness to feedback limiting the accuracy of their feedback.

4.
Cell Rep ; 35(12): 109269, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34161768

ABSTRACT

Projection neuron subtype identities in the cerebral cortex are established by expressing pan-cortical and subtype-specific effector genes that execute terminal differentiation programs bestowing neurons with a glutamatergic neuron phenotype and subtype-specific morphology, physiology, and axonal projections. Whether pan-cortical glutamatergic and subtype-specific characteristics are regulated by the same genes or controlled by distinct programs remains largely unknown. Here, we show that FEZF2 functions as a transcriptional repressor, and it regulates subtype-specific identities of both corticothalamic and subcerebral neurons by selectively repressing expression of genes inappropriate for each neuronal subtype. We report that TLE4, specifically expressed in layer 6 corticothalamic neurons, is recruited by FEZF2 to inhibit layer 5 subcerebral neuronal genes. Together with previous studies, our results indicate that a cortical glutamatergic identity is specified by multiple parallel pathways active in progenitor cells, whereas projection neuron subtype-specific identity is achieved through selectively repressing genes associated with alternate identities in differentiating neurons.


Subject(s)
Cerebral Cortex/cytology , DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Transcription, Genetic , Alleles , Animals , Cell Differentiation/genetics , Electrophysiological Phenomena , Gene Expression Regulation , Mice, Knockout , Mitosis/genetics , Neurons/cytology , Protein Binding , Repressor Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 117(40): 25074-25084, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32948690

ABSTRACT

We are only just beginning to catalog the vast diversity of cell types in the cerebral cortex. Such categorization is a first step toward understanding how diversification relates to function. All cortical projection neurons arise from a uniform pool of progenitor cells that lines the ventricles of the forebrain. It is still unclear how these progenitor cells generate the more than 50 unique types of mature cortical projection neurons defined by their distinct gene-expression profiles. Moreover, exactly how and when neurons diversify their function during development is unknown. Here we relate gene expression and chromatin accessibility of two subclasses of projection neurons with divergent morphological and functional features as they develop in the mouse brain between embryonic day 13 and postnatal day 5 in order to identify transcriptional networks that diversify neuron cell fate. We compare these gene-expression profiles with published profiles of single cells isolated from similar populations and establish that layer-defined cell classes encompass cell subtypes and developmental trajectories identified using single-cell sequencing. Given the depth of our sequencing, we identify groups of transcription factors with particularly dense subclass-specific regulation and subclass-enriched transcription factor binding motifs. We also describe transcription factor-adjacent long noncoding RNAs that define each subclass and validate the function of Myt1l in balancing the ratio of the two subclasses in vitro. Our multidimensional approach supports an evolving model of progressive restriction of cell fate competence through inherited transcriptional identities.


Subject(s)
Nerve Tissue Proteins/genetics , Neurons/metabolism , Single-Cell Analysis , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental/genetics , Mice , RNA-Seq/methods
6.
Cereb Cortex ; 27(2): 1686-1699, 2017 02 01.
Article in English | MEDLINE | ID: mdl-26830346

ABSTRACT

Although many genes that specify neocortical projection neuron subtypes have been identified, the downstream effectors that control differentiation of those subtypes remain largely unknown. Here, we demonstrate that the LIM domain-binding proteins Ldb1 and Ldb2 exhibit dynamic and inversely correlated expression patterns during cerebral cortical development. Ldb1-deficient brains display severe defects in proliferation and changes in regionalization, phenotypes resembling those of Lhx mutants. Ldb2-deficient brains, on the other hand, exhibit striking phenotypes affecting layer 5 pyramidal neurons: Immature neurons have an impaired capacity to segregate into mature callosal and subcerebral projection neurons. The analysis of Ldb2 single-mutant mice reveals a compensatory role of Ldb1 for Ldb2 during corticospinal motor neuron (CSMN) differentiation. Animals lacking both Ldb1 and Ldb2 uncover the requirement for Ldb2 during CSMN differentiation, manifested as incomplete CSMN differentiation, and ultimately leading to a failure of the corticospinal tract.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/deficiency , Gene Expression Regulation, Developmental/physiology , LIM Domain Proteins/deficiency , Motor Neurons/metabolism , Pyramidal Tracts/metabolism , Transcription Factors/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Differentiation/physiology , Mice, Transgenic , Neurogenesis/physiology , Transcription Factors/metabolism
7.
Genome Res ; 26(8): 1013-22, 2016 08.
Article in English | MEDLINE | ID: mdl-27325115

ABSTRACT

Exome sequencing studies have identified multiple genes harboring de novo loss-of-function (LoF) variants in individuals with autism spectrum disorders (ASD), including TBR1, a master regulator of cortical development. We performed ChIP-seq for TBR1 during mouse cortical neurogenesis and show that TBR1-bound regions are enriched adjacent to ASD genes. ASD genes were also enriched among genes that are differentially expressed in Tbr1 knockouts, which together with the ChIP-seq data, suggests direct transcriptional regulation. Of the nine ASD genes examined, seven were misexpressed in the cortices of Tbr1 knockout mice, including six with increased expression in the deep cortical layers. ASD genes with adjacent cortical TBR1 ChIP-seq peaks also showed unusually low levels of LoF mutations in a reference human population and among Icelanders. We then leveraged TBR1 binding to identify an appealing subset of candidate ASD genes. Our findings highlight a TBR1-regulated network of ASD genes in the developing neocortex that are relatively intolerant to LoF mutations, indicating that these genes may play critical roles in normal cortical development.


Subject(s)
Autism Spectrum Disorder/genetics , DNA-Binding Proteins/genetics , Neocortex/physiopathology , Neurogenesis/genetics , Animals , Autism Spectrum Disorder/physiopathology , Disease Models, Animal , Exome/genetics , Gene Expression Regulation , Gene Knockout Techniques , Humans , Mice , Mutation , Neocortex/growth & development , Neurons/metabolism , Neurons/pathology , Risk Factors , T-Box Domain Proteins
8.
Cereb Cortex ; 25(10): 3406-19, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25037921

ABSTRACT

The chromatin-remodeling protein Satb2 plays a role in the generation of distinct subtypes of neocortical pyramidal neurons. Previous studies have shown that Satb2 is required for normal development of callosal projection neurons (CPNs), which fail to extend axons callosally in the absence of Satb2 and instead project subcortically. Here we conditionally delete Satb2 from the developing neocortex and find that neurons in the upper layers adopt some electrophysiological properties characteristic of deep layer neurons, but projections from the superficial layers do not contribute to the aberrant subcortical projections seen in Satb2 mutants. Instead, axons from deep layer CPNs descend subcortically in the absence of Satb2. These data demonstrate distinct developmental roles of Satb2 in regulating the fates of upper and deep layer neurons. Unexpectedly, Satb2 mutant brains also display changes in gene expression by subcerebral projection neurons (SCPNs), accompanied by a failure of corticospinal tract (CST) formation. Altering the timing of Satb2 ablation reveals that SCPNs require an early expression of Satb2 for differentiation and extension of the CST, suggesting that early transient expression of Satb2 in these cells plays an essential role in development. Collectively these data show that Satb2 is required by both CPNs and SCPNs for proper differentiation and axon pathfinding.


Subject(s)
Axons/physiology , Cell Differentiation , Cerebral Cortex/embryology , Corpus Callosum/embryology , Matrix Attachment Region Binding Proteins/physiology , Neurons/physiology , Transcription Factors/physiology , Animals , Axons/metabolism , Brain/embryology , Brain/metabolism , Cerebral Cortex/metabolism , Corpus Callosum/metabolism , Female , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice, Transgenic , Neural Pathways/embryology , Neural Pathways/metabolism , Neurons/metabolism , Somatosensory Cortex/embryology , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Mol Cell Neurosci ; 61: 85-96, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24867253

ABSTRACT

There are two main subgroups of midbrain dopaminergic (DA) neurons: the more medially located ventral tegmental area (VTA) DA neurons, which have axons that innervate the ventral-lateral (VL) striatum, and the more laterally located substantia nigra (SN) DA neurons, which preferentially degenerate in Parkinson's disease (PD) and have axons that project to the dorsal-medial (DM) striatum. DA axonal projections in the striatum are not discretely localized and they arborize widely, however they do not stray from one zone to the other so that VTA axons remain in the VL zone and SN axons in the DM zone. Here we provide evidence that Netrin-1 acts in a novel fashion to topographically pattern midbrain DA axons into these two striatal zones by means of a gradient of Netrin-1 in the striatum and by differential attraction of the axons to Netrin-1. Midbrain DA neurons are attracted to the striatum in culture and this attraction is blocked by an anti-DCC (Netrin receptor) antibody. Mechanistically, outgrowth of both VTA and SN DA axons is stimulated by Netrin-1, but the two populations of DA axons respond optimally to overlapping but distinct concentrations of Netrin-1, with SN axons preferring lower concentrations and VTA axons preferring higher concentrations. In vivo this differential preference is closely mirrored by differences in Netrin-1 expression in their respective striatal target fields. In vivo in mice lacking Netrin-1, DA axons that reach the striatum fail to segregate into two terminal zones and to fully innervate the striatum. Our results reveal novel actions for Netrin-1 and provide evidence for a mechanism through which DA axons can selectively innervate one of two terminal zones in the striatum but have free reign to arborize widely within a terminal zone.


Subject(s)
Axons/physiology , Corpus Striatum/cytology , Dopaminergic Neurons/physiology , Gene Expression Regulation, Developmental/genetics , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , Age Factors , Animals , COS Cells , Chickens , Chlorocebus aethiops , DCC Receptor , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Embryo, Mammalian , In Vitro Techniques , Mice , Mice, Inbred C57BL , Nerve Growth Factors/genetics , Netrin-1 , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tyrosine 3-Monooxygenase/metabolism
10.
Proc Natl Acad Sci U S A ; 109(47): 19071-8, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23144223

ABSTRACT

Neurons within each layer in the mammalian cortex have stereotypic projections. Four genes-Fezf2, Ctip2, Tbr1, and Satb2-regulate these projection identities. These genes also interact with each other, and it is unclear how these interactions shape the final projection identity. Here we show, by generating double mutants of Fezf2, Ctip2, and Satb2, that cortical neurons deploy a complex genetic switch that uses mutual repression to produce subcortical or callosal projections. We discovered that Tbr1, EphA4, and Unc5H3 are critical downstream targets of Satb2 in callosal fate specification. This represents a unique role for Tbr1, implicated previously in specifying corticothalamic projections. We further show that Tbr1 expression is dually regulated by Satb2 and Ctip2 in layers 2-5. Finally, we show that Satb2 and Fezf2 regulate two disease-related genes, Auts2 (Autistic Susceptibility Gene2) and Bhlhb5 (mutated in Hereditary Spastic Paraplegia), providing a molecular handle to investigate circuit disorders in neurodevelopmental diseases.


Subject(s)
Gene Regulatory Networks , Neocortex/growth & development , Neocortex/metabolism , Neurons/metabolism , Repressor Proteins/metabolism , Alkaline Phosphatase/metabolism , Animals , Axons/enzymology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/metabolism , Cytoskeletal Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Developmental , Genetic Loci/genetics , Isoenzymes/metabolism , Mice , Mutation/genetics , Nerve Tissue Proteins/metabolism , Netrin Receptors , Nuclear Proteins/metabolism , Protein Binding , Receptor, EphA4/metabolism , Receptors, Cell Surface/metabolism , Repressor Proteins/genetics , T-Box Domain Proteins , Thalamus/metabolism , Transcription Factors , Tumor Suppressor Proteins/metabolism
11.
Neuron ; 73(6): 1116-26, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22445340

ABSTRACT

VIDEO ABSTRACT: The precise connectivity of inputs and outputs is critical for cerebral cortex function; however, the cellular mechanisms that establish these connections are poorly understood. Here, we show that the secreted molecule Sonic Hedgehog (Shh) is involved in synapse formation of a specific cortical circuit. Shh is expressed in layer V corticofugal projection neurons and the Shh receptor, Brother of CDO (Boc), is expressed in local and callosal projection neurons of layer II/III that synapse onto the subcortical projection neurons. Layer V neurons of mice lacking functional Shh exhibit decreased synapses. Conversely, the loss of functional Boc leads to a reduction in the strength of synaptic connections onto layer Vb, but not layer II/III, pyramidal neurons. These results demonstrate that Shh is expressed in postsynaptic target cells while Boc is expressed in a complementary population of presynaptic input neurons, and they function to guide the formation of cortical microcircuitry.


Subject(s)
Cerebral Cortex/cytology , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Nerve Net/metabolism , Neurons/metabolism , Pyramidal Tracts/physiology , Age Factors , Animals , Animals, Newborn , Cerebral Cortex/growth & development , Channelrhodopsins , Corpus Callosum/cytology , Corpus Callosum/growth & development , DNA-Binding Proteins/metabolism , Dendritic Spines/metabolism , Dendritic Spines/physiology , Electric Stimulation , Electroporation/methods , Fluorobenzenes/metabolism , Functional Laterality/genetics , Furans/metabolism , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/genetics , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , In Vitro Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Membrane Potentials/genetics , Mice , Mice, Transgenic , Mutation/genetics , Nerve Net/cytology , Neurons/ultrastructure , Nuclear Proteins/metabolism , Patch-Clamp Techniques , Phosphopyruvate Hydratase/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Repressor Proteins/metabolism , Silver Staining/methods , Stilbamidines/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptophysin/genetics , Synaptophysin/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases , gamma-Aminobutyric Acid/metabolism
12.
Dev Neurobiol ; 72(9): 1196-212, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21976438

ABSTRACT

During neural development patterning, neurogenesis, and overall growth are highly regulated and coordinated between different brain regions. Here, we show that primary cilia and the regulation of Gli activity are necessary for the normal expansion of the cerebral cortex. We show that loss of Kif3a, an important functional component of primary cilia, leads to the degeneration of primary cilia, marked overgrowth of the cortex, and altered cell cycle kinetics within cortical progenitors. The G1 phase of the cell cycle is shortened through a mechanism likely involving reduced Gli3 activity and a resulting increase in expression of cyclin D1 and Fgf15. The defects in Gli3 activity alone are sufficient to accelerate cell cycle kinetics and cause the molecular changes seen in brains that lack cilia. Finally, we show that levels of full-length and repressor Gli3 proteins are tightly regulated during normal development and correlate with changes in expression of two known Shh-target genes, CyclinD1 and Fgf15, and with the normal lengthening of the cell cycle during corticogenesis. These data suggest that Gli3 activity is regulated through the primary cilium to control cell cycle length in the cortex and thus determine cortical size.


Subject(s)
Cerebral Cortex/growth & development , Kruppel-Like Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Animals , Cerebral Cortex/cytology , Cerebral Cortex/physiopathology , Cilia/physiology , Female , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nervous System Malformations/genetics , Nervous System Malformations/physiopathology , Neurogenesis/physiology , Organ Size/physiology , Zinc Finger Protein Gli3
13.
J Comp Neurol ; 519(10): 1829-46, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21452247

ABSTRACT

The murine olfactory system consists of main and accessory systems that perform distinct and overlapping functions. The main olfactory epithelium (MOE) is primarily involved in the detection of volatile odorants, while neurons in the vomeronasal organ (VNO), part of the accessory olfactory system, are important for pheromone detection. During development, the MOE and VNO both originate from the olfactory pit; however, the mechanisms regulating development of these anatomically distinct organs from a common olfactory primordium are unknown. Here we report that two closely related zinc-finger transcription factors, FEZF1 and FEZF2, regulate the identity of MOE sensory neurons and are essential for the survival of VNO neurons respectively. Fezf1 is predominantly expressed in the MOE while Fezf2 expression is restricted to the VNO. In Fezf1-deficient mice, olfactory neurons fail to mature and also express markers of functional VNO neurons. In Fezf2-deficient mice, VNO neurons degenerate prior to birth. These results identify Fezf1 and Fezf2 as important regulators of olfactory system development and sensory neuron identity.


Subject(s)
DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Olfactory Mucosa/physiology , Olfactory Pathways/physiology , Sensory Receptor Cells/physiology , Smell/physiology , Vomeronasal Organ/physiology , Amino Acid Sequence , Animals , Apoptosis , Cell Proliferation , DNA-Binding Proteins/genetics , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Olfactory Mucosa/anatomy & histology , Olfactory Pathways/anatomy & histology , Repressor Proteins , Sensory Receptor Cells/cytology , Sequence Alignment , Vomeronasal Organ/anatomy & histology
14.
PLoS One ; 6(3): e17802, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21445347

ABSTRACT

Newborn neurons migrate from their birthplace to their final location to form a properly functioning nervous system. During these movements, young neurons must attach and subsequently detach from their substrate to facilitate migration, but little is known about the mechanisms cells use to release their attachments. We show that the machinery for clathrin-mediated endocytosis is positioned to regulate the distribution of adhesion proteins in a subcellular region just proximal to the neuronal cell body. Inhibiting clathrin or dynamin function impedes the movement of migrating neurons both in vitro and in vivo. Inhibiting dynamin function in vitro shifts the distribution of adhesion proteins to the rear of the cell. These results suggest that endocytosis may play a critical role in regulating substrate detachment to enable cell body translocation in migrating neurons.


Subject(s)
Cell Adhesion , Endocytosis , Neurons/metabolism , Clathrin/physiology , Dynamins/physiology , Electroporation , Humans , Immunohistochemistry , Microscopy, Electron
15.
Mol Cell Neurosci ; 45(4): 324-34, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20637284

ABSTRACT

Dopaminergic neurons derived from human embryonic stem cells will be useful in future transplantation studies of Parkinson's disease patients. As newly generated neurons must integrate and reconnect with host cells, the ability of hESC-derived neurons to respond to axon guidance cues will be critical. Both Netrin-1 and Slit-2 guide rodent embryonic dopaminergic (DA) neurons in vitro and in vivo, but very little is known about the response of hESC-derived DA neurons to any axonal guidance cues. Here we examined the ability of Netrin-1 and Slit-2 to affect human ESC DA axons in vitro. hESC DA neurons mature over time in culture with the developmental profile of DA neurons in vivo, including expression of the DA neuron markers FoxA2, En-1 and Nurr-1, and receptors for both Netrin and Slit. hESC DA neurons respond to exogenous Netrin-1 and Slit-2, showing an increased responsiveness to Netrin-1 as the neurons mature in culture. These responses were maintained in the presence of pro-inflammatory cytokines that might be encountered in the diseased brain. These studies are the first to evaluate and confirm that suitably matured human ES-derived DA neurons can respond appropriately to axon guidance cues.


Subject(s)
Axons/ultrastructure , Embryonic Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Axons/metabolism , Cell Differentiation/physiology , Cell Line , Cues , Dopamine , Embryonic Stem Cells/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Humans , Neurons/metabolism
16.
Dev Neurobiol ; 70(9): 659-78, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20506362

ABSTRACT

Progenitor cells in the ventricular zone (VZ) and subventricular zone (SVZ) of the developing forebrain give rise to neurons and glial cells, and are characterized by distinct morphologies and proliferative behaviors. The mechanisms that distinguish VZ and SVZ progenitors are not well understood, although the homeodomain transcription factor Cux2 and Cyclin D2, a core component of the cell cycle machinery, are specifically involved in controlling SVZ cell proliferation. Rho GTPases have been implicated in regulating the proliferation, differentiation, and migration of many cell types, and one family member, Cdc42, affects the polarity and proliferation of radial glial cells in the VZ. Here, we show that another family member, Rac1, is required for the normal proliferation and differentiation of SVZ progenitors and for survival of both VZ and SVZ progenitors. A forebrain-specific loss of Rac1 leads to an SVZ-specific reduction in proliferation, a concomitant increase in cell cycle exit, and premature differentiation. In Rac1 mutants, the SVZ and VZ can no longer be delineated, but rather fuse to become a single compact zone of intermingled cells. Cyclin D2 expression, which is normally expressed by both VZ and SVZ progenitors, is reduced in Rac1 mutants, suggesting that the mutant cells differentiate precociously. Rac1-deficient mice can still generate SVZ-derived upper layer neurons, indicating that Rac1 is not required for the acquisition of upper layer neuronal fates, but instead is needed for the normal regulation of proliferation by progenitor cells in the SVZ.


Subject(s)
Cell Proliferation , Neurons/physiology , Neuropeptides/metabolism , Prosencephalon/embryology , Prosencephalon/physiology , Stem Cells/physiology , rac GTP-Binding Proteins/metabolism , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Cell Survival/physiology , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Cyclin D1/metabolism , Cyclin D2/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Neurogenesis/physiology , Neuropeptides/deficiency , Neuropeptides/genetics , Prosencephalon/pathology , Stem Cell Niche/embryology , Stem Cell Niche/pathology , Stem Cell Niche/physiology , rac GTP-Binding Proteins/deficiency , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
17.
Mol Cell Neurosci ; 41(4): 409-19, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19394428

ABSTRACT

Rac1 is a member of the Rho family of small GTPases that are important for structural aspects of the mature neuronal synapse including basal spine density and shape, activity-dependent spine enlargement, and AMPA receptor clustering in vitro. Here we demonstrate that selective elimination of Rac1 in excitatory neurons in the forebrain in vivo not only affects spine structure, but also impairs synaptic plasticity in the hippocampus with consequent defects in hippocampus-dependent spatial learning. Furthermore, Rac1 mutants display deficits in working/episodic-like memory in the delayed matching-to-place (DMP) task suggesting that Rac1 is a central regulator of rapid encoding of novel spatial information in vivo.


Subject(s)
Hippocampus/cytology , Learning/physiology , Memory/physiology , Neuronal Plasticity/physiology , Spatial Behavior/physiology , rac1 GTP-Binding Protein/physiology , Analysis of Variance , Animals , Biophysics/methods , Disks Large Homolog 4 Protein , Electric Stimulation/methods , Green Fluorescent Proteins/genetics , Guanylate Kinases , Hippocampus/physiology , Hippocampus/ultrastructure , Intracellular Signaling Peptides and Proteins/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neurons/physiology , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Reaction Time/genetics , beta-Galactosidase/metabolism , rac1 GTP-Binding Protein/deficiency
18.
Proc Natl Acad Sci U S A ; 105(32): 11382-7, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18678899

ABSTRACT

Pyramidal neurons in the deep layers of the cerebral cortex can be classified into two major classes: callosal projection neurons and long-range subcortical neurons. We and others have shown that a gene expressed specifically by subcortical projection neurons, Fezf2, is required for the formation of axonal projections to the spinal cord, tectum, and pons. Here, we report that Fezf2 regulates a decision between subcortical vs. callosal projection neuron fates. Fezf2(-/-) neurons adopt the fate of callosal projection neurons as assessed by their axonal projections, electrophysiological properties, and acquisition of Satb2 expression. Ctip2 is a major downstream effector of Fezf2 in regulating the extension of axons toward subcortical targets and can rescue the axonal phenotype of Fezf2 mutants. When ectopically expressed, either Fezf2 or Ctip2 can alter the axonal targeting of corticocortical projection neurons and cause them to project to subcortical targets, although Fezf2 can promote a subcortical projection neuron fate in the absence of Ctip2 expression.


Subject(s)
Axons/metabolism , DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Pyramidal Cells/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Phenotype , Pyramidal Cells/cytology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics
19.
Curr Opin Neurobiol ; 18(1): 28-35, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18508260

ABSTRACT

Here we review the mechanisms that determine projection neuron identity during cortical development. Pyramidal neurons in the mammalian cerebral cortex can be classified into two major classes: corticocortical projection neurons, which are concentrated in the upper layers of the cortex, and subcortical projection neurons, which are found in the deep layers. Early progenitor cells in the ventricular zone produce deep layer neurons that express transcription factors including Sox5, Fezf2, and Ctip2, which play important roles in the specification of subcortically projecting axons. Upper layer neurons are produced from progenitors in the subventricular zone, and the expression of Satb2 in these differentiating neurons is required for the formation of axonal projections that connect the two cerebral hemispheres. The Fezf2/Ctip2 and Satb2 pathways appear to be mutually repressive, thus ensuring that individual neurons adopt either a subcortical or callosal projection neuron identity at early times during development. The molecular mechanisms by which Satb2 regulates gene expression involves long-term epigenetic changes in chromatin configuration, which may enable cell fate decisions to be maintained during development.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental/genetics , Pyramidal Cells/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Cerebral Cortex/cytology , Efferent Pathways/cytology , Efferent Pathways/embryology , Efferent Pathways/metabolism , Humans , Phenotype , Pyramidal Cells/cytology , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Development ; 135(10): 1781-90, 2008 May.
Article in English | MEDLINE | ID: mdl-18403412

ABSTRACT

Apicobasal polarity plays an important role in regulating asymmetric cell divisions by neural progenitor cells (NPCs) in invertebrates, but the role of polarity in mammalian NPCs is poorly understood. Here, we characterize the function of the PDZ domain protein MALS-3 in the developing cerebral cortex. We find that MALS-3 is localized to the apical domain of NPCs. Mice lacking all three MALS genes fail to localize the polarity proteins PATJ and PALS1 apically in NPCs, whereas the formation and maintenance of adherens junctions appears normal. In the absence of MALS proteins, early NPCs progressed more slowly through the cell cycle, and their daughter cells were more likely to exit the cell cycle and differentiate into neurons. Interestingly, these effects were transient; NPCs recovered normal cell cycle properties during late neurogenesis. Experiments in which MALS-3 was targeted to the entire membrane resulted in a breakdown of apicobasal polarity, loss of adherens junctions, and a slowing of the cell cycle. Our results suggest that MALS-3 plays a role in maintaining apicobasal polarity and is required for normal neurogenesis in the developing cortex.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Polarity/physiology , Cerebral Cortex/embryology , Membrane Proteins/physiology , Neurons/physiology , Adaptor Proteins, Signal Transducing/genetics , Adherens Junctions/metabolism , Animals , Cell Cycle/physiology , Cell Differentiation , Cell Membrane/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Female , Mice , Mice, Knockout , Neocortex/cytology , Neocortex/embryology , Neocortex/metabolism , Neurons/cytology , Rats , Stem Cells/cytology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...