Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38701782

ABSTRACT

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Subject(s)
Epigenesis, Genetic , Myelin Sheath , Oligodendroglia , Remyelination , Animals , Myelin Sheath/metabolism , Humans , Mice , Remyelination/drug effects , Oligodendroglia/metabolism , Central Nervous System/metabolism , Mice, Inbred C57BL , Rejuvenation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Organoids/metabolism , Organoids/drug effects , Demyelinating Diseases/metabolism , Demyelinating Diseases/genetics , Cell Differentiation/drug effects , Small Molecule Libraries/pharmacology , Male , Regeneration/drug effects , Multiple Sclerosis/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
2.
Eur J Neurosci ; 56(12): 6099-6114, 2022 12.
Article in English | MEDLINE | ID: mdl-36217300

ABSTRACT

Oligodendrocyte production and myelination continues lifelong in the central nervous system (CNS), and all stages of this process can be adaptively regulated by neuronal activity. While artificial exogenous stimulation of neuronal circuits greatly enhances oligodendrocyte progenitor cell (OPC) production and increases myelination during development, the extent to which physiological stimuli replicates this is unclear, particularly in the adult CNS when the rate of new myelin addition slows. Here, we used environmental enrichment (EE) to physiologically stimulate neuronal activity for 6 weeks in 9-week-old C57BL/six male and female mice and found no increase in compact myelin in the corpus callosum or somatosensory cortex. Instead, we observed a global increase in callosal axon diameter with thicker myelin sheaths, elongated paranodes and shortened nodes of Ranvier. These findings indicate that EE induced the dynamic structural remodelling of myelinated axons. Additionally, we observed a global increase in the differentiation of OPCs and pre-myelinating oligodendroglia in the corpus callosum and somatosensory cortex. Our findings of structural remodelling of myelinated axons in response to physiological neural stimuli during young adulthood provide important insights in understanding experience-dependent myelin plasticity throughout the lifespan and provide a platform to investigate axon-myelin interactions in a physiologically relevant context.


Subject(s)
Axons , Myelin Sheath , Animals , Male , Female , Mice , Mice, Inbred C57BL , Axons/physiology , Oligodendroglia/physiology , Brain , Cell Differentiation/physiology
3.
Metabolites ; 12(6)2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35736487

ABSTRACT

Lipid metabolism is profoundly dysregulated in amyotrophic lateral sclerosis (ALS), yet the lipid composition of the white matter, where the myelinated axons of motor neurons are located, remains uncharacterised. We aimed to comprehensively characterise how myelin is altered in ALS by assessing its lipid and protein composition. We isolated white matter from the motor cortex from post-mortem tissue of ALS patients (n = 8 sporadic ALS cases and n = 6 familial ALS cases) and age- and sex-matched controls (n = 8) and conducted targeted lipidomic analyses, qPCR for gene expression of relevant lipid metabolising enzymes and Western blotting for myelin proteins. We also quantified myelin density by using spectral confocal reflectance microscopy (SCoRe). Whilst myelin protein composition was similar in ALS and control tissue, both the lipid levels and the expression of their corresponding enzymes were dysregulated, highlighting altered lipid metabolism in the white matter as well as a likely change in myelin composition. Altered myelin composition could contribute to motor neuron dysfunction, and this highlights how oligodendrocytes may play a critical role in ALS pathogenesis.

4.
J Neurosci Res ; 98(10): 1987-1998, 2020 10.
Article in English | MEDLINE | ID: mdl-32585763

ABSTRACT

The p75 neurotrophin receptor (p75NTR ) is required for maintaining peripheral sensory neuron survival and function; however, the underlying cellular mechanism remains unclear. The general view is that expression of p75NTR by the neuron itself is required for maintaining sensory neuron survival and myelination in the peripheral nervous system (PNS). Adopting a neuronal-specific conditional knockout strategy, we demonstrate the partial depletion of p75NTR in neurons exerts little influence upon maintaining sensory neuron survival and peripheral nerve myelination in health and after demyelinating neuropathy. Our data show that the density and total number of dorsal root ganglion (DRG) neurons in 2-month-old mice is not affected following the deletion of p75NTR in large-diameter myelinating neurons, as assessed by stereology. Adopting experimental autoimmune neuritis induced in adult male mice, an animal model of demyelinating peripheral neuropathy, we identify that deleting p75NTR in myelinating neurons exerts no influence upon the disease progression, the total number of DRG neurons, and the extent of myelin damage in the sciatic nerve, indicating that the expression of neuronal p75NTR is not essential for maintaining peripheral neuron survival and myelination after a demyelinating insult in vivo. Together, results of this study suggest that the survival and myelination of peripheral sensory neurons is independent of p75NTR expressed by a subtype of neurons in vivo. Thus, our findings provide new insights into the mechanism underpinning p75NTR -mediated neuronal survival in the PNS.


Subject(s)
Ganglia, Spinal/metabolism , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Sensory Receptor Cells/metabolism , Animals , Cell Survival/physiology , Female , Gene Deletion , Male , Mice , Mice, Knockout , Mice, Transgenic
5.
Front Mol Neurosci ; 12: 275, 2019.
Article in English | MEDLINE | ID: mdl-31803018

ABSTRACT

Developing a high-throughput approach to quantify the extent of myelin integrity in preclinical models of demyelinating diseases will enhance our capacity to identify novel therapies for myelin repair. In light of the technical limitations of electron microscopy and immunohistochemical analyses of myelination, we have utilized a novel imaging technique, spectral confocal reflectance (SCoRe) microscopy. SCoRe takes advantage of the optically reflective properties of compact myelin, allowing the integrity of compact myelin to be quantified over the course of the cuprizone-induced model of central demyelination. We applied SCoRe imaging on fixed frozen brain sections. SCoRe analysis of control mice identified an increase in corpus callosum myelination during the period of cuprizone administration and recovery, suggesting that the normal developmental processes of myelination are ongoing at this time. Importantly, analysis of mice subjected to cuprizone identified a significant reduction in compact myelin in both rostral and caudal corpus callosum compared to age-matched control mice. SCoRe microscopy also allowed the visualization and quantification of the amount of myelin debris in demyelinating lesions. Combining SCoRe imaging with immunohistochemistry, we quantified the amount of myelin debris within IBA-1+ microglia and found that 11% of myelin debris colocalized in microglia irrespective of the callosal regions, with the vast majority of debris outside of microglia. In summary, we have demonstrated that SCoRe microscopy is an effective and powerful tool to perform both quantitative and qualitative analyses of compact myelin integrity in health or after injury in vivo, demonstrating its future application in high-throughput assessments and screening of the therapeutic efficacy of myelin repair therapies in preclinical animal models of demyelinating diseases.

6.
eNeuro ; 6(2)2019.
Article in English | MEDLINE | ID: mdl-31028086

ABSTRACT

Blocking inhibitory factors within CNS demyelinating lesions is regarded as a promising strategy to promote remyelination. Bone morphogenetic protein 4 (BMP4) is an inhibitory factor present in demyelinating lesions. Noggin, an endogenous antagonist to BMP, has previously been shown to increase the number of oligodendrocytes and promote remyelination in vivo. However, it remains unclear how BMP4 signaling inhibits remyelination. Here we investigated the downstream signaling pathway that mediates the inhibitory effect that BMP4 exerts upon remyelination through pharmacological and transgenic approaches. Using the cuprizone mouse model of central demyelination, we demonstrate that selectively blocking BMP4 signaling via the pharmacological inhibitor LDN-193189 significantly promotes oligodendroglial differentiation and the extent of remyelination in vivo This was accompanied by the downregulation of transcriptional targets that suppress oligodendrocyte differentiation. Further, selective deletion of BMP receptor type IA (BMPRIA) within primary mouse oligodendrocyte progenitor cells (OPCs) significantly enhanced their differentiation and subsequent myelination in vitro Together, the results of this study identify that BMP4 signals via BMPRIA within OPCs to inhibit oligodendroglial differentiation and their capacity to myelinate axons, and suggest that blocking the BMP4/BMPRIA pathway in OPCs is a promising strategy to promote CNS remyelination.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Demyelinating Diseases/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Remyelination , Signal Transduction , Animals , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type I/deficiency , Cell Differentiation/drug effects , Demyelinating Diseases/drug therapy , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/drug effects , Oligodendroglia/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Remyelination/drug effects
7.
J Neurochem ; 148(4): 447-461, 2019 02.
Article in English | MEDLINE | ID: mdl-30225984

ABSTRACT

Myelin, the multilayered membrane surrounding many axons in the nervous system, increases the speed by which electrical signals travel along axons and facilitates neuronal communication between distant regions of the nervous system. However, how neuronal signals influence the myelinating process in the CNS is still largely unclear. Recent studies have significantly advanced this understanding, identifying important roles for neuronal activity in controlling oligodendrocyte development and their capacity of producing myelin in both developing and mature CNS. Here, we review these recent advances, and discuss potential mechanisms underpinning activity-dependent myelination and how remyelination may be stimulated via manipulating axonal activity, raising new questions for future research.


Subject(s)
Central Nervous System , Myelin Sheath , Neurogenesis , Oligodendroglia , Animals , Cell Differentiation/physiology , Humans
8.
J Neurosci ; 38(32): 7088-7099, 2018 08 08.
Article in English | MEDLINE | ID: mdl-29976621

ABSTRACT

Methods to promote myelin regeneration in response to central myelin loss are essential to prevent the progression of clinical disability in demyelinating diseases. The neurotrophin brain-derived neurotrophic factor (BDNF) is known to promote myelination during development via oligodendrocyte expressed TrkB receptors. Here, we use a structural mimetic of BDNF to promote myelin regeneration in a preclinical mouse model of central demyelination. In female mice, we show that selective targeting of TrkB with the BDNF-mimetic enhances remyelination, increasing oligodendrocyte differentiation, the frequency of myelinated axons, and myelin sheath thickness after a demyelinating insult. Treatment with exogenous BDNF exerted an attenuated effect, increasing myelin sheath thickness only. Further, following conditional deletion of TrkB from premyelinating oligodendrocytes, we show the effects of the BDNF-mimetic on oligodendrocyte differentiation and remyelination are lost, indicating these are dependent on oligodendrocyte expression of TrkB. Overall, these studies demonstrate that targeting oligodendrocyte TrkB promotes in vivo remyelination in the brain.SIGNIFICANCE STATEMENT Novel strategies to promote myelin regeneration are required to prevent progressive neurodegeneration and clinical disability in patients with central demyelinating disease. Here, we test whether selectively targeting the TrkB receptor on the myelin-producing oligodendrocytes, can promote remyelination in the brain. Using a structural mimetic of its native ligand, BDNF, we show that stimulation of TrkB enhances remyelination, increasing oligodendrocyte differentiation, the frequency of myelinated axons and thickness of the myelin sheath following a demyelinating insult. Further, we show that these effects are dependent on the phosphorylation of oligodendrocyte expressed TrkB receptors in vivo Overall, we demonstrate that selective targeting of TrkB has therapeutic potential to promote remyelination in the brain.


Subject(s)
Brain-Derived Neurotrophic Factor/therapeutic use , Brain/drug effects , Demyelinating Diseases/drug therapy , Membrane Glycoproteins/agonists , Molecular Targeted Therapy , Myelin Sheath/metabolism , Oligodendroglia/drug effects , Remyelination/drug effects , Animals , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/pharmacology , Cell Division/drug effects , Corpus Callosum/metabolism , Corpus Callosum/pathology , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Female , Infusion Pumps, Implantable , Infusions, Intraventricular , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/biosynthesis , Neural Stem Cells/drug effects , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Phosphorylation , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/metabolism , Specific Pathogen-Free Organisms
9.
Methods Mol Biol ; 1791: 243-250, 2018.
Article in English | MEDLINE | ID: mdl-30006715

ABSTRACT

Mouse models of peripheral demyelinating neuropathy play an important role in enabling the study of disease pathogenesis. Further, induction in transgenic mice allows for the precise interrogation of disease mechanisms, as well as the analysis of the efficacy and mechanisms of potential new therapies. Here we describe a method to successfully induce experimental autoimmune neuritis (EAN) using myelin protein zero (P0)180-199 peptide in combination with Freund's complete adjuvant and pertussis toxin in the C57BL/6 mouse strain. We also outline a sensitive paradigm of accurately assessing the extent of functional deficits occurring in murine EAN.


Subject(s)
Myelin P0 Protein/immunology , Neuritis, Autoimmune, Experimental/immunology , Peptide Fragments/immunology , Amino Acid Sequence , Animals , Disease Models, Animal , Gait , Mice , Mice, Inbred C57BL , Motor Activity , Myelin P0 Protein/chemistry , Myelin P0 Protein/metabolism , Neuritis, Autoimmune, Experimental/diagnosis , Neuritis, Autoimmune, Experimental/metabolism , Neuritis, Autoimmune, Experimental/physiopathology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Pertussis Toxin/administration & dosage , Pertussis Toxin/adverse effects , Pertussis Toxin/immunology
10.
Cell Tissue Res ; 372(2): 171-193, 2018 05.
Article in English | MEDLINE | ID: mdl-28971249

ABSTRACT

The sympathetic division of the autonomic nervous system includes a variety of cells including neurons, endocrine cells and glial cells. A recent study (Furlan et al. 2017) has revised thinking about the developmental origin of these cells. It now appears that sympathetic neurons and chromaffin cells of the adrenal medulla do not have an immediate common ancestor in the form a "sympathoadrenal cell", as has been long believed. Instead, chromaffin cells arise from Schwann cell precursors. This review integrates the new findings with the expanding body of knowledge on the signalling pathways and transcription factors that regulate the origin of cells of the sympathetic division of the autonomic nervous system.


Subject(s)
Signal Transduction , Sympathetic Nervous System/cytology , Animals , Cell Differentiation , Cell Proliferation , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Humans , Neural Crest/cytology , Neural Crest/metabolism
11.
J Vis Exp ; (129)2017 11 09.
Article in English | MEDLINE | ID: mdl-29155769

ABSTRACT

Experimental autoimmune neuritis (EAN) is a well-appreciated experimental model of autoimmune peripheral demyelinating diseases. EAN disease is induced by immunizing mice with neurogenic peptides to direct an inflammatory attack toward components of the peripheral nervous system (PNS). Recent advances have enabled the induction of EAN in the relatively resistant C57BL/6 mouse line using myelin protein zero (P0)106-125 or P0180-199 peptides delivered in adjuvant combined with the injection of pertussis toxin. The ability to induce EAN in the C57BL/6 strain allows for the use of the numerous genetic tools that exist on this mouse background, and thus allows the sophisticated study of disease pathogenesis and interrogation of the mechanistic action of novel therapeutics in combination with transgenic approaches. In this study, we demonstrate a simple approach to successfully induce EAN using the P0180-199 peptide in C57BL/6 mice. We also outline a protocol for the assessment of functional deficits that occur in this model, accompanied by an array of neuropathological features. Thus, this model is a powerful experimental model to study the pathogenesis of human peripheral demyelinating neuropathies, and to determine the efficacy of potential therapies that aim to promote myelin repair and protect against nerve damage in autoimmune neuritis.


Subject(s)
Neuritis, Autoimmune, Experimental/chemically induced , Animals , Humans , Male , Mice , Mice, Inbred C57BL
12.
eNeuro ; 4(3)2017.
Article in English | MEDLINE | ID: mdl-28680965

ABSTRACT

Axonal damage and demyelination are major determinants of disability in patients with peripheral demyelinating neuropathies. The neurotrophin family of growth factors are essential for the normal development and myelination of the peripheral nervous system (PNS), and as such are potential therapeutic candidates for ameliorating axonal and myelin damage. In particular, BDNF promotes peripheral nerve myelination via p75 neurotrophin receptor (p75NTR) receptors. Here, we investigated the therapeutic efficacy of a small structural mimetic of the region of BDNF that binds to p75NTR (cyclo-dPAKKR) in experimental autoimmune neuritis (EAN), an established animal model of peripheral demyelinating neuropathy. Examination of rodents induced with EAN revealed that p75NTR is abundantly expressed in affected peripheral nerves. We found that systemic administration of cyclo-dPAKKR ameliorates EAN disease severity and accelerates recovery. Animals treated with cyclo-dPAKKR displayed significantly better motor performance compared to control animals. Histological assessment revealed that cyclo-dPAKKR administration limits the extent of inflammatory demyelination and axonal damage, and protects against the disruption of nodal architecture in affected peripheral nerves. In contrast, a structural control peptide of cyclo-dPAKKR exerted no influence. Moreover, all the beneficial effects of cyclo-dPAKKR in EAN are abrogated in p75NTR heterozygous mice, strongly suggesting a p75NTR-dependent effect. Taken together, our data demonstrate that cyclo-dPAKKR ameliorates functional and pathological defects of EAN in a p75NTR-dependant manner, suggesting that p75NTR is a therapeutic target to consider for future treatment of peripheral demyelinating diseases and targeting of p75NTR is a strategy worthy of further investigation.


Subject(s)
Axons/drug effects , Demyelinating Diseases/drug therapy , Demyelinating Diseases/etiology , Neuritis, Autoimmune, Experimental , Oligopeptides/therapeutic use , Receptors, Nerve Growth Factor/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Axons/pathology , Axons/ultrastructure , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , Myelin Basic Protein/metabolism , Neuritis, Autoimmune, Experimental/complications , Neuritis, Autoimmune, Experimental/genetics , Neuritis, Autoimmune, Experimental/pathology , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/therapeutic use , Statistics, Nonparametric
13.
J Neuropathol Exp Neurol ; 76(2): 89-100, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28082327

ABSTRACT

We assessed novel disability-based parameters and neuropathological features of the P0180-190 peptide-induced model of experimental autoimmune neuritis (EAN) in C57BL/6 mice. We show that functional assessments such as running capacity provide a more sensitive method for detecting alterations in disease severity than a classical clinical scoring paradigm. We performed detailed ultrastructural analysis and show for the first time that tomaculous neuropathy is a neuropathological feature of this disease model. In addition, we demonstrate that ultrastructural assessments of myelin pathology are sufficiently sensitive to detect significant differences in both mean G-ratio and mean axon diameter between mice with EAN induced with different doses of pertussis toxin. In summary, we have established a comprehensive assessment paradigm for discriminating variations in disease severity and the extent of myelin pathology in this model. Our findings indicate that this model is a powerful tool to study the pathogenesis of human peripheral demyelinating neuropathies and that this assessment paradigm could be used to determine the efficacy of potential therapies that aim to promote myelin repair and protect against nerve damage in autoimmune neuritides.


Subject(s)
Gait/physiology , Neuritis, Autoimmune, Experimental/pathology , Neuritis, Autoimmune, Experimental/physiopathology , Peptide Fragments/toxicity , Recovery of Function/physiology , Running/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Neuritis, Autoimmune, Experimental/chemically induced
14.
Dev Neurobiol ; 76(2): 137-49, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25989220

ABSTRACT

Adrenal medullary chromaffin cells and peripheral sympathetic neurons originate from a common sympathoadrenal (SA) progenitor cell. The timing and phenotypic changes that mark this lineage diversification are not fully understood. The present study investigated the expression patterns of phenotypic markers, and cell cycle dynamics, in the adrenal medulla and the neighboring suprarenal ganglion of embryonic mice. The noradrenergic marker, tyrosine hydroxylase (TH), was detected in both presumptive adrenal medulla and sympathetic ganglion cells, but with significantly stronger immunostaining in the former. There was intense cocaine and amphetamine-regulated transcript (CART) peptide immunostaining in most neuroblasts, whereas very few adrenal chromaffin cells showed detectable CART immunostaining. This phenotypic segregation appeared as early as E12.5, before anatomical segregation of the two cell types. Cell cycle dynamics were also examined. Initially, 88% of Sox10 positive (+) neural crest progenitors were proliferating at E10.5. Many SA progenitor cells withdrew from the cell cycle at E11.5 as they started to express TH. Whereas 70% of neuroblasts (TH+/CART+ cells) were back in the cell cycle at E12.5, only around 20% of chromaffin (CART negative) cells were in the cell cycle at E12.5 and subsequent days. Thus, chromaffin cell and neuroblast lineages showed differences in proliferative behavior from their earliest appearance. We conclude that the intensity of TH immunostaining and the expression of CART permit early discrimination of chromaffin cells and sympathetic neuroblasts, and that developing chromaffin cells exhibit significantly lower proliferative activity relative to sympathetic neuroblasts.


Subject(s)
Adrenal Medulla/cytology , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Lineage/physiology , Chromaffin Cells/cytology , Neurons/cytology , Animals , Female , Mice , Neural Crest/cytology , Neural Crest/embryology , Neural Stem Cells/metabolism , Pregnancy , Transcription Factors/metabolism
15.
BMC Med Educ ; 15: 231, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26698839

ABSTRACT

BACKGROUND: Anatomy in medical curricula is typically taught via pedagogy consisting of didactic lectures combined with a practical component. The practical component often includes traditional cadaveric dissection classes and/or workshops utilizing anatomical models, carefully prosected cadaveric material and radiology. The primary aim of this study was to determine if there is an association between attendance at practical classes in anatomy and student assessment outcomes. A secondary aim was to determine if student assessment outcomes were better when students preferentially attended workshops or prosection style practical classes. METHOD: We retrospectively examined practical attendance records and assessment outcomes from a single large anatomy subject (approx. 450 students) to identify how attendance at anatomy practical classes correlates with assessment outcome. RESULTS: Students who scored above the median mark for each assessment attended significantly more practical classes than students who scored below the median assessment mark (Mann Whitney; p < 0.001), and students who attended more than half the practical classes had significantly higher scores on assessments than students that attended less than half the practical classes (Mann Whitney; P < 0.01). There was a statistically significant positive correlation between attendance at practical classes and outcomes for each assessment (Spearman's correlation; p < 0.01). There was no difference in assessment outcomes for students who preferentially attended more dissection compared to prosection style classes and vice versa (Mann Whitney; p > 0.05). CONCLUSIONS: Our findings show there is an association between student attendance at practical classes and performance on anatomy assessment.


Subject(s)
Anatomy/education , Dissection/education , Education, Medical/methods , Problem-Based Learning/organization & administration , Students, Medical/psychology , Cadaver , Choice Behavior , Education, Medical/organization & administration , Educational Measurement , Humans , Problem-Based Learning/methods , Retrospective Studies , Statistics, Nonparametric , Students, Medical/statistics & numerical data , Teaching/methods , Victoria
16.
Dev Neurobiol ; 75(3): 287-301, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25205394

ABSTRACT

The rate of proliferation of cells depends on the proportion of cycling cells and the frequency of cell division. Here, we describe in detail methods for quantifying the proliferative behavior of specific cell types in situ, and use the method to examine cell cycle dynamics in two neural crest derivatives--dorsal root ganglia (DRG) using frozen sections, and the enteric nervous system (ENS) using wholemount preparations. In DRG, our data reveal a significant increase in cell cycle length and a decrease in the number of cycling Sox10+ progenitor cells at E12.5-E13.5, which coincides with the commencement of glial cell generation. In the ENS, the vast majority of Sox10+ cells remain proliferative during embryonic development, with only relatively minor changes in cell cycle parameters. Previous studies have identified proliferating cells expressing neuronal markers in the developing ENS; our data suggest that most cells undergoing neuronal differentiation in the developing gut commence expression of neuronal markers during G2 phase of their last division. Combined with previous studies, our findings show that different populations of neural crest-derived cells show tissue-specific patterns of proliferation.


Subject(s)
Cell Proliferation/physiology , Enteric Nervous System/cytology , Ganglia, Spinal/cytology , Neural Crest/cytology , Neurogenesis/physiology , Animals , Biomarkers/metabolism , Cell Cycle/physiology , Enteric Nervous System/embryology , Ganglia, Spinal/embryology , Mice , Mice, Inbred C57BL , Neural Crest/embryology , SOXE Transcription Factors/metabolism
17.
J Comp Neurol ; 522(3): 514-27, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-23861145

ABSTRACT

There are many different types of enteric neurons. Previous studies have identified the time at which some enteric neuron subtypes are born (exit the cell cycle) in the mouse, but the birthdates of some major enteric neuron subtypes are still incompletely characterized or unknown. We combined 5-ethynynl-2'-deoxyuridine (EdU) labeling with antibody markers that identify myenteric neuron subtypes to determine when neuron subtypes are born in the mouse small intestine. We found that different neurochemical classes of enteric neuron differed in their birthdates; serotonin neurons were born first with peak cell cycle exit at E11.5, followed by neurofilament-M neurons, calcitonin gene-related peptide neurons (peak cell cycle exit for both at embryonic day [E]12.5-E13.5), tyrosine hydroxylase neurons (E15.5), nitric oxide synthase 1 (NOS1) neurons (E15.5), and calretinin neurons (postnatal day [P]0). The vast majority of myenteric neurons had exited the cell cycle by P10. We did not observe any EdU+/NOS1+ myenteric neurons in the small intestine of adult mice following EdU injection at E10.5 or E11.5, which was unexpected, as previous studies have shown that NOS1 neurons are present in E11.5 mice. Studies using the proliferation marker Ki67 revealed that very few NOS1 neurons in the E11.5 and E12.5 gut were proliferating. However, Cre-lox-based genetic fate-mapping revealed a small subpopulation of myenteric neurons that appears to express NOS1 only transiently. Together, our results confirm a relationship between enteric neuron subtype and birthdate, and suggest that some enteric neurons exhibit neurochemical phenotypes during development that are different from their mature phenotype.


Subject(s)
Intestine, Small , Myenteric Plexus/cytology , Neurons/classification , Neurons/physiology , Age Factors , Animals , Animals, Newborn , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intestine, Small/cytology , Intestine, Small/embryology , Intestine, Small/growth & development , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myenteric Plexus/embryology , Myenteric Plexus/growth & development , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Phenylurea Compounds/metabolism , Pregnancy , Serotonin/metabolism , Tyrosine 3-Monooxygenase/metabolism
18.
J Neurosci ; 33(14): 5969-79, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23554478

ABSTRACT

Cell proliferation during nervous system development is poorly understood outside the mouse neocortex. We measured cell cycle dynamics in the embryonic mouse sympathetic stellate ganglion, where neuroblasts continue to proliferate following neuronal differentiation. At embryonic day (E) 9.5, when neural crest-derived cells were migrating and coalescing into the ganglion primordium, all cells were cycling, cell cycle length was only 10.6 h, and S-phase comprised over 65% of the cell cycle; these values are similar to those previously reported for embryonic stem cells. At E10.5, Sox10(+) cells lengthened their cell cycle to 38 h and reduced the length of S-phase. As cells started to express the neuronal markers Tuj1 and tyrosine hydroxylase (TH) at E10.5, they exited the cell cycle. At E11.5, when >80% of cells in the ganglion were Tuj1(+)/TH(+) neuroblasts, all cells were again cycling. Neuroblast cell cycle length did not change significantly after E11.5, and 98% of Sox10(-)/TH(+) cells had exited the cell cycle by E18.5. The cell cycle length of Sox10(+)/TH(-) cells increased during late embryonic development, and ∼25% were still cycling at E18.5. Loss of Ret increased neuroblast cell cycle length at E16.5 and decreased the number of neuroblasts at E18.5. A mathematical model generated from our data successfully predicted the relative change in proportions of neuroblasts and non-neuroblasts in wild-type mice. Our results show that, like other neurons, sympathetic neuron differentiation is associated with exit from the cell cycle; sympathetic neurons are unusual in that they then re-enter the cell cycle before later permanently exiting.


Subject(s)
Cell Cycle/physiology , Cell Proliferation , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Neurons/physiology , Stellate Ganglion/cytology , Stellate Ganglion/embryology , Transcription Factors/genetics , Age Factors , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Cycle/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phenotype , Phenylurea Compounds/metabolism , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , SOXE Transcription Factors/metabolism , Time Factors , Transcription Factors/metabolism , Tubulin/metabolism , Tyrosine 3-Monooxygenase/metabolism
19.
J Neurosci ; 30(35): 11781-91, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20810898

ABSTRACT

Cocaine and amphetamine-regulated transcript peptide (CART) is present in a subset of sympathetic preganglionic neurons in the rat. We examined the distribution of CART-immunoreactive terminals in rat stellate and superior cervical ganglia and adrenal gland and found that they surround neuropeptide Y-immunoreactive postganglionic neurons and noradrenergic chromaffin cells. The targets of CART-immunoreactive preganglionic neurons in the stellate and superior cervical ganglia were shown to be vasoconstrictor neurons supplying muscle and skin and cardiac-projecting postganglionic neurons: they did not target non-vasoconstrictor neurons innervating salivary glands, piloerector muscle, brown fat, or adrenergic chromaffin cells. Transneuronal tracing using pseudorabies virus demonstrated that many, but not all, preganglionic neurons in the vasoconstrictor pathway to forelimb skeletal muscle were CART immunoreactive. Similarly, analysis with the confocal microscope confirmed that 70% of boutons in contact with vasoconstrictor ganglion cells contained CART, whereas 30% did not. Finally, we show that CART-immunoreactive cells represented 69% of the preganglionic neuron population expressing c-Fos after systemic hypoxia. We conclude that CART is present in most, although not all, cardiovascular preganglionic neurons but not thoracic preganglionic neurons with non-cardiovascular targets. We suggest that CART immunoreactivity may identify the postulated "accessory" preganglionic neurons, whose actions may amplify vasomotor ganglionic transmission.


Subject(s)
Autonomic Fibers, Preganglionic/chemistry , Cardiovascular System/chemistry , Nerve Tissue Proteins/physiology , Neurons/chemistry , Sympathetic Fibers, Postganglionic/chemistry , Adrenergic Fibers/chemistry , Adrenergic Fibers/physiology , Animals , Autonomic Fibers, Preganglionic/physiology , Cardiovascular System/cytology , Female , Immunohistochemistry , Male , Nerve Tissue Proteins/analysis , Neurons/classification , Neurons/physiology , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/chemistry , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/physiology , Sympathetic Fibers, Postganglionic/physiology , Vasoconstriction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...