Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849524

ABSTRACT

In the mouse embryonic forebrain, developmentally distinct oligodendrocyte progenitor cell populations and their progeny, oligodendrocytes, emerge from three distinct regions in a spatiotemporal gradient from ventral to dorsal. However, the functional importance of this oligodendrocyte developmental heterogeneity is unknown. Using a genetic strategy to ablate dorsally derived oligodendrocyte lineage cells (OLCs), we show here that the areas in which dorsally derived OLCs normally reside in the adult central nervous system become populated and myelinated by OLCs of ventral origin. These ectopic oligodendrocytes (eOLs) have a distinctive gene expression profile as well as subtle myelination abnormalities. The failure of eOLs to fully assume the role of the original dorsally derived cells results in locomotor and cognitive deficits in the adult animal. This study reveals the importance of developmental heterogeneity within the oligodendrocyte lineage and its importance for homeostatic brain function.

2.
Curr Opin Neurobiol ; 80: 102703, 2023 06.
Article in English | MEDLINE | ID: mdl-36933450

ABSTRACT

Inhibition in the mammalian cerebral cortex is mediated by a small population of highly diverse GABAergic interneurons. These largely local neurons are interspersed among excitatory projection neurons and exert pivotal regulation on the formation and function of cortical circuits. We are beginning to understand the extent of GABAergic neuron diversity and how this is generated and shaped during brain development in mice and humans. In this review, we summarise recent findings and discuss how new technologies are being used to further advance our knowledge. Understanding how inhibitory neurons are generated in the embryo is an essential pre-requisite of stem cell therapy, an evolving area of research, aimed at correcting human disorders that result in inhibitory dysfunction.


Subject(s)
Big Data , Interneurons , Mice , Animals , Humans , Mice, Transgenic , Interneurons/physiology , Cerebral Cortex/physiology , GABAergic Neurons/physiology , Mammals
3.
Commun Biol ; 5(1): 1137, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36302841

ABSTRACT

The septum is a key structure at the core of the forebrain that integrates inputs and relays information to other brain areas to support cognition and behaviours such as feeding and locomotion. Underlying these functions is a rich diversity of neuronal types and an intricate complexity of wiring across and within the septal region. We currently have very little understanding of how septal neuronal diversity emerges during development. Using transgenic mice expressing Cre in different subsets of telencephalic precursors we explored the origins of the three main neuronal types of the septal complex: GABAergic, cholinergic and glutamatergic neurons. We find that septal neurons originate from distinct neuroepithelial domains of the developing septum and are born at different embryonic time points. An exception to this is the GABAergic medial septal Parvalbumin-expressing population which is generated outside the septum from surrounding germinal zones. We identify the transcription factor BSX as being expressed in the developing glutamatergic neuron population. Embryonic elimination of BSX in the septum results in a reduction of septal glutamatergic cell numbers and a consequent deficit in locomotion. Further refinement of septal neuron diversity is needed to understand the multiple roles of septal neurons and their contribution to distinct behaviours.


Subject(s)
Neurons , Parvalbumins , Mice , Animals , Neurons/physiology , Prosencephalon , Mice, Transgenic
4.
Nat Commun ; 13(1): 5217, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36064547

ABSTRACT

Cortical interneurons originating in the embryonic medial ganglionic eminence (MGE) diverge into a range of different subtypes found in the adult mouse cerebral cortex. The mechanisms underlying this divergence and the timing when subtype identity is set up remain unclear. We identify the highly conserved transcriptional co-factor MTG8 as being pivotal in the development of a large subset of MGE cortical interneurons that co-expresses Somatostatin (SST) and Neuropeptide Y (NPY). MTG8 interacts with the pan-MGE transcription factor LHX6 and together the two factors are sufficient to promote expression of critical cortical interneuron subtype identity genes. The SST-NPY cortical interneuron fate is initiated early, well before interneurons migrate into the cortex, demonstrating an early onset specification program. Our findings suggest that transcriptional co-factors and modifiers of generic lineage specification programs may hold the key to the emergence of cortical interneuron heterogeneity from the embryonic telencephalic germinal zones.


Subject(s)
Cerebral Cortex , Interneurons , LIM-Homeodomain Proteins , Median Eminence , Transcription Factors , Animals , Cerebral Cortex/metabolism , DNA-Binding Proteins/metabolism , Interneurons/physiology , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Median Eminence/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neuropeptide Y/metabolism , Proto-Oncogene Proteins/metabolism , Somatostatin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Science ; 375(6579): 383-384, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35084977

ABSTRACT

[Figure: see text].


Subject(s)
Cerebral Cortex , Interneurons , Humans , Neurogenesis
6.
Cell Rep ; 35(11): 109249, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34133916

ABSTRACT

Cortical GABAergic interneurons are generated in large numbers in the ganglionic eminences and migrate into the cerebral cortex during embryogenesis. At early postnatal stages, during neuronal circuit maturation, autonomous and activity-dependent mechanisms operate within the cortex to adjust cell numbers by eliminating naturally occurring neuron excess. Here, we show that when cortical interneurons are generated in aberrantly high numbers-due to a defect in precursor cell proliferation during embryogenesis-extra parvalbumin interneurons persist in the postnatal mouse cortex during critical periods of cortical network maturation. Even though cell numbers are subsequently normalized, behavioral abnormalities remain in adulthood. This suggests that timely clearance of excess cortical interneurons is critical for correct functional maturation of circuits that drive adult behavior.


Subject(s)
Behavior, Animal/physiology , Cerebral Cortex/growth & development , Interneurons/pathology , Animals , Animals, Newborn , Cell Count , Homeodomain Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/metabolism , Parvalbumins/metabolism
7.
Stem Cell Reports ; 13(5): 793-802, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31631021

ABSTRACT

Oriens lacunosum-moleculare (O-LM) interneurons constitute 40% of hippocampal interneurons expressing Somatostatin (SST). Recent evidence has indicated a dual origin for these cells in the medial and caudal ganglionic eminences (MGE and CGE), with expression of Htr3a as a distinguishing factor. This is strikingly different from cortical SST interneurons that have a single origin within the MGE/preoptic area (POA). We reassessed the origin of hippocampal SST interneurons using a range of genetic lineage-tracing mice combined with single-cell transcriptomic analysis. We find a common origin for all hippocampal SST interneurons in NKX2-1-expressing progenitors of the telencephalic neuroepithelium and an MGE/POA-like transcriptomic signature for all SST clusters. This suggests that functional heterogeneity within the SST CA1 population cannot be attributed to a differential MGE/CGE genetic origin.


Subject(s)
CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/embryology , Interneurons/cytology , Somatostatin/analysis , Animals , CA1 Region, Hippocampal/metabolism , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Interneurons/metabolism , Mice , Mice, Transgenic , Preoptic Area/cytology , Preoptic Area/embryology , Preoptic Area/metabolism , Receptors, Serotonin, 5-HT3/analysis , Receptors, Serotonin, 5-HT3/genetics , Transcriptome
8.
J Comp Neurol ; 527(10): 1610-1620, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30520039

ABSTRACT

The murine subplate contains some of the earliest generated populations of neurons in the cerebral cortex, which play an important role in the maturation of cortical inhibition. Here we present multiple lines of evidence, that the subplate itself is only very sparsely populated with GABAergic neurons at postnatal day (P)8. We used three different transgenic mouse lines, each of which labels a subset of GABAergic, ganglionic eminence derived neurons. Dlx5/6-eGFP labels the most neurons in cortex (on average 11% of NEUN+ cells across all layers at P8) whereas CGE-derived Lhx6-Cre::Dlx1-Venusfl cells are the sparsest (2% of NEUN+ cells across all layers at P8). There is significant variability in the layer distribution of labeled interneurons, with Dlx5/6-eGFP and Lhx6-Cre::R26R-YFP being expressed most abundantly in Layer 5, whereas CGE-derived Lhx6-Cre::Dlx1-Venusfl cells are least abundant in that layer. All three lines label at most 3% of NEUN+ neurons in the subplate, in contrast to L5, in which up to 30% of neurons are GFP+ in Dlx5/6-eGFP. We assessed all three GABAergic populations for expression of the subplate neuron marker connective tissue growth factor (CTGF). CTGF labels up to two-thirds of NEUN+ cells in the subplate, but was never found to colocalize with labeled GABAergic neurons in any of the three transgenic strains. Despite the GABAergic neuronal population in the subplate being sparse, long-distance axonal connection tracing with carbocyanine dyes revealed that some Gad65-GFP+ subplate cells form long-range axonal projections to the internal capsule or callosum.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/growth & development , GABAergic Neurons/cytology , Animals , Mice , Mice, Transgenic
9.
PLoS Biol ; 16(6): e2006387, 2018 06.
Article in English | MEDLINE | ID: mdl-29912866

ABSTRACT

Understanding any brain circuit will require a categorization of its constituent neurons. In hippocampal area CA1, at least 23 classes of GABAergic neuron have been proposed to date. However, this list may be incomplete; additionally, it is unclear whether discrete classes are sufficient to describe the diversity of cortical inhibitory neurons or whether continuous modes of variability are also required. We studied the transcriptomes of 3,663 CA1 inhibitory cells, revealing 10 major GABAergic groups that divided into 49 fine-scale clusters. All previously described and several novel cell classes were identified, with three previously described classes unexpectedly found to be identical. A division into discrete classes, however, was not sufficient to describe the diversity of these cells, as continuous variation also occurred between and within classes. Latent factor analysis revealed that a single continuous variable could predict the expression levels of several genes, which correlated similarly with it across multiple cell types. Analysis of the genes correlating with this variable suggested it reflects a range from metabolically highly active faster-spiking cells that proximally target pyramidal cells to slower-spiking cells targeting distal dendrites or interneurons. These results elucidate the complexity of inhibitory neurons in one of the simplest cortical structures and show that characterizing these cells requires continuous modes of variation as well as discrete cell classes.


Subject(s)
CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , GABAergic Neurons/classification , GABAergic Neurons/metabolism , Action Potentials , Algorithms , Animals , Chemokines, CXC/genetics , Dendrites/metabolism , GABAergic Neurons/cytology , Interneurons/cytology , Interneurons/metabolism , Mice , Mice, Transgenic , Models, Neurological , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Synaptic Transmission , Transcriptome , Vasoactive Intestinal Peptide/genetics
10.
Cell ; 172(5): 1063-1078.e19, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474907

ABSTRACT

Interneurons navigate along multiple tangential paths to settle into appropriate cortical layers. They undergo a saltatory migration paced by intermittent nuclear jumps whose regulation relies on interplay between extracellular cues and genetic-encoded information. It remains unclear how cycles of pause and movement are coordinated at the molecular level. Post-translational modification of proteins contributes to cell migration regulation. The present study uncovers that carboxypeptidase 1, which promotes post-translational protein deglutamylation, controls the pausing of migrating cortical interneurons. Moreover, we demonstrate that pausing during migration attenuates movement simultaneity at the population level, thereby controlling the flow of interneurons invading the cortex. Interfering with the regulation of pausing not only affects the size of the cortical interneuron cohort but also impairs the generation of age-matched projection neurons of the upper layers.


Subject(s)
Cell Movement , Cerebral Cortex/cytology , Interneurons/cytology , Morphogenesis , Actomyosin/metabolism , Animals , Carboxypeptidases/metabolism , Cell Cycle , Chemotactic Factors/metabolism , Embryo, Mammalian/cytology , Female , Gene Deletion , Interneurons/metabolism , Mice , Mice, Knockout , Myosin-Light-Chain Kinase/metabolism , Neurogenesis , Phenotype
11.
J Comp Neurol ; 526(3): 397-411, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-28921616

ABSTRACT

In mammals, thalamic axons are guided internally toward their neocortical target by corridor (Co) neurons that act as axonal guideposts. The existence of Co-like neurons in non-mammalian species, in which thalamic axons do not grow internally, raised the possibility that Co cells might have an ancestral role. Here, we investigated the contribution of corridor (Co) cells to mature brain circuits using a combination of genetic fate-mapping and assays in mice. We unexpectedly found that Co neurons contribute to striatal-like projection neurons in the central extended amygdala. In particular, Co-like neurons participate in specific nuclei of the bed nucleus of the stria terminalis, which plays essential roles in anxiety circuits. Our study shows that Co neurons possess an evolutionary conserved role in anxiety circuits independently from an acquired guidepost function. It furthermore highlights that neurons can have multiple sequential functions during brain wiring and supports a general role of tangential migration in the building of subpallial circuits.


Subject(s)
Afferent Pathways/physiology , Axon Guidance/genetics , Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Pontine Tegmentum , Thalamus , Animals , Animals, Newborn , Cholera Toxin/metabolism , Deoxyuridine/analogs & derivatives , Deoxyuridine/metabolism , Embryo, Mammalian , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pontine Tegmentum/cytology , Pontine Tegmentum/embryology , Pontine Tegmentum/growth & development , Pregnancy , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Thalamus/cytology , Thalamus/embryology , Thalamus/growth & development , Thyroid Nuclear Factor 1/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Cell Rep ; 20(7): 1572-1584, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813670

ABSTRACT

The transcription factor NKX2-1 is best known for its role in the specification of subsets of cortical, striatal, and pallidal neurons. We demonstrate through genetic fate mapping and intersectional focal septal deletion that NKX2-1 is selectively required in the embryonic septal neuroepithelium for the development of cholinergic septohippocampal projection neurons and large subsets of basal forebrain cholinergic neurons. In the absence of NKX2-1, these neurons fail to develop, causing alterations in hippocampal theta rhythms and severe deficiencies in learning and memory. Our results demonstrate that learning and memory are dependent on NKX2-1 function in the embryonic septum and suggest that cognitive deficiencies that are sometimes associated with pathogenic mutations in NKX2-1 in humans may be a direct consequence of loss of NKX2-1 function.


Subject(s)
Cholinergic Neurons/metabolism , Gene Expression Regulation, Developmental , Hippocampus/metabolism , Memory/physiology , Septum of Brain/metabolism , Thyroid Nuclear Factor 1/genetics , Acetylcholine/metabolism , Animals , Cholinergic Neurons/pathology , Cognition/physiology , Electrodes, Implanted , Embryo, Mammalian , Female , Hippocampus/pathology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rotarod Performance Test , Septum of Brain/pathology , Stereotaxic Techniques , Theta Rhythm/physiology , Thyroid Nuclear Factor 1/deficiency
13.
Front Neurosci ; 11: 55, 2017.
Article in English | MEDLINE | ID: mdl-28232786

ABSTRACT

Although, the suprachiasmatic nucleus (SCN) of the hypothalamus acts as the central clock in mammals, the circadian expression of clock genes has been demonstrated not only in the SCN, but also in peripheral tissues and brain regions outside the SCN. However, the physiological roles of extra-SCN circadian clocks in the brain remain largely elusive. In response, we generated Nkx2.1-Bmal1-/- mice in which Bmal1, an essential clock component, was genetically deleted specifically in the ventral forebrain, including the preoptic area, nucleus of the diagonal band, and most of the hypothalamus except the SCN. In these mice, as expected, PER2::LUC oscillation was drastically attenuated in the explants of mediobasal hypothalamus, whereas it was maintained in those of the SCN. Although, Nkx2.1-Bmal1-/- mice were rhythmic and nocturnal, they showed altered patterns of locomotor activity during the night in a 12:12-h light:dark cycle and during subjective night in constant darkness. Control mice were more active during the first half than the second half of the dark phase or subjective night, whereas Nkx2.1-Bmal1-/- mice showed the opposite pattern of locomotor activity. Temporal patterns of sleep-wakefulness and feeding also changed accordingly. Such results suggest that along with mechanisms in the SCN, local Bmal1-dependent clocks in the ventral forebrain are critical for generating precise temporal patterns of circadian behaviors.

14.
Proc Natl Acad Sci U S A ; 113(47): 13414-13419, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821771

ABSTRACT

In the adult rodent brain, new neurons are born in two germinal regions that are associated with blood vessels, and blood vessels and vessel-derived factors are thought to regulate the activity of adult neural stem cells. Recently, it has been proposed that a vascular niche also regulates prenatal neurogenesis. Here we identify the mouse embryo hindbrain as a powerful model to study embryonic neurogenesis and define the relationship between neural progenitor cell (NPC) behavior and vessel growth. Using this model, we show that a subventricular vascular plexus (SVP) extends through a hindbrain germinal zone populated by NPCs whose peak mitotic activity follows a surge in SVP growth. Hindbrains genetically defective in SVP formation owing to constitutive NRP1 loss showed a premature decline in both NPC activity and hindbrain growth downstream of precocious cell cycle exit, premature neuronal differentiation, and abnormal mitosis patterns. Defective regulation of NPC activity was not observed in mice lacking NRP1 expression by NPCs, but instead in mice lacking NRP1 selectively in endothelial cells, yet was independent of vascular roles in hindbrain oxygenation. Therefore, germinal zone vascularization sustains NPC proliferation in the prenatal brain.


Subject(s)
Blood Vessels/physiology , Neurogenesis , Rhombencephalon/blood supply , Rhombencephalon/embryology , Animals , Cell Proliferation , Cell Self Renewal , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Mice , Mice, Inbred C57BL , Mitosis , Neovascularization, Physiologic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuropilin-1/metabolism , Oxygen/metabolism , Time Factors
15.
Development ; 142(21): 3746-57, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26534986

ABSTRACT

Transcription factors act during cortical development as master regulatory genes that specify cortical arealization and cellular identities. Although numerous transcription factors have been identified as being crucial for cortical development, little is known about their downstream targets and how they mediate the emergence of specific neuronal connections via selective axon guidance. The EMX transcription factors are essential for early patterning of the cerebral cortex, but whether EMX1 mediates interhemispheric connectivity by controlling corpus callosum formation remains unclear. Here, we demonstrate that in mice on the C57Bl/6 background EMX1 plays an essential role in the midline crossing of an axonal subpopulation of the corpus callosum derived from the anterior cingulate cortex. In the absence of EMX1, cingulate axons display reduced expression of the axon guidance receptor NRP1 and form aberrant axonal bundles within the rostral corpus callosum. EMX1 also functions as a transcriptional activator of Nrp1 expression in vitro, and overexpression of this protein in Emx1 knockout mice rescues the midline-crossing phenotype. These findings reveal a novel role for the EMX1 transcription factor in establishing cortical connectivity by regulating the interhemispheric wiring of a subpopulation of neurons within the mouse anterior cingulate cortex.


Subject(s)
Gyrus Cinguli/metabolism , Homeodomain Proteins/metabolism , Neuropilin-1/metabolism , Transcription Factors/metabolism , Agenesis of Corpus Callosum/embryology , Agenesis of Corpus Callosum/genetics , Animals , Axons/metabolism , Mice, Inbred C57BL , Mice, Knockout , Semaphorins/metabolism
16.
Neuron ; 86(2): 501-13, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25843402

ABSTRACT

Transcriptional codes initiated during brain development are ultimately realized in adulthood as distinct cell types performing specialized roles in behavior. Focusing on the mouse external globus pallidus (GPe), we demonstrate that the potential contributions of two GABAergic GPe cell types to voluntary action are fated from early life to be distinct. Prototypic GPe neurons derive from the medial ganglionic eminence of the embryonic subpallium and express the transcription factor Nkx2-1. These neurons fire at high rates during alert rest, and encode movements through heterogeneous firing rate changes, with many neurons decreasing their activity. In contrast, arkypallidal GPe neurons originate from lateral/caudal ganglionic eminences, express the transcription factor FoxP2, fire at low rates during rest, and encode movements with robust increases in firing. We conclude that developmental diversity positions prototypic and arkypallidal neurons to fulfil distinct roles in behavior via their disparate regulation of GABA release onto different basal ganglia targets.


Subject(s)
Forkhead Transcription Factors/metabolism , Globus Pallidus/cytology , Globus Pallidus/growth & development , Movement/physiology , Neurons/classification , Neurons/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Action Potentials/physiology , Animals , Cell Lineage/physiology , Enkephalins/metabolism , Globus Pallidus/embryology , Mice , Protein Precursors/metabolism , ROC Curve , Thyroid Nuclear Factor 1 , gamma-Aminobutyric Acid/metabolism
17.
Cereb Cortex ; 25(9): 2370-82, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24626607

ABSTRACT

Cortical interneurons are characterized by extraordinary functional and morphological diversity. Although tremendous progress has been made in uncovering molecular and cellular mechanisms implicated in interneuron generation and function, several questions still remain open. Rho-GTPases have been implicated as intracellular mediators of numerous developmental processes such as cytoskeleton organization, vesicle trafficking, transcription, cell cycle progression, and apoptosis. Specifically in cortical interneurons, we have recently shown a cell-autonomous and stage-specific requirement for Rac1 activity within proliferating interneuron precursors. Conditional ablation of Rac1 in the medial ganglionic eminence leads to a 50% reduction of GABAergic interneurons in the postnatal cortex. Here we examine the additional role of Rac3 by analyzing Rac1/Rac3 double-mutant mice. We show that in the absence of both Rac proteins, the embryonic migration of medial ganglionic eminence-derived interneurons is further impaired. Postnatally, double-mutant mice display a dramatic loss of cortical interneurons. In addition, Rac1/Rac3-deficient interneurons show gross cytoskeletal defects in vitro, with the length of their leading processes significantly reduced and a clear multipolar morphology. We propose that in the absence of Rac1/Rac3, cortical interneurons fail to migrate tangentially towards the pallium due to defects in actin and microtubule cytoskeletal dynamics.


Subject(s)
Axons/physiology , Cerebral Cortex/cytology , Interneurons/cytology , Microtubules/physiology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Axons/ultrastructure , Cell Cycle/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Interneurons/metabolism , Interneurons/ultrastructure , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Median Eminence/cytology , Mice , Mice, Transgenic , Microtubules/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Paclitaxel/pharmacology , Pregnancy , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolism , Tubulin Modulators/pharmacology , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein/genetics
18.
Proc Natl Acad Sci U S A ; 111(11): 4239-44, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591640

ABSTRACT

Phospholipase Cε (PLCε) has been characterized as a direct effector of Ras in vitro and in cellular systems; however, the role of PLCε in tumorigenesis and its link to Ras in this context remain unclear. To assess the role of PLCε in Ras-driven cancers, we generated two new mouse strains: one carrying a targeted deletion of Plce (Plce(-/-)) and the other carrying mutant alleles of Plce unable to bind to Ras (Plce(RAm/RAm)). The Plce(-/-) and, to a lesser degree, Plce(RAm/RAm) transgenic mice exhibited increased susceptibility to tumor formation in the two-stage skin carcinogenesis protocol, revealing a tumor suppressor function for this PLC. This result also suggests that in this context Ras binding in part regulates functions of PLCε. Although significant differences were not seen in the LSL-Kras(G12D) nonsmall cell lung carcinoma model, down-regulation of PLCε was found in animal tumors and in cellular systems following expression of the oncogenic Ras. An inhibitory impact of PLCε on cell growth requires intact lipase activity and is likely mediated by protein kinase C enzymes. Further cellular studies suggest involvement of histone deacetylase in the mechanism of PLCε down-regulation. Taken together, our results show a previously unidentified tumor suppressor role for this PLC in animal models and, together with observations of marked down-regulation in colorectal, lung, and skin tumors, suggest its use as a biological marker in cancer.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Genes, Tumor Suppressor/physiology , Genes, ras/genetics , Neoplasms/genetics , Phosphoinositide Phospholipase C/physiology , Animals , Cell Proliferation , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Phosphoinositide Phospholipase C/genetics , Protein Kinase C/metabolism , Real-Time Polymerase Chain Reaction
19.
BMC Neurosci ; 15: 12, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24423059

ABSTRACT

BACKGROUND: Olig1 and Olig2, encoding closely related basic helix-loop-helix transcription factors, were originally identified in screens for glial-specific genes. Olig1 and Olig2 are both expressed in restricted parts of the neuroepithelium of the embryonic spinal cord and telencephalon and subsequently in oligodendrocyte lineage cells throughout life. In the spinal cord, Olig2 plays a crucial role in the development of oligodendrocytes and motor neurons, and both cell types are lost from Olig2 null mutant mice. The role of Olig1 has been more cryptic. It was initially reported that Olig1 null mice (with a Cre-Pgk-Neo cassette at the Olig1 locus) have a mild developmental phenotype characterized by a slight delay in oligodendrocyte differentiation. However, a subsequent study of the same line following removal of Pgk-Neo (leaving Olig1-Cre) found severe disruption of oligodendrocyte production, myelination failure and early postnatal lethality. A plausible explanation was proposed, that the highly expressed Pgk-Neo cassette in the original line might have up-regulated the neighbouring Olig2 gene, compensating for loss of Olig1. However, this was not tested, so the importance of Olig1 for oligodendrocyte development has remained unclear. RESULTS: We generated two independent lines of Olig1 null mice. Both lines had a mild phenotype featuring slightly delayed oligodendrocyte differentiation and maturation but no long-term effect. In addition, we found that Olig2 transcripts were not up-regulated in our Olig1 null mice. CONCLUSIONS: Our findings support the original conclusion that Olig1 plays a minor and non-essential role in oligodendrocyte development and have implications for the interpretation of studies based on Olig1 deficient mice (and perhaps Olig1-Cre mice) from different sources.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Brain/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Spinal Cord/cytology , Spinal Cord/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Cells, Cultured , Mice , Mice, Knockout
20.
Curr Opin Neurobiol ; 26: 79-87, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24440413

ABSTRACT

The origins of cortical interneurons in rodents have been localized to the embryonic subcortical telencephalon where distinct neuroepithelial precursors generate defined interneuron subsets. A swathe of research activity aimed at identifying molecular determinants of subtype identity has uncovered a number of transcription factors that function at different stages of interneuron development. Pathways that lead to the acquisition of mature interneuron traits are therefore beginning to emerge. As genetic programs are influenced by external factors the search continues not only into genetic determinants but also extrinsic influences and the interplay between the two in cell fate specification.


Subject(s)
Cell Differentiation/genetics , Cerebral Cortex/cytology , Genetic Phenomena/physiology , Interneurons/physiology , Animals , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...