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










Publication year range
1.
Immunity ; 57(1): 86-105.e9, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38159572

ABSTRACT

Triggering receptor expressed on myeloid cells 2 (Trem2) is a myeloid cell-specific gene expressed in brain microglia, with variants that are associated with neurodegenerative diseases, including Alzheimer's disease. Trem2 is essential for microglia-mediated synaptic refinement, but whether Trem2 contributes to shaping neuronal development remains unclear. Here, we demonstrate that Trem2 plays a key role in controlling the bioenergetic profile of pyramidal neurons during development. In the absence of Trem2, developing neurons in the hippocampal cornus ammonis (CA)1 but not in CA3 subfield displayed compromised energetic metabolism, accompanied by reduced mitochondrial mass and abnormal organelle ultrastructure. This was paralleled by the transcriptional rearrangement of hippocampal pyramidal neurons at birth, with a pervasive alteration of metabolic, oxidative phosphorylation, and mitochondrial gene signatures, accompanied by a delay in the maturation of CA1 neurons. Our results unveil a role of Trem2 in controlling neuronal development by regulating the metabolic fitness of neurons in a region-specific manner.


Subject(s)
Alzheimer Disease , Microglia , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Brain/metabolism , Energy Metabolism , Microglia/metabolism , Neurons/metabolism , Animals , Mice
2.
Brief Bioinform ; 25(1)2023 11 22.
Article in English | MEDLINE | ID: mdl-38102070

ABSTRACT

Mass spectrometry imaging (MSI) is commonly used to map the spatial distribution of small molecules within complex biological matrices. One of the major challenges in imaging MS-based spatial metabolomics is molecular identification and metabolite annotation, to address this limitation, annotation is often complemented with parallel bulk LC-MS2-based metabolomics to confirm and validate identifications. Here we applied MSI method, utilizing data-dependent acquisition, to visualize and identify unknown molecules in a single instrument run. To reach this aim we developed MSIpixel, a fully automated pipeline for compound annotation and quantitation in MSI experiments. It overcomes challenges in molecular identification, and improving reliability and comprehensiveness in MSI-based spatial metabolomics.


Subject(s)
Metabolomics , Reproducibility of Results , Mass Spectrometry , Metabolomics/methods
3.
Cell Mol Life Sci ; 80(12): 373, 2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38007410

ABSTRACT

Mitofusin-2 (MFN2) is an outer mitochondrial membrane protein essential for mitochondrial networking in most cells. Autosomal dominant mutations in the MFN2 gene cause Charcot-Marie-Tooth type 2A disease (CMT2A), a severe and disabling sensory-motor neuropathy that impacts the entire nervous system. Here, we propose a novel therapeutic strategy tailored to correcting the root genetic defect of CMT2A. Though mutant and wild-type MFN2 mRNA are inhibited by RNA interference (RNAi), the wild-type protein is restored by overexpressing cDNA encoding functional MFN2 modified to be resistant to RNAi. We tested this strategy in CMT2A patient-specific human induced pluripotent stem cell (iPSC)-differentiated motor neurons (MNs), demonstrating the correct silencing of endogenous MFN2 and replacement with an exogenous copy of the functional wild-type gene. This approach significantly rescues the CMT2A MN phenotype in vitro, stabilizing the altered axonal mitochondrial distribution and correcting abnormal mitophagic processes. The MFN2 molecular correction was also properly confirmed in vivo in the MitoCharc1 CMT2A transgenic mouse model after cerebrospinal fluid (CSF) delivery of the constructs into newborn mice using adeno-associated virus 9 (AAV9). Altogether, our data support the feasibility of a combined RNAi and gene therapy strategy for treating the broad spectrum of human diseases associated with MFN2 mutations.


Subject(s)
Charcot-Marie-Tooth Disease , Induced Pluripotent Stem Cells , Humans , Mice , Animals , RNA Interference , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Induced Pluripotent Stem Cells/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/therapy , Charcot-Marie-Tooth Disease/metabolism , Mutation , Hydrolases/genetics , Mice, Transgenic
4.
Nat Neurosci ; 25(8): 1049-1058, 2022 08.
Article in English | MEDLINE | ID: mdl-35915179

ABSTRACT

Mammalian neocortical neurons span one of the most diverse cell type spectra of any tissue. Cortical neurons are born during embryonic development, and their maturation extends into postnatal life. The regulatory strategies underlying progressive neuronal development and maturation remain unclear. Here we present an integrated single-cell epigenomic and transcriptional analysis of individual mouse and marmoset cortical neuron classes, spanning both early postmitotic stages of identity acquisition and later stages of neuronal plasticity and circuit integration. We found that, in both species, the regulatory strategies controlling early and late stages of pan-neuronal development diverge. Early postmitotic neurons use more widely shared and evolutionarily conserved molecular regulatory programs. In contrast, programs active during later neuronal maturation are more brain- and neuron-specific and more evolutionarily divergent. Our work uncovers a temporal shift in regulatory choices during neuronal diversification and maturation in both mice and marmosets, which likely reflects unique evolutionary constraints on distinct events of neuronal development in the neocortex.


Subject(s)
Neocortex , Animals , Callithrix , Mammals , Mice , Neurogenesis/physiology , Neuronal Plasticity , Neurons/physiology
5.
Fac Rev ; 11: 13, 2022.
Article in English | MEDLINE | ID: mdl-35719130

ABSTRACT

Decoding the complexity of the brain requires an understanding of the architecture, function, and development of its neuronal circuits. Neuronal classifications that group neurons based on specific features/behaviors have become essential to further analyze the different subtypes in a systematic and reproducible way. A comprehensive taxonomic framework, accounting for multiple defining and quantitative features, will provide the reference to infer generalized rules for cells ascribed to the same neuronal type, and eventually predict cellular behaviors, even in the absence of experimental measures. Technologies that enable cell-type classification in the nervous system are rapidly evolving in scalability and resolution. While these approaches depict astonishing diversity in neuronal morphology, electrophysiology, and gene expression, a robust metric of the coherence between different profiling modalities leading to a unified classification is still largely missing. Focusing on GABAergic neurons of the cerebral cortex, Gouwens et al.1 pioneered the first integrated cell-type classification based on the simultaneous analysis of the transcriptional networks, the recording of intrinsic electrophysiological properties, and the reconstruction of 3D morphologies of the same cell. Their comprehensive and high-quality data provide a new framework to shed light on what may be considered a "neuronal cell type."

6.
Immunity ; 54(11): 2611-2631.e8, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758338

ABSTRACT

Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.


Subject(s)
Hippocampus/metabolism , Interleukin-6/biosynthesis , Maternal Exposure , Neurons/metabolism , Prenatal Exposure Delayed Effects , Synapses/metabolism , Animals , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility , Female , Hippocampus/physiopathology , Inflammation Mediators/metabolism , Mice , Pregnancy , Signal Transduction , Synaptic Transmission
7.
Science ; 374(6566): 439-448, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34672740

ABSTRACT

Up to 40% of patients with inflammatory bowel disease present with psychosocial disturbances. We previously identified a gut vascular barrier that controls the dissemination of bacteria from the intestine to the liver. Here, we describe a vascular barrier in the brain choroid plexus (PVB) that is modulated in response to intestinal inflammation through bacteria-derived lipopolysaccharide. The inflammatory response induces PVB closure after gut vascular barrier opening by the up-regulation of the wingless-type, catenin-beta 1 (Wnt/ß-catenin) signaling pathway, rendering it inaccessible to large molecules. In a model of genetically driven closure of choroid plexus endothelial cells, we observed a deficit in short-term memory and anxiety-like behavior, suggesting that PVB closure may correlate with mental deficits. Inflammatory bowel disease­related mental symptoms may thus be the consequence of a deregulated gut­brain vascular axis.


Subject(s)
Choroid Plexus/blood supply , Choroid Plexus/physiopathology , Colitis, Ulcerative/physiopathology , Colitis, Ulcerative/psychology , Intestines/physiopathology , Memory Disorders/physiopathology , Memory, Short-Term , Animals , Anxiety/etiology , Anxiety/physiopathology , Blood-Brain Barrier/pathology , Colitis, Ulcerative/complications , Dextrans , Disease Models, Animal , Humans , Lipopolysaccharides , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Microglia/pathology , Signal Transduction , Tight Junctions/pathology , Wnt Proteins/metabolism , beta Catenin/metabolism
8.
BMC Med ; 19(1): 42, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33541338

ABSTRACT

BACKGROUND: Preterm birth affects almost 9-11% of newborns and is one of the leading causes of childhood neurodevelopmental disabilities; the underlying molecular networks are poorly defined. In neurons, retrotransposons LINE-1 (L1) are an active source of genomic mosaicism that is deregulated in several neurological disorders; early life experience has been shown to regulate L1 activity in mice. METHODS: Very preterm infants were randomized to receive standard care or early intervention. L1 methylation was measured at birth and at hospital discharge. At 12 and 36 months, infants' neurodevelopment was evaluated with the Griffiths Scales. L1 methylation and CNVs were measured in mouse brain areas at embryonic and postnatal stages. RESULTS: Here we report that L1 promoter is hypomethylated in preterm infants at birth and that an early intervention program, based on enhanced maternal care and positive multisensory stimulation, restores L1 methylation levels comparable to healthy newborns and ameliorates neurodevelopment in childhood. We further show that L1 activity is fine-tuned in the perinatal mouse brain, suggesting a sensitive and vulnerable window for the L1 epigenetic setting. CONCLUSIONS: Our results open the field on the inspection of L1 activity as a novel molecular and predictive approach to infants' prematurity-related neurodevelopmental outcomes. TRIAL REGISTRATION: ClinicalTrial.gov ( NCT02983513 ). Registered on 6 December 2016, retrospectively registered.


Subject(s)
Child Development/physiology , DNA Methylation/physiology , Infant Care/methods , Infant, Premature/physiology , Neurodevelopmental Disorders/prevention & control , Female , Humans , Infant , Infant, Newborn , Infant, Premature/growth & development , Methylation , Patient Discharge/statistics & numerical data , Pregnancy , Premature Birth
9.
Science ; 370(6520)2020 11 27.
Article in English | MEDLINE | ID: mdl-33243861

ABSTRACT

The number of disease risk genes and loci identified through human genetic studies far outstrips the capacity to systematically study their functions. We applied a scalable genetic screening approach, in vivo Perturb-Seq, to functionally evaluate 35 autism spectrum disorder/neurodevelopmental delay (ASD/ND) de novo loss-of-function risk genes. Using CRISPR-Cas9, we introduced frameshift mutations in these risk genes in pools, within the developing mouse brain in utero, followed by single-cell RNA-sequencing of perturbed cells in the postnatal brain. We identified cell type-specific and evolutionarily conserved gene modules from both neuronal and glial cell classes. Recurrent gene modules and cell types are affected across this cohort of perturbations, representing key cellular effects across sets of ASD/ND risk genes. In vivo Perturb-Seq allows us to investigate how diverse mutations affect cell types and states in the developing organism.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/pathology , Brain/abnormalities , Neuroglia/pathology , Neurons/pathology , Animals , Ankyrins/genetics , Ankyrins/metabolism , CRISPR-Cas Systems , DNA-Binding Proteins/genetics , Frameshift Mutation , Gene Expression Profiling , Genetic Loci , Humans , Mice , Neuroglia/metabolism , Neurons/metabolism , Repressor Proteins/genetics , Risk , Transcription Factors/genetics
10.
Curr Opin Neurobiol ; 53: 146-155, 2018 12.
Article in English | MEDLINE | ID: mdl-30165269

ABSTRACT

The neocortex is by far one of the most complex regions of the mammalian brain, characterized by an extraordinary diversity of neuronal and non-neuronal cell types, whose coordinated development and function guarantee the execution of high-order cognitive, sensory, and motor behaviours. Decoding its heterogeneity and understanding the molecular strategies upon which the cerebral cortex is built during development have been at the core of neuroscientists' work for decades. Here, we will focus on the current classification of neuronal types (both excitatory and inhibitory) of the neocortex in light of the insights provided by recent single-cell omic technologies, which have offered - with unprecedented resolution - an extended framework to interpret cortical diversity and its developmental origin. We will cover the impact of neuronal subtype identity on generating specific neuronal networks (neuron-to-neuron interaction), as well as their effect on the development of the non-neuronal populations in the cerebral cortex.


Subject(s)
Neocortex/cytology , Neocortex/growth & development , Nerve Net/cytology , Nerve Net/growth & development , Neural Stem Cells/physiology , Neurons/classification , Animals , Humans
11.
Annu Rev Cell Dev Biol ; 31: 699-720, 2015.
Article in English | MEDLINE | ID: mdl-26359774

ABSTRACT

The neocortex is the part of the brain responsible for execution of higher-order brain functions, including cognition, sensory perception, and sophisticated motor control. During evolution, the neocortex has developed an unparalleled neuronal diversity, which still remains partly unclassified and unmapped at the functional level. Here, we broadly review the structural blueprint of the neocortex and discuss the current classification of its neuronal diversity. We then cover the principles and mechanisms that build neuronal diversity during cortical development and consider the impact of neuronal class-specific identity in shaping cortical connectivity and function.


Subject(s)
Mammals/physiology , Neocortex/physiology , Nerve Net/physiology , Neurons/physiology , Animals , Biological Evolution , Humans
12.
Trends Neurosci ; 38(2): 117-25, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25529141

ABSTRACT

The mammalian cerebral cortex is responsible for the highest levels of associative, cognitive and motor functions. In the central nervous system (CNS) the cortex stands as a prime example of extreme neuronal diversity, broadly classified into excitatory projection neurons (PNs) and inhibitory interneurons (INs). We review here recent progress made in understanding the strategies and mechanisms that shape PN diversity during embryogenesis, and discuss how PN classes may be maintained, postnatally, for the life of the organism. In addition, we consider the intriguing possibility that PNs may be amenable to directed reprogramming of their class-specific features to allow enhanced cortical plasticity in the adult.


Subject(s)
Cerebral Cortex/physiology , Nerve Net/physiology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Animals , Humans , Interneurons/physiology
13.
Nat Neurosci ; 17(8): 1046-54, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997765

ABSTRACT

The neocortex contains an unparalleled diversity of neuronal subtypes, each defined by distinct traits that are developmentally acquired under the control of subtype-specific and pan-neuronal genes. The regulatory logic that orchestrates the expression of these unique combinations of genes is unknown for any class of cortical neuron. Here, we report that Fezf2 is a selector gene able to regulate the expression of gene sets that collectively define mouse corticospinal motor neurons (CSMN). We find that Fezf2 directly induces the glutamatergic identity of CSMN via activation of Vglut1 (Slc17a7) and inhibits a GABAergic fate by repressing transcription of Gad1. In addition, we identify the axon guidance receptor EphB1 as a target of Fezf2 necessary to execute the ipsilateral extension of the corticospinal tract. Our data indicate that co-regulated expression of neuron subtype-specific and pan-neuronal gene batteries by a single transcription factor is one component of the regulatory logic responsible for the establishment of CSMN identity.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Motor Neurons/metabolism , Nerve Tissue Proteins/physiology , Neurotransmitter Agents/genetics , Pyramidal Tracts/metabolism , Signal Transduction/genetics , Animals , DNA-Binding Proteins/genetics , Genes, Suppressor , Mice , Mice, Knockout , Mice, Transgenic , Motor Neurons/classification , Motor Neurons/physiology , Nerve Tissue Proteins/genetics , Neurotransmitter Agents/biosynthesis , Neurotransmitter Agents/metabolism , Promoter Regions, Genetic/genetics , Pyramidal Tracts/cytology , Pyramidal Tracts/physiology
14.
Neuron ; 77(1): 1-3, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23312509

ABSTRACT

Cortical and striatal interneurons are both generated within the ventral telencephalon, but their migratory journey takes them to very different destinations. Two articles in this issue (van den Berge et al., 2013; McKinsey et al., 2013) add an important molecular component to our understanding of how, during development, interneurons reach the cerebral cortex.

15.
Elife ; 2: e01749, 2013 Dec 31.
Article in English | MEDLINE | ID: mdl-24381249

ABSTRACT

Many studies are uncovering functional roles for long noncoding RNAs (lncRNAs), yet few have been tested for in vivo relevance through genetic ablation in animal models. To investigate the functional relevance of lncRNAs in various physiological conditions, we have developed a collection of 18 lncRNA knockout strains in which the locus is maintained transcriptionally active. Initial characterization revealed peri- and postnatal lethal phenotypes in three mutant strains (Fendrr, Peril, and Mdgt), the latter two exhibiting incomplete penetrance and growth defects in survivors. We also report growth defects for two additional mutant strains (linc-Brn1b and linc-Pint). Further analysis revealed defects in lung, gastrointestinal tract, and heart in Fendrr(-/-) neonates, whereas linc-Brn1b(-/-) mutants displayed distinct abnormalities in the generation of upper layer II-IV neurons in the neocortex. This study demonstrates that lncRNAs play critical roles in vivo and provides a framework and impetus for future larger-scale functional investigation into the roles of lncRNA molecules. DOI: http://dx.doi.org/10.7554/eLife.01749.001.


Subject(s)
Brain/growth & development , RNA, Long Noncoding/physiology , Animals , Mice , Mice, Knockout , RNA, Long Noncoding/genetics
16.
J Neurosci ; 31(12): 4650-62, 2011 Mar 23.
Article in English | MEDLINE | ID: mdl-21430164

ABSTRACT

In rodents, cortical interneurons originate from the medial ganglionic eminence (MGE) and caudal ganglionic eminence (CGE) according to precise temporal schedules. The mechanisms controlling the specification of CGE-derived interneurons and their role in cortical circuitry are still unknown. Here, we show that COUP-TFI expression becomes restricted to the dorsal MGE and CGE at embryonic day 13.5 in the basal telencephalon. Conditional loss of function of COUP-TFI in subventricular precursors and postmitotic cells leads to a decrease of late-born, CGE-derived, VIP (vasoactive intestinal peptide)- and CR (calretinin)-expressing bipolar cortical neurons, compensated by the concurrent increase of early-born MGE-derived, PV (parvalbumin)-expressing interneurons. Strikingly, COUP-TFI mutants are more resistant to pharmacologically induced seizures, a phenotype that is dependent on GABAergic signaling. Together, our data indicate that COUP-TFI controls the delicate balance between MGE- and CGE-derived cortical interneurons by regulating intermediate progenitor divisions and ultimately affecting the activity of the cortical inhibitory circuitry.


Subject(s)
COUP Transcription Factor I/genetics , COUP Transcription Factor I/physiology , Cerebral Cortex/physiology , Epilepsy/genetics , Epilepsy/physiopathology , Interneurons/physiology , Median Eminence/physiology , Animals , Antimetabolites , Bromodeoxyuridine , Cell Proliferation , Cerebral Cortex/cytology , Convulsants/pharmacology , Drug Resistance/genetics , Electroencephalography , Electrophysiological Phenomena , Epilepsy/chemically induced , Immunohistochemistry , In Situ Hybridization , Median Eminence/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/cytology , Nerve Net/physiology , Receptors, GABA-A/genetics , Receptors, GABA-A/physiology , Receptors, GABA-B/genetics , Receptors, GABA-B/physiology , Telencephalon/cytology , Telencephalon/physiology , gamma-Aminobutyric Acid/physiology
17.
Neuron ; 69(4): 763-79, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21338885

ABSTRACT

In the mammalian cerebral cortex, the developmental events governing the integration of excitatory projection neurons and inhibitory interneurons into balanced local circuitry are poorly understood. We report that different subtypes of projection neurons uniquely and differentially determine the laminar distribution of cortical interneurons. We find that in Fezf2⁻/⁻ cortex, the exclusive absence of subcerebral projection neurons and their replacement by callosal projection neurons cause distinctly abnormal lamination of interneurons and altered GABAergic inhibition. In addition, experimental generation of either corticofugal neurons or callosal neurons below the cortex is sufficient to recruit cortical interneurons to these ectopic locations. Strikingly, the identity of the projection neurons generated, rather than strictly their birthdate, determines the specific types of interneurons recruited. These data demonstrate that in the neocortex individual populations of projection neurons cell-extrinsically control the laminar fate of interneurons and the assembly of local inhibitory circuitry.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Gene Expression Regulation, Developmental/physiology , Interneurons/physiology , Nerve Net/physiology , Neural Inhibition/physiology , Age Factors , Animals , Animals, Newborn , Cell Count , Cell Movement/genetics , Corpus Callosum/cytology , Corpus Callosum/growth & development , DNA-Binding Proteins/deficiency , Electric Stimulation , Electroporation/methods , Embryo, Mammalian , Female , Functional Laterality/genetics , Gene Expression Regulation, Developmental/genetics , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Membrane Potentials/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Pathways/physiology , Neurons/classification , Neurons/physiology , Patch-Clamp Techniques , Pregnancy
18.
Nat Biotechnol ; 29(2): 149-53, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21248753

ABSTRACT

The ability to direct functional proteins to specific DNA sequences is a long-sought goal in the study and engineering of biological processes. Transcription activator-like effectors (TALEs) from Xanthomonas sp. are site-specific DNA-binding proteins that can be readily designed to target new sequences. Because TALEs contain a large number of repeat domains, it can be difficult to synthesize new variants. Here we describe a method that overcomes this problem. We leverage codon degeneracy and type IIs restriction enzymes to generate orthogonal ligation linkers between individual repeat monomers, thus allowing full-length, customized, repeat domains to be constructed by hierarchical ligation. We synthesized 17 TALEs that are customized to recognize specific DNA-binding sites, and demonstrate that they can specifically modulate transcription of endogenous genes (SOX2 and KLF4) in human cells.


Subject(s)
Genetic Engineering , Transcription Factors/genetics , Transcription, Genetic , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Binding Sites , Cell Line , DNA/genetics , DNA/metabolism , High-Throughput Nucleotide Sequencing , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Microscopy, Fluorescence , Molecular Sequence Data , Promoter Regions, Genetic , SOXB1 Transcription Factors/genetics , Transcription Factors/metabolism , Xanthomonas/genetics , Xanthomonas/metabolism
19.
Sci Prog ; 93(Pt 2): 151-69, 2010.
Article in English | MEDLINE | ID: mdl-20681320

ABSTRACT

The idea of repairing damaged neuronal circuitry in the mammalian central nervous system (CNS) has challenged neuroscientists for centuries. This is mainly due to the notorious inability of neurons to regenerate and the unparalleled cellular diversity of the nervous system. In the mammalian cerebral cortex, one of the most complex areas of the CNS, multipotent neural stem and progenitor cells undergo progressive specification during development to generate the staggering variety of projection neuron subtypes that are found in the adult. How is this process orchestrated in the embryo? And, can developmental signals be used to regenerate projection neuron subtypes in the adult or in the dish? Here, we first provide an overview of the diversity and fate potential of neural progenitors of the cerebral cortex during development. Further, we discuss the plasticity of neural progenitors and the roles of intrinsic and extrinsic signals over progenitor fate. Finally, we discuss the relevance of developmental signals for efforts to direct the differentiation of pluripotent stem cells into specific types of cortical projection neurons for therapeutic benefit.


Subject(s)
Cerebral Cortex/cytology , Neurons/cytology , Neurons/physiology , Animals , Embryonic Development , Nerve Regeneration , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology
20.
Proc Natl Acad Sci U S A ; 107(8): 3576-81, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133588

ABSTRACT

Transcription factors with gradients of expression in neocortical progenitors give rise to distinct motor and sensory cortical areas by controlling the area-specific differentiation of distinct neuronal subtypes. However, the molecular mechanisms underlying this area-restricted control are still unclear. Here, we show that COUP-TFI controls the timing of birth and specification of corticospinal motor neurons (CSMN) in somatosensory cortex via repression of a CSMN differentiation program. Loss of COUP-TFI function causes an area-specific premature generation of neurons with cardinal features of CSMN, which project to subcerebral structures, including the spinal cord. Concurrently, genuine CSMN differentiate imprecisely and do not project beyond the pons, together resulting in impaired skilled motor function in adult mice with cortical COUP-TFI loss-of-function. Our findings indicate that COUP-TFI exerts critical areal and temporal control over the precise differentiation of CSMN during corticogenesis, thereby enabling the area-specific functional features of motor and sensory areas to arise.


Subject(s)
COUP Transcription Factor I/metabolism , Gene Expression Regulation, Developmental , Motor Neurons/cytology , Neurogenesis/genetics , Pyramidal Tracts/cytology , Temporal Lobe/growth & development , Animals , COUP Transcription Factor I/genetics , Mice , Mice, Knockout , Motor Neurons/metabolism , Pyramidal Tracts/metabolism , Temporal Lobe/metabolism , Thalamus/growth & development , Thalamus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL