Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 4087, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34471112

ABSTRACT

We utilized forebrain organoids generated from induced pluripotent stem cells of patients with a syndromic form of Autism Spectrum Disorder (ASD) with a homozygous protein-truncating mutation in CNTNAP2, to study its effects on embryonic cortical development. Patients with this mutation present with clinical characteristics of brain overgrowth. Patient-derived forebrain organoids displayed an increase in volume and total cell number that is driven by increased neural progenitor proliferation. Single-cell RNA sequencing revealed PFC-excitatory neurons to be the key cell types expressing CNTNAP2. Gene ontology analysis of differentially expressed genes (DEgenes) corroborates aberrant cellular proliferation. Moreover, the DEgenes are enriched for ASD-associated genes. The cell-type-specific signature genes of the CNTNAP2-expressing neurons are associated with clinical phenotypes previously described in patients. The organoid overgrowth phenotypes were largely rescued after correction of the mutation using CRISPR-Cas9. This CNTNAP2-organoid model provides opportunity for further mechanistic inquiry and development of new therapeutic strategies for ASD.


Subject(s)
Autism Spectrum Disorder/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Organoids/metabolism , Prosencephalon/metabolism , Adolescent , Autism Spectrum Disorder/genetics , Cell Differentiation , Cell Proliferation , Child , Female , Genetic Predisposition to Disease/genetics , Humans , Induced Pluripotent Stem Cells , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phenotype , Sequence Analysis, RNA
2.
Elife ; 102021 04 27.
Article in English | MEDLINE | ID: mdl-33904394

ABSTRACT

Cortical interneurons establish inhibitory microcircuits throughout the neocortex and their dysfunction has been implicated in epilepsy and neuropsychiatric diseases. Developmentally, interneurons migrate from a distal progenitor domain in order to populate the neocortex - a process that occurs at a slower rate in humans than in mice. In this study, we sought to identify factors that regulate the rate of interneuron maturation across the two species. Using embryonic mouse development as a model system, we found that the process of initiating interneuron migration is regulated by blood vessels of the medial ganglionic eminence (MGE), an interneuron progenitor domain. We identified two endothelial cell-derived paracrine factors, SPARC and SerpinE1, that enhance interneuron migration in mouse MGE explants and organotypic cultures. Moreover, pre-treatment of human stem cell-derived interneurons (hSC-interneurons) with SPARC and SerpinE1 prior to transplantation into neonatal mouse cortex enhanced their migration and morphological elaboration in the host cortex. Further, SPARC and SerpinE1-treated hSC-interneurons also exhibited more mature electrophysiological characteristics compared to controls. Overall, our studies suggest a critical role for CNS vasculature in regulating interneuron developmental maturation in both mice and humans.


Subject(s)
Cell Movement/drug effects , Cerebral Cortex/metabolism , Induced Pluripotent Stem Cells/drug effects , Interneurons/drug effects , Median Eminence/blood supply , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Osteonectin/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Action Potentials , Animals , Cerebral Cortex/embryology , Cerebral Cortex/surgery , Endothelial Cells/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Interneurons/metabolism , Interneurons/transplantation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Median Eminence/embryology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Neovascularization, Physiologic , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Osteonectin/metabolism , Paracrine Communication , Plasminogen Activator Inhibitor 1/metabolism , Signal Transduction
3.
Dev Neurobiol ; 76(5): 533-50, 2016 May.
Article in English | MEDLINE | ID: mdl-26243376

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated member of the basic-helix-loop-helix/PER-ARNT-SIM(PAS) transcription factor superfamily that also mediates the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Increasing evidence suggests that AhR influences the development of many tissues, including the central nervous system. Our previous studies suggest that sustained AhR activation by TCDD and/or AhR deletion disrupts cerebellar granule neuron precursor (GNP) development. In the current study, to determine whether endogenous AhR controls GNP development in a cell-autonomous manner, we created a GNP-specific AhR deletion mouse, AhR(fx/fx) /Math1(CRE/+) (AhR CKO). Selective AhR deletion in GNPs produced abnormalities in proliferation and differentiation. Specifically, fewer GNPs were engaged in S-phase, as demonstrated by ∼25% reductions in thymidine (in vitro) and Bromodeoxyuridine (in vivo) incorporation. Furthermore, total granule neuron numbers in the internal granule layer at PND21 and PND60 were diminished in AhR conditional knockout (CKO) mice compared with controls. Conversely, differentiation was enhanced, including ∼40% increase in neurite outgrowth and 50% increase in GABARα6 receptor expression in deletion mutants. Our results suggest that AhR activity plays a role in regulating granule neuron number and differentiation, possibly by coordinating this GNP developmental transition. These studies provide novel insights for understanding the normal roles of AhR signaling during cerebellar granule cell neurogenesis and may have important implications for the effects of environmental factors in cerebellar dysgenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cerebellum/growth & development , Cerebellum/physiopathology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Receptors, Aryl Hydrocarbon/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Proliferation , Cells, Cultured , Cerebellum/pathology , Mice, Inbred C57BL , Mice, Knockout , Neurites/pathology , Neurites/physiology , Neurons/pathology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Receptors, GABA-A/metabolism
4.
Hum Mol Genet ; 24(20): 5805-27, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26220976

ABSTRACT

Many genes involved in brain development have been associated with human neurodevelopmental disorders, but underlying pathophysiological mechanisms remain undefined. Human genetic and mouse behavioral analyses suggest that ENGRAILED-2 (EN2) contributes to neurodevelopmental disorders, especially autism spectrum disorder. In mouse, En2 exhibits dynamic spatiotemporal expression in embryonic mid-hindbrain regions where monoamine neurons emerge. Considering their importance in neuropsychiatric disorders, we characterized monoamine systems in relation to forebrain neurogenesis in En2-knockout (En2-KO) mice. Transmitter levels of serotonin, dopamine and norepinephrine (NE) were dysregulated from Postnatal day 7 (P7) to P21 in En2-KO, though NE exhibited the greatest abnormalities. While NE levels were reduced ∼35% in forebrain, they were increased 40 -: 75% in hindbrain and cerebellum, and these patterns paralleled changes in locus coeruleus (LC) fiber innervation, respectively. Although En2 promoter was active in Embryonic day 14.5 -: 15.5 LC neurons, expression diminished thereafter and gene deletion did not alter brainstem NE neuron numbers. Significantly, in parallel with reduced NE levels, En2-KO forebrain regions exhibited reduced growth, particularly hippocampus, where P21 dentate gyrus granule neurons were decreased 16%, suggesting abnormal neurogenesis. Indeed, hippocampal neurogenic regions showed increased cell death (+77%) and unexpectedly, increased proliferation. Excess proliferation was restricted to early Sox2/Tbr2 progenitors whereas increased apoptosis occurred in differentiating (Dcx) neuroblasts, accompanied by reduced newborn neuron survival. Abnormal neurogenesis may reflect NE deficits because intra-hippocampal injections of ß-adrenergic agonists reversed cell death. These studies suggest that disruption of hindbrain patterning genes can alter monoamine system development and thereby produce forebrain defects that are relevant to human neurodevelopmental disorders.


Subject(s)
Dopaminergic Neurons/metabolism , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Neurogenesis , Prosencephalon/metabolism , Serotonergic Neurons/metabolism , Animals , Dopaminergic Neurons/physiology , Doublecortin Protein , Female , Gene Deletion , Humans , Male , Mice , Mice, Knockout , Norepinephrine/metabolism , Prosencephalon/growth & development , Prosencephalon/pathology , Prosencephalon/physiopathology , Serotonergic Neurons/physiology , Swimming
5.
PLoS One ; 7(7): e40914, 2012.
Article in English | MEDLINE | ID: mdl-22829897

ABSTRACT

ENGRAILED 2 (En2), a homeobox transcription factor, functions as a patterning gene in the early development and connectivity of rodent hindbrain and cerebellum, and regulates neurogenesis and development of monoaminergic pathways. To further understand the neurobiological functions of En2, we conducted neuroanatomical expression profiling of En2 wildtype mice. RTQPCR assays demonstrated that En2 is expressed in adult brain structures including the somatosensory cortex, hippocampus, striatum, thalamus, hypothalamus and brainstem. Human genetic studies indicate that EN2 is associated with autism. To determine the consequences of En2 mutations on mouse behaviors, including outcomes potentially relevant to autism, we conducted comprehensive phenotyping of social, communication, repetitive, and cognitive behaviors. En2 null mutants exhibited robust deficits in reciprocal social interactions as juveniles and adults, and absence of sociability in adults, replicated in two independent cohorts. Fear conditioning and water maze learning were impaired in En2 null mutants. High immobility in the forced swim test, reduced prepulse inhibition, mild motor coordination impairments and reduced grip strength were detected in En2 null mutants. No genotype differences were found on measures of ultrasonic vocalizations in social contexts, and no stereotyped or repetitive behaviors were observed. Developmental milestones, general health, olfactory abilities, exploratory locomotor activity, anxiety-like behaviors and pain responses did not differ across genotypes, indicating that the behavioral abnormalities detected in En2 null mutants were not attributable to physical or procedural confounds. Our findings provide new insight into the role of En2 in complex behaviors and suggest that disturbances in En2 signaling may contribute to neuropsychiatric disorders marked by social and cognitive deficits, including autism spectrum disorders.


Subject(s)
Autistic Disorder/physiopathology , Cognition Disorders/physiopathology , Animals , Autistic Disorder/genetics , Brain/physiology , Brain Stem/physiology , Cognition Disorders/genetics , Hippocampus/physiology , Homeodomain Proteins , Hypothalamus/physiology , Maze Learning/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins , Signal Transduction , Social Behavior , Somatosensory Cortex/physiology
6.
Development ; 139(3): 475-87, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22223678

ABSTRACT

During cerebral cortex development, precise control of precursor cell cycle length and cell cycle exit is required for balanced precursor pool expansion and layer-specific neurogenesis. Here, we defined the roles of cyclin-dependent kinase inhibitor (CKI) p57(KIP2), an important regulator of G1 phase, using deletion mutant mice. Mutant mice displayed macroencephaly associated with cortical hyperplasia during late embryogenesis and postnatal development. Embryonically, proliferation of radial glial cells (RGC) and intermediate precursors (IPC) was increased, expanding both populations, with greater effect on IPCs. Furthermore, cell cycle re-entry was increased during early corticogenesis, whereas cell cycle exit was augmented at middle stage. Consequently, neurogenesis was reduced early, whereas it was enhanced during later development. In agreement, the timetable of early neurogenesis, indicated by birthdating analysis, was delayed. Cell cycle dynamics analyses in mutants indicated that p57(KIP2) regulates cell cycle length in both RGCs and IPCs. By contrast, related CKI p27(KIP1) controlled IPC proliferation exclusively. Furthermore, p57(KIP2) deficiency markedly increased RGC and IPC divisions at E14.5, whereas p27(KIP1) increased IPC proliferation at E16.5. Consequently, loss of p57(KIP2) increased primarily layer 5-6 neuron production, whereas loss of p27(KIP1) increased neurons specifically in layers 2-5. In conclusion, our observations suggest that p57(KIP2) and p27(KIP1) control neuronal output for distinct cortical layers by regulating different stages of precursor proliferation, and support a model in which IPCs contribute to both lower and upper layer neuron generation.


Subject(s)
Cell Cycle , Cerebral Cortex/growth & development , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Neurogenesis , Neuroglia/metabolism , Stem Cells/metabolism , Animals , Brain/abnormalities , Brain/growth & development , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p57/genetics , Female , Mice , Sequence Deletion
7.
J Neurosci ; 31(31): 11144-58, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21813676

ABSTRACT

The precise control of motor neuron (MN) death and survival following initial innervation of skeletal muscle targets is a key step in sculpting a functional motor system, but how this is regulated at the level of individual motor pools remains unclear. Hepatocyte growth factor (HGF) and its receptor Met play key developmental roles in both muscle and MNs. We generated mice (termed "Nes-Met") in which met is inactivated from midembryonic stages onward in the CNS only. Adult animals showed motor behavioral defects suggestive of impaired innervation of pectoral muscles. Correspondingly, in neonatal spinal cords of Nes-Met mutants, we observed death of a discrete population of pea3-expressing MNs at brachial levels. Axonal tracing using pea3 reporter mice revealed a novel target muscle of pea3-expressing MNs: the pectoralis minor muscle. In Nes-Met mice, the pectoralis minor pool initially innervated its target muscle, but required HGF/Met for survival, hence for proper maintenance of muscle innervation. In contrast, HGF/Met was dispensable for the survival of neighboring Met-expressing MN pools, despite its earlier functions for their specification and axon growth. Our results demonstrate the exquisite degree to which outcomes of signaling by receptor tyrosine kinases are regulated on a cell-by-cell basis. They also provide a model for one way in which the multiplicity of neurotrophic factors may allow for regulation of MN numbers in a pool-specific manner.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Motor Neurons/drug effects , Motor Neurons/physiology , Nerve Growth Factors/pharmacology , Animals , Animals, Newborn , Axons/physiology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Hand Strength/physiology , Hepatocyte Growth Factor/genetics , Intermediate Filament Proteins/genetics , Male , Mice , Mice, Transgenic , Models, Biological , Motor Activity/genetics , Muscle Strength/genetics , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Mutation/genetics , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nestin , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-met/genetics , Psychomotor Performance/physiology , Reaction Time , Rotarod Performance Test , Spinal Cord/cytology , Transcription Factors/genetics
8.
J Neurochem ; 117(5): 892-903, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21443522

ABSTRACT

Hepatocyte growth factor (HGF) is known to promote the survival and foster neuritic outgrowth of different subpopulations of CNS neurons during development. Together with its corresponding receptor c-mesenchymal-epithelial transition factor (Met), it is expressed in the developing and the adult murine, rat and human CNS. We have studied the role of HGF in paradigms of retinal ganglion cell (RGC) regeneration and cell death in vitro and in vivo. After application of recombinant HGF in vitro, survival of serum-deprived RGC-5 cells and of growth factor-deprived primary RGC was significantly increased. This was shown to be correlated to the phosphorylation of c-Met and subsequent activation of serine/threonine protein kinase Akt and MAPK downstream signalling pathways involved in neuronal survival. Furthermore, neurite outgrowth of primary RGC was stimulated by HGF. In vivo, c-Met expression in RGC was up-regulated after optic nerve axotomy lesion. Here, treatment with HGF significantly improved survival of axotomized RGC and enhanced axonal regeneration after optic nerve crush. Our data demonstrates that exogenously applied HGF has a neuroprotective and regeneration-promoting function for lesioned CNS neurons. We provide strong evidence that HGF may represent a trophic factor for adult CNS neurons, which may play a role as therapeutic target in the treatment of neurotraumatic and neurodegenerative CNS disorders.


Subject(s)
Axons/drug effects , Hepatocyte Growth Factor/pharmacology , Nerve Regeneration/drug effects , Neurons/drug effects , Neuroprotective Agents , Retinal Ganglion Cells/drug effects , Animals , Animals, Genetically Modified , Axotomy , Blotting, Western , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , In Situ Hybridization , Mice , Nerve Crush , Optic Nerve/cytology , Optic Nerve/pathology , Optic Nerve Injuries/pathology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Rats , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
9.
Genesis ; 45(3): 107-12, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17304534

ABSTRACT

Extra-toes is a semidominant mutation that affects the Gli3 gene and provokes limb and brain abnormalities. Among the different alleles of this mutation, Xt(H) is due to a deletion that has not yet been fully characterized. Using a PCR-based strategy, we undertook a high-resolution mapping of this deletion and confirmed that Xt(H) is a null allele of Gli3. We further designed a PCR test to identify unequivocally heterozygous and homozygous embryos from their wild-type littermates. Despite the length of the Xt(H) deletion, available data on the mouse genome indicate that no genes other than Gli3 are deleted in Xt(H) mutants. Thus, the Xt(H) mutation can be used as a model for studying the effects that absence of Gli3 function has during development.


Subject(s)
Chromosome Mapping/methods , Gene Deletion , Kruppel-Like Transcription Factors/genetics , Models, Animal , Nerve Tissue Proteins/genetics , Alleles , Animals , Base Sequence , Heterozygote , Homozygote , Mice , Mice, Mutant Strains , Molecular Sequence Data , Polymerase Chain Reaction , Zinc Finger Protein Gli3
SELECTION OF CITATIONS
SEARCH DETAIL
...