134 results on '"Angélique Bordey"'
Search Results
2. Convergent and Divergent Mechanisms of Epileptogenesis in mTORopathies
- Author
-
Lena H. Nguyen and Angélique Bordey
- Subjects
neuron migration ,tuberous sclerosis complex ,focal cortical dysplasia ,GATOR1 complex ,in utero electroporation ,mTOR ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Human anatomy ,QM1-695 - Abstract
Hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1) due to mutations in genes along the PI3K-mTOR pathway and the GATOR1 complex causes a spectrum of neurodevelopmental disorders (termed mTORopathies) associated with malformation of cortical development and intractable epilepsy. Despite these gene variants’ converging impact on mTORC1 activity, emerging findings suggest that these variants contribute to epilepsy through both mTORC1-dependent and -independent mechanisms. Here, we review the literature on in utero electroporation-based animal models of mTORopathies, which recapitulate the brain mosaic pattern of mTORC1 hyperactivity, and compare the effects of distinct PI3K-mTOR pathway and GATOR1 complex gene variants on cortical development and epilepsy. We report the outcomes on cortical pyramidal neuronal placement, morphology, and electrophysiological phenotypes, and discuss some of the converging and diverging mechanisms responsible for these alterations and their contribution to epileptogenesis. We also discuss potential therapeutic strategies for epilepsy, beyond mTORC1 inhibition with rapamycin or everolimus, that could offer personalized medicine based on the gene variant.
- Published
- 2021
- Full Text
- View/download PDF
3. Rab27a-Dependent Paracrine Communication Controls Dendritic Spine Formation and Sensory Responses in the Barrel Cortex
- Author
-
Longbo Zhang, Xiaobing Zhang, Lawrence S. Hsieh, Tiffany V. Lin, and Angélique Bordey
- Subjects
exosomes ,dendritic spine ,excitatory synapse ,somatosensory cortex ,whisker stimulation ,Cytology ,QH573-671 - Abstract
Rab27a is an evolutionarily conserved small GTPase that regulates vesicle trafficking, and copy number variants of RAB27a are associated with increased risk of autism. However, the function of Rab27a on brain development is unknown. Here, we identified a form of paracrine communication that regulates spine development between distinct populations of developing cortical neurons. In the developing somatosensory cortex of mice, we show that decreasing Rab27a levels in late-born pyramidal neurons destined for layer (L) 2/3 had no cell-autonomous effect on their synaptic integration but increased excitatory synaptic transmission onto L4 neurons that receive somatosensory information. This effect resulted in an increased number of L4 neurons activated by whisker stimulation in juvenile mice. In addition, we found that Rab27a, the level of which decreases as neurons mature, regulates the release of small extracellular vesicles (sEVs) in developing neurons in vitro and decreasing Rab27a levels led to the accumulation of CD63-positive vesicular compartments in L2/3 neurons in vivo. Together, our study reveals that Rab27a-mediated paracrine communication regulates the development of synaptic connectivity, ultimately tuning responses to sensory stimulation, possibly via controlling the release of sEVs.
- Published
- 2021
- Full Text
- View/download PDF
4. mTORC1 Targets the Translational Repressor 4E-BP2, but Not S6 Kinase 1/2, to Regulate Neural Stem Cell Self-Renewal In Vivo
- Author
-
Nathaniel W. Hartman, Tiffany V. Lin, Longbo Zhang, Grace E. Paquelet, David M. Feliciano, and Angélique Bordey
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The mammalian target of rapamycin complex 1 (mTORC1) integrates signals important for cell growth, and its dysregulation in neural stem cells (NSCs) is implicated in several neurological disorders associated with abnormal neurogenesis and brain size. However, the function of mTORC1 on NSC self-renewal and the downstream regulatory mechanisms are ill defined. Here, we found that genetically decreasing mTORC1 activity in neonatal NSCs prevented their differentiation, resulting in reduced lineage expansion and aborted neuron production. Constitutive activation of the translational repressor 4E-BP1, which blocked cap-dependent translation, had similar effects and prevented hyperactive mTORC1 induction of NSC differentiation and promoted self-renewal. Although 4E-BP2 knockdown promoted NSC differentiation, p70 S6 kinase 1 and 2 (S6K1/S6K2) knockdown did not affect NSC differentiation but reduced NSC soma size and prevented hyperactive mTORC1-induced increase in soma size. These data demonstrate a crucial role of mTORC1 and 4E-BP for switching on and off cap-dependent translation in NSC differentiation.
- Published
- 2013
- Full Text
- View/download PDF
5. The Postnatal Subventricular Zone: A Source of New Cells in This Old Brain
- Author
-
Angélique Bordey
- Subjects
brain ,neurogenesis ,subventricular zone ,Surgery ,RD1-811 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Findings over the past decades demonstrating persistent neurogenesis in the adult brain have challenged the view of a fixed circuitry and raise hopes for self-renewal following brain injury. The subventricular zone (SVZ, also called subependymal layer, SEL) lining the lateral wall of the lateral ventricle is the largest germinal center where stem cells displaying astrocytic traits have been identified. These astrocyte-like cells ensheath neuroblasts, which migrate throughout the SVZ and along the rostral migratory stream to the olfactory bulb where they differentiate into interneurons. The cellular architecture of the SVZ has been essential for the development of hypotheses to explain how intercellular signaling and non-synaptic communication could regulate neurogenesis. An array of signaling molecules have recently been identified that may offer future strategies to promote neurogenesis and reroute neuroblasts to higher cognitive centers.
- Published
- 2005
- Full Text
- View/download PDF
6. Embryonic cerebrospinal fluid nanovesicles carry evolutionarily conserved molecules and promote neural stem cell amplification.
- Author
-
David M Feliciano, Shiliang Zhang, Carole M Nasrallah, Steven N Lisgo, and Angélique Bordey
- Subjects
Medicine ,Science - Abstract
During brain development, neural stem cells (NSCs) receive on-or-off signals important for regulating their amplification and reaching adequate neuron density. However, how a coordinated regulation of intracellular pathways and genetic programs is achieved has remained elusive. Here, we found that the embryonic (e) CSF contains 10¹² nanoparticles/ml (77 nm diameter), some of which were identified as exosome nanovesicles that contain evolutionarily conserved molecules important for coordinating intracellular pathways. eCSF nanovesicles collected from rodent and human embryos encapsulate protein and microRNA components of the insulin-like growth factor (IGF) signaling pathway. Supplementation of eCSF nanovesicles to a mixed culture containing eNSCs activated the IGF-mammalian target of rapamycin complex 1 (mTORC1) pathway in eNSCs and expanded the pool of proliferative eNSCs. These data show that the eCSF serves as a medium for the distribution of nanovesicles, including exosomes, and the coordinated transfer of evolutionary conserved molecules that regulate eNSC amplification during corticogenesis.
- Published
- 2014
- Full Text
- View/download PDF
7. FMRP S499 is phosphorylated independent of mTORC1-S6K1 activity.
- Author
-
Christopher M Bartley, Rachel A O'Keefe, and Angélique Bordey
- Subjects
Medicine ,Science - Abstract
Hyperactive mammalian target of rapamycin (mTOR) is associated with cognitive deficits in several neurological disorders including tuberous sclerosis complex (TSC). The phosphorylation of the mRNA-binding protein FMRP reportedly depends on mTOR complex 1 (mTORC1) activity via p70 S6 kinase 1 (S6K1). Because this phosphorylation is thought to regulate the translation of messages important for synaptic plasticity, we explored whether FMRP phosphorylation of the S6K1-dependent residue (S499) is altered in TSC and states of dysregulated TSC-mTORC1 signaling. Surprisingly, we found that FMRP S499 phosphorylation was unchanged in heterozygous and conditional Tsc1 knockout mice despite significantly elevated mTORC1-S6K1 activity. Neither up- nor down-regulation of the mTORC1-S6K1 axis in vivo or in vitro had any effect on phospho-FMRP S499 levels. In addition, FMRP S499 phosphorylation was unaltered in S6K1-knockout mice. Collectively, these data strongly suggest that FMRP S499 phosphorylation is independent of mTORC1-S6K1 activity and is not altered in TSC.
- Published
- 2014
- Full Text
- View/download PDF
8. miR-132 enhances dendritic morphogenesis, spine density, synaptic integration, and survival of newborn olfactory bulb neurons.
- Author
-
Manavendra Pathania, Juan Torres-Reveron, Lily Yan, Tomoki Kimura, Tiffany V Lin, Valerie Gordon, Zhao-Qian Teng, Xinyu Zhao, Tudor A Fulga, David Van Vactor, and Angélique Bordey
- Subjects
Medicine ,Science - Abstract
An array of signals regulating the early stages of postnatal subventricular zone (SVZ) neurogenesis has been identified, but much less is known regarding the molecules controlling late stages. Here, we investigated the function of the activity-dependent and morphogenic microRNA miR-132 on the synaptic integration and survival of olfactory bulb (OB) neurons born in the neonatal SVZ. In situ hybridization revealed that miR-132 expression occurs at the onset of synaptic integration in the OB. Using in vivo electroporation we found that sequestration of miR-132 using a sponge-based strategy led to a reduced dendritic complexity and spine density while overexpression had the opposite effects. These effects were mirrored with respective changes in the frequency of GABAergic and glutamatergic synaptic inputs reflecting altered synaptic integration. In addition, timely directed overexpression of miR-132 at the onset of synaptic integration using an inducible approach led to a significant increase in the survival of newborn neurons. These data suggest that miR-132 forms the basis of a structural plasticity program seen in SVZ-OB postnatal neurogenesis. miR-132 overexpression in transplanted neurons may thus hold promise for enhancing neuronal survival and improving the outcome of transplant therapies.
- Published
- 2012
- Full Text
- View/download PDF
9. Expression of 4E-BP1 in juvenile mice alleviates mTOR-induced neuronal dysfunction and epilepsy
- Author
-
Heather A. Born, Lena H. Nguyen, Youfen Xu, Travorn Mahadeo, Angélique Bordey, Tiffany V. Lin, Anne E. Anderson, and Longbo Zhang
- Subjects
Cell Cycle Proteins ,Mice ,Epilepsy ,Seizures ,Gene expression ,Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels ,medicine ,Animals ,Humans ,Phosphorylation ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Neurons ,biology ,Effector ,business.industry ,TOR Serine-Threonine Kinases ,Brain ,Translation (biology) ,medicine.disease ,Electrophysiology ,biology.protein ,Original Article ,Neurology (clinical) ,business ,Neuroscience - Abstract
Hyperactivation of the mTOR pathway during foetal neurodevelopment alters neuron structure and function, leading to focal malformation of cortical development and intractable epilepsy. Recent evidence suggests a role for dysregulated cap-dependent translation downstream of mTOR signalling in the formation of focal malformation of cortical development and seizures. However, it is unknown whether modifying translation once the developmental pathologies are established can reverse neuronal abnormalities and seizures. Addressing these issues is crucial with regards to therapeutics because these neurodevelopmental disorders are predominantly diagnosed during childhood, when patients present with symptoms. Here, we report increased phosphorylation of the mTOR effector and translational repressor, 4E-BP1, in patient focal malformation of cortical development tissue and in a mouse model of focal malformation of cortical development. Using temporally regulated conditional gene expression systems, we found that expression of a constitutively active form of 4E-BP1 that resists phosphorylation by focal malformation of cortical development in juvenile mice reduced neuronal cytomegaly and corrected several neuronal electrophysiological alterations, including depolarized resting membrane potential, irregular firing pattern and aberrant expression of HCN4 ion channels. Further, 4E-BP1 expression in juvenile focal malformation of cortical development mice after epilepsy onset resulted in improved cortical spectral activity and decreased spontaneous seizure frequency in adults. Overall, our study uncovered a remarkable plasticity of the juvenile brain that facilitates novel therapeutic opportunities to treat focal malformation of cortical development-related epilepsy during childhood with potentially long-lasting effects in adults.
- Published
- 2021
- Full Text
- View/download PDF
10. Loss of PTEN phosphorylation via single point mutation alters cortical connectivity and behaviour
- Author
-
Matthew Binder and Angélique Bordey
- Subjects
Male ,Neurons ,Threonine ,Alanine ,PTEN Phosphohydrolase ,Lipids ,Scientific Commentary ,Mice ,Tensins ,Animals ,Humans ,Point Mutation ,Original Article ,Neurology (clinical) ,Phosphorylation - Abstract
The lipid phosphatase PTEN (phosphatase and tensin homologue on chromosome 10) is a key tumour suppressor gene and an important regulator of neuronal signalling. PTEN mutations have been identified in patients with autism spectrum disorders, characterized by macrocephaly, impaired social interactions and communication, repetitive behaviour, intellectual disability, and epilepsy. PTEN enzymatic activity is regulated by a cluster of phosphorylation sites at the C-terminus of the protein. Here, we focused on the role of PTEN T366 phosphorylation and generated a knock-in mouse line in which Pten T366 was substituted with alanine (Pten(T366A/T366A)). We identify that phosphorylation of PTEN at T366 controls neuron size and connectivity of brain circuits involved in sensory processing. We show in behavioural tests that Pten(T366/T366A) mice exhibit cognitive deficits and selective sensory impairments, with significant differences in male individuals. We identify restricted cellular overgrowth of cortical neurons in Pten(T366A/T366A) brains, linked to increases in both dendritic arborization and soma size. In a combinatorial approach of anterograde and retrograde monosynaptic tracing using rabies virus, we characterize differences in connectivity to the primary somatosensory cortex of Pten(T366A/T366A) brains, with imbalances in long-range cortico-cortical input to neurons. We conclude that phosphorylation of PTEN at T366 controls neuron size and connectivity of brain circuits involved in sensory processing and propose that PTEN T366 signalling may account for a subset of autism-related functions of PTEN.
- Published
- 2022
11. Small Extracellular Vesicles Control Dendritic Spine Development through Regulation of HDAC2 Signaling
- Author
-
Rémy Sadoul, Qianying Yuan, TuKiet T. Lam, Angélique Bordey, Longbo Zhang, and Tiffany V. Lin
- Subjects
Male ,Proteomics ,0301 basic medicine ,Cytoplasm ,Dendritic spine ,Dendritic Spines ,viruses ,Primary Cell Culture ,Paracrine Communication ,Histone Deacetylase 2 ,Biology ,Exosomes ,Exosome ,Synapse ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Research Articles ,General Neuroscience ,Gene Expression Regulation, Developmental ,virus diseases ,respiratory system ,Microvesicles ,Cortex (botany) ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Synapses ,Excitatory postsynaptic potential ,Female ,Neuron ,Extracellular Space ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
The release of small extracellular vesicles (sEVs) has recently been reported, but knowledge of their function in neuron development remains limited. Using LC-MS/MS, we found that sEVs released from developing cortical neuronsin vitroobtained from mice of both sexes were enriched in cytoplasm, exosome, and protein-binding and DNA/RNA-binding pathways. The latter included HDAC2, which was of particular interest, because HDAC2 regulates spine development, and populations of neurons expressing different levels of HDAC2 co-existin vivoduring the period of spine growth. Here, we found that HDAC2 levels decrease in neurons as they acquire synapses and that sEVs from HDAC2-rich neurons regulate HDAC2 signaling in HDAC2-low neurons possibly through HDAC2 transfer. This regulation led to a transcriptional decrease in HDAC2 synaptic targets and the density of excitatory synapses. These data suggest that sEVs provide inductive cell-cell signaling that coordinates the development of dendritic spines via the activation of HDAC2-dependent transcriptional programs.SIGNIFICANCE STATEMENTA role of small extracellular vesicles (sEVs; also called exosomes) in neuronal development is of particular interest, because sEVs could provide a major signaling modality between developing neurons when synapses are not fully functional or immature. However, knowledge of sEVs on neuron, and more precisely spine development, is limited. We provide several lines of evidence that sEVs released from developing cortical neurons regulate the development of dendritic spines via the regulation of HDAC2 signaling. This paracrine communication is temporally restricted during development because of the age-dependent decrease in sEV release as neurons mature and acquire spines.
- Published
- 2021
- Full Text
- View/download PDF
12. Imaging and optogenetic modulation of vascular mural cells in the live brain
- Author
-
Lei Tong, Katie N. Murray, Robert A. Hill, Peng Yuan, Angélique Bordey, Jaime Grutzendler, and Eyiyemisi C. Damisah
- Subjects
Cell physiology ,0303 health sciences ,Context (language use) ,Biology ,Optogenetics ,General Biochemistry, Genetics and Molecular Biology ,Mural cell ,03 medical and health sciences ,0302 clinical medicine ,Calcium imaging ,medicine.anatomical_structure ,Cerebral blood flow ,cardiovascular system ,medicine ,Myocyte ,Pericyte ,Neuroscience ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Mural cells (smooth muscle cells and pericytes) are integral components of brain blood vessels that play important roles in vascular formation, blood-brain barrier maintenance, and regulation of regional cerebral blood flow (rCBF). These cells are implicated in conditions ranging from developmental vascular disorders to age-related neurodegenerative diseases. Here we present complementary tools for cell labeling with transgenic mice and organic dyes that allow high-resolution intravital imaging of the different mural cell subtypes. We also provide detailed methodologies for imaging of spontaneous and neural activity-evoked calcium transients in mural cells. In addition, we describe strategies for single- and two-photon optogenetics that allow manipulation of the activity of individual and small clusters of mural cells. Together with measurements of diameter and flow in individual brain microvessels, calcium imaging and optogenetics allow the investigation of pericyte and smooth muscle cell physiology and their role in regulating rCBF. We also demonstrate the utility of these tools to investigate mural cells in the context of Alzheimer's disease and cerebral ischemia mouse models. Thus, these methods can be used to reveal the functional and structural heterogeneity of mural cells in vivo, and allow detailed cellular studies of the normal function and pathophysiology of mural cells in a variety of disease models. The implementation of this protocol can take from several hours to days depending on the intended applications.
- Published
- 2020
- Full Text
- View/download PDF
13. CD-1 Outbred Mice Produce Less Variable Ultrasonic Vocalizations Than FVB Inbred Mice, While Displaying a Similar Developmental Trajectory
- Author
-
Matthew S. Binder, Hannah D. Shi, and Angélique Bordey
- Subjects
Psychiatry ,Psychiatry and Mental health ,Developmental trajectory ,communication ,behavior reproducibility ,neurodevelopmental disorders ,RC435-571 ,Physiology ,Biology ,USV ,Original Research ,methods ,neonatal vocalization - Abstract
The production of ultrasonic vocalizations (USVs) in neonatal mice is a critical means of communication that is used to elicit maternal care. Alterations in neonatal USV production is also an indicator of neurological deficits. However, USVs have been predominately assessed in inbred animals and are significantly understudied in outbred mice, even though outbred animals better represent the genetic diversity of humans and are used in several neurological disorder models. To determine the reproducibility of USVs across models, we compared male and female CD-1 (outbred) and FVB (inbred) mice on postnatal days (PD) 4, 8, 12, 16, and 20. We found that CD-1 and FVB mice displayed a similar developmental trajectory of USVs. However, CD1 mice emitted more USVs on PD 12 than FVB mice. In addition, FVB mice emitted a longer duration of calls on PD 4 and 8 and a higher overall maximum and minimum frequency of USVs than CD-1 mice. No differences in mean amplitude were found between groups. We also detected numerous significant differences between outbred and inbred mice when comparing each group's call composition. We next assessed the relative variability of mouse vocalizations between groups, finding that outbred mice were less variable than inbred mice. For the spectral and temporal characteristics of the USVs, variability was similar between groups. Altogether, we found that CD-1 outbred mice display a similar, if not lower, degree of variability than FVB inbred mice when assessing neonatal USVs.
- Published
- 2021
14. Genetic expression of 4E-BP1 in juvenile mice alleviates mTOR-induced neuronal dysfunction and epilepsy
- Author
-
Anne E. Anderson, Lena H. Nguyen, Heather A. Born, Youfen Xu, Angélique Bordey, Longbo Zhang, Travorn Mahadeo, and Tiffany V. Lin
- Subjects
Fetus ,Electrophysiology ,Epilepsy ,Hyperactivation ,Gene expression ,medicine ,Phosphorylation ,Translation (biology) ,Biology ,medicine.disease ,Neuroscience ,PI3K/AKT/mTOR pathway - Abstract
SUMMARYHyperactivation of mTOR signaling during fetal neurodevelopment alters neuron structure and function, leading to focal malformation of cortical development (FMCD) and intractable epilepsy. Recent evidence suggests increased cap-dependent translation downstream of mTOR contributes to FMCD formation and seizures. However, whether reducing overactive translation once the developmental pathologies are established reverses neuronal abnormalities and seizures is unknown. Here, we found that the translational repressor 4E-BP1, which is inactivated by mTOR-mediated phosphorylation, is hyperphosphorylated in patient FMCD tissue and in a mouse model of FMCD. Expressing constitutive active 4E-BP1 to repress aberrant translation in juvenile mice with FMCD reduced neuronal cytomegaly and corrected several electrophysiological alterations, including depolarized resting membrane potential, irregular firing pattern, and aberrant HCN4 channel expression. This was accompanied by improved cortical spectral activity and decreased seizures. Although mTOR controls multiple pathways, our study shows that targeting 4E-BP1-mediated translation alone is sufficient to alleviate neuronal dysfunction and ongoing epilepsy.
- Published
- 2021
- Full Text
- View/download PDF
15. <scp>GATOR</scp> opathies: The role of amino acid regulatory gene mutations in epilepsy and cortical malformations
- Author
-
Angélique Bordey, Peter B. Crino, Philip H. Iffland, and Vincent J Carson
- Subjects
0301 basic medicine ,Article ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,medicine ,Animals ,Humans ,Megalencephaly ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,biology ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,GTPase-Activating Proteins ,Cortical dysplasia ,NPRL3 ,medicine.disease ,DEPDC5 ,Malformations of Cortical Development ,030104 developmental biology ,Neurology ,Mutation ,biology.protein ,Cancer research ,Neurology (clinical) ,030217 neurology & neurosurgery ,RHEB - Abstract
The mechanistic target of rapamycin (mTOR) pathway has been implicated in a growing number of malformations of cortical development (MCD) associated with intractable epilepsy. Mutations in single genes encoding mTOR pathway regulatory proteins have been linked to MCD such as focal cortical dysplasia (FCD) types IIa and IIb, hemimegalencephaly (HME), and megalencephaly. Recent studies have demonstrated that the GATOR1 protein complex, comprised of DEPDC5, NPRL3, and NPRL2, plays a pivotal role in regulating mTOR signaling in response to cellular amino acid levels and that mutations in DEPDC5, NPRL3, or NPRL2 are linked to FCD, HME, and seizures. Histopathological analysis of FCD and HME tissue specimens resected from individuals harboring DEPDC5, NPRL3, or NPRL2 gene mutations reveals hyperactivation of mTOR pathway signaling. Family pedigrees carrying mutations in either DEPDC5 or NPRL3 share clinical phenotypes of epilepsy and MCD, as well as intellectual and neuropsychiatric disabilities. Interestingly, some individuals with seizures associated with DEPDC5, NPRL3, or NPRL2 variants exhibit normal brain imaging suggesting either occult MCD or a role for these genes in non-lesional neocortical epilepsy. Mouse models resulting from knockdown or knockout of either Depdc5 or Nprl3 exhibit altered cortical lamination, neuronal dysmorphogenesis, and enhanced neuronal excitability as reported in models resulting from direct mTOR activation through expression of its canonical activator RHEB. The role of the GATOR1 proteins in regulating mTOR signaling suggest plausible options for mTOR inhibition in the treatment of epilepsy associated with mutations in DEPDC5, NPRL3, or NPRL2.
- Published
- 2019
- Full Text
- View/download PDF
16. In utero electroporation-based translating ribosome affinity purification identifies age-dependent mRNA expression in cortical pyramidal neurons
- Author
-
Tianxiang Huang, Angélique Bordey, Tiffany V. Lin, Matthew R. Sarkisian, Longbo Zhang, Xuan Gong, Lena H. Nguyen, Joshua J. Breunig, and Gi Bum Kim
- Subjects
Male ,Ribosomal Proteins ,0301 basic medicine ,Doublecortin Protein ,Ribosomal Protein L10 ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Morphogenesis ,Embryonic Development ,Ribosome ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Ribosomal protein ,Animals ,RNA, Messenger ,Oligonucleotide Array Sequence Analysis ,Gene knockdown ,biology ,Gene Expression Profiling ,Pyramidal Cells ,General Neuroscience ,Electroporation ,Brain ,Gene Expression Regulation, Developmental ,Somatosensory Cortex ,General Medicine ,Synapsins ,Cell biology ,Corticogenesis ,030104 developmental biology ,Protein Biosynthesis ,biology.protein ,Female ,TBR1 ,Disks Large Homolog 4 Protein ,030217 neurology & neurosurgery - Abstract
We combined translating ribosome affinity purification (TRAP) with in utero electroporation (IUE), called iTRAP to identify the molecular profile of specific neuronal populations during neonatal development without the need for viral approaches and FACS sorting. We electroporated a plasmid encoding EGFP-tagged ribosomal protein L10a at embryonic day (E) 14–15 to target layer 2–4 cortical neurons of the somatosensory cortex. At three postnatal (P) ages—P0, P7, and P14—when morphogenesis occurs and synapses are forming, TRAP and molecular profiling was performed from electroporated regions. We found that ribosome bound (Ribo)-mRNAs from ~7,300 genes were significantly altered over time and included classical neuronal genes known to decrease (e.g., Tbrl, Dcx) or increase (e.g., Eno2, Camk2a, Synl) as neurons mature. This approach led to the identification of specific developmental patterns for Ribo-mRNAs not previously reported to be developmentally regulated in neurons, providing rationale for future examination of their role in selective biological processes. These include upregulation of Lynxl, Nrnl, Cntnapl over time; downregulation of St8sia2 and Draxin; and bidirectional changes to Fkbplb. iTRAP is a versatile approach that allows researchers to easily assess the molecular profile of specific neuronal populations in selective brain regions under various conditions, including overexpression and knockdown of target genes, and in disease settings.
- Published
- 2019
- Full Text
- View/download PDF
17. mTOR Hyperactivity Levels Influence the Severity of Epilepsy and Associated Neuropathology in an Experimental Model of Tuberous Sclerosis Complex and Focal Cortical Dysplasia
- Author
-
Travorn Mahadeo, Angélique Bordey, and Lena H. Nguyen
- Subjects
Male ,0301 basic medicine ,mTORC1 ,Neuropathology ,Severity of Illness Index ,Mice ,03 medical and health sciences ,Tuberous sclerosis ,Epilepsy ,0302 clinical medicine ,Tuberous Sclerosis ,medicine ,Animals ,Mechanistic target of rapamycin ,Research Articles ,PI3K/AKT/mTOR pathway ,biology ,business.industry ,TOR Serine-Threonine Kinases ,General Neuroscience ,Electroencephalography ,Cortical dysplasia ,medicine.disease ,Malformations of Cortical Development ,Disease Models, Animal ,030104 developmental biology ,biology.protein ,Cancer research ,Female ,business ,030217 neurology & neurosurgery ,RHEB - Abstract
Tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) are focal malformations of cortical development (FMCDs) that are highly associated with intractable epilepsy. TSC and FCD are mTORopathies caused by a spectrum of pathogenic variants in the mechanistic target of rapamycin (mTOR) pathway genes leading to differential activation of mTOR signaling. However, whether the degree of mTOR hyperactivity influences disease severity remains unclear. Here, we examined the effects of differential mTOR hyperactivity levels on epilepsy and associated neuropathology in a mouse model of TSC and FCD. Constitutively active Rheb (RhebCA), the canonical activator of mTOR complex 1 (mTORC1), was expressed in mouse embryos of either sex viain uteroelectroporation at low, intermediate, and high concentrations to induce different mTORC1 activity levels in developing cortical neurons. We found that RhebCAexpression induced mTORC1 hyperactivation and increased neuronal soma size and misplacement in a dose-dependent manner. No seizures were detected in the low RhebCAmice, whereas the intermediate and high RhebCAmice displayed spontaneous, recurrent seizures that significantly increased with higher RhebCAconcentrations. Seizures were associated with a global increase in microglial activation that was notably higher in the regions containing RhebCA-expressing neurons. These data demonstrate that neuronal mTOR hyperactivity levels influence the severity of epilepsy and associated neuropathology in experimental TSC and FCD. Overall, these findings highlight the importance of evaluating the outcome of individual variants on mTOR activity levels and support personalized medicine strategies based on patient variants and mTOR activity level for TSC, FCD, and potentially other mTORopathies.SIGNIFICANCE STATEMENTTuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) are epileptogenic cortical malformations caused by pathogenic variants in mechanistic target of rapamycin (mTOR) pathway genes leading to differential mTOR hyperactivation. Here, we present novel findings that neuronal mTOR hyperactivity levels correlate with the severity of epilepsy and associated neuropathology in a mouse model of TSC and FCD. Our findings suggest the need to evaluate the outcome of individual variants on mTOR activity levels in clinical assessments and support personalized medicine strategies based on patient variants and mTOR activity level. Additionally, we present useful modifications to a previously described mouse model of TSC and FCD that allows for titration of seizure frequency and generation of a mild to severe epilepsy phenotype as applicable for preclinical drug testing and mechanistic studies.
- Published
- 2019
- Full Text
- View/download PDF
18. Rab27a-Dependent Paracrine Communication Controls Dendritic Spine Formation and Sensory Responses in the Barrel Cortex
- Author
-
Tiffany V. Lin, Longbo Zhang, Angélique Bordey, Xiaobing Zhang, and Lawrence S. Hsieh
- Subjects
0301 basic medicine ,endocrine system ,Dendritic spine ,Sensory Receptor Cells ,somatosensory cortex ,Dendritic Spines ,Paracrine Communication ,Stimulation ,Sensory system ,Gestational Age ,excitatory synapse ,exosomes ,Biology ,Somatosensory system ,Synaptic Transmission ,Article ,rab27 GTP-Binding Proteins ,03 medical and health sciences ,Extracellular Vesicles ,Mice ,0302 clinical medicine ,Excitatory synapse ,Pregnancy ,Animals ,whisker stimulation ,lcsh:QH301-705.5 ,Cells, Cultured ,Sensory stimulation therapy ,dendritic spine ,Pyramidal Cells ,Excitatory Postsynaptic Potentials ,Gene Expression Regulation, Developmental ,General Medicine ,Barrel cortex ,Cell biology ,030104 developmental biology ,lcsh:Biology (General) ,Vibrissae ,Female ,030217 neurology & neurosurgery - Abstract
Rab27a is an evolutionarily conserved small GTPase that regulates vesicle trafficking, and copy number variants of RAB27a are associated with increased risk of autism. However, the function of Rab27a on brain development is unknown. Here, we identified a form of paracrine communication that regulates spine development between distinct populations of developing cortical neurons. In the developing somatosensory cortex of mice, we show that decreasing Rab27a levels in late-born pyramidal neurons destined for layer (L) 2/3 had no cell-autonomous effect on their synaptic integration but increased excitatory synaptic transmission onto L4 neurons that receive somatosensory information. This effect resulted in an increased number of L4 neurons activated by whisker stimulation in juvenile mice. In addition, we found that Rab27a, the level of which decreases as neurons mature, regulates the release of small extracellular vesicles (sEVs) in developing neurons in vitro and decreasing Rab27a levels led to the accumulation of CD63-positive vesicular compartments in L2/3 neurons in vivo. Together, our study reveals that Rab27a-mediated paracrine communication regulates the development of synaptic connectivity, ultimately tuning responses to sensory stimulation, possibly via controlling the release of sEVs.
- Published
- 2021
- Full Text
- View/download PDF
19. Ectopic HCN4 expression drives mTOR-dependent epilepsy in mice
- Author
-
Dennis D. Spencer, Stephanie A. Getz, Lawrence S. Hsieh, John H. Wen, Ying Wang, Longbo Zhang, Lena H. Nguyen, Juan Torres-Reveron, and Angélique Bordey
- Subjects
0301 basic medicine ,Gene isoform ,Potassium Channels ,Muscle Proteins ,Gating ,Article ,03 medical and health sciences ,Tuberous sclerosis ,Epilepsy ,0302 clinical medicine ,Tuberous Sclerosis ,Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels ,medicine ,Humans ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,biology ,TOR Serine-Threonine Kinases ,General Medicine ,Cortical dysplasia ,medicine.disease ,Potassium channel ,030104 developmental biology ,Malformations of Cortical Development, Group I ,biology.protein ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The causative link between focal cortical malformations (FCM) and epilepsy is well-accepted, especially among patients with focal cortical dysplasia type II (FCDII) and tuberous sclerosis complex (TSC). However, the mechanisms underlying seizures remain unclear. Using a mouse model of TSC- and FCDII-associated FCM, we showed that FCM neurons were responsible for seizure activity via their unexpected abnormal expression of the hyperpolarization-activated cyclic nucleotide-gated potassium channel isoform 4 (HCN4), which is normally not present in cortical pyramidal neurons after birth. Increasing intracellular cAMP concentrations, which preferentially affects HCN4 gating relative to the other isoforms, drove repetitive firing of FCM neurons but not control pyramidal neurons. Ectopic HCN4 expression was dependent on the mechanistic target of rapamycin (mTOR), preceded the onset of seizures, and was also found in diseased neurons in tissue resected from patients with TSC and FCDII. Finally, blocking HCN4 channel activity in FCM neurons prevented epilepsy in the mouse model. These findings suggest that HCN4 play a main role in seizure and identify a cAMP-dependent seizure mechanism in TSC and FCDII. Furthermore, the unique expression of HCN4 exclusively in FCM neurons suggests that gene therapy targeting HCN4 might be effective in reducing seizures in FCDII or TSC.
- Published
- 2020
- Full Text
- View/download PDF
20. Treating Post-Traumatic Seizures to Limit Tau Accumulation in Larval Zebrafish
- Author
-
Angélique Bordey
- Subjects
Dynamins ,0301 basic medicine ,Green Fluorescent Proteins ,tau Proteins ,Animals, Genetically Modified ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Seizures ,Brain Injuries, Traumatic ,Zebrafish larvae ,medicine ,Animals ,Zebrafish ,Cell Death ,business.industry ,medicine.disease ,Current Literature in Basic Science ,nervous system diseases ,030104 developmental biology ,Tauopathies ,Larva ,Anticonvulsants ,Neurology (clinical) ,Tauopathy ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Traumatic brain injury (TBI) is a prominent risk factor for dementias including tauopathies like chronic traumatic encephalopathy (CTE). The mechanisms that promote prion-like spreading of Tau aggregates after TBI are not fully understood, in part due to lack of tractable animal models. Here, we test the putative role of seizures in promoting the spread of tauopathy. We introduce 'tauopathy reporter' zebrafish expressing a genetically encoded fluorescent Tau biosensor that reliably reports accumulation of human Tau species when seeded via intraventricular brain injections. Subjecting zebrafish larvae to a novel TBI paradigm produced various TBI features including cell death, post-traumatic seizures, and Tau inclusions. Bath application of dynamin inhibitors or anticonvulsant drugs rescued TBI-induced tauopathy and cell death. These data suggest a role for seizure activity in the prion-like seeding and spreading of tauopathy following TBI. Further work is warranted regarding anti-convulsants that dampen post-traumatic seizures as a route to moderating subsequent tauopathy.Traumatic brain injury can result from direct head concussions, rapid head movements, or a blast wave generated by an explosion. Traumatic brain injury often causes seizures in the short term and is a risk factor for certain dementias, including Alzheimer’s disease and chronic traumatic encephalopathy in the long term. A protein called Tau undergoes a series of chemical changes in these dementias that makes it accumulate, form toxic filaments and kill neurons. The toxic abnormal Tau proteins are initially found only in certain regions of the brain, but they spread as the disease progresses. Previous studies in Alzheimer’s disease and other diseases where Tau proteins are abnormal suggest that Tau can spread between neighboring neurons and this can be promoted by neuron activity. However, scientists do not know whether similar mechanisms are at work following traumatic brain injury. Given that seizures are very common following traumatic brain injury, could they be partly responsible for promoting dementia? To investigate this, researchers need animal models in which they can measure neural activity associated with traumatic brain injury and observe the spread of abnormal Tau proteins. Alyenbaawi et al. engineered zebrafish so that their Tau proteins would be fluorescent, making it possible to track the accumulation of aggregated Tau protein in the brain. Next, they invented a simple way to perform traumatic brain injury on zebrafish larvae by using a syringe to produce a pressure wave. After this procedure, many of the fish exhibited features consistent with progression towards dementia, and seizure-like behaviors. The results showed that post-traumatic seizures are linked to the spread of aggregates of abnormal Tau following traumatic brain injury. Alyenbaawi et al. also found that anticonvulsant drugs can lower the levels of abnormal Tau proteins in neurons, preventing cell death, and could potentially ameliorate dementias associated with traumatic brain injury. These drugs are already being used to prevent post-traumatic epilepsy, but more research is needed to confirm whether they reduce the risk or severity of Tau-related neurodegeneration.
- Published
- 2021
- Full Text
- View/download PDF
21. Treating Seizures With Low-Frequency Electrical Stimulation
- Author
-
Angélique Bordey
- Subjects
0301 basic medicine ,business.industry ,Stimulation ,Low frequency ,Hippocampal formation ,Temporal lobe ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Medicine ,Neurology (clinical) ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Hippocampal Low-Frequency Stimulation Prevents Seizure Generation in a Mouse Model of Mesial Temporal Lobe EpilepsyPaschen E, Elgueta C, Heining K, et al. Elife. 2020;9:e54518. doi:10.7554/eLife.54...
- Published
- 2021
- Full Text
- View/download PDF
22. Semi-natural housing rescues social behavior and reduces repetitive exploratory behavior of BTBR autistic-like mice
- Author
-
Matthew S. Binder and Angelique Bordey
- Subjects
Medicine ,Science - Abstract
Abstract Environmental enrichment confers numerous benefits when implemented in murine models and can reduce behavioral symptomatology in models of disease, such as autism spectrum disorder (ASD). However, previous work did not examine the impact of early-life environmental enrichment on each core feature of ASD. We thus implemented a social and physical enrichment at birth, modeling a semi-natural housing, and examined its impact on communicative, social, sensory, and repetitive behaviors using BTBR (autistic-like) and C57BL/6 J (B6, wildtype) mice, comparing them to standard housing conditions. We found that environmental enrichment alleviated the social deficit of juvenile BTBR mice and reduced their repetitive exploratory behavior but did not affect their rough versus smooth texture preference nor the number of maternal isolation-induced pup calls. Environmental enrichment only affected the call characteristics of B6 mice. One interpretation of these data is that early-life environmental enrichment has significant therapeutic potential to treat selective core features of ASD. Another interpretation is that reducing environmental complexity causes selective behavioral deficits in ASD-prone mice suggesting that current standard housing may be suboptimal. Overall, our data illustrate the extent to which the environment influences behavior and highlights the importance of considering housing conditions when designing experiments and interpreting behavioral results.
- Published
- 2023
- Full Text
- View/download PDF
23. Outbred CD1 mice are as suitable as inbred C57BL/6J mice in performing social tasks
- Author
-
Angélique Bordey, Paul J. Lombroso, Laura Miyares, Lawrence S. Hsieh, and John H. Wen
- Subjects
Male ,0301 basic medicine ,Population ,Behavioral testing ,CD1 ,Male mice ,Mice, Inbred Strains ,C57bl 6j ,Article ,Developmental psychology ,03 medical and health sciences ,0302 clinical medicine ,Task Performance and Analysis ,Animals ,Autistic Disorder ,Social Behavior ,education ,Genetics ,education.field_of_study ,Behavior, Animal ,General Neuroscience ,Novel object ,Novelty ,Social relation ,Disease Models, Animal ,030104 developmental biology ,Exploratory Behavior ,Psychology ,030217 neurology & neurosurgery - Abstract
Inbred mouse strains have been used preferentially for behavioral testing over outbred counterparts, even though outbred mice reflect the genetic diversity in the human population better. Here, we compare the sociability of widely available outbred CD1 mice with the commonly used inbred C57BL/6J (C57) mice in the one-chamber social interaction test and the three-chamber sociability test. In the one-chamber task, intra-strain pairs of juvenile, non-littermate, male CD1 or C57 mice display a series of social and aggressive behaviors. While CD1 and C57 pairs spend equal amount of time socializing, CD1 pairs spend significantly more time engaged in aggressive behaviors than C57 mice. In the three-chamber task, sociability of C57 mice was less dependent on acclimation paradigms than CD1 mice. Following acclimation to all three chambers, both groups of age-matched male mice spent more time in the chamber containing a stranger mouse than in the empty chamber, suggesting that CD1 mice are sociable like C57 mice. However, the observed power suggests that it is easier to achieve statistical significance with C57 than CD1 mice. Because the stranger mouse could be considered as a novel object, we assessed for a novelty effect by adding an object. CD1 mice spend more time in the chamber with a stranger mouse than that a novel object, suggesting that their preference is social in nature. Thus, outbred CD1 mice are as appropriate as inbred C57 mice for studying social behavior using either the single or the three-chamber test using a specific acclimation paradigm.
- Published
- 2017
- Full Text
- View/download PDF
24. Ectopic HCN4 expression drives mTOR-dependent epilepsy
- Author
-
Lawrence S. Hsieh, Dennis D. Spencer, Lena H. Nguyen, John H. Wen, Longbo Zhang, Angélique Bordey, and Juan Torres-Reveron
- Subjects
Gene isoform ,0303 health sciences ,Gating ,Cortical dysplasia ,Biology ,medicine.disease ,Potassium channel ,3. Good health ,03 medical and health sciences ,Tuberous sclerosis ,Epilepsy ,0302 clinical medicine ,medicine ,Neuroscience ,030217 neurology & neurosurgery ,PI3K/AKT/mTOR pathway ,Intracellular ,030304 developmental biology - Abstract
The causative link between focal cortical malformations (FCM) and epilepsy is well-accepted, especially among patients with focal cortical dysplasia type II (FCDII) and tuberous sclerosis complex (TSC). However, the mechanisms underlying seizures remain unclear. Using a mouse model of TSC- and FCDII-associated FCM, we show that FCM neurons are responsible for seizure activity via their unexpected abnormal expression of the hyperpolarization-activated cyclic nucleotide-gated potassium channel isoform 4 (HCN4), which is normally not present in cortical pyramidal neurons after birth. Increasing intracellular cAMP levels, which preferentially affects HCN4 gating relative to the other isoforms, drove repetitive firing of FCM neurons but not that of control pyramidal neurons. Ectopic HCN4 expression was mTOR-dependent, preceded the onset of seizures, and was also found in diseased neurons in tissue resected for epilepsy treatment from TSC and FCDII patients. Finally, blocking HCN4 channel activity in FCM neurons prevented epilepsy in mice. These findings that demonstrate HCN4 acquisition as seizure-genic, identify a novel cAMP-dependent seizure mechanism in TSC and FCDII. Furthermore, the unique expression of HCN4 exclusively in FCM neurons provides opportunities for using HCN4 as a gene therapy target to treat epilepsy in individuals with FCDII or TSC.One Sentence SummaryOur data provide a novel cAMP-dependent mechanism of seizure initiation in focal cortical dysplasia and tuberous sclerosis complex due to the unexpected ectopic expression of HCN4 channels only in diseased neurons. HCN4 channels are thus promising candidates for gene therapy to treat epilepsy generated by mTOR-driven focal malformations.
- Published
- 2019
- Full Text
- View/download PDF
25. Preimplantation factor modulates oligodendrocytes by H19-induced demethylation of NCOR2
- Author
-
Sara Ornaghi, Martin Mueller, Fahmeed Hyder, Andreina Schoeberlein, Eytan R. Barnea, Keller Irene, Daniel Surbek, Daniel Coman, Longbo Zhang, Valérie Haesler, Michael J. Paidas, Angélique Bordey, Marialuigia Spinelli, Céline Boucard, Spinelli, M, Boucard, C, Ornaghi, S, Schoeberlein, A, Irene, K, Coman, D, Hyder, F, Zhang, L, Haesler, V, Bordey, A, Barnea, E, Paidas, M, Surbek, D, and Mueller, M
- Subjects
Neurodevelopment ,610 Medicine & health ,Therapeutics ,Biology ,Myelin ,Mice ,Pregnancy ,medicine ,Animals ,Humans ,Nuclear Receptor Co-Repressor 2 ,Gliogenesis ,Oligodendrocyte differentiation ,RNA ,General Medicine ,Methylation ,Cell biology ,Oligodendroglia ,DNA demethylation ,medicine.anatomical_structure ,Nuclear receptor ,embryonic structures ,Female ,RNA, Long Noncoding ,Drug therapy ,Peptides ,Corepressor ,Research Article ,Neuroscience - Abstract
Failed or altered gliogenesis is a major characteristic of diffuse white matter injury in survivors of premature birth. The developmentally regulated long noncoding RNA (lncRNA) H19 inhibits S-adenosylhomocysteine hydrolase (SAHH) and contributes to methylation of diverse cellular components, such as DNA, RNA, proteins, lipids, and neurotransmitters. We showed that the pregnancy-derived synthetic PreImplantation Factor (sPIF) induces expression of the nuclear receptor corepressor 2 (NCOR2) via H19/SAHH-mediated DNA demethylation. In turn, NCOR2 affects oligodendrocyte differentiation markers. Accordingly, after hypoxic-ischemic brain injury in rodents, myelin protection and oligodendrocytes' fate are in part modulated by sPIF and H19. Our results revealed an unexpected mechanism of the H19/SAHH axis underlying myelin preservation during brain recovery and its use in treating neurodegenerative diseases can be envisioned.
- Published
- 2019
- Full Text
- View/download PDF
26. Filamin A inhibition reduces seizure activity in a mouse model of focal cortical malformations
- Author
-
Tianxiang Huang, Shannon Teaw, Xuan Gong, Lawrence S. Hsieh, Lena H. Nguyen, Lindsay H. Burns, Longbo Zhang, and Angélique Bordey
- Subjects
Gene knockdown ,Epilepsy ,biology ,business.industry ,Filamins ,General Medicine ,Cortical dysplasia ,medicine.disease ,Filamin ,Mice ,Phosphatidylinositol 3-Kinases ,Seizures ,Malformations of Cortical Development, Group I ,medicine ,biology.protein ,Cancer research ,FLNA ,Animals ,Humans ,business ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,RHEB - Abstract
Epilepsy treatments for patients with mechanistic target of rapamycin (mTOR) disorders, such as tuberous sclerosis complex (TSC) or focal cortical dysplasia type II (FCDII), are urgently needed. In these patients, the presence of focal cortical malformations is associated with the occurrence of lifelong epilepsy, leading to severe neurological comorbidities. Here, we show that the expression of the actin cross-linking protein filamin A (FLNA) is increased in resected cortical tissue that is responsible for seizures in patients with FCDII and in mice modeling TSC and FCDII with mutations in phosphoinositide 3-kinase (PI3K)-ras homolog enriched in brain (Rheb) pathway genes. Normalizing FLNA expression in these mice through genetic knockdown limited cell misplacement and neuronal dysmorphogenesis, two hallmarks of focal cortical malformations. In addition, Flna knockdown reduced seizure frequency independently of mTOR signaling. Treating mice with a small molecule targeting FLNA, PTI-125, before the onset of seizures alleviated neuronal abnormalities and reduced seizure frequency compared to vehicle-treated mice. In addition, the treatment was also effective when injected after seizure onset in juvenile and adult mice. These data suggest that targeting FLNA with either short hairpin RNAs or the small molecule PTI-125 might be effective in reducing seizures in patients with TSC and FCDII bearing mutations in PI3K-Rheb pathway genes.
- Published
- 2019
27. The mTOR pathway genes MTOR, Rheb, Depdc5, Pten, and Tsc1 have convergent and divergent impacts on cortical neuron development and function
- Author
-
Lena H Nguyen, Youfen Xu, Maanasi Nair, and Angelique Bordey
- Subjects
malformation of cortical development ,epilepsy ,seizures ,HCN channels ,Ih current ,action potential ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Brain somatic mutations in various components of the mTOR complex 1 (mTORC1) pathway have emerged as major causes of focal malformations of cortical development and intractable epilepsy. While these distinct gene mutations converge on excessive mTORC1 signaling and lead to common clinical manifestations, it remains unclear whether they cause similar cellular and synaptic disruptions underlying cortical network hyperexcitability. Here, we show that in utero activation of the mTORC1 activator genes, Rheb or MTOR, or biallelic inactivation of the mTORC1 repressor genes, Depdc5, Tsc1, or Pten in the mouse medial prefrontal cortex leads to shared alterations in pyramidal neuron morphology, positioning, and membrane excitability but different changes in excitatory synaptic transmission. Our findings suggest that, despite converging on mTORC1 signaling, mutations in different mTORC1 pathway genes differentially impact cortical excitatory synaptic activity, which may confer gene-specific mechanisms of hyperexcitability and responses to therapeutic intervention.
- Published
- 2024
- Full Text
- View/download PDF
28. An epigenetic mechanism mediates developmental nicotine effects on neuronal structure and behavior
- Author
-
Daniel Coman, Qiaoping Yuan, Angela M. Lee, Yonwoo Jung, Fahmeed Hyder, Marina R. Picciotto, Zhifeng Zhou, Angélique Bordey, David Goldman, Christopher J. Heath, Lawrence S. Hsieh, and Yann S. Mineur
- Subjects
0301 basic medicine ,Nicotine ,Behavioral epigenetics ,Biology ,Methylation ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,medicine ,Animals ,Histone methyltransferase complex ,Epigenetics ,Promoter Regions, Genetic ,Gene knockdown ,General Neuroscience ,Nuclear Proteins ,Histone-Lysine N-Methyltransferase ,DNA-Binding Proteins ,Mice, Inbred C57BL ,030104 developmental biology ,Nicotinic agonist ,H3K4me3 ,sense organs ,Neuroscience ,030217 neurology & neurosurgery ,Transcription Factors ,medicine.drug - Abstract
Developmental nicotine exposure causes persistent changes in cortical neuron morphology and in behavior. We used microarray screening to identify master transcriptional or epigenetic regulators mediating these effects of nicotine and discovered increases in Ash2l mRNA, encoding a component of a histone methyltransferase complex. We therefore examined genome-wide changes in trimethylation of histone H3 on Lys4 (H3K4me3), a mark induced by the Ash2l complex associated with increased gene transcription. A large proportion of regulated promoter sites were involved in synapse maintenance. We found that Mef2c interacts with Ash2l and mediates changes in H3K4me3. Knockdown of Ash2l or Mef2c abolished nicotine-mediated alterations of dendritic complexity in vitro and in vivo, and attenuated nicotine-dependent changes in passive avoidance behavior. In contrast, overexpression mimicked nicotine-mediated alterations of neuronal structure and passive avoidance behavior. These studies identify Ash2l as a target induced by nicotinic stimulation that couples developmental nicotine exposure to changes in brain epigenetic marks, neuronal structure and behavior.
- Published
- 2016
- Full Text
- View/download PDF
29. Hypoxia-inducible factor-1a contributes to dendritic overgrowth in tuberous sclerosis
- Author
-
Shiliang Zhang, Longbo Zhang, David M. Feliciano, Angélique Bordey, and Tianxiang Huang
- Subjects
Male ,0301 basic medicine ,Genetically modified mouse ,congenital, hereditary, and neonatal diseases and abnormalities ,Mice, Transgenic ,mTORC1 ,Biology ,Tuberous Sclerosis Complex 1 Protein ,Article ,03 medical and health sciences ,Tuberous sclerosis ,0302 clinical medicine ,Tuberous Sclerosis ,medicine ,Animals ,PI3K/AKT/mTOR pathway ,Tumor Suppressor Proteins ,General Neuroscience ,Dendrites ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Olfactory bulb ,Cell biology ,030104 developmental biology ,HIF1A ,medicine.anatomical_structure ,Animals, Newborn ,nervous system ,Hypoxia-inducible factors ,Gene Knockdown Techniques ,Female ,TSC1 ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Expression of hypoxia-inducible factor 1a (HIF1a) is increased under several pathological conditions such as hyperactive mechanistic target of rapamycin complex 1 (mTORC1) in tuberous sclerosis complex (TSC). Hyperactive mTORC1 and the resulting increased dendritic complexity of neurons are shared molecular and cellular alterations in several neurological disorders associated with cognitive disabilities. Despite some evidence that HIF1a contributes to dendritic overgrowth in vitro, it remains unknown whether increased HIF1a in TSC neurons could contribute to their increased dendritic complexity. To address this use in vivo, we generated TSC neurons by deleting Tsc1 in newborn olfactory bulb (OB) neurons of conditional Tsc1 transgenic mice using neonatal electroporation. In addition to their increased dendritic complexity, Tsc1(null) neurons have been reported to display increased Hif1a mRNA level and HIF1a transcriptional activity. We found that Tsc1(null)-dependent dendritic overgrowth was prevented by knocking down HIF1a or expressing a dominant negative HIF1a. In addition, overexpressing HIF1a in wild-type developing neurons resulted in increased dendritic complexity in vivo. These data highlight that an increase in HIF1a levels contributes to abnormal dendritic patterning in developing neurons under normal conditions and hyperactive mTORC1 conditions as in TSC.
- Published
- 2016
- Full Text
- View/download PDF
30. Hypervascularization in mTOR-dependent focal and global cortical malformations displays differential rapamycin sensitivity
- Author
-
Angélique Bordey, Tianxiang Huang, Shannon Teaw, and Longbo Zhang
- Subjects
0301 basic medicine ,Genetically modified mouse ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Mice, Transgenic ,Article ,03 medical and health sciences ,Tuberous sclerosis ,Epilepsy ,Mice ,0302 clinical medicine ,Pregnancy ,Seizures ,Tuberous Sclerosis ,Medicine ,Animals ,Microvessel ,PI3K/AKT/mTOR pathway ,Cell Size ,Neurons ,Sirolimus ,Neovascularization, Pathologic ,business.industry ,TOR Serine-Threonine Kinases ,Dendrites ,Somatosensory Cortex ,medicine.disease ,Malformations of Cortical Development ,Corticogenesis ,030104 developmental biology ,Electroporation ,Neurology ,Forebrain ,Blood Vessels ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Plasmids - Abstract
Objectives Patients with mammalian target of rapamycin (mTOR)-dependent malformations of cortical development (MCDs) associated with seizures display hyperperfusion and increased vessel density of the dysmorphic cortical tissue. Some studies have suggested that the vascular defect occurred independently of seizures. Here, we further examined whether hypervascularization occurs in animal models of global and focal MCD with and without seizures, and whether it is sensitive to the mTOR blocker, rapamycin, that is approved for epilepsy treatment in tuberous sclerosis complex. Methods We used two experimental models of mTOR-dependent MCD consisting of conditional transgenic mice containing Tsc1null cells in the forebrain generating a global malformation associated with seizures and of wild-type mice containing a focal malformation in the somatosensory cortex generated by in utero electroporation (IUE) that does not lead to seizures. Alterations in blood vessels and the effects of a 2-week-long rapamycin treatment on these phenotypes were assessed in juvenile mice. Results Blood vessels in both the focal and global MCDs of postnatal day 14 mice displayed significant increase in vessel density, branching index, total vessel length, and decreased tissue lacunarity. In addition, rapamycin treatment (0.5 mg/kg, every 2 days) partially rescued vessel abnormalities in the focal MCD model, but it did not ameliorate the vessel abnormalities in the global MCD model that required higher rapamycin dosage for a partial rescue. Significance Here, we identified hypervascularization in mTOR-dependent MCD in the absence of seizures in young mice, suggesting that increased angiogenesis occurs during development in parallel to alterations in corticogenesis. In addition, a predictive functional outcome is that dysplastic neurons forming MCD will have better access to oxygen and metabolic supplies via their closer proximity to blood vessels. Finally, the difference in rapamycin sensitivity between a focal and global MCD suggest that rapamycin treatment will need to be titrated to match the type of MCD.
- Published
- 2018
31. Activating the translational repressor 4E-BP or reducing S6K-GSK3β activity prevents accelerated axon growth induced by hyperactive mTORin vivo
- Author
-
John H. Wen, Angélique Bordey, Tiffany V. Lin, Longbo Zhang, Xuan Gong, Lawrence S. Hsieh, Laura Miyares, and Tianxiang Huang
- Subjects
Male ,Regulator ,Cell Cycle Proteins ,P70-S6 Kinase 1 ,Cell Growth Processes ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Ribosomal Protein S6 Kinases, 90-kDa ,Glycogen Synthase Kinase 3 ,Mice ,Eukaryotic initiation factor ,Genetics ,medicine ,Animals ,Eukaryotic Initiation Factors ,Axon ,Molecular Biology ,Genetics (clinical) ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Glycogen Synthase Kinase 3 beta ,TOR Serine-Threonine Kinases ,Translation (biology) ,Articles ,General Medicine ,Phosphoproteins ,Axons ,Cell biology ,Corticogenesis ,medicine.anatomical_structure ,Gene Expression Regulation ,nervous system ,Multiprotein Complexes ,Female ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins ,Signal Transduction - Abstract
Abnormal axonal connectivity and hyperactive mTOR complex 1 (mTORC1) are shared features of several neurological disorders. Hyperactive mTORC1 alters axon length and polarity of hippocampal neurons in vitro, but the impact of hyperactive mTORC1 on axon growth in vivo and the mechanisms underlying those effects remain unclear. Using in utero electroporation during corticogenesis, we show that increasing mTORC1 activity accelerates axon growth without multiple axon formation. This was prevented by counteracting mTORC1 signaling through p70S6Ks (S6K1/2) or eukaryotic initiation factor 4E-binding protein (4E-BP1/2), which both regulate translation. In addition to regulating translational targets, S6K1 indirectly signals through GSK3β, a regulator of axogenesis. Although blocking GSK3β activity did not alter axon growth under physiological conditions in vivo, blocking it using a dominant-negative mutant or lithium chloride prevented mTORC1-induced accelerated axon growth. These data reveal the contribution of translational and non-translational downstream effectors such as GSK3β to abnormal axon growth in neurodevelopmental mTORopathies and open new therapeutic options for restoring long-range connectivity.
- Published
- 2015
- Full Text
- View/download PDF
32. Voltage-dependent K+currents contribute to heterogeneity of olfactory ensheathing cells
- Author
-
Angélique Bordey, Charles A. Greer, Lorena Rela, and Ana Paula Piantanida
- Subjects
Inward-rectifier potassium ion channel ,Gap junction ,Biology ,Sensory neuron ,Potassium channel ,Cell membrane ,Cellular and Molecular Neuroscience ,medicine.anatomical_structure ,Neurology ,Olfactory nerve ,medicine ,Olfactory ensheathing glia ,Patch clamp ,Neuroscience - Abstract
The olfactory nerve is permissive for axon growth throughout life. This has been attributed in part to the olfactory ensheathing glial cells that encompass the olfactory sensory neuron fascicles. Olfactory ensheathing cells (OECs) also promote axon growth in vitro and when transplanted in vivo to sites of injury. The mechanisms involved remain largely unidentified owing in part to the limited knowledge of the physiological properties of ensheathing cells. Glial cells rely for many functions on the properties of the potassium channels expressed; however, those expressed in ensheathing cells are unknown. Here we show that OECs express voltage-dependent potassium currents compatible with inward rectifier (Kir ) and delayed rectifier (KDR ) channels. Together with gap junction coupling, these contribute to the heterogeneity of membrane properties observed in OECs. The relevance of K(+) currents expressed by ensheathing cells is discussed in relation to plasticity of the olfactory nerve.
- Published
- 2015
- Full Text
- View/download PDF
33. Valnoctamide inhibits cytomegalovirus infection in developing brain and attenuates neurobehavioral dysfunctions and brain abnormalities
- Author
-
Michael J. Paidas, Lawrence S. Hsieh, Anthony N. van den Pol, Angélique Bordey, Patrizia Vergani, Sara Ornaghi, Ornaghi, S, Hsieh, L, Bordey, A, Vergani, P, Paidas, M, and van den Pol, A
- Subjects
Male ,0301 basic medicine ,endocrine system ,Congenital cytomegalovirus infection ,Biology ,Antiviral Agents ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Valnoctamide ,Encephalitis, Viral ,Adverse effect ,Research Articles ,Fetus ,Dose-Response Relationship, Drug ,General Neuroscience ,Cytomegalovirus, Brain Development, Neurobehavioral Dysfunctions, Brain Abnormalities ,medicine.disease ,Amides ,Mice, Inbred C57BL ,Treatment Outcome ,030104 developmental biology ,Animals, Newborn ,Viral replication ,Cytomegalovirus Infections ,Immunology ,Toxicity ,Brain size ,Female ,Cerebellar hypoplasia (non-human) ,Cognition Disorders ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Cytomegalovirus (CMV) is the most common infectious cause of brain defects and neurological dysfunction in developing human babies. Due to the teratogenicity and toxicity of available CMV antiviral agents, treatment options during early development are markedly limited. Valnoctamide (VCD), a neuroactive mood stabilizer with no known teratogenic activity, was recently demonstrated to have anti-CMV potential. However, it is not known whether this can be translated into an efficacious therapeutic effect to improve CMV-induced adverse neurological outcomes. Using multiple models of CMV infection in the developing mouse brain, we show that subcutaneous low-dose VCD suppresses CMV by reducing the level of virus available for entry into the brain and by acting directly within the brain to block virus replication and dispersal. VCD during the first 3 weeks of life restored timely acquisition of neurological milestones in neonatal male and female mice and rescued long-term motor and behavioral outcomes in juvenile male mice. CMV-mediated brain defects, including decreased brain size, cerebellar hypoplasia, and neuronal loss, were substantially attenuated by VCD. No adverse side effects on neurodevelopment of uninfected control mice receiving VCD were detected. Treatment of CMV-infected human fetal astrocytes with VCD reduced both viral infectivity and replication by blocking viral particle attachment to the cell, a mechanism that differs from available anti-CMV drugs. These data suggest that VCD during critical periods of neurodevelopment can effectively suppress CMV replication in the brain and safely improve both immediate and long-term neurological outcomes.SIGNIFICANCE STATEMENT Cytomegalovirus (CMV) can irreversibly damage the developing brain. No anti-CMV drugs are available for use during fetal development, and treatment during the neonatal period has substantial limitations. We studied the anti-CMV actions of valnoctamide (VCD), a psychiatric sedative that appears to lack teratogenicity and toxicity, in the newborn mouse brain, a developmental period that parallels that of an early second-trimester human fetus. In infected mice, subcutaneous VCD reaches the brain and suppresses viral replication within the CNS, rescuing the animals from CMV-induced brain defects and neurological problems. Treatment of uninfected control animals exerts no detectable adverse effects. VCD also blocks CMV replication in human fetal brain cells.
- Published
- 2017
34. mTORC1 Targets the Translational Repressor 4E-BP2, but Not S6 Kinase 1/2, to Regulate Neural Stem Cell Self-Renewal In Vivo
- Author
-
David M. Feliciano, Longbo Zhang, Angélique Bordey, Nathaniel W. Hartman, Tiffany V. Lin, and Grace E. Paquelet
- Subjects
Cell Cycle Proteins ,P70-S6 Kinase 1 ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Ribosomal Protein S6 Kinases, 90-kDa ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Neural Stem Cells ,medicine ,Animals ,Eukaryotic Initiation Factors ,Phosphorylation ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Cells, Cultured ,reproductive and urinary physiology ,Adaptor Proteins, Signal Transducing ,Monomeric GTP-Binding Proteins ,Sirolimus ,Gene knockdown ,Kinase ,Cell growth ,TOR Serine-Threonine Kinases ,Neuropeptides ,Neurogenesis ,Cell Differentiation ,Phosphoproteins ,Neural stem cell ,nervous system diseases ,Cell biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,nervous system ,Biochemistry ,Multiprotein Complexes ,RNA Interference ,Ras Homolog Enriched in Brain Protein ,Neuron ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins - Abstract
SummaryThe mammalian target of rapamycin complex 1 (mTORC1) integrates signals important for cell growth, and its dysregulation in neural stem cells (NSCs) is implicated in several neurological disorders associated with abnormal neurogenesis and brain size. However, the function of mTORC1 on NSC self-renewal and the downstream regulatory mechanisms are ill defined. Here, we found that genetically decreasing mTORC1 activity in neonatal NSCs prevented their differentiation, resulting in reduced lineage expansion and aborted neuron production. Constitutive activation of the translational repressor 4E-BP1, which blocked cap-dependent translation, had similar effects and prevented hyperactive mTORC1 induction of NSC differentiation and promoted self-renewal. Although 4E-BP2 knockdown promoted NSC differentiation, p70 S6 kinase 1 and 2 (S6K1/S6K2) knockdown did not affect NSC differentiation but reduced NSC soma size and prevented hyperactive mTORC1-induced increase in soma size. These data demonstrate a crucial role of mTORC1 and 4E-BP for switching on and off cap-dependent translation in NSC differentiation.
- Published
- 2013
- Full Text
- View/download PDF
35. Rheb Activation in Subventricular Zone Progenitors Leads to Heterotopia, Ectopic Neuronal Differentiation, and Rapamycin-Sensitive Olfactory Micronodules and Dendrite Hypertrophy of Newborn Neurons
- Author
-
David M. Feliciano, Lawrence S. Hsieh, Carlos Lafourcade, Angélique Bordey, Tiffany V. Lin, and Longbo Zhang
- Subjects
Male ,Rostral migratory stream ,Neurogenesis ,Subventricular zone ,Cell Enlargement ,Article ,Cerebral Ventricles ,Mice ,Neural Stem Cells ,Neuroblast ,Cell Movement ,Cell Line, Tumor ,Neuroblast migration ,medicine ,Animals ,Monomeric GTP-Binding Proteins ,Neurons ,Sirolimus ,biology ,Stem Cells ,TOR Serine-Threonine Kinases ,General Neuroscience ,Neuropeptides ,Cell Differentiation ,Dendrites ,Hypertrophy ,Olfactory Bulb ,Neural stem cell ,Electroporation ,medicine.anatomical_structure ,Animals, Newborn ,biology.protein ,Female ,Ras Homolog Enriched in Brain Protein ,TSC1 ,Neuroscience ,RHEB - Abstract
Mammalian target of rapamycin (mTOR) hyperactivity in perinatal neural progenitor cells (NPCs) of tuberous sclerosis complex 1 (Tsc1) heterozygote mice leads to heterotopia and abnormal neuronal morphogenesis as seen in patients with tuberous sclerosis. Considering that pathological hyperactive mTOR also occurs in individuals carrying no genetic mutations, we examined whether increasing mTOR activity in neonatal NPCs of wild-type mice would recapitulate the above phenotypes. Electroporation of a plasmid encoding constitutively active Ras-homolog enriched in brain (RhebCA) into subventricular zone NPCs increased mTOR activity in newborn cells. At 19 d post-electroporation (dpe), heterotopia and ectopic cells with a neuronal morphology were observed along the migratory path [rostral migratory stream (RMS)] and in the olfactory bulb (OB). These ectopic cells displayed action potentials and received synaptic inputs identifying them as synaptically integrated neurons. RMS heterotopias contained astrocytes, neurons, and entrapped neuroblasts. Immunostaining at 3 dpe revealed the presence of Mash1+Olig2−cells in the migratory route accompanied by ectopic neuronal differentiation and altered direction and speed of neuroblast migration at 7 dpe, suggesting a non-cell-autonomous disruption of migration. At >19 dpe, newborn RhebCA-expressing neurons displayed altered distribution and formed micronodules in the OB. In addition, they displayed increased dendritic complexity along with altered membrane biophysics and increased frequency of GABAergic synaptic inputs. OB heterotopia, micronodules, and dendrite hypertrophy were notably prevented by rapamycin treatment, suggesting their mTOR dependence. Collectively, these data show that increasing mTOR activity in neonatal NPCs of wild-type mice recapitulate the pathologies observed inTsc1mutant mice. In addition, increased mTOR activity in individuals without known mutations could significantly impact neurogenesis and circuit formation.
- Published
- 2013
- Full Text
- View/download PDF
36. 4E-BP1 expression in embryonic postmitotic neurons mitigates mTORC1-induced cortical malformations and behavioral seizure severity but does not prevent epilepsy in mice
- Author
-
Lena H. Nguyen, Manas Sharma, and Angelique Bordey
- Subjects
mTOR ,4E-BP ,seizures ,epilepsy ,corticogenesis ,cortical development ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1) pathway during neurodevelopment leads to focal cortical malformations associated with intractable seizures. Recent evidence suggests that dysregulated cap-dependent translation downstream of mTORC1 contributes to cytoarchitectural abnormalities and seizure activity. Here, we examined whether reducing cap-dependent translation by expressing a constitutively active form of the translational repressor, 4E-BP1, downstream of mTORC1 would prevent the development of cortical malformations and seizures. 4E-BP1CA was expressed embryonically either in radial glia (neural progenitor cells) that generate cortical layer 2/3 pyramidal neurons or in migrating neurons destined to layer 2/3 using a conditional expression system. In both conditions, 4E-BP1CA expression reduced mTORC1-induced neuronal hypertrophy and alleviated cortical mislamination, but a subset of ectopic neurons persisted in the deep layers and the white matter. Despite the above improvements, 4E-BP1CA expression in radial glia had no effects on seizure frequency and further exacerbated behavioral seizure severity associated with mTORC1 hyperactivation. In contrast, conditional 4E-BP1CA expression in migratory neurons mitigated the severity of behavioral seizures but the seizure frequency remained unchanged. These findings advise against targeting 4E-BPs by 4E-BP1CA expression during embryonic development for seizure prevention and suggest the presence of a development-dependent role for 4E-BPs in mTORC1-induced epilepsy.
- Published
- 2023
- Full Text
- View/download PDF
37. Normalizing translation through 4E-BP prevents mTOR-driven cortical mislamination and ameliorates aberrant neuron integration
- Author
-
Tiffany V. Lin, Taylor J. Malone, Angélique Bordey, Lawrence S. Hsieh, and Tomoki Kimura
- Subjects
0301 basic medicine ,RNA Caps ,Dendritic Spines ,Green Fluorescent Proteins ,Cell Cycle Proteins ,mTORC1 ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Eukaryotic initiation factor ,medicine ,Animals ,Eukaryotic Initiation Factors ,RNA, Small Interfering ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Neurons ,Gene knockdown ,Multidisciplinary ,TOR Serine-Threonine Kinases ,Excitatory Postsynaptic Potentials ,Translation (biology) ,Matrix Attachment Region Binding Proteins ,Biological Sciences ,Phosphoproteins ,Corticogenesis ,030104 developmental biology ,medicine.anatomical_structure ,Cytoarchitecture ,Gene Knockdown Techniques ,Protein Biosynthesis ,Ras Homolog Enriched in Brain Protein ,Neuron ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins ,Neuroscience ,Neuroglia ,030217 neurology & neurosurgery ,Signal Transduction ,Transcription Factors - Abstract
Hyperactive mammalian target of rapamycin complex 1 (mTORC1) is a shared molecular hallmark in several neurodevelopmental disorders characterized by abnormal brain cytoarchitecture. The mechanisms downstream of mTORC1 that are responsible for these defects remain unclear. We show that focally increasing mTORC1 activity during late corticogenesis leads to ectopic placement of upper-layer cortical neurons that does not require altered signaling in radial glia and is accompanied by changes in layer-specific molecular identity. Importantly, we found that decreasing cap-dependent translation by expressing a constitutively active mutant of the translational repressor eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) prevents neuronal misplacement and soma enlargement, while partially rescuing dendritic hypertrophy induced by hyperactive mTORC1. Furthermore, overactivation of translation alone through knockdown of 4E-BP2 was sufficient to induce neuronal misplacement. These data show that many aspects of abnormal brain cytoarchitecture can be prevented by manipulating a single intracellular process downstream of mTORC1, cap-dependent translation.
- Published
- 2016
38. miR-132 enhances dendritic morphogenesis, spine density, synaptic integration, and survival of newborn olfactory bulb neurons
- Author
-
Lily D. Yan, Zhao-Qian Teng, Tiffany V. Lin, Valerie Gordon, Tomoki Kimura, Manavendra Pathania, David Van Vactor, Angélique Bordey, Tudor A. Fulga, Juan Torres-Reveron, and Xinyu Zhao
- Subjects
Dendritic spine ,Mouse ,lcsh:Medicine ,Cerebral Ventricles ,miR-132 ,Mice ,0302 clinical medicine ,RNA interference ,Molecular Cell Biology ,Cyclic AMP Response Element-Binding Protein ,lcsh:Science ,Neurons ,0303 health sciences ,Multidisciplinary ,Neuronal Plasticity ,Neurogenesis ,Gene Expression Regulation, Developmental ,Animal Models ,Olfactory Bulb ,Cell biology ,medicine.anatomical_structure ,Gene Knockdown Techniques ,Cellular Types ,Research Article ,Cell Survival ,Dendritic Spines ,Subventricular zone ,Biology ,03 medical and health sciences ,Glutamatergic ,Model Organisms ,Developmental Neuroscience ,Cellular neuroscience ,Neuroplasticity ,medicine ,Genetics ,Animals ,030304 developmental biology ,lcsh:R ,Olfactory bulb ,MicroRNAs ,Animals, Newborn ,nervous system ,Cellular Neuroscience ,Synapses ,lcsh:Q ,Gene expression ,030217 neurology & neurosurgery ,Developmental Biology ,Neuroscience - Abstract
An array of signals regulating the early stages of postnatal subventricular zone (SVZ) neurogenesis has been identified, but much less is known regarding the molecules controlling late stages. Here, we investigated the function of the activity-dependent and morphogenic microRNA miR-132 on the synaptic integration and survival of olfactory bulb (OB) neurons born in the neonatal SVZ. In situ hybridization revealed that miR-132 expression occurs at the onset of synaptic integration in the OB. Using in vivo electroporation we found that sequestration of miR-132 using a sponge-based strategy led to a reduced dendritic complexity and spine density while overexpression had the opposite effects. These effects were mirrored with respective changes in the frequency of GABAergic and glutamatergic synaptic inputs reflecting altered synaptic integration. In addition, timely directed overexpression of miR-132 at the onset of synaptic integration using an inducible approach led to a significant increase in the survival of newborn neurons. These data suggest that miR-132 forms the basis of a structural plasticity program seen in SVZ-OB postnatal neurogenesis. miR-132 overexpression in transplanted neurons may thus hold promise for enhancing neuronal survival and improving the outcome of transplant therapies.
- Published
- 2016
- Full Text
- View/download PDF
39. Tsc1 haploinsufficiency is sufficient to increase dendritic patterning and Filamin A levels
- Author
-
Tianxiang Huang, Longbo Zhang, and Angélique Bordey
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Filamins ,Mice, Transgenic ,Haploinsufficiency ,Biology ,Filamin ,Article ,Tuberous Sclerosis Complex 1 Protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Tuberous Sclerosis ,medicine ,FLNA ,Animals ,General Neuroscience ,Tumor Suppressor Proteins ,Neurogenesis ,Wild type ,Heterozygote advantage ,Dendrites ,Olfactory Bulb ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,TSC1 ,TSC2 ,030217 neurology & neurosurgery - Abstract
Most individuals with tuberous sclerosis complex (TSC) are born with a mutant allele of either TSC1 or TSC2 and a mosaic of psychological and cognitive defects. Tsc1 loss of heterozygosity contributes to severe dendritic abnormalities that are rescued by normalizing the levels of the actin-cross linking protein, Filamin A (FLNA). However, it is unclear whether dendrites and FLNA levels are abnormal in an heterozygote Tsc1 condition. Here, we examined dendritic morphology and FLNA levels in the olfactory bulb of Tsc1 wild type and heterozygote mice. Using in vivo neonatal electroporation to label newborn neurons followed by sholl analysis, we found that Tsc1 haploinsufficiency is associated with increased dendritic complexity and total dendritic length as well as increased FLNA levels. Since reducing FLNA levels has been shown to decrease Tsc1(+/-) dendritic complexity, these data suggest that increased FLNA levels in Tsc1(+/-) mice contribute to abnormal dendritic patterning in the Tsc1 heterozygote condition of individuals with TSC.
- Published
- 2016
40. Convulsive seizures from experimental focal cortical dysplasia occur independently of cell misplacement
- Author
-
Gordon F. Buchanan, Felicia A. Harrsch, Yuegao Huang, Lawrence S. Hsieh, Kumiko Claycomb, Fahmeed Hyder, John H. Wen, Angélique Bordey, and Janice R. Naegele
- Subjects
Male ,0301 basic medicine ,Science ,Green Fluorescent Proteins ,Prefrontal Cortex ,General Physics and Astronomy ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,White matter ,Mice ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Cell Movement ,Genes, Reporter ,Seizures ,medicine ,Animals ,Humans ,Cognitive Dysfunction ,Prefrontal cortex ,PI3K/AKT/mTOR pathway ,Neurons ,Sirolimus ,Multidisciplinary ,TOR Serine-Threonine Kinases ,General Chemistry ,Cortical dysplasia ,medicine.disease ,White Matter ,3. Good health ,Malformations of Cortical Development ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Heterotopia (medicine) ,Gene Expression Regulation ,Female ,Neuroscience ,Neurocognitive ,030217 neurology & neurosurgery ,Signal Transduction ,medicine.drug - Abstract
Focal cortical dysplasia (FCD), a local malformation of cortical development, is the most common cause of pharmacoresistant epilepsy associated with life-long neurocognitive impairments. It remains unclear whether neuronal misplacement is required for seizure activity. Here we show that dyslamination and white matter heterotopia are not necessary for seizure generation in a murine model of type II FCDs. These experimental FCDs generated by increasing mTOR activity in layer 2/3 neurons of the medial prefrontal cortex are associated with tonic-clonic seizures and a normal survival rate. Preventing all FCD-related defects, including neuronal misplacement and dysmorphogenesis, with rapamycin treatments from birth eliminates seizures, but seizures recur after rapamycin withdrawal. In addition, bypassing neuronal misplacement and heterotopia using inducible vectors do not prevent seizure occurrence. Collectively, data obtained using our new experimental FCD-associated epilepsy suggest that life-long treatment to reduce neuronal dysmorphogenesis is required to suppress seizures in individuals with FCD., The etiology of focal cortical dysplasia (FCD) is not fully understood. Here authors generate an mTORC1 overactivation mouse model that recapitulates hallmarks of type II FCDs, including spontaneous seizures, and suggest that neuronal defects, rather than macrostructural changes, lead to seizures.
- Published
- 2016
- Full Text
- View/download PDF
41. Mammalian FMRP S499 Is Phosphorylated by CK2 and Promotes Secondary Phosphorylation of FMRP
- Author
-
Xuan Gong, Christopher M. Bartley, Mihaela-Rita Mihailescu, Rachel A. O’Keefe, Angélique Bordey, Laura Miyares, Anna C. Blice-Baum, and Esra Karaca
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Time Factors ,Blotting, Western ,fragile X ,translation ,Neuronal Excitability ,Biology ,Receptors, Metabotropic Glutamate ,Mass Spectrometry ,03 medical and health sciences ,Fragile X Mental Retardation Protein ,Mice ,0302 clinical medicine ,Animals ,Humans ,Naphthyridines ,Phosphorylation ,Casein Kinase II ,Protein Kinase Inhibitors ,Cells, Cultured ,030304 developmental biology ,Genetics ,Cerebral Cortex ,0303 health sciences ,Kinase ,General Neuroscience ,HEK 293 cells ,Translation (biology) ,General Medicine ,Protein phosphatase 2 ,New Research ,Recombinant Proteins ,nervous system diseases ,HEK293 Cells ,Metabotropic glutamate receptor ,Protein Biosynthesis ,mTOR ,Phenazines ,Casein kinase 1 ,Casein kinase 2 ,FMRP ,030217 neurology & neurosurgery ,casein kinase - Abstract
Visual Abstract, The fragile X mental retardation protein (FMRP) is an mRNA-binding regulator of protein translation that associates with 4-6% of brain transcripts and is central to neurodevelopment. Autism risk genes’ transcripts are overrepresented among FMRP-binding mRNAs, and FMRP loss-of-function mutations are responsible for fragile X syndrome, the most common cause of monogenetic autism. It is thought that FMRP-dependent translational repression is governed by the phosphorylation of serine residue 499 (S499). However, recent evidence suggests that S499 phosphorylation is not modulated by metabotropic glutamate receptor class I (mGluR-I) or protein phosphatase 2A (PP2A), two molecules shown to regulate FMRP translational repression. Moreover, the mammalian FMRP S499 kinase remains unknown. We found that casein kinase II (CK2) phosphorylates murine FMRP S499. Further, we show that phosphorylation of FMRP S499 permits phosphorylation of additional, nearby residues. Evidence suggests that these nearby residues are modulated by mGluR-I and PP2A pathways. These data support an alternative phosphodynamic model of FMRP that is harmonious with prior studies and serves as a framework for further investigation.
- Published
- 2016
42. Transient mGlu5R inhibition enhances the survival of granule cell precursors in the neonatal cerebellum
- Author
-
Amanda L. Hernandez, Cathryn Kubera, Vibol Heng, and Angélique Bordey
- Subjects
Male ,Cerebellum ,Cell Survival ,Neurogenesis ,Receptor, Metabotropic Glutamate 5 ,Apoptosis ,Biology ,Real-Time Polymerase Chain Reaction ,Receptors, Metabotropic Glutamate ,Article ,Mice ,chemistry.chemical_compound ,Neural Stem Cells ,In Situ Nick-End Labeling ,medicine ,Animals ,TUNEL assay ,Reverse Transcriptase Polymerase Chain Reaction ,Metabotropic glutamate receptor 5 ,General Neuroscience ,Glutamate receptor ,Granule cell ,Molecular biology ,medicine.anatomical_structure ,Animals, Newborn ,Terminal deoxynucleotidyl transferase ,chemistry ,Metabotropic glutamate receptor ,Female ,Bromodeoxyuridine - Abstract
The generation of the most abundant neurons of the cerebellum, the granule cells, relies on a balance between clonal expansion and apoptosis during the first 10 days after birth in the external germinal layer (EGL). The amino acid glutamate controls such critical phases of cell development in other systems through specific receptors such as metabotropic glutamate receptor 5 (mGlu(5)R). However, the function of mGlu(5)Rs on the proliferation and survival of granule cell precursors (GCPs) remains elusive. We found mGlu(5)R mRNA transcripts in EGL using RT-PCR and observed mGlu(5)R-mediated Ca(2+) responses in GCPs in acute slices as early as postnatal day (P) 2-3. Using in vivo injections of the selective non-competitive mGlu(5)R antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP) in P7-P9 mice, we found a 20% increase in the number of proliferative GCPs labeled at P7 with the S-phase marker bromodeoxyuridine (BrdU), but no increase in cell proliferation examined 2h following a BrdU injection. Furthermore, similar treatments led to a significant 70% decrease in the number of apoptotic GCPs in the EGL as determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. In contrast, in vivo treatment with the mGlu(5)R agonist (RS)-2-chloro-5-hydroxyphenylglycine (CHPG) resulted in a ∼60% increase in the number of TUNEL-labeled GCPs compared to control. These findings identify a unique role for glutamate acting at mGlu(5)Rs as a functional switch regulating GCP survival in the EGL, thus controlling the total number of cerebellar granule cells produced.
- Published
- 2012
- Full Text
- View/download PDF
43. Gap junction-mediated calcium waves define communication networks among murine postnatal neural progenitor cells
- Author
-
Benjamin Lacar, Stephanie Z. Young, Jean-Claude Platel, and Angélique Bordey
- Subjects
Cell type ,General Neuroscience ,Neurogenesis ,Gap junction ,Subventricular zone ,Connexin ,Biology ,Neural stem cell ,Cell biology ,B-1 cell ,Calcium imaging ,medicine.anatomical_structure ,medicine ,Neuroscience - Abstract
In the postnatal neurogenic niche, two populations of astrocyte-like cells (B cells) persist, one acting as neural progenitor cells (NPCs, B1 cells) and one forming a structural boundary between the neurogenic niche and the striatum (B2 cells, niche astrocytes). Despite being viewed as two distinct entities, we found that B1 and B2 cells express the gap junction protein connexin 43 and display functional coupling involving 50-60 cells. Using neonatal electroporation to label slowly cycling radial glia-derived B1 cells, which send a basal process onto blood vessels, we further confirmed dye coupling between NPCs. To assess the functionality of the coupling, we used calcium imaging in a preparation preserving the three-dimensional architecture of the subventricular zone. Intercellular calcium waves were observed among B cells. These waves travelled bidirectionally between B1 and B2 cells and propagated on blood vessels. Inter-B-cell calcium waves were absent in the presence of a gap junction blocker but persisted with purinergic receptor blockers. These findings show that privileged microdomains of communication networks exist among NPCs and niche astrocytes. Such functional coupling between these two cell types suggests that niche astrocytes do not merely have a structural role, but may play an active role in shaping the behavior of NPCs.
- Published
- 2011
- Full Text
- View/download PDF
44. Astroglial cells in the external granular layer are precursors of cerebellar granule neurons in neonates
- Author
-
M. Morgan Taylor, Tristan G. Heintz, John Silbereis, Yosif Ganat, Laura R. Ment, Angélique Bordey, and Flora M. Vaccarino
- Subjects
Cerebellum ,Time Factors ,Neurogenesis ,Green Fluorescent Proteins ,Mice, Transgenic ,Biology ,Article ,Mice ,Cellular and Molecular Neuroscience ,Genes, Reporter ,Glial Fibrillary Acidic Protein ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Cell Lineage ,Promoter Regions, Genetic ,Molecular Biology ,Rhombic lip ,Neurons ,Integrases ,Glial fibrillary acidic protein ,Stem Cells ,fungi ,Cell Biology ,beta-Galactosidase ,Granule cell ,Molecular biology ,Embryonic stem cell ,Neural stem cell ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Animals, Newborn ,Astrocytes ,biology.protein ,Stem cell - Abstract
It is well established that cerebellar granule cell precursors (GCPs) initially derive from progenitors in the rhombic lip of the embryonic cerebellar primordium. GCPs proliferate and migrate tangentially across the cerebellum to form the external granule cell layer (EGL) in late embryogenesis and early postnatal development. It is unclear whether GCPs are specified exclusively in the embryonic rhombic lip or whether their precursor persists in the neonate. Using transgenic mice expressing DsRed under the human glial fibrillary acidic protein (hGFAP) promoter, we found 2 populations of DsRed(+) cells in the EGL in the first postnatal week defined by bright and faint DsRed-fluorescent signal. Bright DsRed(+) cells have a protein expression profile and electrophysiological characteristics typical of astrocytes, but faint DsRed(+) cells in the EGL and internal granule cell layer (IGL) express markers and physiological properties of immature neurons. To determine if these astroglial cells gave rise to GCPs, we genetically tagged them with EGFP or betagal reporter genes at postnatal day (P)3-P5 using a hGFAP promoter driven inducible Cre recombinase. We found that GFAP promoter(+) cells in the EGL are proliferative and express glial and neural stem cell markers. In addition, immature granule cells (GCs) en route to the IGL at P12 as well as GCs in the mature cerebellum, 30days after recombination, express the reporter protein, suggesting that GFAP promoter(+) cells in the EGL generate a subset of granule cells. The identification of glial cells which function as neuronal progenitor cells profoundly impacts our understanding of cellular plasticity in the developing cerebellum.
- Published
- 2010
- Full Text
- View/download PDF
45. MicroRNA miR-137 Regulates Neuronal Maturation by Targeting Ubiquitin Ligase Mind Bomb-1
- Author
-
Junmin Peng, Keith E. Szulwach, Richard D. Smrt, Rebecca L. Pfeiffer, Angélique Bordey, Xuekun Li, Yuping Luo, Manavendra Pathania, Xinyu Zhao, Weixiang Guo, Zhao-Qian Teng, and Peng Jin
- Subjects
Ubiquitin-Protein Ligases ,Cellular differentiation ,Synaptogenesis ,Article ,Mice ,microRNA ,Animals ,Cells, Cultured ,Neurons ,Regulation of gene expression ,Messenger RNA ,Base Sequence ,biology ,Neurogenesis ,Cell Differentiation ,Dendrites ,Cell Biology ,Ubiquitin ligase ,Cell biology ,Mice, Inbred C57BL ,MicroRNAs ,Phenotype ,Gene Expression Regulation ,Protein Biosynthesis ,Synaptic plasticity ,biology.protein ,Molecular Medicine ,Developmental Biology - Abstract
The maturation of young neurons is regulated by complex mechanisms and dysregulation of this process is frequently found in neurodevepmental disorders. MicroRNAs have been implicated in several steps of neuronal maturation including dendritic and axonal growth, spine development, and synaptogenesis. We demonstrate that one brain-enriched microRNA, miR-137, has a significant role in regulating neuronal maturation. Overexpression of miR-137 inhibits dendritic morphogenesis, phenotypic maturation, and spine development both in brain and cultured primary neurons. On the other hand, a reduction in miR-137 had opposite effects. We further show that miR-137 targets the Mind bomb one (Mib1) protein through the conserved target site located in the 3′ untranslated region of Mib1 messenger RNA. Mib1 is an ubiquitin ligase known to be important for neurodevelopment. We show that exogenously expressed Mib1 could partially rescue the phenotypes associated with miR-137 overexpression. These results demonstrate a novel miRNA-mediated mechanism involving miR-137 and Mib1 that function to regulate neuronal maturation and dendritic morphogenesis during development.
- Published
- 2010
- Full Text
- View/download PDF
46. NMDA Receptors Activated by Subventricular Zone Astrocytic Glutamate Are Critical for Neuroblast Survival Prior to Entering a Synaptic Network
- Author
-
Angélique Bordey, Maria E. Rubio, Kathleen A. Dave, Valerie Gordon, Benjamin Lacar, and Jean-Claude Platel
- Subjects
animal structures ,Cell Survival ,Nerve net ,Neurogenesis ,Neuroscience(all) ,Glutamic Acid ,Subventricular zone ,Mice, Transgenic ,DEVBIO ,Biology ,Receptors, N-Methyl-D-Aspartate ,Article ,MOLNEURO ,Cerebral Ventricles ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neuroblast ,Cell Movement ,medicine ,Animals ,030304 developmental biology ,Neurons ,0303 health sciences ,musculoskeletal, neural, and ocular physiology ,General Neuroscience ,Glutamate receptor ,Cell Differentiation ,Olfactory Bulb ,Olfactory bulb ,medicine.anatomical_structure ,Animals, Newborn ,nervous system ,Astrocytes ,Synapses ,NMDA receptor ,Nerve Net ,Neuroscience ,Ganglion mother cell ,030217 neurology & neurosurgery - Abstract
Summary Even before integrating into existing circuitry, adult-born neurons express receptors for neurotransmitters, but the intercellular mechanisms and their impact on neurogenesis remain largely unexplored. Here, we show that neuroblasts born in the postnatal subventricular zone (SVZ) acquire NMDA receptors (NMDARs) during their migration to the olfactory bulb. Along their route, neuroblasts are ensheathed by astrocyte-like cells expressing vesicular glutamate release machinery. Increasing calcium in these specialized astrocytes induced NMDAR activity in neuroblasts, and blocking astrocytic vesicular release eliminated spontaneous NMDAR activity. Single-cell knockout of NMDARs using neonatal electroporation resulted in neuroblast apoptosis at the time of NMDAR acquisition. This cumulated in a 40% loss of neuroblasts along their migratory route, demonstrating that NMDAR acquisition is critical for neuroblast survival prior to entering a synaptic network. In addition, our findings suggest an unexpected mechanism wherein SVZ astrocytes use glutamate signaling through NMDARs to control the number of adult-born neurons reaching their final destination.
- Published
- 2010
- Full Text
- View/download PDF
47. GABA increases Ca2+in cerebellar granule cell precursors via depolarization: Implications for proliferation
- Author
-
Angélique Bordey and Kathleen A. Dave
- Subjects
Cerebellum ,Cell signaling ,Clinical Biochemistry ,Glutamic Acid ,Biology ,Biochemistry ,Article ,gamma-Aminobutyric acid ,GABA receptor ,Genetics ,medicine ,Humans ,Cerebellar Neoplasms ,Molecular Biology ,Cells, Cultured ,gamma-Aminobutyric Acid ,Cell Proliferation ,Metabotropic glutamate receptor 6 ,Glutamate receptor ,Cell Biology ,Receptors, GABA-A ,Granule cell ,Cell biology ,medicine.anatomical_structure ,nervous system ,Metabotropic glutamate receptor ,Calcium ,Medulloblastoma ,Signal Transduction ,medicine.drug - Abstract
The amino acids glutamate and gamma-aminobutyric acid (GABA) have primarily been characterized as the most prevalent excitatory and inhibitory, respectively, neurotransmitters in the vertebrate central nervous system. However, the role of these signaling molecules extends far beyond the synapse. GABA, glutamate, and their complement of receptors are essential signaling molecules that regulate developmental processes in both embryonic and young adult mammals. In this review, we describe the current knowledge on the role of GABA and glutamate in development, focusing on the perinatal cerebellum. We will then present novel data suggesting that GABA depolarizes granule cell precursors via GABA(A) receptors, which leads to calcium increases in these cells. Finally, we will consider the role of GABA and glutamate signaling on cell proliferation and perhaps neural cancers. From our review of the literature and these data, we hypothesize that GABA(A) receptors and metabotropic glutamate receptors may be a novel target for the pharmacological regulation of the cerebellar tumors, medulloblastomas.
- Published
- 2009
- Full Text
- View/download PDF
48. Tonic activation of GLUK5kainate receptors decreases neuroblast migration in whole-mounts of the subventricular zone
- Author
-
Jean Claude Platel, Tristan G. Heintz, Valerie Gordon, Stephanie Z. Young, and Angélique Bordey
- Subjects
animal structures ,Physiology ,animal diseases ,fungi ,Subventricular zone ,Kainate receptor ,Bicuculline ,Biology ,Cell biology ,medicine.anatomical_structure ,nervous system ,Neuroblast ,Metabotropic glutamate receptor ,Neuroblast migration ,mental disorders ,medicine ,Receptor ,Neuroscience ,Ganglion mother cell ,medicine.drug - Abstract
In the postnatal subventricular zone (SVZ), neuroblasts migrate in chains along the lateral ventricle towards the olfactory bulb. AMPA/kainate receptors as well as metabotropic glutamate receptors subtype 5 (mGluR5) are expressed in SVZ cells. However, the cells expressing these receptors and the function of these receptors remain unexplored. We thus examined whether SVZ neuroblasts express mGluR5 and Ca2+-permeable kainate receptors in mouse slices. Doublecortin (DCX)-immunopositive cells (i.e. neuroblasts) immunostained positive for mGluR5 and GLUK5-7-containing kainate receptors. RT-PCR from ∼10 GFP-fluorescent cell aspirates obtained in acute slices from transgenic mice expressing green fluorescent protein (GFP) under the DCX promoter showed mGluR5 and GLUK5 receptor mRNA in SVZ neuroblasts. Patch-clamp data suggest that ∼60% of neuroblasts express functional GLUK5-containing receptors. Activation of mGluR5 and GLUK5-containing receptors induced Ca2+ increases in 50% and 60% of SVZ neuroblasts, respectively, while most neuroblasts displayed GABAA-mediated Ca2+ responses. To examine the effects of these receptors on the speed of neuroblast migration, we developed a whole-mount preparation of the entire lateral ventricle from postnatal day (P) 20–25 DCX-GFP mice. The GABAA receptor (GABAAR) antagonist bicuculline increased the speed of neuroblast migration by 27%, as previously reported in acute slices. While the mGluR5 antagonist MPEP did not affect the speed of neuroblast migration, the homomeric and heteromeric GLUK5 receptor antagonists, NS3763 and UB302, respectively, increased the migration speed by 38%. These data show that although both GLUK5 receptor and mGluR5 activations increase Ca2+ in neuroblasts, only GLUK5 receptors tonically reduce the speed of neuroblast migration along the lateral ventricle.
- Published
- 2008
- Full Text
- View/download PDF
49. Control of neuroblast production and migration by converging GABA and glutamate signals in the postnatal forebrain
- Author
-
Kathleen A. Dave, Angélique Bordey, and Jean-Claude Platel
- Subjects
animal structures ,Physiology ,Cellular differentiation ,Neurogenesis ,Glutamate receptor ,Biology ,Neural stem cell ,medicine.anatomical_structure ,nervous system ,Neuroblast ,medicine ,Neuron ,Stem cell ,Ganglion mother cell ,Neuroscience - Abstract
The production of adult-born neurons is an ongoing process accounting for > 10 000 immature neurons migrating to the olfactory bulb every day. This high turnover rate necessitates profound control mechanisms converging onto neural stem cells and neuroblasts to achieve adequate adult-born neuron production. Here, we elaborate on a novel epigenetic control of adult neurogenesis via highly coordinated, non-synaptic, intercellular signalling. This communication engages the neurotransmitters GABA and glutamate, whose extracellular concentrations depend on neuroblast number and high affinity uptake systems in stem cells. Previous studies show that neuroblasts release GABA providing a negative feedback control of stem cell proliferation. Recent findings show an unexpected mosaic expression of glutamate receptors leading to calcium elevations in migrating neuroblasts. We speculate that stem cells release glutamate that activates glutamate receptors on migrating neuroblasts providing them with migratory and survival cues. In addition, we propose that the timing of neurotransmitter release and their spatial diffusion will determine the convergent coactivation of neuroblasts and stem cells, and provide a steady-state level of neuroblast production. Upon external impact or injury this signalling may adjust to a new steady-state level, thus providing non-synaptic scaling of neuroblast production.
- Published
- 2008
- Full Text
- View/download PDF
50. GABA and glutamate signaling: homeostatic control of adult forebrain neurogenesis
- Author
-
Benjamin Lacar, Jean-Claude Platel, and Angélique Bordey
- Subjects
medicine.medical_specialty ,Histology ,Physiology ,Organogenesis ,animal diseases ,Glutamic Acid ,Subventricular zone ,Kainate receptor ,Biology ,Models, Biological ,Article ,Cerebral Ventricles ,Mice ,Prosencephalon ,Internal medicine ,medicine ,Animals ,Homeostasis ,GABA transporter ,gamma-Aminobutyric Acid ,Neurons ,Metabotropic glutamate receptor 5 ,Metabotropic glutamate receptor 6 ,Glutamate receptor ,Cell Biology ,General Medicine ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,nervous system ,Metabotropic glutamate receptor ,Astrocytes ,biology.protein ,GABAergic ,Neuroscience ,Signal Transduction - Abstract
The neurotransmitter GABA exerts a strong negative influence on the production of adult-born olfactory bulb interneurons via tightly regulated, non-synaptic GABAergic signaling. After discussing some findings on GABAergic signaling in the neurogenic subventricular zone (SVZ), we provide data suggesting ambient GABA clearance via two GABA transporter subtypes and further support for a non-vesicular mechanism of GABA release from neuroblasts. While GABA works in cooperation with the neurotransmitter glutamate during embryonic cortical development, the role of glutamate in adult forebrain neurogenesis remains obscure. Only one of the eight metabotropic glutamate receptors (mGluRs), mGluR5, has been reported to tonically increase the number of proliferative SVZ cells in vivo, suggesting a local source of glutamate in the SVZ. We show here that glutamate antibodies strongly label subventricular zone (SVZ) astrocytes, some of which are stem cells. We also show that some SVZ neuroblasts express one of the ionotropic glutamate receptors, AMPA/kainate receptors, earlier than previously thought. Collectively, these findings suggest that neuroblast-to-astrocyte GABAergic signaling may cooperate with astrocyte-to-neuroblast glutamatergic signaling to provide strong homeostatic control on the production of adult-born olfactory bulb interneurons.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.