Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Brain ; 145(4): 1310-1325, 2022 05 24.
Article in English | MEDLINE | ID: mdl-34849602

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.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Epilepsy , TOR Serine-Threonine Kinases , Adaptor Proteins, Signal Transducing/genetics , Animals , Brain/pathology , Cell Cycle Proteins/genetics , Epilepsy/pathology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mice , Neurons/metabolism , Phosphorylation , Seizures/chemically induced , Seizures/genetics , Seizures/metabolism , TOR Serine-Threonine Kinases/metabolism
2.
Cells ; 10(3)2021 03 11.
Article in English | MEDLINE | ID: mdl-33799820

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.


Subject(s)
Dendritic Spines/metabolism , Paracrine Communication , Pyramidal Cells/metabolism , Sensory Receptor Cells/metabolism , Somatosensory Cortex/metabolism , Synaptic Transmission , Vibrissae/innervation , rab27 GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials , Extracellular Vesicles/metabolism , Female , Gene Expression Regulation, Developmental , Gestational Age , Mice , Pregnancy , Somatosensory Cortex/cytology , rab27 GTP-Binding Proteins/genetics
3.
J Neurosci ; 41(17): 3799-3807, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33741723

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 neurons in vitro obtained 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-exist in vivo during 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 STATEMENT A 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.


Subject(s)
Dendritic Spines/physiology , Extracellular Space/physiology , Histone Deacetylase 2/genetics , Histone Deacetylase 2/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Animals , Cytoplasm/metabolism , Exosomes/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Male , Mice , Primary Cell Culture , Proteomics , Synapses/physiology
4.
Neurosci Res ; 143: 44-52, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29857015

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 ∼7300 genes were significantly altered over time and included classical neuronal genes known to decrease (e.g., Tbr1, Dcx) or increase (e.g., Eno2, Camk2a, Syn1) 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 Lynx1, Nrn1, Cntnap1 over time; downregulation of St8sia2 and Draxin; and bidirectional changes to Fkbp1b. 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.


Subject(s)
Electroporation/methods , Protein Biosynthesis , Pyramidal Cells/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Animals , Brain/embryology , Brain/metabolism , Disks Large Homolog 4 Protein/metabolism , Doublecortin Protein , Embryonic Development , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Oligonucleotide Array Sequence Analysis/methods , Pyramidal Cells/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Protein L10 , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism , Synapsins/metabolism
5.
Proc Natl Acad Sci U S A ; 113(40): 11330-11335, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647922

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.


Subject(s)
Carrier Proteins/metabolism , Eukaryotic Initiation Factors/metabolism , Neurons/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , Dendritic Spines/metabolism , Excitatory Postsynaptic Potentials , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Neuroglia/metabolism , RNA Caps/metabolism , RNA, Small Interfering/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Signal Transduction , Transcription Factors/metabolism
6.
Hum Mol Genet ; 24(20): 5746-58, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26220974

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 GSK3ß, a regulator of axogenesis. Although blocking GSK3ß 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 GSK3ß to abnormal axon growth in neurodevelopmental mTORopathies and open new therapeutic options for restoring long-range connectivity.


Subject(s)
Axons/physiology , Carrier Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Axons/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Growth Processes , Eukaryotic Initiation Factors , Female , Gene Expression Regulation , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Phosphoproteins/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction
7.
PLoS One ; 10(3): e0120693, 2015.
Article in English | MEDLINE | ID: mdl-25822989

ABSTRACT

Both the development and relief of stress-related psychiatric conditions such as major depression (MD) and post-traumatic stress disorder (PTSD) have been linked to neuroplastic changes in the brain. One such change involves the birth of new neurons (neurogenesis), which occurs throughout adulthood within discrete areas of the mammalian brain, including the dorsal hippocampus (HIP). Stress can trigger MD and PTSD in humans, and there is considerable evidence that it can decrease HIP neurogenesis in laboratory animals. In contrast, antidepressant treatments increase HIP neurogenesis, and their efficacy is eliminated by ablation of this process. These findings have led to the working hypothesis that HIP neurogenesis serves as a biomarker of neuroplasticity and stress resistance. Here we report that local alterations in the expression of Sprouty2 (SPRY2), an intracellular inhibitor of growth factor function, produces profound effects on both HIP neurogenesis and behaviors that reflect sensitivity to stressors. Viral vector-mediated disruption of endogenous Sprouty2 function (via a dominant negative construct) within the dorsal HIP of adult rats stimulates neurogenesis and produces signs of stress resilience including enhanced extinction of conditioned fear. Conversely, viral vector-mediated elevation of SPRY2 expression intensifies the behavioral consequences of stress. Studies of these manipulations in HIP primary cultures indicate that SPRY2 negatively regulates fibroblast growth factor-2 (FGF2), which has been previously shown to produce antidepressant- and anxiolytic-like effects via actions in the HIP. Our findings strengthen the relationship between HIP plasticity and stress responsiveness, and identify a specific intracellular pathway that could be targeted to study and treat stress-related disorders.


Subject(s)
Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Stress, Psychological/metabolism , Animals , Depression/metabolism , Depression/physiopathology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Fibroblast Growth Factor 2/metabolism , Hippocampus/physiopathology , Male , Neuronal Plasticity/physiology , Neurons/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/physiopathology
8.
Neuron ; 84(1): 78-91, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25277454

ABSTRACT

Abnormal dendritic complexity is a shared feature of many neurodevelopmental disorders associated with neurological defects. Here, we found that the actin-crosslinking protein filamin A (FLNA) is overexpressed in tuberous sclerosis complex (TSC) mice, a PI3K-mTOR model of neurodevelopmental disease that is associated with abnormal dendritic complexity. Both under- and overexpression of FLNA in wild-type neurons led to more complex dendritic arbors in vivo, suggesting that an optimal level of FLNA expression is required for normal dendritogenesis. In Tsc1(null) neurons, knocking down FLNA in vivo prevented dendritic abnormalities. Surprisingly, FLNA overexpression in Tsc1(null) neurons was dependent on MEK1/2 but not mTOR activity, despite both pathways being hyperactive. In addition, increasing MEK-ERK1/2 activity led to dendritic abnormalities via FLNA, and decreasing MEK-ERK1/2 signaling in Tsc1(null) neurons rescued dendritic defects. These data demonstrate that altered FLNA expression increases dendritic complexity and contributes to pathologic dendritic patterning in TSC in an mTOR-independent, ERK1/2-dependent manner.


Subject(s)
Dendrites/metabolism , Filamins/biosynthesis , Gene Expression Regulation , MAP Kinase Signaling System/physiology , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism , Adult , Animals , Animals, Newborn , Cell Line, Tumor , Dendrites/pathology , Female , Filamins/genetics , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Organ Culture Techniques , Tuberous Sclerosis/pathology , Young Adult
9.
Front Cell Neurosci ; 8: 10, 2014.
Article in English | MEDLINE | ID: mdl-24478632

ABSTRACT

GABA regulates the behavior of neuroblasts and neural progenitor cells in the postnatal neurogenic subventricular zone (SVZ) through GABAA receptor (GABAAR)-mediated calcium increases. However, the source of GABA necessary for sufficient GABAAR-mediated depolarization and calcium increase has remained speculative. Here, we explored whether GABAergic striatal neurons functionally connect with SVZ cells. Using patch clamp recordings or single cell electroporation, striatal neurons along the SVZ were filled with a fluorescent dye revealing that they send both dendrites and axons into the SVZ. About 93% of the recorded neurons were medium spiny or aspiny GABAergic neurons and each neuron sent 3-4 processes into the SVZ covering ~56 µm. Using calcium imaging, we found that depolarization of striatal neurons led to increased calcium activity in SVZ cells that were mediated by GABAAR activation. Collectively, these findings undercover a novel mode of signaling in the SVZ providing a mechanism of brain activity-mediated regulation of postnatal neurogenesis through GABAergic striatal activity.

10.
Cell Rep ; 5(2): 433-44, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24139800

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.


Subject(s)
Eukaryotic Initiation Factors/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Differentiation/drug effects , Cells, Cultured , Eukaryotic Initiation Factors/antagonists & inhibitors , Eukaryotic Initiation Factors/genetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Ras Homolog Enriched in Brain Protein , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sirolimus/pharmacology
11.
Int J Dev Neurosci ; 31(7): 667-78, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23485365

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant monogenetic disorder that is characterized by the formation of benign tumors in several organs as well as brain malformations and neuronal defects. TSC is caused by inactivating mutations in one of two genes, TSC1 and TSC2, resulting in increased activity of the mammalian Target of Rapamycin (mTOR). Here, we explore the cytoarchitectural and functional CNS aberrations that may account for the neurological presentations of TSC, notably seizures, hydrocephalus, and cognitive and psychological impairments. In particular, recent mouse models of brain lesions are presented with an emphasis on using electroporation to allow the generation of discrete lesions resulting from loss of heterozygosity during perinatal development. Cortical lesions are thought to contribute to epileptogenesis and worsening of cognitive defects. However, it has recently been suggested that being born with a mutant allele without loss of heterozygosity and associated cortical lesions is sufficient to generate cognitive and neuropsychiatric problems. We will thus discuss the function of mTOR hyperactivity on neuronal circuit formation and the potential consequences of being born heterozygous on neuronal function and the biochemistry of synaptic plasticity, the cellular substrate of learning and memory. Ultimately, a major goal of TSC research is to identify the cellular and molecular mechanisms downstream of mTOR underlying the neurological manifestations observed in TSC patients and identify novel therapeutic targets to prevent the formation of brain lesions and restore neuronal function.


Subject(s)
Central Nervous System/metabolism , Cognition Disorders/etiology , Epilepsy/etiology , Tuberous Sclerosis , Animals , Central Nervous System/pathology , Cognition Disorders/genetics , Disease Models, Animal , Epilepsy/genetics , Humans , Mice , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/complications , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology
12.
J Neurosci ; 33(6): 2419-31, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23392671

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 (Rheb(CA)) 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 Rheb(CA)-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 in Tsc1 mutant mice. In addition, increased mTOR activity in individuals without known mutations could significantly impact neurogenesis and circuit formation.


Subject(s)
Dendrites/metabolism , Monomeric GTP-Binding Proteins/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Olfactory Bulb/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Enlargement/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cerebral Ventricles/metabolism , Cerebral Ventricles/pathology , Dendrites/drug effects , Dendrites/pathology , Electroporation/methods , Female , Hypertrophy/pathology , Male , Mice , Monomeric GTP-Binding Proteins/physiology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/pathology , Neuropeptides/physiology , Olfactory Bulb/drug effects , Olfactory Bulb/pathology , Ras Homolog Enriched in Brain Protein , Sirolimus/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism
13.
PLoS One ; 7(5): e38174, 2012.
Article in English | MEDLINE | ID: mdl-22693596

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.


Subject(s)
Dendritic Spines/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/cytology , Neurons/metabolism , Olfactory Bulb/cytology , Synapses/metabolism , Animals , Animals, Newborn , Cell Survival/genetics , Cerebral Ventricles/cytology , Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dendritic Spines/genetics , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Mice , Neurogenesis/genetics , Neuronal Plasticity/genetics , Olfactory Bulb/growth & development , Synapses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...