Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
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
2.
Sci Transl Med ; 12(570)2020 11 18.
Article in English | MEDLINE | ID: mdl-33208499

ABSTRACT

The causative link between focal cortical malformations (FCMs) 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. Last, 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.


Subject(s)
Epilepsy , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Malformations of Cortical Development, Group I , Tuberous Sclerosis , Epilepsy/genetics , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Muscle Proteins , Potassium Channels/genetics , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/genetics
3.
Sci Transl Med ; 12(531)2020 02 19.
Article in English | MEDLINE | ID: mdl-32075941

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.


Subject(s)
Epilepsy , Malformations of Cortical Development, Group I , Animals , Filamins , Humans , Mice , Phosphatidylinositol 3-Kinases , Seizures/drug therapy
4.
J Neurosci ; 37(29): 6877-6893, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28630251

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.


Subject(s)
Amides/administration & dosage , Cognition Disorders/prevention & control , Cognition Disorders/physiopathology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/physiopathology , Encephalitis, Viral/drug therapy , Encephalitis, Viral/physiopathology , Animals , Animals, Newborn , Antiviral Agents/administration & dosage , Cognition Disorders/pathology , Cytomegalovirus Infections/pathology , Dose-Response Relationship, Drug , Encephalitis, Viral/pathology , Female , Male , Mice , Mice, Inbred C57BL , Treatment Outcome
5.
Neurosci Lett ; 637: 142-147, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27871995

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.


Subject(s)
Autistic Disorder/physiopathology , Behavior, Animal/physiology , Exploratory Behavior/physiology , Social Behavior , Animals , Disease Models, Animal , Male , Mice, Inbred Strains , Task Performance and Analysis
6.
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
7.
Nat Commun ; 7: 11753, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27249187

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.


Subject(s)
Cognitive Dysfunction/drug therapy , Malformations of Cortical Development/drug therapy , Neurons/metabolism , Seizures/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , Animals , Cell Movement , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Malformations of Cortical Development/genetics , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/pathology , Mice , Neurons/drug effects , Neurons/pathology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Seizures/genetics , Seizures/metabolism , Seizures/pathology , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , White Matter/drug effects , White Matter/metabolism , White Matter/pathology
8.
Nat Neurosci ; 19(7): 905-14, 2016 07.
Article in English | MEDLINE | ID: mdl-27239938

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.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic/drug effects , Histone-Lysine N-Methyltransferase/genetics , Nicotine/pharmacology , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Animals , Histones/metabolism , Methylation/drug effects , Mice, Inbred C57BL
9.
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
10.
J Comp Neurol ; 523(8): 1145-61, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25420934

ABSTRACT

Odor information relayed by olfactory bulb projection neurons, mitral and tufted cells (M/T), is modulated by pairs of reciprocal dendrodendritic synaptic circuits in the external plexiform layer (EPL). Interneurons, which are accounted for largely by granule cells, receive depolarizing input from M/T dendrites and in turn inhibit current spread in M/T dendrites via hyperpolarizing reciprocal dendrodendritic synapses. Because the location of dendrodendritic synapses may significantly affect the cascade of odor information, we assessed synaptic properties and density within sublaminae of the EPL and along the length of M/T secondary dendrites. In electron micrographs the M/T to granule cell synapse appeared to predominate and was equivalent in both the outer and inner EPL. However, the dendrodendritic synapses from granule cell spines onto M/T dendrites were more prevalent in the outer EPL. In contrast, individual gephyrin-immunoreactive (IR) puncta, a postsynaptic scaffolding protein at inhibitory synapses used here as a proxy for the granule to M/T dendritic synapse was equally distributed throughout the EPL. Of significance to the organization of intrabulbar circuits, gephyrin-IR synapses are not uniformly distributed along M/T secondary dendrites. Synaptic density, expressed as a function of surface area, increases distal to the cell body. Furthermore, the distributions of gephyrin-IR puncta are heterogeneous and appear as clusters along the length of the M/T dendrites. Consistent with computational models, our data suggest that temporal coding in M/T cells is achieved by precisely located inhibitory input and that distance from the soma is compensated for by an increase in synaptic density.


Subject(s)
Dendrites/ultrastructure , Olfactory Bulb/ultrastructure , Synapses/ultrastructure , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Dendrites/metabolism , Female , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Olfactory Bulb/anatomy & histology , Olfactory Bulb/metabolism , Photomicrography , Synapses/metabolism
11.
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
12.
PLoS One ; 8(8): e69989, 2013.
Article in English | MEDLINE | ID: mdl-23936366

ABSTRACT

In neurons, specific RNAs are assembled into granules, which are translated in dendrites, however the functional consequences of granule assembly are not known. Tumor overexpressed gene (TOG) is a granule-associated protein containing multiple binding sites for heterogeneous nuclear ribonucleoprotein (hnRNP) A2, another granule component that recognizes cis-acting sequences called hnRNP A2 response elements (A2REs) present in several granule RNAs. Translation in granules is sporadic, which is believed to reflect monosomal translation, with occasional bursts, which are believed to reflect polysomal translation. In this study, TOG expression was conditionally knocked out (TOG cKO) in mouse hippocampal neurons using cre/lox technology. In TOG cKO cultured neurons granule assembly and bursty translation of activity-regulated cytoskeletal associated (ARC) mRNA, an A2RE RNA, are disrupted. In TOG cKO brain slices synaptic sensitivity and long term potentiation (LTP) are reduced. TOG cKO mice exhibit hyperactivity, perseveration and impaired short term habituation. These results suggest that in hippocampal neurons TOG is required for granule assembly, granule translation and synaptic plasticity, and affects behavior.


Subject(s)
Gene Knockout Techniques , Habituation, Psychophysiologic/genetics , Long-Term Potentiation/genetics , Microtubule-Associated Proteins/genetics , Neurons/metabolism , Protein Biosynthesis/genetics , RNA/metabolism , Animals , Behavior, Animal/physiology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Cytoskeleton/metabolism , Excitatory Postsynaptic Potentials/genetics , Female , Male , Mice , Microtubule-Associated Proteins/deficiency , Neurons/cytology , RNA/genetics , Synapses/physiology
13.
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
14.
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
15.
Cereb Cortex ; 23(7): 1731-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22693342

ABSTRACT

Endocannabinoids (eCBs) play a prominent role in regulating synaptic signaling throughout the brain. In layer 2/3 of the neocortex, eCB-mediated suppression of GABA release results in an enhanced excitability of pyramidal neurons (PNs). The eCB system is also involved in spike timing-dependent plasticity that is dependent on backpropagating action potentials (bAPs). Dendritic backpropagation plays an important role in many aspects of neuronal function, and can be modulated by intrinsic dendritic conductances as well as by synaptic inputs. The present studies explored a role for the eCB system in modulating backpropagation in PN dendrites. Using dendritic calcium imaging and somatic patch clamp recordings from mouse somatosensory cortical slices, we found that activation of type 1 cannabinoid receptors potentiated bAP-induced calcium transients in apical dendrites of layer 2/3 but not layer 5 PNs. This effect was mediated by suppression of GABAergic transmission, because it was prevented by a GABAA receptor antagonist and was correlated with cannabinoid suppression of inhibitory synaptic activity. Finally, we found that activity-dependent eCB release during depolarization-induced suppression of inhibition enhanced bAP-induced dendritic calcium transients. Taken together, these results point to a potentially important role for the eCB system in regulating dendritic backpropagation in layer 2/3 PNs.


Subject(s)
Action Potentials/physiology , Calcium/metabolism , Endocannabinoids/metabolism , Pyramidal Cells/metabolism , Somatosensory Cortex/metabolism , Animals , Dendrites/metabolism , Mice , Patch-Clamp Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...