Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters










Publication year range
1.
Elife ; 122024 Feb 27.
Article in English | MEDLINE | ID: mdl-38411613

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.


Subject(s)
Drug Resistant Epilepsy , Neurons , Animals , Mice , Pyramidal Cells , Brain , Mechanistic Target of Rapamycin Complex 1/genetics
2.
bioRxiv ; 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-37609221

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 activators, Rheb or mTOR, or biallelic inactivation of the mTORC1 repressors, Depdc5, Tsc1, or Pten in 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.

3.
Sci Rep ; 13(1): 16260, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37758896

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.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Mice , Animals , Housing , Exploratory Behavior , Mice, Inbred C57BL , Social Behavior , Mice, Inbred Strains , Disease Models, Animal
4.
Front Neurosci ; 17: 1257056, 2023.
Article in English | MEDLINE | ID: mdl-37680968

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.

6.
eNeuro ; 10(8)2023 08.
Article in English | MEDLINE | ID: mdl-37620147

ABSTRACT

Several neurodevelopmental disorders are associated with increased mTOR activity that results in pathogenic neuronal dysmorphogenesis (i.e., soma and dendrite overgrowth), leading to circuit alterations associated with epilepsy and neurologic disabilities. Although an mTOR analog is approved for the treatment of epilepsy in one of these disorders, it has limited efficacy and is associated with a wide range of side effects. There is a need to develop novel agents for the treatment of mTOR-pathway related disorders. Here, we developed a medium-throughput phenotypic assay to test drug efficacy on neurite morphogenesis of mouse neurons in a hyperactive mTOR condition. Our assay involved in utero electroporation (IUE) of a selective population of cortical pyramidal neurons with a plasmid encoding the constitutively active mTOR activator, Rheb, and tdTomato. Labeled neurons from the somatosensory cortex (SSC) were cultured onto 96-well plates and fixed at various days in vitro or following Torin 1 treatment. Automated systems were used for image acquisition and neuron morphologic measurements. We validated our automated approach using traditional manual methods of neuron morphologic assessment. Both automated and manual analyses showed increased neurite length and complexity over time, and decreased neurite overgrowth and soma size with Torin 1. These data validate the accuracy of our automated approach that takes hours compared with weeks when using traditional manual methods. Taken together, this assay can be scaled to screen 32 compounds simultaneously in two weeks, highlighting its robustness and efficiency for medium-throughput screening of candidate therapeutics on a defined population of wild-type or diseased neurons.


Subject(s)
Neurites , Neurons , Animals , Mice , Pyramidal Cells , Electroporation , TOR Serine-Threonine Kinases
7.
eNeuro ; 10(8)2023 08.
Article in English | MEDLINE | ID: mdl-37596047

ABSTRACT

One of the most prevalent deficits in autism spectrum disorder (ASD) are sensitivities to sensory stimuli. Despite the prevalence of sensory deficits in autism, there are few paradigms capable of easily assessing sensory behaviors in ASD-like mouse models. We addressed this need by creating the Somatosensory Nose-poke Adapted Paradigm (SNAP), which consists of an elevated platform with 6 holes in the center, half of which are lined with sandpaper and half are smooth, requiring mice to use their whiskers to sense the texture. The SNAP paradigm assesses tactile sensory preferences as well as stereotypy, anxiety, and locomotion. We used two wild-type (neurotypical) mouse strains, C57BL/6J (C57) inbred and CD-1 outbred mice, and two ASD mouse models, BTBR (a model of idiopathic ASD) and Cntnap2 -/- mice (a model of syndromic ASD). We found that both ASD models produced more nose pokes into the rough condition than the smooth condition, suggesting an increased preference for complex tactile stimulation when compared with the neurotypical groups, wherein no differences were observed. Furthermore, we found increased stereotypy and time spent in the center, suggestive of decreased anxiety, only for BTBR mice compared with the other mouse strains. Overall, SNAP is an easy to implement task to assess the degree of preference for complex tactile stimulation in ASD mouse models that can be further modified to exclude possible confounding effects of novelty or anxiety on the sensory preferences.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Mice , Mice, Inbred C57BL , Stereotyped Behavior , Anxiety , Disease Models, Animal
8.
Biochim Biophys Acta Rev Cancer ; 1878(5): 188951, 2023 09.
Article in English | MEDLINE | ID: mdl-37433417

ABSTRACT

Glioma is the most prevalent type of neurological malignancies. Despite decades of efforts in neurosurgery, chemotherapy and radiation therapy, glioma remains one of the most treatment-resistant brain tumors with unfavorable outcomes. Recent progresses in genomic and epigenetic profiling have revealed new concepts of genetic events involved in the etiology of gliomas in humans, meanwhile, revolutionary technologies in gene editing and delivery allows to code these genetic "events" in animals to genetically engineer glioma models. This approach models the initiation and progression of gliomas in a natural microenvironment with an intact immune system and facilitates probing therapeutic strategies. In this review, we focus on recent advances in in vivo electroporation-based glioma modeling and outline the established genetically engineered glioma models (GEGMs).


Subject(s)
Brain Neoplasms , Glioma , Animals , Humans , Glioma/drug therapy , Brain Neoplasms/pathology , Genetic Engineering , Electroporation , Immune System , Tumor Microenvironment
9.
Epilepsia ; 64(6): 1432-1443, 2023 06.
Article in English | MEDLINE | ID: mdl-36869624

ABSTRACT

The hippocampal formation plays a central role in the development of temporal lobe epilepsy (TLE), a disease characterized by recurrent, unprovoked epileptic discharges. TLE is a neurologic disorder characterized by acute long-lasting seizures (i.e., abnormal electrical activity in the brain) or seizures that occur in close proximity without recovery, typically after a brain injury or status epilepticus. After status epilepticus, epileptogenic hyperexcitability develops gradually over the following months to years, resulting in the emergence of chronic, recurrent seizures. Acting as a filter or gate, the hippocampal dentate gyrus (DG) normally prevents excessive excitation from propagating through the hippocampus, and is considered a critical region in the progression of epileptogenesis in pathological conditions. Importantly, lipid-derived endogenous cannabinoids (endocannabinoids), which are produced on demand as retrograde messengers, are central regulators of neuronal activity in the DG circuit. In this review, we summarize recent findings concerning the role of the DG in controlling hyperexcitability and propose how DG regulation by cannabinoids (CBs) could provide avenues for therapeutic interventions. We also highlight possible pathways and manipulations that could be relevant for the control of hyperexcitation. The use of CB compounds to treat epilepsies is controversial, as anecdotal evidence is not always validated by clinical trials. Recent publications shed light on the importance of the DG as a region regulating incoming hippocampal excitability during epileptogenesis. We review recent findings concerning the modulation of the hippocampal DG circuitry by CBs and discuss putative underlying pathways. A better understanding of the mechanisms by which CBs exert their action during seizures may be useful to improve therapies.


Subject(s)
Cannabinoids , Epilepsy, Temporal Lobe , Epilepsy , Status Epilepticus , Humans , Animals , Hippocampus/pathology , Seizures/pathology , Epilepsy/etiology , Epilepsy/pathology , Epilepsy, Temporal Lobe/pathology , Neurons/pathology , Status Epilepticus/pathology , Dentate Gyrus/pathology , Disease Models, Animal
10.
Epilepsy Curr ; 22(4): 234-240, 2022.
Article in English | MEDLINE | ID: mdl-36187145

ABSTRACT

Focal cortical dysplasia (FCD) is a malformation of cortical development that is a prevalent cause of intractable epilepsy in children. Of the three FCD subtypes, understanding the etiology and pathogenesis of FCD type II has seen the most progress owing to the recent advances in identifying gene mutations along the mTOR signaling pathway as a frequent cause of this disorder. Accordingly, numerous animal models of FCD type II based on genetic manipulation of the mTOR signaling pathway have emerged to investigate the mechanisms of epileptogenesis and novel therapeutics for epilepsy. These include transgenic and in utero electroporation-based animal models. Here, we review the histopathological and electroclinical features of existing FCD type II animal models and discuss the scientific and technical considerations, clinical applications, and limitations of current models. We also highlight other models of FCD based on early life acquired factors.

12.
13.
Epilepsy Res ; 181: 106890, 2022 03.
Article in English | MEDLINE | ID: mdl-35219048

ABSTRACT

Tuberous sclerosis complex (TSC) is a monogenic disorder characterized by hyperactivation of the mTOR signaling pathway and developmental brain malformations leading to intractable epilepsy. Although treatment with the recently approved mTOR inhibitor, everolimus, results in clinically relevant seizure suppression in up to 40% of TSC patients, seizures remain uncontrolled in a large number of cases, underscoring the need to identify novel treatment targets. The MEK-ERK signaling pathway has been found to be aberrantly activated in TSC and inhibition of MEK-ERK activity independently of mTOR rescued neuronal dendrite overgrowth in mice modeling TSC neuropathology. Here, we evaluated the efficacy of MEK-ERK inhibition on seizures in two mouse models of TSC. We found that treatment with the MEK inhibitor PD0325901 (mirdametinib) significantly reduced seizure activity in both TSC mouse models. These findings support inhibiting MEK-ERK activity as a potential alternative strategy to treat seizures in TSC.


Subject(s)
Tuberous Sclerosis , Animals , Disease Models, Animal , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Seizures/drug therapy , Signal Transduction , Tuberous Sclerosis/complications , Tuberous Sclerosis/drug therapy
14.
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
15.
Epilepsy Curr ; 21(3): 197-198, 2021.
Article in English | MEDLINE | ID: mdl-34867103
16.
17.
JCI Insight ; 6(20)2021 10 22.
Article in English | MEDLINE | ID: mdl-34676826

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.


Subject(s)
Nuclear Receptor Co-Repressor 2/metabolism , Oligodendroglia/physiology , Peptides/physiology , RNA, Long Noncoding/genetics , Animals , Female , Humans , Mice , Pregnancy
18.
Front Psychiatry ; 12: 687060, 2021.
Article in English | MEDLINE | ID: mdl-34475829

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.

19.
Front Neuroanat ; 15: 715363, 2021.
Article in English | MEDLINE | ID: mdl-34295225

ABSTRACT

[This corrects the article DOI: 10.3389/fnana.2021.664695.].

20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...