Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Transl Neurosci ; 15(1): 20220330, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38283997

ABSTRACT

Objective: Heterozygous mutations within the voltage-gated sodium channel α subunit (SCN1A) are responsible for the majority of cases of Dravet syndrome (DS), a severe developmental and epileptic encephalopathy. Development of novel therapeutic approaches is mandatory in order to directly target the molecular consequences of the genetic defect. The aim of the present study was to investigate whether cis-acting long non-coding RNAs (lncRNAs) of SCN1A are expressed in brain specimens of children and adolescent with epilepsy as these molecules comprise possible targets for precision-based therapy approaches. Methods: We investigated SCN1A mRNA expression and expression of two SCN1A related antisense RNAs in brain tissues in different age groups of pediatric non-Dravet patients who underwent surgery for drug resistant epilepsy. The effect of different antisense oligonucleotides (ASOs) directed against SCN1A specific antisense RNAs on SCN1A expression was tested. Results: The SCN1A related antisense RNAs SCN1A-dsAS (downstream antisense, RefSeq identifier: NR_110598) and SCN1A-usAS (upstream AS, SCN1A-AS, RefSeq identifier: NR_110260) were widely expressed in the brain of pediatric patients. Expression patterns revealed a negative correlation of SCN1A-dsAS and a positive correlation of lncRNA SCN1A-usAS with SCN1A mRNA expression. Transfection of SK-N-AS cells with an ASO targeted against SCN1A-dsAS was associated with a significant enhancement of SCN1A mRNA expression and reduction in SCN1A-dsAS transcripts. Conclusion: These findings support the role of SCN1A-dsAS in the suppression of SCN1A mRNA generation. Considering the haploinsufficiency in genetic SCN1A related DS, SCN1A-dsAS is an interesting target candidate for the development of ASOs (AntagoNATs) based precision medicine therapeutic approaches aiming to enhance SCN1A expression in DS.

2.
Cell Rep ; 15(1): 86-95, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27052171

ABSTRACT

Hyperfunction of the mTORC1 pathway has been associated with idiopathic and syndromic forms of autism spectrum disorder (ASD), including tuberous sclerosis, caused by loss of either TSC1 or TSC2. It remains largely unknown how developmental processes and biochemical signaling affected by mTORC1 dysregulation contribute to human neuronal dysfunction. Here, we have characterized multiple stages of neurogenesis and synapse formation in human neurons derived from TSC2-deleted pluripotent stem cells. Homozygous TSC2 deletion causes severe developmental abnormalities that recapitulate pathological hallmarks of cortical malformations in patients. Both TSC2(+/-) and TSC2(-/-) neurons display altered synaptic transmission paralleled by molecular changes in pathways associated with autism, suggesting the convergence of pathological mechanisms in ASD. Pharmacological inhibition of mTORC1 corrects developmental abnormalities and synaptic dysfunction during independent developmental stages. Our results uncouple stage-specific roles of mTORC1 in human neuronal development and contribute to a better understanding of the onset of neuronal pathophysiology in tuberous sclerosis.


Subject(s)
Multiprotein Complexes/antagonists & inhibitors , Neural Stem Cells/metabolism , Neurogenesis , Synapses/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tuberous Sclerosis/metabolism , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Synapses/physiology , Synaptic Transmission , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
3.
Cancer Cell ; 28(6): 730-742, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26669487

ABSTRACT

In the brain, Notch signaling maintains normal neural stem cells, but also brain cancer stem cells, indicating an oncogenic role. Here, we identify an unexpected tumor suppressor function for Notch in forebrain tumor subtypes. Genetic inactivation of RBP-Jκ, a key Notch mediator, or Notch1 and Notch2 receptors accelerates PDGF-driven glioma growth in mice. Conversely, genetic activation of the Notch pathway reduces glioma growth and increases survival. In humans, high Notch activity strongly correlates with distinct glioma subtypes, increased patient survival, and lower tumor grade. Additionally, simultaneous inactivation of RBP-Jκ and p53 induces primitive neuroectodermal-like tumors in mice. Hence, Notch signaling cooperates with p53 to restrict cell proliferation and tumor growth in mouse models of human brain tumors.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Prosencephalon/metabolism , Receptors, Notch/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Proliferation , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Glioma/genetics , Glioma/mortality , Glioma/pathology , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Infusions, Intraventricular , Kaplan-Meier Estimate , Mice, Knockout , Neoplasm Grading , Neoplastic Stem Cells/pathology , Neural Stem Cells/pathology , Phenotype , Platelet-Derived Growth Factor/administration & dosage , Prosencephalon/pathology , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptors, Notch/genetics , Recombinant Proteins/administration & dosage , Repressor Proteins/genetics , Repressor Proteins/metabolism , Time Factors , Tumor Burden , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
4.
Handb Exp Pharmacol ; 228: 99-155, 2015.
Article in English | MEDLINE | ID: mdl-25977081

ABSTRACT

Adult hippocampal neurogenesis is a remarkable form of brain structural plasticity by which new functional neurons are generated from adult neural stem cells/precursors. Although the precise role of this process remains elusive, adult hippocampal neurogenesis is important for learning and memory and it is affected in disease conditions associated with cognitive impairment, depression, and anxiety. Immature neurons in the adult brain exhibit an enhanced structural and synaptic plasticity during their maturation representing a unique population of neurons to mediate specific hippocampal function. Compelling preclinical evidence suggests that hippocampal neurogenesis is modulated by a broad range of physiological stimuli which are relevant in cognitive and emotional states. Moreover, multiple pharmacological interventions targeting cognition modulate adult hippocampal neurogenesis. In addition, recent genetic approaches have shown that promoting neurogenesis can positively modulate cognition associated with both physiology and disease. Thus the discovery of signaling pathways that enhance adult neurogenesis may lead to therapeutic strategies for improving memory loss due to aging or disease. This chapter endeavors to review the literature in the field, with particular focus on (1) the role of hippocampal neurogenesis in cognition in physiology and disease; (2) extrinsic and intrinsic signals that modulate hippocampal neurogenesis with a focus on pharmacological targets; and (3) efforts toward novel strategies pharmacologically targeting neurogenesis and identification of biomarkers of human neurogenesis.


Subject(s)
Biomarkers/metabolism , Cognition/drug effects , Hippocampus/drug effects , Mental Disorders/drug therapy , Neurogenesis/drug effects , Nootropic Agents/therapeutic use , Animals , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Drug Design , Gene Expression Regulation , Hippocampus/physiopathology , Humans , Mental Disorders/genetics , Mental Disorders/metabolism , Mental Disorders/physiopathology , Mental Disorders/psychology , Molecular Targeted Therapy , Signal Transduction/drug effects
5.
Proteomics Clin Appl ; 9(7-8): 684-94, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25684324

ABSTRACT

PURPOSE: Human pluripotent stem cell (hPSC)-derived cellular models have great potential to enable drug discovery and improve translation of preclinical insights to the clinic. We have developed a hPSC-derived neural precursor cell model for studying early events in human brain development. We present protein-level characterization of this model, using a multiplexed SRM approach, to establish reproducibility and physiological relevance; essential prerequisites for utilization of the neuronal development model in phenotypic screening-based drug discovery. EXPERIMENTAL DESIGN: Profiles of 246 proteins across three key stages of in vitro neuron differentiation were analyzed by SRM. Three independently hPSC-derived isogenic neural stem cell (NSC) lines were analyzed across five to nine independent neuronal differentiations. RESULTS: One hundred seventy-five proteins were reliably quantified revealing a time-dependent pattern of protein regulation that reflected protein dynamics during in vivo brain development and that was conserved across replicate differentiations and multiple cell lines. CONCLUSIONS AND CLINICAL RELEVANCE: SRM-based protein profiling enabled establishment of the reproducibility and physiological relevance of the hPSC-derived neuronal model. Combined with the successful quantification of proteins relevant to neurodevelopmental diseases, this validates the platform for use as a model to enable neuroscience drug discovery.


Subject(s)
Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proteomics/methods , Cell Differentiation , Cell Line , Cluster Analysis , Humans , Models, Biological , Principal Component Analysis , Time Factors
6.
Stem Cells ; 32(1): 70-84, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23964022

ABSTRACT

Neural stem cells (NSCs) in the ventricular domain of the subventricular zone (V-SVZ) of rodents produce neurons throughout life while those in humans become largely inactive or may be lost during infancy. Most adult NSCs are quiescent, express glial markers, and depend on Notch signaling for their self-renewal and the generation of neurons. Using genetic markers and lineage tracing, we identified subpopulations of adult V-SVZ NSCs (type 1, 2, and 3) indicating a striking heterogeneity including activated, brain lipid binding protein (BLBP, FABP7) expressing stem cells. BLBP(+) NSCs are mitotically active components of pinwheel structures in the lateral ventricle walls and persistently generate neurons in adulthood. BLBP(+) NSCs express epidermal growth factor (EGF) receptor, proliferate in response to EGF, and are a major clonogenic population in the SVZ. We also find BLBP expressed by proliferative ventricular and subventricular progenitors in the fetal and postnatal human brain. Loss of BLBP(+) stem/progenitor cells correlates with reduced neurogenesis in aging rodents and postnatal humans. These findings of molecular heterogeneity and proliferative differences subdivide the NSC population and have implications for neurogenesis in the forebrain of mammals during aging.


Subject(s)
Neural Stem Cells/cytology , Neurons/cytology , Prosencephalon/cytology , Animals , Cell Growth Processes/physiology , Humans , Mice , Mice, Transgenic , Neural Stem Cells/metabolism , Neurogenesis , Neurons/metabolism , Prosencephalon/metabolism , Signal Transduction
7.
Nat Neurosci ; 15(7): 962-9, 2012 Jun 17.
Article in English | MEDLINE | ID: mdl-22706270

ABSTRACT

Temporal regulation of embryonic neurogenesis is controlled by hypostable transcription factors. The mechanism of the process is unclear. Here we show that the RNase III Drosha and DGCR8 (also known as Pasha), key components of the microRNA (miRNA) microprocessor, have important functions in mouse neurogenesis. Loss of microprocessor in forebrain neural progenitors resulted in a loss of stem cell character and precocious differentiation whereas Dicer deficiency did not. Drosha negatively regulated expression of the transcription factors Neurogenin 2 (Ngn2) and NeuroD1 whereas forced Ngn2 expression phenocopied the loss of Drosha. Neurog2 mRNA contains evolutionarily conserved hairpins with similarities to pri-miRNAs, and associates with the microprocessor in neural progenitors. We uncovered a Drosha-dependent destabilization of Neurog2 mRNAs consistent with microprocessor cleavage at hairpins. Our findings implicate direct and miRNA-independent destabilization of proneural mRNAs by the microprocessor, which facilitates neural stem cell (NSC) maintenance by blocking accumulation of differentiation and determination factors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Gene Expression Regulation , MicroRNAs , Nerve Tissue Proteins/biosynthesis , Neurogenesis/physiology , Ribonuclease III/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/genetics , MicroRNAs/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Pregnancy , Ribonuclease III/antagonists & inhibitors
8.
Nat Commun ; 3: 670, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22334073

ABSTRACT

Neural stem/progenitor cells generate neurons in the adult hippocampus. Neural stem cells produce transient intermediate progenitors (type-2 cells), which generate neuroblasts (type-3 cells) that exit the cell cycle, and differentiate into neurons. The precise dynamics of neuron production from the neural stem cells remains controversial. Here we lineage trace Notch-dependent neural stem cells in the dentate gyrus and show that over 7-21 days, the progeny of the neural stem cells progress through an Ascl1(high) intermediate stage (type-2a) to neuroblasts. However, contrary to predictions, this Ascl1(high) population is not an amplifying intermediate, but it differentiates into mitotic Tbr2(+) early neuroblasts, which in turn expand the lineage. After 100 days, the majority of the neural stem cell progeny are neuroblasts or postmitotic neurons. Hence, the neural stem cells require many weeks to generate differentiated neurons. On the basis of this temporal delay in differentiation and population expansion, we propose that the neural stem cell and early neuroblast divisions drive dentate gyrus neurogenesis and not the amplification of type-2a intermediate progenitors as was previously thought.


Subject(s)
Hippocampus/metabolism , Neurogenesis , Stem Cells/cytology , Animals , Animals, Newborn , Bromodeoxyuridine/pharmacology , Cell Differentiation/genetics , Cell Lineage , Developmental Biology/methods , Mice , Mice, Transgenic , Mitosis , Neural Stem Cells , Neurons/cytology , Neurons/metabolism
9.
Cell Stem Cell ; 8(5): 464-5, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21549320
10.
Cell Stem Cell ; 6(5): 445-56, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20452319

ABSTRACT

New neurons are generated in the adult hippocampus throughout life by neural stem/progenitor cells (NSCs), and neurogenesis is a plastic process responsive to external stimuli. We show that canonical Notch signaling through RBP-J is required for hippocampal neurogenesis. Notch signaling distinguishes morphologically distinct Sox2(+) NSCs, and within these pools subpopulations can shuttle between mitotically active or quiescent. Radial and horizontal NSCs respond selectively to neurogenic stimuli. Physical exercise activates the quiescent radial population whereas epileptic seizures induce expansion of the horizontal NSC pool. Surprisingly, reduced neurogenesis correlates with a loss of active horizontal NSCs in aged mice rather than a total loss of stem cells, and the transition to a quiescent state is reversible to rejuvenate neurogenesis in the brain. The discovery of multiple NSC populations with Notch dependence but selective responses to stimuli and reversible quiescence has important implications for the mechanisms of adaptive learning and also for regenerative therapy.


Subject(s)
Aging/pathology , Cell Shape , Hippocampus/pathology , Neurons/pathology , Physical Conditioning, Animal , Seizures/pathology , Stem Cells/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Proliferation , Green Fluorescent Proteins/metabolism , Mice , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Neurogenesis , Neurons/metabolism , Receptors, Notch/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Seizures/metabolism , Signal Transduction , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL