Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Sci Rep ; 14(1): 570, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38177237

ABSTRACT

Familial dysautonomia (FD) is a rare recessive neurodevelopmental disease caused by a splice mutation in the Elongator acetyltransferase complex subunit 1 (ELP1) gene. This mutation results in a tissue-specific reduction of ELP1 protein, with the lowest levels in the central and peripheral nervous systems (CNS and PNS, respectively). FD patients exhibit complex neurological phenotypes due to the loss of sensory and autonomic neurons. Disease symptoms include decreased pain and temperature perception, impaired or absent myotatic reflexes, proprioceptive ataxia, and progressive retinal degeneration. While the involvement of the PNS in FD pathogenesis has been clearly recognized, the underlying mechanisms responsible for the preferential neuronal loss remain unknown. In this study, we aimed to elucidate the molecular mechanisms underlying FD by conducting a comprehensive transcriptome analysis of neuronal tissues from the phenotypic mouse model TgFD9; Elp1Δ20/flox. This mouse recapitulates the same tissue-specific ELP1 mis-splicing observed in patients while modeling many of the disease manifestations. Comparison of FD and control transcriptomes from dorsal root ganglion (DRG), trigeminal ganglion (TG), medulla (MED), cortex, and spinal cord (SC) showed significantly more differentially expressed genes (DEGs) in the PNS than the CNS. We then identified genes that were tightly co-expressed and functionally dependent on the level of full-length ELP1 transcript. These genes, defined as ELP1 dose-responsive genes, were combined with the DEGs to generate tissue-specific dysregulated FD signature genes and networks. Within the PNS networks, we observed direct connections between Elp1 and genes involved in tRNA synthesis and genes related to amine metabolism and synaptic signaling. Importantly, transcriptomic dysregulation in PNS tissues exhibited enrichment for neuronal subtype markers associated with peptidergic nociceptors and myelinated sensory neurons, which are known to be affected in FD. In summary, this study has identified critical tissue-specific gene networks underlying the etiology of FD and provides new insights into the molecular basis of the disease.


Subject(s)
Dysautonomia, Familial , Humans , Mice , Animals , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Dysautonomia, Familial/pathology , Carrier Proteins/metabolism , Peripheral Nervous System/metabolism , Sensory Receptor Cells/metabolism , Gene Expression Profiling , Gene Expression
4.
bioRxiv ; 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37808686

ABSTRACT

Familial dysautonomia (FD) is a rare recessive neurodevelopmental disease caused by a splice mutation in the Elongator acetyltransferase complex subunit 1 ( ELP1 ) gene. This mutation results in a tissue-specific reduction of ELP1 protein, with the lowest levels in the central and peripheral nervous systems (CNS and PNS, respectively). FD patients exhibit complex neurological phenotypes due to the loss of sensory and autonomic neurons. Disease symptoms include decreased pain and temperature perception, impaired or absent myotatic reflexes, proprioceptive ataxia, and progressive retinal degeneration. While the involvement of the PNS in FD pathogenesis has been clearly recognized, the underlying mechanisms responsible for the preferential neuronal loss remain unknown. In this study, we aimed to elucidate the molecular mechanisms underlying FD by conducting a comprehensive transcriptome analysis of neuronal tissues from the phenotypic mouse model TgFD9 ; Elp1 Δ 20/flox . This mouse recapitulates the same tissue-specific ELP1 mis-splicing observed in patients while modeling many of the disease manifestations. Comparison of FD and control transcriptomes from dorsal root ganglion (DRG), trigeminal ganglion (TG), medulla (MED), cortex, and spinal cord (SC) showed significantly more differentially expressed genes (DEGs) in the PNS than the CNS. We then identified genes that were tightly co-expressed and functionally dependent on the level of full-length ELP1 transcript. These genes, defined as ELP1 dose-responsive genes, were combined with the DEGs to generate tissue-specific dysregulated FD signature genes and networks. Within the PNS networks, we observed direct connections between Elp1 and genes involved in tRNA synthesis and genes related to amine metabolism and synaptic signaling. Importantly, transcriptomic dysregulation in PNS tissues exhibited enrichment for neuronal subtype markers associated with peptidergic nociceptors and myelinated sensory neurons, which are known to be affected in FD. In summary, this study has identified critical tissue-specific gene networks underlying the etiology of FD and provides new insights into the molecular basis of the disease.

5.
Am J Hum Genet ; 110(3): 531-547, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36809767

ABSTRACT

Familial dysautonomia (FD) is a rare neurodegenerative disease caused by a splicing mutation in elongator acetyltransferase complex subunit 1 (ELP1). This mutation leads to the skipping of exon 20 and a tissue-specific reduction of ELP1, mainly in the central and peripheral nervous systems. FD is a complex neurological disorder accompanied by severe gait ataxia and retinal degeneration. There is currently no effective treatment to restore ELP1 production in individuals with FD, and the disease is ultimately fatal. After identifying kinetin as a small molecule able to correct the ELP1 splicing defect, we worked on its optimization to generate novel splicing modulator compounds (SMCs) that can be used in individuals with FD. Here, we optimize the potency, efficacy, and bio-distribution of second-generation kinetin derivatives to develop an oral treatment for FD that can efficiently pass the blood-brain barrier and correct the ELP1 splicing defect in the nervous system. We demonstrate that the novel compound PTC258 efficiently restores correct ELP1 splicing in mouse tissues, including brain, and most importantly, prevents the progressive neuronal degeneration that is characteristic of FD. Postnatal oral administration of PTC258 to the phenotypic mouse model TgFD9;Elp1Δ20/flox increases full-length ELP1 transcript in a dose-dependent manner and leads to a 2-fold increase in functional ELP1 in the brain. Remarkably, PTC258 treatment improves survival, gait ataxia, and retinal degeneration in the phenotypic FD mice. Our findings highlight the great therapeutic potential of this novel class of small molecules as an oral treatment for FD.


Subject(s)
Dysautonomia, Familial , Neurodegenerative Diseases , Retinal Degeneration , Mice , Animals , Dysautonomia, Familial/genetics , Kinetin , Gait Ataxia , Administration, Oral
6.
Am J Hum Genet ; 109(11): 2049-2067, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36283406

ABSTRACT

Point mutations and structural variants that directly disrupt the coding sequence of MEF2C have been associated with a spectrum of neurodevelopmental disorders (NDDs). However, the impact of MEF2C haploinsufficiency on neurodevelopmental pathways and synaptic processes is not well understood, nor are the complex mechanisms that govern its regulation. To explore the functional changes associated with structural variants that alter MEF2C expression and/or regulation, we generated an allelic series of 204 isogenic human induced pluripotent stem cell (hiPSC)-derived neural stem cells and glutamatergic induced neurons. These neuronal models harbored CRISPR-engineered mutations that involved direct deletion of MEF2C or deletion of the boundary points for topologically associating domains (TADs) and chromatin loops encompassing MEF2C. Systematic profiling of mutation-specific alterations, contrasted to unedited controls that were exposed to the same guide RNAs for each edit, revealed that deletion of MEF2C caused differential expression of genes associated with neurodevelopmental pathways and synaptic function. We also discovered significant reduction in synaptic activity measured by multielectrode arrays (MEAs) in neuronal cells. By contrast, we observed robust buffering against MEF2C regulatory disruption following deletion of a distal 5q14.3 TAD and loop boundary, whereas homozygous loss of a proximal loop boundary resulted in down-regulation of MEF2C expression and reduced electrophysiological activity on MEA that was comparable to direct gene disruption. Collectively, these studies highlight the considerable functional impact of MEF2C deletion in neuronal cells and systematically characterize the complex interactions that challenge a priori predictions of regulatory consequences from structural variants that disrupt three-dimensional genome organization.


Subject(s)
Induced Pluripotent Stem Cells , Neural Stem Cells , Humans , Genome , Haploinsufficiency , MEF2 Transcription Factors/genetics , Neurons , Transcription, Genetic
7.
Hum Mol Genet ; 31(11): 1776-1787, 2022 06 04.
Article in English | MEDLINE | ID: mdl-34908112

ABSTRACT

Familial dysautonomia (FD) is an autosomal recessive neurodegenerative disease caused by a splicing mutation in the gene encoding Elongator complex protein 1 (ELP1, also known as IKBKAP). This mutation results in tissue-specific skipping of exon 20 with a corresponding reduction of ELP1 protein, predominantly in the central and peripheral nervous system. Although FD patients have a complex neurological phenotype caused by continuous depletion of sensory and autonomic neurons, progressive visual decline leading to blindness is one of the most problematic aspects of the disease, as it severely affects their quality of life. To better understand the disease mechanism as well as to test the in vivo efficacy of targeted therapies for FD, we have recently generated a novel phenotypic mouse model, TgFD9; IkbkapΔ20/flox. This mouse exhibits most of the clinical features of the disease and accurately recapitulates the tissue-specific splicing defect observed in FD patients. Driven by the dire need to develop therapies targeting retinal degeneration in FD, herein, we comprehensively characterized the progression of the retinal phenotype in this mouse, and we demonstrated that it is possible to correct ELP1 splicing defect in the retina using the splicing modulator compound (SMC) BPN-15477.


Subject(s)
Dysautonomia, Familial , Intracellular Signaling Peptides and Proteins , Neurodegenerative Diseases , Optic Nerve Diseases , Retinal Ganglion Cells , Animals , Disease Models, Animal , Dysautonomia, Familial/pathology , Humans , Mice , Neurodegenerative Diseases/pathology , Optic Nerve Diseases/pathology , Retinal Ganglion Cells/pathology
8.
J Genet Genomics ; 49(7): 654-665, 2022 07.
Article in English | MEDLINE | ID: mdl-34896608

ABSTRACT

Familial dysautonomia (FD), a hereditary sensory and autonomic neuropathy, is caused by a mutation in the Elongator complex protein 1 (ELP1) gene that leads to a tissue-specific reduction of ELP1 protein. Our work to generate a phenotypic mouse model for FD headed to the discovery that homozygous deletion of the mouse Elp1 gene leads to embryonic lethality prior to mid-gestation. Given that FD is caused by a reduction, not loss, of ELP1, we generated two new mouse models by introducing different copy numbers of the human FD ELP1 transgene into the Elp1 knockout mouse (Elp1-/-) and observed that human ELP1 expression rescues embryonic development in a dose-dependent manner. We then conducted a comprehensive transcriptome analysis in mouse embryos to identify genes and pathways whose expression correlates with the amount of ELP1. We found that ELP1 is essential for the expression of genes responsible for nervous system development. Further, gene length analysis of the differentially expressed genes showed that the loss of Elp1 mainly impacts the expression of long genes and that by gradually restoring Elongator, their expression is progressively rescued. Finally, through evaluation of co-expression modules, we identified gene sets with unique expression patterns that depended on ELP1 expression.


Subject(s)
Carrier Proteins , Dysautonomia, Familial , Animals , Carrier Proteins/genetics , Disease Models, Animal , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Gene Expression , Homozygote , Humans , Mice , Sequence Deletion
9.
Nat Commun ; 12(1): 3332, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099697

ABSTRACT

Pre-mRNA splicing is a key controller of human gene expression. Disturbances in splicing due to mutation lead to dysregulated protein expression and contribute to a substantial fraction of human disease. Several classes of splicing modulator compounds (SMCs) have been recently identified and establish that pre-mRNA splicing represents a target for therapy. We describe herein the identification of BPN-15477, a SMC that restores correct splicing of ELP1 exon 20. Using transcriptome sequencing from treated fibroblast cells and a machine learning approach, we identify BPN-15477 responsive sequence signatures. We then leverage this model to discover 155 human disease genes harboring ClinVar mutations predicted to alter pre-mRNA splicing as targets for BPN-15477. Splicing assays confirm successful correction of splicing defects caused by mutations in CFTR, LIPA, MLH1 and MAPT. Subsequent validations in two disease-relevant cellular models demonstrate that BPN-15477 increases functional protein, confirming the clinical potential of our predictions.


Subject(s)
Deep Learning , Gene Targeting/methods , RNA Splicing , Animals , Computational Biology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Exons , HEK293 Cells , Humans , Mice , Mice, Transgenic , MutL Protein Homolog 1/genetics , Mutation , Phenethylamines/administration & dosage , Pyridazines/administration & dosage , Sterol Esterase/genetics , Transcriptome , tau Proteins/genetics
10.
Hum Mol Genet ; 30(10): 908-922, 2021 05 29.
Article in English | MEDLINE | ID: mdl-33822942

ABSTRACT

Mucolipidosis IV (MLIV) is an orphan disease leading to debilitating psychomotor deficits and vision loss. It is caused by loss-of-function mutations in the MCOLN1 gene that encodes the lysosomal transient receptor potential channel mucolipin1, or TRPML1. With no existing therapy, the unmet need in this disease is very high. Here, we showed that AAV-mediated CNS-targeted gene transfer of the human MCOLN1 gene rescued motor function and alleviated brain pathology in the MLIV mouse model. Using the AAV-PHP.b vector in symptomatic mice, we showed long-term reversal of declined motor function and significant delay of paralysis. Next, using self-complementary AAV9 clinical candidate vector, we showed that its intracerebroventricular administration in post-natal day 1 mice significantly improved motor function, myelination and reduced lysosomal storage load in the MLIV mouse brain. Based on our data and general advancements in the gene therapy field, we propose scAAV9-mediated CSF-targeted MCOLN1 gene transfer as a therapeutic strategy in MLIV.


Subject(s)
Genetic Therapy , Mucolipidoses/therapy , Nervous System Diseases/therapy , Transient Receptor Potential Channels/genetics , Animals , Brain/metabolism , Brain/pathology , Dependovirus/genetics , Disease Models, Animal , Humans , Loss of Function Mutation/genetics , Lysosomes/genetics , Lysosomes/pathology , Mice , Mucolipidoses/cerebrospinal fluid , Mucolipidoses/genetics , Mucolipidoses/pathology , Nervous System Diseases/cerebrospinal fluid , Nervous System Diseases/genetics , Nervous System Diseases/pathology
11.
Am J Hum Genet ; 104(4): 638-650, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905397

ABSTRACT

Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a splice mutation in Elongator complex protein 1 (ELP1, also known as IKBKAP); this mutation leads to variable skipping of exon 20 and to a drastic reduction of ELP1 in the nervous system. Clinically, many of the debilitating aspects of the disease are related to a progressive loss of proprioception; this loss leads to severe gait ataxia, spinal deformities, and respiratory insufficiency due to neuromuscular incoordination. There is currently no effective treatment for FD, and the disease is ultimately fatal. The development of a drug that targets the underlying molecular defect provides hope that the drastic peripheral neurodegeneration characteristic of FD can be halted. We demonstrate herein that the FD mouse TgFD9;IkbkapΔ20/flox recapitulates the proprioceptive impairment observed in individuals with FD, and we provide the in vivo evidence that postnatal correction, promoted by the small molecule kinetin, of the mutant ELP1 splicing can rescue neurological phenotypes in FD. Daily administration of kinetin starting at birth improves sensory-motor coordination and prevents the onset of spinal abnormalities by stopping the loss of proprioceptive neurons. These phenotypic improvements correlate with increased amounts of full-length ELP1 mRNA and protein in multiple tissues, including in the peripheral nervous system (PNS). Our results show that postnatal correction of the underlying ELP1 splicing defect can rescue devastating disease phenotypes and is therefore a viable therapeutic approach for persons with FD.


Subject(s)
Dysautonomia, Familial/therapy , Kinetin/therapeutic use , Proprioception , RNA Splicing , Transcriptional Elongation Factors/genetics , Alleles , Animals , Behavior, Animal , Cell Line , Crosses, Genetic , Disease Models, Animal , Dysautonomia, Familial/genetics , Exons , Fibroblasts , Genotype , Humans , Introns , Kinetin/genetics , Male , Mice , Mice, Inbred C57BL , Mutation , Neurons/metabolism , Phenotype
12.
SLAS Discov ; 24(1): 57-67, 2019 01.
Article in English | MEDLINE | ID: mdl-30085848

ABSTRACT

Familial dysautonomia (FD) is an autonomic and sensory neuropathy caused by a mutation in the splice donor site of intron 20 of the ELP1 gene. Variable skipping of exon 20 leads to a tissue-specific reduction in the level of ELP1 protein. We have shown that the plant cytokinin kinetin is able to increase cellular ELP1 protein levels in vivo and in vitro through correction of ELP1 splicing. Studies in FD patients determined that kinetin is not a practical therapy due to low potency and rapid elimination. To identify molecules with improved potency and efficacy, we developed a cell-based luciferase splicing assay by inserting renilla (Rluc) and firefly (Fluc) luciferase reporters into our previously well-characterized ELP1 minigene construct. Evaluation of the Fluc/Rluc signal ratio enables a fast and accurate way to measure exon 20 inclusion. Further, we developed a secondary assay that measures ELP1 splicing in FD patient-derived fibroblasts. Here we demonstrate the quality and reproducibility of our screening method. Development and implementation of this screening platform has allowed us to efficiently screen for new compounds that robustly and specifically enhance ELP1 pre-mRNA splicing.


Subject(s)
Drug Evaluation, Preclinical/methods , Dysautonomia, Familial/genetics , RNA Precursors/genetics , RNA Splicing/drug effects , RNA, Messenger/genetics , Small Molecule Libraries/pharmacology , Transcriptional Elongation Factors/genetics , Cell Line , Cytokinins/pharmacology , Exons/drug effects , Exons/genetics , HEK293 Cells , Humans , Kinetin/pharmacology , RNA Splicing/genetics
13.
Hum Mol Genet ; 25(6): 1116-28, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26769677

ABSTRACT

Familial dysautonomia (FD) is an autosomal recessive neurodegenerative disease that affects the development and survival of sensory and autonomic neurons. FD is caused by an mRNA splicing mutation in intron 20 of the IKBKAP gene that results in a tissue-specific skipping of exon 20 and a corresponding reduction of the inhibitor of kappaB kinase complex-associated protein (IKAP), also known as Elongator complex protein 1. To date, several promising therapeutic candidates for FD have been identified that target the underlying mRNA splicing defect, and increase functional IKAP protein. Despite these remarkable advances in drug discovery for FD, we lacked a phenotypic mouse model in which we could manipulate IKBKAP mRNA splicing to evaluate potential efficacy. We have, therefore, engineered a new mouse model that, for the first time, will permit to evaluate the phenotypic effects of splicing modulators and provide a crucial platform for preclinical testing of new therapies. This new mouse model, TgFD9; Ikbkap(Δ20/flox) was created by introducing the complete human IKBKAP transgene with the major FD splice mutation (TgFD9) into a mouse that expresses extremely low levels of endogenous Ikbkap (Ikbkap(Δ20/flox)). The TgFD9; Ikbkap(Δ20/flox) mouse recapitulates many phenotypic features of the human disease, including reduced growth rate, reduced number of fungiform papillae, spinal abnormalities, and sensory and sympathetic impairments, and recreates the same tissue-specific mis-splicing defect seen in FD patients. This is the first mouse model that can be used to evaluate in vivo the therapeutic effect of increasing IKAP levels by correcting the underlying FD splicing defect.


Subject(s)
Disease Models, Animal , Dysautonomia, Familial/metabolism , Dysautonomia, Familial/pathology , Alternative Splicing , Animals , Autonomic Pathways/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dysautonomia, Familial/genetics , Exons , Humans , Intracellular Signaling Peptides and Proteins , Introns , Male , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , RNA Splicing/genetics , RNA, Messenger/metabolism , Sensory Receptor Cells/metabolism
14.
Nature ; 525(7567): 109-13, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26258302

ABSTRACT

Mitral valve prolapse (MVP) is a common cardiac valve disease that affects nearly 1 in 40 individuals. It can manifest as mitral regurgitation and is the leading indication for mitral valve surgery. Despite a clear heritable component, the genetic aetiology leading to non-syndromic MVP has remained elusive. Four affected individuals from a large multigenerational family segregating non-syndromic MVP underwent capture sequencing of the linked interval on chromosome 11. We report a missense mutation in the DCHS1 gene, the human homologue of the Drosophila cell polarity gene dachsous (ds), that segregates with MVP in the family. Morpholino knockdown of the zebrafish homologue dachsous1b resulted in a cardiac atrioventricular canal defect that could be rescued by wild-type human DCHS1, but not by DCHS1 messenger RNA with the familial mutation. Further genetic studies identified two additional families in which a second deleterious DCHS1 mutation segregates with MVP. Both DCHS1 mutations reduce protein stability as demonstrated in zebrafish, cultured cells and, notably, in mitral valve interstitial cells (MVICs) obtained during mitral valve repair surgery of a proband. Dchs1(+/-) mice had prolapse of thickened mitral leaflets, which could be traced back to developmental errors in valve morphogenesis. DCHS1 deficiency in MVP patient MVICs, as well as in Dchs1(+/-) mouse MVICs, result in altered migration and cellular patterning, supporting these processes as aetiological underpinnings for the disease. Understanding the role of DCHS1 in mitral valve development and MVP pathogenesis holds potential for therapeutic insights for this very common disease.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Mitral Valve Prolapse/genetics , Mitral Valve Prolapse/pathology , Mutation/genetics , Animals , Body Patterning/genetics , Cadherin Related Proteins , Cadherins/deficiency , Cell Movement/genetics , Chromosomes, Human, Pair 11/genetics , Female , Humans , Male , Mice , Mitral Valve/abnormalities , Mitral Valve/embryology , Mitral Valve/pathology , Mitral Valve/surgery , Pedigree , Phenotype , Protein Stability , RNA, Messenger/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
15.
Hum Mol Genet ; 19(16): 3114-23, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20504995

ABSTRACT

Rett syndrome is a severe neurodevelopmental disorder mainly caused by mutations in the transcriptional regulator MeCP2. Although there is no effective therapy for Rett syndrome, the recently discovered disease reversibility in mice suggests that there are therapeutic possibilities. Identification of MeCP2 targets or modifiers of the phenotype can facilitate the design of curative strategies. To identify possible novel MeCP2 interactors, we exploited a bioinformatic approach and selected Ying Yang 1 (YY1) as an interesting candidate. We demonstrate that MeCP2 interacts in vitro and in vivo with YY1, a ubiquitous zinc-finger epigenetic factor regulating the expression of several genes. We show that MeCP2 cooperates with YY1 in repressing the ANT1 gene encoding a mitochondrial adenine nucleotide translocase. Importantly, ANT1 mRNA levels are increased in human and mouse cell lines devoid of MeCP2, in Rett patient fibroblasts and in the brain of Mecp2-null mice. We further demonstrate that ANT1 protein levels are upregulated in Mecp2-null mice. Finally, the identified MeCP2-YY1 interaction, together with the well-known involvement of YY1 in the regulation of D4Z4-associated genes at 4q35, led us to discover the anomalous depression of FRG2, a subtelomeric gene of unknown function, in Rett fibroblasts. Collectively, our data indicate that mutations in MeCP2 might cause the aberrant overexpression of genes located at a specific locus, thus providing new candidates for the pathogenesis of Rett syndrome. As both ANT1 mutations and overexpression have been associated with human diseases, we consider it highly relevant to address the consequences of ANT1 deregulation in Rett syndrome.


Subject(s)
Adenine Nucleotide Translocator 1/metabolism , Chromosomes, Human, Pair 4/genetics , Methyl-CpG-Binding Protein 2/metabolism , YY1 Transcription Factor/metabolism , Adenine Nucleotide Translocator 1/genetics , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cells, Cultured , Chromatin Immunoprecipitation , Female , Fibroblasts/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Rett Syndrome/genetics , Rett Syndrome/metabolism , Reverse Transcriptase Polymerase Chain Reaction , YY1 Transcription Factor/genetics
16.
J Neurochem ; 108(3): 821-34, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19187099

ABSTRACT

Neurotransmitters are considered part of the signaling system active in nervous system development and we have previously reported that acetylcholine (ACh) is capable of enhancing neuronal differentiation in cultures of sensory neurons and N18TG2 neuroblastoma cells. To study the mechanism of ACh action, in this study, we demonstrate the ability of choline acetyltransferase-transfected N18TG2 clones (e.g. 2/4 clone) to release ACh. Analysis of muscarinic receptors showed the presence of M1-M4 subtypes and the activation of both IP(3) and cAMP signal transduction pathways. Muscarinic receptor activation increases early growth response factor-1 (EGR-1) levels and treatments with agonists, antagonists, and signal transduction enzyme inhibitors suggest a role for M3 subtype in EGR-1 induction. The role of EGR-1 in the enhancement of differentiation was investigated transfecting in N18TG2 cells a construct for EGR-1. EGR-1 clones show increased neurite extension and a decrease in Repressor Element-1 silencing transcription factor (REST) expression: both these features have also been observed for the 2/4 clone. Transfection of this latter with EGR zinc-finger domain, a dominant negative inhibitor of EGR-1 action, increases REST expression, and decreases fiber outgrowth. The data reported suggest that progression of the clone 2/4 in the developmental program is dependent on ACh release and the ensuing activation of muscarinic receptors, which in turn modulate the level of EGR-1 and REST transcription factors.


Subject(s)
Acetylcholine/pharmacology , Brain Neoplasms/metabolism , Cell Differentiation/drug effects , Early Growth Response Protein 1/biosynthesis , Muscarinic Agonists , Neuroblastoma/metabolism , Neurons/drug effects , Receptors, Muscarinic/drug effects , Repressor Proteins/biosynthesis , Acetylcholine/metabolism , Acetylcholine/physiology , Binding, Competitive/drug effects , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Size , Cyclic AMP/metabolism , Early Growth Response Protein 1/genetics , Humans , Neuroblastoma/pathology , Protein Kinase Inhibitors/pharmacology , Quinuclidinyl Benzilate/metabolism , Repressor Proteins/genetics , Signal Transduction/drug effects , Transfection
17.
Biochem Biophys Res Commun ; 362(2): 269-74, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17707768

ABSTRACT

The matrix metalloproteinases (MMPs), responsible for the degradation of extracellular matrix (ECM) proteins, may regulate brain cellular functions. Choline acetyltransferase (ChAT) transfected murine neuroblastoma cell line N18TG2, that synthesize acetylcholine and show enhancement of several neurospecific markers (i.e., sinapsin I, voltage gated Na(+) channels, high affinity choline uptake) and fiber outgrowth, were studied for the MMP regulation during neuronal differentiation. Zymography of N18TG2 culture medium revealed no gelatinolytic activity, whereas after carbachol treatment of cells both MMP-9 and activated MMP-2 forms were detected. ChAT-transfected clone culture medium contains three MMP forms at 230, 92, and 66kDa. Carbachol treatment increased MMP-2 and MMP-9 gene expression in N18TG2 cells and higher levels for both genes were also observed in ChAT transfected cells. The data are consistent with the hypothesis that acetylcholine brings about the activation of an autocrine loop modulating MMP expression.


Subject(s)
Acetylcholine/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neurites/drug effects , Animals , Atropine/pharmacology , Blotting, Western , Carbachol/pharmacology , Cell Line, Tumor , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , Molecular Weight , Neurites/enzymology , Neurites/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
18.
J Mol Neurosci ; 30(1-2): 75-6, 2006.
Article in English | MEDLINE | ID: mdl-17192635

ABSTRACT

The gene of mammalian acetylcholinesterase (AChE) generates multiple molecular forms, by alternative splicing of its transcripts and association of the tailed variant (AChET) with structural proteins. In the mammalian brain, the major AChE species consists of AChET tetramers anchored to the cell membrane of neurons by the PRiMA protein (Perrier et al., 2002). Stress and anticholinesterase inhibitors have been reported to induce rapid and long-lasting expression of the readthrough variant (AChER) in the mouse brain (Kaufer et al., 1998). In the readthrough transcript, there is no splicing after the last exon encoding the catalytic domain, so that the entire alternatively spliced 3' region is maintained. It encodes a C-terminal peptide with no specific interaction properties: COS cells transfected with AChER produce a soluble, nonamphiphilic monomeric form. We quantified AChER and total AChE expression in the mouse brain after an immobilization stress and after heat shock in neuroblastoma cells, and compared the observed effects with those induced by irreversible AChE inhibition (Perrier et al., 2005).


Subject(s)
Acetylcholinesterase/genetics , Brain/enzymology , Cholinesterase Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Stress, Psychological/enzymology , Animals , Cell Line, Tumor , Male , Mice , Neuroblastoma , RNA, Messenger/genetics , Restraint, Physical , Reverse Transcriptase Polymerase Chain Reaction
19.
J Neurochem ; 94(3): 629-38, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16001972

ABSTRACT

Acetylcholinesterase (AChE) exists in various molecular forms, depending on alternative splicing of its transcripts and association with structural proteins. Tetramers of the 'tailed' variant (AChE(T)), which are anchored in the cell membrane of neurons by the PRiMA (Proline Rich Membrane Anchor) protein, constitute the main form of AChE in the mammalian brain. In the mouse brain, stress and anticholinesterase inhibitors have been reported to induce expression of the unspliced 'readthrough' variant (AChE(R)) mRNA which produces a monomeric form. To generalize this observation, we attempted to quantify AChE(R) and AChE(T) after organophosphate intoxication in the mouse brain and compared the observed effects with those of stress induced by swimming or immobilization; we also analyzed the effects of heat shock and AChE inhibition on neuroblastoma cells. Active AChE molecular forms were characterized by sedimentation and non-denaturing electrophoresis, and AChE transcripts were quantified by real-time PCR. We observed a moderate increase of the AChE(R) transcript in some cases, both in the mouse brain and in neuroblastoma cultures, but we did not detect any increase of the corresponding active enzyme.


Subject(s)
Acetylcholinesterase/metabolism , Alternative Splicing/drug effects , Cholinesterase Inhibitors/pharmacology , Hot Temperature , Soman/pharmacology , Stress, Physiological/enzymology , Acetylcholinesterase/chemistry , Acetylcholinesterase/genetics , Acetylcholinesterase/pharmacology , Alternative Splicing/physiology , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Deoxycholic Acid/pharmacology , Detergents/pharmacology , Drug Interactions , Male , Mice , Mice, Inbred BALB C , Neuroblastoma , Octoxynol/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/biosynthesis , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...