Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(19): e2311685121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38683994

ABSTRACT

Neural crest cells exemplify cellular diversification from a multipotent progenitor population. However, the full sequence of early molecular choices orchestrating the emergence of neural crest heterogeneity from the embryonic ectoderm remains elusive. Gene-regulatory-networks (GRN) govern early development and cell specification toward definitive neural crest. Here, we combine ultradense single-cell transcriptomes with machine-learning and large-scale transcriptomic and epigenomic experimental validation of selected trajectories, to provide the general principles and highlight specific features of the GRN underlying neural crest fate diversification from induction to early migration stages using Xenopus frog embryos as a model. During gastrulation, a transient neural border zone state precedes the choice between neural crest and placodes which includes multiple converging gene programs. During neurulation, transcription factor connectome, and bifurcation analyses demonstrate the early emergence of neural crest fates at the neural plate stage, alongside an unbiased multipotent-like lineage persisting until epithelial-mesenchymal transition stage. We also decipher circuits driving cranial and vagal neural crest formation and provide a broadly applicable high-throughput validation strategy for investigating single-cell transcriptomes in vertebrate GRNs in development, evolution, and disease.


Subject(s)
Neural Crest , Single-Cell Analysis , Xenopus laevis , Animals , Neural Crest/cytology , Neural Crest/metabolism , Single-Cell Analysis/methods , Xenopus laevis/embryology , Gene Expression Regulation, Developmental , Cell Movement , Gene Regulatory Networks , Transcriptome , Gastrulation , Neural Plate/metabolism , Neural Plate/embryology , Neural Plate/cytology , Epithelial-Mesenchymal Transition/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/cytology , Neurulation/genetics , Neurulation/physiology , Cell Differentiation
2.
Life Sci Alliance ; 5(12)2022 08 01.
Article in English | MEDLINE | ID: mdl-35914811

ABSTRACT

Cell migration is a complex process, tightly regulated during embryonic development and abnormally activated during cancer metastasis. RAS-dependent signaling is a major nexus controlling essential cell parameters including proliferation, survival, and migration, utilizing downstream effectors such as the PI3K/AKT signaling pathway. In melanoma, oncogenic mutations frequently enhance RAS, PI3K/AKT, or MAP kinase signaling and trigger other cancer hallmarks among which the activation of metabolism regulators. PFKFB4 is one of these critical regulators of glycolysis and of the Warburg effect. Here, however, we explore a novel function of PFKFB4 in melanoma cell migration. We find that PFKFB4 interacts with ICMT, a posttranslational modifier of RAS. PFKFB4 promotes ICMT/RAS interaction, controls RAS localization at the plasma membrane, activates AKT signaling and enhances cell migration. We thus provide evidence of a novel and glycolysis-independent function of PFKFB4 in human cancer cells. This unconventional activity links the metabolic regulator PFKFB4 to RAS-AKT signaling and impacts melanoma cell migration.


Subject(s)
Melanoma , Proto-Oncogene Proteins c-akt , Cell Line, Tumor , Cell Movement/physiology , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Protein Methyltransferases , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
3.
Mol Psychiatry ; 27(2): 918-928, 2022 02.
Article in English | MEDLINE | ID: mdl-34785784

ABSTRACT

The persistent and experience-dependent nature of drug addiction may result in part from epigenetic alterations, including non-coding micro-RNAs (miRNAs), which are both critical for neuronal function and modulated by cocaine in the striatum. Two major striatal cell populations, the striato-nigral and striato-pallidal projection neurons, express, respectively, the D1 (D1-SPNs) and D2 (D2-SPNs) dopamine receptor, and display distinct but complementary functions in drug-evoked responses. However, a cell-type-specific role for miRNAs action has yet to be clarified. Here, we evaluated the expression of a subset of miRNAs proposed to modulate cocaine effects in the nucleus accumbens (NAc) and dorsal striatum (DS) upon sustained cocaine exposure in mice and showed that these selected miRNAs were preferentially upregulated in the NAc. We focused on miR-1 considering the important role of some of its predicted mRNA targets, Fosb and Npas4, in the effects of cocaine. We validated these targets in vitro and in vivo. We explored the potential of miR-1 to regulate cocaine-induced behavior by overexpressing it in specific striatal cell populations. In DS D1-SPNs miR-1 overexpression downregulated Fosb and Npas4 and reduced cocaine-induced CPP reinstatement, but increased cue-induced cocaine seeking. In DS D2-SPNs miR-1 overexpression reduced the motivation to self-administer cocaine. Our results indicate a role of miR1 and its target genes, Fosb and Npas4, in these behaviors and highlight a precise cell-type- and region-specific modulatory role of miR-1, illustrating the importance of cell-specific investigations.


Subject(s)
Cocaine , MicroRNAs , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cocaine/metabolism , Cocaine/pharmacology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/metabolism , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Self Administration
4.
Sci Adv ; 7(43): eabg5970, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34669474

ABSTRACT

Addictive drugs increase dopamine in the nucleus accumbens (NAc), where it persistently shapes excitatory glutamate transmission and hijacks natural reward processing. Here, we provide evidence, from mice to humans, that an underlying mechanism relies on drug-evoked heteromerization of glutamate N-methyl-d-aspartate receptors (NMDAR) with dopamine receptor 1 (D1R) or 2 (D2R). Using temporally controlled inhibition of D1R-NMDAR heteromerization, we unraveled their selective implication in early phases of cocaine-mediated synaptic, morphological, and behavioral responses. In contrast, preventing D2R-NMDAR heteromerization blocked the persistence of these adaptations. Interfering with these heteromers spared natural reward processing. Notably, we established that D2R-NMDAR complexes exist in human samples and showed that, despite a decreased D2R protein expression in the NAc, individuals with psychostimulant use disorder display a higher proportion of D2R forming heteromers with NMDAR. These findings contribute to a better understanding of molecular mechanisms underlying addiction and uncover D2R-NMDAR heteromers as targets with potential therapeutic value.

5.
Proc Natl Acad Sci U S A ; 116(49): 24840-24851, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31744868

ABSTRACT

Huntington's disease (HD) is a chronic neurodegenerative disorder characterized by a late clinical onset despite ubiquitous expression of the mutant Huntingtin gene (HTT) from birth. Transcriptional dysregulation is a pivotal feature of HD. Yet, the genes that are altered in the prodromal period and their regulators, which present opportunities for therapeutic intervention, remain to be elucidated. Using transcriptional and chromatin profiling, we found aberrant transcription and changes in histone H3K27acetylation in the striatum of R6/1 mice during the presymptomatic disease stages. Integrating these data, we identified the Elk-1 transcription factor as a candidate regulator of prodromal changes in HD. Exogenous expression of Elk-1 exerted beneficial effects in a primary striatal cell culture model of HD, and adeno-associated virus-mediated Elk-1 overexpression alleviated transcriptional dysregulation in R6/1 mice. Collectively, our work demonstrates that aberrant gene expression precedes overt disease onset in HD, identifies the Elk-1 transcription factor as a key regulator linked to early epigenetic and transcriptional changes in HD, and presents evidence for Elk-1 as a target for alleviating molecular pathology in HD.


Subject(s)
Epigenomics , Huntington Disease/genetics , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism , Animals , Corpus Striatum/metabolism , Dependovirus , Disease Models, Animal , Histones/metabolism , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Mice , Mice, Transgenic , Neurons/metabolism , Nuclear Proteins/metabolism
6.
Brain ; 142(8): 2432-2450, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31286142

ABSTRACT

Dysfunctions in brain cholesterol homeostasis have been extensively related to brain disorders. The main pathway for brain cholesterol elimination is its hydroxylation into 24S-hydroxycholesterol by the cholesterol 24-hydrolase, CYP46A1. Increasing evidence suggests that CYP46A1 has a role in the pathogenesis and progression of neurodegenerative disorders, and that increasing its levels in the brain is neuroprotective. However, the mechanisms underlying this neuroprotection remain to be fully understood. Huntington's disease is a fatal autosomal dominant neurodegenerative disease caused by an abnormal CAG expansion in huntingtin's gene. Among the multiple cellular and molecular dysfunctions caused by this mutation, altered brain cholesterol homeostasis has been described in patients and animal models as a critical event in Huntington's disease. Here, we demonstrate that a gene therapy approach based on the delivery of CYP46A1, the rate-limiting enzyme for cholesterol degradation in the brain, has a long-lasting neuroprotective effect in Huntington's disease and counteracts multiple detrimental effects of the mutated huntingtin. In zQ175 Huntington's disease knock-in mice, CYP46A1 prevented neuronal dysfunctions and restored cholesterol homeostasis. These events were associated to a specific striatal transcriptomic signature that compensates for multiple mHTT-induced dysfunctions. We thus explored the mechanisms for these compensations and showed an improvement of synaptic activity and connectivity along with the stimulation of the proteasome and autophagy machineries, which participate to the clearance of mutant huntingtin (mHTT) aggregates. Furthermore, BDNF vesicle axonal transport and TrkB endosome trafficking were restored in a cellular model of Huntington's disease. These results highlight the large-scale beneficial effect of restoring cholesterol homeostasis in neurodegenerative diseases and give new opportunities for developing innovative disease-modifying strategies in Huntington's disease.


Subject(s)
Brain/metabolism , Cholesterol 24-Hydroxylase/therapeutic use , Cholesterol/metabolism , Genetic Therapy , Genetic Vectors/therapeutic use , Huntington Disease/therapy , Neuroprotective Agents/therapeutic use , Animals , Autophagy , Axonal Transport , Brain-Derived Neurotrophic Factor/physiology , Cells, Cultured , Cerebral Cortex/physiopathology , Cholesterol 24-Hydroxylase/genetics , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Dependovirus/genetics , Endosomes/metabolism , Gene Knock-In Techniques , Genetic Vectors/genetics , Humans , Huntingtin Protein/genetics , Huntington Disease/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/physiopathology , Neuroprotective Agents/administration & dosage , Oxysterols/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Aggregation, Pathological , Protein-Tyrosine Kinases/physiology , Rotarod Performance Test , Synaptic Transmission , Transcriptome
7.
Biol Psychiatry ; 81(7): 573-584, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27567310

ABSTRACT

BACKGROUND: Addiction relies on persistent alterations of neuronal properties, which depends on gene regulation. Activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene that modulates neuronal plasticity underlying learning and memory. Its role in cocaine-induced neuronal and behavioral adaptations remains elusive. METHODS: Acute cocaine-treated mice were used for quantitative reverse-transcriptase polymerase chain reaction, immunocytochemistry, and confocal imaging from striatum. Live imaging and transfection assays for Arc overexpression were performed from primary cultures. Molecular and behavioral adaptations to cocaine were studied from Arc-deficient mice and their wild-type littermates. RESULTS: Arc messenger RNA and proteins are rapidly induced in the striatum after acute cocaine administration, via an extracellular-signal regulated kinase-dependent de novo protein synthesis. Although detected in dendrites, Arc accumulates in the nucleus in active zones of transcription, where it colocalizes with phospho-Ser10-histone H3, an important component of nucleosomal response. In vitro, Arc overexpression downregulates phospho-Ser10-histone H3 without modifying extracellular-signal regulated kinase phosphorylation in the nucleus. In vivo, Arc-deficient mice display decreased heterochromatin domains, a high RNA-polymerase II activity and enhanced c-Fos expression. These mice presented an exacerbated psychomotor sensitization and conditioned place preference induced by low doses of cocaine. CONCLUSIONS: Cocaine induces the rapid induction of Arc and its nuclear accumulation in striatal neurons. Locally, it alters the nucleosomal response, and acts as a brake on chromatin remodeling and gene regulation. These original observations posit Arc as a major homeostatic modulator of molecular and behavioral responses to cocaine. Thus, modulating Arc levels may provide promising therapeutic approaches in drug addiction.


Subject(s)
Behavior, Animal/drug effects , Chromatin Assembly and Disassembly , Cocaine/administration & dosage , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Conditioning, Classical/drug effects , Histones/metabolism , Locomotion/drug effects , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , RNA, Messenger
8.
J Neurosci ; 31(40): 14296-307, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21976515

ABSTRACT

Activation of the extracellular signal-regulated kinase (ERK) signaling pathway in the striatum is crucial for molecular adaptations and long-term behavioral alterations induced by cocaine. In response to cocaine, ERK controls the phosphorylation levels of both mitogen and stress-activated protein kinase 1 (MSK-1), a nuclear kinase involved in histone H3 (Ser10) and cAMP response element binding protein phosphorylation, and Elk-1, a transcription factor involved in serum response element (SRE)-driven gene regulations. We recently characterized the phenotype of msk-1 knock-out mice in response to cocaine. Herein, we wanted to address the role of Elk-1 phosphorylation in cocaine-induced molecular, morphological, and behavioral responses. We used a cell-penetrating peptide, named TAT-DEF-Elk-1 (TDE), which corresponds to the DEF docking domain of Elk-1 toward ERK and inhibits Elk-1 phosphorylation induced by ERKs without modifying ERK or MSK-1 in vitro. The peptide was injected in vivo before cocaine administration in mice. Immunocytochemical, molecular, morphological, and behavioral studies were performed. The TDE inhibited Elk-1 and H3 (Ser10) phosphorylation induced by cocaine, sparing ERK and MSK-1 activation. Consequently, TDE altered cocaine-induced regulation of genes bearing SRE site(s) in their promoters, including c-fos, zif268, ΔFosB, and arc/arg3.1 (activity-regulated cytoskeleton-associated protein). In a chronic cocaine administration paradigm, TDE reversed cocaine-induced increase in dendritic spine density. Finally, the TDE delayed the establishment of cocaine-induced psychomotor sensitization and conditioned-place preference. We conclude that Elk-1 phosphorylation downstream from ERK is a key molecular event involved in long-term neuronal and behavioral adaptations to cocaine.


Subject(s)
Cocaine/pharmacology , Neural Inhibition/physiology , ets-Domain Protein Elk-1/antagonists & inhibitors , ets-Domain Protein Elk-1/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neural Inhibition/drug effects , Peptides/metabolism , Peptides/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology
9.
Mol Cell Neurosci ; 41(3): 325-36, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398002

ABSTRACT

Nuclear translocation of activated extracellular signal-regulated kinases (ERK) in neurons is critical for gene regulations underlying long-term neuronal adaptation and memory formation. However, it is unknown how activated ERK travel from the post-synaptic elements where their activation occurs, to the nucleus where they translocate to exert their transcriptional roles. In cultured neurons, we identified endocytosis as a prime event in glutamate-induced nuclear trafficking of ERK2. We show that glutamate triggers a rapid recruitment of ERK2 to a protein complex comprising markers of the clathrin-dependent endocytotic and AMPA/glutamate receptor subtype. Inhibition of endocytosis results in a neuritic withholding of activated ERK2 without modification of ERK2 activity. As a consequence, endocytosis blockade alters ERK-dependent nuclear events, such as mitogen and stressed-activated kinase-1 (MSK-1) activation, histone H3 phosphorylation and gene regulations. Our data provide the first evidence that the endocytic pathway controls ERK nuclear translocation and ERK-dependent gene regulations induced by glutamate.


Subject(s)
Cell Nucleus/metabolism , Endocytosis/drug effects , Glutamic Acid/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Animals , Cell Culture Techniques , Cell Line , Cell Nucleus/drug effects , Chlorocebus aethiops , Chromatin Assembly and Disassembly , Enzyme Activation , Neurons/drug effects , Neurons/metabolism , Protein Transport/drug effects , Rats , Receptors, AMPA/metabolism , Receptors, Glutamate/metabolism , ets-Domain Protein Elk-1/metabolism
10.
J Neurosci ; 27(52): 14448-58, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18160653

ABSTRACT

The transcription factor Elk-1 plays a key role in cell differentiation, proliferation and apoptosis. This role is thought to arise from its phosphorylation by activated extracellular signal-regulated kinases (ERKs), a critical posttranslational event for the transcriptional activity of the ternary complex composed of Elk-1 and a dimer of serum response factor (SRF) at the serum response element (SRE) regulatory site of transcription. In addition to its nuclear localization, Elk-1 is found in the dendrites and soma of neuronal cells and recent evidence implicate a cytoplasmic proapoptotic function of Elk-1, via its association with the mitochondrial permeability transition pore complex. Thus, the nuclear versus cytoplasmic localization of Elk-1 seems to be crucial for its biological function. In this study we show that the excitatory neurotransmitter, glutamate, induces an ERK-dependent Elk-1 activation and nuclear relocalization. We demonstrate that Elk-1 phosphorylation on Ser383/389 has a dual function and triggers both Elk-1 nuclear translocation and SRE-dependent gene expression. Mutating these sites into inactive residues or using a synthetic penetrating peptide (TAT-DEF-Elk-1), which specifically interferes with the DEF docking domain of Elk-1, prevents Elk-1 nuclear translocation without interfering with ERK nor MSK1 (mitogen- and stress-activated protein kinase 1), a CREB kinase downstream from ERK- activation. This results in a differential regulation of glutamate-induced IEG regulation when compared with classical inhibitors of the ERK pathway. Using the TAT-DEF-Elk-1 peptide or the dominant-negative version of Elk-1, we show that Elk-1 phosphorylation controls dendritic elongation, SRF and Actin expression levels as well as cytoskeleton dynamics.


Subject(s)
Cytoskeleton/physiology , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Peptides/metabolism , ets-Domain Protein Elk-1/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cocaine/pharmacology , Corpus Striatum/cytology , Dopamine Uptake Inhibitors/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Glutamic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Peptides/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Serine/metabolism , Serine/pharmacology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Transfection/methods , Transfection/statistics & numerical data
11.
J Neurochem ; 86(5): 1233-46, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12911631

ABSTRACT

Neurosteroids are steroids produced within the nervous system. Based on behavioural responses evoked in animals by synthetic steroid injections, several studies suggested neurosteroid involvement in important neurophysiological processes. These observations should be correlated only to neuroactive effects of the injected steroids. Neurosteroids mostly control the CNS activity through allosteric modulation of neurotransmitter receptors within concentration ranges used by neurotransmitters themselves. Therefore, neurosteroid production within pathways controlling a neurophysiological process is necessary to consider neurosteroid involvement in that process. Because of the increasing speculation about pain modulation by neurosteroids based on pharmacological observations, we decided to clarify the situation by investigating neurosteroidogenesis occurrence in sensory pathways, particularly in nociceptive structures. We studied the presence and activity of cytochrome P450side chain cleavage (P450scc) in rat pain pathways. P450scc-immunoreactive cells were localized in dorsal root ganglia (DRG), spinal cord (SC) dorsal horn, nociceptive supraspinal nuclei (SSN) and somatosensory cortex. Incubation of DRG, SSN or SC tissue homogenates with [3H]cholesterol yielded the formation of radioactive metabolites including [3H]pregnenolone of which the synthesis was reduced in presence of aminogluthetimide, a P450scc inhibitor. These first neuroanatomical and neurochemical results demonstrate the occurrence of neurosteroidogenesis in nociceptive pathways and strongly suggest that neurosteroids may control pain mechanisms.


Subject(s)
Cholesterol Side-Chain Cleavage Enzyme/metabolism , Neural Pathways/enzymology , Neurons, Afferent/enzymology , Animals , Brain Stem/cytology , Brain Stem/enzymology , Cholesterol/metabolism , Cholesterol Side-Chain Cleavage Enzyme/analysis , Cholesterol Side-Chain Cleavage Enzyme/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/enzymology , Immunohistochemistry , Male , Neural Pathways/cytology , Neurons, Afferent/cytology , Organ Specificity , Pregnenolone/analysis , Pregnenolone/biosynthesis , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/cytology , Somatosensory Cortex/enzymology , Spinal Cord/cytology , Spinal Cord/enzymology , Thalamus/cytology , Thalamus/enzymology , Tritium
12.
J Am Soc Nephrol ; 11(4): 632-639, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10752522

ABSTRACT

The binding of Fc fragments of Ig on glomerular epithelial cells (GEC) was observed previously, but the receptor could not be identified. In immunofluorescence and immunohistochemical studies using normal adult human kidney sections, the presence of the so-called neonatal Fc receptor (FcRn) was demonstrated on GEC as well as in the brush border of proximal tubular cells. FcRn transcripts were also detected on isolated glomeruli by reverse transcription-PCR. Using an immortalized GEC line, the presence of the FcRn was confirmed by flow cytometry, reverse transcription-PCR, Western blotting, and by the pH dependence of the binding of heat-aggregated IgG. Because it is well established that the FcRn is involved in IgG transcytosis, it is hypothesized that the FcRn in the kidney may play a role in the reabsorption of IgG. Ongoing studies should clarify the role of the FcRn as a potential target for immune complexes on GEC and should assess its relevance in physiology and pathology.


Subject(s)
Aging/metabolism , Kidney/metabolism , Receptors, Fc/metabolism , Adult , Cell Line , Cell Line, Transformed , Humans , Infant, Newborn , Kidney/cytology , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Microvilli/metabolism , RNA, Messenger/metabolism , Receptors, Fc/genetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...