Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 883
Filtrer
1.
Methods Mol Biol ; 2831: 199-208, 2024.
Article de Anglais | MEDLINE | ID: mdl-39134851

RÉSUMÉ

The use of time-lapse live imaging enables us to track the dynamic changes in neurites during their formation. Ex vivo live imaging with acute brain slices provides a more physiological environment than cultured cells. To accomplish this, a certain method of labeling is necessary to visualize and identify neurite morphology. To understand the dynamics of neurite structure at early stages of neurite formation, we describe in this chapter ex vivo live imaging using a confocal microscope at P0 in combination with in utero electroporation (IUE).


Sujet(s)
Encéphale , Électroporation , Neurites , Animaux , Électroporation/méthodes , Neurites/métabolisme , Encéphale/cytologie , Encéphale/embryologie , Encéphale/imagerie diagnostique , Souris , Femelle , Microscopie confocale/méthodes , Imagerie accélérée/méthodes , Grossesse , Neurogenèse
2.
Methods Mol Biol ; 2831: 301-313, 2024.
Article de Anglais | MEDLINE | ID: mdl-39134858

RÉSUMÉ

Isolation and culture of dorsal root ganglion (DRG) neurons from adult animals is a useful experimental system for evaluating neural plasticity after axonal injury, as well as the neurological dysfunction resulting from aging and various types of disease. In this chapter, we will introduce a detailed method for the culture of mature rat DRG neurons. About 30-40 ganglia are dissected from a rat and mechanically and enzymatically digested. Subsequently, density gradient centrifugation of the digested tissue using 30% Percoll efficiently eliminates myelin debris and non-neuronal cells, to afford neuronal cells with a high yield and purity.


Sujet(s)
Techniques de culture cellulaire , Séparation cellulaire , Ganglions sensitifs des nerfs spinaux , Régénération nerveuse , Neurones , Animaux , Ganglions sensitifs des nerfs spinaux/cytologie , Rats , Neurones/cytologie , Neurones/physiologie , Techniques de culture cellulaire/méthodes , Régénération nerveuse/physiologie , Séparation cellulaire/méthodes , Dégénérescence nerveuse/anatomopathologie , Cellules cultivées , Centrifugation en gradient de densité/méthodes
3.
Front Neurosci ; 18: 1425525, 2024.
Article de Anglais | MEDLINE | ID: mdl-39027325

RÉSUMÉ

The S100B is a member of the S100 family of "E" helix-loop- "F" helix structure (EF) hand calcium-binding proteins expressed in diverse glial, selected neuronal, and various peripheral cells, exerting differential effects. In particular, this review compiles descriptions of the detection of S100B in different brain cells localized in specific regions during the development of humans, mice, and rats. Then, it summarizes S100B's actions on the differentiation, growth, and maturation of glial and neuronal cells in humans and rodents. Particular emphasis is placed on S100B regulation of the differentiation and maturation of astrocytes, oligodendrocytes (OL), and the stimulation of dendritic development in serotoninergic and cerebellar neurons during embryogenesis. We also summarized reports that associate morphological alterations (impaired neurite outgrowth, neuronal migration, altered radial glial cell morphology) of specific neural cell groups during neurodevelopment and functional disturbances (slower rate of weight gain, impaired spatial learning) with changes in the expression of S100B caused by different conditions and stimuli as exposure to stress, ethanol, cocaine and congenital conditions such as Down's Syndrome. Taken together, this evidence highlights the impact of the expression and early actions of S100B in astrocytes, OL, and neurons during brain development, which is reflected in the alterations in differentiation, growth, and maturation of these cells. This allows the integration of a spatiotemporal panorama of S100B actions in glial and neuronal cells in the developing brain.

4.
J Biol Chem ; 300(8): 107537, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38971314

RÉSUMÉ

Neurite outgrowth is a critical step in neural development, leading to the generation of neurite branches that allow individual neurons to make contacts with multiple neurons within the target region. Polyglutamine-binding protein 1 (PQBP1) is a highly conserved protein with a key role in neural development. Our recent mass spectrometric analysis showed that PQBP1 associates with neural Wiskott-Aldrich syndrome protein (N-WASP), an important actin polymerization-promoting factor involved in neurite outgrowth. Here, we report that the WW domain of PQBP1 directly interacts with the proline-rich domain of N-WASP. The disruption of this interaction leads to impaired neurite outgrowth and growth cone size. Furthermore, we demonstrate that PQBP1/N-WASP interaction is critical for the recruitment of N-WASP to the growth cone, but does not affect N-WASP protein levels or N-WASP-induced actin polymerization. Our results indicated that PQBP1 regulates neurite outgrowth by recruiting N-WASP to the growth cone, thus representing an alternative molecular mechanism via which PQBP1-mediates neurite outgrowth.

5.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891838

RÉSUMÉ

Nanoparticles (NPs) are becoming increasingly important novel materials for many purposes, including basic research, medicine, agriculture, and engineering. Increasing human and environmental exposure to these promising compounds requires assessment of their potential health risks. While the general direct cytotoxicity of NPs is often routinely measured, more indirect possible long-term effects, such as reproductive or developmental neurotoxicity (DNT), have been studied only occasionally and, if so, mostly on non-human animal models, such as zebrafish embryos. In this present study, we employed a well-characterized human neuronal precursor cell line to test the concentration-dependent DNT of green-manufactured copper sulfide (CuS) nanoparticles on crucial early events in human brain development. CuS NPs turned out to be generally cytotoxic in the low ppm range. Using an established prediction model, we found a clear DNT potential of CuS NPs on neuronal precursor cell migration and neurite outgrowth, with IC50 values 10 times and 5 times, respectively, lower for the specific DNT endpoint than for general cytotoxicity. We conclude that, in addition to the opportunities of NPs, their risks to human health should be carefully considered.


Sujet(s)
Cuivre , Nanoparticules métalliques , Neurones , Humains , Cuivre/toxicité , Nanoparticules métalliques/toxicité , Nanoparticules métalliques/composition chimique , Neurones/effets des médicaments et des substances chimiques , Sulfures/toxicité , Sulfures/composition chimique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/anatomopathologie , Nanoparticules/toxicité , Nanoparticules/composition chimique , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques
6.
Mol Cell ; 84(12): 2304-2319.e8, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38838666

RÉSUMÉ

Circular RNAs (circRNAs) are upregulated during neurogenesis. Where and how circRNAs are localized and what roles they play during this process have remained elusive. Comparing the nuclear and cytoplasmic circRNAs between H9 cells and H9-derived forebrain (FB) neurons, we identify that a subset of adenosine (A)-rich circRNAs are restricted in H9 nuclei but exported to cytosols upon differentiation. Such a subcellular relocation of circRNAs is modulated by the poly(A)-binding protein PABPC1. In the H9 nucleus, newly produced (A)-rich circRNAs are bound by PABPC1 and trapped by the nuclear basket protein TPR to prevent their export. Modulating (A)-rich motifs in circRNAs alters their subcellular localization, and introducing (A)-rich circRNAs in H9 cytosols results in mRNA translation suppression. Moreover, decreased nuclear PABPC1 upon neuronal differentiation enables the export of (A)-rich circRNAs, including circRTN4(2,3), which is required for neurite outgrowth. These findings uncover subcellular localization features of circRNAs, linking their processing and function during neurogenesis.


Sujet(s)
Transport nucléaire actif , Adénosine , Noyau de la cellule , Neurogenèse , Neurones , Protéine-1 de liaison au poly(A) , ARN circulaire , ARN , ARN circulaire/métabolisme , ARN circulaire/génétique , Neurones/métabolisme , Adénosine/métabolisme , Noyau de la cellule/métabolisme , Humains , Protéine-1 de liaison au poly(A)/métabolisme , Protéine-1 de liaison au poly(A)/génétique , Animaux , ARN/métabolisme , ARN/génétique , Lignée cellulaire , Différenciation cellulaire , Cytoplasme/métabolisme , Prosencéphale/métabolisme
7.
Biomolecules ; 14(6)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38927091

RÉSUMÉ

BACKGROUND: Induced pluripotent stem cell (iPSC) based neuronal differentiation is valuable for studying neuropsychiatric disorders and pharmacological mechanisms at the cellular level. We aimed to examine the effects of typical and atypical antipsychotics on human iPSC-derived neural progenitor cells (NPCs). METHODS: Proliferation and neurite outgrowth were measured by live cell imaging, and gene expression levels related to neuronal identity were analyzed by RT-QPCR and immunocytochemistry during differentiation into hippocampal dentate gyrus granule cells following treatment of low- and high-dose antipsychotics (haloperidol, olanzapine, and risperidone). RESULTS: Antipsychotics did not modify the growth properties of NPCs after 3 days of treatment. However, the characteristics of neurite outgrowth changed significantly in response to haloperidol and olanzapine. After three weeks of differentiation, mRNA expression levels of the selected neuronal markers increased (except for MAP2), while antipsychotics caused only subtle changes. Additionally, we found no changes in MAP2 or GFAP protein expression levels as a result of antipsychotic treatment. CONCLUSIONS: Altogether, antipsychotic medications promoted neurogenesis in vitro by influencing neurite outgrowth rather than changing cell survival or gene expression. This study provides insights into the effects of antipsychotics on neuronal differentiation and highlights the importance of considering neurite outgrowth as a potential target of action.


Sujet(s)
Neuroleptiques , Différenciation cellulaire , Halopéridol , Hippocampe , Cellules souches pluripotentes induites , Cellules souches neurales , Neurogenèse , Olanzapine , Rispéridone , Humains , Olanzapine/pharmacologie , Rispéridone/pharmacologie , Neurogenèse/effets des médicaments et des substances chimiques , Hippocampe/cytologie , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Halopéridol/pharmacologie , Neuroleptiques/pharmacologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Excroissance neuronale/effets des médicaments et des substances chimiques
8.
Cell Calcium ; 121: 102894, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38728789

RÉSUMÉ

TRPV2 voltage-insensitive, calcium-permeable ion channels play important roles in cancer progression, immune response, and neuronal development. Despite TRPV2's physiological impact, underlying endogenous proteins mediating TRPV2 responses and affected signaling pathways remain elusive. Using quantitative peroxidase-catalyzed (APEX2) proximity proteomics we uncover dynamic changes in the TRPV2-proximal proteome and identify calcium signaling and cell adhesion factors recruited to the molecular channel neighborhood in response to activation. Quantitative TRPV2 proximity proteomics further revealed activation-induced enrichment of protein clusters with biological functions in neural and cellular projection. We demonstrate a functional connection between TRPV2 and the neural immunoglobulin cell adhesion molecules NCAM and L1CAM. NCAM and L1CAM stimulation robustly induces TRPV2 [Ca2+]I flux in neuronal PC12 cells and this TRPV2-specific [Ca2+]I flux requires activation of the protein kinase PKCα. TRPV2 expression directly impacts neurite lengths that are modulated by NCAM or L1CAM stimulation. Hence, TRPV2's calcium signaling plays a previously undescribed, yet vital role in cell adhesion, and TRPV2 calcium flux and neurite development are intricately linked via NCAM and L1CAM cell adhesion proteins.


Sujet(s)
Calcium , Molécule d'adhérence cellulaire neurale L-1 , Molécules d'adhérence cellulaire neurales , Excroissance neuronale , Protéome , Canaux cationiques TRPV , Animaux , Humains , Rats , Calcium/métabolisme , Signalisation calcique , Adhérence cellulaire , Molécule d'adhérence cellulaire neurale L-1/métabolisme , Molécules d'adhérence cellulaire neurales/métabolisme , Neurites/métabolisme , Cellules PC12 , Protein kinase C-alpha/métabolisme , Protéome/métabolisme , Canaux cationiques TRPV/métabolisme , Antigènes CD56/métabolisme
9.
Traffic ; 25(5): e12936, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38725127

RÉSUMÉ

Endosomal trafficking of TrkA is a critical process for nerve growth factor (NGF)-dependent neuronal cell survival and differentiation. The small GTPase ADP-ribosylation factor 6 (Arf6) is implicated in NGF-dependent processes in PC12 cells through endosomal trafficking and actin cytoskeleton reorganization. However, the regulatory mechanism for Arf6 in NGF signaling is largely unknown. In this study, we demonstrated that EFA6A, an Arf6-specific guanine nucleotide exchange factor, was abundantly expressed in PC12 cells and that knockdown of EFA6A significantly inhibited NGF-dependent Arf6 activation, TrkA recycling from early endosomes to the cell surface, prolonged ERK1/2 phosphorylation, and neurite outgrowth. We also demonstrated that EFA6A forms a protein complex with TrkA through its N-terminal region, thereby enhancing its catalytic activity for Arf6. Similarly, we demonstrated that EFA6A forms a protein complex with TrkA in cultured dorsal root ganglion (DRG) neurons. Furthermore, cultured DRG neurons from EFA6A knockout mice exhibited disturbed NGF-dependent TrkA trafficking compared with wild-type neurons. These findings provide the first evidence for EFA6A as a key regulator of NGF-dependent TrkA trafficking and signaling.


Sujet(s)
Facteur-6 de ribosylation de l'ADP , Facteurs d'ADP-ribosylation , Endosomes , Facteurs d'échange de nucléotides guanyliques , Facteur de croissance nerveuse , Excroissance neuronale , Récepteur trkA , Animaux , Souris , Rats , Facteurs d'ADP-ribosylation/métabolisme , Facteurs d'ADP-ribosylation/génétique , Endosomes/métabolisme , Ganglions sensitifs des nerfs spinaux/métabolisme , Facteurs d'échange de nucléotides guanyliques/métabolisme , Facteurs d'échange de nucléotides guanyliques/génétique , Souris knockout , Facteur de croissance nerveuse/métabolisme , Cellules PC12 , Transport des protéines , Récepteur trkA/métabolisme
10.
Mol Neurobiol ; 2024 Apr 06.
Article de Anglais | MEDLINE | ID: mdl-38581538

RÉSUMÉ

Spinal cord injury (SCI) constitutes a significant clinical challenge, and there is extensive research focused on identifying molecular activities that can facilitate the repair of spinal cord injuries. Mammalian sterile 20-like kinase 2 (MST2), a core component of the Hippo signaling pathway, plays a key role in apoptosis and cell growth. However, its role in neurite outgrowth after spinal cord injury remains unknown. Through comprehensive in vitro and in vivo experiments, we demonstrated that MST2, predominantly expressed in neurons, actively participated in the natural development of the CNS. Post-SCI, MST2 expression significantly increased, indicating its activation and potential role in the early stages of neural recovery. Detailed analyses showed that MST2 knockdown impaired neurite outgrowth and motor function recovery, whereas MST2 overexpression led to the opposite effects, underscoring MST2's neuroprotective role in enhancing neural repair. Further, we elucidated the mechanism underlying MST2's action, revealing its interaction with AKT and positive regulation of AKT activity, a well-established promoter of neurite outgrowth. Notably, MST2's promotion of neurite outgrowth and motor functional recovery was diminished by AKT inhibitors, highlighting the dependency of MST2's neuroprotective effects on AKT signaling. In conclusion, our findings affirmed MST2's pivotal role in fostering neuronal neurite outgrowth and facilitating functional recovery after SCI, mediated through its positive modulation of AKT activity. In conclusion, our findings confirmed MST2's crucial role in neural protection, promoting neurite outgrowth and functional recovery after SCI through positive AKT activity modulation. These results position MST2 as a potential therapeutic target for SCI, offering new insights into strategies for enhancing neuroregeneration and functional restoration.

11.
Life Sci ; 345: 122606, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38574884

RÉSUMÉ

AIMS: Alzheimer's disease (AD), the most common neurodegenerative disorder associated with aging, is characterized by amyloid-ß (Aß) plaques in the hippocampus. Ergosterol, a mushroom sterol, exhibits neuroprotective activities; however, the underlying mechanisms of ergosterol in promoting neurite outgrowth and preventing Aß-associated aging have never been investigated. We aim to determine the beneficial activities of ergosterol in neuronal cells and Caenorhabditis elegans (C. elegans). MATERIALS AND METHODS: The neuritogenesis and molecular mechanisms of ergosterol were investigated in wild-type and Aß precursor protein (APP)-overexpressing Neuro2a cells. The anti-amyloidosis properties of ergosterol were determined by evaluating in vitro Aß production and the potential inhibition of Aß-producing enzymes. Additionally, AD-associated transgenic C. elegans was utilized to investigate the in vivo attenuating effects of ergosterol. KEY FINDINGS: Ergosterol promoted neurite outgrowth in Neuro2a cells through the upregulation of the transmembrane protein Teneurin-4 (Ten-4) mRNA and protein expressions, phosphorylation of the extracellular signal-regulated kinases (ERKs), activity of cAMP response element (CRE), and growth-associated protein-43 (GAP-43). Furthermore, ergosterol enhanced neurite outgrowth in transgenic Neuro2A cells overexpressing either the wild-type APP (Neuro2a-APPwt) or the Swedish mutant APP (Neuro2a-APPswe) through the Ten-4/ERK/CREB/GAP-43 signaling pathway. Interestingly, ergosterol inhibited Aß synthesis in Neuro2a-APPwt cells. In silico analysis indicated that ergosterol can interact with the catalytic sites of ß- and γ-secretases. In Aß-overexpressing C. elegans, ergosterol decreased Aß accumulation, increased chemotaxis behavior, and prolonged lifespan. SIGNIFICANCE: Ergosterol is a potential candidate compound that might benefit AD patients by promoting neurite outgrowth, inhibiting Aß synthesis, and enhancing longevity.


Sujet(s)
Maladie d'Alzheimer , Animaux , Humains , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/métabolisme , Amyloid precursor protein secretases/métabolisme , Animal génétiquement modifié/métabolisme , Caenorhabditis elegans/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Protéine GAP-43 , Longévité , Neuroblastome , Excroissance neuronale , Lignée cellulaire tumorale
12.
Endocr Regul ; 58(1): 105-114, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38656256

RÉSUMÉ

Oxytocin plays an important role in brain development and is associated with various neurotransmitter systems in the brain. Abnormalities in the production, secretion, and distribution of oxytocin in the brain, at least during some stages of the development, are critical for the pathogenesis of neuropsychiatric diseases, particularly in the autism spectrum disorder. The etiology of autism includes changes in local sensory and dopaminergic areas of the brain, which are also supplied by the hypothalamic sources of oxytocin. It is very important to understand their mutual relationship. In this review, the relationship of oxytocin with several components of the dopaminergic system, gamma-aminobutyric acid (GABA) inhibitory neurotransmission and their alterations in the autism spectrum disorder is discussed. Special attention has been paid to the results describing a reduced expression of inhibitory GABAergic markers in the brain in the context of dopaminergic areas in various models of autism. It is presumed that the altered GABAergic neurotransmission, due to the absence or dysfunction of oxytocin at certain developmental stages, disinhibits the dopaminergic signaling and contributes to the autism symptoms.


Sujet(s)
Trouble autistique , Encéphale , Dopamine , Ocytocine , Acide gamma-amino-butyrique , Ocytocine/métabolisme , Ocytocine/physiologie , Humains , Dopamine/métabolisme , Acide gamma-amino-butyrique/métabolisme , Trouble autistique/métabolisme , Encéphale/métabolisme , Animaux , Transmission synaptique/physiologie , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/étiologie
13.
J Nat Med ; 78(3): 599-607, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38662302

RÉSUMÉ

In this study, the effects of 3,5,7,3',4'-pentamethoxyflavone (KP1), a major bioactive ingredient isolated from the Kaempferia parviflora rhizomes, on a neurite outgrowth in Neuro2a cells and its mechanism have been investigated. KP1 increased concentration-dependently the percentage of neurite-bearing cells. KP1 showed a remarkable capability to elicit neurite outgrowth in Neuro2a cells, as evidenced by morphological alterations and immunostaining using anti-class III ß-tubulin and anti-NeuN antibodies. KP1 also displayed a higher neurogenic activity than retinoic acid (RA), a promoter of neurite outgrowth in Neuro2a cells. KP1 treatment caused significant elevation in phosphorylation of extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (p38 MAPK) and glycogen synthase kinase-3ß (GSK-3ß). However, KP1-triggered neurite outgrowth was markedly inhibited by treatment with the ERK inhibitor U0126, whereas p38 MAPK inhibitor SB203580 and GSK-3ß inhibitor SB216763 did not influence KP1-induced neurite outgrowth. These results demonstrate that KP1 elicits neurite outgrowth and triggers cell differentiation of Neuro2a cells through ERK signal pathway.


Sujet(s)
Système de signalisation des MAP kinases , Excroissance neuronale , Animaux , Excroissance neuronale/effets des médicaments et des substances chimiques , Souris , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , p38 Mitogen-Activated Protein Kinases/métabolisme , Neurites/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Flavonoïdes/pharmacologie , Flavones/pharmacologie , Flavones/composition chimique , Lignée cellulaire tumorale , Glycogen synthase kinase 3 beta/métabolisme , Lignée cellulaire
14.
Food Chem Toxicol ; 187: 114623, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38554842

RÉSUMÉ

Acrylamide (ACR) is a known neurotoxicant and developmental neurotoxicant. As a soft electrophile, ACR reacts with thiol groups in cysteine. One hypothesis of ACR induced neurotoxicity and developmental neurotoxicity (DNT) is conjugation with reduced glutathione (GSH) leading to GSH depletion, increased reactive oxygen species (ROS) production and further oxidative stress and cellular damage. In this regard, we have investigated the effect of ACR on neuronal differentiation, glutathione levels and ROS production in the human neuroblastoma SH-SY5Y cell model. After 9 days of differentiation and exposure, ACR significantly impaired area neurites per cell at non-cytotoxic concentrations (0.33 µM and 10 µM). Furthermore, 10 µM ACR dysregulated 9 mRNA markers important for neuronal development, 5 of them being associated with cytoskeleton organization and axonal guidance. At the non-cytotoxic concentrations that significantly attenuate neuronal differentiation, ACR did neither decrease the level of GSH or total glutathione levels, nor increased ROS production. In addition, the expression of 5 mRNA markers for cellular stress was assessed with no significant altered regulation after ACR exposure up to 320 µM. Thus, ACR-induced DNT is not due to GSH depletion and increased ROS production, neither at non-cytotoxic nor cytotoxic concentrations, in the SH-SH5Y model during differentiation.


Sujet(s)
Acrylamide , Neuroblastome , Humains , Espèces réactives de l'oxygène/métabolisme , Acrylamide/toxicité , Neuroblastome/métabolisme , Stress oxydatif , Glutathion/métabolisme , ARN messager/métabolisme , Lignée cellulaire tumorale
15.
Front Mol Neurosci ; 17: 1371135, 2024.
Article de Anglais | MEDLINE | ID: mdl-38516042

RÉSUMÉ

Canonical retinoid signaling via nuclear receptors and gene regulation is critical for the initiation of developmental processes such as cellular differentiation, patterning and neurite outgrowth, but also mediates nerve regeneration and synaptic functions in adult nervous systems. In addition to canonical transcriptional regulation, retinoids also exert rapid effects, and there are now multiple lines of evidence supporting non-canonical retinoid actions outside of the nucleus, including in dendrites and axons. Together, canonical and non-canonical retinoid signaling provide the precise temporal and spatial control necessary to achieve the fine cellular coordination required for proper nervous system function. Here, we examine and discuss the evidence supporting non-canonical actions of retinoids in neural development and regeneration as well as synaptic function, including a review of the proposed molecular mechanisms involved.

16.
Biomed Pharmacother ; 173: 116455, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38503234

RÉSUMÉ

The sigma-1 receptor (σ-1R), a chaperone protein located at the mitochondria-associated membrane (MAM) of the endoplasmic reticulum, can interact with and modify the signaling pathways of various proteins, thereby modulating many disease pathologies, including Alzheimer's disease (AD). The σ-1R ligand dipentylammonium (DPA) was analyzed for its anti-AD properties using PC12 cells (in vitro) and Caenorhabditis elegans (in vivo) models along with molecular docking (in silico) analysis. DPA at 1 and 10 µM concentrations was able to significantly potentiate NGF-induced neurite growth length by 137.7 ± 12.0 and 187.8 ± 16.4, respectively, when compared to the control 76.9 ± 7.4. DPA also regulated neurite damage caused by Aß(25-35) treatment in differentiated PC12 cells by improving cell viability and neurite length. In C. elegans, DPA could significantly extend the median and maximum lifespan of Aß transgenic strain CL2006 without impacting wild-type nematodes. Additionally, it could significantly reduce the paralysis phenotype of another Aß transgenic strain, CL4176, thereby improving the overall health in AD pathogenesis. This effect depended on σ-1R, as DPA could not modulate the lifespan of σ-1R mutant TM3443. This was further confirmed using agonist PRE084 and antagonist BD1047, wherein the agonist alone could extend the lifespan of CL2006, while the antagonist suppressed the effect of DPA in CL2006. Interestingly, neither had an TM3443. Further, molecular docking analysis showed that DPA had a similar binding affinity as that of PRE084, BD1047 and pentazocine against the σ-1R receptor in humans and C. elegans, which collectively suggests the anti-AD properties of DPA.


Sujet(s)
Maladie d'Alzheimer , Composés d'ammonium , Éthylènediamines , Neuroprotecteurs , Récepteur sigma , Animaux , Rats , Humains , Maladie d'Alzheimer/traitement médicamenteux , , Caenorhabditis elegans , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Ligands , Simulation de docking moléculaire , Animal génétiquement modifié/métabolisme , Techniques de culture cellulaire , Peptides bêta-amyloïdes/métabolisme , Récepteur sigma/métabolisme
17.
Dis Model Mech ; 17(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38511331

RÉSUMÉ

CDK13-related disorder, also known as congenital heart defects, dysmorphic facial features and intellectual developmental disorder (CHDFIDD) is associated with mutations in the CDK13 gene encoding transcription-regulating cyclin-dependent kinase 13 (CDK13). Here, we focused on the development of craniofacial structures and analyzed early embryonic stages in CHDFIDD mouse models, with one model comprising a hypomorphic mutation in Cdk13 and exhibiting cleft lip/palate, and another model comprising knockout of Cdk13, featuring a stronger phenotype including midfacial cleft. Cdk13 was found to be physiologically expressed at high levels in the mouse embryonic craniofacial structures, namely in the forebrain, nasal epithelium and maxillary mesenchyme. We also uncovered that Cdk13 deficiency leads to development of hypoplastic branches of the trigeminal nerve including the maxillary branch. Additionally, we detected significant changes in the expression levels of genes involved in neurogenesis (Ache, Dcx, Mef2c, Neurog1, Ntn1, Pou4f1) within the developing palatal shelves. These results, together with changes in the expression pattern of other key face-specific genes (Fgf8, Foxd1, Msx1, Meis2 and Shh) at early stages in Cdk13 mutant embryos, demonstrate a key role of CDK13 in the regulation of craniofacial morphogenesis.


Sujet(s)
Modèles animaux de maladie humaine , Développement embryonnaire , Régulation de l'expression des gènes au cours du développement , Neurogenèse , Animaux , Neurogenèse/génétique , Développement embryonnaire/génétique , Kinases cyclines-dépendantes/métabolisme , Kinases cyclines-dépendantes/génétique , Crâne/embryologie , Crâne/anatomopathologie , Souris , Fente palatine/génétique , Fente palatine/anatomopathologie , Fente palatine/embryologie , Bec-de-lièvre/génétique , Bec-de-lièvre/anatomopathologie , Bec-de-lièvre/embryologie , Nerf trijumeau/embryologie , Embryon de mammifère/métabolisme , Face/embryologie , Face/malformations , Phénotype , Déficience intellectuelle/génétique , Mutation/génétique , Protéine doublecortine
18.
J Cancer ; 15(5): 1153-1168, 2024.
Article de Anglais | MEDLINE | ID: mdl-38356706

RÉSUMÉ

We conducted a high-content screening (HCS) in neuroblastoma BE(2)-C cells to identify cell cycle regulators that control cell differentiation using a library of siRNAs against cell cycle-regulatory genes. We discovered that knocking down expression of cyclin dependent kinase inhibitor 3 (CDKN3) showed the most potent effect in inducing neurite outgrowth, the morphological cell differentiation marker of neuroblastoma cells. We then demonstrated that CDKN3 knockdown increased expression of neuroblastoma molecular differentiation markers, neuron specific enolase (NSE), ßIII-tubulin and growth associated protein 43 (GAP43). We further showed that CDKN3 knockdown reduced expression of cell proliferation markers Ki67 and proliferating cell nuclear antigen (PCNA), and reduced colony formation of neuroblastoma cells. More importantly, we observed a correlation of high tumor CDKN3 mRNA levels with poor patient survival in the investigation of public neuroblastoma patient datasets. In exploring the mechanisms that regulate CDKN3 expression, we found that multiple strong differentiation-inducing molecules, including miR-506-3p and retinoic acid, down-regulated CDKN3 expression. In addition, we found that N-Myc promoted CDKN3 expression at the transcriptional level by directly binding to the CDKN3 promoter. Furthermore, we found that CDKN3 and two additional differentiation-regulating cell cycle proteins identified in our HCS, CDC6 and CDK4, form an interactive network to promote expression of each other. In summary, we for the first time discovered the function of CDKN3 in regulating neuroblastoma cell differentiation and characterized the transcriptional regulation of CDKN3 expression by N-Myc in neuroblastoma cells. Our findings support that CDKN3 plays a role in modulating neuroblastoma cell differentiation and that overexpression of CDKN3 may contribute to neuroblastoma progression.

19.
Prog Chem Org Nat Prod ; 123: 1-473, 2024.
Article de Anglais | MEDLINE | ID: mdl-38340248

RÉSUMÉ

Neurotrophins (NGF, BDNF, NT3, NT4) can decrease cell death, induce differentiation, as well as sustain the structure and function of neurons, which make them promising therapeutic agents for the treatment of neurodegenerative disorders. However, neurotrophins have not been very effective in clinical trials mostly because they cannot pass through the blood-brain barrier owing to being high-molecular-weight proteins. Thus, neurotrophin-mimic small molecules, which stimulate the synthesis of endogenous neurotrophins or enhance neurotrophic actions, may serve as promising alternatives to neurotrophins. Small-molecular-weight natural products, which have been used in dietary functional foods or in traditional medicines over the course of human history, have a great potential for the development of new therapeutic agents against neurodegenerative diseases such as Alzheimer's disease. In this contribution, a variety of natural products possessing neurotrophic properties such as neurogenesis, neurite outgrowth promotion (neuritogenesis), and neuroprotection are described, and a focus is made on the chemistry and biology of several neurotrophic natural products.


Sujet(s)
Produits biologiques , Humains , Produits biologiques/pharmacologie , Facteurs de croissance nerveuse/pharmacologie , Facteurs de croissance nerveuse/métabolisme , Neurones/métabolisme , Neurogenèse , Différenciation cellulaire/physiologie
20.
Bioorg Med Chem Lett ; 102: 129670, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38387692

RÉSUMÉ

Histone deacetylase 6 (HDAC6) has drawn more and more attention for its potential application in Alzheimer's disease (AD) therapy. A series of tetrahydro-ß-carboline (THßC) hydroxamic acids with aryl linker were synthesized. In enzymatic assay, all compounds exhibited nanomolar IC50 values. The most promising compound 11d preferentially inhibited HDAC6 (IC50, 8.64 nM) with approximately 149-fold selectivity over HDAC1. Molecular simulation revealed that the hydroxamic acid of 11d could bind to the zinc ion by a bidentate chelating manner. In vitro, 11d induced neurite outgrowth of PC12 cells without producing toxic effects and showed obvious neuroprotective activity in a model of H2O2-induced oxidative stress.


Sujet(s)
Carbolines , Inhibiteurs de désacétylase d'histone , Peroxyde d'hydrogène , Rats , Animaux , Histone deacetylase 6 , Inhibiteurs de désacétylase d'histone/pharmacologie , Peroxyde d'hydrogène/pharmacologie , Acides hydroxamiques/pharmacologie , Excroissance neuronale , Histone Deacetylase 1/métabolisme , Relation structure-activité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE