Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres











Gamme d'année
1.
Int J Mol Sci ; 25(7)2024 Apr 02.
Article de Anglais | MEDLINE | ID: mdl-38612763

RÉSUMÉ

Idiopathic intellectual disability (IID) encompasses the cases of intellectual disability (ID) without a known cause and represents approximately 50% of all cases. Neural progenitor cells (NPCs) from the olfactory neuroepithelium (NEO) contain the same information as the cells found in the brain, but they are more accessible. Some miRNAs have been identified and associated with ID of known etiology. However, in idiopathic ID, the effect of miRNAs is poorly understood. The aim of this study was to determine the miRNAs regulating the expression of mRNAs that may be involved in development of IID. Expression profiles were obtained using NPC-NEO cells from IID patients and healthy controls by microarray. A total of 796 miRNAs and 28,869 mRNAs were analyzed. Several miRNAs were overexpressed in the IID patients compared to controls. miR-25 had the greatest expression. In silico analysis showed that ROBO2 was the target for miR-25, with the highest specificity and being the most down-regulated. In vitro assay showed an increase of miR-25 expression induced a decrease in ROBO2 expression. In neurodevelopment, ROBO2 plays a crucial role in episodic learning and memory, so its down-regulation, caused by miR-25, could have a fundamental role in the intellectual disability that, until now, has been considered idiopathic.


Sujet(s)
Déficience intellectuelle , microARN , Humains , Déficience intellectuelle/génétique , microARN/génétique , Encéphale , Régulation négative/génétique , Apprentissage , ARN messager , Roundabout Proteins , Récepteurs immunologiques/génétique
2.
Microorganisms ; 11(11)2023 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-38004683

RÉSUMÉ

During Toxoplasma gondii chronic infection, certain internal factors that trigger the proliferation of neural progenitor cells (NPCs), such as brain inflammation, cell death, and changes in cytokine levels, are observed. NPCs give rise to neuronal cell types in the adult brain of some mammals. NPCs are capable of dividing and differentiating into a restricted repertoire of neuronal and glial cell types. In this study, the proliferation of NPCs was evaluated in CD-1 adult male mice chronically infected with the T. gondii ME49 strain. Histological brain sections from the infected mice were evaluated in order to observe T. gondii tissue cysts. Sagittal and coronal sections from the subventricular zone of the lateral ventricles and from the subgranular zone of the hippocampal dentate gyrus, as well as sagittal sections from the rostral migratory stream, were obtained from infected and non-infected mice previously injected with bromodeoxyuridine (BrdU). A flotation immunofluorescence technique was used to identify BrdU+ NPC. The scanning of BrdU+ cells was conducted using a confocal microscope, and the counting was performed with ImageJ® software (version 1.48q). In all the evaluated zones from the infected mice, a significant proliferation of the NPCs was observed when compared with that of the control group. We concluded that chronic infection with T. gondii increased the proliferation of NPCs in the three evaluated zones. Regardless of the role these cells are playing, our results could be useful to better understand the pathogenesis of chronic toxoplasmosis.

3.
Stem Cell Rev Rep ; 19(6): 1800-1811, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37129730

RÉSUMÉ

Proteins involved in the Alzheimer's disease (AD), such as amyloid precursor protein (APP) and presenilin-1 (PS1), play critical roles in early development of the central nervous system (CNS), as well as in innate immune and glial cell responses. Familial AD is associated with the presence of APPswe and PS1dE9 mutations. However, it is still unknown whether these mutations cause deficits in CNS development of carriers. We studied genome-wide gene expression profiles of differentiated neural progenitor cells (NPCs) from wild-type and APPswe/PS1dE9 mouse embryo telencephalon. The occurrence of strong innate immune and glial cell responses in APPswe/PS1dE9 neurospheres mainly involves microglial activation, inflammatory mediators and chemokines. APPswe/PS1dE9 neurospheres augmented up to 100-fold CCL12, CCL5, CCL3, C3, CX3CR1, TLR2 and TNF-alpha expression levels, when compared to WT neurospheres. Expression levels of the glia cell marker GFAP and microglia marker Iba-1 were up to 20-fold upregulated in APPswe/PS1dE9 neurospheres. The secretome of differentiated APPswe/PS1dE9 NPCs revealed enhanced chemoattraction of peripheral blood mononuclear cells. When evaluating the inferred protein interaction networks constructed from the array data, an improvement in astrocyte differentiation in APPswe/PS1dE9 neurospheres was evident in view of increased GFAP expression. Transgenic NPCs differentiated into neural phenotypes presented expression patterns of cytokine, glial cells, and inflammatory mediators characteristic of APPswe/PS1dE9 adult animals. Consequently, the neurogenic niche obtained from differentiation of embryonic APPswe/PS1dE9 neurospheres spontaneously presents several alterations observed in adult AD brains. Finally, our data strengthen pathophysiological hypotheses that propose an early neurodevelopmental origin for familial AD.


Sujet(s)
Maladie d'Alzheimer , Souris , Animaux , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/complications , Maladie d'Alzheimer/métabolisme , Agranulocytes/métabolisme , Souris transgéniques , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Névroglie/métabolisme , Différenciation cellulaire/génétique , Médiateurs de l'inflammation , Immunité innée/génétique
4.
Braz. j. otorhinolaryngol. (Impr.) ; Braz. j. otorhinolaryngol. (Impr.);88(6): 962-967, Nov.-Dec. 2022. graf
Article de Anglais | LILACS-Express | LILACS | ID: biblio-1420782

RÉSUMÉ

Abstract Introduction: Inner ear progenitor cells have the potential for multi-directional differentiation. Retinoic acid is an important requirement for the development of the inner ear. Blocking the Curtyr's retinoic acid signaling pathway can significantly reduce the number of hair cells. Therefore, we believe that retinoic acid may induce the regeneration of inner ear hair cells. Objective: To investigate whether the cochlear neural progenitor cells maintain the characteristics of stem cells during recovery and subculture, whether retinoic acid can induce cochlear neural progenitor cells into hair cells in vitro, and whether retinoic acid promotes or inhibits the proliferation of cochlear neural progenitor cells during differentiation. Methods: Cochlear neural progenitor cells were cultured and induced in DMEM/F12 + RA (10−6M) and then detected the expressions of hair cell markers (Math1 and MyosinVIIa) by immunofluorescence cytochemistry and realtime-polymerase chain reaction, and the proliferation of cochlear neural progenitor cells was detected by Brdu. Results: The nestin of cochlear neural progenitor cells was positively expressed. The ratios of Math1-positive cells in the control group and experimental group were 1.5% and 63%, respectively; the ratios of MyosinVIIa-positive cells in the control group and experimental group were 0.96% and 56%, respectively (p <0.05). The ratios of Brdu+-labeled cells in retinoic acid group, group PBS, and group FBS were 20.6%, 29.9%, and 54.3%, respectively; however, the proliferation rate in the experimental group decreased. Conclusion: Retinoic acid can promote cochlear neural progenitor cells to differentiate into the hair cells.


Resumo Introdução: As células progenitoras da orelha interna têm potencial para diferenciação multidirecional. O ácido retinoico é uma condição importante para o desenvolvimento da orelha interna. O bloqueio da via de sinalização do ácido retinoico no órgão de Corti pode reduzir significativamente o número de células ciliadas. Portanto, acreditamos que o ácido retinoico pode induzir a regeneração das células ciliadas do ouvido interno. Objetivo: Investigar se as células progenitoras neurais cocleares mantêm as características das células-tronco durante a recuperação e subcultura, se o ácido retinoico pode induzir a transformação de células progenitoras neurais cocleares em células ciliadas in vitro e se o ácido retinoico promove ou inibe a proliferação das células progenitoras durante a diferenciação. Método: As células progenitoras neurais cocleares foram cultivadas e induzidas em DMEM/F12+AR (106M) e, então, foram detectadas as expressões de marcadores das células ciliadas (Math1 e Myosin?a) com o uso de citoquímica por imunofluorescência e real time -polymerase chain reaction e a proliferação de células progenitoras neurais cocleares foi detectada pelo teste Brdu. Resultados: A nestina das células progenitoras neurais cocleares foi expressa positivamente. As proporções de células positivas para Math1 no grupo controle e no grupo experimental foram 1,5% e 63%, respectivamente; as proporções de células positivas para Myosin?a no grupo controle e no grupo experimental foram de 0,96% e 56%, respectivamente (p <0,05). As proporções de células marcadas com Brdu+ no grupo ácido retinoico, grupo PBS e grupo FBS foram de 20,6%, 29,9% e 54,3%, respectivamente; no entanto, a taxa de proliferação no grupo experimental diminuiu. Conclusões: O ácido retinoico pode promover a diferenciação das células progenitoras neurais cocleares em células ciliadas.

5.
Front Neurosci ; 16: 828646, 2022.
Article de Anglais | MEDLINE | ID: mdl-35360153

RÉSUMÉ

Prenatal exposure to maternal immune activation (MIA) has been suggested to increase the probability of autism spectrum disorder (ASD). Recent evidence from animal studies indicates a key role for interleukin-17a (IL-17a) in promoting MIA-induced behavioral and brain abnormalities reminiscent of ASD. However, it is still unclear how IL-17a acts on the human developing brain and the cell types directly affected by IL-17a signaling. In this study, we used iPSC-derived neural progenitor cells (NPCs) from individuals with ASD of known and unknown genetic cause as well as from neurotypical controls to examine the effects of exogenous IL-17a on NPC proliferation, migration and neuronal differentiation, and whether IL-17a and genetic risk factors for ASD interact exacerbating alterations in NPC function. We observed that ASD and control NPCs endogenously express IL-17a receptor (IL17RA), and that IL-17a/IL17RA activation modulates downstream ERK1/2 and mTORC1 signaling pathways. Exogenous IL-17a did not induce abnormal proliferation and migration of ASD and control NPCs but, on the other hand, it significantly increased the expression of synaptic (Synaptophysin-1, Synapsin-1) and neuronal polarity (MAP2) proteins in these cells. Also, as we observed that ASD and control NPCs exhibited similar responses to exogenous IL-17a, it is possible that a more inflammatory environment containing other immune molecules besides IL-17a may be needed to trigger gene-environment interactions during neurodevelopment. In conclusion, our results suggest that exogenous IL-17a positively regulates the neuronal differentiation of human NPCs, which may disturb normal neuronal and synaptic development and contribute to MIA-related changes in brain function and behavior.

6.
Braz J Otorhinolaryngol ; 88(6): 962-967, 2022.
Article de Anglais | MEDLINE | ID: mdl-33707121

RÉSUMÉ

INTRODUCTION: Inner ear progenitor cells have the potential for multi-directional differentiation. Retinoic acid is an important requirement for the development of the inner ear. Blocking the Curtyr's retinoic acid signaling pathway can significantly reduce the number of hair cells. Therefore, we believe that retinoic acid may induce the regeneration of inner ear hair cells. OBJECTIVE: To investigate whether the cochlear neural progenitor cells maintain the characteristics of stem cells during recovery and subculture, whether retinoic acid can induce cochlear neural progenitor cells into hair cells in vitro, and whether retinoic acid promotes or inhibits the proliferation of cochlear neural progenitor cells during differentiation. METHODS: Cochlear neural progenitor cells were cultured and induced in DMEM/F12+RA (10-6M) and then detected the expressions of hair cell markers (Math1 and MyosinVIIa) by immunofluorescence cytochemistry and realtime-polymerase chain reaction, and the proliferation of cochlear neural progenitor cells was detected by Brdu. RESULTS: The nestin of cochlear neural progenitor cells was positively expressed. The ratios of Math1-positive cells in the control group and experimental group were 1.5% and 63%, respectively; the ratios of MyosinVIIa-positive cells in the control group and experimental group were 0.96% and 56%, respectively (p<0.05). The ratios of Brdu+-labeled cells in retinoic acid group, group PBS, and group FBS were 20.6%, 29.9%, and 54.3%, respectively; however, the proliferation rate in the experimental group decreased. CONCLUSION: Retinoic acid can promote cochlear neural progenitor cells to differentiate into the hair cells.


Sujet(s)
Cellules souches neurales , Trétinoïne , Humains , Trétinoïne/pharmacologie , Broxuridine , Cellules cultivées , Différenciation cellulaire
7.
Adv Exp Med Biol ; 1326: 55-72, 2021.
Article de Anglais | MEDLINE | ID: mdl-33378003

RÉSUMÉ

Lin28 is a highly conserved RNA binding protein that regulates stemness whose molecular role has been widely studied in vitro. However, the regulation and the molecular role of Lin28 during the development of the vertebrate central nervous system (CNS) in vivo are not completely understood. Here, the expression and the putative role of Lin28 in the development of the mammalian CNS are reviewed in the context of recent results showing the progressive cellular and molecular changes in neural progenitor cells. Downstream genes that may play a role during CNS development and the effect of misregulated expression of Lin28 are discussed. Evidence suggests that Lin28 promotes symmetric divisions over asymmetric divisions, increasing the number of progenitors during early neurogenesis. Future quantitative analysis of Lin28 isoforms levels and stabilities together with single cell transcriptomics data, cell cycle dynamics and cell fate analysis in Lin28 gain- and loss-of-function experiments will provide a better understanding of the molecular role of Lin28 during development.


Sujet(s)
microARN , Cellules souches neurales , Animaux , Différenciation cellulaire , Prolifération cellulaire , Système nerveux central
8.
Front Cell Infect Microbiol ; 10: 558324, 2020.
Article de Anglais | MEDLINE | ID: mdl-33251156

RÉSUMÉ

Zika virus (ZIKV), a member of the Flaviviridae family, was brought into the spotlight due to its widespread and increased pathogenicity, including Guillain-Barré syndrome and microcephaly. Neural progenitor cells (NPCs), which are multipotent cells capable of differentiating into the major neural phenotypes, are very susceptible to ZIKV infection. Given the complications of ZIKV infection and potential harm to public health, effective treatment options are urgently needed. Betulinic acid (BA), an abundant terpenoid of the lupane group, displays several biological activities, including neuroprotective effects. Here we demonstrate that Sox2+ NPCs, which are highly susceptible to ZIKV when compared to their neuronal counterparts, are protected against ZIKV-induced cell death when treated with BA. Similarly, the population of Sox2+ and Casp3+ NPCs found in ZIKV-infected cerebral organoids was significantly higher in the presence of BA than in untreated controls. Moreover, well-preserved structures were found in BA-treated organoids in contrast to ZIKV-infected controls. Bioinformatics analysis indicated Akt pathway activation by BA treatment. This was confirmed by phosphorylated Akt analysis, both in BA-treated NPCs and brain organoids, as shown by immunoblotting and immunofluorescence analyses, respectively. Taken together, these data suggest a neuroprotective role of BA in ZIKV-infected NPCs.


Sujet(s)
Microcéphalie , Cellules souches neurales , Infection par le virus Zika , Virus Zika , Humains , Triterpènes pentacycliques , Infection par le virus Zika/traitement médicamenteux , Acide bétulinique
9.
J Tissue Eng Regen Med ; 14(12): 1880-1891, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33049106

RÉSUMÉ

In this study, porcine embryonic fibroblasts (pEFs) were reprogrammed into porcine-induced pluripotent stem cells (piPSCs) using either human or mouse specific sequences for the OCT4, SOX2, c-Myc, and KLF4 transcription factors. In total, three pEFs lines were reprogrammed, cultured for at least 15 passages, and characterized regarding their pluripotency status (alkaline phosphatase expression, embryoid body formation, expression of exogenous and endogenous genes, and immunofluorescence). Two piPSC lines were further differentiated, using chemical inhibitors, into putative neural progenitor-like (NPC-like) cells with subsequent analyses of their morphology and expression of neural markers such as NESTIN and GFAP as well as immunofluorescent labeling of NESTIN, ß-TUBULIN III, and VIMENTIN. NPC-like cells were positive for all the neural markers tested. These results evidence of the generation of porcine NPC-like cells after in vitro induction with chemical inhibitors, representing an adequate model for future regenerative and translational medicine research.


Sujet(s)
Différenciation cellulaire , Cellules souches pluripotentes induites/cytologie , Cellules souches neurales/cytologie , Phosphatase alcaline/métabolisme , Animaux , Marqueurs biologiques/métabolisme , Différenciation cellulaire/génétique , Lignée cellulaire , Forme de la cellule , Reprogrammation cellulaire , Corps embryoïdes/cytologie , Régulation de l'expression des gènes , Cellules souches pluripotentes induites/métabolisme , Facteur-4 de type Kruppel , Cellules souches neurales/métabolisme , Neurones/cytologie , Suidae
10.
J Mol Histol ; 51(4): 437-453, 2020 Aug.
Article de Anglais | MEDLINE | ID: mdl-32653982

RÉSUMÉ

The hilus plays an important role modulating the excitability of the hippocampal dentate gyrus (DG). It also harbors proliferative cells whose proliferation rate is modified during pathological events. However, the characterization of these cells, in terms of cellular identity, lineage, and fate, as well as the morphology and proportion of each cell subpopulation has been poorly studied. Therefore, a deeper investigation of hilar proliferative cells might expand the knowledge not only in the physiology, but in the pathophysiological processes related to the hippocampus too. The aim of this work was to perform an integrative study characterizing the identity of proliferative cells populations harbored in the hilus, along with morphology and proportion. In addition, this study provides comparative evidence of the subgranular zone (SGZ) of the DG. Quantified cells included proliferative, neural precursor, Type 1, oligodendrocyte progenitor (OPCs), neural progenitor (NPCs), and proliferative mature astrocytes in the hilus and SGZ of Wistar adult rats. Our results showed that 84% of the hilar proliferative cells correspond to neural precursor cells, OPCs and NPCs being the most abundant at 54 and 45%, respectively, unlike the SGZ, where OPCs represent only 11%. Proliferative mature astrocytes and Type 1-like cells were rarely observed in the hilus. Together, our results lay the basis for future studies focused on the lineage and fate of hilar proliferative cells and suggest that the hilus could be relevant to the formation of new cells that modulate multiple physiological processes governed by the hippocampus.


Sujet(s)
Prolifération cellulaire/physiologie , Gyrus denté/physiologie , Animaux , Astrocytes/physiologie , Numération cellulaire/méthodes , Cellules souches neurales/physiologie , Neurogenèse/physiologie , Neurones/physiologie , Rats , Rat Wistar , Cellules souches/physiologie
11.
Microb Pathog ; 136: 103703, 2019 Nov.
Article de Anglais | MEDLINE | ID: mdl-31476377

RÉSUMÉ

Trypanosoma evansi appears to have a significant tropism for brain tissue in its chronic and acute phases. The most common symptoms of this brain infection are motor incoordination, meningoencephalitis, demyelination, and anemia. There have only been few studies of the effects of T. evansi infection on neuronal differentiation and brain plasticity. Here, we investigated the impact of the congenital T. evansi infection on brain development in mice. We collected telencephalon-derived neural progenitor cells (NPCs) from T. evansi uninfected and infected mice, and cultivated them into neurospheres. We found that T. evansi significantly decreased the number of cells during development of neurospheres. Analysis of neurosphere differentiation revealed that T. evansi infection significantly increased neural migration. We also observed that T. evansi promoted expression of glial fibrillary acidic protein (GFAP) in infected cells. These data suggest that congenital T. evansi infection may affect embryonic brain development.


Sujet(s)
Interactions hôte-pathogène , Cellules souches neurales/anatomopathologie , Cellules souches neurales/parasitologie , Trypanosoma/croissance et développement , Animaux , Différenciation cellulaire , Souris
12.
Cell Transplant ; 28(3): 269-285, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30574805

RÉSUMÉ

Individuals with Parkinson's disease (PD) suffer from motor and mental disturbances due to degeneration of dopaminergic and non-dopaminergic neuronal systems. Although they provide temporary symptom relief, current treatments fail to control motor and non-motor alterations or to arrest disease progression. Aiming to explore safety and possible motor and neuropsychological benefits of a novel strategy to improve the PD condition, a case series study was designed for brain grafting of human neural progenitor cells (NPCs) to a group of eight patients with moderate PD. A NPC line, expressing Oct-4 and Sox-2, was manufactured and characterized. Using stereotactic surgery, NPC suspensions were bilaterally injected into patients' dorsal putamina. Cyclosporine A was given for 10 days prior to surgery and continued for 1 month thereafter. Neurological, neuropsychological, and brain imaging evaluations were performed pre-operatively, 1, 2, and 4 years post-surgery. Seven of eight patients have completed 4-year follow-up. The procedure proved to be safe, with no immune responses against the transplant, and no adverse effects. One year after cell grafting, all but one of the seven patients completing the study showed various degrees of motor improvement, and five of them showed better response to medication. PET imaging showed a trend toward enhanced midbrain dopaminergic activity. By their 4-year evaluation, improvements somewhat decreased but remained better than at baseline. Neuropsychological changes were minor, if at all. The intervention appears to be safe. At 4 years post-transplantation we report that undifferentiated NPCs can be delivered safely by stereotaxis to both putamina of patients with PD without causing adverse effects. In 6/7 patients in OFF condition improvement in UPDRS III was observed. PET functional scans suggest enhanced putaminal dopaminergic neurotransmission that could correlate with improved motor function, and better response to L-DOPA. Patients' neuropsychological scores were unaffected by grafting. Trial Registration: Fetal derived stem cells for Parkinson's disease https://doi.org/10.1186/ISRCTN39104513Reg#ISRCTN39104513.


Sujet(s)
Mésencéphale , Cellules souches neurales , Maladie de Parkinson , Putamen , Adolescent , Adulte , Sujet âgé , Allogreffes , Dopamine/métabolisme , Femelle , Études de suivi , Humains , Mâle , Mésencéphale/métabolisme , Mésencéphale/anatomopathologie , Mésencéphale/chirurgie , Adulte d'âge moyen , Cellules souches neurales/métabolisme , Cellules souches neurales/anatomopathologie , Cellules souches neurales/transplantation , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Maladie de Parkinson/chirurgie , Putamen/métabolisme , Putamen/anatomopathologie , Putamen/chirurgie
13.
Hum Mutat ; 39(10): 1372-1383, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-29969175

RÉSUMÉ

The Reelin-DAB1 signaling pathway plays a crucial role in regulating neuronal migration and synapse function. Although many rare heterozygous variants in the Reelin gene (RELN) have been identified in patients with autism spectrum disorder (ASD), most variants are still of unknown clinical significance. Also, genetic data suggest that heterozygous variants in RELN alone appear to be insufficient to cause ASD. Here, we describe the identification and functional characterization of rare compound heterozygous missense variants in RELN in a patient with ASD in whom we have previously reported hyperfunctional mTORC1 signaling of yet unknown etiology. Using iPSC-derived neural progenitor cells (NPCs) from this patient, we provide experimental evidence that the identified variants are deleterious and lead to diminished Reelin secretion and impaired Reelin-DAB1 signal transduction. Also, our results suggest that mTORC1 pathway overactivation may function as a second hit event contributing to downregulation of the Reelin-DAB1 cascade in patient-derived NPCs, and that inhibition of mTORC1 by rapamycin attenuates Reelin-DAB1 signaling impairment. Taken together, our findings point to an abnormal interplay between Reelin-DAB1 and mTORC1 networks in nonsyndromic ASD.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Trouble du spectre autistique/génétique , Trouble du spectre autistique/métabolisme , Molécules d'adhérence cellulaire neuronale/génétique , Molécules d'adhérence cellulaire neuronale/métabolisme , Protéines de la matrice extracellulaire/génétique , Protéines de la matrice extracellulaire/métabolisme , Variation génétique , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Serine endopeptidases/génétique , Serine endopeptidases/métabolisme , Transduction du signal , Protéines adaptatrices de la transduction du signal/composition chimique , Allèles , Trouble du spectre autistique/diagnostic , Marqueurs biologiques , Études cas-témoins , Molécules d'adhérence cellulaire neuronale/composition chimique , Enfant , Enfant d'âge préscolaire , Protéines de la matrice extracellulaire/composition chimique , Femelle , Expression des gènes , Hétérozygote , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Mâle , Modèles moléculaires , Protéines de tissu nerveux/composition chimique , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Conformation des protéines , Protéines proto-oncogènes c-akt/métabolisme , Protéine reeline , Serine endopeptidases/composition chimique , Relation structure-activité , Sérine-thréonine kinases TOR/métabolisme
14.
Neurobiol Learn Mem ; 140: 82-91, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-28254465

RÉSUMÉ

Putrescine, spermidine and spermine are organic cations implicated in learning, memory consolidation, reconsolidation and neurogenesis. These physiological processes are closely related, and convincing evidence indicates that neurogenesis is implicated both, in the establishment and maintenance of remote contextual fear memory. Although brain-derived neurotrophic factor (BDNF) is a key mediator involved in both neurogenesis and memory consolidation, effects of spermidine on persistence of memory after reactivation (reconsolidation) and possible involvement of BDNF have not been investigated. Here, we investigated whether the intrahippocampal infusion of spermidine improves the persistence of reconsolidated contextual fear conditioning memory in rats and whether these possible changes depend on BDNF/TrkB signaling in the hippocampus. The infusion of spermidine immediately and 12h post-reactivation improved fear memory of the animals tested seven but not two days after reactivation. The facilitatory effect of spermidine on the persistence of reconsolidated memory was blocked by the TrkB inhibitor ANA-12 (73.6pmol/site) and accompanied by mature BDNF level increase in the hippocampus, indicating that it depends on the BDNF/TrkB pathway. We also investigated whether spermidine alters BDNF levels and neural progenitor cell differentiation in vitro. Spermidine increased BDNF levels in vitro, facilitating neuritogenesis and neural migration. Spermidine-induced neuritogenesis in vitro was also blocked by ANA-12 (10µM). Since spermidine increases BDNF levels and facilitates neural differentiation in vitro, similar mechanisms may be involved in spermidine-induced facilitation of the persistence of reconsolidated memory.


Sujet(s)
Facteur neurotrophique dérivé du cerveau/métabolisme , Peur/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Consolidation de la mémoire/effets des médicaments et des substances chimiques , Neurogenèse/effets des médicaments et des substances chimiques , Spermidine/pharmacologie , Animaux , Azépines/pharmacologie , Benzamides/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Conditionnement classique/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Mâle , Rats , Rat Wistar , Récepteur trkB/antagonistes et inhibiteurs
15.
Neurochem Res ; 42(2): 415-427, 2017 Feb.
Article de Anglais | MEDLINE | ID: mdl-27804011

RÉSUMÉ

Neural progenitors (NP), found in fetal and adult brain, differentiate into neurons potentially able to be used in cell replacement therapies. This approach however, raises technical and ethical problems which limit their potential therapeutic use. Alternately, NPs can be obtained by transdifferentiation of non-neural somatic cells evading these difficulties. Human bone marrow mesenchymal stromal cells (MSCs) are suggested to transdifferentiate into NP-like cells, which however, have a low proliferation capacity. The present study demonstrates the requisite of cell adhesion for proliferation and survival of NP-like cells and re-evaluates some neuronal features after differentiation by standard procedures. Mature neuronal markers, though, were not detected by these procedures. A chemical differentiation approach was used in this study to convert MSCs-derived NP-like cells into neurons by using a cocktail of six molecules, CHIR99021, I-BET151, RepSox, DbcAMP, forskolin and Y-27632, defined after screening combinations of 22 small molecules. Direct transdifferentiation of MSCs into neuronal cells was obtained with the small molecule cocktail, without requiring the NP-like intermediate stage.


Sujet(s)
Prolifération cellulaire/physiologie , Transdifférenciation cellulaire/physiologie , Cellules souches mésenchymateuses/physiologie , Cellules souches neurales/physiologie , Neurones/physiologie , Adolescent , Adulte , Amides/administration et posologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transdifférenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Colforsine/administration et posologie , Association médicamenteuse , Composés hétérocycliques avec 4 noyaux ou plus/administration et posologie , Humains , Mâle , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches neurales/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Pyridines/administration et posologie , Jeune adulte
16.
Mol Brain ; 9: 29, 2016 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-26980182

RÉSUMÉ

BACKGROUND: In the adult hippocampus new neurons are continuously generated from neural stem cells (NSCs) present at the subgranular zone of the dentate gyrus. This process is controlled by Wnt signaling, which plays a complex role in regulating multiple steps of neurogenesis including maintenance, proliferation and differentiation of progenitor cells and the development of newborn neurons. Differential effects of Wnt signaling during progression of neurogenesis could be mediated by cell-type specific expression of Wnt receptors. Here we studied the potential role of Frizzled-1 (FZD1) receptor in adult hippocampal neurogenesis. RESULTS: In the adult dentate gyrus, we determined that FZD1 is highly expressed in NSCs, neural progenitors and immature neurons. Accordingly, FZD1 is expressed in cultured adult hippocampal progenitors isolated from mouse brain. To evaluate the role of this receptor in vivo we targeted FZD1 in newborn cells using retroviral-mediated RNA interference. FZD1 knockdown resulted in a marked decrease in the differentiation of newborn cells into neurons and increased the generation of astrocytes, suggesting a regulatory role for the receptor in cell fate commitment. In addition, FZD1 knockdown induced an extended migration of adult-born neurons within the granule cell layer. However, no differences were observed in total dendritic length and dendritic arbor complexity between control and FZD1-deficient newborn neurons. CONCLUSIONS: Our results show that FZD1 regulates specific stages of adult hippocampal neurogenesis, being required for neuronal differentiation and positioning of newborn neurons into the granule cell layer, but not for morphological development of adult-born granule neurons.


Sujet(s)
Vieillissement/métabolisme , Récepteurs Frizzled/métabolisme , Hippocampe/métabolisme , Neurogenèse , Animaux , Animaux nouveau-nés , Différenciation cellulaire , Mouvement cellulaire , Dendrites/métabolisme , Gyrus denté/cytologie , Gyrus denté/métabolisme , Techniques de knock-down de gènes , Souris de lignée C57BL , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme
17.
J Neurochem ; 137(4): 506-17, 2016 05.
Article de Anglais | MEDLINE | ID: mdl-26875993

RÉSUMÉ

The generation of abnormally high levels of reactive oxygen species (ROS) is linked to cellular dysfunction, including neuronal toxicity and neurodegeneration. However, physiological ROS production modulates redox-sensitive roles of several molecules such as transcription factors, signaling proteins, and cytoskeletal components. Changes in the functions of redox-sensitive proteins may be important for defining key aspects of stem cell proliferation and differentiation, neuronal maturation, and neuronal plasticity. In neurons, most of the studies have been focused on the pathological implications of such modifications and only very recently their essential roles in neuronal development and plasticity has been recognized. In this review, we discuss the participation of NADPH oxidases (NOXs) and a family of protein-methionine sulfoxide oxidases, named molecule interacting with CasLs, as regulated enzymatic sources of ROS production in neurons, and describes the contribution of ROS signaling to neurogenesis and differentiation, neurite outgrowth, and neuronal plasticity. We review the role of reactive oxygen species (ROS) in neurogenesis, axon growth, and guidance and NMDA-receptor-mediated plasticity, LTP, and memory. ROS participation is presented in the context of NADPH oxidase and MICAL functions and their importance for brain functions.


Sujet(s)
Neurogenèse/physiologie , Plasticité neuronale/physiologie , Neurones/métabolisme , Espèces réactives de l'oxygène/métabolisme , Transduction du signal/physiologie , Animaux , Humains , Oxydoréduction
18.
PeerJ ; 3: e1486, 2015.
Article de Anglais | MEDLINE | ID: mdl-26713239

RÉSUMÉ

Oxygen concentration should be carefully regulated in all living tissues, beginning at the early embryonic stages. Unbalances in oxygen regulation can lead to cell death and disease. However, to date, few studies have investigated the consequences of variations in oxygen levels for fetal-like cells. Therefore, in the present work, human neural progenitor cells (NPCs) derived from pluripotent stem cells grown in 3% oxygen (v/v) were compared with NPCs cultured in 21% (v/v) oxygen. Low oxygen concentrations altered the mitochondrial content and oxidative functions of the cells, which led to improved ATP production, while reducing generation of reactive oxygen species (ROS). NPCs cultured in both conditions showed no differences in proliferation and glucose metabolism. Furthermore, antioxidant enzymatic activity was not altered in NPCs cultured in 3% oxygen under normal conditions, however, when exposed to external agents known to induce oxidative stress, greater susceptibility to DNA damage was observed. Our findings indicate that the management of oxygen levels should be considered for in vitro models of neuronal development and drug screening.

19.
Cytometry A ; 87(10): 929-35, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26189685

RÉSUMÉ

Neural stem cells proliferate and differentiate into neurons and glial cells, being responsible for embryonic and postnatal development of the central nervous system (CNS) as well as for regeneration in the adult brain. These cells also play a key role in maintaining the physiological integrity of the CNS in face of injury or disease. The previous study has demonstrated that bradykinin (BK) treatment simultaneously induces neuronal enrichment (indicating that BK contributes to neurogenesis) and reduced proliferation rates during in vitro differentiation of rat embryonic telencephalon neural precursor cells (NPCs). Here, we provide a mechanism for the unresolved question whether (i) the low rate of proliferation is owed to enhanced neurogenesis or, conversely, (ii) the alteration of the population ratio could result from low proliferation of NPCs and glial cells. In agreement with the previous study, BK promoted neuron-specific ß3-tubulin and MAP2 expression in differentiating embryonic mouse neurospheres, whereas glial protein expression and global proliferation rates decreased. Furthermore, BK augmented the global frequency of cells in G0 -phase of cell cycle after differentiation. Heterogeneous cell populations were observed at this stage, including neurons that always remaining a quiescent state (G0 -phase). It is noteworthy that BK did not interfere with proliferation of any particular cell type, evidenced by coimmunostaining for nestin, ß3-tubulin, glial fibrillary acidic protein (GFAP), and 5-ethynyl-2'-deoxyuridine (EdU). Thus, we conclude that neuronal enrichment is owing only to the fostering of neurogenesis, and that the low proliferation rate on the seventh day of differentiation is a consequence and not the cause of BK-induced neuronal enrichment.


Sujet(s)
Bradykinine/administration et posologie , Cellules souches neurales/effets des médicaments et des substances chimiques , Neurogenèse/génétique , Névroglie/effets des médicaments et des substances chimiques , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Système nerveux central/effets des médicaments et des substances chimiques , Système nerveux central/croissance et développement , Cytométrie en flux , Régulation de l'expression des gènes au cours du développement , Souris , Protéines associées aux microtubules/biosynthèse , Cellules souches neurales/cytologie , Névroglie/cytologie , Neurones/effets des médicaments et des substances chimiques , Rats , Récepteur de la bradykinine de type B2/métabolisme , Tubuline/biosynthèse
20.
J Neurosci Res ; 92(5): 607-18, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24452691

RÉSUMÉ

Neural progenitor cell (NPC) transplantation is a promising therapeutic strategy for spinal cord injury (SCI) because of the potential for cell replacement and restoration of connectivity. Our previous studies have shown that transplants of NPC, composed of neuron- and glia-restricted progenitors derived from the embryonic spinal cord, survived well in partial lesion models and generated graft-derived neurons, which could be used to form a functional relay. We have now examined the properties of a similar NPC transplant using a complete transection model in juvenile and adult rats. We found poor survival of grafted cells despite using a variety of lesion methods, matrices, and delays of transplantation. If, instead of cultured progenitor cells, the transplants were composed of segmental or dissociated segments of fetal spinal cord (FSC) derived from similar-staged embryos, grafted cells survived and integrated well with host tissue in juvenile and adult rats. FSC transplants differentiated into neurons and glial cells, including astrocytes and oligodendrocytes. Graft-derived neurons expressed glutaminergic and GABAergic markers. Grafted cells also migrated and extended processes into host tissue. Analysis of axon growth from the host spinal cord showed serotonin-positive fibers and biotinylated dextran amine-traced propriospinal axons growing into the transplants. These results suggest that in treating severe SCI, such as complete transection, NPC grafting faces major challenges related to cell survival and formation of a functional relay. Lessons learned from the efficacy of FSC transplants could be used to develop a therapeutic strategy based on neural progenitor cells for severe SCI.


Sujet(s)
Régénération nerveuse/physiologie , Traumatismes de la moelle épinière/chirurgie , Transplantation de cellules souches/méthodes , Cellules souches/physiologie , Facteurs âges , Animaux , Différenciation cellulaire , Mouvement cellulaire , Prolifération cellulaire , Choline O-acetyltransferase/métabolisme , Modèles animaux de maladie humaine , Embryon de mammifère , Femelle , Peptide libérant la gastrine/métabolisme , Protéines de tissu nerveux/métabolisme , Rats , Rats de lignée F344 , Rat Sprague-Dawley , Rats transgéniques , Sérotonine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE