Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 112
Filtrer
1.
Life Sci ; 350: 122750, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38801982

RÉSUMÉ

C-Jun-N-terminal-kinases (JNKs), members of the mitogen-activated-protein-kinase family, are significantly linked with neurological and neurodegenerative pathologies and cancer progression. However, JNKs serve key roles under physiological conditions, particularly within the central-nervous-system (CNS), where they are critical in governing neural proliferation and differentiation during both embryogenesis and adult stages. These processes control the development of CNS, avoiding neurodevelopment disorders. JNK are key to maintain the proper activity of neural-stem-cells (NSC) and neural-progenitors (NPC) that exist in adults, which keep the convenient brain plasticity and homeostasis. This review underscores how the interaction of JNK with upstream and downstream molecules acts as a regulatory mechanism to manage the self-renewal capacity and differentiation of NSC/NPC during CNS development and in adult neurogenic niches. Evidence suggests that JNK is reliant on non-canonical Wnt components, Fbw7-ubiquitin-ligase, and WDR62-scaffold-protein, regulating substrates such as transcription factors and cytoskeletal proteins. Therefore, understanding which pathways and molecules interact with JNK will bring knowledge on how JNK activation orchestrates neuronal processes that occur in CNS development and brain disorders.


Sujet(s)
Différenciation cellulaire , Cellules souches neurales , Neurogenèse , Humains , Animaux , Différenciation cellulaire/physiologie , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Neurogenèse/physiologie , Système de signalisation des MAP kinases/physiologie , JNK Mitogen-Activated Protein Kinases/métabolisme , Neurones/métabolisme , Neurones/cytologie
2.
Int J Mol Sci ; 25(10)2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38791505

RÉSUMÉ

In contrast to the hypothesis that aging results from cell-autonomous deterioration processes, the programmed longevity theory proposes that aging arises from a partial inactivation of a "longevity program" aimed at maintaining youthfulness in organisms. Supporting this hypothesis, age-related changes in organisms can be reversed by factors circulating in young blood. Concordantly, the endocrine secretion of exosomal microRNAs (miRNAs) by hypothalamic neural stem cells (htNSCs) regulates the aging rate by enhancing physiological fitness in young animals. However, the specific molecular mechanisms through which hypothalamic-derived miRNAs exert their anti-aging effects remain unexplored. Using experimentally validated miRNA-target gene interactions and single-cell transcriptomic data of brain cells during aging and heterochronic parabiosis, we identify the main pathways controlled by these miRNAs and the cell-type-specific gene networks that are altered due to age-related loss of htNSCs and the subsequent decline in specific miRNA levels in the cerebrospinal fluid (CSF). Our bioinformatics analysis suggests that these miRNAs modulate pathways associated with senescence and cellular stress response, targeting crucial genes such as Cdkn2a, Rps27, and Txnip. The oligodendrocyte lineage appears to be the most responsive to age-dependent loss of exosomal miRNA, leading to significant derepression of several miRNA target genes. Furthermore, heterochronic parabiosis can reverse age-related upregulation of specific miRNA-targeted genes, predominantly in brain endothelial cells, including senescence promoting genes such as Cdkn1a and Btg2. Our findings support the presence of an anti-senescence mechanism triggered by the endocrine secretion of htNSC-derived exosomal miRNAs, which is associated with a youthful transcriptional signature.


Sujet(s)
Vieillissement , Exosomes , Hypothalamus , microARN , Cellules souches neurales , microARN/génétique , microARN/métabolisme , Animaux , Exosomes/métabolisme , Hypothalamus/métabolisme , Vieillissement/génétique , Vieillissement/métabolisme , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Réseaux de régulation génique , Vieillissement de la cellule/génétique , Encéphale/métabolisme , Souris , Parabiose , Oligodendroglie/métabolisme , Transcriptome , Régulation de l'expression des gènes , Analyse de profil d'expression de gènes
3.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article de Anglais | MEDLINE | ID: mdl-34768919

RÉSUMÉ

The generation of new neurons in the adult brain is a currently accepted phenomenon. Over the past few decades, the subventricular zone and the hippocampal dentate gyrus have been described as the two main neurogenic niches. Neurogenic niches generate new neurons through an asymmetric division process involving several developmental steps. This process occurs throughout life in several species, including humans. These new neurons possess unique properties that contribute to the local circuitry. Despite several efforts, no other neurogenic zones have been observed in many years; the lack of observation is probably due to technical issues. However, in recent years, more brain niches have been described, once again breaking the current paradigms. Currently, a debate in the scientific community about new neurogenic areas of the brain, namely, human adult neurogenesis, is ongoing. Thus, several open questions regarding new neurogenic niches, as well as this phenomenon in adult humans, their functional relevance, and their mechanisms, remain to be answered. In this review, we discuss the literature and provide a compressive overview of the known neurogenic zones, traditional zones, and newly described zones. Additionally, we will review the regulatory roles of some molecular mechanisms, such as miRNAs, neurotrophic factors, and neurotrophins. We also join the debate on human adult neurogenesis, and we will identify similarities and differences in the literature and summarize the knowledge regarding these interesting topics.


Sujet(s)
Gyrus denté/cytologie , Ventricules latéraux/cytologie , Neurogenèse/physiologie , Neurones/cytologie , Striatum ventral/cytologie , Adulte , Animaux , Hippocampe/cytologie , Humains , Souris , microARN/génétique , Cellules souches neurales/cytologie , Neurogenèse/génétique , Rats
4.
Neurosci Lett ; 765: 136293, 2021 11 20.
Article de Anglais | MEDLINE | ID: mdl-34662661

RÉSUMÉ

Neurogenesis in adult mammals occurs mainly in the subventricular and subgranular areas of the brain, but there are also reports of its occurrence in the spinal cord. In a study on rats, neural stem cells and neuroprogenitor cells could be obtained through primary spinal cord culture, but there are no studies on these cells in canine species, to date. Dogs represent an appropriate animal model for studies on neurogenesis and neurological disorders. In addition, they are animals of great affective value, and the therapeutic use of neural stem cells can represent a breakthrough in regenerative veterinary medicine. Therefore, this study aimed to determine a protocol for the isolation, culture, and characterization of neural and neuroprogenitor stem cells derived from the spinal cord of canine fetuses. The cells were isolated from spinal cord fragments and cultured in serum-free culture medium supplemented with EGF and FGF-2 growth factors. These cells were observed daily by optical microscopy to analyze their morphological characteristics. From the third day in vitro, it was possible to observe translucent cell groupings, similar to the neurospheres, which approximately ranged from 50 µm to 200 µm at seven days in vitro. Throughout the culture period, the neurospheres developed ribbons in their periphery that migrated and communicated with other neurospheres. RT-PCR revealed that the cells expressed the characteristic genes SOX2, NESTIN, and GFAP. In addition to gene expression, the cells were phenotypically marked in the immunofluorescence assay for the proteins Nestin, GFAP, and ß-tubulin III, characterizing them as neurospheres. Our results suggest that the spinal cord may be a source of neural stem cells and neural progenitor cells in canine fetuses. These cells may be an interesting option for neurogenesis and neuroregenerative therapy studies.


Sujet(s)
Chiens , Cellules souches neurales/cytologie , Moelle spinale/cytologie , Animaux , Techniques de culture cellulaire , Foetus
5.
Biochem Biophys Res Commun ; 560: 152-158, 2021 06 30.
Article de Anglais | MEDLINE | ID: mdl-33989907

RÉSUMÉ

Dp71 and Dp40 are the main products of the DMD gene in the central nervous system, and they are developmentally regulated from the early stages of embryonic development to adulthood. To further study the roles of Dp71 and Dp40 during cell proliferation and neural differentiation, we analyzed Dp71/Dp40 isoform expression at the mRNA level by RT-PCR assays to identify alternative splicing (AS) in the isoforms expressed in rat neural stem/progenitor cells (NSPCs) and in differentiated cells (neurons and glia). We found that proliferating NSPCs expressed Dp71d, Dp71dΔ71, Dp71f, Dp71fΔ71, Dp71dΔ74 and Dp40, as well as two Dp40 isoforms: Dp40Δ63,64 and Dp40Δ64-67. In differentiated cells we also found the expression of Dp71d, Dp71dΔ71, Dp71f, Dp71fΔ71 and Dp40. However, the expression frequencies were different in both stages. In addition, in differentiated cells, we found Dp71fΔ71-74, and interestingly, we did not find the expression of Dp71dΔ74 or the newly identified Dp40 isoforms. In this work we show that NSPC differentiation is accompanied by changes in Dp71/Dp40 isoform expression, suggesting different roles for these isoforms in NSPCs proliferation and neuronal differentiation, and we describe, for the first time, alternative splicing of Dp40.


Sujet(s)
Épissage alternatif , Dystrophine/génétique , Cellules souches neurales/métabolisme , Animaux , Différenciation cellulaire , Prolifération cellulaire , Dystrophine/métabolisme , Cellules souches neurales/cytologie , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Isoformes d'ARN/métabolisme , Rat Wistar
6.
Methods Mol Biol ; 2174: 19-29, 2021.
Article de Anglais | MEDLINE | ID: mdl-32813242

RÉSUMÉ

Glioblastomas (GBM) are the most frequent and aggressive brain tumors due to their recurrence and resistance to current therapies. These characteristics are associated with the presence of glioma stem cells (GSCs), mainly identified by the detection of the membrane antigens CD133 and CD15. The main source of GSCs has been biopsies of tumors. However, alternatives are sought from cell lines because more homogeneous populations can be obtained with high yields. This chapter describes a method for the enrichment and characterization of GSCs from cell lines derived from human GBM by selective culture with serum-free neural stem cell medium and growth factors. The technique offers alternatives for the enrichment and characterization of GSCs, that could contribute to a better understanding of the biology of GBMs.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Régulation de l'expression des gènes tumoraux/génétique , Glioblastome/anatomopathologie , Cellules souches tumorales/anatomopathologie , RT-PCR/méthodes , Antigène AC133/analyse , Tumeurs du cerveau/génétique , Lignée cellulaire tumorale , Milieux de culture sans sérum/composition chimique , Milieux de culture sans sérum/pharmacologie , Cytométrie en flux , Glioblastome/génétique , Humains , Protéines et peptides de signalisation intercellulaire/pharmacologie , Antigènes CD15/analyse , Cellules souches tumorales/physiologie , Cellules souches neurales/cytologie
7.
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
8.
Mol Neurobiol ; 57(9): 3814-3826, 2020 Sep.
Article de Anglais | MEDLINE | ID: mdl-32592125

RÉSUMÉ

Neural stem cells can generate new neurons in the mouse adult brain in a complex multistep process called neurogenesis. Several factors regulate this process, including neurotransmitters, hormones, neurotrophic factors, pharmacological agents, and environmental factors. Purinergic signaling, mainly the adenosinergic system, takes part in neurogenesis, being involved in cell proliferation, migration, and differentiation. However, the role of the purine nucleoside guanosine in neurogenesis remains unclear. Here, we examined the effect of guanosine by using the neurosphere assay derived from neural stem cells of adult mice. We found that continuous treatment with guanosine increased the number of neurospheres, neural stem cell proliferation, and neuronal differentiation. The effect of guanosine to increase the number of neurospheres was reduced by removing adenosine from the culture medium. We next traced the neurogenic effect of guanosine in vivo. The intraperitoneal treatment of adult C57BL/6 mice with guanosine (8 mg/kg) for 26 days increased the number of dividing bromodeoxyuridine (BrdU)-positive cells and also increased neurogenesis, as identified by measuring doublecortin (DCX)-positive cells in the dentate gyrus (DG) of the hippocampus. Antidepressant-like behavior in adult mice accompanied the guanosine-induced neurogenesis in the DG. These results provide new evidence of a pro-neurogenic effect of guanosine on neural stem/progenitor cells, and it was associated in vivo with antidepressant-like effects.


Sujet(s)
Vieillissement/physiologie , Guanosine/pharmacologie , Hippocampe/cytologie , Cellules souches neurales/cytologie , Neurogenèse , Animaux , Antidépresseurs/pharmacologie , Comportement animal/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Gyrus denté/cytologie , Protéine doublecortine , Femelle , Mâle , Souris de lignée C57BL , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/métabolisme , Neurogenèse/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/effets des médicaments et des substances chimiques
9.
Cells ; 9(6)2020 05 26.
Article de Anglais | MEDLINE | ID: mdl-32466390

RÉSUMÉ

Neural precursor cells differentiate into several cell types that display distinct functions. However, little is known about how cell surface mechanics vary during the differentiation process. Here, by precisely measuring membrane tension and bending modulus, we map their variations and correlate them with changes in neural precursor cell morphology along their distinct differentiation fates. Both cells maintained in culture as neural precursors as well as those plated in neurobasal medium reveal a decrease in membrane tension over the first hours of culture followed by stabilization, with no change in bending modulus. During astrocyte differentiation, membrane tension initially decreases and then increases after 72 h, accompanied by consolidation of glial fibrillary acidic protein expression and striking actin reorganization, while bending modulus increases following observed alterations. For oligodendrocytes, the changes in membrane tension are less abrupt over the first hours, but their values subsequently decrease, correlating with a shift from oligodendrocyte marker O4 to myelin basic protein expressions and a remarkable actin reorganization, while bending modulus remains constant. Oligodendrocytes at later differentiation stages show membrane vesicles with similar membrane tension but higher bending modulus as compared to the cell surface. Altogether, our results display an entire spectrum of how membrane elastic properties are varying, thus contributing to a better understanding of neural differentiation from a mechanobiological perspective.


Sujet(s)
Différenciation cellulaire , Membrane cellulaire/physiologie , Élasticité , Cellules souches neurales/cytologie , Animaux , Astrocytes/cytologie , Marqueurs biologiques/métabolisme , Phénomènes biomécaniques , Cellules cultivées , Milieux de culture , Cytosquelette/métabolisme , Souris , Pinces optiques
10.
Brain Res Bull ; 157: 140-148, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-31945407

RÉSUMÉ

MicroRNAs (miRNAs) plays an important role in the human brain from the embryonic period to adulthood. In this sense, they influence the development of neural stem cells (NSCs), regulating cellular differentiation and survival. Therefore, due to the importance of better comprehending the regulation of miRNAs in NSCs differentiation and the lack of studies that show the panorama of miRNAs and their signaling pathways studied until now we aimed to systematically review the literature to identify which miRNAs are currently being associated with neuronal differentiation and using bioinformatics analysis to identify their related pathways. A search was carried out in the following databases: Scientific Electronic Library Online (Scielo), National Library of Medicine National Institutes of Health (PubMed), Scopus, Web of Science and Science Direct, using the descriptors "(microRNA [MeSH])" and "(neurogenesis [MeSH])". From the articles found, two independent and previously calibrated reviewers, using the EndNote X7 (Thomson Reuters, New York, NY, US), selected those that concern miRNA in the development of NSCs, based on in vitro studies. After, bioinformatic analysis was performed using the software DIANA Tools, mirPath v.3. Subsequently, data was tabulated, analyzed and interpreted. Among the 106 miRNAs cited by included studies, 55 were up-regulated and 47 were down-regulated. The bioinformatics analysis revealed that among the up-regulated miRNAs there were 24 total and 6 union pathways, and 3 presented a statistically significant difference (p ≤ 0.05). Among the down-regulated miRNAs, 46 total and 13 union pathways were found, with 7 presenting a significant difference (p ≤ 0.05). The miR-125a-5p, miR-423-5p, miR-320 were the most frequently found miRNAs in the pathways determined by bioinformatics. In this study a panel of altered miRNAs in neuronal differentiation was created with their related pathways, which could be a step towards understanding the complex network of miRNAs in neuronal differentiation.


Sujet(s)
Différenciation cellulaire/physiologie , Biologie informatique , Analyse de profil d'expression de gènes , microARN/métabolisme , Cellules souches neurales/cytologie , Animaux , Biologie informatique/méthodes , Humains , Neurogenèse/physiologie
11.
Curr Neurovasc Res ; 16(4): 291-300, 2019.
Article de Anglais | MEDLINE | ID: mdl-31633476

RÉSUMÉ

BACKGROUND: In the developing cerebral cortex, Radial Glia (RG) multipotent neural stem cell, among other functions, differentiate into astrocytes and serve as a scaffold for blood vessel development. After some time, blood vessel Endothelial Cells (ECs) become associated with astrocytes to form the neurovascular Blood-Brain Barrier (BBB) unit. OBJECTIVE: Since little is known about the mechanisms underlying bidirectional RG-ECs interactions in both vascular development and astrocyte differentiation, this study investigated the impact of interactions between RG and ECs mediated by secreted factors on EC maturation and gliogenesis control. METHODS: First, we demonstrated that immature vasculature in the murine embryonic cerebral cortex physically interacts with Nestin positive RG neural stem cells in vivo. Isolated Microcapillary Brain Endothelial Cells (MBEC) treated with the conditioned medium from RG cultures (RG-CM) displayed decreased proliferation, reduction in the protein levels of the endothelial tip cell marker Delta-like 4 (Dll4), and decreased expression levels of the vascular permeability associated gene, plasmalemma vesicle-associated protein-1 (PLVAP1). These events were also accompanied by increased levels of the tight junction protein expression, zonula occludens-1 (ZO-1). RESULTS: Finally, we demonstrated that isolated RG cells cultures treated with MBEC conditioned medium promoted the differentiation of astrocytes in a Vascular Endothelial Growth Factor-A (VEGF-A) dependent manner. CONCLUSION: These results suggest that the bidirectional interaction between RG and ECs is essential to induce vascular maturation and astrocyte generation, which may be an essential cell-cell communication mechanism to promote BBB establishment.


Sujet(s)
Astrocytes/cytologie , Barrière hémato-encéphalique/cytologie , Différenciation cellulaire/physiologie , Cellules endothéliales/cytologie , Animaux , Encéphale/cytologie , Encéphale/métabolisme , Perméabilité capillaire/physiologie , Cellules cultivées , Souris , Cellules souches neurales/cytologie , Neurogenèse/physiologie
12.
Front Immunol ; 10: 1928, 2019.
Article de Anglais | MEDLINE | ID: mdl-31474994

RÉSUMÉ

Zika virus (ZIKV) infection during pregnancy is associated with microcephaly, a congenital malformation resulting from neuroinflammation and direct effects of virus replication on the developing central nervous system (CNS). However, the exact changes in the affected CNS remain unknown. Here, we show by transcriptome analysis (at 48 h post-infection) and multiplex immune profiling that human induced-neuroprogenitor stem cells (hiNPCs) respond to ZIKV infection with a strong induction of type-I interferons (IFNs) and several type-I IFNs stimulated genes (ISGs), notably cytokines and the pro-apoptotic chemokines CXCL9 and CXCL10. By comparing the inflammatory profile induced by a ZIKV Brazilian strain with an ancestral strain isolated from Cambodia in 2010, we observed that the response magnitude differs among them. Compared to ZIKV/Cambodia, the experimental infection of hiNPCs with ZIKV/Brazil resulted in a diminished induction of ISGs and lower induction of several cytokines (IFN-α, IL-1α/ß, IL-6, IL-8, and IL-15), consequently favoring virus replication. From ZIKV-confirmed infant microcephaly cases, we detected a similar profile characterized by the presence of IFN-α, CXCL10, and CXCL9 in cerebrospinal fluid (CSF) samples collected after birth, evidencing a sustained CNS inflammation. Altogether, our data suggest that the CNS may be directly affected due to an unbalanced and chronic local inflammatory response, elicited by ZIKV infection, which contributes to damage to the fetal brain.


Sujet(s)
Système nerveux central/immunologie , Cellules souches pluripotentes induites/cytologie , Microcéphalie/immunologie , Cellules souches neurales/cytologie , Virus Zika/immunologie , Brésil , Cambodge , Cellules cultivées , Système nerveux central/anatomopathologie , Système nerveux central/virologie , Chimiokine CXCL10/liquide cérébrospinal , Chimiokine CXCL10/immunologie , Chimiokine CXCL9/liquide cérébrospinal , Chimiokine CXCL9/immunologie , Cytokines/analyse , Femelle , Analyse de profil d'expression de gènes , Humains , Nourrisson , Inflammation/immunologie , Inflammation/anatomopathologie , Interféron alpha/liquide cérébrospinal , Interféron alpha/immunologie , Interféron bêta/immunologie , Mâle , Microcéphalie/anatomopathologie , Grossesse , Complications infectieuses de la grossesse/virologie , Réplication virale/immunologie , Infection par le virus Zika/immunologie
13.
Stem Cell Res Ther ; 10(1): 146, 2019 05 21.
Article de Anglais | MEDLINE | ID: mdl-31113444

RÉSUMÉ

BACKGROUND: Survival and therapeutic actions of bone marrow-derived mesenchymal stem cells (BMMSCs) can be limited by the hostile microenvironment present during acute spinal cord injury (SCI). Here, we investigated whether BMMSCs overexpressing insulin-like growth factor 1 (IGF-1), a cytokine involved in neural development and injury repair, improved the therapeutic effects of BMMSCs in SCI. METHODS: Using a SCI contusion model in C57Bl/6 mice, we transplanted IGF-1 overexpressing or wild-type BMMSCs into the lesion site following SCI and evaluated cell survival, proliferation, immunomodulation, oxidative stress, myelination, and functional outcomes. RESULTS: BMMSC-IGF1 transplantation was associated with increased cell survival and recruitment of endogenous neural progenitor cells compared to BMMSC- or saline-treated controls. Modulation of gene expression of pro- and anti-inflammatory mediators was observed after BMMSC-IGF1 and compared to saline- and BMMSC-treated mice. Treatment with BMMSC-IGF1 restored spinal cord redox homeostasis by upregulating antioxidant defense genes. BMMSC-IGF1 protected against SCI-induced myelin loss, showing more compact myelin 28 days after SCI. Functional analyses demonstrated significant gains in BMS score and gait analysis in BMMSC-IGF1, compared to BMMSC or saline treatment. CONCLUSIONS: Overexpression of IGF-1 in BMMSC resulted in increased cell survival, immunomodulation, myelination, and functional improvements, suggesting that IGF-1 facilitates the regenerative actions of BMMSC in acute SCI.


Sujet(s)
Facteur de croissance IGF-I/génétique , Transplantation de cellules souches mésenchymateuses , Cellules souches neurales/transplantation , Traumatismes de la moelle épinière/thérapie , Animaux , Cellules de la moelle osseuse/cytologie , Différenciation cellulaire/génétique , Modèles animaux de maladie humaine , Humains , Cellules souches mésenchymateuses/cytologie , Souris , Gaine de myéline/génétique , Gaine de myéline/anatomopathologie , Cellules souches neurales/cytologie , Récupération fonctionnelle , Régénération/génétique , Traumatismes de la moelle épinière/génétique , Traumatismes de la moelle épinière/anatomopathologie
14.
Stem Cells Dev ; 28(16): 1104-1115, 2019 08 15.
Article de Anglais | MEDLINE | ID: mdl-31140356

RÉSUMÉ

Transplantation of dopaminergic (DA) cells into the striatum can rescue from dopamine deficiency in a Parkinson's disease condition, but this is not a suitable procedure for regaining the full control of motor activity. The minimal condition toward recovering the nigrostriatal pathway is the proper innervation of transplanted DA neurons or their precursors from the substancia nigra pars compacta (SNpc) to their target areas. However, functional integration of transplanted cells would require first that the host SNpc is suitable for their survival and/or differentiation. We recently reported that the intact adult SNpc holds a strong neurogenic environment, but primed embryonic stem cells (ie, embryoid body cells, EBCs) could not derive into DA neurons. In this study, we transplanted into the intact or lesioned SNpc, EBCs derived from embryonic stem cells that were prompt to differentiate into DA neurons by the forced expression of Lmx1a in neural precursor cells (R1B5/NesE-Lmx1a). We observed that, 6 days posttransplantation (dpt), R1B5 or R1B5/NesE-Lmx1a EBCs gave rise to Nes+ and Dcx+ cells within the host SNpc, but a large number of Th+ cells derived only from EBCs exogenously expressing Lmx1a. In contrast, when transplantation was carried out into the 6-hydroxidopamine-lesioned SNpc, the emergence of Th+ cells from EBCs was independent of exogenous Lmx1a expression, although these cells were not found by 15 dpt. These results suggest that the adult SNpc is not only a permissive niche for initiation of DA differentiation of non-neuralized cells but also releases factors upon damage that promote the acquisition of DA characteristics by transplanted EBCs.


Sujet(s)
Différenciation cellulaire/physiologie , Dopamine/métabolisme , Cellules souches embryonnaires/cytologie , Substantia nigra/cytologie , Animaux , Cellules cultivées , Corps strié/cytologie , Corps strié/métabolisme , Neurones dopaminergiques/cytologie , Neurones dopaminergiques/métabolisme , Protéine doublecortine , Cellules souches embryonnaires/métabolisme , Souris , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Neurogenèse/physiologie , Maladie de Parkinson/métabolisme , Substantia nigra/métabolisme , Facteurs de transcription/métabolisme
15.
J Chem Neuroanat ; 98: 117-123, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-31047946

RÉSUMÉ

In the adult hippocampus new neurons are generated in the dentate gyrus from neural progenitor cells. Adult-born neurons integrate into the hippocampal circuitry and contribute to hippocampal function. PSD95 is a major postsynaptic scaffold protein that is crucial for morphological maturation and synaptic development of hippocampal neurons. Here we study the function of PSD95 in adult hippocampal neurogenesis by downregulating PSD95 expression in newborn cells using retroviral-mediated RNA interference. Retroviruses coding for a control shRNA or an shRNA targeting PSD95 (shPSD95) were stereotaxically injected into the dorsal dentate gyrus of 2-month-old C57BL/6 mice. PSD95 knockdown did not affect neuronal differentiation of newborn cells into neurons, or migration of newborn neurons into the granule cell layer. Morphological analysis revealed that newborn neurons expressing shPSD95 showed increased dendritic length and increased number of high-order dendrites. Concomitantly, dendrites from shPSD95-expressing newborn granule neurons showed a reduction in the density of dendritic spines. These results suggest that PSD95 is required for proper dendritic and spine maturation of adult-born neurons, but not for early stages of neurogenesis in the hippocampus.


Sujet(s)
Homologue-4 de la protéine Disks Large/métabolisme , Hippocampe/cytologie , Cellules souches neurales/cytologie , Neurogenèse/physiologie , Neurones/cytologie , Cellules souches adultes/cytologie , Cellules souches adultes/métabolisme , Animaux , Différenciation cellulaire/physiologie , Mouvement cellulaire/physiologie , Hippocampe/métabolisme , Souris , Souris de lignée C57BL , Cellules souches neurales/métabolisme , Neurones/métabolisme
16.
Methods Mol Biol ; 1919: 97-118, 2019.
Article de Anglais | MEDLINE | ID: mdl-30656624

RÉSUMÉ

The work with midbrain dopaminergic neurons (mDAN) differentiation might seem to be hard. There are about 40 different published protocols for mDAN differentiation, which are eventually modified according to the respective laboratory. In many cases, protocols are not fully described, failing to provide essential tips for researchers starting in the field. Considering that commercial kits produce low mDAN percentages (20-50%), we chose to follow a mix of four main protocols based on Kriks and colleagues' protocol, from which the resulting mDAN were engrafted with success in three different animal models of Parkinson's disease. We present a differential step-by-step methodology for generating mDAN directly from human-induced pluripotent stem cells cultured with E8 medium on Geltrex, without culture on primary mouse embryonic fibroblasts prior to mDAN differentiation, and subsequent exposure of neurons to rock inhibitor during passages for improving cell viability. The protocol described here allows obtaining mDAN with phenotypical and functional characteristics suitable for in vitro modeling, cell transplantation, and drug screening.


Sujet(s)
Différenciation cellulaire , Neurones dopaminergiques/cytologie , Cellules souches pluripotentes induites/cytologie , Mésencéphale/cytologie , Animaux , Marqueurs biologiques , Calcium/métabolisme , Signalisation calcique , Techniques de culture cellulaire , Séparation cellulaire , Cellules cultivées , Neurones dopaminergiques/métabolisme , Humains , Cellules souches pluripotentes induites/métabolisme , Mésencéphale/métabolisme , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Maladie de Parkinson
17.
Semin Cell Dev Biol ; 95: 42-53, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-30639325

RÉSUMÉ

In mammals, new neurons can be generated from neural stem cells in specific regions of the adult brain. Neural stem cells are characterized by their abilities to differentiate into all neural lineages and to self-renew. The specific microenvironments regulating neural stem cells, commonly referred to as neurogenic niches, comprise multiple cell populations whose precise contributions are under active current exploration. Understanding the cross-talk between neural stem cells and their niche components is essential for the development of therapies against neurological disorders in which neural stem cells function is altered. In this review, we describe and discuss recent studies that identified novel components in the neural stem cell niche. These discoveries bring new concepts to the field. Here, we evaluate these recent advances that change our understanding of the neural stem cell niche heterogeneity and its influence on neural stem cell function.


Sujet(s)
Cellules souches neurales/cytologie , Niche de cellules souches , Animaux , Communication autocrine , Liquide cérébrospinal/cytologie , Humains , Cellules souches neurales/métabolisme , Neurones/cytologie , Neurones/métabolisme , Transduction du signal
18.
Mol Biol Rep ; 46(1): 191-198, 2019 Feb.
Article de Anglais | MEDLINE | ID: mdl-30421128

RÉSUMÉ

Cell transplantation offers a promising approach in many neurological disorders. Neural stem (NS) cells are potential candidates for cell therapy. The ability to track the grafted cells in the host tissue will refine this therapy. Superparamagnetic iron oxide nanoparticles (SPION) have been suggested as a feasible method, but there is no consensus about its safety. Here we investigated the feasibility of label NS cells with SPION and track by MRI after transplantation into mouse striatum with SPION cells and its therapeutic effects by grafting the cells into mouse striatum. We demonstrated that SPION-labeled NS cells display normal patterns of cellular processes including proliferation, migration, differentiation and neurosphere formation. Transmission electron microscopy reveals SPION in the cytoplasm of the cells, which was confirmed by microanalysis. Neurons and astrocytes generated from SPION-labeled NS cells were able to carry nanoparticles after 7 days under differentiation. SPION-labeled NS cells transplanted into striatum of mice were detected by magnetic resonance imaging (MRI) and microscopy 51 days later. In agreement with others reports, we demonstrated that NS cells are able to incorporate SPION in vitro without altering the stemness, and can survive and be tracked by MRI after they have been grafted into mice striatum.


Sujet(s)
Suivi cellulaire/méthodes , Nanoparticules de magnétite/composition chimique , Cellules souches neurales/physiologie , Animaux , Différenciation cellulaire , Survie cellulaire , Cellules cultivées , Composés du fer III/métabolisme , Fer/métabolisme , Imagerie par résonance magnétique/méthodes , Souris , Microscopie électronique à transmission/méthodes , Cellules souches neurales/cytologie , Neurones/physiologie
19.
Semin Cell Dev Biol ; 95: 54-73, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-29782926

RÉSUMÉ

Oligodendrocytes are fundamental for the functioning of the nervous system; they participate in several cellular processes, including axonal myelination and metabolic maintenance for astrocytes and neurons. In the mammalian nervous system, they are produced through waves of proliferation and differentiation, which occur during embryogenesis. However, oligodendrocytes and their precursors continue to be generated during adulthood from specific niches of stem cells that were not recruited during development. Deficiencies in the formation and maturation of these cells can generate pathologies mainly related to myelination. Understanding the mechanisms involved in oligodendrocyte development, from the precursor to mature cell level, will allow inferring therapies and treatments for associated pathologies and disorders. Such mechanisms include cell signalling pathways that involve many growth factors, small metabolic molecules, non-coding RNAs, and transcription factors, as well as specific elements of the extracellular matrix, which act in a coordinated temporal and spatial manner according to a given stimulus. Deciphering those aspects will allow researchers to replicate them in vitro in a controlled environment and thus mimic oligodendrocyte maturation to understand the role of oligodendrocytes in myelination in pathologies and normal conditions. In this study, we review these aspects, based on the most recent in vivo and in vitro data on oligodendrocyte generation and differentiation.


Sujet(s)
Différenciation cellulaire , Oligodendroglie/cytologie , Oligodendroglie/métabolisme , Transduction du signal , Animaux , Matrice extracellulaire/métabolisme , Humains , Gaine de myéline/métabolisme , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme
20.
Eur J Neurosci ; 48(12): 3514-3533, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30402991

RÉSUMÉ

Understanding endogenous neurogenesis and neuronal replacement to mature circuits is a topic of discussion as a therapeutic alternative under acute and chronic neurodegenerative disorders. Adaptive neurogenic response may result as a result of ischemia which could support long-term recovery of behavioral functions. Endogenous sources of neural progenitors may be stimulated by changes in blood flow or neuromodulation. Using a mouse model of unilateral cortical devascularization, we have observed reactive neurogenesis in the perilesional cortex and subventricular zone neurogenic niche. C57BL/6L 4 weeks old male mice were craneotomized at 1 mm caudal from frontal suture and 1 mm lateral from midline to generate a window of 3 mm side. Brain injury was produced by removal of the meninges and superficial vasculature of dorsal parietal cortex. BrdU agent (50 mg/kg, ip) was injected to lesioned and sham animals, during days 0 and 1 after surgery. Sagittal sections were analyzed at 1, 4, 7, and 10 days post-injury. A time-dependent increase in BrdU+ cells in the perilesional parietal cortex was accompanied by augmented BrdU+ cells in the sub ventricular and rostral migratory stream of ipsilateral and contralateral hemispheres. Neural progenitors and neuroblasts proliferated in the lesioned and non-lesioned subventricular zone and rostral migratory stream on day 4 after injury. Augmented contralateral neurogenesis was associated with an increase in vesicular monoamine transporter 2 protein in the striosomal sub ventricular neurogenic niche of non-lesioned hemisphere.


Sujet(s)
Encéphalopathie ischémique/anatomopathologie , Cortex cérébral/métabolisme , Dopamine/métabolisme , Neurogenèse/physiologie , Transmission synaptique/physiologie , Animaux , Lésions encéphaliques/métabolisme , Encéphalopathie ischémique/métabolisme , Différenciation cellulaire/physiologie , Mouvement cellulaire/physiologie , Prolifération cellulaire/physiologie , Modèles animaux de maladie humaine , Mâle , Souris de lignée C57BL , Cellules souches neurales/cytologie , Neurones/cytologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE