Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Methods Mol Biol ; 2520: 335-360, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35579839

RESUMO

The unique properties of stem cells to self-renew and differentiate hold great promise in disease modelling and regenerative medicine. However, more information about basic stem cell biology and thorough characterization of available stem cell lines is needed. This is especially essential to ensure safety before any possible clinical use of stem cells or partially committed cell lines. As proteins are the key effector molecules in the cell, the proteomic characterization of cell lines, cell compartments or cell secretome and microenvironment is highly beneficial to answer above mentioned questions. Nowadays, method of choice for large-scale discovery-based proteomic analysis is mass spectrometry (MS) with data-independent acquisition (DIA). DIA is a robust, highly reproducible, high-throughput quantitative MS approach that enables relative quantification of thousands of proteins in one sample. In the current protocol, we describe a specific variant of DIA known as SWATH-MS for characterization of neural stem cell differentiation. The protocol covers the whole process from cell culture, sample preparation for MS analysis, the SWATH-MS data acquisition on TTOF 5600, the complete SWATH-MS data processing and quality control using Skyline software and the basic statistical analysis in R and MSstats package. The protocol for SWATH-MS data acquisition and analysis can be easily adapted to other samples amenable to MS-based proteomics.


Assuntos
Células-Tronco Neurais , Proteômica , Software , Diferenciação Celular , Humanos , Espectrometria de Massas/métodos , Células-Tronco Neurais/química , Células-Tronco Neurais/metabolismo , Proteoma/análise , Proteômica/métodos , Controle de Qualidade
2.
Cell Commun Signal ; 20(1): 66, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35585601

RESUMO

Notch signalling pathway plays a key role in metazoan biology by contributing to resolution of binary decisions in the life cycle of cells during development. Outcomes such as proliferation/differentiation dichotomy are resolved by transcriptional remodelling that follows a switch from Notchon to Notchoff state, characterised by dissociation of Notch intracellular domain (NICD) from DNA-bound RBPJ. Here we provide evidence that transitioning to the Notchoff state is regulated by heat flux, a phenomenon that aligns resolution of fate dichotomies to mitochondrial activity. A combination of phylogenetic analysis and computational biochemistry was utilised to disclose structural adaptations of Notch1 ankyrin domain that enabled function as a sensor of heat flux. We then employed DNA-based micro-thermography to measure heat flux during brain development, followed by analysis in vitro of the temperature-dependent behaviour of Notch1 in mouse neural progenitor cells. The structural capacity of NICD to operate as a thermodynamic sensor in metazoans stems from characteristic enrichment of charged acidic amino acids in ß-hairpins of the ankyrin domain that amplify destabilising inter-residue electrostatic interactions and render the domain thermolabile. The instability emerges upon mitochondrial activity which raises the perinuclear and nuclear temperatures to 50 °C and 39 °C, respectively, leading to destabilization of Notch1 transcriptional complex and transitioning to the Notchoff state. Notch1 functions a metazoan thermodynamic sensor that is switched on by intercellular contacts, inputs heat flux as a proxy for mitochondrial activity in the Notchon state via the ankyrin domain and is eventually switched off in a temperature-dependent manner. Video abstract.


Assuntos
Anquirinas , Células-Tronco Neurais , Receptores Notch , Animais , Anquirinas/química , Anquirinas/metabolismo , Camundongos , Células-Tronco Neurais/química , Células-Tronco Neurais/metabolismo , Filogenia , Domínios Proteicos , Receptores Notch/química , Receptores Notch/metabolismo , Transdução de Sinais , Termodinâmica
3.
Bioengineered ; 13(1): 1062-1072, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34990302

RESUMO

Recent studies suggested that propofol, one of the most widely used anesthetics, may cause neurotoxicity in the developing brain, leading to cognitive deficits in adults. However, the underlying mechanisms remain unclear. In this study, we aimed to evaluate the mechanisms of propofol neurotoxicity in the neural stem cells (NSCs). The mRNA and protein expression levels of microRNA-9-5p (miR-9-5p) and chemokine CXC receptor 4 (CXCR4) were determined by quantitative reverse transcription-polymerase chain reaction and Western blotting analyses. Cell viability and apoptosis were evaluated using the cell counting kit-8 and Hoechst staining kits. The levels of apoptosis-related proteins B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein, and caspase-3 were detected by Western blotting analysis. These results confirmed that propofol activated cell apoptosis in a dose-dependent manner. A significant increase in the miR-9-5p and CXCR4 expression was observed in the propofol-treated cells. The overexpression of miR-9-5p induced apoptosis in NSCs, accompanied by elevated apoptosis-related protein activity. Furthermore, mitigated CXCR4 expression reduced propofol-induced cell apoptosis. We conclude that propofol induces cell death in NSCs, and overexpression of miR-9-5p/CXCR4 contributes to propofol-induced cell apoptosis, which might be a target for developing novel strategies to treat propofol neurotoxicity.


Assuntos
MicroRNAs/genética , Células-Tronco Neurais/citologia , Propofol/efeitos adversos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Células-Tronco Neurais/química , Células-Tronco Neurais/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima , Proteína X Associada a bcl-2/metabolismo
4.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-34948008

RESUMO

Recent studies have begun to reveal surprising levels of cell diversity in the human brain, both in adults and during development. Distinctive cellular phenotypes point to complex molecular profiles, cellular hierarchies and signaling pathways in neural stem cells, progenitor cells, neuronal and glial cells. Several recent reports have suggested that neural stem and progenitor cell types found in the developing and adult brain share several properties and phenotypes with cells from brain primary tumors, such as gliomas. This transcriptomic crosstalk may help us to better understand the cell hierarchies and signaling pathways in both gliomas and the normal brain, and, by clarifying the phenotypes of cells at the origin of the tumor, to therapeutically address their most relevant signaling pathways.


Assuntos
Neoplasias Encefálicas/genética , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Glioma/genética , Células-Tronco Neurais/química , Neoplasias Encefálicas/patologia , Comunicação Celular , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Células-Tronco Neoplásicas/química , Células-Tronco Neoplásicas/patologia , Fenótipo , Transdução de Sinais , Telencéfalo/química , Telencéfalo/citologia , Telencéfalo/patologia
5.
Elife ; 102021 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34259628

RESUMO

The ventricular-subventricular zone (V-SVZ), on the walls of the lateral ventricles, harbors the largest neurogenic niche in the adult mouse brain. Previous work has shown that neural stem/progenitor cells (NSPCs) in different locations within the V-SVZ produce different subtypes of new neurons for the olfactory bulb. The molecular signatures that underlie this regional heterogeneity remain largely unknown. Here, we present a single-cell RNA-sequencing dataset of the adult mouse V-SVZ revealing two populations of NSPCs that reside in largely non-overlapping domains in either the dorsal or ventral V-SVZ. These regional differences in gene expression were further validated using a single-nucleus RNA-sequencing reference dataset of regionally microdissected domains of the V-SVZ and by immunocytochemistry and RNAscope localization. We also identify two subpopulations of young neurons that have gene expression profiles consistent with a dorsal or ventral origin. Interestingly, a subset of genes are dynamically expressed, but maintained, in the ventral or dorsal lineages. The study provides novel markers and territories to understand the region-specific regulation of adult neurogenesis.


Nerve cells, or neurons, are the central building blocks of brain circuits. Their damage, death or loss of function leads to cognitive decline. Neural stem/progenitor cells (NSPCs) first appear during embryo development, generating most of the neurons found in the nervous system. However, the adult brain retains a small subpopulation of NSPCs, which in some species are an important source of new neurons throughout life. In the adult mouse brain the largest population of NSPCs, known as B cells, is found in an area called the ventricular-subventricular zone (V-SVZ). These V-SVZ B cells have properties of specialized support cells known as astrocytes, but they can also divide and generate intermediate 'progenitor cells' called C cells. These, in turn, divide to generate large numbers of young 'A cells' neurons that undertake a long and complex migration from V-SVZ to the olfactory bulb, the first relay in the central nervous system for the processing of smells. Depending on their location in the V-SVZ, B cells can generate different kinds of neurons, leading to at least ten subtypes of neurons. Why this is the case is still poorly understood. To examine this question, Cebrián-Silla, Nascimento, Redmond, Mansky et al. determined which genes were expressed in B, C and A cells from different parts of the V-SVZ. While cells within each of these populations had different expression patterns, those that originated in the same V-SVZ locations shared a set of genes, many of which associated with regional specification in the developing brain. Some, however, were intriguingly linked to hormonal regulation. Salient differences between B cells depended on whether the cells originated closer to the top ('dorsal' position) or to the bottom of the brain ('ventral' position). This information was used to stain slices of mouse brains for the RNA and proteins produced by these genes in different regions. These experiments revealed dorsal and ventral territories containing B cells with distinct 'gene expression'. This study highlights the heterogeneity of NSPCs, revealing key molecular differences among B cells in dorsal and ventral areas of the V-SVZ and reinforcing the concept that the location of NSPCs determines the types of neuron they generate. Furthermore, the birth of specific types of neurons from B cells that are so strictly localized highlights the importance of neuronal migration to ensure that young neurons with specific properties reach their appropriate destination in the olfactory bulb. The work by Cebrián-Silla, Nascimento, Redmond, Mansky et al. has identified sets of genes that are differentially expressed in dorsal and ventral regions which may contribute to regional regulation. Furthering the understanding of how adult NSPCs differ according to their location will help determine how various neuron types emerge in the adult brain.


Assuntos
Ventrículos Laterais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Transcriptoma/genética , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Microdissecção , Células-Tronco Neurais/química , Células-Tronco Neurais/citologia , Análise de Célula Única
6.
Theranostics ; 11(13): 6507-6521, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995671

RESUMO

Ischemic stroke remains a major cause of death, and anti-inflammatory strategies hold great promise for preventing major brain injury during reperfusion. In the past decade, stem cell-derived extracellular vesicles (EVs) have emerged as novel therapeutic effectors in immune modulation. However, the intravenous delivery of EVs into the ischemic brain remains a challenge due to poor targeting of unmodified EVs, and the costs of large-scale production of stem cell-derived EVs hinder their clinical application. Methods: EVs were isolated from a human neural progenitor cell line, and their anti-inflammatory effects were verified in vitro. To attach targeting ligands onto EVs, we generated a recombinant fusion protein containing the arginine-glycine-aspartic acid (RGD)-4C peptide (ACDCRGDCFC) fused to the phosphatidylserine (PS)-binding domains of lactadherin (C1C2), which readily self-associates onto the EV membrane. Subsequently, in a middle cerebral artery occlusion (MCAO) mouse model, the RGD-C1C2-bound EVs (RGD-EV) were intravenously injected through the tail vein, followed by fluorescence imaging and assessment of proinflammatory cytokines expression and microglia activation. Results: The neural progenitor cell-derived EVs showed intrinsic anti-inflammatory activity. The RGD-EV targeted the lesion region of the ischemic brain after intravenous administration, and resulted in a strong suppression of the inflammatory response. Furthermore, RNA sequencing revealed a set of 7 miRNAs packaged in the EVs inhibited MAPK, an inflammation related pathway. Conclusion: These results point to a rapid and easy strategy to produce targeting EVs and suggest a potential therapeutic agent for ischemic stroke.


Assuntos
Isquemia Encefálica/terapia , Vesículas Extracelulares/fisiologia , Infarto da Artéria Cerebral Média/terapia , Inflamação/prevenção & controle , Células-Tronco Neurais/citologia , Animais , Antígenos de Superfície/química , Antígenos de Superfície/farmacologia , Isquemia Encefálica/complicações , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Genes Reporter , Células HEK293 , Humanos , Infarto da Artéria Cerebral Média/complicações , Inflamação/etiologia , Injeções Intravenosas , Lipopolissacarídeos/toxicidade , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteínas do Leite/química , Proteínas do Leite/farmacologia , Nanopartículas , Células-Tronco Neurais/química , Oligopeptídeos/farmacologia , Fosfatidilserinas/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia
7.
J Environ Sci (China) ; 101: 72-86, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33334539

RESUMO

Intensive agriculture activities, industrialization and growing numbers of wastewater treatment plants along river banks collectively contribute to the elevated levels of neurotoxic pollutants in natural water reservoirs across Europe. We established an in vitro bioassay based upon neural stem cells isolated from the subventricular zone of the postnatal mouse to evaluate the neurotoxic potential of raw wastewater, treated sewage effluent, groundwater and drinking water. The toxic potential of water samples was evaluated employing viability, proliferation, differentiation and migration assays. We found that raw wastewater could reduce the viability and proliferation of neural stem cells, and decreased the neuronal and astrocyte differentiation, neuronal neurite growth, astrocyte growth and cell migration. Treated sewage water also showed inhibitory effects on cell proliferation and migration. Our results indicated that relatively high concentrations of nitrogenous substances, pesticides, mercuric compounds, bisphenol-A, and phthalates, along with some other pollutants in raw wastewater and treated sewage water, might be the reason for the neuroinhibitory effects of these water samples. Our model successfully predicted the neurotoxicity of water samples collected from different sources and also revealed that the incomplete removal of contaminants from wastewater can be problematic for the developing nervous system. The presented data also provides strong evidence that more effective treatments should be used to minimize the contamination of water before release into major water bodies which may be considered as water reservoirs for human usage in the future.


Assuntos
Células-Tronco Neurais , Poluentes Químicos da Água , Animais , Bioensaio , Monitoramento Ambiental , Camundongos , Células-Tronco Neurais/química , Esgotos/análise , Águas Residuárias , Água , Poluentes Químicos da Água/análise , Poluentes Químicos da Água/toxicidade
8.
Environ Pollut ; 270: 116179, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33348142

RESUMO

The developing nervous system is highly vulnerable to environmental toxicants especially pesticides. Glyphosate pesticide induces neurotoxicity both in humans and rodents, but so far only when exposed to higher concentrations. A few studies, however, have also reported the risk of general toxicity of glyphosate at concentrations comparable to allowable limits set up by environmental protection authorities. In vitro data regarding glyphosate neurotoxicity at concentrations comparable to maximum permissible concentrations in drinking water is lacking. In the present study, we established an in vitro assay based upon neural stem cells (NSCs) from the subventricular zone of the postnatal mouse to decipher the effects of two maximum permissible concentrations of glyphosate in drinking water on the basic neurogenesis processes. Our results demonstrated that maximum permissible concentrations of glyphosate recognized by environmental protection authorities significantly reduced the cell migration and differentiation of NSCs as demonstrated by the downregulation of the expression levels of the neuronal ß-tubulin III and the astrocytic S100B genes. The expression of the cytoprotective gene CYP1A1 was downregulated whilst the expression of oxidative stresses indicator gene SOD1 was upregulated. The concentration comparable to non-toxic human plasma concentration significantly induced cytotoxicity and activated Ca2+ signalling in the differentiated culture. Our findings demonstrated that the permissible concentrations of glyphosate in drinking water recognized by environmental protection authorities are capable of inducing neurotoxicity in the developing nervous system.


Assuntos
Água Potável , Herbicidas , Células-Tronco Neurais , Animais , Água Potável/análise , Glicina/análogos & derivados , Glicina/toxicidade , Herbicidas/análise , Ventrículos Laterais/química , Camundongos , Células-Tronco Neurais/química , Glifosato
9.
Reprod Toxicol ; 100: 17-34, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33333158

RESUMO

Halogenated persistent organic pollutants (POPs) like perfluorinated alkylated substances (PFASs), brominated flame retardants (BFRs), organochlorine pesticides and polychlorinated biphenyls (PCBs) are known to cause cancer, immunotoxicity, neurotoxicity and interfere with reproduction and development. Concerns have been raised about the impact of POPs upon brain development and possibly neurodevelopmental disorders. The developing brain is a particularly vulnerable organ due to dynamic and complex neurodevelopmental processes occurring early in life. However, very few studies have reported on the effects of POP mixtures at human relevant exposures, and their impact on key neurodevelopmental processes using human in vitro test systems. Aiming to reduce this knowledge gap, we exposed mixed neuronal/glial cultures differentiated from neural stem cells (NSCs) derived from human induced pluripotent stem cells (hiPSCs) to reconstructed mixtures of 29 different POPs using concentrations comparable to Scandinavian human blood levels. Effects of the POP mixtures on neuronal proliferation, differentiation and synaptogenesis were evaluated using in vitro assays anchored to common key events identified in the existing developmental neurotoxicity (DNT) adverse outcome pathways (AOPs). The present study showed that mixtures of POPs (in particular brominated and chlorinated compounds) at human relevant concentrations increased proliferation of NSCs and decreased synapse number. Based on a mathematical modelling, synaptogenesis and neurite outgrowth seem to be the most sensitive DNT in vitro endpoints. Our results indicate that prenatal exposure to POPs may affect human brain development, potentially contributing to recently observed learning and memory deficits in children.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Halogenação , Células-Tronco Neurais/fisiologia , Poluentes Orgânicos Persistentes/toxicidade , Sinapses/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Fator Neurotrófico Derivado do Encéfalo/análise , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Teóricos , Células-Tronco Neurais/química , Neuritos/efeitos dos fármacos , Transtornos do Neurodesenvolvimento/induzido quimicamente , Poluentes Orgânicos Persistentes/sangue , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Receptores de Hidrocarboneto Arílico/genética
10.
Biotechnol Bioeng ; 117(9): 2741-2748, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32572957

RESUMO

Neural progenitor cells (NPCs) are considered to be a promising source for stem cell-based regenerative therapy for central nervous disorders. However, the widespread clinical application of NPCs requires another technology that permits the efficient production of pure NPCs in large quantities. In this study, culture substrates were designed by immobilizing epidermal growth factor (EGF) onto the substrate and evaluated for their feasibility of expanding NPCs obtained through the neurosphere culture of induced pluripotent stem (iPS) cells. After three passages we obtained neurospheres that contained cells abundantly expressing an EGF receptor. The neurospheres were dissociated into single cells and seeded onto the EGF-immobilized substrates. It was observed that neurosphere-forming cells seeded and cultured on the EGF-immobilized surface formed a two-dimensional cellular network characteristic of NPCs. These cells were found to be capable of being subcultured, while remaining their proliferation potential. Furthermore, a majority of cells (~99% of total cells) on the substrate was shown to express an NPC marker, nestin, whereas a limited number of cells (~1% of total cells) expressed neuronal marker, ß-tubulin III. These results as a whole demonstrate that the EGF-immobilized substrate allows for iPS cell-derived NPCs to efficiently proliferate while maintaining the undifferentiated state.


Assuntos
Técnicas Citológicas/métodos , Fator de Crescimento Epidérmico/metabolismo , Proteínas Imobilizadas/metabolismo , Células-Tronco Pluripotentes Induzidas , Células-Tronco Neurais , Animais , Proliferação de Células , Células Cultivadas , Fator de Crescimento Epidérmico/química , Receptores ErbB/química , Receptores ErbB/metabolismo , Proteínas Imobilizadas/química , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Células-Tronco Neurais/química , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
11.
Chem Commun (Camb) ; 56(15): 2344-2347, 2020 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-31993612

RESUMO

In our report, we found a distinct difference in azido sugar metabolic rate between neural stem cells and fibroblasts, which can be used for selective removal of fibroblasts from neural stem cell mixtures. Chemically induced neural stem cells (ciNSCs) serve as a highly valuable source of NSCs. Incompletely induced fibroblasts could interfere with ciNSC differentiation and become tumorigenic. Herein, we applied our method for the decontamination of ciNSCs and it exhibited excellent selectivity for ciNSCs. The results demonstrate that the ciNSC population can be efficiently purified to 98.1%. As far as we know, this is the highest purity obtained so far. We envision that, in the future, our method could be used as a safe, effective, and chemically-defined tool for decontaminating ciNSCs in both fundamental research and clinical stem cell therapy.


Assuntos
Azidas/metabolismo , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/metabolismo , Açúcares/metabolismo , Células 3T3 , Animais , Azidas/química , Proliferação de Células , Fibroblastos/química , Células-Tronco Pluripotentes Induzidas/química , Camundongos , Células-Tronco Neurais/química , Açúcares/química
12.
Proc Natl Acad Sci U S A ; 116(51): 25688-25696, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31772009

RESUMO

Neural stem cells (NSCs) generate neurons and glial cells throughout embryonic and postnatal brain development. The role of S-palmitoylation (also referred to as S-acylation), a reversible posttranslational lipid modification of proteins, in regulating the fate and activity of NSCs remains largely unknown. We used an unbiased screening approach to identify proteins that are S-acylated in mouse NSCs and showed that bone morphogenic protein receptor 1a (BMPR1a), a core mediator of BMP signaling, is palmitoylated. Genetic manipulation of S-acylated sites affects the localization and trafficking of BMPR1a and leads to altered BMP signaling. Strikingly, defective palmitoylation of BMPR1a modulates NSC function within the mouse brain, resulting in enhanced oligodendrogenesis. Thus, we identified a mechanism regulating the behavior of NSCs and provided the framework to characterize dynamic posttranslational lipid modifications of proteins in the context of NSC biology.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Lipoilação/fisiologia , Células-Tronco Neurais , Neurogênese/fisiologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/química , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Células Cultivadas , Camundongos , Células-Tronco Neurais/química , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo
13.
J Vis Exp ; (151)2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31609309

RESUMO

Multiple DNA modifications have been identified in the mammalian genome. Of that, 5-methylcytosine and 5-hydroxymethylcytosine-mediated epigenetic mechanisms have been intensively studied. 5-hydroxymethylcytosine displays dynamic features during embryonic and postnatal development of the brain, plays a regulatory function in gene expression, and is involved in multiple neurological disorders. Here, we describe the detailed methods including immunofluorescence staining and DNA dot-blot to detect 5-hydroxymethylcytosine in cultured cells and brain tissues of mouse.


Assuntos
5-Metilcitosina/análogos & derivados , Química Encefálica , Encéfalo/imunologia , Células-Tronco Neurais/química , Células-Tronco Neurais/imunologia , 5-Metilcitosina/análise , 5-Metilcitosina/imunologia , Fatores Etários , Animais , Linhagem Celular , Células Cultivadas , Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Immunoblotting/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
EMBO J ; 38(17): e100481, 2019 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-31304985

RESUMO

Regulation of adult neural stem cell (NSC) number is critical for lifelong neurogenesis. Here, we identified a post-transcriptional control mechanism, centered around the microRNA 204 (miR-204), to control the maintenance of quiescent (q)NSCs. miR-204 regulates a spectrum of transcripts involved in cell cycle regulation, neuronal migration, and differentiation in qNSCs. Importantly, inhibition of miR-204 function reduced the number of qNSCs in the subependymal zone (SEZ) by inducing pre-mature activation and differentiation of NSCs without changing their neurogenic potential. Strikingly, we identified the choroid plexus of the mouse lateral ventricle as the major source of miR-204 that is released into the cerebrospinal fluid to control number of NSCs within the SEZ. Taken together, our results describe a novel mechanism to maintain adult somatic stem cells by a niche-specific miRNA repressing activation and differentiation of stem cells.


Assuntos
Plexo Corióideo/química , MicroRNAs/genética , Células-Tronco Neurais/citologia , Adulto , Animais , Ciclo Celular , Diferenciação Celular , Movimento Celular , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , MicroRNAs/líquido cefalorraquidiano , Pessoa de Meia-Idade , Células-Tronco Neurais/química , Nicho de Células-Tronco
15.
ACS Nano ; 13(8): 8793-8803, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31361458

RESUMO

The full realization of stem cell-based treatments for neurodegenerative diseases requires precise control and characterization of stem cell fate. Herein, we report a multifunctional magneto-plasmonic nanorod (NR)-based detection platform to address the limitations associated with the current destructive characterization methods of stem cell neurogenesis. Exosomes and their inner contents have been discovered to play critical roles in cell-cell interactions and intrinsic cellular regulations and have received wide attention as next-generation biomarkers. Moreover, exosomal microRNAs (miRNA) also offer an essential avenue for nondestructive molecular analyses of cell cytoplasm components. To this end, our developed nondestructive, selective, and sensitive detection platform has (i) an immunomagnetic active component for exosome isolation and (ii) a plasmonic/metal-enhanced fluorescence component for sensitive exosomal miRNA detection to characterize stem cell differentiation. In a proof-of-concept demonstration, our multifunctional magneto-plasmonic NR successfully detected the expression level of miRNA-124 and characterized neurogenesis of human-induced pluripotent stem cell-derived neural stem cells in a nondestructive and efficient manner. Furthermore, we demonstrated the versatility and feasibility of our multifunctional magneto-plasmonic NRs by characterizing a heterogeneous population of neural cells in an ex vivo rodent model. Collectively, we believe our multifunctional magneto-plasmonic NR-based exosomal miRNA detection platform has a great potential to investigate the function of cell-cell interactions and intrinsic cellular regulators for controlling stem cell differentiation.


Assuntos
Técnicas Biossensoriais , Nanopartículas de Magnetita/química , MicroRNAs/isolamento & purificação , Neurogênese/genética , Biomarcadores/química , Comunicação Celular/genética , Diferenciação Celular/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Nanoestruturas/química , Células-Tronco Neurais/química , Células-Tronco Neurais/metabolismo
16.
Nat Commun ; 10(1): 2835, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31249377

RESUMO

During embryogenesis cells make fate decisions within complex tissue environments. The levels and dynamics of transcription factor expression regulate these decisions. Here, we use single cell live imaging of an endogenous HES5 reporter and absolute protein quantification to gain a dynamic view of neurogenesis in the embryonic mammalian spinal cord. We report that dividing neural progenitors show both aperiodic and periodic HES5 protein fluctuations. Mathematical modelling suggests that in progenitor cells the HES5 oscillator operates close to its bifurcation boundary where stochastic conversions between dynamics are possible. HES5 expression becomes more frequently periodic as cells transition to differentiation which, coupled with an overall decline in HES5 expression, creates a transient period of oscillations with higher fold expression change. This increases the decoding capacity of HES5 oscillations and correlates with interneuron versus motor neuron cell fate. Thus, HES5 undergoes complex changes in gene expression dynamics as cells differentiate.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Proteínas Repressoras/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos/embriologia , Camundongos/metabolismo , Camundongos Endogâmicos ICR , Camundongos Knockout , Células-Tronco Neurais/química , Células-Tronco Neurais/citologia , Proteínas Repressoras/química , Proteínas Repressoras/genética , Análise de Célula Única
17.
J Comp Neurol ; 527(18): 2931-2947, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31132148

RESUMO

Neural stem cells in the subventricular zone (SVZ) of the lateral ventricle generate new interneurons, which migrate tangentially through the rostral migratory stream (RMS) to the olfactory bulb (OB). The PROK2 (prokineticin 2) and PROKR2 (prokineticin receptor 2) signaling pathway has been identified to cause human Kallmann syndrome, a developmental disease that associates hypogonadism with anosmia (OB developmental defects). However, the identities and properties of PROK2+ and PROKR2+ cells in the SVZ-RMS-OB remain largely unknown. Here we examine the expression patterns of Prok2 and Prokr2 in the SVZ-RMS-OB using Prok2EGFP transgenic and Prokr2LacZ/+ knockin mice. Our results show that Prokr2 is expressed in postmitotic immature interneurons in the SVZ-RMS-OB. Prok2 is not expressed in the SVZ, but a few PROK2+ cells are found in the medial part of the RMS; they are not neural progenitors or migrating neuroblasts. In the OB, Prok2 is expressed in a subset of granule cells and tufted cells, but no coexpression of Prok2 and Prokr2 in the OB cells is observed. In Prok2 and Prokr2 mutant mice, severe tangential and radial migration defects of neuroblasts in the SVZ-RMS-OB result in loss of ~75% of GABAergic interneurons in the OB. These analyses demonstrate that PROK2/PROKR2 signaling is crucial for the tangential and radial migration of OB interneurons.


Assuntos
Movimento Celular/fisiologia , Hormônios Gastrointestinais/biossíntese , Interneurônios/metabolismo , Células-Tronco Neurais/metabolismo , Neuropeptídeos/biossíntese , Bulbo Olfatório/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Receptores de Peptídeos/biossíntese , Animais , Hormônios Gastrointestinais/genética , Interneurônios/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neurais/química , Neuropeptídeos/genética , Bulbo Olfatório/química , Bulbo Olfatório/citologia , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Transdução de Sinais/fisiologia
18.
Clin Transl Oncol ; 21(11): 1551-1560, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30945128

RESUMO

PURPOSE: Neural stem cells (NSCs) have been characterized with the ability of self-renewal and neurogenesis, which has inspired lots of studies to clarify the functions of NSCs in neural injury, ischemic stroke, brain inflammation and neurodegenerative diseases. We focused on the relationship of NSCs with glioblastoma, since we have discovered that recurrent glioblastomas were inclined to be derived from subventricular zone (SVZ), where NSCs reside. We want to clarify whether NSCs are involved in glioblastoma relapse. METHODS: Immunocytochemistry was used to confirm the stemness of NSCs. The Cell Counting Kit-8 was used to measure the proliferation of cells. Migration abilities were examined by wound healing and transwell assays, and tumor formation abilities were confirmed in nude mice. RESULTS: We found in experiments that NSCs promoted proliferation of a glioblastoma cell line-Ln229, the migration ability of Ln229 cells was motivated by co-cultured with NSCs. Tumor formation of Ln229 cells was also accelerated in nude mice when co-transplanted with NSCs. In immunohistochemistry, we found that the Sox2- and Ki67-positive cells were much higher in co-transplanted groups than that of control groups. CONCLUSIONS: These results imply the potential role that NSCs play in speeding up tumor formation in the process of glioblastoma relapse, providing the basis for dealing with newly diagnosed glioblastoma patients, which may help postpone the recurrence of glioblastoma as far as possible through preprocessing the tumor-adjacent SVZ tissue.


Assuntos
Movimento Celular , Proliferação de Células , Glioblastoma/etiologia , Células-Tronco Neurais/fisiologia , Animais , Linhagem Celular Tumoral , Glioblastoma/química , Antígeno Ki-67/análise , Ventrículos Laterais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neurais/química , Fatores de Transcrição SOXB1/análise , Cicatrização
19.
J Comp Neurol ; 527(14): 2317-2333, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30843204

RESUMO

Adiponectin and its receptors (adipor) have been initially characterized for their role in lipid and glucose metabolism. More recently, adiponectin signaling was shown to display anti-inflammatory effects and to participate in brain homeostasis and neuroprotection. In this study, we investigated adipor gene expression and its regulation under inflammatory conditions in two complementary models: mouse and zebrafish. We demonstrate that adipor1a, adipor1b, and adipor2 are widely distributed throughout the brain of adult fish, in neurons and also in radial glia, behaving as neural stem cells. We also show that telencephalic injury results in a decrease in adipor gene expression, inhibited by an anti-inflammatory treatment (Dexamethasone). Interestingly, adiponectin injection after brain injury led to a consistent decrease (a) in the recruitment of microglial cells at the lesioned site and (b) in the proliferation of neural progenitors, arguing for a neuroprotective role of adiponectin. In a comparative approach, we investigate Adipor1 and Adipor2 gene distribution in the brain of mice and demonstrated their expression in regions shared with fish including neurogenic regions. We also document Adipor gene expression in mice after middle cerebral artery occlusion and lipopolysaccharide injection. In contrast to zebrafish, these inflammatory stimuli do no impact cerebral adiponectin receptor gene expression in mouse. This work provides new insights regarding adipor expression in the brain of fish, and demonstrates evolutionary conserved distribution of adipor with mouse. This is the first report of adipor expression in adult neural stem cells of fish, suggesting a potential role of adiponectin signaling during vertebrate neurogenesis. It also suggests a potential contribution of inflammation in the regulation of adipor in fish.


Assuntos
Encéfalo/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Receptores de Adiponectina/biossíntese , Fatores Etários , Animais , Encéfalo/citologia , Química Encefálica/fisiologia , Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/química , Receptores de Adiponectina/análise , Receptores de Adiponectina/genética , Especificidade da Espécie , Peixe-Zebra
20.
J Neurosci ; 38(44): 9338-9345, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30381425

RESUMO

Sonic Hedgehog (SHH) signaling has been most widely known for its role in specifying region and cell-type identity during embryonic morphogenesis. This mini-review accompanies a 2018 SFN mini-symposium that addresses an emerging body of research focused on understanding the diverse roles for Shh signaling in a wide range of contexts in neurodevelopment and, more recently, in the mature CNS. Such research shows that Shh affects the function of brain circuits, including the production and maintenance of diverse cell types and the establishment of wiring specificity. Here, we review these novel and unexpected functions and the unanswered questions regarding the role of SHH and its signaling pathway members in these cases.


Assuntos
Proteínas Hedgehog/fisiologia , Morfogênese/fisiologia , Células-Tronco Neurais/metabolismo , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Animais , Humanos , Células-Tronco Neurais/química , Sinapses/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA