Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50.644
Filtrar
1.
Methods Mol Biol ; 2799: 47-54, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38727902

RESUMEN

Transfection allows the introduction of foreign nucleic acid into eukaryotic cells. It is an important tool in understanding the roles of NMDARs in neurons. Here we describe using lipofection-mediated transfection to introduce cDNA encoding NMDAR subunits into postmitotic rodent primary cortical neurons maintained in culture.


Asunto(s)
Neuronas , Transfección , Neuronas/metabolismo , Neuronas/citología , Animales , Transfección/métodos , Células Cultivadas , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Ratones , Cultivo Primario de Células/métodos , ADN Complementario/genética
2.
Epigenetics Chromatin ; 17(1): 18, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783373

RESUMEN

The three-dimensional organization of the genome plays a central role in the regulation of cellular functions, particularly in the human brain. This review explores the intricacies of chromatin organization, highlighting the distinct structural patterns observed between neuronal and non-neuronal brain cells. We integrate findings from recent studies to elucidate the characteristics of various levels of chromatin organization, from differential compartmentalization and topologically associating domains (TADs) to chromatin loop formation. By defining the unique chromatin landscapes of neuronal and non-neuronal brain cells, these distinct structures contribute to the regulation of gene expression specific to each cell type. In particular, we discuss potential functional implications of unique neuronal chromatin organization characteristics, such as weaker compartmentalization, neuron-specific TAD boundaries enriched with active histone marks, and an increased number of chromatin loops. Additionally, we explore the role of Polycomb group (PcG) proteins in shaping cell-type-specific chromatin patterns. This review further emphasizes the impact of variations in chromatin architecture between neuronal and non-neuronal cells on brain development and the onset of neurological disorders. It highlights the need for further research to elucidate the details of chromatin organization in the human brain in order to unravel the complexities of brain function and the genetic mechanisms underlying neurological disorders. This research will help bridge a significant gap in our comprehension of the interplay between chromatin structure and cell functions.


Asunto(s)
Encéfalo , Cromatina , Neuronas , Humanos , Neuronas/metabolismo , Neuronas/citología , Cromatina/metabolismo , Animales , Encéfalo/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteínas del Grupo Polycomb/genética , Ensamble y Desensamble de Cromatina
3.
Cells ; 13(10)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38786037

RESUMEN

Intestinal homeostasis results from the proper interplay among epithelial cells, the enteric nervous system (ENS), interstitial cells of Cajal (ICCs), smooth muscle cells, the immune system, and the microbiota. The disruption of this balance underpins the onset of gastrointestinal-related diseases. The scarcity of models replicating the intricate interplay between the ENS and the intestinal epithelium highlights the imperative for developing novel methods. We have pioneered a sophisticated tridimensional in vitro technique, coculturing small intestinal organoids with myenteric and submucosal neurons. Notably, we have made significant advances in (1) refining the isolation technique for culturing the myenteric plexus, (2) enhancing the isolation of the submucosal plexus-both yielding mixed cultures of enteric neurons and glial cells from both plexuses, and (3) subsequently co-culturing myenteric and submucosal neurons with small intestinal organoids. This co-culture system establishes neural innervations with intestinal organoids, allowing for the investigation of regulatory interactions in the context of gastrointestinal diseases. Furthermore, we have developed a method for microinjecting the luminal space of small intestinal organoids with fluorescently labeled compounds. This technique possesses broad applicability such as the assessment of intestinal permeability, transcytosis, and immunocytochemical and immunofluorescence applications. This microinjection method could be extended to alternative experimental setups, incorporating bacterial species, or applying treatments to study ENS-small intestinal epithelium interactions. Therefore, this technique serves as a valuable tool for evaluating the intricate interplay between neuronal and intestinal epithelial cells (IECs) and shows great potential for drug screening, gene editing, the development of novel therapies, the modeling of infectious diseases, and significant advances in regenerative medicine. The co-culture establishment process spans twelve days, making it a powerful asset for comprehensive research in this critical field.


Asunto(s)
Técnicas de Cocultivo , Intestino Delgado , Plexo Mientérico , Organoides , Animales , Organoides/citología , Técnicas de Cocultivo/métodos , Ratones , Plexo Mientérico/citología , Intestino Delgado/citología , Plexo Submucoso/citología , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/citología , Neuronas/citología , Neuronas/metabolismo
4.
J Vis Exp ; (207)2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38767374

RESUMEN

The neuromodulatory effects of focused ultrasound (FUS) have been demonstrated in animal models, and FUS has been used successfully to treat movement and psychiatric disorders in humans. However, despite the success of FUS, the mechanism underlying its effects on neurons remains poorly understood, making treatment optimization by tuning FUS parameters difficult. To address this gap in knowledge, we studied human neurons in vitro using neurons cultured from human-induced pluripotent stem cells (HiPSCs). Using HiPSCs allows for the study of human-specific neuronal behaviors in both physiologic and pathologic states. This report presents a protocol for using a high-throughput system that enables the monitoring and quantification of the neuromodulatory effects of FUS on HiPSC neurons. By varying the FUS parameters and manipulating the HiPSC neurons through pharmaceutical and genetic modifications, researchers can evaluate the neural responses and elucidate the neuro-modulatory effects of FUS on HiPSC neurons. This research could have significant implications for the development of safe and effective FUS-based therapies for a range of neurological and psychiatric disorders.


Asunto(s)
Células Madre Pluripotentes Inducidas , Microelectrodos , Neuronas , Humanos , Neuronas/fisiología , Neuronas/citología , Células Madre Pluripotentes Inducidas/citología , Ondas Ultrasónicas
5.
Nat Commun ; 15(1): 4102, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778027

RESUMEN

The development of robust tools for segmenting cellular and sub-cellular neuronal structures lags behind the massive production of high-resolution 3D images of neurons in brain tissue. The challenges are principally related to high neuronal density and low signal-to-noise characteristics in thick samples, as well as the heterogeneity of data acquired with different imaging methods. To address this issue, we design a framework which includes sample preparation for high resolution imaging and image analysis. Specifically, we set up a method for labeling thick samples and develop SENPAI, a scalable algorithm for segmenting neurons at cellular and sub-cellular scales in conventional and super-resolution STimulated Emission Depletion (STED) microscopy images of brain tissues. Further, we propose a validation paradigm for testing segmentation performance when a manual ground-truth may not exhaustively describe neuronal arborization. We show that SENPAI provides accurate multi-scale segmentation, from entire neurons down to spines, outperforming state-of-the-art tools. The framework will empower image processing of complex neuronal circuitries.


Asunto(s)
Algoritmos , Encéfalo , Imagenología Tridimensional , Neuronas , Neuronas/citología , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/citología , Imagenología Tridimensional/métodos , Ratones , Procesamiento de Imagen Asistido por Computador/métodos
6.
Sci Adv ; 10(21): eadk2149, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38781326

RESUMEN

Understanding the genetic programs that drive neuronal diversification into classes and subclasses is key to understand nervous system development. All neurons can be classified into two types: commissural and ipsilateral, based on whether their axons cross the midline or not. However, the gene regulatory program underlying this binary division is poorly understood. We identified a pair of basic helix-loop-helix transcription factors, Nhlh1 and Nhlh2, as a global transcriptional mechanism that controls the laterality of all floor plate-crossing commissural axons in mice. Mechanistically, Nhlh1/2 play an essential role in the expression of Robo3, the key guidance molecule for commissural axon projections. This genetic program appears to be evolutionarily conserved in chick. We further discovered that Isl1, primarily expressed in ipsilateral neurons within neural tubes, negatively regulates the Robo3 induction by Nhlh1/2. Our findings elucidate a gene regulatory strategy where a conserved global mechanism intersects with neuron class-specific regulators to control the partitioning of neurons based on axon laterality.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Regulación del Desarrollo de la Expresión Génica , Neuronas , Animales , Neuronas/metabolismo , Neuronas/citología , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Axones/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Embrión de Pollo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Redes Reguladoras de Genes
7.
PLoS One ; 19(5): e0298274, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38753762

RESUMEN

The membrane peroxisomal proteins PEX11, play a crucial role in peroxisome proliferation by regulating elongation, membrane constriction, and fission of pre-existing peroxisomes. In this study, we evaluated the function of PEX11B gene in neural differentiation of human embryonic stem cell (hESC) by inducing shRNAi-mediated knockdown of PEX11B expression. Our results demonstrate that loss of PEX11B expression led to a significant decrease in the expression of peroxisomal-related genes including ACOX1, PMP70, PEX1, and PEX7, as well as neural tube-like structures and neuronal markers. Inhibition of SIRT1 using pharmacological agents counteracted the effects of PEX11B knockdown, resulting in a relative increase in PEX11B expression and an increase in differentiated neural tube-like structures. However, the neuroprotective effects of SIRT1 were eliminated by PPAR inhibition, indicating that PPARÉ£ may mediate the interaction between PEX11B and SIRT1. Our findings suggest that both SIRT1 and PPARÉ£ have neuroprotective effects, and also this study provides the first indication for a potential interaction between PEX11B, SIRT1, and PPARÉ£ during hESC neural differentiation.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias Humanas , Proteínas de la Membrana , PPAR gamma , Sirtuina 1 , Humanos , Sirtuina 1/metabolismo , Sirtuina 1/genética , PPAR gamma/metabolismo , PPAR gamma/genética , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Neuronas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Línea Celular , Peroxisomas/metabolismo
8.
Lab Chip ; 24(10): 2811-2824, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38700452

RESUMEN

The aging process has broad physiological impacts, including a significant decline in sensory function, which threatens both physical health and quality of life. One ideal model to study aging, neuronal function, and gene expression is the nematode Caenorhabditis elegans, which has a short lifespan and relatively simple, thoroughly mapped nervous system and genome. Previous works have identified that mechanosensory neuronal structure changes with age, but importantly, the actual age-related changes in the function and health of neurons, as well as the underlying genetic mechanisms responsible for these declines, are not fully understood. While advanced techniques such as single-cell RNA-sequencing have been developed to quantify gene expression, it is difficult to relate this information to functional changes in aging due to a lack of tools available. To address these limitations, we present a platform capable of measuring both physiological function and its associated gene expression throughout the aging process in individuals. Using our pipeline, we investigate the age-related changes in function of the mechanosensing ALM neuron in C. elegans, as well as some relevant gene expression patterns (mec-4 and mec-10). Using a series of devices for animals of different ages, we examined subtle changes in neuronal function and found that while the magnitude of neuronal response to a large stimulus declines with age, sensory capability does not significantly decline with age; further, gene expression is well maintained throughout aging. Additionally, we examine PVD, a harsh-touch mechanosensory neuron, and find that it exhibits a similar age-related decline in magnitude of neuronal response. Together, our data demonstrate that our strategy is useful for identifying genetic factors involved in the decline in neuronal health. We envision that this framework could be applied to other systems as a useful tool for discovering new biology.


Asunto(s)
Envejecimiento , Caenorhabditis elegans , Dispositivos Laboratorio en un Chip , Neuronas , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Caenorhabditis elegans/metabolismo , Envejecimiento/fisiología , Neuronas/metabolismo , Neuronas/citología , Mecanotransducción Celular , Técnicas Analíticas Microfluídicas/instrumentación
9.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732109

RESUMEN

Adipose-derived mesenchymal stem cells (ASCs) are adult multipotent stem cells, able to differentiate toward neural elements other than cells of mesodermal lineage. The aim of this research was to test ASC neural differentiation using melatonin combined with conditioned media (CM) from glial cells. Isolated from the lipoaspirate of healthy donors, ASCs were expanded in a basal growth medium before undergoing neural differentiation procedures. For this purpose, CM obtained from olfactory ensheathing cells and from Schwann cells were used. In some samples, 1 µM of melatonin was added. After 1 and 7 days of culture, cells were studied using immunocytochemistry and flow cytometry to evaluate neural marker expression (Nestin, MAP2, Synapsin I, GFAP) under different conditions. The results confirmed that a successful neural differentiation was achieved by glial CM, whereas the addition of melatonin alone did not induce appreciable changes. When melatonin was combined with CM, ASC neural differentiation was enhanced, as demonstrated by a further improvement of neuronal marker expression, whereas glial differentiation was attenuated. A dynamic modulation was also observed, testing the expression of melatonin receptors. In conclusion, our data suggest that melatonin's neurogenic differentiation ability can be usefully exploited to obtain neuronal-like differentiated ASCs for potential therapeutic strategies.


Asunto(s)
Diferenciación Celular , Melatonina , Células Madre Mesenquimatosas , Melatonina/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Humanos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Tejido Adiposo/citología , Neuronas/citología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Células de Schwann/citología , Células de Schwann/metabolismo , Células de Schwann/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Adulto , Nestina/metabolismo , Nestina/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/citología , Neuroglía/metabolismo , Sinapsinas/metabolismo
10.
Cell Stem Cell ; 31(5): 754-771.e6, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701759

RESUMEN

Development of embryonic stem cells (ESCs) into neurons requires intricate regulation of transcription, splicing, and translation, but how these processes interconnect is not understood. We found that polypyrimidine tract binding protein 1 (PTBP1) controls splicing of DPF2, a subunit of BRG1/BRM-associated factor (BAF) chromatin remodeling complexes. Dpf2 exon 7 splicing is inhibited by PTBP1 to produce the DPF2-S isoform early in development. During neuronal differentiation, loss of PTBP1 allows exon 7 inclusion and DPF2-L expression. Different cellular phenotypes and gene expression programs were induced by these alternative DPF2 isoforms. We identified chromatin binding sites enriched for each DPF2 isoform, as well as sites bound by both. In ESC, DPF2-S preferential sites were bound by pluripotency factors. In neuronal progenitors, DPF2-S sites were bound by nuclear factor I (NFI), while DPF2-L sites were bound by CCCTC-binding factor (CTCF). DPF2-S sites exhibited enhancer modifications, while DPF2-L sites showed promoter modifications. Thus, alternative splicing redirects BAF complex targeting to impact chromatin organization during neuronal development.


Asunto(s)
Empalme Alternativo , Diferenciación Celular , Cromatina , Ribonucleoproteínas Nucleares Heterogéneas , Neuronas , Proteína de Unión al Tracto de Polipirimidina , Factores de Transcripción , Empalme Alternativo/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Animales , Diferenciación Celular/genética , Cromatina/metabolismo , Ratones , Neuronas/metabolismo , Neuronas/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Transcripción Genética , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/citología , Exones/genética , Humanos , Autorrenovación de las Células/genética
11.
Biosci Rep ; 44(5)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38700092

RESUMEN

Pre-eclampsia (PE) is a hypertensive disorder of pregnancy which is associated with increased risk of neurodevelopmental disorders in exposed offspring. The pathophysiological mechanisms mediating this relationship are currently unknown, and one potential candidate is the anti-angiogenic factor soluble Fms-like tyrosine kinase 1 (sFlt-1), which is highly elevated in PE. While sFlt-1 can impair angiogenesis via inhibition of VEGFA signalling, it is unclear whether it can directly affect neuronal development independently of its effects on the vasculature. To test this hypothesis, the current study differentiated the human neural progenitor cell (NPC) line ReNcell® VM into a mixed culture of mature neurons and glia, and exposed them to sFlt-1 during development. Outcomes measured were neurite growth, cytotoxicity, mRNA expression of nestin, MBP, GFAP, and ßIII-tubulin, and neurosphere differentiation. sFlt-1 induced a significant reduction in neurite growth and this effect was timing- and dose-dependent up to 100 ng/ml, with no effect on cytotoxicity. sFlt-1 (100 ng/ml) also reduced ßIII-tubulin mRNA and neuronal differentiation of neurospheres. Undifferentiated NPCs and mature neurons/glia expressed VEGFA and VEGFR-2, required for endogenous autocrine and paracrine VEGFA signalling, while sFlt-1 treatment prevented the neurogenic effects of exogenous VEGFA. Overall, these data provide the first experimental evidence for a direct effect of sFlt-1 on neurite growth and neuronal differentiation in human neurons through inhibition of VEGFA signalling, clarifying our understanding of the potential role of sFlt-1 as a mechanism by which PE can affect neuronal development.


Asunto(s)
Diferenciación Celular , Células-Madre Neurales , Neuronas , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Humanos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/citología , Diferenciación Celular/efectos de los fármacos , Neuritas/metabolismo , Neuritas/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Femenino , Preeclampsia/metabolismo , Preeclampsia/patología , Embarazo , Línea Celular Tumoral , Transducción de Señal
12.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38739758

RESUMEN

The complicated process of neuronal development is initiated early in life, with the genetic mechanisms governing this process yet to be fully elucidated. Single-cell RNA sequencing (scRNA-seq) is a potent instrument for pinpointing biomarkers that exhibit differential expression across various cell types and developmental stages. By employing scRNA-seq on human embryonic stem cells, we aim to identify differentially expressed genes (DEGs) crucial for early-stage neuronal development. Our focus extends beyond simply identifying DEGs. We strive to investigate the functional roles of these genes through enrichment analysis and construct gene regulatory networks to understand their interactions. Ultimately, this comprehensive approach aspires to illuminate the molecular mechanisms and transcriptional dynamics governing early human brain development. By uncovering potential links between these DEGs and intelligence, mental disorders, and neurodevelopmental disorders, we hope to shed light on human neurological health and disease. In this study, we have used scRNA-seq to identify DEGs involved in early-stage neuronal development in hESCs. The scRNA-seq data, collected on days 26 (D26) and 54 (D54), of the in vitro differentiation of hESCs to neurons were analyzed. Our analysis identified 539 DEGs between D26 and D54. Functional enrichment of those DEG biomarkers indicated that the up-regulated DEGs participated in neurogenesis, while the down-regulated DEGs were linked to synapse regulation. The Reactome pathway analysis revealed that down-regulated DEGs were involved in the interactions between proteins located in synapse pathways. We also discovered interactions between DEGs and miRNA, transcriptional factors (TFs) and DEGs, and between TF and miRNA. Our study identified 20 significant transcription factors, shedding light on early brain development genetics. The identified DEGs and gene regulatory networks are valuable resources for future research into human brain development and neurodevelopmental disorders.


Asunto(s)
Biomarcadores , Encéfalo , Redes Reguladoras de Genes , Células Madre Embrionarias Humanas , Análisis de la Célula Individual , Humanos , Análisis de la Célula Individual/métodos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Encéfalo/metabolismo , Encéfalo/embriología , Encéfalo/citología , Biomarcadores/metabolismo , Neuronas/metabolismo , Neuronas/citología , Diferenciación Celular/genética , RNA-Seq , Neurogénesis/genética , Regulación del Desarrollo de la Expresión Génica , Perfilación de la Expresión Génica , Análisis de Secuencia de ARN/métodos , Análisis de Expresión Génica de una Sola Célula
13.
Nat Commun ; 15(1): 4047, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38744873

RESUMEN

Human hippocampal organoids (hHOs) derived from human induced pluripotent stem cells (hiPSCs) have emerged as promising models for investigating neurodegenerative disorders, such as schizophrenia and Alzheimer's disease. However, obtaining the electrical information of these free-floating organoids in a noninvasive manner remains a challenge using commercial multi-electrode arrays (MEAs). The three-dimensional (3D) MEAs developed recently acquired only a few neural signals due to limited channel numbers. Here, we report a hippocampal cyborg organoid (cyb-organoid) platform coupling a liquid metal-polymer conductor (MPC)-based mesh neuro-interface with hHOs. The mesh MPC (mMPC) integrates 128-channel multielectrode arrays distributed on a small surface area (~2*2 mm). Stretchability (up to 500%) and flexibility of the mMPC enable its attachment to hHOs. Furthermore, we show that under Wnt3a and SHH activator induction, hHOs produce HOPX+ and PAX6+ progenitors and ZBTB20+PROX1+ dentate gyrus (DG) granule neurons. The transcriptomic signatures of hHOs reveal high similarity to the developing human hippocampus. We successfully detect neural activities from hHOs via the mMPC from this cyb-organoid. Compared with traditional planar devices, our non-invasive coupling offers an adaptor for recording neural signals from 3D models.


Asunto(s)
Hipocampo , Células Madre Pluripotentes Inducidas , Organoides , Humanos , Organoides/metabolismo , Organoides/citología , Hipocampo/citología , Hipocampo/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Neuronas/citología , Metales/química , Transcriptoma , Giro Dentado/citología , Giro Dentado/metabolismo
14.
Nat Commun ; 15(1): 3473, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724563

RESUMEN

Neuronal differentiation-the development of neurons from neural stem cells-involves neurite outgrowth and is a key process during the development and regeneration of neural functions. In addition to various chemical signaling mechanisms, it has been suggested that thermal stimuli induce neuronal differentiation. However, the function of physiological subcellular thermogenesis during neuronal differentiation remains unknown. Here we create methods to manipulate and observe local intracellular temperature, and investigate the effects of noninvasive temperature changes on neuronal differentiation using neuron-like PC12 cells. Using quantitative heating with an infrared laser, we find an increase in local temperature (especially in the nucleus) facilitates neurite outgrowth. Intracellular thermometry reveals that neuronal differentiation is accompanied by intracellular thermogenesis associated with transcription and translation. Suppression of intracellular temperature increase during neuronal differentiation inhibits neurite outgrowth. Furthermore, spontaneous intracellular temperature elevation is involved in neurite outgrowth of primary mouse cortical neurons. These results offer a model for understanding neuronal differentiation induced by intracellular thermal signaling.


Asunto(s)
Diferenciación Celular , Neuronas , Transducción de Señal , Temperatura , Animales , Células PC12 , Neuronas/fisiología , Neuronas/citología , Ratones , Ratas , Proyección Neuronal , Neurogénesis/fisiología , Neuritas/metabolismo , Neuritas/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Termometría/métodos , Termogénesis/fisiología
15.
Sci Rep ; 14(1): 12113, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802572

RESUMEN

SH-SY5Y, a neuroblastoma cell line, can be converted into mature neuronal phenotypes, characterized by the expression of mature neuronal and neurotransmitter markers. However, the mature phenotypes described across multiple studies appear inconsistent. As this cell line expresses common neuronal markers after a simple induction, there is a high chance of misinterpreting its maturity. Therefore, sole reliance on common neuronal markers is presumably inadequate. The Alzheimer's disease (AD) central gene, amyloid precursor protein (APP), has shown contrasting transcript variant dynamics in various cell types. We differentiated SH-SY5Y cells into mature neuron-like cells using a concise protocol and observed the upregulation of total APP throughout differentiation. However, APP transcript variant-1 was upregulated only during the early to middle stages of differentiation and declined in later stages. We identified the maturity state where this post-transcriptional shift occurs, terming it "true maturity." At this stage, we observed a predominant expression of mature neuronal and cholinergic markers, along with a distinct APP variant pattern. Our findings emphasize the necessity of using a differentiation state-sensitive marker system to precisely characterize SH-SY5Y differentiation. Moreover, this study offers an APP-guided, alternative neuronal marker system to enhance the accuracy of the conventional markers.


Asunto(s)
Precursor de Proteína beta-Amiloide , Diferenciación Celular , Neuronas , Humanos , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Neuronas/metabolismo , Neuronas/citología , Línea Celular Tumoral , Neuroblastoma/metabolismo , Neuroblastoma/genética , Neuroblastoma/patología , Biomarcadores/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Empalme Alternativo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética
16.
Eur J Histochem ; 68(2)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38766720

RESUMEN

Previous studies on the granular layer of the cerebellar cortex have revealed a wide distribution of different subpopulations of less-known large neuron types, called "non-traditional large neurons", which are distributed in three different zones of the granular layer. These neuron types are mainly involved in the formation of intrinsiccircuits inside the cerebellar cortex. A subpopulation of these neuron types is represented by the synarmotic neuron, which could play a projective role within the cerebellar circuitry. The synarmotic neuron cell body map within the internal zone of the granular layer or in the subjacent white substance. Furthermore, the axon crosses the granular layer and runs in the subcortical white substance, to reenter in an adjacent granular layer, associating two cortico-cerebellar regions of the same folium or of different folia, or could project to the intrinsic cerebellar nuclei. Therefore, along with the Purkinje neuron, the traditional projective neuron type of the cerebellar cortex, the synarmotic neuron is candidate to represent the second projective neuron type of the cerebellar cortex. Studies of chemical neuroanatomy evidenced a predominant inhibitory GABAergic nature of the synarmotic neuron, suggesting that it may mediate an inhibitory GABAergic output of cerebellar cortex within cortico-cortical interconnections or in projections towards intrinsic cerebellar nuclei. On this basis, the present minireview mainly focuses on the morphofunctional and neurochemical data of the synarmotic neuron, and explores its potential involvement in some forms of cerebellar ataxias.


Asunto(s)
Corteza Cerebelosa , Neuronas , Corteza Cerebelosa/citología , Animales , Humanos , Neuronas/citología , Neuronas/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/citología
17.
Bioelectrochemistry ; 158: 108713, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38688079

RESUMEN

Boron doped diamond has been considered as a fouling-resistive electrode material for in vitro and in vivo detection of neurotransmitters. In this study, its performance in electrochemical detection of dopamine and serotonin in neuron cultivation media Neurobasal™ before and after cultivation of rat neurons was investigated. For differential pulse voltammetry the limits of detection in neat Neurobasal™ medium of 2 µM and 0.2 µM for dopamine and serotonin, respectively, were achieved on the polished surface, which is comparable with physiological values. On oxidized surface twofold higher values, but increased repeatabilities of the signals were obtained. However, in Neurobasal™ media with peptides-containing supplements necessary for cell cultivation, the voltammograms were notably worse shaped due to biofouling, especially in the medium isolated after neuron growth. In these complex media, the amperometric detection mode at +0.75 V (vs. Ag/AgCl) allowed to detect portion-wise additions of dopamine and serotonin (as low as 1-2 µM), mimicking neurotransmitter release from vesicles despite the lower sensitivity in comparison with neat NeurobasalTM. The results indicate substantial differences in detection on boron doped diamond electrode in the presence and absence of proteins, and the necessity of studies in real media for successful implementation to neuron-electrode interfaces.


Asunto(s)
Incrustaciones Biológicas , Boro , Medios de Cultivo , Diamante , Dopamina , Electrodos , Neuronas , Serotonina , Serotonina/análisis , Dopamina/análisis , Boro/química , Diamante/química , Animales , Neuronas/citología , Neuronas/metabolismo , Ratas , Incrustaciones Biológicas/prevención & control , Medios de Cultivo/química , Técnicas Electroquímicas/métodos
18.
Nature ; 629(8010): 121-126, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38632395

RESUMEN

The neural crest is an embryonic stem cell population unique to vertebrates1 whose expansion and diversification are thought to have promoted vertebrate evolution by enabling emergence of new cell types and structures such as jaws and peripheral ganglia2. Although jawless vertebrates have sensory ganglia, convention has it that trunk sympathetic chain ganglia arose only in jawed vertebrates3-8. Here, by contrast, we report the presence of trunk sympathetic neurons in the sea lamprey, Petromyzon marinus, an extant jawless vertebrate. These neurons arise from sympathoblasts near the dorsal aorta that undergo noradrenergic specification through a transcriptional program homologous to that described in gnathostomes. Lamprey sympathoblasts populate the extracardiac space and extend along the length of the trunk in bilateral streams, expressing the catecholamine biosynthetic pathway enzymes tyrosine hydroxylase and dopamine ß-hydroxylase. CM-DiI lineage tracing analysis further confirmed that these cells derive from the trunk neural crest. RNA sequencing of isolated ammocoete trunk sympathoblasts revealed gene profiles characteristic of sympathetic neuron function. Our findings challenge the prevailing dogma that posits that sympathetic ganglia are a gnathostome innovation, instead suggesting that a late-developing rudimentary sympathetic nervous system may have been characteristic of the earliest vertebrates.


Asunto(s)
Linaje de la Célula , Ganglios Simpáticos , Cresta Neural , Neuronas , Petromyzon , Sistema Nervioso Simpático , Tirosina 3-Monooxigenasa , Animales , Cresta Neural/citología , Cresta Neural/metabolismo , Ganglios Simpáticos/citología , Ganglios Simpáticos/metabolismo , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Tirosina 3-Monooxigenasa/genética , Neuronas/citología , Neuronas/metabolismo , Dopamina beta-Hidroxilasa/metabolismo , Dopamina beta-Hidroxilasa/genética , Vertebrados , Evolución Biológica , Norepinefrina/metabolismo
19.
Lab Chip ; 24(9): 2397-2417, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38623840

RESUMEN

Optical techniques, such as optogenetic stimulation and functional fluorescence imaging, have been revolutionary for neuroscience by enabling neural circuit analysis with cell-type specificity. To probe deep brain regions, implantable light sources are crucial. Silicon photonics, commonly used for data communications, shows great promise in creating implantable devices with complex optical systems in a compact form factor compatible with high volume manufacturing practices. This article reviews recent developments of wafer-scale multifunctional nanophotonic neural probes. The probes can be realized on 200 or 300 mm wafers in commercial foundries and integrate light emitters for photostimulation, microelectrodes for electrophysiological recording, and microfluidic channels for chemical delivery and sampling. By integrating active optical devices to the probes, denser emitter arrays, enhanced on-chip biosensing, and increased ease of use may be realized. Silicon photonics technology makes possible highly versatile implantable neural probes that can transform neuroscience experiments.


Asunto(s)
Encéfalo , Encéfalo/fisiología , Humanos , Animales , Mapeo Encefálico/instrumentación , Neuronas/fisiología , Neuronas/citología , Silicio/química , Nanotecnología/instrumentación , Optogenética/instrumentación
20.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38664022

RESUMEN

Neural stem cells (NSCs) reside in discrete regions of the adult mammalian brain where they can differentiate into neurons, astrocytes, and oligodendrocytes. Several studies suggest that mitochondria have a major role in regulating NSC fate. Here, we evaluated mitochondrial properties throughout NSC differentiation and in lineage-specific cells. For this, we used the neurosphere assay model to isolate, expand, and differentiate mouse subventricular zone postnatal NSCs. We found that the levels of proteins involved in mitochondrial fusion (Mitofusin [Mfn] 1 and Mfn 2) increased, whereas proteins involved in fission (dynamin-related protein 1 [DRP1]) decreased along differentiation. Importantly, changes in mitochondrial dynamics correlated with distinct patterns of mitochondrial morphology in each lineage. Particularly, we found that the number of branched and unbranched mitochondria increased during astroglial and neuronal differentiation, whereas the area occupied by mitochondrial structures significantly reduced with oligodendrocyte maturation. In addition, comparing the three lineages, neurons revealed to be the most energetically flexible, whereas astrocytes presented the highest ATP content. Our work identified putative mitochondrial targets to enhance lineage-directed differentiation of mouse subventricular zone-derived NSCs.


Asunto(s)
Astrocitos , Diferenciación Celular , Linaje de la Célula , Dinaminas , Mitocondrias , Dinámicas Mitocondriales , Células-Madre Neurales , Neuronas , Oligodendroglía , Animales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Mitocondrias/metabolismo , Ratones , Diferenciación Celular/genética , Linaje de la Célula/genética , Astrocitos/metabolismo , Astrocitos/citología , Oligodendroglía/metabolismo , Oligodendroglía/citología , Neuronas/metabolismo , Neuronas/citología , Células Cultivadas , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , GTP Fosfohidrolasas/metabolismo , GTP Fosfohidrolasas/genética , Neurogénesis , Ventrículos Laterales/citología , Ventrículos Laterales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...