ng2 cells
Recently Published Documents


TOTAL DOCUMENTS

90
(FIVE YEARS 14)

H-INDEX

27
(FIVE YEARS 2)

Author(s):  
Chengcai Li ◽  
Shaoxin Huang ◽  
Wu Zhou ◽  
Zhiping Xie ◽  
Shenke Xie ◽  
...  

PLoS ONE ◽  
2021 ◽  
Vol 16 (4) ◽  
pp. e0249729
Author(s):  
Yasuhisa Tamura ◽  
Kumi Takata ◽  
Asami Eguchi ◽  
Mitsuyo Maeda ◽  
Yosky Kataoka

NG2 immunoreactive cells (NG2 cells) are found in the brain and peripheral tissues including the skin, intestinal tracts, and bladder. In a previous study, we observed the presence of NG2 cells in the stomach using bioluminescence imaging techniques in NG2-firefly luciferase (fLuc) transgenic (Tg) rats. Here, we aimed to identify and characterize NG2 cells in the adult rat stomach. Immunohistochemical studies showed that NG2 cells were mainly present in the lamina propria and most of the cells were gastric telocytes, co-expressing CD34, and platelet-derived growth factor receptor alpha (PDGFRα), with a small oval-shaped cell body and extremely long and thin cellular prolongations. In the rat stomach, NG2-expressing telocytes comprised two subpopulations: NG2+/CD34+/PDGFRα+ and NG2+/CD34+/PDGFRα-. Furthermore, we showed that the expression of NG2 gene in the aged rat stomach decreased relative to that of the young rat stomach and the decline of NG2 expression in aged rats was mainly observed in NG2+/CD34+/PDGFRα+ telocytes. These findings suggested age-related alterations in NG2+/CD34+/PDGFRα+ telocytes of rat stomach.


2020 ◽  
Vol 17 (1) ◽  
Author(s):  
Laura A. Bell ◽  
Glenna J. Wallis ◽  
Karen S. Wilcox

Abstract Background Neuron-glial antigen 2 (NG2) cells are a glial cell type tiled throughout the gray and white matter of the central nervous system (CNS). NG2 cells are known for their ability to differentiate into oligodendrocytes and are commonly referred to as oligodendrocyte precursor cells. However, recent investigations have begun to identify additional functions of NG2 cells in CNS health and pathology. NG2 cells form physical and functional connections with neurons and other glial cell types throughout the CNS, allowing them to monitor and respond to the neural environment. Growing evidence indicates that NG2 cells become reactive under pathological conditions, though their specific roles are only beginning to be elucidated. While reactive microglia and astrocytes are well-established contributors to neuroinflammation and the development of epilepsy following CNS infection, the dynamics of NG2 cells remain unclear. Therefore, we investigated NG2 cell reactivity in a viral-induced mouse model of temporal lobe epilepsy. Methods C57BL6/J mice were injected intracortically with Theiler’s murine encephalomyelitis virus (TMEV) or PBS. Mice were graded twice daily for seizures between 3 and 7 days post-injection (dpi). At 4 and 14 dpi, brains were fixed and stained for NG2, the microglia/macrophage marker IBA1, and the proliferation marker Ki-67. Confocal z stacks were acquired in both the hippocampus and the overlying cortex. Total field areas stained by each cell marker and total field area of colocalized pixels between NG2 and Ki67 were compared between groups. Results Both NG2 cells and microglia/macrophages displayed increased immunoreactivity and reactive morphologies in the hippocampus of TMEV-injected mice. While increased immunoreactivity for IBA1 was also present in the cortex, there was no significant change in NG2 immunoreactivity in the cortex following TMEV infection. Colocalization analysis for NG2 and Ki-67 revealed a significant increase in overlap between NG2 and Ki-67 in the hippocampus of TMEV-injected mice at both time points, but no significant differences in cortex. Conclusions NG2 cells acquire a reactive phenotype and proliferate in response to TMEV infection. These results suggest that NG2 cells alter their function in response to viral encephalopathy, making them potential targets to prevent the development of epilepsy following viral infection.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Rebeca Sánchez-González ◽  
Nieves Salvador ◽  
Laura López-Mascaraque

Abstract NG2-glia, also referred to as oligodendrocyte precursor cells or polydendrocytes, represent a large pool of proliferative neural cells in the adult brain that lie outside of the two major adult neurogenic niches. Although their roles are not fully understood, we previously reported significant clonal expansion of adult NG2-cells from embryonic pallial progenitors using the StarTrack lineage-tracing tool. To define the contribution of early postnatal progenitors to the specific NG2-glia lineage, we used NG2-StarTrack. A temporal clonal analysis of single postnatal progenitor cells revealed the production of different glial cell types in distinct areas of the dorsal cortex but not neurons. Moreover, the dispersion and size of the different NG2 derived clonal cell clusters increased with age. Indeed, clonally-related NG2-glia were located throughout the corpus callosum and the deeper layers of the cortex. In summary, our data reveal that postnatally derived NG2-glia are proliferative cells that give rise to NG2-cells and astrocytes but not neurons. These progenitors undergo clonal cell expansion and dispersion throughout the adult dorsal cortex in a manner that was related to aging and cell identity, adding new information about the ontogeny of these cells. Thus, identification of clonally-related cells from specific progenitors is important to reveal the NG2-glia heterogeneity.


2020 ◽  
Author(s):  
Laura A Bell ◽  
Glenna J Wallis ◽  
Karen S Wilcox

Abstract BackgroundNeuron-Glial Antigen 2 (NG2) cells are a glial cell type tiled throughout the grey and white matter of the Central Nervous System (CNS). NG2 cells are known for their ability to differentiate into oligodendrocytes and are commonly referred to as oligodendrocyte precursor cells. However, recent investigations have begun to identify additional functions of NG2 cells in CNS health and pathology. NG2 cells form physical and functional connections with neurons and other glial cell types throughout the CNS, allowing them to monitor and respond to the neural environment. Growing evidence indicates that NG2 cells become reactive under pathological conditions, though their specific roles are only beginning to be elucidated. While reactive microglia and astrocytes are well-established contributors to neuroinflammation and the development of epilepsy following CNS infection, the dynamics of NG2 cells remain unclear. Therefore, we investigated NG2 cell reactivity in a viral-induced mouse model of temporal lobe epilepsy. MethodsC57BL6/J mice were injected intracortically with Theiler’s Murine Encephalomyelitis 36 Virus (TMEV) or PBS. Mice were graded twice daily for seizures between 3–7 days post-injection (dpi). At 4 and 14 dpi, brains were fixed and stained for NG2, the microglia/macrophage marker IBA1, and the proliferation marker Ki-67. Confocal z-stacks were acquired in both the hippocampus and the overlying cortex. Total field areas stained by each cell marker and total field area of colocalized pixels between NG2 and Ki67 were compared between groups. ResultsBoth NG2 cells and microglia/macrophages displayed increased immunoreactivity and reactive morphologies in the hippocampus of TMEV-injected mice. While increased immunoreactivity for IBA1 was also present in the cortex, there was no significant change in NG2 immunoreactivity in the cortex following TMEV-infection. Colocalization analysis for NG2 and Ki-67 revealed a significant increase in overlap between NG2 and Ki-67 in the hippocampus of TMEV-injected mice at both timepoints, but no significant differences in cortex.ConclusionsNG2 cells acquire a reactive phenotype and proliferate in response to TMEV-infection. These results suggest that NG2 cells alter their function in response to viral encephalopathy, making them potential targets to prevent the development of epilepsy following viral infection.


2020 ◽  
Author(s):  
Giulia Treccani ◽  
Hatice Yigit ◽  
Thomas Lingner ◽  
Vanessa Schleuβner ◽  
Malin Wennström ◽  
...  

AbstractThe precise mechanisms underlying the detrimental effects of early life stress (ELS) on adult mental health remain still elusive. To date, most studies have exclusively targeted neuronal populations and not considered neuron-glia crosstalk as a crucially important element for the integrity of stress-related brain function. Here, we have investigated the impact of ELS on a glial subpopulation with unique properties in brain homeostasis, the NG2+ cells. ELS shifted the NG2+ transcriptome towards more mature stages, and these transcriptional effects were dependent on stress-induced glucocorticoids. The functional relevance of one candidate gene, Scn7a, could be confirmed by an increase in the density of voltage-gated sodium (Nav) channel activated currents in hippocampal NG2+ cells. Scn7a remained upregulated until adulthood in ELS animals, and these same animals displayed impaired cognitive performance. Considering that Nav channels are important for NG2+ cell-to-neuron communication, our findings suggest novel insights into the pathophysiology of stress-related mental disorders.


2020 ◽  
Vol 127 (Suppl_1) ◽  
Author(s):  
Linda Alex ◽  
Ya Su ◽  
Nikolaos G Frangogiannis

Repair of the infarcted heart is dependent on inflammation-driven activation of myofibroblasts (MFs) and subsequent formation of a scar. Though pericytes have been implicated in injury-associated fibroblast activation in several organs, their potential role in cardiac repair and fibrosis has not been studied. We hypothesized that myocardial infarction (MI) may induce pericyte activation, contributing to repair through pericyte to MF conversion, secretion of fibrogenic mediators, or regulation of angiogenesis. In order to test the hypothesis, we generated pericyte/fibroblast reporter mice (NG2 DsRed ;PDGFRα GFP ). In normal myocardium, NG2 labeled peri-endothelial mural cells that coexpressed PDGFRβ, whereas PDGFRα identified interstitial cells with fibroblast characteristics. Pericytes and fibroblasts had distinct transcriptomic profiles: NG2+/PDGFRα- pericytes expressed αSMA and low amounts of extracellular matrix (ECM) genes, whereas PDGFRα+/NG2- fibroblasts synthesized collagens. Pericyte rarefaction was noted in the necrotic core 3 days after non-reperfused MI. 3-7 days post MI, expansion of the NG2+ population in the infarct zone was associated with emergence of non-mural NG2+/αSMA+ cells with MF characteristics. FACS-sorted NG2+/PDGFRα- cells from 7-day infarcts expressed higher levels of ColIα2 (7.2±1.0-fold) and ColIIIα1 (8.9±1.14-fold), when compared to NG2+/PDGFRα- cells from normal hearts. NG2+ cells had high mRNA levels of integrins α1, αV, β1, and β5, and of MMP14, reflecting an activated migratory phenotype. To examine whether expression of ECM genes by infarct pericytes is due to fibroblast conversion, we did lineage tracing studies using NG2CreER TM ;Rosa tdTomato mice bred with the PDGFRα GFP line for reliable fibroblast identification. 7 days post MI, 5.7%±1.04 of PDGFRα+ fibroblasts were derived from NG2+ cells. Also, αSMA staining showed that 10.49%±2.73 of infarct MFs were derived from NG2+ lineage. The majority of mural cells wrapping neovessels were derived from NG2+ cells, suggesting a role for resident pericytes in infarct angiogenesis. In conclusion, upon MI, pericytes become activated and contribute to repair by undergoing conversion to a subset of myofibroblasts and by coating infarct neovessels.


2020 ◽  
Vol 50 ◽  
pp. 53-60 ◽  
Author(s):  
Denisa Kirdajova ◽  
Miroslava Anderova
Keyword(s):  

2019 ◽  
Author(s):  
Rui Gong ◽  
Min Zhang ◽  
Kexin Sun ◽  
Wei Qi ◽  
Shanquan Sun ◽  
...  

Abstract Purpose Investigating the roles of phosphorylated epidermal growth factor receptor (pEGFR) in the recovery of neural function after decompression of CSCI, therefore provide experimental basis for the development of therapeutic strategies and medicines for treating CSCI. Methods A CSCI model was established with a customized device, and was then subjected to spinal decompression. The motor functions were monitored by the Basso, Beattie & Bresnahan(BBB) locomotor rating scale; the number of axonal myelinated fibers was estimated by staining with luxol fast blue (LFB); pEGFR and phosphorylated Akt1 (pAkt1) were detected by Western blot; pEGFR+-NG2+(NG2+ cells are precursor to oligodendrocytes and pAkt1+-NG2+ cells were detected by double-labeling immunefluorescence assay. Results After decompression of CSCI, the BBB scores and the number of myelinated nerve fibers gradually increased with time. Meanwhile, the expression of pEGFR and pAkt1 were up-regulated and the number of pEGFR+-NG2+ and pAkt1+-NG2+ cells increased consistent with the changes of motor functions and the number of myelinated nerve fibers. Whereas, significant decreases in BBB scores, expression level of pAkt1, as well as numbers of myelinated nerve fibers, and pAkt1+-NG2+ cells were observed after inhibition of expression. Conclusions Up-regulated expression of pEGFR can promote recovery of neurological functions in rats with CSCI. This effect is achieved by activation of pAkt1(a downstream signal molecule of pEGFR), which subsequently promotes the proliferation of oligodendrocyte precursor cells (OPCs).


Sign in / Sign up

Export Citation Format

Share Document