Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Psychiatry ; 2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36878967

ABSTRACT

Maternal immune activation (MIA) during critical windows of gestation is correlated with long-term neurodevelopmental deficits in the offspring, including increased risk for autism spectrum disorder (ASD) in humans. Interleukin 6 (IL-6) derived from the gestational parent is one of the major molecular mediators by which MIA alters the developing brain. In this study, we establish a human three-dimensional (3D) in vitro model of MIA by treating induced pluripotent stem cell-derived dorsal forebrain organoids with a constitutively active form of IL-6, Hyper-IL-6. We validate our model by showing that dorsal forebrain organoids express the molecular machinery necessary for responding to Hyper-IL-6 and activate STAT signaling upon Hyper-IL-6 treatment. RNA sequencing analysis reveals the upregulation of major histocompatibility complex class I (MHCI) genes in response to Hyper-IL-6 exposure, which have been implicated with ASD. We find a small increase in the proportion of radial glia cells after Hyper-IL-6 treatment through immunohistochemistry and single-cell RNA-sequencing. We further show that radial glia cells are the cell type with the highest number of differentially expressed genes, and Hyper-IL-6 treatment leads to the downregulation of genes related to protein translation in line with a mouse model of MIA. Additionally, we identify differentially expressed genes not found in mouse models of MIA, which might drive species-specific responses to MIA. Finally, we show abnormal cortical layering as a long-term consequence of Hyper-IL-6 treatment. In summary, we establish a human 3D model of MIA, which can be used to study the cellular and molecular mechanisms underlying the increased risk for developing disorders such as ASD.

2.
J Cell Physiol ; 236(3): 2036-2050, 2021 03.
Article in English | MEDLINE | ID: mdl-32761631

ABSTRACT

The electrogenic Na+ /HCO3- cotransporter (NBCe1) in astrocytes is crucial in regulation of acid-base homeostasis in the brain. Since many pathophysiological conditions in the brain have been associated with pH shifts we exposed primary mouse cortical and hippocampal astrocytes to prolonged low or high extracellular pH (pHo ) at constant extracellular bicarbonate concentration and investigated activation of astrocytes and regulation of NBCe1 by immunoblotting, biotinylation of surface proteins, and intracellular H+ recordings. High pHo at constant extracellular bicarbonate caused upregulation of NBCe1 protein, surface expression and activity via upregulation of the astrocytic activation markers signal transducer and activator of transcription 3 (STAT3) signaling and glial fibrillary acidic protein expression. High pHo -induced increased NBCe1 protein expression was prevented in astrocytes from Stat3flox/flox ::GfapCre/+ mice. In vitro, basal and high pHo -induced increased NBCe1 functional expression was impaired following inhibition of STAT3 phosphorylation. These results provide a novel regulation mode of NBCe1 protein and activity, highlight the importance of astrocyte reactivity on regulation of NBCe1 and implicate roles for NBCe1 in altering/modulating extracellular pH during development as well as of the microenvironment at sites of brain injuries and other pathophysiological conditions.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/cytology , STAT3 Transcription Factor/metabolism , Sodium-Bicarbonate Symporters/metabolism , Animals , Astrocytes/drug effects , Bicarbonates/pharmacology , Biomarkers/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Extracellular Space/metabolism , Hippocampus/cytology , Hydrogen-Ion Concentration , Mice, Inbred C57BL , Models, Biological , Protein Transport/drug effects , Signal Transduction/drug effects
3.
Sci Rep ; 10(1): 17661, 2020 10 19.
Article in English | MEDLINE | ID: mdl-33077786

ABSTRACT

Different types of carbon materials are biocompatible with neural cells and can promote maturation. The mechanism of this effect is not clear. Here we have tested the capacity of a carbon material composed of amorphous sp3 carbon backbone, embedded with a percolating network of sp2 carbon domains to sustain neuronal cultures. We found that cortical neurons survive and develop faster on this novel carbon material. After 3 days in culture, there is a precocious increase in the frequency of neuronal activity and in the expression of maturation marker KCC2 on carbon films as compared to a commonly used glass surface. Accelerated development is accompanied by a dramatic increase in neuronal dendrite arborization. The mechanism for the precocious maturation involves the activation of intracellular calcium oscillations by the carbon material already after 1 day in culture. Carbon-induced oscillations are independent of network activity and reflect intrinsic spontaneous activation of developing neurons. Thus, these results reveal a novel mechanism for carbon material-induced neuronal survival and maturation.


Subject(s)
Calcium/metabolism , Carbon , Cell Differentiation , Neurons/physiology , Dendrites/physiology , Humans , Nerve Net , Neurons/metabolism
4.
Brain Res ; 1742: 146803, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32240655

ABSTRACT

Rodent models have catalyzed major discoveries in the neocortex, a brain region unique to mammals. However, since the neocortex has expanded considerably in primates, employing rodent models has limitations. Human fetal brain tissue is a scarce resource with limitations for experimental manipulations. In order to create an experimentally tractable representation of human brain development, a number of labs have recently created in vitro models of the developing human brain. These models, generated using human embryonic stem cells or induced pluripotent stem cells, are called "organoids". Organoids have successfully and rapidly uncovered new mechanisms of human brain development in health and disease. In the future, we envision that this strategy will enable faster and more efficient translation of basic neuroscience findings to therapeutic applications. In this review, we discuss the generation of the first human cerebral organoids, progress since their debut, and challenges to be overcome in the future.


Subject(s)
Cell Culture Techniques/methods , Neocortex/growth & development , Organoids/growth & development , Brain/growth & development , Humans , Induced Pluripotent Stem Cells , Models, Biological , Neocortex/physiology , Neurogenesis , Organoids/physiology
5.
Glia ; 67(12): 2264-2278, 2019 12.
Article in English | MEDLINE | ID: mdl-31318482

ABSTRACT

The electrogenic sodium bicarbonate cotransporter 1, NBCe1 (SLC4A4), is the major bicarbonate transporter expressed in astrocytes. It is highly sensitive for bicarbonate and the main regulator of intracellular, extracellular, and synaptic pH, thereby modulating neuronal excitability. However, despite these essential functions, the molecular mechanisms underlying NBCe1-mediated astrocytic response to extracellular pH changes are mostly unknown. Using primary mouse cortical astrocyte cultures, we investigated the effect of long-term extracellular metabolic alkalosis on regulation of NBCe1 and elucidated the underlying molecular mechanisms by immunoblotting, biotinylation of surface proteins, intracellular H+ recording using the H+ -sensitive dye 2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein, and phosphoproteomic analysis. The results showed significant downregulation of NBCe1 activity following metabolic alkalosis without influencing protein abundance or surface expression of NBCe1. During alkalosis, the rate of intracellular H+ changes upon challenging NBCe1 was decreased in wild-type astrocytes, but not in cortical astrocytes from NBCe1-deficient mice. Alkalosis-induced decrease of NBCe1 activity was rescued after activation of mTOR signaling. Moreover, mass spectrometry revealed constitutively phosphorylated S255-257 and mutational analysis uncovered these residues being crucial for NBCe1 transport activity. Our results demonstrate a novel mTOR-regulated mechanism by which NBCe1 functional expression is regulated. Such mechanism likely applies not only for NBCe1 in astrocytes, but in epithelial cells as well.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Sodium-Bicarbonate Symporters/biosynthesis , TOR Serine-Threonine Kinases/physiology , Alkalosis/metabolism , Alkalosis/pathology , Animals , Cells, Cultured , Female , Gene Expression , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/physiology , Sodium-Bicarbonate Symporters/genetics
6.
J Cell Physiol ; 234(9): 15061-15079, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30648263

ABSTRACT

Bicarbonate concentration in saliva is controlled by the action of acid-base transporters in salivary duct cells. We show for the first time expression of ATP6V1B1 in submandibular gland and introduce transforming growth factor-beta (TGF-ß) as a novel regulator of V-ATPase subunits. Using QRT-PCR, immunoblotting, biotinylation of surface proteins, immunofluorescence, chromatin immunoprecipitation, and intracellular H(+ ) recording with H(+ )-sensitive dye 2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein we show that in the human submandibular gland (HSG) cell line, activation of TGF-ß signaling upregulates ATP6V1E1 and ATP6V1B2, downregulates ATP6V1B1, and has no effect on ATP6V1A. TGF-ß1 effects on ATP6V1B1 are mediated through the canonical, the soluble adenylate cyclase, and ERK signaling. A CREB binding sequence was identified in the ATP6V1B1 promoter and CREB binding decreased after TGF-ß1 treatment. Following acidosis, a bafilomycin-sensitive and Na+ -independent cell pH recovery was observed in HSG cells, an effect that was not influenced after disruption of acidic lysosomes. Moreover, neutralization of TGF-ßs, inhibition of TGF-ß receptor, or inhibition of the canonical pathway decreased membrane expression of ATP6V1A and prevented the acidosis-induced increased V-ATPase activity. The results suggest multiple modes of action of TGF-ß1 on V-ATPase subunits in HSG cells: TGF-ß1 may regulate transcription or protein synthesis of certain subunits and trafficking of other subunits in a context-dependent manner. Moreover, surface V-ATPase is active in salivary duct cells and involved in intracellular pH regulation following acidosis.

7.
Cell Tissue Res ; 370(2): 211-225, 2017 11.
Article in English | MEDLINE | ID: mdl-28799057

ABSTRACT

The temporal dynamic expression of Sonic Hedgehog (SHH) and signaling during early midbrain dopaminergic (mDA) neuron development is one of the key players in establishing mDA progenitor diversity. However, whether SHH signaling is also required during later developmental stages and in mature mDA neurons is less understood. We study the expression of SHH receptors Ptch1 and Gas1 (growth arrest-specific 1) and of the transcription factors Gli1, Gli2 and Gli3 in mouse midbrain during embryonic development [embryonic day (E) 12.5 onwards)], in newborn and adult mice using in situ hybridization and immunohistochemistry. Moreover, we examine the expression and regulation of dopaminergic neuronal progenitor markers, midbrain dopaminergic neuronal markers and markers of the SHH signaling pathway in undifferentiated and butyric acid-treated (differentiated) MN9D cells in the presence or absence of exogenous SHH in vitro by RT-PCR, immunoblotting and immunocytochemistry. Gli1 was expressed in the lateral mesencephalic domains, whereas Gli2 and Gli3 were expressed dorsolaterally and complemented by ventrolateral expression of Ptch1. Co-localization with tyrosine hydroxylase could not be observed. GAS1 was exclusively expressed in the dorsal mesencephalon at E11.5 and co-localized with Ki67. In contrast, MN9D cells expressed all the genes investigated and treatment of the cells with butyric acid significantly upregulated their expression. The results suggest that SHH is only indirectly involved in the differentiation and survival of mDA neurons and that the MN9D cell line is a valuable model for investigating early development but not the differentiation and survival of mDA neurons.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Mesencephalon/growth & development , Animals , Animals, Newborn , Cell Line , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Hedgehog Proteins/analysis , Immunohistochemistry , In Situ Hybridization , Mesencephalon/chemistry , Mesencephalon/embryology , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
8.
Glia ; 65(8): 1361-1375, 2017 08.
Article in English | MEDLINE | ID: mdl-28568893

ABSTRACT

The electrogenic sodium bicarbonate cotransporter NBCe1 (SLC4A4) expressed in astrocytes regulates intracellular and extracellular pH. Here, we introduce transforming growth factor beta (TGF-ß) as a novel regulator of NBCe1 transcription and functional expression. Using hippocampal slices and primary hippocampal and cortical astrocyte cultures, we investigated regulation of NBCe1 and elucidated the underlying signaling pathways by RT-PCR, immunoblotting, immunofluorescence, intracellular H(+ ) recording using the H(+ ) -sensitive dye 2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein, mink lung epithelial cell (MLEC) assay, and chromatin immunoprecipitation. Activation of TGF-ß signaling significantly upregulated transcript, protein, and surface expression of NBCe1. These effects were TGF-ß receptor-mediated and suppressed following inhibition of JNK and Smad signaling. Moreover, 4-aminopyridine (4AP)-dependent NBCe1 regulation requires TGF-ß. TGF-ß increased the rate and amplitude of intracellular H+ changes upon challenging NBCe1 in wild-type astrocytes but not in cortical astrocytes from Slc4a4-deficient mice. A Smad4 binding sequence was identified in the NBCe1 promoter and Smad4 binding increased after activation of TGF-ß signaling. The data show for the first time that NBCe1 is a direct target of TGF-ß/Smad4 signaling. Through activation of the canonical pathway TGF-ß acts directly on NBCe1 by binding of Smad4 to the NBCe1 promoter and regulating its transcription, followed by increased protein expression and transport activity.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation/physiology , Signal Transduction/physiology , Sodium-Bicarbonate Symporters/metabolism , Transforming Growth Factor beta/metabolism , 4-Aminopyridine/pharmacology , Aldehyde Dehydrogenase 1 Family , Animals , Benzamides/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Chloride-Bicarbonate Antiporters/pharmacology , Dioxoles/pharmacology , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Hydrogen-Ion Concentration , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium Channel Blockers/pharmacology , Retinal Dehydrogenase/metabolism , Signal Transduction/drug effects , Smad4 Protein/metabolism , Sodium-Bicarbonate Symporters/antagonists & inhibitors , Sodium-Bicarbonate Symporters/genetics , Transforming Growth Factor beta/genetics
9.
J Cell Sci ; 129(18): 3485-98, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27505893

ABSTRACT

Functional activation of the neuronal K(+)-Cl(-) co-transporter KCC2 (also known as SLC12A5) is a prerequisite for shifting GABAA responses from depolarizing to hyperpolarizing during development. Here, we introduce transforming growth factor ß2 (TGF-ß2) as a new regulator of KCC2 membrane trafficking and functional activation. TGF-ß2 controls membrane trafficking, surface expression and activity of KCC2 in developing and mature mouse primary hippocampal neurons, as determined by immunoblotting, immunofluorescence, biotinylation of surface proteins and KCC2-mediated Cl(-) extrusion. We also identify the signaling pathway from TGF-ß2 to cAMP-response-element-binding protein (CREB) and Ras-associated binding protein 11b (Rab11b) as the underlying mechanism for TGF-ß2-mediated KCC2 trafficking and functional activation. TGF-ß2 increases colocalization and interaction of KCC2 with Rab11b, as determined by 3D stimulated emission depletion (STED) microscopy and co-immunoprecipitation, respectively, induces CREB phosphorylation, and enhances Rab11b gene expression. Loss of function of either CREB1 or Rab11b suppressed TGF-ß2-dependent KCC2 trafficking, surface expression and functionality. Thus, TGF-ß2 is a new regulatory factor for KCC2 functional activation and membrane trafficking, and a putative indispensable molecular determinant for the developmental shift of GABAergic transmission.


Subject(s)
Cell Membrane/metabolism , Symporters/metabolism , Transforming Growth Factor beta2/pharmacology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Hippocampus/cytology , Humans , Intracellular Space/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Symporters/drug effects , rab GTP-Binding Proteins/metabolism , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...