Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Neuroscience ; 381: 124-137, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29689292

ABSTRACT

Molecular and functional diversity within midbrain dopaminergic (mDA) and hindbrain serotonergic (5-HT) neurons has emerged as a relevant feature that could underlie selective vulnerability of neurons in clinical disorders. We have investigated the role of transforming growth factor beta (TGF-ß) during development of mDA and 5-HT subgroups. We have generated TßRIIflox/flox::En1cre/+ mice where type II TGF-ß receptor is conditionally deleted from engrailed 1-expressing cells and have investigated the hindbrain serotonergic system of these mice together with Tgf-ß2-/- mice. The results show a significant decrease in the number of 5-HT neurons in TGF-ß2-deficient mice at embryonic day (E) 12 and a selective significant decrease in the hindbrain paramedian raphe 5-HT neurons at E18, compared to wild type. Moreover, conditional deletion of TGF-ß signaling from midbrain and rhombomere 1 leads to inactive TGF-ß signaling in cre-expressing cells, impaired development of mouse mDA neuron subgroups and of dorsal raphe 5-HT neuron subgroups in a temporal manner. These results highlight a selective growth factor dependency of individual rostral hindbrain serotonergic subpopulations, emphasize the impact of TGF-ß signaling during development of mDA and 5-HT subgroups, and suggest TGF-ßs as potent candidates to establish diversity within the hindbrain serotonergic system. Thus, the data contribute to a better understanding of development and degeneration of mDA neurons and 5-HT-associated clinical disorders.


Subject(s)
Dopaminergic Neurons/cytology , Mesencephalon/embryology , Neurogenesis/physiology , Rhombencephalon/embryology , Serotonergic Neurons/cytology , Transforming Growth Factor beta/metabolism , Animals , Embryo, Mammalian , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Rhombencephalon/cytology , Signal Transduction/physiology
2.
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
3.
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
4.
Int J Biochem Cell Biol ; 44(8): 1254-65, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22561749

ABSTRACT

Changes in systemic acid-base homeostasis cause a series of organ-specific cellular responses, among them changes of acid-base transporter activities, and recruitment or retrieval of these transporters from intracellular pools to the plasma membrane and vice versa. The purpose of this study was to investigate the impact of protein phosphorylation in the acidosis-induced translocation of vacuolar-type H(+)-ATPase (V-ATPase) in salivary ducts and to identify molecular targets. Therefore, the human submandibular gland cell line HSG was exposed to acidosis and V-ATPase trafficking was investigated in the presence or absence of inhibitors and activators of sAC/PKA and Src/ERK signaling pathways. Putative target genes have been identified by RT-PCR and immunoblotting, and validated by loss-of-function experiments. Acidosis caused activation of cAMP/PKA and Src signaling and inhibition of either pathway significantly impaired acidosis-induced V-ATPase redistribution and incorporation into the plasma membrane. Activation of ERK1/2 was Src-independent, whereas activation of PKA caused phosphorylation of cAMP response element-binding (CREB) and activation to regulate Rab11b transcription. Loss-of-function of CREB down-regulated Rab11b transcript and protein and significantly impaired acidosis-induced V-ATPase translocation in HSG cells. These data demonstrate that the cAMP/PKA/CREB signaling pathway initiates acidosis-induced V-ATPase trafficking in salivary ducts via regulation of Rab11b expression and provide first evidence for a molecular mechanism underlying cAMP/PKA-dependent transporter trafficking that could account for accumulation and activity of transporters in other cellular systems as well.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Salivary Ducts/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , rab GTP-Binding Proteins/metabolism , Acidosis , Cell Line , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique , Humans , Hydrogen-Ion Concentration , Immunoblotting , Isoquinolines/pharmacology , Membrane Glycoproteins/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Subunits/metabolism , Protein Transport/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Salivary Ducts/cytology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Time Factors , rab GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...