Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 10678, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38724551

ABSTRACT

Mutations in LRBA, a BEACH domain protein, cause severe immune deficiency in humans. LRBA is expressed in many tissues and organs according to biochemical analysis, but little is known about its cellular and subcellular localization, and its deficiency phenotype outside the immune system. By LacZ histochemistry of Lrba gene-trap mice, we performed a comprehensive survey of LRBA expression in numerous tissues, detecting it in many if not all epithelia, in exocrine and endocrine cells, and in subpopulations of neurons. Immunofluorescence microscopy of the exocrine and endocrine pancreas, salivary glands, and intestinal segments, confirmed these patterns of cellular expression and provided information on the subcellular localizations of the LRBA protein. Immuno-electron microscopy demonstrated that in neurons and endocrine cells, which co-express LRBA and its closest relative, neurobeachin, both proteins display partial association with endomembranes in complementary, rather than overlapping, subcellular distributions. Prominent manifestations of human LRBA deficiency, such as inflammatory bowel disease or endocrinopathies, are believed to be primarily due to immune dysregulation. However, as essentially all affected tissues also express LRBA, it is possible that LRBA deficiency enhances their vulnerability and contributes to the pathogenesis.


Subject(s)
Endocrine Glands , Epithelium , Exocrine Glands , Immunologic Deficiency Syndromes , Neurons , Animals , Humans , Mice , Endocrine Glands/metabolism , Epithelium/metabolism , Exocrine Glands/metabolism , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Mutation , Neurons/metabolism
2.
Cells ; 12(23)2023 11 21.
Article in English | MEDLINE | ID: mdl-38067105

ABSTRACT

Ischemic conditions cause an increase in the sodium concentration of astrocytes, driving the breakdown of ionic homeostasis and exacerbating cellular damage. Astrocytes express high levels of the electrogenic sodium-bicarbonate cotransporter1 (NBCe1), which couples intracellular Na+ homeostasis to regulation of pH and operates close to its reversal potential under physiological conditions. Here, we analyzed its mode of operation during transient energy deprivation via imaging astrocytic pH, Na+, and ATP in organotypic slice cultures of the mouse neocortex, complemented with patch-clamp and ion-selective microelectrode recordings and computational modeling. We found that a 2 min period of metabolic failure resulted in a transient acidosis accompanied by a Na+ increase in astrocytes. Inhibition of NBCe1 increased the acidosis while decreasing the Na+ load. Similar results were obtained when comparing ion changes in wild-type and Nbce1-deficient mice. Mathematical modeling replicated these findings and further predicted that NBCe1 activation contributes to the loss of cellular ATP under ischemic conditions, a result confirmed experimentally using FRET-based imaging of ATP. Altogether, our data demonstrate that transient energy failure stimulates the inward operation of NBCe1 in astrocytes. This causes a significant amelioration of ischemia-induced astrocytic acidification, albeit at the expense of increased Na+ influx and a decline in cellular ATP.


Subject(s)
Acidosis , Neocortex , Mice , Animals , Astrocytes/metabolism , Sodium-Bicarbonate Symporters/metabolism , Mice, Knockout , Neocortex/metabolism , Ions/metabolism , Sodium/metabolism , Acidosis/metabolism , Adenosine Triphosphate/metabolism
3.
Front Cell Neurosci ; 17: 1253424, 2023.
Article in English | MEDLINE | ID: mdl-37881493

ABSTRACT

K+/Cl- cotransporter 2 (KCC2) is a major Cl- extruder in mature neurons and is responsible for the establishment of low intracellular [Cl-], necessary for fast hyperpolarizing GABAA-receptor mediated synaptic inhibition. Electrogenic sodium bicarbonate cotransporter 1 (NBCe1) is a pH regulatory protein expressed in neurons and glial cells. An interactome study identified NBCe1 as a possible interaction partner of KCC2. In this study, we investigated the putative effect of KCC2/NBCe1 interaction in baseline and the stimulus-induced phosphorylation pattern and function of KCC2. Primary mouse hippocampal neuronal cultures from wildtype (WT) and Nbce1-deficient mice, as well as HEK-293 cells stably transfected with KCC2WT, were used. The results show that KCC2 and NBCe1 are interaction partners in the mouse brain. In HEKKCC2 cells, pharmacological inhibition of NBCs with S0859 prevented staurosporine- and 4-aminopyridine (4AP)-induced KCC2 activation. In mature cultures of hippocampal neurons, however, S0859 completely inhibited postsynaptic GABAAR and, thus, could not be used as a tool to investigate the role of NBCs in GABA-dependent neuronal networks. In Nbce1-deficient immature hippocampal neurons, baseline phosphorylation of KCC2 at S940 was downregulated, compared to WT, and exposure to staurosporine failed to reduce pKCC2 S940 and T1007. In Nbce1-deficient mature neurons, baseline levels of pKCC2 S940 and T1007 were upregulated compared to WT, whereas after 4AP treatment, pKCC2 S940 was downregulated, and pKCC2 T1007 was further upregulated. Functional experiments showed that the levels of GABAAR reversal potential, baseline intracellular [Cl-], Cl- extrusion, and baseline intracellular pH were similar between WT and Nbce1-deficient neurons. Altogether, our data provide a primary description of the properties of KCC2/NBCe1 protein-protein interaction and implicate modulation of stimulus-mediated phosphorylation of KCC2 by NBCe1/KCC2 interaction-a mechanism with putative pathophysiological relevance.

4.
Am J Physiol Renal Physiol ; 325(5): F564-F577, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37589051

ABSTRACT

The transmembrane protein SLC22A17 [or the neutrophil gelatinase-associated lipocalin/lipocalin-2 (LCN2)/24p3 receptor] is an atypical member of the SLC22 family of organic anion and cation transporters: it does not carry typical substrates of SLC22 transporters but mediates receptor-mediated endocytosis (RME) of LCN2. One important task of the kidney is the prevention of urinary loss of proteins filtered by the glomerulus by bulk reabsorption of multiple ligands via megalin:cubilin:amnionless-mediated endocytosis in the proximal tubule (PT). Accordingly, overflow, glomerular, or PT damage, as in Fanconi syndrome, results in proteinuria. Strikingly, up to 20% of filtered proteins escape the PT under physiological conditions and are reabsorbed by the distal nephron. The renal distal tubule and collecting duct express SLC22A17, which mediates RME of filtered proteins that evade the PT but with limited capacity to prevent proteinuria under pathological conditions. The kidney also prevents excretion of filtered essential and nonessential transition metals, such as iron or cadmium, respectively, that are largely bound to proteins with high affinity, e.g., LCN2, transferrin, or metallothionein, or low affinity, e.g., microglobulins or albumin. Hence, increased uptake of transition metals may cause nephrotoxicity. Here, we assess the literature on SLC22A17 structure, topology, tissue distribution, regulation, and assumed functions, emphasizing renal SLC22A17, which has relevance for physiology, pathology, and nephrotoxicity due to the accumulation of proteins complexed with transition metals, e.g., cadmium or iron. Other putative renal functions of SLC22A17, such as its contribution to osmotic stress adaptation, protection against urinary tract infection, or renal carcinogenesis, are discussed.


Subject(s)
Metalloproteins , Nephrosis , Humans , Lipocalin-2/metabolism , Metalloproteins/metabolism , Cadmium/metabolism , Iron/metabolism , Metallothionein/metabolism , Kidney Tubules, Proximal/metabolism , Proteinuria/metabolism , Nephrosis/metabolism , Endocytosis , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Organic Cation Transport Proteins/metabolism
5.
Cells ; 11(23)2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36497119

ABSTRACT

KCC2 mediates extrusion of K+ and Cl- and assuresthe developmental "switch" in GABA function during neuronal maturation. However, the molecular mechanisms underlying KCC2 regulation are not fully elucidated. We investigated the impact of transforming growth factor beta 2 (TGF-ß2) on KCC2 during neuronal maturation using quantitative RT-PCR, immunoblotting, immunofluorescence and chromatin immunoprecipitation in primary mouse hippocampal neurons and brain tissue from Tgf-ß2-deficient mice. Inhibition of TGF-ß/activin signaling downregulates Kcc2 transcript in immature neurons. In the forebrain of Tgf-ß2-/- mice, expression of Kcc2, transcription factor Ap2ß and KCC2 protein is downregulated. AP2ß binds to Kcc2 promoter, a binding absent in Tgf-ß2-/-. In hindbrain/brainstem tissue of Tgf-ß2-/- mice, KCC2 phosphorylation at T1007 is increased and approximately half of pre-Bötzinger-complex neurons lack membrane KCC2 phenotypes rescued through exogenous TGF-ß2. These results demonstrate that TGF-ß2 regulates KCC2 transcription in immature neurons, possibly acting upstream of AP2ß, and contributes to the developmental dephosphorylation of KCC2 at T1007. The present work suggests multiple and divergent roles for TGF-ß2 on KCC2 during neuronal maturation and provides novel mechanistic insights for TGF-ß2-mediated regulation of KCC2 gene expression, posttranslational modification and surface expression. We propose TGF-ß2 as a major regulator of KCC2 with putative implications for pathophysiological conditions.


Subject(s)
Neural Stem Cells , Symporters , Transforming Growth Factor beta2 , Animals , Mice , Hippocampus/cytology , Hippocampus/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Phosphorylation , Transforming Growth Factor beta2/metabolism , Symporters/metabolism , K Cl- Cotransporters
6.
Int J Mol Sci ; 23(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36012235

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and malignant brain tumour. It is characterised by transcriptionally distinct cell populations. In tumour cells, physiological pH gradients between the intracellular and extracellular compartments are reversed, compared to non-cancer cells. Intracellular pH in tumour cells is alkaline, whereas extracellular pH is acidic. Consequently, the function and/or expression of pH regulating transporters might be altered. Here, we investigated protein expression and regulation of the electrogenic sodium/bicarbonate cotransporter 1 (NBCe1) in mesenchymal (MES)-like hypoxia-dependent and -independent cells, as well as in astrocyte-like glioblastoma cells following chemical hypoxia, acidosis and elucidated putative underlying molecular pathways. Immunoblotting, immunocytochemistry, and intracellular pH recording with the H+-sensitive dye 2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein were applied. The results show NBCe1 protein abundance and active NBCe1 transport. Hypoxia upregulated NBCe1 protein and activity in MES-like hypoxia-dependent GBM cells. This effect was positively correlated with HIF-1α protein levels, was mediated by TGF-ß signalling, and was prevented by extracellular acidosis. In MES-like hypoxia-independent GBM cells, acidosis (but not hypoxia) regulated NBCe1 activity in an HIF-1α-independent manner. These results demonstrate a cell-specific adaptation of NBCe1 expression and activity to the microenvironment challenge of hypoxia and acidosis that depends on their transcriptional signature in GBM.


Subject(s)
Acidosis , Glioblastoma , Symporters , Humans , Sodium/metabolism , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism , Tumor Microenvironment
7.
J Cell Biochem ; 123(6): 1120-1129, 2022 06.
Article in English | MEDLINE | ID: mdl-35533251

ABSTRACT

Permanent degeneration and loss of dopaminergic (DA) neurons in substantia nigra is the main cause of Parkinson's disease. Considering the therapeutic application of stem cells in neurodegeneration, we sought to examine the neurogenic differentiation potential of the newly introduced neural crest originated mesenchymal stem cells (MSCs), namely, trabecular meshwork-derived mesenchymal stem cells (TM-MSCs) compared to two other sources of MSCs, adipose tissue-derived stem cells (ADSCs) and bone marrow-derived mesenchymal stem cells (BM-MSCs). The three types of cells were therefore cultured in the presence and absence of a neural induction medium followed by the analysis of their differentiation potentials. Our results showed that TM-MSCs exhibited enhanced neural morphologies as well as higher expressions of MAP2 as the general neuron marker and Nurr-1 as an early DA marker compared to the adipose tissue-derived mesenchymal stem cells (AD-MSCs) and bone marrow-derived stem cells (BMSCs). Also, analysis of Nurr-1 immunostaining showed more intense Nurr-1 stained nuclei in the neurally induced TM-MSCs compared to those in the AD-MSCs, BMSCs, and noninduced control TM-MSCs. To examine if Wnt/beta-catenin pathway drives TM-MSCs towards a DA fate, we treated them with the Wnt agonist (CHIR, 3 µM) and the Wnt antagonist (IWP-2, 3 µM). Our results showed that the expressions of Nurr-1 and MAP2, as well as the Wnt/beta-catenin target genes, c-Myc and Cyclin D1, were significantly increased in the CHIR-treated TM-MSCs, but significantly reduced in those treated with IWP-2. Altogether, we declare first a higher neural potency of TM-MSCs compared to the more commonly used MSCs, BMSCs and ADSCs, and second that Wnt/beta-catenin activation directs the neurally induced TM-MSCs towards a DA fate.


Subject(s)
Mesenchymal Stem Cells , Wnt Signaling Pathway , Cell Differentiation , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Trabecular Meshwork/metabolism , beta Catenin/metabolism
8.
J Cell Physiol ; 237(2): 1372-1388, 2022 02.
Article in English | MEDLINE | ID: mdl-34642952

ABSTRACT

Astrocytes are pivotal responders to alterations of extracellular pH, primarily by regulation of their principal acid-base transporter, the membrane-bound electrogenic Na+ /bicarbonate cotransporter 1 (NBCe1). Here, we describe amammalian target of rapamycin (mTOR)-dependent and NBCe1-mediated astroglial response to extracellular acidosis. Using primary mouse cortical astrocytes, we investigated the effect of long-term extracellular metabolic acidosis 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 increase of NBCe1-mediated recovery of intracellular pH from acidification in WT astrocytes, but not in cortical astrocytes from NBCe1-deficient mice. Acidosis-induced upregulation of NBCe1 activity was prevented following inhibition of mTOR signaling by rapamycin. Yet, during acidosis or following exposure of astrocytes to rapamycin, surface protein abundance of NBCe1 remained -unchanged. Mutational analysis in HeLa cells suggested that NBCe1 activity was dependent on phosphorylation state of Ser245 , a residue conserved in all NBCe1 variants. Moreover, phosphorylation state of Ser245 is regulated by mTOR and is inversely correlated with NBCe1 transport activity. Our results identify pSer245 as a novel regulator of NBCe1 functional expression. We propose that context-dependent and mTOR-mediated multisite phosphorylation of serine residues of NBCe1 is likely to be a potent mechanism contributing to the response of astrocytes to acid/base challenges during pathophysiological conditions.


Subject(s)
Acidosis , Symporters , Acidosis/metabolism , Animals , Cerebral Cortex , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Sirolimus/pharmacology , Sodium/metabolism , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism , Symporters/metabolism , TOR Serine-Threonine Kinases/metabolism
9.
Arch Toxicol ; 95(8): 2719-2735, 2021 08.
Article in English | MEDLINE | ID: mdl-34181029

ABSTRACT

The liver hormone hepcidin regulates systemic iron homeostasis. Hepcidin is also expressed by the kidney, but exclusively in distal nephron segments. Several studies suggest hepcidin protects against kidney damage involving Fe2+ overload. The nephrotoxic non-essential metal ion Cd2+ can displace Fe2+ from cellular biomolecules, causing oxidative stress and cell death. The role of hepcidin in Fe2+ and Cd2+ toxicity was assessed in mouse renal cortical [mCCD(cl.1)] and inner medullary [mIMCD3] collecting duct cell lines. Cells were exposed to equipotent Cd2+ (0.5-5 µmol/l) and/or Fe2+ (50-100 µmol/l) for 4-24 h. Hepcidin (Hamp1) was transiently silenced by RNAi or overexpressed by plasmid transfection. Hepcidin or catalase expression were evaluated by RT-PCR, qPCR, immunoblotting or immunofluorescence microscopy, and cell fate by MTT, apoptosis and necrosis assays. Reactive oxygen species (ROS) were detected using CellROX™ Green and catalase activity by fluorometry. Hepcidin upregulation protected against Fe2+-induced mIMCD3 cell death by increasing catalase activity and reducing ROS, but exacerbated Cd2+-induced catalase dysfunction, increasing ROS and cell death. Opposite effects were observed with Hamp1 siRNA. Similar to Hamp1 silencing, increased intracellular Fe2+ prevented Cd2+ damage, ROS formation and catalase disruption whereas chelation of intracellular Fe2+ with desferrioxamine augmented Cd2+ damage, corresponding to hepcidin upregulation. Comparable effects were observed in mCCD(cl.1) cells, indicating equivalent functions of renal hepcidin in different collecting duct segments. In conclusion, hepcidin likely binds Fe2+, but not Cd2+. Because Fe2+ and Cd2+ compete for functional binding sites in proteins, hepcidin affects their free metal ion pools and differentially impacts downstream processes and cell fate.


Subject(s)
Cadmium/toxicity , Hepcidins/genetics , Iron/toxicity , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Binding Sites , Binding, Competitive , Cadmium/administration & dosage , Cell Death/drug effects , Cell Line , Cells, Cultured , Deferoxamine/pharmacology , Female , Gene Silencing , Iron/administration & dosage , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
10.
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
11.
Int J Mol Sci ; 21(8)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326436

ABSTRACT

Calcium homeostasis is a cellular process required for proper cell function and survival, maintained by the coordinated action of several transporters, among them members of the Na+/Ca2+-exchanger family, such as SLC8A3. Transforming growth factor beta (TGF-ß) signaling defines neuronal development and survival and may regulate the expression of channels and transporters. We investigated the regulation of SLC8A3 by TGF-ß in a conditional knockout mouse with deletion of TGF-ß signaling from Engrailed 1-expressing cells, i.e., in cells from the midbrain and rhombomere 1, and elucidated the underlying molecular mechanisms. The results show that SLC8A3 is significantly downregulated in developing dopaminergic and dorsal raphe serotonergic neurons in mutants and that low SLC8A3 abundance prevents the expression of the anti-apoptotic protein Bcl-xL. TGF-ß signaling affects SLC8A3 via the canonical and p38 signaling pathway and may increase the binding of Smad4 to the Slc8a3 promoter. Expression of the lipid peroxidation marker malondialdehyde (MDA) was increased following knockdown of Slc8a3 expression in vitro. In neurons lacking TGF-ß signaling, the number of MDA- and 4-hydroxynonenal (4-HNE)-positive cells was significantly increased, accompanied with increased cellular 4-HNE abundance. These results suggest that TGF-ß contributes to the regulation of SLC8A3 expression in developing dopaminergic and dorsal raphe serotonergic neurons, thereby preventing oxidative stress.


Subject(s)
Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Neurogenesis/genetics , Oxidative Stress/genetics , Serotonergic Neurons/metabolism , Sodium-Calcium Exchanger/metabolism , Transforming Growth Factor beta/metabolism , Aldehydes/metabolism , Animals , Apoptosis/genetics , Calcium/metabolism , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , Dopaminergic Neurons/drug effects , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeostasis , Humans , Immunohistochemistry , Malondialdehyde/metabolism , Mesencephalon/drug effects , Mesencephalon/growth & development , Mice , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Serotonergic Neurons/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Smad4 Protein/metabolism , Sodium-Calcium Exchanger/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , bcl-X Protein/metabolism
12.
J Mol Neurosci ; 70(5): 769-777, 2020 May.
Article in English | MEDLINE | ID: mdl-32043204

ABSTRACT

So far no evidence is available as to whether TGFß and Wnt signaling pathways cooperatively modulate dopaminergic differentiation of the adult stem cells. To investigate the interaction between the two pathways in early dopaminergic differentiation, we cultured the newly introduced unrestricted somatic stem cells (USSCs) in neuron differentiation media followed by treatments with inducers and inhibitors of Wnt and TGF beta pathways either alone or in combinations. Our results showed that the level of Nurr-1 as a marker for dopaminergic neuron precursors and that of the nuclear ß-catenin as the key effector of the active Wnt pathway were significantly elevated following the treatment with either TGFß or BIO (the Wnt pathway inducer). Conversely, Nurr-1 expression was significantly reduced following the combined treatments with SB431542 (the TGFß inhibitor) plus BIO or with TGFß plus Dkk1 (the specific Wnt inhibitor). Nuclear ß-catenin was also significantly reduced following combined treatments with SB431542 plus either BIO or TGFß. Altogether, our results imply that Wnt and TGFß signaling pathways cooperatively ensure the early dopaminergic differentiation of the USSC adult stem cells.


Subject(s)
Dopaminergic Neurons/metabolism , Mesenchymal Stem Cells/metabolism , Neurogenesis , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway , Benzamides/pharmacology , Cells, Cultured , Dioxoles/pharmacology , Dopaminergic Neurons/cytology , Fetal Blood/cytology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
13.
Front Cell Neurosci ; 13: 427, 2019.
Article in English | MEDLINE | ID: mdl-31619968

ABSTRACT

Transforming growth factor betas are integral molecular components of the signalling cascades defining development and survival of several neuronal groups. Among TGF-ß ligands, TGF-ß2 has been considered as relatively more important during development. We have generated a conditional knockout mouse of the Tgf-ß2 gene with knock-in of an EGFP reporter and subsequently a mouse line with cell-type specific deletion of TGF-ß2 ligand from Krox20 expressing cells (i.e., in cells from rhombomeres r3 and r5). We performed a phenotypic analysis of the hindbrain serotonergic system during development and in adulthood, determined the neurochemical profile in hindbrain and forebrain, and assessed behavioural performance of wild type and mutant mice. Mutant mice revealed significantly decreased number of caudal 5-HT neurons at embryonic day (E) 14, and impaired development of caudal dorsal raphe, median raphe, raphe magnus, and raphe obscurus neurons at E18, a phenotype that was largely restored and even overshot in dorsal raphe of mutant adult mice. Serotonin levels were decreased in hindbrain but significantly increased in cortex of adult mutant mice, though without any behavioural consequences. These results highlight differential and temporal dependency of developing and adult neurons on TGF-ß2. The results also indicate TGF-ß2 being directly or indirectly potent to modulate neurotransmitter synthesis and metabolism. The novel floxed TGF-ß2 mouse model is a suitable tool for analysing the in vivo functions of TGF-ß2 during development and in adulthood in many organs.

14.
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
15.
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.

16.
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
17.
Open Biol ; 8(3)2018 03.
Article in English | MEDLINE | ID: mdl-29593116

ABSTRACT

The optic fissure is a transient gap in the developing vertebrate eye, which must be closed as development proceeds. A persisting optic fissure, coloboma, is a major cause for blindness in children. Although many genes have been linked to coloboma, the process of optic fissure fusion is still little appreciated, especially on a molecular level. We identified a coloboma in mice with a targeted inactivation of transforming growth factor ß2 (TGFß2). Notably, here the optic fissure margins must have touched, however failed to fuse. Transcriptomic analyses indicated an effect on remodelling of the extracellular matrix (ECM) as an underlying mechanism. TGFß signalling is well known for its effect on ECM remodelling, but it is at the same time often inhibited by bone morphogenetic protein (BMP) signalling. Notably, we also identified two BMP antagonists among the downregulated genes. For further functional analyses we made use of zebrafish, in which we found TGFß ligands expressed in the developing eye, and the ligand binding receptor in the optic fissure margins where we also found active TGFß signalling and, notably, also gremlin 2b (grem2b) and follistatin a (fsta), homologues of the regulated BMP antagonists. We hypothesized that TGFß is locally inducing expression of BMP antagonists within the margins to relieve the inhibition from its regulatory capacity regarding ECM remodelling. We tested our hypothesis and found that induced BMP expression is sufficient to inhibit optic fissure fusion, resulting in coloboma. Our findings can likely be applied also to other fusion processes, especially when TGFß signalling or BMP antagonism is involved, as in fusion processes during orofacial development.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Coloboma/genetics , Gene Expression Profiling/methods , Transforming Growth Factor beta2/genetics , Animals , Coloboma/drug therapy , Disease Models, Animal , Extracellular Matrix/metabolism , Follistatin/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Signal Transduction , Zebrafish/metabolism , Zebrafish Proteins/metabolism
18.
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
19.
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
20.
J Endod ; 43(5): 715-722, 2017 May.
Article in English | MEDLINE | ID: mdl-28325578

ABSTRACT

INTRODUCTION: The aim of this study was to elucidate whether the use of mineral trioxide aggregate (MTA) in endodontic therapy in human teeth leads to the same regeneration of the apical tissues as observed in animals. METHODS: Four human teeth were identified in a policlinic that had been treated endodontically with MTA and had to be extracted for other reasons than just endodontic failure. All teeth were processed for histologic and one for immunohistochemical analyses to analyze the histologic response of the periapical structure to the former treatment with MTA. RESULTS: All identified teeth showed clinical and radiographic signs of healing at the time of extraction. In the histologic evaluation, all teeth showed a layer of cementlike tissues at least on the MTA surface. Further double immunofluorescence analyses for collagen type I and type III revealed protein expression and colocalization of the 2 proteins, implicating formation of periodontal ligamentlike tissue, presumably fibers. CONCLUSIONS: Histologic healing of the human periodontium to MTA corresponds to the healing pattern shown in animal studies. Cementlike tissues were formed on the surface of MTA, which proves regeneration of the periodontal ligament.


Subject(s)
Aluminum Compounds/therapeutic use , Calcium Compounds/therapeutic use , Oxides/therapeutic use , Periodontium/physiology , Root Canal Filling Materials/therapeutic use , Silicates/therapeutic use , Adult , Aged, 80 and over , Drug Combinations , Fluorescent Antibody Technique , Humans , Middle Aged , Periodontal Ligament/drug effects , Periodontal Ligament/physiology , Periodontium/drug effects , Periodontium/pathology , Regeneration/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...