Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
J Biol Chem ; 297(6): 101355, 2021 12.
Article in English | MEDLINE | ID: mdl-34717959

ABSTRACT

The ion pump Na+,K+-ATPase is a critical determinant of neuronal excitability; however, its role in the etiology of diseases of the central nervous system (CNS) is largely unknown. We describe here the molecular phenotype of a Trp931Arg mutation of the Na+,K+-ATPase catalytic α1 subunit in an infant diagnosed with therapy-resistant lethal epilepsy. In addition to the pathological CNS phenotype, we also detected renal wasting of Mg2+. We found that membrane expression of the mutant α1 protein was low, and ion pumping activity was lost. Arginine insertion into membrane proteins can generate water-filled pores in the plasma membrane, and our molecular dynamic (MD) simulations of the principle states of Na+,K+-ATPase transport demonstrated massive water inflow into mutant α1 and destabilization of the ion-binding sites. MD simulations also indicated that a water pathway was created between the mutant arginine residue and the cytoplasm, and analysis of oocytes expressing mutant α1 detected a nonspecific cation current. Finally, neurons expressing mutant α1 were observed to be depolarized compared with neurons expressing wild-type protein, compatible with a lowered threshold for epileptic seizures. The results imply that Na+,K+-ATPase should be considered a neuronal locus minoris resistentia in diseases associated with epilepsy and with loss of plasma membrane integrity.


Subject(s)
Epilepsy/genetics , Mutation, Missense , Sodium-Potassium-Exchanging ATPase/genetics , Animals , Anticonvulsants/pharmacology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cells, Cultured , Drug Resistance , Epilepsy/drug therapy , Epilepsy/pathology , Humans , Infant , Molecular Dynamics Simulation , Mutation, Missense/drug effects , Protein Subunits/analysis , Protein Subunits/genetics , Sodium-Potassium-Exchanging ATPase/analysis , Xenopus
2.
FASEB J ; 33(3): 4089-4096, 2019 03.
Article in English | MEDLINE | ID: mdl-30496703

ABSTRACT

The central role of calcium signaling during development of early vertebrates is well documented, but little is known about its role in mammalian embryogenesis. We have used immunofluorescence and time-lapse calcium imaging of cultured explanted embryonic rat kidneys to study the role of calcium signaling for branching morphogenesis. In mesenchymal cells, we recorded spontaneous calcium activity that was characterized by irregular calcium transients. The calcium signals were dependent on release of calcium from intracellular stores in the endoplasmic reticulum. Down-regulation of the calcium activity, both by blocking the sarco-endoplasmic reticulum Ca2+-ATPase and by chelating cytosolic calcium, resulted in retardation of branching morphogenesis and a reduced formation of primitive nephrons but had no effect on cell proliferation. We propose that spontaneous calcium activity contributes with a stochastic factor to the self-organizing process that controls branching morphogenesis, a major determinant of the ultimate number of nephrons in the kidney.-Fontana, J. M., Khodus, G. R., Unnersjö-Jess, D., Blom, H., Aperia, A., Brismar, H. Spontaneous calcium activity in metanephric mesenchymal cells regulates branching morphogenesis in the embryonic kidney.


Subject(s)
Calcium Signaling , Embryonic Stem Cells/metabolism , Kidney/metabolism , Morphogenesis , Animals , Endoplasmic Reticulum/metabolism , Kidney/cytology , Kidney/embryology , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
3.
FASEB J ; 33(9): 10193-10206, 2019 09.
Article in English | MEDLINE | ID: mdl-31199885

ABSTRACT

The ion pump Na+, K+-ATPase (NKA) is a receptor for the cardiotonic steroid ouabain. Subsaturating concentration of ouabain triggers intracellular calcium oscillations, stimulates cell proliferation and adhesion, and protects from apoptosis. However, it is controversial whether ouabain-bound NKA is considered a signal transducer. To address this question, we performed a global analysis of protein phosphorylation in COS-7 cells, identifying 2580 regulated phosphorylation events on 1242 proteins upon 10- and 20-min treatment with ouabain. Regulated phosphorylated proteins include the inositol triphosphate receptor and stromal interaction molecule, which are essential for initiating calcium oscillations. Hierarchical clustering revealed that ouabain triggers a structured phosphorylation response that occurs in a well-defined, time-dependent manner and affects specific cellular processes, including cell proliferation and cell-cell junctions. We additionally identify regulation of the phosphorylation of several calcium and calmodulin-dependent protein kinases (CAMKs), including 2 sites of CAMK type II-γ (CAMK2G), a protein known to regulate apoptosis. To verify the significance of this result, CAMK2G was knocked down in primary kidney cells. CAMK2G knockdown impaired ouabain-dependent protection from apoptosis upon treatment with high glucose or serum deprivation. In conclusion, we establish NKA as the coordinator of a broad, tightly regulated phosphorylation response in cells and define CAMK2G as a downstream effector of NKA.-Panizza, E., Zhang, L., Fontana, J. M., Hamada, K., Svensson, D., Akkuratov, E. E., Scott, L., Mikoshiba, K., Brismar, H., Lehtiö, J., Aperia, A. Ouabain-regulated phosphoproteome reveals molecular mechanisms for Na+, K+-ATPase control of cell adhesion, proliferation, and survival.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Ouabain/pharmacology , Protein Kinases/metabolism , Protein Processing, Post-Translational/drug effects , Sodium-Potassium-Exchanging ATPase/physiology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis/physiology , COS Cells , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Survival/drug effects , Cell Survival/physiology , Chlorocebus aethiops , Down-Regulation/drug effects , Glucose/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/enzymology , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/genetics , Models, Molecular , Phosphorylation , Protein Conformation , Protein Kinases/drug effects , Proteome , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats , Sodium-Potassium-Exchanging ATPase/drug effects
4.
Am J Physiol Renal Physiol ; 316(5): F1078-F1089, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30864838

ABSTRACT

It is generally believed that cells that are unable to downregulate glucose transport are particularly vulnerable to hyperglycemia. Yet, little is known about the relation between expression of glucose transporters and acute toxic effects of high glucose exposure. In the present ex vivo study of rat renal cells, we compared the apoptotic response to a moderate increase in glucose concentration. We studied cell types that commonly are targeted in diabetic kidney disease (DKD): proximal tubule cells, which express Na+-dependent glucose transporter (SGLT)2, mesangial cells, which express SGLT1, and podocytes, which lack SGLT and take up glucose via insulin-dependent glucose transporter 4. Proximal tubule cells and mesangial cells responded within 4-8 h of exposure to 15 mM glucose with translocation of the apoptotic protein Bax to mitochondria and an increased apoptotic index. SGLT downregulation and exposure to SGLT inhibitors abolished the apoptotic response. The onset of overt DKD generally coincides with the onset of albuminuria. Albumin had an additive effect on the apoptotic response. Ouabain, which interferes with the apoptotic onset, rescued from the apoptotic response. Insulin-supplemented podocytes remained resistant to 15 and 30 mM glucose for at least 24 h. Our study points to a previously unappreciated role of SGLT-dependent glucose uptake as a risk factor for diabetic complications and highlights the importance of therapeutic approaches that specifically target the different cell types in DKD.


Subject(s)
Apoptosis/drug effects , Diabetic Nephropathies/metabolism , Epithelial Cells/drug effects , Glucose/toxicity , Kidney Tubules, Proximal/drug effects , Mesangial Cells/drug effects , Podocytes/drug effects , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2/metabolism , Animals , Cells, Cultured , Diabetic Nephropathies/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Insulin/pharmacology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mesangial Cells/metabolism , Mesangial Cells/pathology , Ouabain/pharmacology , Podocytes/metabolism , Podocytes/pathology , Rats, Sprague-Dawley , Signal Transduction , Time Factors
5.
EMBO J ; 34(19): 2408-23, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26323479

ABSTRACT

Extracellular α-synuclein (α-syn) assemblies can be up-taken by neurons; however, their interaction with the plasma membrane and proteins has not been studied specifically. Here we demonstrate that α-syn assemblies form clusters within the plasma membrane of neurons. Using a proteomic-based approach, we identify the α3-subunit of Na+/K+-ATPase (NKA) as a cell surface partner of α-syn assemblies. The interaction strength depended on the state of α-syn, fibrils being the strongest, oligomers weak, and monomers none. Mutations within the neuron-specific α3-subunit are linked to rapid-onset dystonia Parkinsonism (RDP) and alternating hemiplegia of childhood (AHC). We show that freely diffusing α3-NKA are trapped within α-syn clusters resulting in α3-NKA redistribution and formation of larger nanoclusters. This creates regions within the plasma membrane with reduced local densities of α3-NKA, thereby decreasing the efficiency of Na+ extrusion following stimulus. Thus, interactions of α3-NKA with extracellular α-syn assemblies reduce its pumping activity as its mutations in RDP/AHC.


Subject(s)
Hemiplegia/metabolism , Mutation , Neurons/metabolism , Parkinsonian Disorders/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , alpha-Synuclein/metabolism , Hemiplegia/genetics , Hemiplegia/pathology , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurons/pathology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Sodium-Potassium-Exchanging ATPase/genetics , alpha-Synuclein/genetics
6.
Int J Mol Sci ; 19(8)2018 Jul 29.
Article in English | MEDLINE | ID: mdl-30060621

ABSTRACT

Neuronal activity leads to an influx of Na⁺ that needs to be rapidly cleared. The sodium-potassium ATPase (Na,K-ATPase) exports three Na⁺ ions and imports two K⁺ ions at the expense of one ATP molecule. Na,K-ATPase turnover accounts for the majority of energy used by the brain. To prevent an energy crisis, the energy expense for Na⁺ clearance must provide an optimal effect. Here we report that in rat primary hippocampal neurons, the clearance of Na⁺ ions is more efficient if Na,K-ATPase is laterally mobile in the membrane than if it is clustered. Using fluorescence recovery after photobleaching and single particle tracking analysis, we show that the ubiquitous α1 and the neuron-specific α3 catalytic subunits as well as the supportive ß1 subunit of Na,K-ATPase are highly mobile in the plasma membrane. We show that cross-linking of the ß1 subunit with polyclonal antibodies or exposure to Modulator of Na,K-ATPase (MONaKA), a secreted protein which binds to the extracellular domain of the ß subunit, clusters the α3 subunit in the membrane and restricts its mobility. We demonstrate that clustering, caused by cross-linking or by exposure to MONaKA, reduces the efficiency in restoring intracellular Na⁺. These results demonstrate that extracellular interactions with Na,K-ATPase regulate the Na⁺ extrusion efficiency with consequences for neuronal energy balance.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Neurons/cytology , Protein Subunits/metabolism , Protein Transport , Rats, Sprague-Dawley
7.
BMC Cardiovasc Disord ; 17(1): 126, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28514967

ABSTRACT

BACKGROUND: Blockers of angiotensin II type 1 receptor (AT1R) and the voltage gated calcium channel 1.2 (CaV1.2) are commonly used for treatment of hypertension. Yet there is little information about the effect of physiological concentrations of angiotensin II (AngII) on AT1R signaling and whether there is a reciprocal regulation of AT1R signaling by CaV1.2. METHODS: To elucidate these questions, we have studied the Ca2+ signaling response to physiological and pharmacological AngII doses in HEK293a cells, vascular smooth muscle cells and cardiomyocytes using a Ca2+ sensitive dye as the principal sensor. Intra-cellular calcium recordings were performed in presence and absence of CaV1.2 blockers. Semi-quantitative imaging methods were used to assess the plasma membrane expression of AT1R and G-protein activation. RESULTS: Repeated exposure to pharmacological (100 nM) concentrations of AngII caused, as expected, a down-regulation of the Ca2+ response. In contrast, repeated exposure to physiological (1 nM) AngII concentration resulted in an enhancement of the Ca2+ response. The up-regulation of the Ca2+ response to repeated 1 nM AngII doses and the down-regulation of the Ca2+ response to repeated 100 nM Angll doses were not accompanied by a parallel change of the AT1R plasma membrane expression. The Ca2+ response to 1 nM of AngII was amplified in the presence of therapeutic concentrations of the CaV1.2 blockers, nifedipine and verapamil, in vascular smooth muscle cells, cardiomyocytes and HEK293a cells. Amplification of the AT1R response was also observed following inhibition of the calcium permeable transient receptor potential cation channels, suggesting that the activity of AT1R is sensitive to calcium influx. CONCLUSIONS: Our findings have implications for the understanding of hyperactivity of the angiotensin system and for use of Ca2+ channel blockers as mono-therapy in hypertension.


Subject(s)
Angiotensin II/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Smooth Muscle/drug effects , Nifedipine/pharmacology , Receptor, Angiotensin, Type 1/agonists , Verapamil/pharmacology , Animals , Animals, Newborn , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/metabolism , Time Factors , Transfection
8.
Am J Physiol Cell Physiol ; 310(7): C491-5, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26791490

ABSTRACT

The Na(+)-K(+)-ATPase (NKA) differs from most other ion transporters, not only in its capacity to maintain a steep electrochemical gradient across the plasma membrane, but also as a receptor for a family of cardiotonic steroids, to which ouabain belongs. Studies from many groups, performed during the last 15 years, have demonstrated that ouabain, a member of the cardiotonic steroid family, can activate a network of signaling molecules, and that NKA will also serve as a signal transducer that can provide a feedback loop between NKA and the mitochondria. This brief review summarizes the current knowledge and controversies with regard to the understanding of NKA signaling.


Subject(s)
Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Humans
9.
Kidney Int ; 90(1): 135-48, 2016 07.
Article in English | MEDLINE | ID: mdl-27217195

ABSTRACT

There is a great need for treatment that arrests progression of chronic kidney disease. Increased albumin in urine leads to apoptosis and fibrosis of podocytes and tubular cells and is a major cause of functional deterioration. There have been many attempts to target fibrosis, but because of the lack of appropriate agents, few have targeted apoptosis. Our group has described an ouabain-activated Na,K-ATPase/IP3R signalosome, which protects from apoptosis. Here we show that albumin uptake in primary rat renal epithelial cells is accompanied by a time- and dose-dependent mitochondrial accumulation of the apoptotic factor Bax, down-regulation of the antiapoptotic factor Bcl-xL and mitochondrial membrane depolarization. Ouabain opposes these effects and protects from apoptosis in albumin-exposed proximal tubule cells and podocytes. The efficacy of ouabain as an antiapoptotic and kidney-protective therapeutic tool was then tested in rats with passive Heymann nephritis, a model of proteinuric chronic kidney disease. Chronic ouabain treatment preserved renal function, protected from renal cortical apoptosis, up-regulated Bax, down-regulated Bcl-xL, and rescued from glomerular tubular disconnection and podocyte loss. Thus we have identified a novel clinically feasible therapeutic tool, which has the potential to protect from apoptosis and rescue from loss of functional tissue in chronic proteinuric kidney disease.


Subject(s)
Apoptosis/drug effects , Cardiotonic Agents/therapeutic use , Glomerulonephritis, Membranous/drug therapy , Kidney Glomerulus/drug effects , Kidney Tubules, Proximal/drug effects , Ouabain/therapeutic use , Proteinuria/drug therapy , Animals , Down-Regulation , Drug Evaluation, Preclinical , Humans , Kidney Diseases/physiopathology , Male , Podocytes/physiology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase , Up-Regulation , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
10.
J Neurochem ; 132(6): 677-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25639954

ABSTRACT

Metabotropic glutamate receptor 5 (mGluR5) regulates excitatory post-synaptic signaling in the central nervous system (CNS) and is implicated in various CNS disorders. Protein kinase A (PKA) signaling is known to play a critical role in neuropsychiatric disorders such as Parkinson's disease, schizophrenia, and addiction. Dopamine signaling is known to modulate the properties of mGluR5 in a cAMP- and PKA-dependent manner, suggesting that mGluR5 may be a direct target for PKA. Our study identifies mGluR5 at Ser870 as a direct substrate for PKA phosphorylation and demonstrates that this phosphorylation plays a critical role in the PKA-mediated modulation of mGluR5 functions such as extracellular signal-regulated kinase phosphorylation and intracellular Ca(2+) oscillations. The identification of the molecular mechanism by which PKA signaling modulates mGluR5-mediated cellular responses contributes to the understanding of the interaction between dopaminergic and glutamatergic neuronal signaling. We identified serine residue 870 (S870) in metabotropic glutamate receptor 5 (mGluR5) as a direct substrate for protein kinase A (PKA). The phosphorylation of this site regulates the ability of mGluR5 to induce extracellular signal-regulated kinase (ERK) phosphorylation and intracellular Ca(2+) oscillations. This study provides a direct molecular mechanism by which PKA signaling interacts with glutamate neurotransmission.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Receptor, Metabotropic Glutamate 5/physiology , Amino Acid Sequence , Animals , Binding Sites/physiology , Cyclic AMP-Dependent Protein Kinases/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Phosphorylation/physiology
11.
Trends Biochem Sci ; 35(12): 653-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20542436

ABSTRACT

Na(+),K(+)-ATPase (NKA) has a fundamental role in ion transport across the plasma membrane of animal cells and uses approximately 50% of brain energy consumption. Recent work has uncovered additional roles for NKA in signal transduction. How might such different functions of the sodium-potassium pump be connected and regulated? We envision an integrated model of ion pumping and signaling, considering in particular the recently discovered regulation of the sodium-potassium pump by agrin, a protein that is cleaved specifically by neurotrypsin at the synapse. Based on the recently solved structure of NKA and sequence analysis, we propose a molecular model for the agrin-NKA interaction, in which agrin displaces the NKA ß-subunit and exploits the ouabain-binding pocket.


Subject(s)
Agrin/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/metabolism , Agrin/chemistry , Animals , Models, Molecular , Sodium-Potassium-Exchanging ATPase/chemistry
12.
J Biol Chem ; 288(4): 2734-43, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23195960

ABSTRACT

Most neurons co-express two catalytic isoforms of Na,K-ATPase, the ubiquitous α1, and the more selectively expressed α3. Although neurological syndromes are associated with α3 mutations, the specific role of this isoform is not completely understood. Here, we used electrophysiological and Na(+) imaging techniques to study the role of α3 in central nervous system neurons expressing both isoforms. Under basal conditions, selective inhibition of α3 using a low concentration of the cardiac glycoside, ouabain, resulted in a modest increase in intracellular Na(+) concentration ([Na(+)](i)) accompanied by membrane potential depolarization. When neurons were challenged with a large rapid increase in [Na(+)](i), similar to what could be expected following suprathreshold neuronal activity, selective inhibition of α3 almost completely abolished the capacity to restore [Na(+)](i) in soma and dendrite. Recordings of Na,K-ATPase specific current supported the notion that when [Na(+)](i) is elevated in the neuron, α3 is the predominant isoform responsible for rapid extrusion of Na(+). Low concentrations of ouabain were also found to disrupt cortical network oscillations, providing further support for the importance of α3 function in the central nervous system. The α isoforms express a well conserved protein kinase A consensus site, which is structurally associated with an Na(+) binding site. Following activation of protein kinase A, both the α3-dependent current and restoration of dendritic [Na(+)](i) were significantly attenuated, indicating that α3 is a target for phosphorylation and may participate in short term regulation of neuronal function.


Subject(s)
Neurons/metabolism , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Calibration , Catalysis , Cyclic AMP-Dependent Protein Kinases/metabolism , Electrophysiology/methods , Hippocampus/metabolism , Membrane Potentials , Models, Biological , Ouabain/pharmacology , Protein Isoforms , Rats , Rats, Sprague-Dawley , Sodium/chemistry , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry
13.
BMC Neurosci ; 15: 45, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24670218

ABSTRACT

BACKGROUND: Norbin is a neuron-specific, cytosolic protein that interacts with the metabotropic glutamate receptor 5 (mGluR5) and has a profound impact on mGluR5 signaling. Yet, little is known about its synaptic distribution. RESULTS: Here we have analyzed the spatial relationship between Norbin, postsynaptic density protein 95 (PSD-95), actin and mGluR5 in spines using super-resolution microscopy. Norbin was found to have a high degree of colocalization with actin and a lower degree of colocalization with PSD-95. Co-immunoprecipitation studies confirmed that interaction occurs between Norbin and actin, but not between Norbin and PSD-95. Norbin was also found to have a high degree of colocalization with the perisynaptically located mGluR5. Findings based on structured illumination microscopy (3D-SIM) of exogenous expressed Norbin-GFP were confirmed by stimulated emission depletion microscopy (STED) of immunolabeled endogenous Norbin. CONCLUSIONS: Norbin associates with actin rather than with PSD-95 in dendritic spines. Results regarding protein localization and colocalization performed with conventional confocal microscopy must be interpreted with great caution. The now available super-resolution microscopy techniques provide more accurate information about sub-cellular protein localization than previously was possible.


Subject(s)
Actins/metabolism , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Neuropeptides/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Subcellular Fractions/metabolism , Animals , Cells, Cultured , Female , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
14.
J Am Soc Nephrol ; 24(9): 1413-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23744887

ABSTRACT

Hemolytic uremic syndrome, a life-threatening disease often accompanied by acute renal failure, usually occurs after gastrointestinal infection with Shiga toxin 2 (Stx2)-producing Escherichia coli. Stx2 binds to the glycosphingolipid globotriaosylceramide receptor, expressed by renal epithelial cells, and triggers apoptosis by activating the apoptotic factor Bax. Signaling via the ouabain/Na,K-ATPase/IP3R/NF-κB pathway increases expression of Bcl-xL, an inhibitor of Bax, suggesting that ouabain might protect renal cells from Stx2-triggered apoptosis. Here, exposing rat proximal tubular cells to Stx2 in vitro resulted in massive apoptosis, upregulation of the apoptotic factor Bax, increased cleaved caspase-3, and downregulation of the survival factor Bcl-xL; co-incubation with ouabain prevented all of these effects. Ouabain activated the NF-κB antiapoptotic subunit p65, and the inhibition of p65 DNA binding abolished the antiapoptotic effect of ouabain in Stx2-exposed tubular cells. Furthermore, in vivo, administration of ouabain reversed the imbalance between Bax and Bcl-xL in Stx2-treated mice. Taken together, these results suggest that ouabain can protect the kidney from the apoptotic effects of Stx2.


Subject(s)
Apoptosis/drug effects , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Ouabain/pharmacology , Shiga Toxin 2/pharmacology , bcl-2-Associated X Protein/physiology , bcl-X Protein/physiology , Animals , Apoptosis/physiology , Caspase 3/physiology , Caspase 8/physiology , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors/physiology , Kidney Tubules, Proximal/drug effects , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/physiology , bcl-2-Associated X Protein/drug effects , bcl-X Protein/drug effects
15.
J Neurosci ; 32(50): 17998-8008, 2012 Dec 12.
Article in English | MEDLINE | ID: mdl-23238716

ABSTRACT

Postsynaptic receptor trafficking plays an essential role in tuning neurotransmission and signal plasticity and has emerged as a potential therapeutic target in neuropsychiatric disease. Using a novel application of fluorescence recovery after photobleaching in rat hippocampal neurons, we examined transport from the soma to dendrites of seven G-protein-coupled receptors (GPCRs) implicated in mood disorders. Most GPCRs were delivered to dendrites via lateral diffusion, but one GPCR, the serotonin 1B receptor (5-HT(1B)), was delivered to the dendrites in secretory vesicles. Within the dendrites, 5-HT(1B) were stored in a reservoir of accessible vesicles that were recruited to preferential sites in plasma membrane, as observed with superecliptic pHluorin labeling. After membrane recruitment, 5-HT(1B) transport via lateral diffusion and temporal confinement to inhibitory and excitatory synapses was monitored by single particle tracking. These results suggest an alternative mechanism for control of neuronal activity via a GPCR that has been implicated in mood regulation.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Serotonin/metabolism , Synaptic Membranes/metabolism , Synaptic Transmission/physiology , Animals , Immunohistochemistry , Immunoprecipitation , Microscopy, Confocal , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
16.
BMC Nephrol ; 14: 34, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23398808

ABSTRACT

BACKGROUND: Polycystin-2 (PC2), encoded by the gene that is mutated in autosomal dominant polycystic kidney disease (ADPKD), functions as a calcium (Ca(2+)) permeable ion channel. Considerable controversy remains regarding the subcellular localization and signaling function of PC2 in kidney cells. METHODS: We investigated the subcellular PC2 localization by immunocytochemistry and confocal microscopy in primary cultures of human and rat proximal tubule cells after stimulating cytosolic Ca(2+) signaling. Plasma membrane (PM) Ca(2+) permeability was evaluated by Fura-2 manganese quenching using time-lapse fluorescence microscopy. RESULTS: We demonstrated that PC2 exhibits a dynamic subcellular localization pattern. In unstimulated human or rat proximal tubule cells, PC2 exhibited a cytosolic/reticular distribution. Treatments with agents that in various ways affect the Ca(2+) signaling machinery, those being ATP, bradykinin, ionomycin, CPA or thapsigargin, resulted in increased PC2 immunostaining in the PM. Exposing cells to the steroid hormone ouabain, known to trigger Ca(2+) oscillations in kidney cells, caused increased PC2 in the PM and increased PM Ca(2+) permeability. Intracellular Ca(2+) buffering with BAPTA, inositol 1,4,5-trisphosphate receptor (InsP3R) inhibition with 2-aminoethoxydiphenyl borate (2-APB) or Ca(2+)/Calmodulin-dependent kinase inhibition with KN-93 completely abolished ouabain-stimulated PC2 translocation to the PM. CONCLUSIONS: These novel findings demonstrate intracellular Ca(2+)-dependent PC2 trafficking in human and rat kidney cells, which may provide new insight into cyst formations in ADPKD.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Cell Membrane/metabolism , Kidney/cytology , Kidney/metabolism , Animals , Cells, Cultured , Humans , Protein Transport/physiology , Rats
17.
J Am Soc Nephrol ; 23(8): 1283-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22745476

ABSTRACT

The ability of cells to maintain sharp ion gradients across their membranes is the foundation for the molecular transport and electrical excitability. Across animal species and cell types, Na(+),K(+)-adenosine triphosphatase (ATPase) is arguably the most powerful contributor to this phenomenon. By producing a steep concentration difference of sodium and potassium between the intracellular and extracellular milieu, Na(+),K(+)-ATPase in the tubules provides the driving force for renal sodium reabsorption. Pump activity is downregulated by natriuretic hormones, such as dopamine, and is upregulated by antinatriuretic hormones, such as angiotensin. In the past decade, studies have revealed a novel and surprising role: that Na(+),K(+)-ATPase is a transducer of signals from extracellular to intracellular compartments. The signaling function of Na(+),K(+)-ATPase is activated by ouabain, a mammalian steroid hormone, at far lower concentrations than those that inhibit pump activity. By promoting growth and inhibiting apoptosis, activation of Na(+),K(+)-ATPase exerts tissue-protective effects. Ouabain-stimulated Na(+),K(+)-ATPase signaling has recently shown clinical promise by protecting the malnourished embryonic kidney from adverse developmental programming. A deeper understanding of the tissue-protective role of Na(+),K(+)-ATPase signaling and the regulation of Na(+),K(+)-ATPase pumping activity is of fundamental importance for the understanding and treatment of kidney diseases and kidney-related hypertension.


Subject(s)
Kidney/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Apoptosis , Calcium Signaling , Cell Proliferation , Homeostasis , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Kidney/growth & development , Kidney Diseases/enzymology , Sodium/metabolism
18.
J Am Soc Nephrol ; 23(3): 421-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22193384

ABSTRACT

Signaling through both angiotensin AT1 receptors (AT1R) and dopamine D1 receptors (D1R) modulates renal sodium excretion and arterial BP. AT1R and D1R form heterodimers, but whether treatment with AT1R antagonists functionally modifies D1R via allosterism is unknown. In this study, the AT1R antagonist losartan strengthened the interaction between AT1R and D1R and increased expression of D1R on the plasma membrane in vitro. In rat proximal tubule cells that express endogenous AT1R and D1R, losartan increased cAMP generation. Losartan increased cAMP in HEK 293a cells transfected with both AT1R and D1R, but it did not increase cAMP in cells transfected with either receptor alone, suggesting that losartan induces D1R activation. Furthermore, losartan did not increase cAMP in HEK 293a cells expressing AT1R and mutant S397/S398A D1R, which disrupts the physical interaction between AT1R and D1R. In vivo, administration of a D1R antagonist significantly attenuated the antihypertensive effect of losartan in rats with renal hypertension. Taken together, these data imply that losartan might exert its antihypertensive effect both by inhibiting AT1R signaling and by enhancing D1R signaling.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/metabolism , Kidney Tubules, Proximal/metabolism , Kidney/metabolism , Losartan/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Dopamine D1/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aortic Coarctation/complications , Benzazepines/pharmacology , Benzazepines/therapeutic use , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Hypertension/drug therapy , Hypertension/etiology , In Vitro Techniques , Kidney/cytology , Kidney/drug effects , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Losartan/pharmacology , Losartan/therapeutic use , Male , Protein Binding , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/drug effects , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/drug effects , Signal Transduction/drug effects
19.
Proc Natl Acad Sci U S A ; 106(5): 1602-7, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19164545

ABSTRACT

Disturbed brain water homeostasis with swelling of astroglial cells is a common complication in stroke, trauma, and meningitis and is considered to be a major cause of permanent brain damage. Astroglial cells possess the water channel aquaporin 4 (AQP4). Recent studies from our laboratory have shown that glutamate, acting on group I metabotropic glutamate receptors (mGluRs), increases the permeability of astrocyte AQP4, which, in situations of hypoxia-ischemia, will increase astrocyte water uptake. Here we report that erythropoietin (EPO), which in recent years has emerged as a potent neuro-protective agent, antagonizes the effect of a group I mGluR agonist on astrocyte water permeability. Activation of group I mGluRs triggers fast and highly regular intracellular calcium oscillations and we show that EPO interferes with this signaling event by altering the frequency of the oscillations. These effects of EPO are immediate, in contrast to the neuroprotective effects of EPO that are known to depend upon gene activation. Our findings indicate that EPO may directly reduce the risk of astrocyte swelling in stroke and other brain insults. In support of this conclusion we found that EPO reduced the neurological symptoms in a mouse model of primary brain edema known to depend upon AQP4 water transport.


Subject(s)
Astrocytes/metabolism , Erythropoietin/physiology , Water/metabolism , Animals , Aquaporin 4/metabolism , Brain Edema/physiopathology , Calcium Signaling , Cells, Cultured , Female , Hippocampus/metabolism , In Vitro Techniques , Mice , Mice, Inbred C3H , Permeability , Rats , Receptors, Glutamate/metabolism
20.
BMC Neurosci ; 12: 16, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21272290

ABSTRACT

BACKGROUND: The Na+,K+-ATPase plays an important role for ion homeostasis in virtually all mammalian cells, including neurons. Despite this, there is as yet little known about the isoform specific distribution in neurons. RESULTS: With help of superresolving stimulated emission depletion microscopy the spatial distribution of Na+,K+-ATPase in dendritic spines of cultured striatum neurons have been dissected. The found compartmentalized distribution provides a strong evidence for the confinement of neuronal Na+,K+-ATPase (α3 isoform) in the postsynaptic region of the spine. CONCLUSIONS: A compartmentalized distribution may have implications for the generation of local sodium gradients within the spine and for the structural and functional interaction between the sodium pump and other synaptic proteins. Superresolution microscopy has thus opened up a new perspective to elucidate the nature of the physiological function, regulation and signaling role of Na+,K+-ATPase from its topological distribution in dendritic spines.


Subject(s)
Dendritic Spines/enzymology , Nanotechnology/methods , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dendritic Spines/ultrastructure , Image Interpretation, Computer-Assisted/instrumentation , Image Interpretation, Computer-Assisted/methods , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL