Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
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.
Cell Death Dis ; 12(8): 739, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34315852

ABSTRACT

Activation of the apoptotic pathway is a major cause of progressive loss of function in chronic diseases such as neurodegenerative and diabetic kidney diseases. There is an unmet need for an anti-apoptotic drug that acts in the early stage of the apoptotic process. The multifunctional protein Na+,K+-ATPase has, in addition to its role as a transporter, a signaling function that is activated by its ligand, the cardiotonic steroid ouabain. Several lines of evidence suggest that sub-saturating concentrations of ouabain protect against apoptosis of renal epithelial cells, a common complication and major cause of death in diabetic patients. Here, we induced apoptosis in primary rat renal epithelial cells by exposing them to an elevated glucose concentration (20 mM) and visualized the early steps in the apoptotic process using super-resolution microscopy. Treatment with 10 nM ouabain interfered with the onset of the apoptotic process by inhibiting the activation of the BH3-only protein Bad and its translocation to mitochondria. This occurred before the pro-apoptotic protein Bax had been recruited to mitochondria. Two ouabain regulated and Akt activating Ca2+/calmodulin-dependent kinases were found to play an essential role in the ouabain anti-apoptotic effect. Our results set the stage for further exploration of ouabain as an anti-apoptotic drug in diabetic kidney disease as well as in other chronic diseases associated with excessive apoptosis.


Subject(s)
Apoptosis , Cytoprotection , Glucose/toxicity , Microscopy , Signal Transduction , Sodium-Potassium-Exchanging ATPase/metabolism , bcl-Associated Death Protein/metabolism , Animals , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cytoprotection/drug effects , Cytosol/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Ouabain/pharmacology , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
3.
Cell Death Dis ; 12(4): 292, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33731672

ABSTRACT

Mutations in PRoline Rich Transmembrane protein 2 (PRRT2) cause pleiotropic syndromes including benign infantile epilepsy, paroxysmal kinesigenic dyskinesia, episodic ataxia, that share the paroxysmal character of the clinical manifestations. PRRT2 is a neuronal protein that plays multiple roles in the regulation of neuronal development, excitability, and neurotransmitter release. To better understand the physiopathology of these clinical phenotypes, we investigated PRRT2 interactome in mouse brain by a pulldown-based proteomic approach and identified α1 and α3 Na+/K+ ATPase (NKA) pumps as major PRRT2-binding proteins. We confirmed PRRT2 and NKA interaction by biochemical approaches and showed their colocalization at neuronal plasma membrane. The acute or constitutive inactivation of PRRT2 had a functional impact on NKA. While PRRT2-deficiency did not modify NKA expression and surface exposure, it caused an increased clustering of α3-NKA on the plasma membrane. Electrophysiological recordings showed that PRRT2-deficiency in primary neurons impaired NKA function during neuronal stimulation without affecting pump activity under resting conditions. Both phenotypes were fully normalized by re-expression of PRRT2 in PRRT2-deficient neurons. In addition, the NKA-dependent afterhyperpolarization that follows high-frequency firing was also reduced in PRRT2-silenced neurons. Taken together, these results demonstrate that PRRT2 is a physiological modulator of NKA function and suggest that an impaired NKA activity contributes to the hyperexcitability phenotype caused by PRRT2 deficiency.


Subject(s)
Adenosine Triphosphatases/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Proteomics/methods , Humans , Synaptic Transmission
4.
Mol Neurobiol ; 57(10): 4018-4030, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32651756

ABSTRACT

The N-methyl-D-aspartate (NMDA) receptor plays an essential role in glutamatergic transmission and synaptic plasticity and researchers are seeking for different modulators of NMDA receptor function. One possible mechanism for its regulation could be through adjacent membrane proteins. NMDA receptors coprecipitate with Na,K-ATPase, indicating a potential interaction of these two proteins. Ouabain, a mammalian cardiotonic steroid that specifically binds to Na,K-ATPase and affects its conformation, can protect from some toxic effects of NMDA receptor activation. Here we have examined whether NMDA receptor activity and downstream effects can be modulated by physiological ouabain concentrations. The spatial colocalization between NMDA receptors and the Na,K-ATPase catalytic subunits on dendrites of cultured rat hippocampal neurons was analyzed with super-resolution dSTORM microscopy. The functional interaction was analyzed with calcium imaging of single hippocampal neurons exposed to 10 µM NMDA in presence and absence of ouabain and by determination of the ouabain effect on NMDA receptor-dependent long-term potentiation. We show that NMDA receptors and the Na,K-ATPase catalytic subunits alpha1 and alpha3 exist in same protein complex and that ouabain in nanomolar concentration consistently reduces the calcium response to NMDA. Downregulation of the NMDA response is not associated with internalization of the receptor or with alterations in its state of Src phosphorylation. Ouabain in nanomolar concentration elicits a long-term potentiation response. Our findings suggest that ouabain binding to a fraction of Na,K-ATPase molecules that cluster with the NMDA receptors will, via a conformational effect on the NMDA receptors, cause moderate but consistent reduction of NMDA receptor response at synaptic activation.


Subject(s)
Ouabain/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Calcium/metabolism , Down-Regulation/drug effects , Hippocampus/cytology , Models, Biological , N-Methylaspartate/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/metabolism , src-Family Kinases/metabolism
5.
Cell Calcium ; 88: 102210, 2020 Apr 26.
Article in English | MEDLINE | ID: mdl-32380435

ABSTRACT

Na,K-ATPase is a ubiquitous multifunctional protein that acts both as an ion pump and as a signal transducer. The signaling function is activated by ouabain in non-toxic concentrations. In epithelial cells the ouabain-bound Na,K-ATPase connects with the inositol 1,4,5-trisphosphate receptor via a short linear motif to activate low frequency Ca2+ oscillations. Within a couple of minutes this ouabain mediated signal has resulted in phosphorylation or dephosphorylation of 2580 phospho-sites. Proteins that control cell proliferation and cell adhesion and calmodulin regulated proteins are enriched among the ouabain phosphor-regulated proteins. The inositol 1,4,5-trisphosphate receptor and the stromal interaction molecule, which are both essential for the initiation of Ca2+ oscillations, belong to the ouabain phosphor-regulated proteins. Downstream effects of the ouabain-evoked Ca2+ signal in epithelial cells include interference with the intrinsic mitochondrial apoptotic process and stimulation of embryonic growth processes. The dual function of Na,K-ATPase as an ion pump and a signal transducer is now well established and evaluation of the physiological and pathophysiological consequences of this universal signal emerges as an urgent topic for future studies.

6.
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
7.
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
8.
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
9.
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
10.
Neuron ; 95(1): 33-50, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683268

ABSTRACT

Several neurodegenerative disorders, such as Alzheimer's and Parkinson's disease, are characterized by prominent loss of synapses and neurons associated with the presence of abnormally structured or misfolded protein assemblies. Cell-to-cell transfer of misfolded proteins has been proposed for the intra-cerebral propagation of these diseases. When released, misfolded proteins diffuse in the 3D extracellular space before binding to the plasma membrane of neighboring cells, where they diffuse on a 2D plane. This reduction in diffusion dimension and the cell surface molecular crowding promote deleterious interactions with native membrane proteins, favoring clustering and further aggregation of misfolded protein assemblies. These processes open up new avenues for therapeutics development targeting the initial interactions of deleterious proteins with the plasma membrane or the subsequent pathological signaling.


Subject(s)
Cell Membrane/metabolism , Neurodegenerative Diseases/metabolism , Prions/metabolism , Protein Aggregation, Pathological/metabolism , Protein Folding , Protein Transport , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Extracellular Space/metabolism , Humans , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Parkinson Disease/metabolism , Superoxide Dismutase-1/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism
11.
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
12.
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
13.
Data Brief ; 7: 221-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26958642

ABSTRACT

α-Synuclein (α-syn) is the principal component of Lewy bodies, the pathophysiological hallmark of individuals affected by Parkinson disease (PD). This neuropathologic form of α-syn contributes to PD progression and propagation of α-syn assemblies between neurons. The data we present here support the proteomic analysis used to identify neuronal proteins that specifically interact with extracellularly applied oligomeric or fibrillar α-syn assemblies (conditions 1 and 2, respectively) (doi: 10.15252/embj.201591397[1]). α-syn assemblies and their cellular partner proteins were pulled down from neuronal cell lysed shortly after exposure to exogenous α-syn assemblies and the associated proteins were identified by mass spectrometry using a shotgun proteomic-based approach. We also performed experiments on pure cultures of astrocytes to identify astrocyte-specific proteins interacting with oligomeric or fibrillar α-syn (conditions 3 and 4, respectively). For each condition, proteins interacting selectively with α-syn assemblies were identified by comparison to proteins pulled-down from untreated cells used as controls. The mass spectrometry data, the database search and the peak lists have been deposited to the ProteomeXchange Consortium database via the PRIDE partner repository with the dataset identifiers PRIDE: PXD002256 to PRIDE: PXD002263 and doi: 10.6019/PXD002256 to 10.6019/PXD002263.

14.
Sci Rep ; 6: 22047, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26911348

ABSTRACT

Migraine is a complex brain disorder, and understanding the complexity of this prevalent disease could improve quality of life for millions of people. Familial Hemiplegic Migraine type 2 (FHM2) is a subtype of migraine with aura and co-morbidities like epilepsy/seizures, cognitive impairments and psychiatric manifestations, such as obsessive-compulsive disorder (OCD). FHM2 disease-mutations locate to the ATP1A2 gene encoding the astrocyte-located α2-isoform of the sodium-potassium pump (α2Na(+)/K(+)-ATPase). We show that knock-in mice heterozygous for the FHM2-associated G301R-mutation (α2(+/G301R)) phenocopy several FHM2-relevant disease traits e.g., by mimicking mood depression and OCD. In vitro studies showed impaired glutamate uptake in hippocampal mixed astrocyte-neuron cultures from α2(G301R/G301R) E17 embryonic mice, and moreover, induction of cortical spreading depression (CSD) resulted in reduced recovery in α2(+/G301R) male mice. Moreover, NMDA-type glutamate receptor antagonists or progestin-only treatment reverted specific α2(+/G301R) behavioral phenotypes. Our findings demonstrate that studies of an in vivo relevant FHM2 disease knock-in mouse model provide a link between the female sex hormone cycle and the glutamate system and a link to co-morbid psychiatric manifestations of FHM2.


Subject(s)
Glutamic Acid/metabolism , Migraine with Aura/genetics , Migraine with Aura/metabolism , Mutation , Phenotype , Acoustic Stimulation , Animals , Behavior, Animal , Biological Transport , Cerebrovascular Circulation , Computational Biology/methods , Cortical Spreading Depression/genetics , Disease Models, Animal , Female , Gonadal Steroid Hormones/metabolism , Male , Mice , Mice, Transgenic , Migraine with Aura/diagnosis , Migraine with Aura/drug therapy , Motor Activity , Reaction Time , Sodium-Potassium-Exchanging ATPase/genetics , Stress, Physiological
15.
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
16.
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
17.
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
18.
PLoS One ; 9(6): e98469, 2014.
Article in English | MEDLINE | ID: mdl-24901986

ABSTRACT

Glutamate released during neuronal activity is cleared from the synaptic space via the astrocytic glutamate/Na(+) co-transporters. This transport is driven by the transmembrane Na(+) gradient mediated by Na,K-ATPase. Astrocytes express two isoforms of the catalytic Na,K-ATPase α subunits; the ubiquitously expressed α1 subunit and the α2 subunit that has a more specific expression profile. In the brain α2 is predominantly expressed in astrocytes. The isoforms differ with regard to Na+ affinity, which is lower for α2. The relative roles of the α1 and α2 isoforms in astrocytes are not well understood. Here we present evidence that the presence of the α2 isoform may contribute to a more efficient restoration of glutamate triggered increases in intracellular sodium concentration [Na(+)]i. Studies were performed on primary astrocytes derived from E17 rat striatum expressing Na,K-ATPase α1 and α2 and the glutamate/Na(+) co-transporter GLAST. Selective inhibition of α2 resulted in a modest increase of [Na(+)]i accompanied by a disproportionately large decrease in uptake of aspartate, an indicator of glutamate uptake. To compare the capacity of α1 and α2 to handle increases in [Na(+)]i triggered by glutamate, primary astrocytes overexpressing either α1 or α2 were used. Exposure to glutamate 200 µM caused a significantly larger increase in [Na(+)]i in α1 than in α2 overexpressing cells, and as a consequence restoration of [Na(+)]i, after glutamate exposure was discontinued, took longer time in α1 than in α2 overexpressing cells. Both α1 and α2 interacted with astrocyte glutamate/Na(+) co-transporters via the 1st intracellular loop.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Aspartic Acid/metabolism , Biological Transport , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Gene Expression , Intracellular Space/metabolism , Isoenzymes , Protein Binding , Rats , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics
19.
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
20.
PLoS One ; 8(9): e75155, 2013.
Article in English | MEDLINE | ID: mdl-24058659

ABSTRACT

The phosphoprotein DARPP-32 (dopamine and cyclic adenosine 3´, 5´-monophosphate-regulated phosphoprotein, 32 kDa) is an important component in the molecular regulation of postsynaptic signaling in neostriatum. Despite the importance of this phosphoprotein, there is as yet little known about the nanoscale distribution of DARPP-32. In this study we applied superresolution stimulated emission depletion microscopy (STED) to assess the expression and distribution of DARPP-32 in striatal neurons. Primary culture of striatal neurons were immunofluorescently labeled for DARPP-32 with Alexa-594 and for the dopamine D1 receptor (D1R) with atto-647N. Dual-color STED microscopy revealed discrete localizations of DARPP-32 and D1R in the spine structure, with clustered distributions in both head and neck. Dissected spine structures reveal that the DARPP-32 signal rarely overlapped with the D1R signal. The D1R receptor is positioned in an "aggregated" manner primarily in the spine head and to some extent in the neck, while DARPP-32 forms several neighboring small nanoclusters spanning the whole spine structure. The DARPP-32 clusters have a mean size of 52 +/- 6 nm, which is close to the resolution limit of the microscope and corresponds to the physical size of a few individual phosphoprotein immunocomplexes. Dissection of synaptic proteins using superresolution microscopy gives possibilities to reveal in better detail biologically relevant information, as compared to diffraction-limited microscopy. In this work, the dissected postsynaptic topology of the DARPP-32 phosphoprotein provides strong evidence for a compartmentalized and confined distribution in dendritic spines. The protein topology and the relatively low copy number of phosphoprotein provides a conception of DARPP-32's possibilities to fine-tune the regulation of synaptic signaling, which should have an impact on the performance of the neuronal circuits in which it is expressed.


Subject(s)
Corpus Striatum/metabolism , Dendritic Spines/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Animals , Corpus Striatum/cytology , Female , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/metabolism , Spine/cytology , Spine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...