1
|
Erazo-Toscano R, Fomenko M, Core S, Calabrese RL, Cymbalyuk G. Bursting Dynamics Based on the Persistent Na + and Na +/K + Pump Currents: A Dynamic Clamp Approach. eNeuro 2023; 10:ENEURO.0331-22.2023. [PMID: 37433684 PMCID: PMC10444573 DOI: 10.1523/eneuro.0331-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 06/04/2023] [Accepted: 06/16/2023] [Indexed: 07/13/2023] Open
Abstract
Life-supporting rhythmic motor functions like heart-beating in invertebrates and breathing in vertebrates require an indefatigable generation of a robust rhythm by specialized oscillatory circuits, central pattern generators (CPGs). These CPGs should be sufficiently flexible to adjust to environmental changes and behavioral goals. Continuous self-sustained operation of bursting neurons requires intracellular Na+ concentration to remain in a functional range and to have checks and balances of the Na+ fluxes met on a cycle-to-cycle basis during bursting. We hypothesize that at a high excitability state, the interaction of the Na+/K+ pump current, Ipump, and persistent Na+ current, INaP, produces a mechanism supporting functional bursting. INaP is a low voltage-activated inward current that initiates and supports the bursting phase. This current does not inactivate and is a significant source of Na+ influx. Ipump is an outward current activated by [Na+]i and is the major source of Na+ efflux. Both currents are active and counteract each other between and during bursts. We apply a combination of electrophysiology, computational modeling, and dynamic clamp to investigate the role of Ipump and INaP in the leech heartbeat CPG interneurons (HN neurons). Applying dynamic clamp to introduce additional Ipump and INaP into the dynamics of living synaptically isolated HN neurons in real time, we show that their joint increase produces transition into a new bursting regime characterized by higher spike frequency and larger amplitude of the membrane potential oscillations. Further increase of Ipump speeds up this rhythm by shortening burst duration (BD) and interburst interval (IBI).
Collapse
Affiliation(s)
- Ricardo Erazo-Toscano
- Neuroscience Institute, Georgia State University, Atlanta, 30302 GA
- Department of Biology, Emory University, Atlanta, 30322 GA
| | - Mykhailo Fomenko
- Neuroscience Institute, Georgia State University, Atlanta, 30302 GA
| | - Samuel Core
- Neuroscience Institute, Georgia State University, Atlanta, 30302 GA
| | | | | |
Collapse
|
2
|
Markina AA, Kazanskaya RB, Timoshina JA, Zavialov VA, Abaimov DA, Volnova AB, Fedorova TN, Gainetdinov RR, Lopachev AV. Na +,K +-ATPase and Cardiotonic Steroids in Models of Dopaminergic System Pathologies. Biomedicines 2023; 11:1820. [PMID: 37509460 PMCID: PMC10377002 DOI: 10.3390/biomedicines11071820] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 07/30/2023] Open
Abstract
In recent years, enough evidence has accumulated to assert that cardiotonic steroids, Na+,K+-ATPase ligands, play an integral role in the physiological and pathophysiological processes in the body. However, little is known about the function of these compounds in the central nervous system. Endogenous cardiotonic steroids are involved in the pathogenesis of affective disorders, including depression and bipolar disorder, which are linked to dopaminergic system dysfunction. Animal models have shown that the cardiotonic steroid ouabain induces mania-like behavior through dopamine-dependent intracellular signaling pathways. In addition, mutations in the alpha subunit of Na+,K+-ATPase lead to the development of neurological pathologies. Evidence from animal models confirms the neurological consequences of mutations in the Na+,K+-ATPase alpha subunit. This review is dedicated to discussing the role of cardiotonic steroids and Na+,K+-ATPase in dopaminergic system pathologies-both the evidence supporting their involvement and potential pathways along which they may exert their effects are evaluated. Since there is an association between affective disorders accompanied by functional alterations in the dopaminergic system and neurological disorders such as Parkinson's disease, we extend our discussion to the role of Na+,K+-ATPase and cardiotonic steroids in neurodegenerative diseases as well.
Collapse
Affiliation(s)
- Alisa A Markina
- Biological Department, Saint Petersburg State University, Universitetskaya Emb. 7/9, 199034 Saint Petersburg, Russia
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya Emb. 7/9, 199034 Saint Petersburg, Russia
| | - Rogneda B Kazanskaya
- Biological Department, Saint Petersburg State University, Universitetskaya Emb. 7/9, 199034 Saint Petersburg, Russia
- Research Center of Neurology, Volokolamskoye Ahosse 80, 125367 Moscow, Russia
| | - Julia A Timoshina
- Research Center of Neurology, Volokolamskoye Ahosse 80, 125367 Moscow, Russia
- Biological Department, Lomonosov Moscow State University, Leninskiye Gory 1, 119991 Moscow, Russia
| | - Vladislav A Zavialov
- Biological Department, Saint Petersburg State University, Universitetskaya Emb. 7/9, 199034 Saint Petersburg, Russia
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya Emb. 7/9, 199034 Saint Petersburg, Russia
| | - Denis A Abaimov
- Research Center of Neurology, Volokolamskoye Ahosse 80, 125367 Moscow, Russia
| | - Anna B Volnova
- Biological Department, Saint Petersburg State University, Universitetskaya Emb. 7/9, 199034 Saint Petersburg, Russia
| | - Tatiana N Fedorova
- Research Center of Neurology, Volokolamskoye Ahosse 80, 125367 Moscow, Russia
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya Emb. 7/9, 199034 Saint Petersburg, Russia
- Saint Petersburg University Hospital, 199034 Saint Petersburg, Russia
| | - Alexander V Lopachev
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya Emb. 7/9, 199034 Saint Petersburg, Russia
- Research Center of Neurology, Volokolamskoye Ahosse 80, 125367 Moscow, Russia
| |
Collapse
|
3
|
Wang J, Miao X, Sun Y, Li S, Wu A, Wei C. Dopaminergic System in Promoting Recovery from General Anesthesia. Brain Sci 2023; 13:brainsci13040538. [PMID: 37190503 DOI: 10.3390/brainsci13040538] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 05/17/2023] Open
Abstract
Dopamine is an important neurotransmitter that plays a biological role by binding to dopamine receptors. The dopaminergic system regulates neural activities, such as reward and punishment, memory, motor control, emotion, and sleep-wake. Numerous studies have confirmed that the dopaminergic system has the function of maintaining wakefulness in the body. In recent years, there has been increasing evidence that the sleep-wake cycle in the brain has similar neurobrain network mechanisms to those associated with the loss and recovery of consciousness induced by general anesthesia. With the continuous development and innovation of neurobiological techniques, the dopaminergic system has now been proved to be involved in the emergence from general anesthesia through the modulation of neuronal activity. This article is an overview of the dopaminergic system and the research progress into its role in wakefulness and general anesthesia recovery. It provides a theoretical basis for interpreting the mechanisms regulating consciousness during general anesthesia.
Collapse
Affiliation(s)
- Jinxu Wang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaolei Miao
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yi Sun
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Sijie Li
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
4
|
Oda S, Funato H. D1- and D2-type dopamine receptors are immunolocalized in pial and layer I astrocytes in the rat cerebral cortex. Front Neuroanat 2023; 17:1111008. [PMID: 36865631 PMCID: PMC9971002 DOI: 10.3389/fnana.2023.1111008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/25/2023] [Indexed: 02/16/2023] Open
Abstract
Pial astrocytes, a cellular component of the cerebral cortex surface structure, are observed in a wide range of mammalian species. Despite being recognized as such, the functional potential of pial astrocytes has long been overlooked. Our previous research demonstrated that pial astrocytes exhibit stronger immunoreactivity for muscarinic acetylcholine receptor M1 than protoplasmic astrocytes, indicating sensitivity to neuromodulators. Here, we examined whether pial astrocytes express receptors for dopamine, another crucial neuromodulator of cortical activity. We investigated the immunolocalization of each dopamine receptor subtype (D1R, D2R, D4R, D5R) in the rat cerebral cortex, and compared the intensity of immunoreactivity between pial astrocytes, protoplasmic astrocytes, and pyramidal cells. Our findings revealed that pial astrocytes and layer I astrocytes exhibit stronger D1R- and D4R-immunoreactivity than D2R and D5R. These immunoreactivities were primarily localized in the somata and thick processes of pial and layer I astrocytes. In contrast, protoplasmic astrocytes located in cortical layers II-VI displayed low or negligible immunoreactivities for dopamine receptors. D4R- and D5R-immunopositivity was distributed throughout pyramidal cells including somata and apical dendrites. These findings suggest that the dopaminergic system may regulate the activity of pial and layer I astrocytes via D1R and D4R.
Collapse
Affiliation(s)
- Satoko Oda
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, Japan,*Correspondence: Satoko Oda Hiromasa Funato
| | - Hiromasa Funato
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, Japan,International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Japan,*Correspondence: Satoko Oda Hiromasa Funato
| |
Collapse
|
5
|
Crespo M, León-Navarro DA, Martín M. Na +/K +- and Mg 2+-ATPases and Their Interaction with AMPA, NMDA and D 2 Dopamine Receptors in an Animal Model of Febrile Seizures. Int J Mol Sci 2022; 23:ijms232314638. [PMID: 36498965 PMCID: PMC9737571 DOI: 10.3390/ijms232314638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/05/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
Febrile seizures (FS) are one of the most common seizure disorders in childhood which are classified into short and prolonged, depending on their duration. Short FS are usually considered as benign. However, epidemiological studies have shown an association between prolonged FS and temporal lobe epilepsy. The development of animal models of FS has been very useful to investigate the mechanisms and the consequences of FS. One of the most used, the "hair dryer model", has revealed that prolonged FS may lead to temporal lobe epilepsy by altering neuronal function. Several pieces of evidence suggest that Na+/ K+-ATPase and Mg2+-ATPase may play a role in this epileptogenic process. In this work, we found that hyperthermia-induced seizures (HIS) significantly increased the activity of Na+/ K+-ATPase and Mg2+-ATPase five and twenty days after hyperthermic insult, respectively. These effects were diminished in response to AMPA, D2 dopamine A1 and A2A receptors activation, respectively. Furthermore, HIS also significantly increased the protein level of the AMPA subunit GluR1. Altogether, the increased Na+/ K+-ATPase and Mg2+-ATPase agree well with the presence of protective mechanisms. However, the reduction in ATPase activities in the presence of NMDA and AMPA suggest an increased propensity for epileptic events in adults.
Collapse
Affiliation(s)
- María Crespo
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, Regional Centre of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - David Agustín León-Navarro
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, Regional Centre of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
- Correspondence: ; Tel.: +34-926-052-114
| | - Mairena Martín
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Centre of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
| |
Collapse
|
6
|
Jomova K, Makova M, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Rhodes CJ, Valko M. Essential metals in health and disease. Chem Biol Interact 2022; 367:110173. [PMID: 36152810 DOI: 10.1016/j.cbi.2022.110173] [Citation(s) in RCA: 221] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/10/2022] [Accepted: 09/05/2022] [Indexed: 11/03/2022]
Abstract
In total, twenty elements appear to be essential for the correct functioning of the human body, half of which are metals and half are non-metals. Among those metals that are currently considered to be essential for normal biological functioning are four main group elements, sodium (Na), potassium (K), magnesium (Mg), and calcium (Ca), and six d-block transition metal elements, manganese (Mn), iron (Fe), cobalt (Co), copper (Cu), zinc (Zn) and molybdenum (Mo). Cells have developed various metallo-regulatory mechanisms for maintaining a necessary homeostasis of metal-ions for diverse cellular processes, most importantly in the central nervous system. Since redox active transition metals (for example Fe and Cu) may participate in electron transfer reactions, their homeostasis must be carefully controlled. The catalytic behaviour of redox metals which have escaped control, e.g. via the Fenton reaction, results in the formation of reactive hydroxyl radicals, which may cause damage to DNA, proteins and membranes. Transition metals are integral parts of the active centers of numerous enzymes (e.g. Cu,Zn-SOD, Mn-SOD, Catalase) which catalyze chemical reactions at physiologically compatible rates. Either a deficiency, or an excess of essential metals may result in various disease states arising in an organism. Some typical ailments that are characterized by a disturbed homeostasis of redox active metals include neurological disorders (Alzheimer's, Parkinson's and Huntington's disorders), mental health problems, cardiovascular diseases, cancer, and diabetes. To comprehend more deeply the mechanisms by which essential metals, acting either alone or in combination, and/or through their interaction with non-essential metals (e.g. chromium) function in biological systems will require the application of a broader, more interdisciplinary approach than has mainly been used so far. It is clear that a stronger cooperation between bioinorganic chemists and biophysicists - who have already achieved great success in understanding the structure and role of metalloenzymes in living systems - with biologists, will access new avenues of research in the systems biology of metal ions. With this in mind, the present paper reviews selected chemical and biological aspects of metal ions and their possible interactions in living systems under normal and pathological conditions.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences and Informatics, Constantine The Philosopher University in Nitra, 949 01, Nitra, Slovakia
| | - Marianna Makova
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, 812 37, Bratislava, Slovakia
| | - Suliman Y Alomar
- King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia
| | - Saleh H Alwasel
- King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | | | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, 812 37, Bratislava, Slovakia; King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia.
| |
Collapse
|
7
|
Ellingson PJ, Barnett WH, Kueh D, Vargas A, Calabrese RL, Cymbalyuk GS. Comodulation of h- and Na +/K + Pump Currents Expands the Range of Functional Bursting in a Central Pattern Generator by Navigating between Dysfunctional Regimes. J Neurosci 2021; 41:6468-6483. [PMID: 34103361 PMCID: PMC8318076 DOI: 10.1523/jneurosci.0158-21.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 11/21/2022] Open
Abstract
Central pattern generators (CPGs), specialized oscillatory neuronal networks controlling rhythmic motor behaviors such as breathing and locomotion, must adjust their patterns of activity to a variable environment and changing behavioral goals. Neuromodulation adjusts these patterns by orchestrating changes in multiple ionic currents. In the medicinal leech, the endogenous neuromodulator myomodulin speeds up the heartbeat CPG by reducing the electrogenic Na+/K+ pump current and increasing h-current in pairs of mutually inhibitory leech heart interneurons (HNs), which form half-center oscillators (HN HCOs). Here we investigate whether the comodulation of two currents could have advantages over a single current in the control of functional bursting patterns of a CPG. We use a conductance-based biophysical model of an HN HCO to explain the experimental effects of myomodulin. We demonstrate that, in the model, comodulation of the Na+/K+ pump current and h-current expands the range of functional bursting activity by avoiding transitions into nonfunctional regimes, such as asymmetric bursting and plateau-containing seizure-like activity. We validate the model by finding parameters that reproduce temporal bursting characteristics matching experimental recordings from HN HCOs under control, three different myomodulin concentrations, and Cs+ treated conditions. The matching cases are located along the border of an asymmetric regime away from the border with more dangerous seizure-like activity. We found a simple comodulation mechanism with an inverse relation between the pump and h-currents makes a good fit of the matching cases and comprises a general mechanism for the robust and flexible control of oscillatory neuronal networks.SIGNIFICANCE STATEMENT Rhythm-generating neuronal circuits adjust their oscillatory patterns to accommodate a changing environment through neuromodulation. In different species, chemical messengers participating in such processes may target two or more membrane currents. In medicinal leeches, the neuromodulator myomodulin speeds up the heartbeat central pattern generator by reducing Na+/K+ pump current and increasing h-current. In a computational model, we show that this comodulation expands the range of central pattern generator's functional activity by navigating the circuit between dysfunctional regimes resulting in a much wider range of cycle period. This control would not be attainable by modulating only one current, emphasizing the synergy of combined effects. Given the prevalence of h-current and Na+/K+ pump current in neurons, similar comodulation mechanisms may exist across species.
Collapse
Affiliation(s)
- Parker J Ellingson
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - William H Barnett
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - Daniel Kueh
- Department of Biology, Emory University, Atlanta, Georgia 30322
| | - Alex Vargas
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | | | | |
Collapse
|
8
|
Kravtsova VV, Krivoi II. Molecular and Functional Heterogeneity of Na,K-ATPase in the Skeletal Muscle. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
Lopachev AV, Lagarkova MA, Lebedeva OS, Ezhova MA, Kazanskaya RB, Timoshina YA, Khutorova AV, Akkuratov EE, Fedorova TN, Gainetdinov RR. Ouabain-Induced Gene Expression Changes in Human iPSC-Derived Neuron Culture Expressing Dopamine and cAMP-Regulated Phosphoprotein 32 and GABA Receptors. Brain Sci 2021; 11:brainsci11020203. [PMID: 33562186 PMCID: PMC7915459 DOI: 10.3390/brainsci11020203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/22/2022] Open
Abstract
Cardiotonic steroids (CTS) are specific inhibitors and endogenous ligands of a key enzyme in the CNS-the Na+, K+-ATPase, which maintains and creates an ion gradient on the plasma membrane of neurons. CTS cause the activation of various signaling cascades and changes in gene expression in neurons and other cell types. It is known that intracerebroventricular injection of cardiotonic steroid ouabain causes mania-like behavior in rodents, in part due to activation of dopamine-related signaling cascades in the dopamine and cAMP-regulated phosphoprotein 32 (DARPP-32) expressing medium spiny neurons in the striatum. Dopaminergic projections in the striatum innervate these GABAergic medium spiny neurons. The objective of this study was to assess changes in the expression of all genes in human iPSC-derived expressing DARPP-32 and GABA receptors neurons under the influence of ouabain. We noted a large number of statistically significant upregulated and downregulated genes after a 16-h incubation with non-toxic concentration (30 nM) of ouabain. These changes in the transcriptional activity were accomplished with activation of MAP-kinase ERK1/2 and transcriptional factor cAMP response element-binding protein (CREB). Thus, it can be concluded that 30 nM ouabain incubated for 16 h with human iPSC-derived expressing DARPP-32 and GABA receptors neurons activates genes associated with neuronal maturation and synapse formation, by increasing the expression of genes associated with translation, vesicular transport, and increased electron transport chain function. At the same time, the expression of genes associated with proliferation, migration, and early development of neurons decreases. These data indicate that non-toxic concentrations of ouabain may induce neuronal maturation, neurite growth, and increased synaptogenesis in dopamine-receptive GABAergic neurons, suggesting formation of plasticity and the establishment of new neuronal junctions.
Collapse
Affiliation(s)
- Alexander V. Lopachev
- Laboratory of Clinical and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia; (Y.A.T.); (A.V.K.); (T.N.F.)
- Correspondence:
| | - Maria A. Lagarkova
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine Federal Medical Biological Agency, 119435 Moscow, Russia; (M.A.L.); (O.S.L.)
| | - Olga S. Lebedeva
- Laboratory of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine Federal Medical Biological Agency, 119435 Moscow, Russia; (M.A.L.); (O.S.L.)
| | - Margarita A. Ezhova
- Laboratory of Plant Genomics, Institute for Information Transmission Problems of the Russian Academy of Sciences, 127051 Moscow, Russia;
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| | - Rogneda B. Kazanskaya
- Biological Department, Saint Petersburg State University, 199034 St. Petersburg, Russia;
| | - Yulia A. Timoshina
- Laboratory of Clinical and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia; (Y.A.T.); (A.V.K.); (T.N.F.)
- Biological Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Anastasiya V. Khutorova
- Laboratory of Clinical and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia; (Y.A.T.); (A.V.K.); (T.N.F.)
- Biological Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Evgeny E. Akkuratov
- Department of Applied Physics, Royal Institute of Technology, Science for Life Laboratory, 171 65 Stockholm, Sweden;
| | - Tatiana N. Fedorova
- Laboratory of Clinical and Experimental Neurochemistry, Research Center of Neurology, 125367 Moscow, Russia; (Y.A.T.); (A.V.K.); (T.N.F.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine and Saint Petersburg University Hospital, Saint Petersburg State University, 199034 St. Petersburg, Russia;
| |
Collapse
|
10
|
Ghaffari H, Grant SC, Petzold LR, Harrington MG. Regulation of CSF and Brain Tissue Sodium Levels by the Blood-CSF and Blood-Brain Barriers During Migraine. Front Comput Neurosci 2020; 14:4. [PMID: 32116618 PMCID: PMC7010722 DOI: 10.3389/fncom.2020.00004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/10/2020] [Indexed: 11/13/2022] Open
Abstract
Cerebrospinal fluid (CSF) and brain tissue sodium levels increase during migraine. However, little is known regarding the underlying mechanisms of sodium homeostasis disturbance in the brain during the onset and propagation of migraine. Exploring the cause of sodium dysregulation in the brain is important, since correction of the altered sodium homeostasis could potentially treat migraine. Under the hypothesis that disturbances in sodium transport mechanisms at the blood-CSF barrier (BCSFB) and/or the blood-brain barrier (BBB) are the underlying cause of the elevated CSF and brain tissue sodium levels during migraines, we developed a mechanistic, differential equation model of a rat's brain to compare the significance of the BCSFB and the BBB in controlling CSF and brain tissue sodium levels. The model includes the ventricular system, subarachnoid space, brain tissue and blood. Sodium transport from blood to CSF across the BCSFB, and from blood to brain tissue across the BBB were modeled by influx permeability coefficients PBCSFB and PBBB, respectively, while sodium movement from CSF into blood across the BCSFB, and from brain tissue to blood across the BBB were modeled by efflux permeability coefficients PBCSFB′ and PBBB′, respectively. We then performed a global sensitivity analysis to investigate the sensitivity of the ventricular CSF, subarachnoid CSF and brain tissue sodium concentrations to pathophysiological variations in PBCSFB, PBBB, PBCSFB′ and PBBB′. Our results show that the ventricular CSF sodium concentration is highly influenced by perturbations of PBCSFB, and to a much lesser extent by perturbations of PBCSFB′. Brain tissue and subarachnoid CSF sodium concentrations are more sensitive to pathophysiological variations of PBBB and PBBB′ than variations of PBCSFB and PBCSFB′ within 30 min of the onset of the perturbations. However, PBCSFB is the most sensitive model parameter, followed by PBBB and PBBB′, in controlling brain tissue and subarachnoid CSF sodium levels within 3 h of the perturbation onset.
Collapse
Affiliation(s)
- Hamed Ghaffari
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Samuel C Grant
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL, United States
| | - Linda R Petzold
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Michael G Harrington
- Neuroscience, Huntington Medical Research Institutes, Pasadena, CA, United States
| |
Collapse
|
11
|
Askari A. The other functions of the sodium pump. Cell Calcium 2019; 84:102105. [DOI: 10.1016/j.ceca.2019.102105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/21/2019] [Accepted: 10/28/2019] [Indexed: 01/14/2023]
|
12
|
Yang Z, Zhou J, Wei B, Cheng Y, Zhang L, Zhen X. Comparative transcriptome analysis reveals osmotic-regulated genes in the gill of Chinese mitten crab (Eriocheir sinensis). PLoS One 2019; 14:e0210469. [PMID: 30629688 PMCID: PMC6328174 DOI: 10.1371/journal.pone.0210469] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 12/24/2018] [Indexed: 11/19/2022] Open
Abstract
Salinity is one of the most important abiotic factors directly affecting the reproduction, molting, growth, immune, physiological and metabolic activities of Chinese mitten crab (Eriocheir sinensis). This species has strong osmoregulatory capacity and can maintain stringent internal homeostasis. However, the mechanisms conferring tolerance to salinity fluctuations are not well understood. To reveal the genes and pathways involved in osmoregulation, adult male crabs (body weight = 110 ± 5 g) were acclimated for 144 h in freshwater (FW, 0 ppt) or seawater (SW, 25 ppt). Changes in the transcriptome of crab gills were then analysed by RNA-Seq, and 174,903 unigenes were obtained. Comparison of genes between FW- SW-acclimated groups identified 932 genes that were significantly differentially expressed in the gill, comprising 433 and 499 up- and downregulated transcripts. Gene Ontology functional enrichment analysis revealed that important biological processes related to salt stress were significantly enriched, including energy metabolism, ion transport, signal transduction and antioxidant activity. Kyoto Encyclopaedia of Genes and Genomes enrichment analysis mapped the differentially expressed genes to 241 specific metabolic pathways, and pathways related to energy metabolism, oxidative phosphorylation and the tricarboxylic acid (TCA)/citrate cycle were significantly enriched. Salinity stress altered the expression of many enzymes involved in energy metabolism, ion transport, signal transduction and antioxidant pathways, including citrate synthase (CS), Na+/K+-ATPase (NKA), Na+-K+-2Cl cotransporter-1 (NKCC1), dopamine receptor D1 (DRD1), synaptic binding protein 1 (STXBP1), Cu2+/Zn2+ superoxide dismutase (SOD1) and glutathione S-transferase (GST). Additionally, the obtained transcriptomic sequencing data provided a useful resource for identification of novel genes, and further physiological analysis of Chinese mitten crab.
Collapse
Affiliation(s)
- Zhigang Yang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
- * E-mail: (ZGY); (YXC)
| | - Junyu Zhou
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Banghong Wei
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Yongxu Cheng
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
- * E-mail: (ZGY); (YXC)
| | - Long Zhang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xiaomin Zhen
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| |
Collapse
|
13
|
Na +/K +-pump and neurotransmitter membrane receptors. INVERTEBRATE NEUROSCIENCE 2018; 19:1. [PMID: 30488358 PMCID: PMC6267510 DOI: 10.1007/s10158-018-0221-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/17/2018] [Indexed: 02/06/2023]
Abstract
Na+/K+-pump is an electrogenic transmembrane ATPase located in the outer plasma membrane of cells. The Na+/K+-ATPase pumps 3 sodium ions out of cells while pumping 2 potassium ions into cells. Both cations move against their concentration gradients. This enzyme's electrogenic nature means that it has a chronic role in stabilizing the resting membrane potential of the cell, in regulating the cell volume and in the signal transduction of the cell. This review will mainly consider the role of the Na+/K+-pump in neurons, with an emphasis on its role in modulating neurotransmitter receptor. Most of the literature on the modulation of neurotransmitter receptors refers to the situation in the mammalian nervous system, but the position is likely to be similar in most, if not all, invertebrate nervous systems.
Collapse
|
14
|
Bolotta A, Visconti P, Fedrizzi G, Ghezzo A, Marini M, Manunta P, Messaggio E, Posar A, Vignini A, Abruzzo PM. Na + , K + -ATPase activity in children with autism spectrum disorder: Searching for the reason(s) of its decrease in blood cells. Autism Res 2018; 11:1388-1403. [PMID: 30120881 PMCID: PMC6221099 DOI: 10.1002/aur.2002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/24/2018] [Accepted: 06/03/2018] [Indexed: 12/27/2022]
Abstract
Na+, K+‐ATPase (NKA) activity, which establishes the sodium and potassium gradient across the cell membrane and is instrumental in the propagation of the nerve impulses, is altered in a number of neurological and neuropsychiatric disorders, including autism spectrum disorders (ASD). In the present work, we examined a wide range of biochemical and cellular parameters in the attempt to understand the reason(s) for the severe decrease in NKA activity in erythrocytes of ASD children that we reported previously. NKA activity in leukocytes was found to be decreased independently from alteration in plasma membrane fluidity. The different subunits were evaluated for gene expression in leukocytes and for protein expression in erythrocytes: small differences in gene expression between ASD and typically developing children were not apparently paralleled by differences in protein expression. Moreover, no gross difference in erythrocyte plasma membrane oxidative modifications was detectable, although oxidative stress in blood samples from ASD children was confirmed by increased expression of NRF2 mRNA. Interestingly, gene expression of some NKA subunits correlated with clinical features. Excess inhibitory metals or ouabain‐like activities, which might account for NKA activity decrease, were ruled out. Plasma membrane cholesterol, but not phosphatidylcholine and phosphatidlserine, was slighty decreased in erythrocytes from ASD children. Although no compelling results were obtained, our data suggest that alteration in the erytrocyte lipid moiety or subtle oxidative modifications in NKA structure are likely candidates for the observed decrease in NKA activity. These findings are discussed in the light of the relevance of NKA in ASD. Autism Res2018, 11: 1388–1403. © 2018 International Society for Autism Research, Wiley Periodicals, Inc. Lay Summary The activity of the cell membrane enzyme NKA, which is instrumental in the propagation of the nerve impulses, is severely decreased in erythrocytes from ASD children and in other brain disorders, yet no explanation has been provided for this observation. We strived to find a biological/biochemical cause of such alteration, but most queries went unsolved because of the complexity of NKA regulation. As NKA activity is altered in many brain disorders, we stress the relevance of studies aimed at understanding its regulation in ASD.
Collapse
Affiliation(s)
- Alessandra Bolotta
- From the Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Paola Visconti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Giorgio Fedrizzi
- Chemical Department, IZSLER Zooprophylactic Experimental Institute for Lombardy and Emilia Romagna, Bologna, Italy
| | - Alessandro Ghezzo
- From the Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Marina Marini
- From the Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Paolo Manunta
- University and Hospital Vita-Salute, Milan, Italy.,Chair of Nephrology, University Vita Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Annio Posar
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Arianna Vignini
- Department of Clinical Sciences - Section of Biochemistry, Biology and Physics, Polytechnic University of Marche, Ancona, Italy
| | - Provvidenza Maria Abruzzo
- From the Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| |
Collapse
|
15
|
Dopamine Pumping Up Spinal Locomotor Network Function. J Neurosci 2018; 37:3103-3105. [PMID: 28330979 DOI: 10.1523/jneurosci.0019-17.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/06/2017] [Accepted: 02/16/2017] [Indexed: 01/11/2023] Open
|
16
|
Sodium Pumps Mediate Activity-Dependent Changes in Mammalian Motor Networks. J Neurosci 2017; 37:906-921. [PMID: 28123025 PMCID: PMC5296784 DOI: 10.1523/jneurosci.2005-16.2016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/29/2016] [Accepted: 12/07/2016] [Indexed: 12/24/2022] Open
Abstract
Ubiquitously expressed sodium pumps are best known for maintaining the ionic gradients and resting membrane potential required for generating action potentials. However, activity- and state-dependent changes in pump activity can also influence neuronal firing and regulate rhythmic network output. Here we demonstrate that changes in sodium pump activity regulate locomotor networks in the spinal cord of neonatal mice. The sodium pump inhibitor, ouabain, increased the frequency and decreased the amplitude of drug-induced locomotor bursting, effects that were dependent on the presence of the neuromodulator dopamine. Conversely, activating the pump with the sodium ionophore monensin decreased burst frequency. When more "natural" locomotor output was evoked using dorsal-root stimulation, ouabain increased burst frequency and extended locomotor episode duration, whereas monensin slowed and shortened episodes. Decreasing the time between dorsal-root stimulation, and therefore interepisode interval, also shortened and slowed activity, suggesting that pump activity encodes information about past network output and contributes to feedforward control of subsequent locomotor bouts. Using whole-cell patch-clamp recordings from spinal motoneurons and interneurons, we describe a long-duration (∼60 s), activity-dependent, TTX- and ouabain-sensitive, hyperpolarization (∼5 mV), which is mediated by spike-dependent increases in pump activity. The duration of this dynamic pump potential is enhanced by dopamine. Our results therefore reveal sodium pumps as dynamic regulators of mammalian spinal motor networks that can also be affected by neuromodulatory systems. Given the involvement of sodium pumps in movement disorders, such as amyotrophic lateral sclerosis and rapid-onset dystonia parkinsonism, knowledge of their contribution to motor network regulation also has considerable clinical importance. SIGNIFICANCE STATEMENT The sodium pump is ubiquitously expressed and responsible for at least half of total brain energy consumption. The pumps maintain ionic gradients and the resting membrane potential of neurons, but increasing evidence suggests that activity- and state-dependent changes in pump activity also influence neuronal firing. Here we demonstrate that changes in sodium pump activity regulate locomotor output in the spinal cord of neonatal mice. We describe a sodium pump-mediated afterhyperpolarization in spinal neurons, mediated by spike-dependent increases in pump activity, which is affected by dopamine. Understanding how sodium pumps contribute to network regulation and are targeted by neuromodulators, including dopamine, has clinical relevance due to the role of the sodium pump in diseases, including amyotrophic lateral sclerosis, parkinsonism, epilepsy, and hemiplegic migraine.
Collapse
|
17
|
Fisetin inhibits liver cancer growth in a mouse model: Relation to dopamine receptor. Oncol Rep 2017; 38:53-62. [PMID: 28560391 PMCID: PMC5492805 DOI: 10.3892/or.2017.5676] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/03/2017] [Indexed: 12/19/2022] Open
Abstract
Fisetin (3,3',4',7-tetrahydroxyflavone), a natural abundant flavonoid, is produced in different vegetables and fruits. Fisetin has been reported to relate to various positive biological effects, including anti-proliferative, anticancer, anti-oxidative and neuroprotective effects. Dopamine receptors (DRs) belonging to G protein‑coupled receptor family, are known as the target of ~50% of all modern medicinal drugs. DRs consist of various proteins, functioning as transduction of intracellular signals for extracellular stimuli. We found that fisetin performed as DR2 agonist to suppress liver cancer cells proliferation, migration and invasion. Caspase-3 signaling was activated to induce apoptosis for fisetin administration. Furthermore, TGF‑β1 was also inhibited in fisetin-treated liver cancer cells, reducing epithelial-mesenchymal transition (EMT). Additionally, fisetin downregulated VEGFR1, p-ERK1/2, p38 and pJNK, ameliorating liver cancer progression. In vivo, the orthotopically implanted tumors from mice were inhibited by fisetin adminisatration accompanied by prolonged survival rate and higher levels of dopamine. Together, the results indicated a novel therapeutic strategy to suppress liver cancer progression associated with DR2 regulation, indicating that dopamine might be of importance in liver cancer progression.
Collapse
|
18
|
Rosin C, López Ordieres MG, Rodríguez de Lores Arnaiz G. Changes in [ 3H]-ouabain and [ 3H]-neurotensin binding to rat cerebral cortex membranes after administration of antipsychotic drugs haloperidol and clozapine. Peptides 2017; 89:82-89. [PMID: 27586561 DOI: 10.1016/j.peptides.2016.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/08/2016] [Accepted: 08/29/2016] [Indexed: 11/17/2022]
Abstract
Evidences indicate the relationship between neurotensinergic and dopaminergic systems. Neurotensin inhibits synaptosomal membrane Na+, K+-ATPase activity, an effect blocked by SR 48692, antagonist for high affinity neurotensin receptor (NTS1) type. Assays of high affinity [3H]-ouabain binding (to analyze K+ site of Na+, K+-ATPase) show that in vitro addition of neurotensin decreases binding. Herein potential interaction between NTS1 receptor, dopaminergic D2 receptor and Na+, K+-ATPase was studied. To test the involvement of dopaminergic D2 receptors in [3H]-ouabain binding inhibition by neurotensin, Wistar rats were administered i.p.with antipsychotic drugs haloperidol (2mg/kg) and clozapine (3, 10 and 30mg/kg). Animals were sacrificed 18h later, cerebral cortices harvested, membrane fractions prepared and high affinity [3H]-ouabain binding assayed in the absence or presence of neurotensin at a 10 micromolar concentration. No differences versus controls for basal binding or for binding inhibition by neurotensin were recorded, except after 10mg/kg clozapine. Rats were administered with neurotensin (3, 10y 30μg, i.c.v.) and 60min later, animals were sacrificed, cerebral cortices harvested and processed to obtain membrane fractions for high affinity [3H]-ouabain binding assays. Results showed a slight but statistically significant decrease in binding with the 30μg neurotensin dose. To analyze the interaction between dopaminergic D2 and NTS1 receptors, [3H]-neurotensin binding to cortical membranes from rats injected with haloperidol (2mg/kg, i.p.) or clozapine (10mg/kg) was assayed. Saturation curves and Scatchard transformation showed that the only statistically significant change occurred in Bmax after haloperidol administration. Hill number was close to the unit in all cases. Results indicated that typical and atypical antipsychotic drugs differentially modulate the interaction between neurotensin and Na+, K+-ATPase. At the same time, support the notion of an interaction among dopaminergic and neurotensinergic systems and Na+, K+-ATPase at central synapses.
Collapse
Affiliation(s)
- Carina Rosin
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis", Facultad de Medicina, CONICET-UBA, Universidad de Buenos Aires, Paraguay 2155, 1121-Buenos Aires, Argentina; Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113-Buenos Aires, Argentina
| | - María Graciela López Ordieres
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis", Facultad de Medicina, CONICET-UBA, Universidad de Buenos Aires, Paraguay 2155, 1121-Buenos Aires, Argentina; Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113-Buenos Aires, Argentina
| | - Georgina Rodríguez de Lores Arnaiz
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis", Facultad de Medicina, CONICET-UBA, Universidad de Buenos Aires, Paraguay 2155, 1121-Buenos Aires, Argentina.
| |
Collapse
|
19
|
Kuter K, Kratochwil M, Marx SH, Hartwig S, Lehr S, Sugawa MD, Dencher NA. Native DIGE proteomic analysis of mitochondria from substantia nigra and striatum during neuronal degeneration and its compensation in an animal model of early Parkinson's disease. Arch Physiol Biochem 2016; 122:238-256. [PMID: 27467289 DOI: 10.1080/13813455.2016.1197948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cause of Parkinson's disease (PD) is still not understood. Motor symptoms are not observed at early stages of disease due to compensatory processes. Dysfunction of mitochondria was indicated already at preclinical PD. Selective toxin 6-OHDA was applied to kill dopaminergic neurons in substantia nigra and disturb neuronal transmission in striatum. Early phase of active degeneration and later stage, when surviving cells adapted to function normally, were analysed. 2D BN/SDS difference gel electrophoresis (DIGE) of mitochondrial proteome enabled to point out crucial processes involved at both time-points in dopaminergic structures. Marker proteins such as DPYSL2, HSP60, ATP1A3, EAAT2 indicated structural remodelling, cytoskeleton rearrangement, organelle trafficking, axon outgrowth and regeneration. Adaptations in dopaminergic and glutamatergic neurotransmission, recycling of synaptic vesicles, along with enlargement of mitochondria mass were proposed as causative for compensation. Changed expression of carbohydrates metabolism and oxidative phosphorylation proteins were described, including their protein-protein interactions and supercomplex assembly.
Collapse
Affiliation(s)
- Katarzyna Kuter
- a Department of Neuropsychopharmacology , Polish Academy of Sciences , Kraków , Poland
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Manuela Kratochwil
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Sven-Hendric Marx
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| | - Sonja Hartwig
- c Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Düsseldorf, Leibniz Center for Diabetes Research , Düsseldorf , Germany
- d German Center for Diabetes Research (DZD) , München, Neuherberg , Germany , and
| | - Stephan Lehr
- c Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Düsseldorf, Leibniz Center for Diabetes Research , Düsseldorf , Germany
- d German Center for Diabetes Research (DZD) , München, Neuherberg , Germany , and
| | - Michiru D Sugawa
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
- e Clinical Neurobiology, Charité-Universitätsmedizin , Berlin , Germany
| | - Norbert A Dencher
- b Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt , Darmstadt , Germany
| |
Collapse
|
20
|
Holm TH, Lykke-Hartmann K. Insights into the Pathology of the α3 Na(+)/K(+)-ATPase Ion Pump in Neurological Disorders; Lessons from Animal Models. Front Physiol 2016; 7:209. [PMID: 27378932 PMCID: PMC4906016 DOI: 10.3389/fphys.2016.00209] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 05/22/2016] [Indexed: 01/08/2023] Open
Abstract
The transmembrane Na(+)-/K(+) ATPase is located at the plasma membrane of all mammalian cells. The Na(+)-/K(+) ATPase utilizes energy from ATP hydrolysis to extrude three Na(+) cations and import two K(+) cations into the cell. The minimum constellation for an active Na(+)-/K(+) ATPase is one alpha (α) and one beta (β) subunit. Mammals express four α isoforms (α1-4), encoded by the ATP1A1-4 genes, respectively. The α1 isoform is ubiquitously expressed in the adult central nervous system (CNS) whereas α2 primarily is expressed in astrocytes and α3 in neurons. Na(+) and K(+) are the principal ions involved in action potential propagation during neuronal depolarization. The α1 and α3 Na(+)-/K(+) ATPases are therefore prime candidates for restoring neuronal membrane potential after depolarization and for maintaining neuronal excitability. The α3 isoform has approximately four-fold lower Na(+) affinity compared to α1 and is specifically required for rapid restoration of large transient increases in [Na(+)]i. Conditions associated with α3 deficiency are therefore likely aggravated by suprathreshold neuronal activity. The α3 isoform been suggested to support re-uptake of neurotransmitters. These processes are required for normal brain activity, and in fact autosomal dominant de novo mutations in ATP1A3 encoding the α3 isoform has been found to cause the three neurological diseases Rapid Onset Dystonia Parkinsonism (RDP), Alternating Hemiplegia of Childhood (AHC), and Cerebellar ataxia, areflexia, pes cavus, optic atrophy, and sensorineural hearing loss (CAPOS). All three diseases cause acute onset of neurological symptoms, but the predominant neurological manifestations differ with particularly early onset of hemiplegic/dystonic episodes and mental decline in AHC, ataxic encephalopathy and impairment of vision and hearing in CAPOS syndrome and late onset of dystonia/parkinsonism in RDP. Several mouse models have been generated to study the in vivo consequences of Atp1a3 modulation. The different mice show varying degrees of hyperactivity, gait problems, and learning disability as well as stress-induced seizures. With the advent of several Atp1a3-gene or chemically modified animal models that closely phenocopy many aspects of the human disorders, we will be able to reach a much better understanding of the etiology of RDP, AHC, and CAPOS syndrome.
Collapse
Affiliation(s)
- Thomas H. Holm
- Department of Biomedicine, Aarhus UniversityAarhus, Denmark
- Department of Molecular Biology and Genetics, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Aarhus UniversityAarhus, Denmark
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus UniversityAarhus, Denmark
- Department of Molecular Biology and Genetics, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Danish National Research Foundation, Aarhus UniversityAarhus, Denmark
- Aarhus Institute of Advanced Studies, Aarhus UniversityAarhus, Denmark
| |
Collapse
|
21
|
Matchkov VV, Krivoi II. Specialized Functional Diversity and Interactions of the Na,K-ATPase. Front Physiol 2016; 7:179. [PMID: 27252653 PMCID: PMC4879863 DOI: 10.3389/fphys.2016.00179] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/04/2016] [Indexed: 12/22/2022] Open
Abstract
Na,K-ATPase is a protein ubiquitously expressed in the plasma membrane of all animal cells and vitally essential for their functions. A specialized functional diversity of the Na,K-ATPase isozymes is provided by molecular heterogeneity, distinct subcellular localizations, and functional interactions with molecular environment. Studies over the last decades clearly demonstrated complex and isoform-specific reciprocal functional interactions between the Na,K-ATPase and neighboring proteins and lipids. These interactions are enabled by a spatially restricted ion homeostasis, direct protein-protein/lipid interactions, and protein kinase signaling pathways. In addition to its "classical" function in ion translocation, the Na,K-ATPase is now considered as one of the most important signaling molecules in neuronal, epithelial, skeletal, cardiac and vascular tissues. Accordingly, the Na,K-ATPase forms specialized sub-cellular multimolecular microdomains which act as receptors to circulating endogenous cardiotonic steroids (CTS) triggering a number of signaling pathways. Changes in these endogenous cardiotonic steroid levels and initiated signaling responses have significant adaptive values for tissues and whole organisms under numerous physiological and pathophysiological conditions. This review discusses recent progress in the studies of functional interactions between the Na,K-ATPase and molecular microenvironment, the Na,K-ATPase-dependent signaling pathways and their significance for diversity of cell function.
Collapse
Affiliation(s)
| | - Igor I Krivoi
- Department of General Physiology, St. Petersburg State University St. Petersburg, Russia
| |
Collapse
|
22
|
Abstract
The variety of physiological functions controlled by dopamine in the brain and periphery is mediated by the D1, D2, D3, D4 and D5 dopamine GPCRs. Drugs acting on dopamine receptors are significant tools for the management of several neuropsychiatric disorders including schizophrenia, bipolar disorder, depression and Parkinson's disease. Recent investigations of dopamine receptor signalling have shown that dopamine receptors, apart from their canonical action on cAMP-mediated signalling, can regulate a myriad of cellular responses to fine-tune the expression of dopamine-associated behaviours and functions. Such signalling mechanisms may involve alternate G protein coupling or non-G protein mechanisms involving ion channels, receptor tyrosine kinases or proteins such as β-arrestins that are classically involved in GPCR desensitization. Another level of complexity is the growing appreciation of the physiological roles played by dopamine receptor heteromers. Applications of new in vivo techniques have significantly furthered the understanding of the physiological functions played by dopamine receptors. Here we provide an update of the current knowledge regarding the complex biology, signalling, physiology and pharmacology of dopamine receptors.
Collapse
|
23
|
Krivoi II. Functional interactions of Na,K-ATPase with molecular environment. Biophysics (Nagoya-shi) 2014. [DOI: 10.1134/s000635091405011x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
24
|
Ennis RC, Asico LD, Armando I, Yang J, Feranil JB, Jurgens JA, Escano CS, Yu P, Wang X, Sibley DR, Jose PA, Villar VAM. Dopamine D₁-like receptors regulate the α₁A-adrenergic receptor in human renal proximal tubule cells and D₁-like dopamine receptor knockout mice. Am J Physiol Renal Physiol 2014; 307:F1238-48. [PMID: 25339698 DOI: 10.1152/ajprenal.00119.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The homeostatic control of blood pressure hinges upon the delicate balance between prohypertensinogenic and antihypertensinogenic systems. D₁-like dopamine receptors [dopamine D₁ and D₅ receptors (D₁Rs and D₅Rs, respectively)] and the α₁A-adrenergic receptor (α₁A-AR) are expressed in the renal proximal tubule and engender opposing effects on Na(+) transport, i.e., natriuresis (via D₁Rs and D5Rs) or antinatriuresis (via α₁A-ARs). We tested the hypothesis that the D₁R/D₅R regulates the α₁A-AR. D₁-like dopamine receptors coimmunoprecipitated, colocalized, and cofractionated with α₁A-ARs in lipid rafts in immortalized human renal proximal tubule cells. Long-term treatment with the D₁R/D₅R agonist fenoldopam resulted in decreased D₁R and D₅R expression but increased α₁A-AR abundance in the plasma membrane. Short-term fenoldopam treatment stimulated the translocation of Na(+)-K(+)-ATPase from the plasma membrane to the cytosol that was partially reversed by an α₁A-AR agonist, which by itself induced Na(+)-K(+)-ATPase translocation from the cytosol to the plasma membrane. The α₁A-AR-specific agonist A610603 also minimized the ability of fenoldopam to inhibit Na(+)-K(+)-ATPase activity. To determine the interaction among D₁Rs, D₅Rs, and α₁A-ARs in vivo, we used phenylephrine and A610603 to decrease Na(+) excretion in several D1-like dopamine receptor knockout mouse strains. Phenylephrine and A61603 treatment resulted in a partial reduction of urinary Na(+) excretion in wild-type mice and its abolition in D1R knockout, D₅R knockout, and D₁R-D₅R double-knockout mice. Our results demonstrate the ability of the D₁-like dopamine receptors to regulate the expression and activity of α₁A-AR. Elucidating the intricacies of the interaction among these receptors is crucial for a better understanding of the crosstalk between anti- and pro-hypertensive systems.
Collapse
Affiliation(s)
- Riley Charles Ennis
- Thomas Jefferson High School for Science and Technology, Alexandria, Virgina
| | - Laureano D Asico
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ines Armando
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jian Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jun B Feranil
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Julie A Jurgens
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Crisanto S Escano
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peiying Yu
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Xiaoyan Wang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Van Anthony M Villar
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland;
| |
Collapse
|
25
|
Laßek M, Weingarten J, Volknandt W. The Proteome of the Murine Presynaptic Active Zone. Proteomes 2014; 2:243-257. [PMID: 28250380 PMCID: PMC5302740 DOI: 10.3390/proteomes2020243] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/09/2014] [Accepted: 04/21/2014] [Indexed: 01/09/2023] Open
Abstract
The proteome of the presynaptic active zone controls neurotransmitter release and the short- and long-term structural and functional dynamics of the nerve terminal. The proteinaceous inventory of the presynaptic active zone has recently been reported. This review will evaluate the subcellular fractionation protocols and the proteomic approaches employed. A breakthrough for the identification of the proteome of the presynaptic active zone was the successful employment of antibodies directed against a cytosolic epitope of membrane integral synaptic vesicle proteins for the immunopurification of synaptic vesicles docked to the presynaptic plasma membrane. Combining immunopurification and subsequent analytical mass spectrometry, hundreds of proteins, including synaptic vesicle proteins, components of the presynaptic fusion and retrieval machinery, proteins involved in intracellular and extracellular signaling and a large variety of adhesion molecules, were identified. Numerous proteins regulating the rearrangement of the cytoskeleton are indicative of the functional and structural dynamics of the presynapse. This review will critically discuss both the experimental approaches and prominent protein candidates identified. Many proteins have not previously been assigned to the presynaptic release sites and may be directly involved in the short- and long-term structural modulation of the presynaptic compartment. The identification of proteinaceous constituents of the presynaptic active zone provides the basis for further analyzing the interaction of presynaptic proteins with their targets and opens novel insights into the functional role of these proteins in neuronal communication.
Collapse
Affiliation(s)
- Melanie Laßek
- Institute for Cell Biology and Neuroscience, Department Molecular and Cellular Neurobiology, Max von Laue Str. 13, 60438 Frankfurt am Main, Germany.
| | - Jens Weingarten
- Institute for Cell Biology and Neuroscience, Department Molecular and Cellular Neurobiology, Max von Laue Str. 13, 60438 Frankfurt am Main, Germany.
| | - Walter Volknandt
- Institute for Cell Biology and Neuroscience, Department Molecular and Cellular Neurobiology, Max von Laue Str. 13, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
26
|
Weingarten J, Lassek M, Mueller BF, Rohmer M, Lunger I, Baeumlisberger D, Dudek S, Gogesch P, Karas M, Volknandt W. The proteome of the presynaptic active zone from mouse brain. Mol Cell Neurosci 2014; 59:106-18. [PMID: 24534009 DOI: 10.1016/j.mcn.2014.02.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 02/05/2014] [Accepted: 02/07/2014] [Indexed: 01/07/2023] Open
Abstract
Neurotransmitter release as well as the structural and functional dynamics of the presynaptic active zone is controlled by proteinaceous components. Here we describe for the first time an experimental approach for the isolation of the presynaptic active zone from individual mouse brains, a prerequisite for understanding the functional inventory of the presynaptic protein network and for the later analysis of changes occurring in mutant mice. Using a monoclonal antibody against the ubiquitous synaptic vesicle protein SV2 we immunopurified synaptic vesicles docked to the presynaptic plasma membrane. Enrichment studies by means of Western blot analysis and mass spectrometry identified 485 proteins belonging to an impressive variety of functional categories. Our data suggest that presynaptic active zones represent focal hot spots that are not only involved in the regulation of neurotransmitter release but also in multiple structural and functional alterations the adult nerve terminal undergoes during neural activity in adult CNS. They furthermore open new avenues for characterizing alterations in the active zone proteome of mutant mice and their corresponding controls, including the various mouse models of neurological diseases.
Collapse
Affiliation(s)
- Jens Weingarten
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | - Melanie Lassek
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | - Benjamin F Mueller
- Institute of Pharmaceutical Chemistry, Cluster of Excellence "Macromolecular Complexes", Goethe-University, Frankfurt am Main, Germany
| | - Marion Rohmer
- Institute of Pharmaceutical Chemistry, Cluster of Excellence "Macromolecular Complexes", Goethe-University, Frankfurt am Main, Germany
| | - Ilaria Lunger
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | | | - Simone Dudek
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | - Patricia Gogesch
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | - Michael Karas
- Institute of Pharmaceutical Chemistry, Cluster of Excellence "Macromolecular Complexes", Goethe-University, Frankfurt am Main, Germany
| | - Walter Volknandt
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany.
| |
Collapse
|
27
|
Zhang LN, Li JX, Hao L, Sun YJ, Xie YH, Wu SM, Liu L, Chen XL, Gao ZB. Crosstalk between dopamine receptors and the Na⁺/K⁺-ATPase (review). Mol Med Rep 2013; 8:1291-9. [PMID: 24065247 DOI: 10.3892/mmr.2013.1697] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/05/2013] [Indexed: 11/06/2022] Open
Abstract
Dopamine (DA) receptors, which belong to the G protein-coupled receptor family, are the target of ~50% of all modern medicinal drugs and constitute a large and diverse class of proteins whose primary function is to transduce extracellular stimuli into intracellular signals. Na+/K+-ATPase (NKA) is ubiquitous and crucial for the maintenance of intracellular ion homeostasis and excitability. Furthermore, it plays a critical role in diverse effects, including clinical cardiotonic and cardioprotective effects, ischemic preconditioning in the brain, natriuresis, lung edema clearance and other processes. NKA regulation is of physiological and pharmacological importance and has species- and tissue-specific variations. The activation of DA receptors regulates NKA expression/activity and trafficking in various tissues and cells, for example in the kidney, lung, intestine, brain, non-pigmented ciliary epithelium and the vascular bed. DA receptor-mediated regulation of NKA mediates a diverse range of cellular responses and includes endocytosis/exocytosis, phosphorylation/dephosphorylation of the α subunit of NKA and multiple signaling pathways, including phosphatidylinositol (PI)-phospholipase C/protein kinase (PK) C, cAMP/PKA, PI3K, adaptor protein 2, tyrosine phosphatase and mitogen-activated protein kinase/extracellular signal-regulated protein kinase. Furthermore, in brain and HEK293T cells, D1 and D2 receptors exist in a complex with NKA. Among D1 and D2 receptors and NKA, regulations are reciprocal, which leads to crosstalk between DA receptors and NKA. In the present study, the current understanding of signaling mechanisms responsible for the crosstalk between DA receptors and NKA, as well as with specific consequent functions, is reviewed.
Collapse
Affiliation(s)
- Li-Nan Zhang
- Department of Pharmacy, College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei 050018, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
de Juan-Sanz J, Núñez E, Villarejo-López L, Pérez-Hernández D, Rodriguez-Fraticelli AE, López-Corcuera B, Vázquez J, Aragón C. Na+/K+-ATPase is a new interacting partner for the neuronal glycine transporter GlyT2 that downregulates its expression in vitro and in vivo. J Neurosci 2013; 33:14269-81. [PMID: 23986260 PMCID: PMC6618510 DOI: 10.1523/jneurosci.1532-13.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/09/2013] [Accepted: 07/18/2013] [Indexed: 01/16/2023] Open
Abstract
The neuronal glycine transporter GlyT2 plays a fundamental role in the glycinergic neurotransmission by recycling the neurotransmitter to the presynaptic terminal. GlyT2 is the main supplier of glycine for vesicle refilling, a process that is absolutely necessary to preserve quantal glycine content in synaptic vesicles. Alterations in GlyT2 activity modify glycinergic neurotransmission and may underlie several neuromuscular disorders, such as hyperekplexia, myoclonus, dystonia, and epilepsy. Indeed, mutations in the gene encoding GlyT2 are the main presynaptic cause of hyperekplexia in humans and produce congenital muscular dystonia type 2 (CMD2) in Belgian Blue cattle. GlyT2 function is strictly coupled to the sodium electrochemical gradient actively generated by the Na+/K+-ATPase (NKA). GlyT2 cotransports 3Na+/Cl-/glycine generating large rises of Na+ inside the presynaptic terminal that must be efficiently reduced by the NKA to preserve Na+ homeostasis. In this work, we have used high-throughput mass spectrometry to identify proteins interacting with GlyT2 in the CNS. NKA was detected as a putative candidate and through reciprocal coimmunoprecipitations and immunocytochemistry analyses the association between GlyT2 and NKA was confirmed. NKA mainly interacts with the raft-associated active pool of GlyT2, and low and high levels of the specific NKA ligand ouabain modulate the endocytosis and total expression of GlyT2 in neurons. The ouabain-mediated downregulation of GlyT2 also occurs in vivo in two different systems: zebrafish embryos and adult rats, indicating that this NKA-mediated regulatory mechanism is evolutionarily conserved and may play a relevant role in the physiological control of inhibitory glycinergic neurotransmission.
Collapse
Affiliation(s)
- Jaime de Juan-Sanz
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Enrique Núñez
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Lucía Villarejo-López
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | | | - Alejo E. Rodriguez-Fraticelli
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Beatriz López-Corcuera
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain, and
| | - Carmen Aragón
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| |
Collapse
|
29
|
Abstract
Manganese is an important metal for human health, being absolutely necessary for development, metabolism, and the antioxidant system. Nevertheless, excessive exposure or intake may lead to a condition known as manganism, a neurodegenerative disorder that causes dopaminergic neuronal death and parkinsonian-like symptoms. Hence, Mn has a paradoxal effect in animals, a Janus-faced metal. Extensive work has been carried out to understand Mn-induced neurotoxicity and to find an effective treatment. This review focuses on the requirement for Mn in human health as well as the diseases associated with excessive exposure to this metal.
Collapse
Affiliation(s)
- Daiana Silva Avila
- Biochemistry Graduation Program, Universidade Federal do Pampa, Uruguaiana, Rio Grande do Sul, Brazil,
| | | | | |
Collapse
|
30
|
Reinhard L, Tidow H, Clausen MJ, Nissen P. Na(+),K (+)-ATPase as a docking station: protein-protein complexes of the Na(+),K (+)-ATPase. Cell Mol Life Sci 2013; 70:205-22. [PMID: 22695678 PMCID: PMC11113973 DOI: 10.1007/s00018-012-1039-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Revised: 05/13/2012] [Accepted: 05/23/2012] [Indexed: 12/13/2022]
Abstract
The Na(+),K(+)-ATPase, or sodium pump, is well known for its role in ion transport across the plasma membrane of animal cells. It carries out the transport of Na(+) ions out of the cell and of K(+) ions into the cell and thus maintains electrolyte and fluid balance. In addition to the fundamental ion-pumping function of the Na(+),K(+)-ATPase, recent work has suggested additional roles for Na(+),K(+)-ATPase in signal transduction and biomembrane structure. Several signaling pathways have been found to involve Na(+),K(+)-ATPase, which serves as a docking station for a fast-growing number of protein interaction partners. In this review, we focus on Na(+),K(+)-ATPase as a signal transducer, but also briefly discuss other Na(+),K(+)-ATPase protein-protein interactions, providing a comprehensive overview of the diverse signaling functions ascribed to this well-known enzyme.
Collapse
Affiliation(s)
- Linda Reinhard
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Henning Tidow
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Michael J. Clausen
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Poul Nissen
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| |
Collapse
|
31
|
Bai YL, Chu QJ, Li J, Chen YY, Li WJ, Zhang Q. A comparison of the effects of digoxin, ouabain and milrinone on naloxone-precipitated withdrawal syndrome in mice. Eur J Pharmacol 2012; 694:69-74. [DOI: 10.1016/j.ejphar.2012.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 08/14/2012] [Accepted: 08/18/2012] [Indexed: 10/27/2022]
|
32
|
|
33
|
Sibarov DA, Bolshakov AE, Abushik PA, Krivoi II, Antonov SM. Na+,K+-ATPase Functionally Interacts with the Plasma Membrane Na+,Ca2+ Exchanger to Prevent Ca2+ Overload and Neuronal Apoptosis in Excitotoxic Stress. J Pharmacol Exp Ther 2012; 343:596-607. [DOI: 10.1124/jpet.112.198341] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
34
|
Abstract
Dopamine is an important regulator of systemic blood pressure via multiple mechanisms. It affects fluid and electrolyte balance by its actions on renal hemodynamics and epithelial ion and water transport and by regulation of hormones and humoral agents. The kidney synthesizes dopamine from circulating or filtered L-DOPA independently from innervation. The major determinants of the renal tubular synthesis/release of dopamine are probably sodium intake and intracellular sodium. Dopamine exerts its actions via two families of cell surface receptors, D1-like receptors comprising D1R and D5R, and D2-like receptors comprising D2R, D3R, and D4R, and by interactions with other G protein-coupled receptors. D1-like receptors are linked to vasodilation, while the effect of D2-like receptors on the vasculature is variable and probably dependent upon the state of nerve activity. Dopamine secreted into the tubular lumen acts mainly via D1-like receptors in an autocrine/paracrine manner to regulate ion transport in the proximal and distal nephron. These effects are mediated mainly by tubular mechanisms and augmented by hemodynamic mechanisms. The natriuretic effect of D1-like receptors is caused by inhibition of ion transport in the apical and basolateral membranes. D2-like receptors participate in the inhibition of ion transport during conditions of euvolemia and moderate volume expansion. Dopamine also controls ion transport and blood pressure by regulating the production of reactive oxygen species and the inflammatory response. Essential hypertension is associated with abnormalities in dopamine production, receptor number, and/or posttranslational modification.
Collapse
Affiliation(s)
- Ines Armando
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| | - Van Anthony M. Villar
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| | - Pedro A. Jose
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| |
Collapse
|
35
|
Kirshenbaum GS, Saltzman K, Rose B, Petersen J, Vilsen B, Roder JC. Decreased neuronal Na+, K+ -ATPase activity in Atp1a3 heterozygous mice increases susceptibility to depression-like endophenotypes by chronic variable stress. GENES BRAIN AND BEHAVIOR 2011; 10:542-50. [PMID: 21418141 DOI: 10.1111/j.1601-183x.2011.00691.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Unipolar depression and bipolar depression are prevalent and debilitating diseases in need of effective novel treatments. It is becoming increasingly evident that depressive disorders manifest from a combination of inherited susceptibility genes and environmental stress. Genetic mutations resulting in decreased neuronal Na(+) ,K(+) -ATPase (sodium-potassium adenosine triphosphatase) activity may put individuals at risk for depression given that decreased Na(+) ,K(+) -ATPase activity is observed in depressive disorders and animal models of depression. Here, we show that Na(+) ,K(+) -ATPase α3 heterozygous mice (Atp1a3(+/-) ), with 15% reduced neuronal Na(+) ,K(+) -ATPase activity, are vulnerable to develop increased depression-like endophenotypes in a chronic variable stress (CVS) paradigm compared to wild-type littermates (Atp1a3(+/+) ). In Atp1a3(+/+) mice CVS did not decrease Na(+) ,K(+) -ATPase activity, however led to despair-like behavior in the tail suspension test (TST), anhedonia in a sucrose preference test and a minimal decrease in sociability, whereas in Atp1a3(+/-) mice CVS decreased neuronal Na(+) ,K(+) -ATPase activity to 33% of wild-type levels, induced despair-like behavior in the TST, anhedonia in a sucrose preference test, anxiety in the elevated plus maze, a memory deficit in a novel object recognition task and sociability deficits in a social interaction test. We found that a mutation that decreases neuronal Na(+) ,K(+) -ATPase activity interacts with stress to exacerbate depression. Furthermore, we observed an interesting correlation between Na(+) ,K(+) -ATPase activity and mood that may relate to both unipolar depression and bipolar disorder. Pharmaceuticals that increase Na(+) ,K(+) -ATPase activity or block endogenous Na(+) , K(+) -ATPase inhibition may provide effective treatment for depressive disorders and preclude depression in susceptible individuals.
Collapse
Affiliation(s)
- G S Kirshenbaum
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital and Institute of Medical Science, University of Toronto, 600 University Avenue, Toronto, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
36
|
Schaefer TL, Lingrel JB, Moseley AE, Vorhees CV, Williams MT. Targeted mutations in the Na,K-ATPase α 2 isoform confer ouabain resistance and result in abnormal behavior in mice. Synapse 2010; 65:520-31. [PMID: 20936682 DOI: 10.1002/syn.20870] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 09/09/2010] [Indexed: 01/09/2023]
Abstract
Sodium and potassium-activated adenosine triphosphatases (Na,K-ATPase) are ubiquitous, participate in osmotic balance and membrane potential, and are composed of α, β, and γ subunits. The α subunit is required for the catalytic and transport properties of the enzyme and contains binding sites for cations, ATP, and digitalis-like compounds including ouabain. There are four known α isoforms; three that are expressed in the CNS in a regional and cell-specific manner. The α2 isoform is most commonly found in astrocytes, pyramidal cells of the hippocampus in adults, and developmentally in several other neuronal types. Ouabain-like compounds are thought to be produced endogenously in mammals, bind the Na,K-ATPase, and function as a stress-related hormone, however, the impact of the Na,K-ATPase ouabain binding site on neurobehavioral function is largely unknown. To determine if the ouabain binding site of the α2 isoform plays a physiological role in CNS function, we examined knock-in mice in which the normally ouabain-sensitive α2 isoform was made resistant (α2(R/R) ) while still retaining basal Na,K-ATPase enzymatic function. Egocentric learning (Cincinnati water maze) was impaired in adult α2(R/R) mice compared to wild type (WT) mice. They also exhibited decreased locomotor activity in a novel environment and increased responsiveness to a challenge with an indirect sympathomimetic agonist (methamphetamine) relative to WT mice. The α2(R/R) mice also demonstrated a blunted acoustic startle reflex and a failure to habituate to repeated acoustic stimuli. The α2(R/R) mice showed no evidence of altered anxiety (elevated zero maze) nor were they impaired in spatial learning or memory in the Morris water maze and neither group could learn in a large Morris maze. These results suggest that the ouabain binding site is involved in specific types of learning and the modulation of dopamine-mediated locomotor behavior.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Neurology, Cincinnati Children's Research Foundation and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229-3039, USA
| | | | | | | | | |
Collapse
|
37
|
Heiny JA, Kravtsova VV, Mandel F, Radzyukevich TL, Benziane B, Prokofiev AV, Pedersen SE, Chibalin AV, Krivoi II. The nicotinic acetylcholine receptor and the Na,K-ATPase alpha2 isoform interact to regulate membrane electrogenesis in skeletal muscle. J Biol Chem 2010; 285:28614-26. [PMID: 20595385 DOI: 10.1074/jbc.m110.150961] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The nicotinic acetylcholine receptor (nAChR) and the Na,K-ATPase functionally interact in skeletal muscle (Krivoi, I. I., Drabkina, T. M., Kravtsova, V. V., Vasiliev, A. N., Eaton, M. J., Skatchkov, S. N., and Mandel, F. (2006) Pflugers Arch. 452, 756-765; Krivoi, I., Vasiliev, A., Kravtsova, V., Dobretsov, M., and Mandel, F. (2003) Ann. N.Y. Acad. Sci. 986, 639-641). In this interaction, the specific binding of nanomolar concentrations of nicotinic agonists to the nAChR stimulates electrogenic transport by the Na,K-ATPase alpha2 isozyme, causing membrane hyperpolarization. This study examines the molecular nature and membrane localization of this interaction. Stimulation of Na,K-ATPase activity by the nAChR does not require ion flow through open nAChRs. It can be induced by nAChR desensitization alone, in the absence of nicotinic agonist, and saturates when the nAChR is fully desensitized. It is enhanced by noncompetitive blockers of the nAChR (proadifen, QX-222), which promote non-conducting or desensitized states; and retarded by tetracaine, which stabilizes the resting nAChR conformation. The interaction operates at the neuromuscular junction as well as on extrajunctional sarcolemma. The Na,K-ATPase alpha2 isozyme is enriched at the postsynaptic neuromuscular junction and co-localizes with nAChRs. The nAChR and Na,K-ATPase alpha subunits specifically coimmunoprecipitate with each other, phospholemman, and caveolin-3. In a purified membrane preparation from Torpedo californica enriched in nAChRs and the Na,K-ATPase, a ouabain-induced conformational change of the Na,K-ATPase enhances a conformational transition of the nAChR to a desensitized state. These results suggest a mechanism by which the nAChR in a desensitized state with high apparent affinity for agonist interacts with the Na,K-ATPase to stimulate active transport. The interaction utilizes a membrane-delimited complex involving protein-protein interactions, either directly or through additional protein partners. This interaction is expected to enhance neuromuscular transmission and muscle excitation.
Collapse
Affiliation(s)
- Judith A Heiny
- Department of General Physiology, St. Petersburg State University, St. Petersburg 199034, Russia
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Rex EB, Rankin ML, Yang Y, Lu Q, Gerfen CR, Jose PA, Sibley DR. Identification of RanBP 9/10 as interacting partners for protein kinase C (PKC) gamma/delta and the D1 dopamine receptor: regulation of PKC-mediated receptor phosphorylation. Mol Pharmacol 2010; 78:69-80. [PMID: 20395553 DOI: 10.1124/mol.110.063727] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We reported previously that ethanol treatment regulates D(1) receptor phosphorylation and signaling in a protein kinase C (PKC) delta- and PKCgamma-dependent fashion by a mechanism that may involve PKC isozyme-specific interacting proteins. Using a PKC isozyme-specific coimmunoprecipitation approach coupled to mass spectrometry, we report the identification of RanBP9 and RanBP10 as novel interacting proteins for both PKCgamma and PKCdelta. Both RanBP9 and RanBP10 were found to specifically coimmunoprecipitate with both PKCgamma and PKCdelta; however, this association did not seem to mediate the ethanol regulation of the PKCs. It is noteworthy that the D(1) receptor was also found to specifically coimmunoprecipitate with RanBP9/10 from human embryonic kidney (HEK) 293T cells and with endogenous RanBP9 from rat kidney. RanBP9 and RanBP10 were also found to colocalize at the cellular level with the D(1) receptor in both kidney and brain tissue. Although overexpression of RanBP9 or RanBP10 in HEK293T cells did not seem to alter the kinase activities of either PKCdelta or PKCgamma, both RanBP proteins regulated D(1) receptor phosphorylation, signaling, and, in the case of RanBP9, expression. Specifically, overexpression of either RanBP9 or RanBP10 enhanced basal D(1) receptor phosphorylation, which was associated with attenuation of D(1) receptor-stimulated cAMP accumulation. Moreover, treatment of cells with select PKC inhibitors blocked the RanBP9/10-dependent increase in basal receptor phosphorylation, suggesting that phosphorylation of the receptor by PKC is regulated by RanBP9/10. These data support the idea that RanBP9 and RanBP10 may function as signaling integrators and dictate the efficient regulation of D(1) receptor signaling by PKCdelta and PKCgamma.
Collapse
Affiliation(s)
- Elizabeth B Rex
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Harrington MG, Fonteh AN, Arakaki X, Cowan RP, Ecke LE, Foster H, Hühmer AF, Biringer RG. Capillary endothelial Na(+), K(+), ATPase transporter homeostasis and a new theory for migraine pathophysiology. Headache 2010; 50:459-78. [PMID: 19845787 PMCID: PMC8020446 DOI: 10.1111/j.1526-4610.2009.01551.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Cerebrospinal fluid sodium concentration ([Na(+)](csf)) increases during migraine, but the cause of the increase is not known. OBJECTIVE Analyze biochemical pathways that influence [Na(+)](csf) to identify mechanisms that are consistent with migraine. METHOD We reviewed sodium physiology and biochemistry publications for links to migraine and pain. RESULTS Increased capillary endothelial cell (CEC) Na(+), K(+), -ATPase transporter (NKAT) activity is probably the primary cause of increased [Na(+)](csf). Physiological fluctuations of all NKAT regulators in blood, many known to be involved in migraine, are monitored by receptors on the luminal wall of brain CECs; signals are then transduced to their abluminal NKATs that alter brain extracellular sodium ([Na(+)](e)) and potassium ([K(+)](e)). CONCLUSIONS We propose a theoretical mechanism for aura and migraine when NKAT activity shifts outside normal limits: (1) CEC NKAT activity below a lower limit increases [K(+)](e), facilitates cortical spreading depression, and causes aura; (2) CEC NKAT activity above an upper limit elevates [Na(+)](e), increases neuronal excitability, and causes migraine; (3) migraine-without-aura may arise from CEC NKAT over-activity without requiring a prior decrease in activity and its consequent spreading depression; (4) migraine triggers disturb, and treatments improve, CEC NKAT homeostasis; (5) CEC NKAT-induced regulation of neural and vasomotor excitability coordinates vascular and neuronal activities, and includes occasional pathology from CEC NKAT-induced apoptosis or cerebral infarction.
Collapse
Affiliation(s)
- Michael G Harrington
- Huntington Medical Research Institutes - Molecular Neurology, Pasadena, CA 91101, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Jose PA, Soares-da-Silva P, Eisner GM, Felder RA. Dopamine and G protein-coupled receptor kinase 4 in the kidney: role in blood pressure regulation. Biochim Biophys Acta Mol Basis Dis 2010; 1802:1259-67. [PMID: 20153824 DOI: 10.1016/j.bbadis.2010.02.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Revised: 02/05/2010] [Accepted: 02/07/2010] [Indexed: 12/11/2022]
Abstract
Complex interactions between genes and environment result in a sodium-induced elevation in blood pressure (salt sensitivity) and/or hypertension that lead to significant morbidity and mortality affecting up to 25% of the middle-aged adult population worldwide. Determining the etiology of genetic and/or environmentally-induced high blood pressure has been difficult because of the many interacting systems involved. Two main pathways have been implicated as principal determinants of blood pressure since they are located in the kidney (the key organ responsible for blood pressure regulation), and have profound effects on sodium balance: the dopaminergic and renin-angiotensin systems. These systems counteract or modulate each other, in concert with a host of intracellular second messenger pathways to regulate sodium and water balance. In particular, the G protein-coupled receptor kinase type 4 (GRK4) appears to play a key role in regulating dopaminergic-mediated natriuresis. Constitutively activated GRK4 gene variants (R65L, A142V, and A486V), by themselves or by their interaction with other genes involved in blood pressure regulation, are associated with essential hypertension and/or salt-sensitive hypertension in several ethnic groups. GRK4γ 142Vtransgenic mice are hypertensive on normal salt intake while GRK4γ 486V transgenic mice develop hypertension only with an increase in salt intake. GRK4 gene variants have been shown to hyperphosphorylate, desensitize, and internalize two members of the dopamine receptor family, the D(1) (D(1)R) and D(3) (D(3)R) dopamine receptors, but also increase the expression of a key receptor of the renin-angiotensin system, the angiotensin type 1 receptor (AT(1)R). Knowledge of the numerous blood pressure regulatory pathways involving angiotensin and dopamine may provide new therapeutic approaches to the pharmacological regulation of sodium excretion and ultimately blood pressure control.
Collapse
Affiliation(s)
- Pedro A Jose
- Children's National Medical Center, George Washington University for the Health Sciences, Washington, DC, USA.
| | | | | | | |
Collapse
|