1
|
Dos Santos GRO, Cararo-Lopes MM, Possebom IR, de Sá Lima L, Scavone C, Kawamoto EM. Sex-dependent changes in AMPAR expression and Na, K-ATPase activity in the cerebellum and hippocampus of α-Klotho-Hypomorphic mice. Neuropharmacology 2024; 258:110097. [PMID: 39094831 DOI: 10.1016/j.neuropharm.2024.110097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Aging is characterized by a functional decline in several physiological systems. α-Klotho-hypomorphic mice (Kl-/-) exhibit accelerated aging and cognitive decline. We evaluated whether male and female α-Klotho-hypomorphic mice show changes in the expression of synaptic proteins, N-methyl-d-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, postsynaptic density protein 95 (PSD-95), synaptophysin and synapsin, and the activity of Na+, K+-ATPase (NaK) isoforms in the cerebellum and hippocampus. In this study, we demonstrated that in the cerebellum, Kl-/- male mice have reduced expression of GluA1 (AMPA) compared to wild-type (Kl+/+) males and Kl-/- females. Also, Kl-/- male and female mice show reduced ɑ2/ɑ3-NaK and Mg2+-ATPase activities in the cerebellum, respectively, and sex-based differences in NaK and Mg2+-ATPase activities in both the regions. Our findings suggest that α-Klotho could influence the expression of AMPAR and the activity of NaK isoforms in the cerebellum in a sex-dependent manner, and these changes may contribute, in part, to cognitive decline.
Collapse
Affiliation(s)
| | - Marina Minto Cararo-Lopes
- Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, 79070-900, Campo Grande, MS, Brazil
| | - Isabela Ribeiro Possebom
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Larissa de Sá Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Elisa Mitiko Kawamoto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil.
| |
Collapse
|
2
|
Wigstrom TP, Roytman S, Bohnen JLB, Paalanen RR, Griggs AM, Vangel R, Barr J, Albin R, Kanel P, Bohnen NI. Impaired mitochondrial function in bipolar disorder and alcohol use disorder: a case study using 18F-BCPP-EF PET imaging of mitochondrial Complex I. PSYCHORADIOLOGY 2024; 4:kkae014. [PMID: 39399447 PMCID: PMC11467810 DOI: 10.1093/psyrad/kkae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/17/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024]
Abstract
Background With bipolar disorder (BD) having a lifetime prevalence of 4.4% and a significant portion of patients being chronically burdened by symptoms, there has been an increased focus on uncovering new targets for intervention in BD. One area that has shown early promise is the mitochondrial hypothesis. However, at the time of publication no studies have utilized positron emission tomography (PET) imaging to assess mitochondrial function in the setting of BD. Case Presentation Our participant is a 58 year-old male with a past medical history notable for alcohol use disorder and BD (unspecified type) who underwent PET imaging with the mitochondrial complex I PET ligand 18F-BCPP-EF. The resulting images demonstrated significant overlap between areas of dysfunction identified with the 18F-BCPP-EF PET ligand and prior functional magnetic resonance imaging (MRI) techniques in the setting of BD. That overlap was seen in both affective and cognitive circuits, with mitochondrial dysfunction in the fronto-limbic, ventral affective, and dorsal cognitive circuits showing particularly significant differences. Conclusions Despite mounting evidence implicating mitochondria in BD, this study represents the first PET imaging study to investigate this mechanistic connection. There were key limitations in the form of comorbid alcohol use disorder, limited statistical power inherent to a case study, no sex matched controls, and the absence of a comprehensive psychiatric history. However, even with these limitations in mind, the significant overlap between dysfunction previously demonstrated on functional MRI and this imaging provides compelling preliminary evidence that strengthens the mechanistic link between mitochondrial dysfunction and BD.
Collapse
Affiliation(s)
- Travis P Wigstrom
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stiven Roytman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffrey L B Bohnen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rebecca R Paalanen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexis M Griggs
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Robert Vangel
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jaimie Barr
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Roger Albin
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Prabesh Kanel
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nicolaas I Bohnen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Neurology Service and GRECC, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI 48109, USA
- Parkinson's Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Nag S, Jia K, Arakawa R, Datta P, Scott D, Shaffer C, Moein MM, Hutchison M, Kaliszczak M, Halldin C. Synthesis of [ 11C]BIIB104, an α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic-Acid-Positive Allosteric Modulator, and Evaluation of the Bio-Distribution in Non-Human Primate Brains Using Positron Emission Tomography. Molecules 2024; 29:427. [PMID: 38257338 PMCID: PMC10818776 DOI: 10.3390/molecules29020427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
The aim of this study was to measure the brain penetrance and kinetics of BIIB104, a first-in-class AMPA receptor potentiator developed for cognitive impairment associated with schizophrenia. It was recently halted in phase 2 clinical development, and there are a lack of tools to directly measure AMPA receptor engagement. To achieve this, the drug candidate was radiolabeled with carbon-11, and its brain penetrance and kinetics were measured in non-human primates via dynamic PET scans. Radiolabeling was achieved through a three-step nucleophilic [11C]cyanation reaction in one pot, resulting in the high radioactivity and radiochemical purity (>99%) of [11C]BIIB104. The study found that [11C]BIIB104 entered the non-human primate brains at 4-5% ID at peak, with a homogeneous distribution. However, a mild regional heterogeneity was observed in the thalamus. The lack of conclusive evidence for a change in regional values after BIIB104 dosing suggests that any specific binding component of BIIB104 is negligible compared to the free and non-specific components in the living brain. Overall, the study demonstrated high brain uptake with minor variability in [11C]BIIB104 distribution across various brain regions, its kinetics were consistent with those of passive diffusion, and the dominating components were the free concentration and non-specific binding. This information is valuable for understanding the potential effects and mechanisms of BIIB104 in the brain.
Collapse
Affiliation(s)
- Sangram Nag
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 171 64 Stockholm, Sweden (C.H.)
| | - Kevin Jia
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 171 64 Stockholm, Sweden (C.H.)
| | - Ryosuke Arakawa
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 171 64 Stockholm, Sweden (C.H.)
| | - Prodip Datta
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 171 64 Stockholm, Sweden (C.H.)
| | - Daniel Scott
- BIOGEN MA Inc., 225 Binney St., Cambridge, MA 02142, USA (C.S.); (M.H.)
| | | | - Mohammad Mahdi Moein
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 171 64 Stockholm, Sweden (C.H.)
| | - Matthew Hutchison
- BIOGEN MA Inc., 225 Binney St., Cambridge, MA 02142, USA (C.S.); (M.H.)
| | - Maciej Kaliszczak
- BIOGEN MA Inc., 225 Binney St., Cambridge, MA 02142, USA (C.S.); (M.H.)
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 171 64 Stockholm, Sweden (C.H.)
| |
Collapse
|
4
|
Warming H, Deinhardt K, Garland P, More J, Bulters D, Galea I, Vargas-Caballero M. Functional effects of haemoglobin can be rescued by haptoglobin in an in vitro model of subarachnoid haemorrhage. J Neurochem 2023; 167:90-103. [PMID: 37702203 DOI: 10.1111/jnc.15936] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
During subarachnoid haemorrhage, a blood clot forms in the subarachnoid space releasing extracellular haemoglobin (Hb), which causes oxidative damage and cell death in surrounding tissues. High rates of disability and cognitive decline in SAH survivors are attributed to loss of neurons and functional connections during secondary brain injury. Haptoglobin sequesters Hb for clearance, but this scavenging system is overwhelmed after a haemorrhage. Whilst exogenous haptoglobin application can attenuate cytotoxicity of Hb in vitro and in vivo, the functional effects of sub-lethal Hb concentrations on surviving neurons and whether cellular function can be protected with haptoglobin treatment remain unclear. Here we use cultured neurons to investigate neuronal health and function across a range of Hb concentrations to establish the thresholds for cellular damage and investigate synaptic function. Hb impairs ATP concentrations and cytoskeletal structure. At clinically relevant but sub-lethal Hb concentrations, we find that synaptic AMPAR-driven currents are reduced, accompanied by a reduction in GluA1 subunit expression. Haptoglobin co-application can prevent these deficits by scavenging free Hb to reduce it to sub-threshold concentrations and does not need to be present at stoichiometric amounts to achieve efficacy. Haptoglobin itself does not impair measures of neuronal health and function at any concentration tested. Our data highlight a role for Hb in modifying synaptic function in surviving neurons, which may link to impaired cognition or plasticity after SAH and support the development of haptoglobin as a therapy for subarachnoid haemorrhage.
Collapse
Affiliation(s)
- Hannah Warming
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Katrin Deinhardt
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | | | - John More
- Bio Products Laboratory Limited, Elstree, UK
| | - Diederik Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Mariana Vargas-Caballero
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
5
|
Bohnen JLB, Wigstrom TP, Griggs AM, Roytman S, Paalanen RR, Andrews HA, Bohnen NI, Franklin JJH, McInnis MG. Ketogenic-Mimicking Diet as a Therapeutic Modality for Bipolar Disorder: Biomechanistic Rationale and Protocol for a Pilot Clinical Trial. Nutrients 2023; 15:3068. [PMID: 37447394 PMCID: PMC10346691 DOI: 10.3390/nu15133068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
There is growing interest in the investigation of ketogenic diets as a potential therapy for bipolar disorder. The overlapping pharmacotherapies utilized for both bipolar disorder and seizures suggest that a mechanistic overlap may exist between these conditions, with fasting and the ketogenic diet representing the most time-proven therapies for seizure control. Recently, preliminary evidence has begun to emerge supporting a potential role for ketogenic diets in treating bipolar disorder. Notably, some patients may struggle to initiate a strict diet in the midst of a mood episode or significant life stressors. The key question addressed by this pilot clinical trial protocol is if benefits can be achieved with a less restrictive diet, as this would allow such an intervention to be accessible for more patients. Recent development of so-called ketone esters, that once ingested is converted to natural ketone bodies, combined with low glycemic index dietary changes has the potential to mimic two foundational components of therapeutic ketosis: high levels of ketones and minimal spiking of glucose/insulin. This pilot clinical trial protocol thus aims to investigate the effect of a 'ketogenic-mimicking diet' (combining supplementation of ketone esters with a low glycemic index dietary intervention) on neural network stability, mood, and biomarker outcomes in the setting of bipolar disorder. Positive findings obtained via this pilot clinical trial protocol may support future target engagement studies of ketogenic-mimicking diets or related ketogenic interventions. A lack of positive findings, in contrast, may justify a focus on more strict dietary interventions for future research.
Collapse
Affiliation(s)
| | | | - Alexis M. Griggs
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Neurology Service and GRECC, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Stiven Roytman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | - Nicolaas I. Bohnen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Neurology Service and GRECC, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Morris K. Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, MI 48109, USA
- Parkinson’s Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Melvin G. McInnis
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, USA
- Heinz C. Prechter Bipolar Research Program, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
The role of post-translational modifications in synaptic AMPA receptor activity. Biochem Soc Trans 2023; 51:315-330. [PMID: 36629507 DOI: 10.1042/bst20220827] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023]
Abstract
AMPA-type receptors for the neurotransmitter glutamate are very dynamic entities, and changes in their synaptic abundance underlie different forms of synaptic plasticity, including long-term synaptic potentiation (LTP), long-term depression (LTD) and homeostatic scaling. The different AMPA receptor subunits (GluA1-GluA4) share a common modular structure and membrane topology, and their intracellular C-terminus tail is responsible for the interaction with intracellular proteins important in receptor trafficking. The latter sequence differs between subunits and contains most sites for post-translational modifications of the receptors, including phosphorylation, O-GlcNAcylation, ubiquitination, acetylation, palmitoylation and nitrosylation, which affect differentially the various subunits. Considering that each single subunit may undergo modifications in multiple sites, and that AMPA receptors may be formed by the assembly of different subunits, this creates multiple layers of regulation of the receptors with impact in synaptic function and plasticity. This review discusses the diversity of mechanisms involved in the post-translational modification of AMPA receptor subunits, and their impact on the subcellular distribution and synaptic activity of the receptors.
Collapse
|
7
|
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
|
8
|
Kinoshita PF, Orellana AM, Andreotti DZ, de Souza GA, de Mello NP, de Sá Lima L, Kawamoto EM, Scavone C. Consequences of the Lack of TNFR1 in Ouabain Response in the Hippocampus of C57BL/6J Mice. Biomedicines 2022; 10:biomedicines10112937. [PMID: 36428505 PMCID: PMC9688030 DOI: 10.3390/biomedicines10112937] [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: 09/09/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Ouabain is a cardiac glycoside that has a protective effect against neuroinflammation at low doses through Na+/K+-ATPase signaling and that can activate tumor necrosis factor (TNF) in the brain. TNF plays an essential role in neuroinflammation and regulates glutamate receptors by acting on two different receptors (tumor necrosis factor receptor 1 [TNFR1] and TNFR2) that have distinct functions and expression. The activation of constitutively and ubiquitously expressed TNFR1 leads to the expression of pro-inflammatory cytokines. Thus, this study aimed to elucidate the effects of ouabain in a TNFR1 knockout (KO) mouse model. Interestingly, the hippocampus of TNFR1 KO mice showed a basal increase in both TNFR2 membrane expression and brain-derived neurotrophic factor (BDNF) release, suggesting a compensatory mechanism. Moreover, ouabain activated TNF-α-converting enzyme/a disintegrin and metalloprotease 17 (TACE/ADAM17), decreased N-methyl-D-aspartate (NMDA) receptor subunit 2A (NR2A) expression, and induced anxiety-like behavior in both genotype animals, independent of the presence of TNFR1. However, ouabain induced an increase in interleukin (IL)-1β in the hippocampus, a decrease in IL-6 in serum, and an increase in NMDA receptor subunit 1 (NR1) only in wild-type (WT) mice, indicating that TNFR1 or TNFR2 expression may be important for some effects of ouabain. Collectively, our results indicate a connection between ouabain signaling and TNFR1, with the effect of ouabain partially dependent on TNFR1.
Collapse
Affiliation(s)
- Paula Fernanda Kinoshita
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | - Ana Maria Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | - Diana Zukas Andreotti
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | - Giovanna Araujo de Souza
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | - Natalia Prudente de Mello
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil
- Correspondence:
| |
Collapse
|
9
|
Historical perspective and progress on protein ubiquitination at glutamatergic synapses. Neuropharmacology 2021; 196:108690. [PMID: 34197891 DOI: 10.1016/j.neuropharm.2021.108690] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/07/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
Transcription-translation coupling leads to the production of proteins that are key for controlling essential neuronal processes that include neuronal development and changes in synaptic strength. Although these events have been a prevailing theme in neuroscience, the regulation of proteins via posttranslational signaling pathways are equally relevant for these neuronal processes. Ubiquitin is one type of posttranslational modification that covalently attaches to its targets/substrates. Ubiquitination of proteins play a key role in multiple signaling pathways, the predominant being removal of its substrates by a large molecular machine called the proteasome. Here, I review 40 years of progress on ubiquitination in the nervous system at glutamatergic synapses focusing on axon pathfinding, synapse formation, presynaptic release, dendritic spine formation, and regulation of postsynaptic glutamate receptors. Finally, I elucidate emerging themes in ubiquitin biology that may challenge our current understanding of ubiquitin signaling in the nervous system.
Collapse
|
10
|
Valvassori SS, Dal-Pont GC, Varela RB, Resende WR, Gava FF, Mina FG, Budni J, Quevedo J. Ouabain induces memory impairment and alter the BDNF signaling pathway in an animal model of bipolar disorder: Cognitive and neurochemical alterations in BD model. J Affect Disord 2021; 282:1195-1202. [PMID: 33601696 DOI: 10.1016/j.jad.2020.12.190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/16/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The present study aims to evaluate the effects of ouabain on memory and neurotrophic parameters in the brains of rats. METHODS Wistar rats received an intracerebroventricular (ICV) injection of ouabain or artificial cerebrospinal fluid (aCSF). Seven and 14 days after ICV administration, the animals were subjected to the open-field and splash tests. Furthermore, the pro-BDNF, BDNF, TrkB, and CREB were assessed in the frontal cortex and hippocampus of the rats, in both seven and 14 days after ICV injection. The memory of the animals was tested by novel object recognition test (NOR) and inhibitory avoidance task (IA), only 14 days after ICV administration. RESULTS Ouabain increased locomotion and exploration in the animals seven days after its administration; however, 14 days after ICV, these behavioral parameters return to the basal level. Seven days after ouabain administration increased grooming behavior in the splash test; on the other hand, seven days after ouabain injection decreased the grooming behavior, which is considered an anhedonic response. Besides, ouabain decreased recognition index in the NOR and decreased aversive memory in the IA, when compared to the control group. The levels of pro-BDNF and BDNF decreased in the frontal cortex seven days after ouabain; but its receptor (TrkB) and CREB decreased seven and 14 days after ouabain, in both cerebral structures evaluated. CONCLUSION Ouabain-induced animal model of BD is an excellent model to assess memory alteration, observed in bipolar patients. Besides, the memory impairment induced by ouabain seems to be related to BDNF signaling pathway alterations.
Collapse
Affiliation(s)
- Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Gustavo C Dal-Pont
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Roger B Varela
- Queensland Brain Institute, The Universty of Queensland, St Lucia, QLD 4072, Australia
| | - Wilson R Resende
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Fernanda F Gava
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Franciele G Mina
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Josiane Budni
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, United States; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States
| |
Collapse
|
11
|
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
|
12
|
Remodeling without destruction: non-proteolytic ubiquitin chains in neural function and brain disorders. Mol Psychiatry 2021; 26:247-264. [PMID: 32709994 PMCID: PMC9229342 DOI: 10.1038/s41380-020-0849-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 12/31/2022]
Abstract
Ubiquitination is a fundamental posttranslational protein modification that regulates diverse biological processes, including those in the CNS. Several topologically and functionally distinct polyubiquitin chains can be assembled on protein substrates, modifying their fates. The classical and most prevalent polyubiquitin chains are those that tag a substrate to the proteasome for degradation, which has been established as a major mechanism driving neural circuit deconstruction and remodeling. In contrast, proteasome-independent non-proteolytic polyubiquitin chains regulate protein scaffolding, signaling complex formation, and kinase activation, and play essential roles in an array of signal transduction processes. Despite being a cornerstone in immune signaling and abundant in the mammalian brain, these non-proteolytic chains are underappreciated in neurons and synapses in the brain. Emerging studies have begun to generate exciting insights about some fundamental roles played by these non-degradative chains in neuronal function and plasticity. In addition, their roles in a number of brain diseases are being recognized. In this article, we discuss recent advances on these nonconventional ubiquitin chains in neural development, function, plasticity, and related pathologies.
Collapse
|
13
|
UPF2 leads to degradation of dendritically targeted mRNAs to regulate synaptic plasticity and cognitive function. Mol Psychiatry 2020; 25:3360-3379. [PMID: 31636381 PMCID: PMC7566522 DOI: 10.1038/s41380-019-0547-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 08/13/2019] [Accepted: 08/19/2019] [Indexed: 12/21/2022]
Abstract
Synaptic plasticity requires a tight control of mRNA levels in dendrites. RNA translation and degradation pathways have been recently linked to neurodevelopmental and neuropsychiatric diseases, suggesting a role for RNA regulation in synaptic plasticity and cognition. While the local translation of specific mRNAs has been implicated in synaptic plasticity, the tightly controlled mechanisms that regulate local quantity of specific mRNAs remain poorly understood. Despite being the only RNA regulatory pathway that is associated with multiple mental illnesses, the nonsense-mediated mRNA decay (NMD) pathway presents an unexplored regulatory mechanism for synaptic function and plasticity. Here, we show that neuron-specific disruption of UPF2, an NMD component, in adulthood attenuates learning, memory, spine density, synaptic plasticity (L-LTP), and potentiates perseverative/repetitive behavior in mice. We report that the NMD pathway operates within dendrites to regulate Glutamate Receptor 1 (GLUR1) surface levels. Specifically, UPF2 modulates the internalization of GLUR1 and promotes its local synthesis in dendrites. We identified neuronal Prkag3 mRNA as a mechanistic substrate for NMD that contributes to the UPF2-mediated regulation of GLUR1 by limiting total GLUR1 levels. These data establish that UPF2 regulates synaptic plasticity, cognition, and local protein synthesis in dendrites, providing fundamental insight into the neuron-specific function of NMD within the brain.
Collapse
|
14
|
O'Connor M, Shentu YP, Wang G, Hu WT, Xu ZD, Wang XC, Liu R, Man HY. Acetylation of AMPA Receptors Regulates Receptor Trafficking and Rescues Memory Deficits in Alzheimer's Disease. iScience 2020; 23:101465. [PMID: 32861999 PMCID: PMC7476873 DOI: 10.1016/j.isci.2020.101465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/21/2020] [Accepted: 08/13/2020] [Indexed: 12/26/2022] Open
Abstract
In Alzheimer's disease (AD), decreases in the amount and synaptic localization of AMPA receptors (AMPARs) result in weakened synaptic activity and dysfunction in synaptic plasticity, leading to impairments in cognitive functions. We have previously found that AMPARs are subject to lysine acetylation, resulting in higher AMPAR stability and protein accumulation. Here we report that AMPAR acetylation was significantly reduced in AD and neurons with Aβ incubation. We identified p300 as the acetyltransferase responsible for AMPAR acetylation and found that enhancing GluA1 acetylation ameliorated Aβ-induced reductions in total and cell-surface AMPARs. Importantly, expression of acetylation mimetic GluA1 (GluA1-4KQ) in APP/PS1 mice rescued impairments in synaptic plasticity and memory. These findings indicate that Aβ-induced reduction in AMPAR acetylation and stability contributes to synaptopathy and memory deficiency in AD, suggesting that AMPAR acetylation may be an effective molecular target for AD therapeutics.
Collapse
Affiliation(s)
- Margaret O'Connor
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Yang-Ping Shentu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guan Wang
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Wen-Ting Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhen-Dong Xu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, 72 East Concord St., L-603, Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Avenue, Boston, MA, USA
| |
Collapse
|
15
|
Felix L, Delekate A, Petzold GC, Rose CR. Sodium Fluctuations in Astroglia and Their Potential Impact on Astrocyte Function. Front Physiol 2020; 11:871. [PMID: 32903427 PMCID: PMC7435049 DOI: 10.3389/fphys.2020.00871] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Astrocytes are the main cell type responsible for the regulation of brain homeostasis, including the maintenance of ion gradients and neurotransmitter clearance. These processes are tightly coupled to changes in the intracellular sodium (Na+) concentration. While activation of the sodium-potassium-ATPase (NKA) in response to an elevation of extracellular K+ may decrease intracellular Na+, the cotransport of transmitters, such as glutamate, together with Na+ results in an increase in astrocytic Na+. This increase in intracellular Na+ can modulate, for instance, metabolic downstream pathways. Thereby, astrocytes are capable to react on a fast time scale to surrounding neuronal activity via intracellular Na+ fluctuations and adjust energy production to the demand of their environment. Beside the well-documented conventional roles of Na+ signaling mainly mediated through changes in its electrochemical gradient, several recent studies have identified more atypical roles for Na+, including protein interactions leading to changes in their biochemical activity or Na+-dependent regulation of gene expression. In this review, we will address both the conventional as well as the atypical functions of astrocytic Na+ signaling, presenting the role of transporters and channels involved and their implications for physiological processes in the central nervous system (CNS). We will also discuss how these important functions are affected under pathological conditions, including stroke and migraine. We postulate that Na+ is an essential player not only in the maintenance of homeostatic processes but also as a messenger for the fast communication between neurons and astrocytes, adjusting the functional properties of various cellular interaction partners to the needs of the surrounding network.
Collapse
Affiliation(s)
- Lisa Felix
- Institute of Neurobiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Andrea Delekate
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Division of Vascular Neurology, Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
16
|
Melone M, Ciriachi C, Pietrobon D, Conti F. Heterogeneity of Astrocytic and Neuronal GLT-1 at Cortical Excitatory Synapses, as Revealed by its Colocalization With Na+/K+-ATPase α Isoforms. Cereb Cortex 2020; 29:3331-3350. [PMID: 30260367 DOI: 10.1093/cercor/bhy203] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 12/29/2022] Open
Abstract
GLT-1, the major glutamate transporter, is expressed at perisynaptic astrocytic processes (PAP) and axon terminals (AxT). GLT-1 is coupled to Na+/K+-ATPase (NKA) α1-3 isoforms, whose subcellular distribution and spatial organization in relationship to GLT-1 are largely unknown. Using several microscopy techniques, we showed that at excitatory synapses α1 and α3 are exclusively neuronal (mainly in dendrites and in some AxT), while α2 is predominantly astrocytic. GLT-1 displayed a differential colocalization with α1-3. GLT-1/α2 and GLT-1/α3 colocalization was higher in GLT-1 positive puncta partially (for GLT-1/α2) or almost totally (for GLT-1/α3) overlapping with VGLUT1 positive terminals than in nonoverlapping ones. GLT-1 colocalized with α2 at PAP, and with α1 and α3 at AxT. GLT-1 and α2 gold particles were ∼1.5-2 times closer than GLT-1/α1 and GLT-1/α3 particles. GLT-1/α2 complexes (edge to edge interdistance of gold particles ≤50 nm) concentrated at the perisynaptic region of PAP membranes, whereas neuronal GLT-1/α1 and GLT-1/α3 complexes were fewer and more uniformly distributed in AxT. These data unveil different composition of GLT-1 and α subunits complexes in the glial and neuronal domains of excitatory synapses. The spatial organization of GLT-1/α1-3 complexes suggests that GLT-1/NKA interaction is more efficient in astrocytes than in neurons, further supporting the dominant role of astrocytic GLT-1 in glutamate homeostasis.
Collapse
Affiliation(s)
- Marcello Melone
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Chiara Ciriachi
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Daniela Pietrobon
- Department of Biomedical Sciences, University of Padova, and CNR Institute of Neuroscience, Padova, Italy
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.,Foundation for Molecular Medicine, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
17
|
Lazarov E, Hillebrand M, Schröder S, Ternka K, Hofhuis J, Ohlenbusch A, Barrantes-Freer A, Pardo LA, Fruergaard MU, Nissen P, Brockmann K, Gärtner J, Rosewich H. Comparative analysis of alternating hemiplegia of childhood and rapid-onset dystonia-parkinsonism ATP1A3 mutations reveals functional deficits, which do not correlate with disease severity. Neurobiol Dis 2020; 143:105012. [PMID: 32653672 DOI: 10.1016/j.nbd.2020.105012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/12/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Heterozygous mutations in the ATP1A3 gene, coding for an alpha subunit isoform (α3) of Na+/K+-ATPase, are the primary genetic cause for rapid-onset dystonia-parkinsonism (RDP) and alternating hemiplegia of childhood (AHC). Recently, cerebellar ataxia, areflexia, pes cavus, optic atrophy and sensorineural hearing loss (CAPOS), early infantile epileptic encephalopathy (EIEE), childhood rapid onset ataxia (CROA) and relapsing encephalopathy with rapid onset ataxia (RECA) extend the clinical spectrum of ATP1A3 related disorders. AHC and RDP demonstrate distinct clinical features, with AHC symptoms being generally more severe compared to RDP. Currently, it is largely unknown what determines the disease severity, and whether severity is linked to the degree of functional impairment of the α3 subunit. Here we compared the effect of twelve different RDP and AHC specific mutations on the expression and function of the α3 Na+/K+-ATPase in transfected HEK cells and oocytes. All studied mutations led to functional impairment of the pump, as reflected by lower survival rate and reduced pump current. No difference in the extent of impairment, nor in the expression level, was found between the two phenotypes, suggesting that these measures of pump dysfunction do not exclusively determine the disease severity.
Collapse
Affiliation(s)
- Elinor Lazarov
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| | - Merle Hillebrand
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| | - Simone Schröder
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| | - Katharina Ternka
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| | - Julia Hofhuis
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| | - Andreas Ohlenbusch
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| | | | - Luis A Pardo
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| | - Marlene U Fruergaard
- DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Dept. Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark.
| | - Poul Nissen
- DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Dept. Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark.
| | - Knut Brockmann
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| | - Jutta Gärtner
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| | - Hendrik Rosewich
- University Medical Center Göttingen, Georg August University, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, Germany.
| |
Collapse
|
18
|
Akkuratov EE, Westin L, Vazquez-Juarez E, de Marothy M, Melnikova AK, Blom H, Lindskog M, Brismar H, Aperia A. Ouabain Modulates the Functional Interaction Between Na,K-ATPase and NMDA Receptor. Mol Neurobiol 2020; 57:4018-4030. [PMID: 32651756 PMCID: PMC7467916 DOI: 10.1007/s12035-020-01984-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022]
Abstract
The N-methyl-D-aspartate (NMDA) receptor plays an essential role in glutamatergic transmission and synaptic plasticity and researchers are seeking for different modulators of NMDA receptor function. One possible mechanism for its regulation could be through adjacent membrane proteins. NMDA receptors coprecipitate with Na,K-ATPase, indicating a potential interaction of these two proteins. Ouabain, a mammalian cardiotonic steroid that specifically binds to Na,K-ATPase and affects its conformation, can protect from some toxic effects of NMDA receptor activation. Here we have examined whether NMDA receptor activity and downstream effects can be modulated by physiological ouabain concentrations. The spatial colocalization between NMDA receptors and the Na,K-ATPase catalytic subunits on dendrites of cultured rat hippocampal neurons was analyzed with super-resolution dSTORM microscopy. The functional interaction was analyzed with calcium imaging of single hippocampal neurons exposed to 10 μM NMDA in presence and absence of ouabain and by determination of the ouabain effect on NMDA receptor–dependent long-term potentiation. We show that NMDA receptors and the Na,K-ATPase catalytic subunits alpha1 and alpha3 exist in same protein complex and that ouabain in nanomolar concentration consistently reduces the calcium response to NMDA. Downregulation of the NMDA response is not associated with internalization of the receptor or with alterations in its state of Src phosphorylation. Ouabain in nanomolar concentration elicits a long-term potentiation response. Our findings suggest that ouabain binding to a fraction of Na,K-ATPase molecules that cluster with the NMDA receptors will, via a conformational effect on the NMDA receptors, cause moderate but consistent reduction of NMDA receptor response at synaptic activation.
Collapse
Affiliation(s)
- Evgeny E Akkuratov
- Science for Life Laboratory, Department of Applied Physics, Kungliga Tekniska Högskolan, Stockholm, Sweden
| | - Linda Westin
- Science for Life Laboratory, Department of Women's and Children's health, Karolinska Institutet, Stockholm, Sweden
| | - Erika Vazquez-Juarez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Minttu de Marothy
- Science for Life Laboratory, Department of Women's and Children's health, Karolinska Institutet, Stockholm, Sweden
| | - Aleksandra K Melnikova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia, 119234
| | - Hans Blom
- Science for Life Laboratory, Department of Applied Physics, Kungliga Tekniska Högskolan, Stockholm, Sweden
| | - Maria Lindskog
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Applied Physics, Kungliga Tekniska Högskolan, Stockholm, Sweden.
| | - Anita Aperia
- Science for Life Laboratory, Department of Women's and Children's health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
Reis AS, Paltian JJ, Domingues WB, Costa GP, Alves D, Giongo JL, Campos VF, Luchese C, Wilhelm EA. Pharmacological modulation of Na +, K +-ATPase as a potential target for OXA-induced neurotoxicity: Correlation between anxiety and cognitive decline and beneficial effects of 7-chloro-4-(phenylselanyl) quinoline. Brain Res Bull 2020; 162:282-290. [PMID: 32628972 DOI: 10.1016/j.brainresbull.2020.06.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/21/2020] [Accepted: 06/29/2020] [Indexed: 01/04/2023]
Abstract
Growing evidence demonstrates that Oxaliplatin (OXA) is commonly associated with neurotoxicity that leads to emotional and cognitive impairments. The aim of the present study was to evaluate the OXA and Na+, K+-ATPase interaction and to correlate anxious behavior and cognitive impairment induced by this chemotherapeutic in Swiss mice. Also, considering the pharmacological modulation of Na+, K+-ATPase as a potential target for OXA-induced neurotoxicity, the therapeutic potential of 7-chloro-4-(phenylselanyl) quinoline (4-PSQ) was evaluated. Mice received OXA (10 mg kg-1) or vehicle by intraperitoneal route (days 0 and 2). Oral administration of 4-PSQ (1 mg kg-1) or vehicle was performed from days 2-14. Behavioral tasks started from day 12 onwards. On day 15, the animals were sacrificed, and the tissues collected. The effects of OXA and 4-PSQ on activity and expression level of Na+, K+-ATPase in the hippocampus and cerebral cortex, and the plasmatic corticosterone levels were determined. The findings demonstrated a significant positive correlation between anxious behavior and cognitive impairment induced by OXA. OXA caused an increase on the plasmatic corticosterone levels and reduced activity and expression level of Na+, K+-ATPase. 4-PSQ reduced both anxious behavior and cognitive impairment induced by OXA. 4-PSQ effect seems to be due to the modulation of Na+, K+-ATPase and reduction of corticosterone levels. Our results helped to expand knowledge about the mechanisms involved in the physiopathology of the OXA-induced neurotoxicity and strongly indicated that 4-PSQ may be a good prototype for the treatment of anxious behavior and cognitive impairment induced by OXA exposure.
Collapse
Affiliation(s)
- Angélica S Reis
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel - CEP, 96010-900, Pelotas, RS, Brazil
| | - Jaini J Paltian
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel - CEP, 96010-900, Pelotas, RS, Brazil
| | - William B Domingues
- Programa de Pós-graduação em Biotecnologia, Laboratório de Genômica Estrutural, Biotecnologia - Universidade Federal de Pelotas, UFPel - CEP, 96010-900, Pelotas, RS, Brazil
| | - Gabriel P Costa
- Programa de Pós-graduação em Química, Laboratório de Síntese Orgânica Limpa, CCQFA - Universidade Federal de Pelotas, UFPel - CEP, 96010-900, Pelotas, RS, Brazil
| | - Diego Alves
- Programa de Pós-graduação em Química, Laboratório de Síntese Orgânica Limpa, CCQFA - Universidade Federal de Pelotas, UFPel - CEP, 96010-900, Pelotas, RS, Brazil
| | - Janice L Giongo
- Pharmacy Department, Faculdade Anhanguera - CEP - 96055000, Pelotas, RS, Brazil
| | - Vinicius F Campos
- Programa de Pós-graduação em Biotecnologia, Laboratório de Genômica Estrutural, Biotecnologia - Universidade Federal de Pelotas, UFPel - CEP, 96010-900, Pelotas, RS, Brazil
| | - Cristiane Luchese
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel - CEP, 96010-900, Pelotas, RS, Brazil.
| | - Ethel A Wilhelm
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA - Universidade Federal de Pelotas, UFPel - CEP, 96010-900, Pelotas, RS, Brazil.
| |
Collapse
|
20
|
Venkataramaiah C. Modulations in the ATPases during ketamine-induced schizophrenia and regulatory effect of "3-(3, 4-dimethoxy phenyl) -1- (4-methoxyphenyl) prop-2-en-1-one": an in vivo and in silico studies. J Recept Signal Transduct Res 2020; 40:148-156. [PMID: 32009493 DOI: 10.1080/10799893.2020.1720242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Schizophrenia is a devastating illness and displays a wide range of psychotic symptoms. Accumulating evidence indicate impairment of bioenergetic pathways including energy storage and usage in the pathogenesis of schizophrenia. Although well-established synthetic drugs are being used for the management of schizophrenia, most of them have several adverse effects. Hence, natural products derived from medicinal plants represent a continuous major source for ethnomedicine-derived pharmaceuticals for different neurological disorders including schizophrenia. In the present study, we have investigated the neuroprotective effect of the novel bioactive compound i.e. "3-(3,4-dimethoxy phenyl) -1- (4-methoxyphenyl) prop-2-en-1-one" of Celastrus paniculata against ketamine-induced schizophrenia with particular reference to the activities of ATPase using in vivo and in silico methods. Ketamine-induced schizophrenia caused significant reduction in the activities of all three ATPases (Na+/K+, Ca2+ and Mg2+) in different regions of brain which reflects the decreased turnover of ATP, presumably due to the inhibition of oxidoreductase system and uncoupling of the same from the electron transport system. On par with the reference compound, clozapine, the activity levels of all three ATPases were restored to normal after pretreatment with the compound suggesting recovery of energy loss that was occurred during ketamine-induced schizophrenia. Besides, the compound has shown strong interaction and exhibited highest binding energies against all the three ATPases with a lowest inhibition constant value than the clozapine. The results of the present study clearly imply that the compound exhibit significant neuroprotective and antischizophrenic effect by modulating bioenergietic pathways that were altered during induced schizophrenia.
Collapse
Affiliation(s)
- Chintha Venkataramaiah
- Division of Molecular Biology, Department of Zoology, Sri Venkateswara University, Tirupati, India
| |
Collapse
|
21
|
Wu T, Donohoe ME. Yy1 regulates Senp1 contributing to AMPA receptor GluR1 expression following neuronal depolarization. J Biomed Sci 2019; 26:79. [PMID: 31629407 PMCID: PMC6800989 DOI: 10.1186/s12929-019-0582-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/09/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuronal activity-induced changes in gene expression patterns are important mediators of neuronal plasticity. Many neuronal genes can be activated or inactivated in response to neuronal depolarization. Mechanisms that activate gene transcription are well established, but activity-dependent mechanisms that silence transcription are less understood. It is also not clear what is the significance of inhibiting these genes during neuronal activity. METHODS Quantitative Real Time-PCR, western blot and immunofluorescence staining were performed to examine the expression of Senp1 and GluR1 in mouse cortical neurons. The alterations of Yy1 phosphorylation upon neuronal depolarization and the interaction of Yy1 with Brd4 were studied by protein co-immunoprecipitation. The regulators of Yy1 phosphorylation were identified by phosphatase inhibitors. Chromatin immunoprecipitation, in vitro DNA binding assay, luciferase assay and gene knockdown experiments were used to validate the roles of Yy1 and its phosphorylation as well as Brd4 in regulating Senp1 expression. RESULTS We report that neuronal depolarization deactivates the transcription of the SUMO protease Senp1, an important component regulating synaptic transmission, scaling, and plasticity, through Yy1. In un-stimulated neurons, Senp1 transcription is activated by a Yy1-Brd4 transcription factor protein complex assembled on the Senp1 promoter. Upon membrane depolarization, however, Yy1 is dephosphorylated and the Yy1-Brd4 complex is evicted from the Senp1 promoter, reducing Senp1 transcription levels. Both Yy1 and Senp1 promote the expression of AMPA receptor subunit GluR1, a pivotal component in learning and memory. CONCLUSIONS These results reveal an axis of Yy1/Brd4-Senp1 which regulates the expression of GluR1 during neuronal depolarization. This implicates a regulation mechanism in silencing gene expression upon neuronal activity.
Collapse
Affiliation(s)
- Tao Wu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, People's Republic of China.
- Burke Medical Research Institute, White Plains, NY, 10605, USA.
- Department of Neuroscience, Brain Mind Research Institute, Department of Cell & Development, Weill Cornell Medical College, New York, NY, 10065, USA.
| | - Mary E Donohoe
- Burke Medical Research Institute, White Plains, NY, 10605, USA.
- Department of Neuroscience, Brain Mind Research Institute, Department of Cell & Development, Weill Cornell Medical College, New York, NY, 10065, USA.
- Present address: Department of Medicine, Division of Regenerative Medicine, University of California San Diego School of Medicine, La Jolla, CA, 92037, USA.
| |
Collapse
|
22
|
Zhang Y, Guo O, Huo Y, Wang G, Man HY. Amyloid-β Induces AMPA Receptor Ubiquitination and Degradation in Primary Neurons and Human Brains of Alzheimer's Disease. J Alzheimers Dis 2019; 62:1789-1801. [PMID: 29614651 DOI: 10.3233/jad-170879] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
As the primary mediator for synaptic transmission, AMPA receptors (AMPARs) are crucial for synaptic plasticity and higher brain functions. A downregulation of AMPAR expression has been indicated as one of the early pathological molecular alterations in Alzheimer's disease (AD), presumably via amyloid-β (Aβ). However, the molecular mechanisms leading to the loss of AMPARs remain less clear. We report that in primary neurons, application of Aβ triggers AMPAR internalization accompanied with a decrease in cell-surface AMPAR expression. Importantly, in both Aβ-treated neurons and human brain tissue from AD patients, we observed a significant decrease in total AMPAR amount and an enhancement in AMPAR ubiquitination. Consistent with facilitated receptor degradation, AMPARs show higher turnover rates in the presence of Aβ. Furthermore, AD brain lysates and Aβ-incubated neurons show increased expression of the AMPAR E3 ligase Nedd4 and decreased expression of AMPAR deubiquitinase USP46. Changes in these enzymes are responsible for the Aβ-dependent AMPAR reduction. These findings indicate that AMPAR ubiquitination acts as the key molecular event leading to the loss of AMPARs and thus suppressed synaptic transmission in AD.
Collapse
Affiliation(s)
- Yanmin Zhang
- Department of Biology, Boston University, Boston, MA, USA.,Department of Histology and Embryology, Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Ouyang Guo
- Department of Biology, Boston University, Boston, MA, USA
| | - Yuda Huo
- Department of Biology, Boston University, Boston, MA, USA
| | - Guan Wang
- Department of Biology, Boston University, Boston, MA, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA.,Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
23
|
Waugh DT. Fluoride Exposure Induces Inhibition of Sodium-and Potassium-Activated Adenosine Triphosphatase (Na +, K +-ATPase) Enzyme Activity: Molecular Mechanisms and Implications for Public Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E1427. [PMID: 31010095 PMCID: PMC6518254 DOI: 10.3390/ijerph16081427] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 12/24/2022]
Abstract
In this study, several lines of evidence are provided to show that Na + , K + -ATPase activity exerts vital roles in normal brain development and function and that loss of enzyme activity is implicated in neurodevelopmental, neuropsychiatric and neurodegenerative disorders, as well as increased risk of cancer, metabolic, pulmonary and cardiovascular disease. Evidence is presented to show that fluoride (F) inhibits Na + , K + -ATPase activity by altering biological pathways through modifying the expression of genes and the activity of glycolytic enzymes, metalloenzymes, hormones, proteins, neuropeptides and cytokines, as well as biological interface interactions that rely on the bioavailability of chemical elements magnesium and manganese to modulate ATP and Na + , K + -ATPase enzyme activity. Taken together, the findings of this study provide unprecedented insights into the molecular mechanisms and biological pathways by which F inhibits Na + , K + -ATPase activity and contributes to the etiology and pathophysiology of diseases associated with impairment of this essential enzyme. Moreover, the findings of this study further suggest that there are windows of susceptibility over the life course where chronic F exposure in pregnancy and early infancy may impair Na + , K + -ATPase activity with both short- and long-term implications for disease and inequalities in health. These findings would warrant considerable attention and potential intervention, not to mention additional research on the potential effects of F intake in contributing to chronic disease.
Collapse
Affiliation(s)
- Declan Timothy Waugh
- EnviroManagement Services, 11 Riverview, Doherty's Rd, P72 YF10 Bandon, Co. Cork, Ireland.
| |
Collapse
|
24
|
Shi M, Cao L, Cao X, Zhu M, Zhang X, Wu Z, Xiong S, Xie Z, Yang Y, Chen J, Wong PTH, Bian JS. DR-region of Na +/K + ATPase is a target to treat excitotoxicity and stroke. Cell Death Dis 2018; 10:6. [PMID: 30584244 PMCID: PMC6315034 DOI: 10.1038/s41419-018-1230-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Abstract
Na+/K+ ATPase (NKA) is important in maintaining cellular functions. We found that loss of NKA activities in NKAα1+/− mice is associated with increased susceptibility to ischemic injuries following transient middle cerebral artery occlusion (tMCAO). This is corroborated by the neuroprotective effects of an antibody raised against an extracellular DR region (897DVEDSYGQQWTYEQR911, sequence number as in rat) of NKAα subunit (DR-Ab) in both preventive and therapeutic settings. DR-Ab protects cortical neurons against glutamate-induced toxicity by stimulating activities of NKA and Na+/Ca2+ exchanger (NCX), which resulted in accelerated Ca2+ extrusion. DR-Ab also enhanced the association between NKA and GluR2 and therefore reduced the internalization of both proteins from membrane induced by glutamate toxicity. The mechanism appears to involve suppression of GluR2 phosphorylation through PKCα/PICK pathway. Our data indicate that DR-region of NKA may be a novel therapeutic target for drug development for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Meimei Shi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Lei Cao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Xu Cao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Mengyuan Zhu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Xingzhou Zhang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Zhiyuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Siping Xiong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Zhizhong Xie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Yong Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Jingyu Chen
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214021, Jiangsu, PR China
| | - Peter T H Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore. .,National University of Singapore (Suzhou) Research Institute, Suzhou, 215123, China.
| |
Collapse
|
25
|
Shrivastava AN, Triller A, Melki R. Cell biology and dynamics of Neuronal Na +/K +-ATPase in health and diseases. Neuropharmacology 2018; 169:107461. [PMID: 30550795 DOI: 10.1016/j.neuropharm.2018.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/17/2018] [Accepted: 12/08/2018] [Indexed: 10/27/2022]
Abstract
Neuronal Na+/K+-ATPase is responsible for the maintenance of ionic gradient across plasma membrane. In doing so, in a healthy brain, Na+/K+-ATPase activity accounts for nearly half of total brain energy consumption. The α3-subunit containing Na+/K+-ATPase expression is restricted to neurons. Heterozygous mutations within α3-subunit leads to Rapid-onset Dystonia Parkinsonism, Alternating Hemiplegia of Childhood and other neurological and neuropsychiatric disorders. Additionally, proteins such as α-synuclein, amyloid-β, tau and SOD1 whose aggregation is associated to neurodegenerative diseases directly bind and impair α3-Na+/K+-ATPase activity. The review will provide a summary of neuronal α3-Na+/K+-ATPase functional properties, expression pattern, protein-protein interactions at the plasma membrane, biophysical properties (distribution and lateral diffusion). Lastly, the role of α3-Na+/K+-ATPase in neurological and neurodegenerative disorders will be discussed. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Amulya Nidhi Shrivastava
- CEA, Institut François Jacob (MIRcen) and CNRS, Laboratory of Neurodegenerative Diseases (U9199), 18 Route du Panorama, 92265, Fontenay-aux-Roses, France.
| | - Antoine Triller
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, INSERM, CNRS, PSL, Research University, 46 Rue d'Ulm, 75005 Paris, France
| | - Ronald Melki
- CEA, Institut François Jacob (MIRcen) and CNRS, Laboratory of Neurodegenerative Diseases (U9199), 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Dash B, Dib-Hajj SD, Waxman SG. Multiple myosin motors interact with sodium/potassium-ATPase alpha 1 subunits. Mol Brain 2018; 11:45. [PMID: 30086768 PMCID: PMC6081954 DOI: 10.1186/s13041-018-0388-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/20/2018] [Indexed: 11/10/2022] Open
Abstract
The alpha1 (α1) subunit of the sodium/potassium ATPase (i.e., Na+/K+-ATPase α1), the prototypical sodium pump, is expressed in each eukaryotic cell. They pump out three sodium ions in exchange for two extracellular potassium ions to establish a cellular electrochemical gradient important for firing of neuronal and cardiac action potentials. We hypothesized that myosin (myo or myh) motor proteins might interact with Na+/K+-ATPase α1 subunits in order for them to play an important role in the transport and trafficking of sodium pump. To this end immunoassays were performed to determine whether class II non-muscle myosins (i.e., NMHC-IIA/myh9, NMHC-IIB/myh10 or NMHC-IIC/myh14), myosin Va (myoVa) and myosin VI (myoVI) would interact with Na+/K+-ATPase α1 subunits. Immunoprecipitation of myh9, myh10, myh14, myoVa and myoVI from rat brain tissues led to the co-immunoprecipitation of Na+/K+-ATPase α1 subunits expressed there. Heterologous expression studies using HEK293 cells indicated that recombinant myh9, myh10, myh14 and myoVI interact with Na+/K+-ATPase α1 subunits expressed in HEK293 cells. Additional results indicated that loss of tail regions in recombinant myh9, myh10, myh14 and myoVI did not affect their interaction with Na+/K+-ATPase α1 subunits. However, recombinant myh9, myh10 and myh14 mutants having reduced or no actin binding ability, as a result of loss of their actin binding sites, displayed greatly reduced or null interaction with Na+/K+-ATPase α1 subunits. These results suggested the involvement of the actin binding site, but not tail regions, of NMHC-IIs in their interaction with Na+/K+-ATPase α1 subunits. Overall these results suggest a role for these diverse myosins in the trafficking and transport of sodium pump in neuronal and non-neuronal tissues.
Collapse
Affiliation(s)
- Bhagirathi Dash
- Department of Neurology, Yale University Schoolof Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research center, VA Connecticut Healthcare System, 950 Campbell Avenue, Bldg. 34, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University Schoolof Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research center, VA Connecticut Healthcare System, 950 Campbell Avenue, Bldg. 34, West Haven, CT, 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University Schoolof Medicine, New Haven, CT, 06510, USA. .,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA. .,Rehabilitation Research center, VA Connecticut Healthcare System, 950 Campbell Avenue, Bldg. 34, West Haven, CT, 06516, USA.
| |
Collapse
|
29
|
Lopachev AV, Abaimov DA, Fedorova TN, Lopacheva OM, Akkuratova NV, Akkuratov EE. Cardiotonic Steroids as Potential Endogenous Regulators in the Nervous System. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418010087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
30
|
Widagdo J, Guntupalli S, Jang SE, Anggono V. Regulation of AMPA Receptor Trafficking by Protein Ubiquitination. Front Mol Neurosci 2017; 10:347. [PMID: 29123470 PMCID: PMC5662755 DOI: 10.3389/fnmol.2017.00347] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/12/2017] [Indexed: 11/27/2022] Open
Abstract
The molecular mechanisms underlying plastic changes in the strength and connectivity of excitatory synapses have been studied extensively for the past few decades and remain the most attractive cellular models of learning and memory. One of the major mechanisms that regulate synaptic plasticity is the dynamic adjustment of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptor content on the neuronal plasma membrane. The expression of surface AMPA receptors (AMPARs) is controlled by the delicate balance between the biosynthesis, dendritic transport, exocytosis, endocytosis, recycling and degradation of the receptors. These processes are dynamically regulated by AMPAR interacting proteins as well as by various post-translational modifications that occur on their cytoplasmic domains. In the last few years, protein ubiquitination has emerged as a major regulator of AMPAR intracellular trafficking. Dysregulation of AMPAR ubiquitination has also been implicated in the pathophysiology of Alzheimer’s disease. Here we review recent advances in the field and provide insights into the role of protein ubiquitination in regulating AMPAR membrane trafficking and function. We also discuss how aberrant ubiquitination of AMPARs contributes to the pathogenesis of various neurological disorders, including Alzheimer’s disease, chronic stress and epilepsy.
Collapse
Affiliation(s)
- Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Sumasri Guntupalli
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Se E Jang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
31
|
Chakraborty D, Fedorova OV, Bagrov AY, Kaphzan H. Selective ligands for Na+/K+-ATPase α isoforms differentially and cooperatively regulate excitability of pyramidal neurons in distinct brain regions. Neuropharmacology 2017; 117:338-351. [DOI: 10.1016/j.neuropharm.2017.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/24/2017] [Accepted: 02/17/2017] [Indexed: 11/28/2022]
|
32
|
Adaptor Complex 2 Controls Dendrite Morphology via mTOR-Dependent Expression of GluA2. Mol Neurobiol 2017; 55:1590-1606. [PMID: 28190237 PMCID: PMC5820378 DOI: 10.1007/s12035-017-0436-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 02/03/2017] [Indexed: 11/26/2022]
Abstract
The formation of dendritic arbors in neurons is a highly regulated process. Among the regulators of dendritogenesis are numerous membrane proteins that are eventually internalized via clathrin-mediated endocytosis. AP2 is an adaptor complex that is responsible for recruiting endocytic machinery to internalized cargo. Its direct involvement in dendritogenesis in mammalian neurons has not yet been tested. We found that the knockdown of AP2b1 (β2-adaptin), an AP2 subunit, reduced the number of dendrites in developing rat hippocampal neurons and decreased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA2 levels by inhibiting mechanistic/mammalian target of rapamycin (mTOR). The dendritic tree abruption that was caused by AP2b1 knockdown was rescued by the overexpression of GluA2 or restoration of the activity of the mTOR effector p70S6 kinase (S6K1). Altogether, this work provides evidence that the AP2 adaptor complex is needed for the dendritogenesis of mammalian neurons and reveals that mTOR-dependent GluA2 biosynthesis contributes to this process.
Collapse
|
33
|
Akkuratov EE, Wu J, Sowa D, Shah ZA, Liu L. Ouabain-Induced Signaling and Cell Survival in SK-N-SH Neuroblastoma Cells Differentiated by Retinoic Acid. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2016; 14:1343-9. [PMID: 26295826 PMCID: PMC5388798 DOI: 10.2174/1871527314666150821103008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/18/2015] [Indexed: 11/22/2022]
Abstract
Ouabain stimulates activation of various signaling cascades such as protein kinase B (Akt) and Extracellular-signaling-regulated kinase 1/2 (ERK 1/2) in various cell lines. Retinoic acid (RA) is commonly used to induce neuroblastoma differentiation in cultures. Upon RA administration, human neuroblastoma cell line, SK-N-SH demonstrated neurite extensions, which is an indicator of neuronal cell differentiation. Here we report that ouabain-induced signaling is altered under the action of 1 μM RA in human neuroblastoma SK-N-SH cells. RA increased the expression of p110α subunit of phosphoinositide 3-kinase (PI3K), Akt and β1 subunit of Na+/K+-ATPase. Ouabain activated Akt and ERK 1/2 in differentiated SK-N-SH cells; this effect was not observed in non-differentiated SK-N-SH cells. Long-term incubation of non-differentiated SK-N-SH with 1 μM ouabain led to a decrease in the number of cells; this effect was reduced in differentiated SK-N-SH cells. Taken together, these results suggest that ouabain leads to cell death in neuroblastoma cells rather than neuronal cells due to the different response to ouabain manifested by activation of Akt and ERK 1/2.
Highlights
• RA increases the expression of p110α subunit of PI3K, Akt and β1 subunit of Na+/K+-ATPase • Ouabain induces activation of Akt and ERK 1/2 in differentiated SK-N-SH cells but not in non-differentiated cells • 1 μM ouabain leads to a decrease in the number of cells in non-differentiated SK-N-SH • Reduction of ouabain-induced cell death in differentiated SK-N-SH
Collapse
Affiliation(s)
| | | | | | | | - Lijun Liu
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
34
|
Hakim V, Cohen LD, Zuchman R, Ziv T, Ziv NE. The effects of proteasomal inhibition on synaptic proteostasis. EMBO J 2016; 35:2238-2262. [PMID: 27613546 PMCID: PMC5069550 DOI: 10.15252/embj.201593594] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 08/08/2016] [Indexed: 01/03/2023] Open
Abstract
Synaptic function crucially depends on uninterrupted synthesis and degradation of synaptic proteins. While much has been learned on synaptic protein synthesis, little is known on the routes by which synaptic proteins are degraded. Here we systematically studied how inhibition of the ubiquitin-proteasome system (UPS) affects the degradation rates of thousands of neuronal and synaptic proteins. We identified a group of proteins, including several proteins related to glutamate receptor trafficking, whose degradation rates were significantly slowed by UPS inhibition. Unexpectedly, however, degradation rates of most synaptic proteins were not significantly affected. Interestingly, many of the differential effects of UPS inhibition were readily explained by a quantitative framework that considered known metabolic turnover rates for the same proteins. In contrast to the limited effects on protein degradation, UPS inhibition profoundly and preferentially suppressed the synthesis of a large number of synaptic proteins. Our findings point to the importance of the UPS in the degradation of certain synaptic proteins, yet indicate that under basal conditions most synaptic proteins might be degraded through alternative pathways.
Collapse
Affiliation(s)
- Vicky Hakim
- The Rappaport Faculty of Medicine and Research Institute, Haifa, Israel.,Network Biology Research Laboratories, Technion - Israel Institute of Technology, Haifa, Israel
| | - Laurie D Cohen
- The Rappaport Faculty of Medicine and Research Institute, Haifa, Israel.,Network Biology Research Laboratories, Technion - Israel Institute of Technology, Haifa, Israel
| | - Rina Zuchman
- Smoler Proteomics Center, Faculty of Biology, Technion, Haifa, Israel
| | - Tamar Ziv
- Smoler Proteomics Center, Faculty of Biology, Technion, Haifa, Israel
| | - Noam E Ziv
- The Rappaport Faculty of Medicine and Research Institute, Haifa, Israel .,Network Biology Research Laboratories, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
35
|
Banerjee B, Medda BK, Zhang J, Tuchscherer V, Babygirija R, Kannampalli P, Sengupta JN, Shaker R. Prolonged esophageal acid exposures induce synaptic downscaling of cortical membrane AMPA receptor subunits in rats. Neurogastroenterol Motil 2016; 28:1356-69. [PMID: 27271201 PMCID: PMC5063079 DOI: 10.1111/nmo.12834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/11/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND We recently reported the involvement of AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor subunit upregulation and phosphorylation in the rostral cingulate cortex (rCC) as the underlying mechanism of acute esophageal acid-induced cortical sensitization. Based on these findings, we proposed to investigate whether prolonged esophageal acid exposures in rats exhibit homeostatic synaptic scaling through downregulation of AMPA receptor expression in rCC neurons. We intended to study further whether this compensatory mechanism is impaired when rats are pre-exposed to repeated esophageal acid exposures neonatally during neuronal development. METHODS Two different esophageal acid exposure protocols in rats were used. Since AMPA receptor trafficking and channel conductance depend on CaMKIIα-mediated phosphorylation of AMPA receptor subunits, we examined the effect of esophageal acid on CaMKIIα activation and AMPA receptor expression in synaptoneurosomes and membrane preparations from rCCs. KEY RESULTS In cortical membrane preparations, GluA1 and pGluA1Ser(831) expression were significantly downregulated following prolonged acid exposures in adult rats; this was accompanied by the significant downregulation of cortical membrane pCaMKIIα expression. No change in GluA1 and pGluA1Ser(831) expression was observed in rCC membrane preparations in rats pre-exposed to acid neonatally followed by adult rechallenge. CONCLUSIONS & INFERENCES This study along with our previous findings suggests that synaptic AMPA receptor subunits expression and phosphorylation may be involved bidirectionally in both esophageal acid-induced neuronal sensitization and acid-dependent homeostatic plasticity in cortical neurons. The impairment of homeostatic compensatory mechanism as observed following early-in-life acid exposure could be the underlying mechanism of heightening cortical sensitization and esophageal hypersensitivity in patients with gastroesophageal reflux disease.
Collapse
Affiliation(s)
- Banani Banerjee
- Gastroenterology & Hepatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bidyut K Medda
- Gastroenterology & Hepatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jian Zhang
- Gastroenterology & Hepatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Reji Babygirija
- Gastroenterology & Hepatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Pradeep Kannampalli
- Gastroenterology & Hepatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jyoti N. Sengupta
- Gastroenterology & Hepatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Reza Shaker
- Gastroenterology & Hepatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
36
|
Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells. PLoS One 2016; 11:e0159034. [PMID: 27428326 PMCID: PMC4948917 DOI: 10.1371/journal.pone.0159034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 06/27/2016] [Indexed: 11/25/2022] Open
Abstract
Although the possibility of developing cardiac steroids/cardiac glycosides as novel cancer therapeutic agents has been recognized, the mechanism of their anticancer activity is still not clear enough. Toad venom extract containing bufadienolides, which belong to cardiac steroids, has actually long been used as traditional Chinese medicine in clinic for cancer therapy in China. The cytotoxicity of arenobufagin, a bufadienolide isolated from toad venom, on human cervical carcinoma HeLa cells was checked. And, the protein expression profile of control HeLa cells and HeLa cells treated with arenobufagin for 48 h was analyzed using two-dimensional electrophoresis, respectively. Differently expressed proteins in HeLa cells treated with arenobufagin were identified and the pathways related to these proteins were mapped from KEGG database. Computational molecular docking was performed to verify the binding of arenobufagin and Na, K-ATPase. The effects of arenobufagin on Na, K-ATPase activity and proteasome activity of HeLa cells were checked. The protein-protein interaction network between Na, K-ATPase and proteasome was constructed and the expression of possible intermediate proteins ataxin-1 and translationally-controlled tumor protein in HeLa cells treated with arenobufagin was then checked. Arenobufagin induced apoptosis and G2/M cell cycle arrest in HeLa cells. The cytotoxic effect of arenobufagin was associated with 25 differently expressed proteins including proteasome-related proteins, calcium ion binding-related proteins, oxidative stress-related proteins, metabolism-related enzymes and others. The results of computational molecular docking revealed that arenobufagin was bound in the cavity formed by the transmembrane alpha subunits of Na, K-ATPase, which blocked the pathway of extracellular Na+/K+ cation exchange and inhibited the function of ion exchange. Arenobufagin inhibited the activity of Na, K-ATPase and proteasome, decreased the expression of Na, K-ATPase α1 and α3 subunits and increased the expression of WEE1 in HeLa cells. Antibodies against Na, K-ATPase α1 and α3 subunits alone or combinated with arenobufagin also inhibited the activity of proteasome. Furthermore, the expression of the possible intermediate proteins ataxin-1 and translationally-controlled tumor protein was increased in HeLa cells treated with arenobufagin by flow cytometry analysis, respectively. These results indicated that arenobufagin might directly bind with Na, K-ATPase α1 and α3 subunits and the inhibitive effect of arenobufagin on proteasomal activity of HeLa cells might be related to its binding with Na, K-ATPase.
Collapse
|
37
|
Sun P, Feng LX, Zhang DM, Liu M, Liu W, Mi T, Wu WY, Jiang BH, Yang M, Hu LH, Guo DA, Liu X. Bufalin derivative BF211 inhibits proteasome activity in human lung cancer cells in vitro by inhibiting β1 subunit expression and disrupting proteasome assembly. Acta Pharmacol Sin 2016; 37:908-18. [PMID: 27238210 DOI: 10.1038/aps.2016.30] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/28/2016] [Indexed: 12/20/2022] Open
Abstract
AIM Bufalin is one of the active components in the traditional Chinese medicine ChanSu that is used to treat arrhythmia, inflammation and cancer. BF211 is a bufalin derivative with stronger cytotoxic activity in cancer cells. The aim of this study was to identify the putative target proteins of BF211 and the signaling pathways in cancer cells. METHODS A549 human lung cancer cells were treated with BF211. A SILAC-based proteomic analysis was used to detect the protein expression profiles of BF211-treated A549 cells. Cellular proteasome activities were examined using fluorogenic peptide substrates, and the binding affinities of BF211 to recombinant proteasome subunit proteins were evaluated using the Biacore assay. The expression levels of proteasome subunits were determined using RT-PCR and Western blotting, and the levels of the integral 26S proteasome were evaluated using native PAGE analysis. RESULTS The proteomic analysis revealed that 1282 proteins were differentially expressed in BF211-treated A549 cells, and the putative target proteins of BF211 were associated with various cellular functions, including transcription, translation, mRNA splicing, ribosomal protein synthesis and proteasome function. In A549 cells, BF211 (5, 10, and 20 nmol/L) dose-dependently inhibited the enzymatic activities of proteasome. But BF211 displayed a moderate affinity in binding to proteasome β1 subunit and no binding affinity to the β2 and β5 subunits. Moreover, BF211 (0.1, 1, and 10 nmol/L) did not inhibit the proteasome activities in the cell lysates. BF211 (5, 10, and 20 nmol/L) significantly decreased the expression level of proteasome β1 subunit and the levels of integral 26S proteasome in A549 cells. Similarly, knockdown of the β1 subunit with siRNA in A549 cells significantly decreased integral 26S proteasome and proteasome activity. CONCLUSION BF211 inhibits proteasome activity in A549 cells by decreasing β1 subunit expression and disrupting proteasome assembly.
Collapse
|
38
|
Kinoshita PF, Leite JA, Orellana AMM, Vasconcelos AR, Quintas LEM, Kawamoto EM, Scavone C. The Influence of Na(+), K(+)-ATPase on Glutamate Signaling in Neurodegenerative Diseases and Senescence. Front Physiol 2016; 7:195. [PMID: 27313535 PMCID: PMC4890531 DOI: 10.3389/fphys.2016.00195] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022] Open
Abstract
Decreased Na(+), K(+)-ATPase (NKA) activity causes energy deficiency, which is commonly observed in neurodegenerative diseases. The NKA is constituted of three subunits: α, β, and γ, with four distinct isoforms of the catalytic α subunit (α1-4). Genetic mutations in the ATP1A2 gene and ATP1A3 gene, encoding the α2 and α3 subunit isoforms, respectively can cause distinct neurological disorders, concurrent to impaired NKA activity. Within the central nervous system (CNS), the α2 isoform is expressed mostly in glial cells and the α3 isoform is neuron-specific. Mutations in ATP1A2 gene can result in familial hemiplegic migraine (FHM2), while mutations in the ATP1A3 gene can cause Rapid-onset dystonia-Parkinsonism (RDP) and alternating hemiplegia of childhood (AHC), as well as the cerebellar ataxia, areflexia, pescavus, optic atrophy and sensorineural hearing loss (CAPOS) syndrome. Data indicates that the central glutamatergic system is affected by mutations in the α2 isoform, however further investigations are required to establish a connection to mutations in the α3 isoform, especially given the diagnostic confusion and overlap with glutamate transporter disease. The age-related decline in brain α2∕3 activity may arise from changes in the cyclic guanosine monophosphate (cGMP) and cGMP-dependent protein kinase (PKG) pathway. Glutamate, through nitric oxide synthase (NOS), cGMP and PKG, stimulates brain α2∕3 activity, with the glutamatergic N-methyl-D-aspartate (NMDA) receptor cascade able to drive an adaptive, neuroprotective response to inflammatory and challenging stimuli, including amyloid-β. Here we review the NKA, both as an ion pump as well as a receptor that interacts with NMDA, including the role of NKA subunits mutations. Failure of the NKA-associated adaptive response mechanisms may render neurons more susceptible to degeneration over the course of aging.
Collapse
Affiliation(s)
- Paula F. Kinoshita
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Jacqueline A. Leite
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Ana Maria M. Orellana
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Andrea R. Vasconcelos
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Luis E. M. Quintas
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Elisa M. Kawamoto
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| |
Collapse
|
39
|
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
|
40
|
Blom H, Bernhem K, Brismar H. Sodium pump organization in dendritic spines. NEUROPHOTONICS 2016; 3:041803. [PMID: 27175374 PMCID: PMC4855081 DOI: 10.1117/1.nph.3.4.041803] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/04/2016] [Indexed: 05/16/2023]
Affiliation(s)
- Hans Blom
- Royal Institute of Technology, Department of Applied Physics, Roslagstullsbacken 21, Stockholm 10691, SwedenbScience for Life Laboratory, Advanced Light Microscopy Facility, Tomtebodavägen 23A, Solna 17165, Sweden
| | - Kristoffer Bernhem
- Royal Institute of Technology, Department of Applied Physics, Roslagstullsbacken 21, Stockholm 10691, Sweden
| | - Hjalmar Brismar
- Royal Institute of Technology, Department of Applied Physics, Roslagstullsbacken 21, Stockholm 10691, SwedenbScience for Life Laboratory, Advanced Light Microscopy Facility, Tomtebodavägen 23A, Solna 17165, SwedencKarolinska Institutet, Department of Wome
| |
Collapse
|
41
|
Abstract
All animals are characterized by steep gradients of Na(+) and K(+) across the plasma membrane, and in spite of their highly similar chemical properties, the ions can be distinguished by numerous channels and transporters. The gradients are generated by the Na(+),K(+)-ATPase, or sodium pump, which pumps out Na(+) and takes up K(+) at the expense of the chemical energy from ATP. Because the membrane is more permeable to K(+) than to Na(+), the uneven ion distribution causes a transmembrane voltage difference, and this membrane potential forms the basis for the action potential and for much of the neuronal signaling in general. The potential energy stored in the concentration gradients is also used to drive a large number of the secondary transporters responsible for transmembrane carriage of solutes ranging from sugars, amino acids, and neurotransmitters to inorganic ions such as chloride, inorganic phosphate, and bicarbonate. Furthermore, Na(+) and K(+) themselves are important enzymatic cofactors that typically lower the energy barrier of substrate binding.In this chapter, we describe the roles of Na(+) and K(+) in the animal cell with emphasis on the creation and usage of the steep gradients across the membrane. More than 50 years of Na(+),K(+)-ATPase research has revealed many details of the molecular machinery and offered insights into how the pump is regulated by post-translational modifications and specific drugs.
Collapse
Affiliation(s)
- Michael Jakob Voldsgaard Clausen
- Centre for Structural Biology, Department of Molecular Biology and Genetics, University of Aarhus, Science Park, Gustav Wieds Vej 10c, Aarhus C, Denmark,
| | | |
Collapse
|
42
|
Miyata S, Taniguchi M, Koyama Y, Shimizu S, Tanaka T, Yasuno F, Yamamoto A, Iida H, Kudo T, Katayama T, Tohyama M. Association between chronic stress-induced structural abnormalities in Ranvier nodes and reduced oligodendrocyte activity in major depression. Sci Rep 2016; 6:23084. [PMID: 26976207 PMCID: PMC4791682 DOI: 10.1038/srep23084] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 02/25/2016] [Indexed: 11/09/2022] Open
Abstract
Repeated stressful events are associated with the onset of major depressive disorder (MDD). We previously showed oligodendrocyte (OL)-specific activation of the serum/glucocorticoid-regulated kinase (SGK)1 cascade, increased expression of axon-myelin adhesion molecules, and elaboration of the oligodendrocytic arbor in the corpus callosum of chronically stressed mice. In the current study, we demonstrate that the nodes and paranodes of Ranvier in the corpus callosum were narrower in these mice. Chronic stress also led to diffuse redistribution of Caspr and Kv 1.1 and decreased the activity in white matter, suggesting a link between morphological changes in OLs and inhibition of axonal activity. OL primary cultures subjected to chronic stress resulted in SGK1 activation and translocation to the nucleus, where it inhibited the transcription of metabotropic glutamate receptors (mGluRs). Furthermore, the cAMP level and membrane potential of OLs were reduced by chronic stress exposure. We showed by diffusion tensor imaging that the corpus callosum of patients with MDD exhibited reduced fractional anisotropy, reflecting compromised white matter integrity possibly caused by axonal damage. Our findings suggest that chronic stress disrupts the organization of the nodes of Ranvier by suppressing mGluR activation in OLs, and that specific white matter abnormalities are closely associated with MDD onset.
Collapse
Affiliation(s)
- Shingo Miyata
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki University, Osaka-sayama, Osaka 589-8511, Japan
| | - Manabu Taniguchi
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yoshihisa Koyama
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shoko Shimizu
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki University, Osaka-sayama, Osaka 589-8511, Japan
| | - Takashi Tanaka
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki University, Osaka-sayama, Osaka 589-8511, Japan
| | - Fumihiko Yasuno
- Department of Psychiatry, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Akihide Yamamoto
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Hidehiro Iida
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Takashi Kudo
- Department of Psychiatry, Osaka University Health Care Center, Toyonaka, Osaka 560-0043, Japan.,Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Taiichi Katayama
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University and Hamamatsu University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masaya Tohyama
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki University, Osaka-sayama, Osaka 589-8511, Japan.,Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University and Hamamatsu University School of Medicine, Suita, Osaka 565-0871, Japan.,Osaka Prefectural Hospital Organization, Osaka 558-8558, Japan
| |
Collapse
|
43
|
Ruegsegger C, Maharjan N, Goswami A, Filézac de L'Etang A, Weis J, Troost D, Heller M, Gut H, Saxena S. Aberrant association of misfolded SOD1 with Na(+)/K(+)ATPase-α3 impairs its activity and contributes to motor neuron vulnerability in ALS. Acta Neuropathol 2016; 131:427-51. [PMID: 26619836 DOI: 10.1007/s00401-015-1510-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 11/03/2015] [Accepted: 11/14/2015] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult onset progressive motor neuron disease with no cure. Transgenic mice overexpressing familial ALS associated human mutant SOD1 are a commonly used model for examining disease mechanisms. Presently, it is well accepted that alterations in motor neuron excitability and spinal circuits are pathological hallmarks of ALS, but the underlying molecular mechanisms remain unresolved. Here, we sought to understand whether the expression of mutant SOD1 protein could contribute to altering processes governing motor neuron excitability. We used the conformation specific antibody B8H10 which recognizes a misfolded state of SOD1 (misfSOD1) to longitudinally identify its interactome during early disease stage in SOD1G93A mice. This strategy identified a direct isozyme-specific association of misfSOD1 with Na(+)/K(+)ATPase-α3 leading to the premature impairment of its ATPase activity. Pharmacological inhibition of Na(+)/K(+)ATPase-α3 altered glutamate receptor 2 expression, modified cholinergic inputs and accelerated disease pathology. After mapping the site of direct association of misfSOD1 with Na(+)/K(+)ATPase-α3 onto a 10 amino acid stretch that is unique to Na(+)/K(+)ATPase-α3 but not found in the closely related Na(+)/K(+)ATPase-α1 isozyme, we generated a misfSOD1 binding deficient, but fully functional Na(+)/K(+)ATPase-α3 pump. Adeno associated virus (AAV)-mediated expression of this chimeric Na(+)/K(+)ATPase-α3 restored Na(+)/K(+)ATPase-α3 activity in the spinal cord, delayed pathological alterations and prolonged survival of SOD1G93A mice. Additionally, altered Na(+)/K(+)ATPase-α3 expression was observed in the spinal cord of individuals with sporadic and familial ALS. A fraction of sporadic ALS cases also presented B8H10 positive misfSOD1 immunoreactivity, suggesting that similar mechanism might contribute to the pathology.
Collapse
Affiliation(s)
- Céline Ruegsegger
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Niran Maharjan
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Anand Goswami
- Institute of Neuropathology, Rheinisch-Westfälische Technische Hochschule, Aachen University Hospital, Aachen, Germany
| | - Audrey Filézac de L'Etang
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Neuroscience, Genentech, Inc., South San Francisco, California, USA
| | - Joachim Weis
- Institute of Neuropathology, Rheinisch-Westfälische Technische Hochschule, Aachen University Hospital, Aachen, Germany
| | - Dirk Troost
- Division of Neuropathology, Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Manfred Heller
- Department of Clinical Research, Inselspital, University of Bern, Bern, Switzerland
| | - Heinz Gut
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Smita Saxena
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| |
Collapse
|
44
|
MicroRNA miR124 is required for the expression of homeostatic synaptic plasticity. Nat Commun 2015; 6:10045. [PMID: 26620774 PMCID: PMC4686673 DOI: 10.1038/ncomms10045] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 10/29/2015] [Indexed: 12/19/2022] Open
Abstract
Homeostatic synaptic plasticity is a compensatory response to alterations in neuronal activity. Chronic deprivation of neuronal activity results in an increase in synaptic AMPA receptors (AMPARs) and postsynaptic currents. The biogenesis of GluA2-lacking, calcium-permeable AMPARs (CP-AMPARs) plays a crucial role in the homeostatic response; however, the mechanisms leading to CP-AMPAR formation remain unclear. Here we show that the microRNA, miR124, is required for the generation of CP-AMPARs and homeostatic plasticity. miR124 suppresses GluA2 expression via targeting its 3′-UTR, leading to the formation of CP-AMPARs. Blockade of miR124 function abolishes the homeostatic response, whereas miR124 overexpression leads to earlier induction of homeostatic plasticity. miR124 transcription is controlled by an inhibitory transcription factor EVI1, acting by association with the deacetylase HDAC1. Our data support a cellular cascade in which inactivity relieves EVI1/HDAC-mediated inhibition of miR124 gene transcription, resulting in enhanced miR124 expression, formation of CP-AMPARs and subsequent induction of homeostatic synaptic plasticity. GluA2-lacking AMPA receptors are known to play a role in homeostatic plasticity. Here, the authors show that spiking activity blockade disinhibits mir124 transcription, which in turn suppresses GluA2 mRNA translation, thereby contributing to synaptic upscaling in hippocampal cells.
Collapse
|
45
|
Goo MS, Scudder SL, Patrick GN. Ubiquitin-dependent trafficking and turnover of ionotropic glutamate receptors. Front Mol Neurosci 2015; 8:60. [PMID: 26528125 PMCID: PMC4607782 DOI: 10.3389/fnmol.2015.00060] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/22/2015] [Indexed: 12/19/2022] Open
Abstract
Changes in synaptic strength underlie the basis of learning and memory and are controlled, in part, by the insertion or removal of AMPA-type glutamate receptors at the postsynaptic membrane of excitatory synapses. Once internalized, these receptors may be recycled back to the plasma membrane by subunit-specific interactions with other proteins or by post-translational modifications such as phosphorylation. Alternatively, these receptors may be targeted for destruction by multiple degradation pathways in the cell. Ubiquitination, another post-translational modification, has recently emerged as a key signal that regulates the recycling and trafficking of glutamate receptors. In this review, we will discuss recent findings on the role of ubiquitination in the trafficking and turnover of ionotropic glutamate receptors and plasticity of excitatory synapses.
Collapse
Affiliation(s)
- Marisa S Goo
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Samantha L Scudder
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Gentry N Patrick
- Section of Neurobiology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
46
|
Tan W, Yao WL, Hu R, Lv YY, Wan L, Zhang CH, Zhu C. Alleviating neuropathic pain mechanical allodynia by increasing Cdh1 in the anterior cingulate cortex. Mol Pain 2015; 11:56. [PMID: 26364211 PMCID: PMC4568074 DOI: 10.1186/s12990-015-0058-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 08/26/2015] [Indexed: 12/30/2022] Open
Abstract
Background Plastic changes in the anterior cingulate cortex (ACC) are critical in the pathogenesis of pain hypersensitivity caused by injury to peripheral nerves. Cdh1, a co-activator subunit of anaphase-promoting complex/cyclosome (APC/C) regulates synaptic differentiation and transmission. Based on this, we hypothesised that the APC/C–Cdh1 played an important role in long-term plastic changes induced by neuropathic pain in ACC. Results We employed spared nerve injury (SNI) model in rat and found Cdh1 protein level in the ACC was down-regulated 3, 7 and 14 days after SNI surgery. We detected increase in c-Fos expression, numerical increase of organelles, swollen myelinated fibre and axon collapse of neuronal cells in the ACC of SNI rat. Additionally, AMPA receptor GluR1 subunit protein level was up-regulated on the membrane through a pathway that involves EphA4 mediated by APC/C–Cdh1, 3 and 7 days after SNI surgery. To confirm the effect of Cdh1 in neuropathic pain, Cdh1-expressing lentivirus was injected into the ACC of SNI rat. Intra-ACC treatment with Cdh1-expressing lentivirus vectors elevated Cdh1 levels, erased synaptic strengthening, as well as alleviating established mechanical allodynia in SNI rats. We also found Cdh1-expressing lentivirus normalised SNI-induced redistribution of AMPA receptor GluR1 subunit in ACC by regulating AMPA receptor trafficking. Conclusions These results provide evidence that Cdh1 in ACC synapses may offer a novel therapeutic strategy for treating chronic neuropathic pain.
Collapse
Affiliation(s)
- Wei Tan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wen-Long Yao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Rong Hu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - You-You Lv
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Li Wan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Chuan-Han Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Chang Zhu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
47
|
Chauvet S, Boonen M, Chevallet M, Jarvis L, Abebe A, Benharouga M, Faller P, Jadot M, Bouron A. The Na+/K+-ATPase and the amyloid-beta peptide aβ1-40 control the cellular distribution, abundance and activity of TRPC6 channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2957-65. [PMID: 26348127 DOI: 10.1016/j.bbamcr.2015.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 11/19/2022]
Abstract
The Na(+)/K(+)-ATPase interacts with the non-selective cation channels TRPC6 but the functional consequences of this association are unknown. Experiments performed with HEK cells over-expressing TRPC6 channels showed that inhibiting the activity of the Na(+)/K(+)-ATPase with ouabain reduced the amount of TRPC6 proteins and depressed Ca(2+) entry through TRPC6. This effect, not mimicked by membrane depolarization with KCl, was abolished by sucrose and bafilomycin-A, and was partially sensitive to the intracellular Ca(2+) chelator BAPTA/AM. Biotinylation and subcellular fractionation experiments showed that ouabain caused a multifaceted redistribution of TRPC6 to the plasma membrane and to an endo/lysosomal compartment where they were degraded. The amyloid beta peptide Aβ(1-40), another inhibitor of the Na(+)/K(+)-ATPase, but not the shorter peptide Aβ1-16, reduced TRPC6 protein levels and depressed TRPC6-mediated responses. In cortical neurons from embryonic mice, ouabain, veratridine (an opener of voltage-gated Na(+) channel), and Aβ(1-40) reduced TRPC6-mediated Ca(2+) responses whereas Aβ(1-16) was ineffective. Furthermore, when Aβ(1-40) was co-added together with zinc acetate it could no longer control TRPC6 activity. Altogether, this work shows the existence of a functional coupling between the Na(+)/K(+)-ATPase and TRPC6. It also suggests that the abundance, distribution and activity of TRPC6 can be regulated by cardiotonic steroids like ouabain and the naturally occurring peptide Aβ(1-40) which underlines the pathophysiological significance of these processes.
Collapse
Affiliation(s)
- Sylvain Chauvet
- Université Grenoble Alpes, F-38000 Grenoble, France; CNRS, F-38000 Grenoble, France; CEA, iRTSV-LCBM, F-38000 Grenoble, France
| | - Marielle Boonen
- URPhyM-Laboratoire de Chimie Physiologique, University of Namur, Belgium
| | - Mireille Chevallet
- Université Grenoble Alpes, F-38000 Grenoble, France; CNRS, F-38000 Grenoble, France; CEA, iRTSV-LCBM, F-38000 Grenoble, France
| | - Louis Jarvis
- Université Grenoble Alpes, F-38000 Grenoble, France; CNRS, F-38000 Grenoble, France; CEA, iRTSV-LCBM, F-38000 Grenoble, France
| | - Addis Abebe
- Université Grenoble Alpes, F-38000 Grenoble, France; CNRS, F-38000 Grenoble, France; CEA, iRTSV-LCBM, F-38000 Grenoble, France
| | - Mohamed Benharouga
- Université Grenoble Alpes, F-38000 Grenoble, France; CNRS, F-38000 Grenoble, France; CEA, iRTSV-LCBM, F-38000 Grenoble, France
| | - Peter Faller
- CNRS, Laboratoire de Chimie de Coordination, Toulouse, France
| | - Michel Jadot
- URPhyM-Laboratoire de Chimie Physiologique, University of Namur, Belgium
| | - Alexandre Bouron
- Université Grenoble Alpes, F-38000 Grenoble, France; CNRS, F-38000 Grenoble, France; CEA, iRTSV-LCBM, F-38000 Grenoble, France.
| |
Collapse
|
48
|
Shrivastava AN, Redeker V, Fritz N, Pieri L, Almeida LG, Spolidoro M, Liebmann T, Bousset L, Renner M, Léna C, Aperia A, Melki R, Triller A. α-synuclein assemblies sequester neuronal α3-Na+/K+-ATPase and impair Na+ gradient. EMBO J 2015; 34:2408-23. [PMID: 26323479 DOI: 10.15252/embj.201591397] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/21/2015] [Indexed: 11/09/2022] Open
Abstract
Extracellular α-synuclein (α-syn) assemblies can be up-taken by neurons; however, their interaction with the plasma membrane and proteins has not been studied specifically. Here we demonstrate that α-syn assemblies form clusters within the plasma membrane of neurons. Using a proteomic-based approach, we identify the α3-subunit of Na+/K+-ATPase (NKA) as a cell surface partner of α-syn assemblies. The interaction strength depended on the state of α-syn, fibrils being the strongest, oligomers weak, and monomers none. Mutations within the neuron-specific α3-subunit are linked to rapid-onset dystonia Parkinsonism (RDP) and alternating hemiplegia of childhood (AHC). We show that freely diffusing α3-NKA are trapped within α-syn clusters resulting in α3-NKA redistribution and formation of larger nanoclusters. This creates regions within the plasma membrane with reduced local densities of α3-NKA, thereby decreasing the efficiency of Na+ extrusion following stimulus. Thus, interactions of α3-NKA with extracellular α-syn assemblies reduce its pumping activity as its mutations in RDP/AHC.
Collapse
Affiliation(s)
- Amulya Nidhi Shrivastava
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
| | - Virginie Redeker
- Paris-Saclay Institute of Neuroscience CNRS, Gif-sur-Yvette, France
| | - Nicolas Fritz
- Department of Women and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Laura Pieri
- Paris-Saclay Institute of Neuroscience CNRS, Gif-sur-Yvette, France
| | - Leandro G Almeida
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
| | - Maria Spolidoro
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
| | - Thomas Liebmann
- Department of Women and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Luc Bousset
- Paris-Saclay Institute of Neuroscience CNRS, Gif-sur-Yvette, France
| | - Marianne Renner
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
| | - Clément Léna
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
| | - Anita Aperia
- Department of Women and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ronald Melki
- Paris-Saclay Institute of Neuroscience CNRS, Gif-sur-Yvette, France
| | - Antoine Triller
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
| |
Collapse
|
49
|
Rosa SG, Quines CB, da Rocha JT, Bortolatto CF, Duarte T, Nogueira CW. Antinociceptive action of diphenyl diselenide in the nociception induced by neonatal administration of monosodium glutamate in rats. Eur J Pharmacol 2015; 758:64-71. [DOI: 10.1016/j.ejphar.2015.03.060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/13/2015] [Accepted: 03/14/2015] [Indexed: 12/21/2022]
|
50
|
Quantitative assessment of single-cell whole genome amplification methods for detecting copy number variation using hippocampal neurons. Sci Rep 2015; 5:11415. [PMID: 26091148 PMCID: PMC4650676 DOI: 10.1038/srep11415] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 05/08/2015] [Indexed: 02/04/2023] Open
Abstract
Single-cell genomic analysis has grown rapidly in recent years and finds widespread applications in various fields of biology, including cancer biology, development, immunology, pre-implantation genetic diagnosis, and neurobiology. To date, the amplification bias, amplification uniformity and reproducibility of the three major single cell whole genome amplification methods (GenomePlex WGA4, MDA and MALBAC) have not been systematically investigated using mammalian cells. In this study, we amplified genomic DNA from individual hippocampal neurons using three single-cell DNA amplification methods, and sequenced them at shallow depth. We then systematically evaluated the GC-bias, reproducibility, and copy number variations among individual neurons. Our results showed that single-cell genome sequencing results obtained from the MALBAC and WGA4 methods are highly reproducible and have a high success rate. The MALBAC displays significant biases towards high GC content. We then attempted to correct the GC bias issue by developing a bioinformatics pipeline, which allows us to call CNVs in single cell sequencing data, and chromosome level and sub-chromosomal level CNVs among individual neurons can be detected. We also proposed a metric to determine the CNV detection limits. Overall, MALBAC and WGA4 have better performance than MDA in detecting CNVs.
Collapse
|