1
|
Burkart ME, Kurzke J, Jacobi R, Vera J, Ashcroft FM, Eilers J, Lippmann K. KATP channel mutation disrupts hippocampal network activity and nocturnal gamma shifts. Brain 2024; 147:4200-4212. [PMID: 38748482 DOI: 10.1093/brain/awae157] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/31/2024] [Accepted: 05/02/2024] [Indexed: 12/14/2024] Open
Abstract
ATP-sensitive potassium (KATP) channels couple cell metabolism to cellular electrical activity. Humans affected by severe activating mutations in KATP channels suffer from developmental delay, epilepsy and neonatal diabetes (DEND syndrome). While the aetiology of diabetes in DEND syndrome is well understood, the pathophysiology of the neurological symptoms remains unclear. We hypothesized that impaired activity of parvalbumin-positive interneurons (PV-INs) may result in seizures and cognitive problems. We found, by performing electrophysiological experiments, that expressing the DEND mutation Kir6.2-V59M selectively in mouse PV-INs reduced intrinsic gamma frequency preference and short-term depression as well as disturbed cognition-associated gamma oscillations and hippocampal sharp waves. Furthermore, the risk of seizures was increased and the day-night shift in gamma activity disrupted. Blocking KATP channels with tolbutamide partially rescued the network oscillations. The non-reversible part may, to some extent, result from observed altered PV-IN dendritic branching and PV-IN arrangement within CA1. In summary, PV-INs play a key role in DEND syndrome, and this provides a framework for establishing treatment options.
Collapse
Affiliation(s)
- Marie-Elisabeth Burkart
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| | - Josephine Kurzke
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| | - Robert Jacobi
- Department for Neurophysiology, Institute for Physiology, Julius-Maximilians-University Würzburg, Würzburg 97070, Germany
| | - Jorge Vera
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Frances M Ashcroft
- Henry Wellcome Centre for Gene Function, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Jens Eilers
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| | - Kristina Lippmann
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| |
Collapse
|
2
|
Lee J, Xue X, Au E, McIntyre WB, Asgariroozbehani R, Tseng GC, Papoulias M, Panganiban K, Agarwal SM, Mccullumsmith R, Freyberg Z, Logan RW, Hahn MK. Central insulin dysregulation in antipsychotic-naïve first-episode psychosis: In silico exploration of gene expression signatures. Psychiatry Res 2024; 331:115636. [PMID: 38104424 PMCID: PMC10984627 DOI: 10.1016/j.psychres.2023.115636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/18/2023] [Accepted: 11/25/2023] [Indexed: 12/19/2023]
Abstract
Antipsychotic drug (AP)-naïve first-episode psychosis (FEP) patients display premorbid cognitive dysfunctions and dysglycemia. Brain insulin resistance may link metabolic and cognitive disorders in humans. This suggests that central insulin dysregulation represents a component of the pathophysiology of psychosis spectrum disorders (PSDs). Nonetheless, the links between central insulin dysregulation, dysglycemia, and cognitive deficits in PSDs are poorly understood. We investigated whether AP-naïve FEP patients share overlapping brain gene expression signatures with central insulin perturbation (CIP) in rodent models. We systematically compiled and meta-analyzed peripheral transcriptomic datasets of AP-naïve FEP patients along with hypothalamic and hippocampal datasets of CIP rodent models to identify common transcriptomic signatures. The common signatures were used for pathway analysis and to identify potential drug treatments with discordant (reverse) signatures. AP-naïve FEP and CIP (hypothalamus and hippocampus) shared 111 and 346 common signatures respectively, which were associated with pathways related to inflammation, endoplasmic reticulum stress, and neuroplasticity. Twenty-two potential drug treatments were identified, including antidiabetic agents. The pathobiology of PSDs may include central insulin dysregulation, which contribute to dysglycemia and cognitive dysfunction independently of AP treatment. The identified treatments may be tested in early psychosis patients to determine if dysglycemia and cognitive deficits can be mitigated.
Collapse
Affiliation(s)
- Jiwon Lee
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | - Xiangning Xue
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States.
| | - Emily Au
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| | - William B McIntyre
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| | - Roshanak Asgariroozbehani
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | - George C Tseng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States.
| | - Maria Papoulias
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | - Kristoffer Panganiban
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | - Sri Mahavir Agarwal
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| | - Robert Mccullumsmith
- Department of Neurosciences, University of Toledo, Toledo, Ohio, United States; ProMedica, Toledo, Ohio, United States.
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States.
| | - Ryan W Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States; Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States.
| | - Margaret K Hahn
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Cognitive deficits and impaired hippocampal long-term potentiation in K ATP-induced DEND syndrome. Proc Natl Acad Sci U S A 2021; 118:2109721118. [PMID: 34732576 PMCID: PMC8609313 DOI: 10.1073/pnas.2109721118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
ATP-sensitive potassium (KATP) gain-of-function (GOF) mutations cause neonatal diabetes, with some individuals exhibiting developmental delay, epilepsy, and neonatal diabetes (DEND) syndrome. Mice expressing KATP-GOF mutations pan-neuronally (nKATP-GOF) demonstrated sensorimotor and cognitive deficits, whereas hippocampus-specific hKATP-GOF mice exhibited mostly learning and memory deficiencies. Both nKATP-GOF and hKATP-GOF mice showed altered neuronal excitability and reduced hippocampal long-term potentiation (LTP). Sulfonylurea therapy, which inhibits KATP, mildly improved sensorimotor but not cognitive deficits in KATP-GOF mice. Mice expressing KATP-GOF mutations in pancreatic β-cells developed severe diabetes but did not show learning and memory deficits, suggesting neuronal KATP-GOF as promoting these features. These findings suggest a possible origin of cognitive dysfunction in DEND and the need for novel drugs to treat neurological features induced by neuronal KATP-GOF.
Collapse
|
4
|
Akyuz E, Villa C, Beker M, Elibol B. Unraveling the Role of Inwardly Rectifying Potassium Channels in the Hippocampus of an Aβ (1-42)-Infused Rat Model of Alzheimer's Disease. Biomedicines 2020; 8:biomedicines8030058. [PMID: 32183098 PMCID: PMC7148495 DOI: 10.3390/biomedicines8030058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder with a complex etiology and characterized by cognitive deficits and memory loss. The pathogenesis of AD is not yet completely elucidated, and no curative treatment is currently available. Inwardly rectifying potassium (Kir) channels are important for playing a key role in maintaining the resting membrane potential and controlling cell excitability, being largely expressed in both excitable and non-excitable tissues, including neurons. Accordingly, the aim of the study is to investigate the role of neuronal Kir channels in AD pathophysiology. The mRNA and protein levels of neuronal Kir2.1, Kir3.1, and Kir6.2 were evaluated by real-time PCR and Western blot analysis from the hippocampus of an amyloid-β(Aβ)(1-42)-infused rat model of AD. Extracellular deposition of Aβ was confirmed by both histological Congo red staining and immunofluorescence analysis. Significant decreased mRNA and protein levels of Kir2.1 and Kir6.2 channels were observed in the rat model of AD, whereas no differences were found in Kir3.1 channel levels as compared with controls. Our results provide in vivo evidence that Aβ can modulate the expression of these channels, which may represent novel potential therapeutic targets in the treatment of AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, Faculty of Medicine, Yozgat Bozok University, Yozgat 66100, Turkey
- Correspondence: (E.A.); (C.V.)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Correspondence: (E.A.); (C.V.)
| | - Merve Beker
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey; (M.B.); (B.E.)
| | - Birsen Elibol
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey; (M.B.); (B.E.)
| |
Collapse
|
5
|
Jayanthi S, Torres OV, Ladenheim B, Cadet JL. A Single Prior Injection of Methamphetamine Enhances Methamphetamine Self-Administration (SA) and Blocks SA-Induced Changes in DNA Methylation and mRNA Expression of Potassium Channels in the Rat Nucleus Accumbens. Mol Neurobiol 2019; 57:1459-1472. [PMID: 31758400 PMCID: PMC7060962 DOI: 10.1007/s12035-019-01830-3] [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: 08/06/2019] [Accepted: 11/01/2019] [Indexed: 12/27/2022]
Abstract
The transition from occasional to escalated psychostimulant use is accelerated by prior drug exposure. These behavioral observations may be related to long-lasting transcriptional and/or epigenetic changes induced by the drug pre-exposure. Herein, we investigated if a single methamphetamine (METH) injection would enhance METH self-administration (SA) and impact any METH SA-induced epigenetic or transcriptional alterations. We thus injected a single METH dose (10 mg/kg) or saline to rats before training them to self-administer METH or saline. There were three experimental groups in SA experiments: (1) a single saline injection followed by saline SA (SS); (2) a single saline injection followed by METH SA (SM); and (3) a single METH injection followed by METH SA (MM). METH-pretreated rats escalated METH SA earlier and took more METH than saline-pretreated animals. Both groups showed similar incubation of cue-induced METH craving. Because compulsive METH takers and METH-abstinent rats show differences in potassium (K+) channel mRNA levels in their nucleus accumbens (NAc), we wondered if K+ channel expression might also help to distinguish between SM and MM groups. We found increases in mRNA and protein expression of shaker-related voltage-gated K+ channels (Kv1: Kcna1, Kcna3, and Kcna6) and calcium-activated K+ channels (Kcnn1) in the SM compared to MM rats. SM rats also showed decreased DNA methylation at the CpG-rich sites near the promoter region of Kcna1, Kcna3 and Kcnn1 genes in comparison to MM rats. Together, these results provide additional evidence for potentially using K+ channels as therapeutic targets against METH use disorder.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Oscar V Torres
- Department of Behavioral Sciences, San Diego Mesa College, San Diego, CA, USA
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA.
| |
Collapse
|
6
|
Kharade SV, Sanchez-Andres JV, Fulton MG, Shelton EL, Blobaum AL, Engers DW, Hofmann CS, Dadi PK, Lantier L, Jacobson DA, Lindsley CW, Denton JS. Structure-Activity Relationships, Pharmacokinetics, and Pharmacodynamics of the Kir6.2/SUR1-Specific Channel Opener VU0071063. J Pharmacol Exp Ther 2019; 370:350-359. [PMID: 31201216 PMCID: PMC6691189 DOI: 10.1124/jpet.119.257204] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/12/2019] [Indexed: 01/14/2023] Open
Abstract
Glucose-stimulated insulin secretion from pancreatic β-cells is controlled by ATP-regulated potassium (KATP) channels composed of Kir6.2 and sulfonylurea receptor 1 (SUR1) subunits. The KATP channel-opener diazoxide is FDA-approved for treating hyperinsulinism and hypoglycemia but suffers from off-target effects on vascular KATP channels and other ion channels. The development of more specific openers would provide critically needed tool compounds for probing the therapeutic potential of Kir6.2/SUR1 activation. Here, we characterize a novel scaffold activator of Kir6.2/SUR1 that our group recently discovered in a high-throughput screen. Optimization efforts with medicinal chemistry identified key structural elements that are essential for VU0071063-dependent opening of Kir6.2/SUR1. VU0071063 has no effects on heterologously expressed Kir6.1/SUR2B channels or ductus arteriole tone, indicating it does not open vascular KATP channels. VU0071063 induces hyperpolarization of β-cell membrane potential and inhibits insulin secretion more potently than diazoxide. VU0071063 exhibits metabolic and pharmacokinetic properties that are favorable for an in vivo probe and is brain penetrant. Administration of VU0071063 inhibits glucose-stimulated insulin secretion and glucose-lowering in mice. Taken together, these studies indicate that VU0071063 is a more potent and specific opener of Kir6.2/SUR1 than diazoxide and should be useful as an in vitro and in vivo tool compound for investigating the therapeutic potential of Kir6.2/SUR1 expressed in the pancreas and brain.
Collapse
Affiliation(s)
- Sujay V Kharade
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Juan Vicente Sanchez-Andres
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Mark G Fulton
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Elaine L Shelton
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Anna L Blobaum
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Darren W Engers
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Christopher S Hofmann
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Prasanna K Dadi
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Louise Lantier
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - David A Jacobson
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Craig W Lindsley
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| | - Jerod S Denton
- Departments of Anesthesiology (S.V.K., J.S.D.) and Pediatrics (E.L.S.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, Jaume I University, Castellon de la Plana, Spain (J.V.S.-A.); Departments of Chemistry (M.G.F., C.W.L.), Pharmacology (M.G.F., A.L.B., D.W.E., C.S.H., C.W.L., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.), and Mouse Metabolic Phenotyping Core (L.L.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Center for Neuroscience Drug Discovery, Franklin, Tennessee (D.W.E., A.L.B., C.W.L.)
| |
Collapse
|
7
|
Jojo GM, Kuppusamy G, Selvaraj K, Baruah UK. Prospective of managing impaired brain insulin signalling in late onset Alzheimers disease with excisting diabetic drugs. J Diabetes Metab Disord 2019; 18:229-242. [PMID: 31275894 DOI: 10.1007/s40200-019-00405-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/12/2019] [Indexed: 12/25/2022]
Abstract
Late onset Alzheimer's disease (AD) is the most common cause of dementia among elderly. The exact cause of the disease is until now unknown and there is no complete cure for the disease. Growing evidence suggest that AD is a metabolic disorder associated with impairment in brain insulin signalling. These findings enriched the scope for the repurposing of diabetic drugs in AD management. Even though many of these drugs are moving in a positive direction in the ongoing clinical studies, the extent of the success has seen to influence by several properties of these drugs since they were originally designed to manage the peripheral insulin resistance. In depth understandings of these properties is hence highly significant to optimise the use of diabetic drugs in the clinical management of AD; which is the primary aim of the present review article.
Collapse
Affiliation(s)
- Gifty M Jojo
- Department of Pharmaceutics, JSS College of pharmacy, Ootacamund, JSS Academy of Higher Education & Research, Mysore, India
| | - Gowthamarajan Kuppusamy
- Department of Pharmaceutics, JSS College of pharmacy, Ootacamund, JSS Academy of Higher Education & Research, Mysore, India
| | - Kousalya Selvaraj
- Department of Pharmaceutics, JSS College of pharmacy, Ootacamund, JSS Academy of Higher Education & Research, Mysore, India
| | - Uday Krishna Baruah
- Department of Pharmaceutics, JSS College of pharmacy, Ootacamund, JSS Academy of Higher Education & Research, Mysore, India
| |
Collapse
|
8
|
Possible involvement of monoamine neurons in the emotional abnormality in Kir6.2-deficient mice. Physiol Behav 2018; 188:251-261. [PMID: 29432787 DOI: 10.1016/j.physbeh.2018.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 02/07/2018] [Accepted: 02/07/2018] [Indexed: 11/24/2022]
Abstract
ATP-sensitive potassium (KATP) channels consist of two structurally different subunits: a pore-forming subunit of the Kir6.0-family (Kir6.1 or Kir6.2) and a regulatory sulfonylurea receptor subunit (SUR1, SUR2A or SUR2B). Although Kir6.2 is widely distributed in the brain, the mechanisms that underlie the impact of Kir6.2 on emotional behavior are not yet fully understood. To clarify the role of Kir6.2 in emotional behavior, in the present study, we investigated the behavioral characteristics of Kir6.2-knockout (Kir6.2-/-) mice. Kir6.2-/- mice showed impaired general behavior in a locomotor activity test and open field test. In addition, anxiety-like behavior was observed in the open field test, elevated plus-maze test and light-dark test. In particular, excessive anxiety-like behavior was observed in female Kir6.2-/- mice. Moreover, we investigated whether Kir6.2 is expressed on monoamine neurons in the brain. Immunohistochemical studies showed that Kir6.2 was co-localized with tryptophan hydroxylase (TPH), a marker of serotonergic neurons, in dorsal raphe nuclei. Kir6.2 was also co-localized with tyrosine hydroxylase (TH), a marker of dopaminergic/noradrenergic neurons, in the ventral tegmental area and locus coeruleus. Next, we checked the protein levels of TH and TPH in the midbrain. Interestingly, TPH expression was significantly elevated in female Kir6.2-/- mice. These results suggest that Kir6.2 in monoamine neurons, especially serotonergic neurons, could play a key role in emotional behavior.
Collapse
|
9
|
Salgado-Puga K, Rodríguez-Colorado J, Prado-Alcalá RA, Peña-Ortega F. Subclinical Doses of ATP-Sensitive Potassium Channel Modulators Prevent Alterations in Memory and Synaptic Plasticity Induced by Amyloid-β. J Alzheimers Dis 2018; 57:205-226. [PMID: 28222502 DOI: 10.3233/jad-160543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In addition to coupling cell metabolism and excitability, ATP-sensitive potassium channels (KATP) are involved in neural function and plasticity. Moreover, alterations in KATP activity and expression have been observed in Alzheimer's disease (AD) and during amyloid-β (Aβ)-induced pathology. Thus, we tested whether KATP modulators can influence Aβ-induced deleterious effects on memory, hippocampal network function, and plasticity. We found that treating animals with subclinical doses (those that did not change glycemia) of a KATP blocker (Tolbutamide) or a KATP opener (Diazoxide) differentially restrained Aβ-induced memory deficit, hippocampal network activity inhibition, and long-term synaptic plasticity unbalance (i.e., inhibition of LTP and promotion of LTD). We found that the protective effect of Tolbutamide against Aβ-induced memory deficit was strong and correlated with the reestablishment of synaptic plasticity balance, whereas Diazoxide treatment produced a mild protection against Aβ-induced memory deficit, which was not related to a complete reestablishment of synaptic plasticity balance. Interestingly, treatment with both KATP modulators renders the hippocampus resistant to Aβ-induced inhibition of hippocampal network activity. These findings indicate that KATP are involved in Aβ-induced pathology and they heighten the potential role of KATP modulation as a plausible therapeutic strategy against AD.
Collapse
Affiliation(s)
- Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Javier Rodríguez-Colorado
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Roberto A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| |
Collapse
|
10
|
Moriguchi S, Ishizuka T, Yabuki Y, Shioda N, Sasaki Y, Tagashira H, Yawo H, Yeh JZ, Sakagami H, Narahashi T, Fukunaga K. Blockade of the K ATP channel Kir6.2 by memantine represents a novel mechanism relevant to Alzheimer's disease therapy. Mol Psychiatry 2018; 23:211-221. [PMID: 27777420 DOI: 10.1038/mp.2016.187] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 08/11/2016] [Accepted: 08/22/2016] [Indexed: 01/09/2023]
Abstract
Here, we report a novel target of the drug memantine, ATP-sensitive K+ (KATP) channels, potentially relevant to memory improvement. We confirmed that memantine antagonizes memory impairment in Alzheimer's model APP23 mice. Memantine increased CaMKII activity in the APP23 mouse hippocampus, and memantine-induced enhancement of hippocampal long-term potentiation (LTP) and CaMKII activity was totally abolished by treatment with pinacidil, a specific opener of KATP channels. Memantine also inhibited Kir6.1 and Kir6.2 KATP channels and elevated intracellular Ca2+ concentrations in neuro2A cells overexpressing Kir6.1 or Kir6.2. Kir6.2 was preferentially expressed at postsynaptic regions of hippocampal neurons, whereas Kir6.1 was predominant in dendrites and cell bodies of pyramidal neurons. Finally, we confirmed that Kir6.2 mutant mice exhibit severe memory deficits and impaired hippocampal LTP, impairments that cannot be rescued by memantine administration. Altogether, our studies show that memantine modulates Kir6.2 activity, and that the Kir6.2 channel is a novel target for therapeutics to improve memory impairment in Alzheimer disease patients.
Collapse
Affiliation(s)
- S Moriguchi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - T Ishizuka
- Department of Developmental Biology and Neuroscience, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Y Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - N Shioda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Y Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - H Tagashira
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - H Yawo
- Department of Developmental Biology and Neuroscience, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - J Z Yeh
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - H Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - T Narahashi
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - K Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
11
|
Hascup KN, Lynn MK, Fitzgerald PJ, Randall S, Kopchick JJ, Boger HA, Bartke A, Hascup ER. Enhanced Cognition and Hypoglutamatergic Signaling in a Growth Hormone Receptor Knockout Mouse Model of Successful Aging. J Gerontol A Biol Sci Med Sci 2017; 72:329-337. [PMID: 27208894 DOI: 10.1093/gerona/glw088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 04/25/2016] [Indexed: 01/19/2023] Open
Abstract
Growth hormone receptor knockout (GHR-KO) mice are long lived with improved health span, making this an excellent model system for understanding biochemical mechanisms important to cognitive reserve. The purpose of the present study was to elucidate differences in cognition and glutamatergic dynamics between aged (20- to 24-month-old) GHR-KO and littermate controls. Glutamate plays a critical role in hippocampal learning and memory and is implicated in several neurodegenerative disorders, including Alzheimer's disease. Spatial learning and memory were assessed using the Morris water maze (MWM), whereas independent dentate gyrus (DG), CA3, and CA1 basal glutamate, release, and uptake measurements were conducted in isoflurane anesthetized mice utilizing an enzyme-based microelectrode array (MEA) coupled with constant potential amperometry. These MEAs have high temporal and low spatial resolution while causing minimal damage to the surrounding parenchyma. Littermate controls performed worse on the memory portion of the MWM behavioral task and had elevated DG, CA3, and CA1 basal glutamate and stimulus-evoked release compared with age-matched GHR-KO mice. CA3 basal glutamate negatively correlated with MWM performance. These results support glutamatergic regulation in learning and memory and may have implications for therapeutic targets to delay the onset of, or reduce cognitive decline, in Alzheimer's disease.
Collapse
Affiliation(s)
- Kevin N Hascup
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield
| | - Mary K Lynn
- Department of Neuroscience, Medical University of South Carolina, Charleston
| | - Patrick J Fitzgerald
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield
| | - Shari Randall
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield
| | - John J Kopchick
- Edison Biotechnology Institute, Department of Biomedical Sciences, Ohio University, Athens
| | - Heather A Boger
- Department of Neuroscience, Medical University of South Carolina, Charleston.,Center on Aging, Medical University of South Carolina, Charleston
| | | | - Erin R Hascup
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield
| |
Collapse
|
12
|
Griffith CM, Xie MX, Qiu WY, Sharp AA, Ma C, Pan A, Yan XX, Patrylo PR. Aberrant expression of the pore-forming KATP channel subunit Kir6.2 in hippocampal reactive astrocytes in the 3xTg-AD mouse model and human Alzheimer’s disease. Neuroscience 2016; 336:81-101. [DOI: 10.1016/j.neuroscience.2016.08.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 08/15/2016] [Accepted: 08/20/2016] [Indexed: 12/21/2022]
|
13
|
Liu R, Wang H, Xu B, Chen W, Turlova E, Dong N, Sun CLF, Lu Y, Fu H, Shi R, Barszczyk A, Yang D, Jin T, Mannucci E, Feng ZP, Sun HS. Cerebrovascular Safety of Sulfonylureas: The Role of KATP Channels in Neuroprotection and the Risk of Stroke in Patients With Type 2 Diabetes. Diabetes 2016; 65:2795-809. [PMID: 27207539 DOI: 10.2337/db15-1737] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/11/2016] [Indexed: 11/13/2022]
Abstract
Sulfonylureas are ATP-sensitive potassium (KATP) channel blockers commonly used in the treatment of type 2 diabetes mellitus (T2DM). Activation of KATP channels plays a neuroprotective role in ischemia; thus, whether sulfonylureas affect the outcomes of stroke in patients with T2DM needs to be further studied. In our study, streptozotocin (STZ)-induced diabetic mice subjected to transient middle cerebral artery occlusion (MCAO) showed larger areas of brain damage and poorer behavioral outcomes. Blocking the KATP channel by tolbutamide increased neuronal injury induced by oxygen-glucose deprivation (OGD) in vitro and permanent MCAO (pMCAO) in vivo. Activating the KATP channel by diazoxide reduced the effects of both the OGD and pMCAO. Western blot analysis in STZ mouse brains indicated an early increase in protein levels of N-methyl-d-aspartate receptor 2B and postsynaptic density protein-95, followed by a decrease in phosphorylation of glycogen synthase kinase 3β. Our systematic meta-analysis indicated that patients with T2DM treated with sulfonylureas had a higher odds ratio for stroke morbidity than those who received comparator drugs. Taken together, these results suggest that sulfonylurea treatment in patients with T2DM may inhibit the neuroprotective effects of KATP channels and increase the risk of stroke.
Collapse
Affiliation(s)
- Rui Liu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Haitao Wang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Baofeng Xu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Wenliang Chen
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ekaterina Turlova
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Nan Dong
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Christopher L F Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yangqingqin Lu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hanhui Fu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ranran Shi
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Barszczyk
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dongzi Yang
- Department of Obstetrics and Gynecology, Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tianru Jin
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Edoardo Mannucci
- Diabetology, Careggi Hospital, University of Florence, Florence, Italy
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
The shifting landscape of KATP channelopathies and the need for 'sharper' therapeutics. Future Med Chem 2016; 8:789-802. [PMID: 27161588 DOI: 10.4155/fmc-2016-0005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
ATP-sensitive potassium (KATP) channels play fundamental roles in the regulation of endocrine, neural and cardiovascular function. Small-molecule inhibitors (e.g., sulfonylurea drugs) or activators (e.g., diazoxide) acting on SUR1 or SUR2 have been used clinically for decades to manage the inappropriate secretion of insulin in patients with Type 2 diabetes, hyperinsulinism and intractable hypertension. More recently, the discovery of rare disease-causing mutations in KATP channel-encoding genes has highlighted the need for new therapeutics for the treatment of certain forms of neonatal diabetes mellitus, congenital hyperinsulinism and Cantu syndrome. Here, we provide a high-level overview of the pathophysiology of these diseases and discuss the development of a flexible high-throughput screening platform to enable the development of new classes of KATP channel modulators.
Collapse
|
15
|
Pál I, Kardos J, Dobolyi Á, Héja L. Appearance of fast astrocytic component in voltage-sensitive dye imaging of neural activity. Mol Brain 2015; 8:35. [PMID: 26043770 PMCID: PMC4455916 DOI: 10.1186/s13041-015-0127-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 05/24/2015] [Indexed: 12/21/2022] Open
Abstract
Background Voltage-sensitive dye (VSD) imaging and intrinsic optical signals (IOS) are widely used methods for monitoring spatiotemporal neural activity in extensive networks. In spite of that, identification of their major cellular and molecular components has not been concluded so far. Results We addressed these issues by imaging spatiotemporal spreading of IOS and VSD transients initiated by Schaffer collateral stimulation in rat hippocampal slices with temporal resolution comparable to standard field potential recordings using a 464-element photodiode array. By exploring the potential neuronal and astroglial molecular players in VSD and IOS generation, we identified multiple astrocytic mechanisms that significantly contribute to the VSD signal, in addition to the expected neuronal targets. Glutamate clearance through the astroglial glutamate transporter EAAT2 has been shown to be a significant player in VSD generation within a very short (<5 ms) time-scale, indicating that astrocytes do contribute to the development of spatiotemporal VSD transients previously thought to be essentially neuronal. In addition, non-specific anion channels, astroglial K+ clearance through Kir4.1 channel and astroglial Na+/K+ ATPase also contribute to IOS and VSD transients. Conclusion VSD imaging cannot be considered as a spatially extended field potential measurement with predominantly neuronal origin, instead it also reflects a fast communication between neurons and astrocytes.
Collapse
Affiliation(s)
- Ildikó Pál
- Group of Functional Pharmacology, Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117, Budapest, Hungary.
| | - Julianna Kardos
- Group of Functional Pharmacology, Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117, Budapest, Hungary.
| | - Árpád Dobolyi
- MTA-ELTE-NAP B Laboratory of Molecular and Systems Neurobiology, H-1117, Budapest, Hungary. .,Department of Anatomy, Human Brain Tissue Bank, Semmelweis University, H-1450, Budapest, Hungary.
| | - László Héja
- Group of Functional Pharmacology, Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117, Budapest, Hungary.
| |
Collapse
|
16
|
Kim DY, Abdelwahab MG, Lee SH, O’Neill D, Thompson RJ, Duff HJ, Sullivan PG, Rho JM. Ketones prevent oxidative impairment of hippocampal synaptic integrity through KATP channels. PLoS One 2015; 10:e0119316. [PMID: 25848768 PMCID: PMC4388385 DOI: 10.1371/journal.pone.0119316] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 01/29/2015] [Indexed: 12/31/2022] Open
Abstract
Dietary and metabolic therapies are increasingly being considered for a variety of neurological disorders, based in part on growing evidence for the neuroprotective properties of the ketogenic diet (KD) and ketones. Earlier, we demonstrated that ketones afford hippocampal synaptic protection against exogenous oxidative stress, but the mechanisms underlying these actions remain unclear. Recent studies have shown that ketones may modulate neuronal firing through interactions with ATP-sensitive potassium (KATP) channels. Here, we used a combination of electrophysiological, pharmacological, and biochemical assays to determine whether hippocampal synaptic protection by ketones is a consequence of KATP channel activation. Ketones dose-dependently reversed oxidative impairment of hippocampal synaptic integrity, neuronal viability, and bioenergetic capacity, and this action was mirrored by the KATP channel activator diazoxide. Inhibition of KATP channels reversed ketone-evoked hippocampal protection, and genetic ablation of the inwardly rectifying K+ channel subunit Kir6.2, a critical component of KATP channels, partially negated the synaptic protection afforded by ketones. This partial protection was completely reversed by co-application of the KATP blocker, 5-hydoxydecanoate (5HD). We conclude that, under conditions of oxidative injury, ketones induce synaptic protection in part through activation of KATP channels.
Collapse
Affiliation(s)
- Do Young Kim
- Departments of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, Arizona, United States of America
- * E-mail:
| | - Mohammed G. Abdelwahab
- Departments of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, Arizona, United States of America
| | - Soo Han Lee
- Departments of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, Arizona, United States of America
| | - Derek O’Neill
- Departments of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, Arizona, United States of America
| | - Roger J. Thompson
- Departments of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Henry J. Duff
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jong M. Rho
- Departments of Pediatrics and Clinical Neurosciences, Alberta Children’s Hospital, Calgary, Alberta, Canada
| |
Collapse
|
17
|
Meloni EG, Gillis TE, Manoukian J, Kaufman MJ. Xenon impairs reconsolidation of fear memories in a rat model of post-traumatic stress disorder (PTSD). PLoS One 2014; 9:e106189. [PMID: 25162644 PMCID: PMC4146606 DOI: 10.1371/journal.pone.0106189] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/29/2014] [Indexed: 12/19/2022] Open
Abstract
Xenon (Xe) is a noble gas that has been developed for use in people as an inhalational anesthestic and a diagnostic imaging agent. Xe inhibits glutamatergic N-methyl-D-aspartate (NMDA) receptors involved in learning and memory and can affect synaptic plasticity in the amygdala and hippocampus, two brain areas known to play a role in fear conditioning models of post-traumatic stress disorder (PTSD). Because glutamate receptors also have been shown to play a role in fear memory reconsolidation--a state in which recalled memories become susceptible to modification--we examined whether Xe administered after fear memory reactivation could affect subsequent expression of fear-like behavior (freezing) in rats. Male Sprague-Dawley rats were trained for contextual and cued fear conditioning and the effects of inhaled Xe (25%, 1 hr) on fear memory reconsolidation were tested using conditioned freezing measured days or weeks after reactivation/Xe administration. Xe administration immediately after fear memory reactivation significantly reduced conditioned freezing when tested 48 h, 96 h or 18 d after reactivation/Xe administration. Xe did not affect freezing when treatment was delayed until 2 h after reactivation or when administered in the absence of fear memory reactivation. These data suggest that Xe substantially and persistently inhibits memory reconsolidation in a reactivation and time-dependent manner, that it could be used as a new research tool to characterize reconsolidation and other memory processes, and that it could be developed to treat people with PTSD and other disorders related to emotional memory.
Collapse
MESH Headings
- Amygdala/drug effects
- Animals
- Conditioning, Classical/drug effects
- Conditioning, Classical/physiology
- Cues
- Disease Models, Animal
- Emotions/physiology
- Extinction, Psychological/drug effects
- Fear/drug effects
- Fear/psychology
- Freezing Reaction, Cataleptic/drug effects
- Freezing Reaction, Cataleptic/physiology
- Hippocampus/drug effects
- Male
- Memory/drug effects
- Memory/physiology
- Rats
- Rats, Sprague-Dawley
- Stress Disorders, Post-Traumatic/drug therapy
- Stress Disorders, Post-Traumatic/physiopathology
- Stress Disorders, Post-Traumatic/psychology
- Tranquilizing Agents/pharmacology
- Xenon/pharmacology
Collapse
Affiliation(s)
- Edward G. Meloni
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, Massachusetts, United States of America
| | - Timothy E. Gillis
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, Massachusetts, United States of America
| | - Jasmine Manoukian
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, Massachusetts, United States of America
| | - Marc J. Kaufman
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, Massachusetts, United States of America
| |
Collapse
|
18
|
de Araujo IE. Contextual fear conditioning: connecting brain glucose sensing and hippocampal-dependent memories. Biol Psychiatry 2014; 75:834-5. [PMID: 24837619 DOI: 10.1016/j.biopsych.2014.03.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Ivan E de Araujo
- John B Pierce Laboratory and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
19
|
Gupta S, Sharma B. Protective effects of phosphodiesterase-1 (PDE1) and ATP sensitive potassium (KATP) channel modulators against 3-nitropropionic acid induced behavioral and biochemical toxicities in experimental Huntington׳s disease. Eur J Pharmacol 2014; 732:111-22. [DOI: 10.1016/j.ejphar.2014.03.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/15/2014] [Accepted: 03/24/2014] [Indexed: 12/16/2022]
|
20
|
Zhang MZ, Zhou ZZ, Yuan X, Cheng YF, Bi BT, Gong MF, Chen YP, Xu JP. Chlorbipram: A novel PDE4 inhibitor with improved safety as a potential antidepressant and cognitive enhancer. Eur J Pharmacol 2013; 721:56-63. [DOI: 10.1016/j.ejphar.2013.09.055] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 09/23/2013] [Accepted: 09/26/2013] [Indexed: 12/23/2022]
|
21
|
Huang CW, Lai MC, Cheng JT, Tsai JJ, Huang CC, Wu SN. Pregabalin attenuates excitotoxicity in diabetes. PLoS One 2013; 8:e65154. [PMID: 23785408 PMCID: PMC3681790 DOI: 10.1371/journal.pone.0065154] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 04/22/2013] [Indexed: 12/15/2022] Open
Abstract
Diabetes can exacerbate seizures and worsen seizure-related brain damage. In the present study, we aimed to determine whether the standard antiepileptic drug pregabalin (PGB) protects against pilocarpine-induced seizures and excitotoxicity in diabetes. Adult male Sprague-Dawley rats were divided into either a streptozotocin (STZ)-induced diabetes group or a normal saline (NS) group. Both groups were further divided into subgroups that were treated intravenously with either PGB (15 mg/kg) or a vehicle; all groups were treated with subcutaneous pilocarpine (60 mg/kg) to induce seizures. To evaluate spontaneous recurrent seizures (SRS), PGB-pretreated rats were fed rat chow containing oral PGB (450 mg) for 28 consecutive days; vehicle-pretreated rats were fed regular chow. SRS frequency was monitored for 2 weeks from post-status epilepticus day 15. We evaluated both acute neuronal loss and chronic mossy fiber sprouting in the CA3 area. In addition, we performed patch clamp recordings to study evoked excitatory postsynaptic currents (eEPSCs) in hippocampal CA1 neurons for both vehicle-treated rats with SRS. Finally, we used an RNA interference knockdown method for Kir6.2 in a hippocampal cell line to evaluate PGB's effects in the presence of high-dose ATP. We found that compared to vehicle-treated rats, PGB-treated rats showed less severe acute seizure activity, reduced acute neuronal loss, and chronic mossy fiber sprouting. In the vehicle-treated STZ rats, eEPSC amplitude was significantly lower after PGB administration, but glibenclamide reversed this effect. The RNA interference study confirmed that PGB could counteract the ATP-sensitive potassium channel (KATP)-closing effect of high-dose ATP. By opening KATP, PGB protects against neuronal excitotoxicity, and is therefore a potential antiepileptogenic in diabetes. These findings might help develop a clinical algorithm for treating patients with epilepsy and comorbid metabolic disorders.
Collapse
Affiliation(s)
- Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Chi Lai
- Department of Pediatrics, Chi-Mei Foundation Medical Center, Tainan City, Taiwan
| | - Juei-Tang Cheng
- Department of Pharmacology, National Cheng Kung University, Tainan, Taiwan
| | - Jing-Jane Tsai
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chao-Ching Huang
- Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
22
|
Mollajew R, Toloe J, Mironov SL. Single KATP channel opening in response to stimulation of AMPA/kainate receptors is mediated by Na+ accumulation and submembrane ATP and ADP changes. J Physiol 2013; 591:2593-609. [PMID: 23507878 DOI: 10.1113/jphysiol.2012.248369] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Excessive stimulation of glutamatergic receptors (GluRs) can overexcite neurons. This can be dampened by KATP channels linking metabolic and neuronal activities, but the cross-talk has not yet been examined on the single channel level. In the brainstem and hippocampal neurons, GluR agonists augmented the open state probability (Popen) of KATP channels with relative efficacy: kainate AMPA > NMDA > t-ACPD. Inhibition of calcium influx and chelation of intracellular calcium did not modify the effects. Kainate did not augment production of reactive oxygen species measured with roGFP1. H2O2 slightly increased Popen, but GluR effects were not modified. GluR actions were abolished in Na(+)-free solutions and after blockade of Na(+)-K(+)-ATPase. KATP channels in open-cell patch-clamp measurements were inhibited by ATP, stimulated by ADP, and kainate was effective only in the presence of ATP. GluR stimulation enhanced ATP consumption that decreased submembrane ATP levels, whereas metabolic poisoning diminished bulk ATP. Modelling showed strong ATP depletion and ADP accumulation near the membrane, and both effects contributed to Popen increases after GluR stimulation. Kainate and hypoxia activated KATP channels in the functional brainstem slices. Inhibition of aerobic ATP production and GluR stimulation were about equally effective in KATP channel opening during hypoxia. Induction of seizure-like activity in hippocampal slices with Mg(2+)-free solutions was accompanied by ATP decrease and KATP channel opening. We propose that KATP channels and GluRs are functionally coupled that can regulate long-lasting changes of neuronal activity in the CNS neurons.
Collapse
Affiliation(s)
- R Mollajew
- DFG-Center of Molecular Physiology of the Brain, Institute of Neuro- and Sensory Physiology, Georg-August-University, Göttingen 37073, Germany
| | | | | |
Collapse
|
23
|
Morris G, Maes M. Increased nuclear factor-κB and loss of p53 are key mechanisms in Myalgic Encephalomyelitis/chronic fatigue syndrome (ME/CFS). Med Hypotheses 2012; 79:607-13. [PMID: 22951418 DOI: 10.1016/j.mehy.2012.07.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 07/14/2012] [Accepted: 07/31/2012] [Indexed: 12/18/2022]
Abstract
Fukuda's criteria are adequate to make a distinction between Myalgic Encephalomyelitis/chronic fatigue syndrome (ME/CFS) and chronic fatigue (CF), but ME/CFS patients should be subdivided into those with (termed ME) and without (termed CFS) post exertional malaise [Maes et al. 2012]. ME/CFS is considered to be a neuro-immune disease. ME/CFS is characterized by activated immuno-inflammatory pathways, including increased levels of pro-inflammatory cytokines, nuclear factor κB (NF-κB) and aberrations in mitochondrial functions, including lowered ATP. These processes may explain typical symptoms of ME/CFS, e.g. fatigue, malaise, hyperalgesia, and neurologic and autonomic symptoms. Here we hypothesize that increased NF-κB together with a loss of p53 are key phenomena in ME/CFS that further explain ME/CFS symptoms, such as fatigue and neurocognitive dysfunction, and explain ME symptoms, such as post-exertional malaise following mental and physical activities. Inactivation of p53 impairs aerobic mitochondrial functions and causes greater dependence on anaerobic glycolysis, elevates lactate levels, reduces mitochondrial density in skeletal muscle and reduces endurance during physical exercise. Lowered p53 and increased NF-κB are associated with elevated reactive oxygen species. Increased NF-κB induces the production of pro-inflammatory cytokines, which increase glycolysis and further compromise mitochondrial functions. All these factors together may contribute to mitochondrial exhaustion and indicate that the demand for extra ATP upon the commencement of increased activity cannot be met. In conditions of chronic inflammation and oxidative stress, high NF-κB and low p53 may conspire to promote neuron and glial cell survival at a price of severely compromised metabolic brain function. Future research should examine p53 signaling in ME/CFS.
Collapse
|
24
|
Towards therapeutic applications of arthropod venom k(+)-channel blockers in CNS neurologic diseases involving memory acquisition and storage. J Toxicol 2012; 2012:756358. [PMID: 22701481 PMCID: PMC3373146 DOI: 10.1155/2012/756358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 02/08/2012] [Indexed: 12/31/2022] Open
Abstract
Potassium channels are the most heterogeneous and widely distributed group of ion channels and play important functions in all cells, in both normal and pathological mechanisms, including learning and memory processes. Being fundamental for many diverse physiological processes, K+-channels are recognized as potential therapeutic targets in the treatment of several Central Nervous System (CNS) diseases, such as multiple sclerosis, Parkinson's and Alzheimer's diseases, schizophrenia, HIV-1-associated dementia, and epilepsy. Blockers of these channels are therefore potential candidates for the symptomatic treatment of these neuropathies, through their neurological effects. Venomous animals have evolved a wide set of toxins for prey capture and defense. These compounds, mainly peptides, act on various pharmacological targets, making them an innumerable source of ligands for answering experimental paradigms, as well as for therapeutic application. This paper provides an overview of CNS K+-channels involved in memory acquisition and storage and aims at evaluating the use of highly selective K+-channel blockers derived from arthropod venoms as potential therapeutic agents for CNS diseases involving learning and memory mechanisms.
Collapse
|
25
|
Su D, Zhao Y, Wang B, Xu H, Li W, Chen J, Wang X. Isoflurane-induced spatial memory impairment in mice is prevented by the acetylcholinesterase inhibitor donepezil. PLoS One 2011; 6:e27632. [PMID: 22114680 PMCID: PMC3219671 DOI: 10.1371/journal.pone.0027632] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 10/20/2011] [Indexed: 11/30/2022] Open
Abstract
Although many studies have shown that isoflurane exposure impairs spatial memory in aged animals, there are no clinical treatments available to prevent this memory deficit. The anticholinergic properties of volatile anesthetics are a biologically plausible cause of cognitive dysfunction in elderly subjects. We hypothesized that pretreatment with the acetylcholinesterase inhibitor donepezil, which has been approved by the Food and Drug Administration (FDA) for the treatment of Alzheimer's disease, prevents isoflurane-induced spatial memory impairment in aged mice. In present study, eighteen-month-old mice were administered donepezil (5 mg/kg) or an equal volume of saline by oral gavage with a feeding needle for four weeks. Then the mice were exposed to isoflurane (1.2%) for six hours. Two weeks later, mice were subjected to the Morris water maze to examine the impairment of spatial memory after exposure to isoflurane. After the behavioral test, the mice were sacrificed, and the protein expression level of acetylcholinesterase (AChE), choline acetylase (ChAT) and α7 nicotinic receptor (α7-nAChR) were measured in the brain. Each group consisted of 12 mice. We found that isoflurane exposure for six hours impaired the spatial memory of the mice. Compared with the control group, isoflurane exposure dramatically decreased the protein level of ChAT, but not AChE or α7-nAChR. Donepezil prevented isoflurane-induced spatial memory impairments and increased ChAT levels, which were downregulated by isoflurane. In conclusions, pretreatment with the AChE inhibitor donepezil prevented isoflurane-induced spatial memory impairment in aged mice. The mechanism was associated with the upregulation of ChAT, which was decreased by isoflurane.
Collapse
Affiliation(s)
- Diansan Su
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Obiang P, Maubert E, Bardou I, Nicole O, Launay S, Bezin L, Vivien D, Agin V. Enriched housing reverses age-associated impairment of cognitive functions and tPA-dependent maturation of BDNF. Neurobiol Learn Mem 2011; 96:121-9. [DOI: 10.1016/j.nlm.2011.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 02/22/2011] [Accepted: 03/21/2011] [Indexed: 01/19/2023]
|
27
|
Su D, Zhao Y, Wang B, Li W, Xiao J, Chen J, Wang X. Repeated but not single isoflurane exposure improved the spatial memory of young adult mice. Acta Anaesthesiol Scand 2011; 55:468-73. [PMID: 21288227 DOI: 10.1111/j.1399-6576.2010.02385.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Exposure to general anesthesia damages the spatial memory of aged rodents, but its role in the young adult has not been well investigated. This study was conducted to determine the effects of single and repeated isoflurane exposure on spatial memory in young adult mice. METHODS The experimental design comprised three series of experiments. In the first series, animals (2 months old) were randomly exposed to 1.4% isoflurane for 2 or 6 h or vehicle gas for 6 h (n=15 in each group). The Morris water maze (MWM) test was performed at 48 h after anesthetic exposure to evaluate spatial memory. In the second part, animals were randomly exposed to 1.4% isoflurane (isoflurane group, n=15) or vehicle gas only (control group, n=15) for 2 h/day on 5 consecutive days. Forty-eight hours later, the MWM was performed. In the third part, animals were randomized into an isoflurane group (n=15) and a control group (n=15) as in the second part of the experiment. Two weeks later, the MWM was performed. RESULTS The mean escape latency time and the mean pathway length measured at 48 h after repeated isoflurane exposure decreased significantly compared with the control. However, there was no difference between the two groups when tested at 2 weeks after repeated isoflurane exposure. A single exposure to isoflurane lasting 2 or 6 h had no effect on the performance of mice in the MWM as compared with the control. CONCLUSION Repeated but not single isoflurane exposure temporarily improved the spatial memory of young adult mice.
Collapse
Affiliation(s)
- D Su
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Chen BS, Wu SN. Functional role of the activity of ATP-sensitive potassium channels in electrical behavior of hippocampal neurons: Experimental and theoretical studies. J Theor Biol 2011; 272:16-25. [DOI: 10.1016/j.jtbi.2010.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 10/13/2010] [Accepted: 12/01/2010] [Indexed: 11/17/2022]
|
29
|
Betourne A, Marty V, Ceccom J, Halley H, Lassalle JM, Zajac JM, Frances B, Mouledous L. Central locomotor and cognitive effects of a NPFF receptor agonist in mouse. Peptides 2010; 31:221-6. [PMID: 19931330 DOI: 10.1016/j.peptides.2009.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 11/09/2009] [Accepted: 11/10/2009] [Indexed: 10/20/2022]
Abstract
NPFF receptors are expressed in several brain regions directly or indirectly involved in cognition and behavior. However, the cognitive effects of the NPFF system have been poorly studied. Therefore, the aim of our study was to analyze the effects of i.c.v. injections of 1 DMe, a stable agonist of NPFF receptors, on behavioral and cognitive performances in C57BL/6J mice. We measured locomotor activity, and an open field with objects was used to estimate the ability of mice to react to spatial changes and to measure short-term retention of information. The Morris navigation task was used to evaluate the acquisition, as well as long-term retention of a hippocampo-dependent spatial memory with a distributed training procedure. Finally, 1 DMe was tested in a contextual fear conditioning paradigm to study its effect on long-term memory of contextual information acquired in a single training session. Altogether, our results demonstrate a small but complex influence of the NPFF system on mouse behavior. 1 DMe injected i.c.v. induces a delayed hyperlocomotion and mildly impairs both short-term and long-term spatial memory processing without affecting contextual fear memory.
Collapse
Affiliation(s)
- Alexandre Betourne
- Université de Toulouse, Centre de Recherches sur la Cognition Animale, France
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Akrouh A, Halcomb SE, Nichols CG, Sala-Rabanal M. Molecular biology of K(ATP) channels and implications for health and disease. IUBMB Life 2009; 61:971-8. [PMID: 19787700 DOI: 10.1002/iub.246] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The ATP-sensitive potassium (K(ATP)) channel is expressed in most excitable tissues and plays a critical role in numerous physiological processes by coupling intracellular energetics to electrical activity. The channel is comprised of four Kir6.x subunits associated with four regulatory sulfonylurea receptors (SUR). Intracellular ATP acts on Kir6.x to inhibit channel activity, while MgADP stimulates channel activity through SUR. Changes in the cytosolic [ATP] to [ADP] ratio thus determine channel activity. Multiple mutations in Kir6.x and SUR genes have implicated K(ATP) channels in various diseases ranging from diabetes and hyperinsulinism to cardiac arrhythmias and cardiovascular disease. Continuing studies of channel physiology and pathology will bring new insights to the molecular basis of K(ATP) channel function, leading to a better understanding of the role that K(ATP) channels play in both health and disease.
Collapse
Affiliation(s)
- Alejandro Akrouh
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
31
|
Group I metabotropic glutamate receptors are involved in TEA-induced long-term potentiation at mossy fiber-CA3 synapses in the rat hippocampus. Brain Res 2009; 1313:45-52. [PMID: 19961834 DOI: 10.1016/j.brainres.2009.11.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 11/19/2009] [Accepted: 11/21/2009] [Indexed: 11/24/2022]
Abstract
Gq-protein-coupled Group I metabotropic glutamate receptors (mGluR) reportedly activate phospholipase C (PLC), leading to Ca(2+) release from intracellular stores and the formation of diacylglycerol (DAG). We electrophysiologically examined the involvement of the Group I mGluR in tetraethylammonium (TEA)-induced long-term potentiation (LTP) at mossy fiber (MF)-CA3 synapses in the rat hippocampus. TEA-induced LTP was almost completely blocked under the selective blockade of either mGluR1 or mGluR5, both of which are Group I mGluR. This result was supported by the blockade of TEA-induced LTP even in the absence of these blockers under low temperature conditions, in which the activation of Group I mGluR is thought not to be fully effective. In addition, the blockade of mGluR1 resulted in lower short-term potentiation (STP) during TEA application compared with the blockade of mGluR5. These results demonstrate the crucial roles of Group I mGluR in the TEA-induced LTP at MF-CA3 synapses and the different contributions of mGluR1 and mGluR5 to the initial component of plasticity.
Collapse
|
32
|
Diazoxide reduces status epilepticus neuron damage in diabetes. Neurotox Res 2009; 17:305-16. [PMID: 19728004 DOI: 10.1007/s12640-009-9104-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 08/20/2009] [Accepted: 08/20/2009] [Indexed: 01/19/2023]
Abstract
Diabetic hyperglycemia is associated with seizure severity and may aggravate brain damage after status epilepticus. Our earlier studies suggest the involvement of ATP-sensitive potassium channels (K(ATP)) in glucose-related neuroexcitability. We aimed to determine whether K(ATP) agonist protects against status epilepticus-induced brain damage. Adult male Sprague-Dawley rats were divided into two groups: the streptozotocin (STZ)-induced diabetes (STZ) group and the normal saline (NS) group. Both groups were treated with either diazoxide (15 mg/kg, i.v.) (STZ + DZX, NS + DZX) or vehicle (STZ + V, NS + V) before lithium-pilocarpine-induced status epilepticus. We evaluated seizure susceptibility, severity, and mortality. The rats underwent Morris water-maze tests and hippocampal histopathology analyses 24 h post-status epilepticus. A multi-electrode recording system was used to study field excitatory postsynaptic synaptic potentials (fEPSP). RNA interference (RNAi) to knockdown Kir 6.2 in a hippocampal cell line was used to evaluate the effect of diazoxide in the presence of high concentration of ATP. Seizures were less severe (P < 0.01), post-status epilepticus learning and memory were better (P < 0.05), and neuron loss in the hippocampal CA3 area was lower (P < 0.05) in the STZ + DZX than the STZ + V group. In contrast, seizure severity, post-status epilepticus learning and memory, and hippocampal CA3 neuron loss were comparable in the NS + DZX and NS + V groups. fEPSP was lower in the STZ + DZX but not in the NS + DZX group. The RNAi study confirmed that diazoxide, with its K(ATP)-opening effects, could counteract the K(ATP)-closing effect by high dose ATP. We conclude that, by opening K(ATP), diazoxide protects against status epilepticus-induced neuron damage during diabetic hyperglycemia.
Collapse
|