1
|
Becchi S, Chieng B, Bradfield LA, Capellán R, Leung BK, Balleine BW. Cognitive effects of thalamostriatal degeneration are ameliorated by normalizing striatal cholinergic activity. SCIENCE ADVANCES 2023; 9:eade8247. [PMID: 37352346 PMCID: PMC10289650 DOI: 10.1126/sciadv.ade8247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 05/19/2023] [Indexed: 06/25/2023]
Abstract
The loss of neurons in parafascicular thalamus (Pf) and their inputs to dorsomedial striatum (DMS) in Lewy body disease (LBD) and Parkinson's disease dementia (PDD) have been linked to the effects of neuroinflammation. We found that, in rats, these inputs were necessary for both the function of striatal cholinergic interneurons (CINs) and the flexible encoding of the action-outcome (AO) associations necessary for goal-directed action, producing a burst-pause pattern of CIN firing but only during the remapping elicited by a shift in AO contingency. Neuroinflammation in the Pf abolished these changes in CIN activity and goal-directed control after the shift in contingency. However, both effects were rescued by either the peripheral or the intra-DMS administration of selegiline, a monoamine oxidase B inhibitor that we found also enhances adenosine triphosphatase activity in CINs. These findings suggest a potential treatment for the cognitive deficits associated with neuroinflammation affecting the function of the Pf and related structures.
Collapse
Affiliation(s)
- Serena Becchi
- School of Psychology, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Billy Chieng
- School of Psychology, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Laura A. Bradfield
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Roberto Capellán
- School of Psychology, Department of Psychobiology, National University for Distance Learning, Madrid, Spain
| | - Beatrice K. Leung
- School of Psychology, Faculty of Science, University of New South Wales, Sydney, Australia
| | - Bernard W. Balleine
- School of Psychology, Faculty of Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
2
|
Poppi LA, Ho-Nguyen KT, Shi A, Daut CT, Tischfield MA. Recurrent Implication of Striatal Cholinergic Interneurons in a Range of Neurodevelopmental, Neurodegenerative, and Neuropsychiatric Disorders. Cells 2021; 10:907. [PMID: 33920757 PMCID: PMC8071147 DOI: 10.3390/cells10040907] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/03/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Cholinergic interneurons are "gatekeepers" for striatal circuitry and play pivotal roles in attention, goal-directed actions, habit formation, and behavioral flexibility. Accordingly, perturbations to striatal cholinergic interneurons have been associated with many neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. The role of acetylcholine in many of these disorders is well known, but the use of drugs targeting cholinergic systems fell out of favor due to adverse side effects and the introduction of other broadly acting compounds. However, in response to recent findings, re-examining the mechanisms of cholinergic interneuron dysfunction may reveal key insights into underlying pathogeneses. Here, we provide an update on striatal cholinergic interneuron function, connectivity, and their putative involvement in several disorders. In doing so, we aim to spotlight recurring physiological themes, circuits, and mechanisms that can be investigated in future studies using new tools and approaches.
Collapse
Affiliation(s)
- Lauren A. Poppi
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA;
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Tourette International Collaborative (TIC) Genetics Study, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Khue Tu Ho-Nguyen
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Anna Shi
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Cynthia T. Daut
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Max A. Tischfield
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Tourette International Collaborative (TIC) Genetics Study, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
3
|
Yildirim C, Özkaya B, Bal R. KATP and TRPM2-like channels couple metabolic status to resting membrane potential of octopus neurons in the mouse ventral cochlear nucleus. Brain Res Bull 2021; 170:115-128. [PMID: 33581312 DOI: 10.1016/j.brainresbull.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/29/2022]
Abstract
ATP-sensitive potassium (KATP) channels and transient receptor potential melastatin 2 (TRPM2) channels are commonly expressed both pre- and postsynaptically in the central nervous system (CNS). We hypothesized that KATP and TRPM2 may couple metabolic status to the resting membrane potential of octopus neurons of the mouse ventral cochlear nucleus (VCN). Therefore, we studied the expression of KATP channels and TRPM2 channels in octopus cells by immunohistochemical techniques and their contribution to neuronal electrical properties by the electrophysiological patch clamp technique. In immunohistochemical staining of octopus cells, labelling with Kir6.2 and SUR1 antibodies was strong, and labelling with the SUR2 antibody was moderate, but labelling with Kir6.1 was very weak. Octopus cells had intense staining with TRPM2 antibodies. In patch clamp recordings, bath application of KATP channel agonists H2O2 (880 μM), ATZ (1 mM), cromakalim (50 μM), diazoxide (200 μM), NNC 55-0118 and NN 414 separately resulted in hyperpolarizations of resting potential to different extents. Application of 8-Bro-cADPR (50 μM), a specific antagonist of TRPM2 channels, in the presence of H2O2 (880 μM) resulted in further hyperpolarization by approximately 1 mV. The amplitudes of H2O2-induced outward KATP currents and ADPR-induced inward currents were 206.1 ± 31.5 pA (n = 4) and 136.8 ± 22.4 pA, respectively, at rest. Their respective reversal potentials were -77 ± 2.6 mV (n = 3) and -6.3 ± 2.9 (n = 3) and -6.3 ± 2.9 (n = 3). In conclusion, octopus cells appear to possess both KATP channels and TRPM2-like channels. KATP might largely be constituted by SUR1-Kir6.2 subunits and SUR2-Kir6.2 subunits. Both KATP and TRPM2-like channels might have a modulatory action in setting the membrane potential.
Collapse
Affiliation(s)
- Caner Yildirim
- Department of Physiology, Faculty of Medicine, Gaziantep University, 27310, Gaziantep, Turkey
| | - Beytullah Özkaya
- Department of Physiology, Faculty of Medicine, Gaziantep University, 27310, Gaziantep, Turkey
| | - Ramazan Bal
- Department of Physiology, Faculty of Medicine, Gaziantep University, 27310, Gaziantep, Turkey.
| |
Collapse
|
4
|
Zhao G, Kaplan A, Greiser M, Lederer WJ. The surprising complexity of KATP channel biology and of genetic diseases. J Clin Invest 2020; 130:1112-1115. [PMID: 32065592 DOI: 10.1172/jci135759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The ATP-sensitive K+ channel (KATP) is formed by the association of four inwardly rectifying K+ channel (Kir6.x) pore subunits with four sulphonylurea receptor (SUR) regulatory subunits. Kir6.x or SUR mutations result in KATP channelopathies, which reflect the physiological roles of these channels, including but not limited to insulin secretion, cardiac protection, and blood flow regulation. In this issue of the JCI, McClenaghan et al. explored one of the channelopathies, namely Cantu syndrome (CS), which is a result of one kind of KATP channel mutation. Using a knockin mouse model, the authors demonstrated that gain-of-function KATP mutations in vascular smooth muscle resulted in cardiac remodeling. Moreover, they were able to reverse the cardiovascular phenotypes by administering the KATP channel blocker glibenclamide. These results exemplify how genetic mutations can have an impact on developmental trajectories, and provide a therapeutic approach to mitigate cardiac hypertrophy in cases of CS.
Collapse
Affiliation(s)
- Guiling Zhao
- Center for Biomedical Engineering and Technology and Department of Physiology
| | - Aaron Kaplan
- Center for Biomedical Engineering and Technology and Department of Physiology.,Department of Medicine and Division of Cardiology, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Maura Greiser
- Center for Biomedical Engineering and Technology and Department of Physiology
| | - W Jonathan Lederer
- Center for Biomedical Engineering and Technology and Department of Physiology
| |
Collapse
|
5
|
Tubert C, Murer MG. What’s wrong with the striatal cholinergic interneurons in Parkinson’s disease? Focus on intrinsic excitability. Eur J Neurosci 2020; 53:2100-2116. [DOI: 10.1111/ejn.14742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Cecilia Tubert
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| | - Mario Gustavo Murer
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| |
Collapse
|
6
|
Ztaou S, Amalric M. Contribution of cholinergic interneurons to striatal pathophysiology in Parkinson's disease. Neurochem Int 2019; 126:1-10. [PMID: 30825602 DOI: 10.1016/j.neuint.2019.02.019] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/25/2019] [Accepted: 02/24/2019] [Indexed: 01/22/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by the loss of nigral dopaminergic neurons innervating the striatum, the main input structure of the basal ganglia. This creates an imbalance between dopaminergic inputs and cholinergic interneurons (ChIs) within the striatum. The efficacy of anticholinergic drugs, one of the earliest therapy for PD before the discovery of L-3,4-dihydroxyphenylalanine (L-DOPA) suggests an increased cholinergic tone in this disease. The dopamine (DA)-acetylcholine (ACh) balance hypothesis is now revisited with the use of novel cutting-edge techniques (optogenetics, pharmacogenetics, new electrophysiological recordings). This review will provide the background of the specific contribution of ChIs to striatal microcircuit organization in physiological and pathological conditions. The second goal of this review is to delve into the respective contributions of nicotinic and muscarinic receptor cholinergic subunits to the control of striatal afferent and efferent neuronal systems. Special attention will be given to the role played by muscarinic acetylcholine receptors (mAChRs) in the regulation of striatal network which may have important implications in the development of novel therapeutic strategies for motor and cognitive impairment in PD.
Collapse
Affiliation(s)
- Samira Ztaou
- Aix Marseille Univ, CNRS, LNC, FR3C, Marseille, France; Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY, 10032, USA
| | | |
Collapse
|
7
|
Abudukeyoumu N, Hernandez-Flores T, Garcia-Munoz M, Arbuthnott GW. Cholinergic modulation of striatal microcircuits. Eur J Neurosci 2018; 49:604-622. [PMID: 29797362 PMCID: PMC6587740 DOI: 10.1111/ejn.13949] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022]
Abstract
The purpose of this review is to bridge the gap between earlier literature on striatal cholinergic interneurons and mechanisms of microcircuit interaction demonstrated with the use of newly available tools. It is well known that the main source of the high level of acetylcholine in the striatum, compared to other brain regions, is the cholinergic interneurons. These interneurons provide an extensive local innervation that suggests they may be a key modulator of striatal microcircuits. Supporting this idea requires the consideration of functional properties of these interneurons, their influence on medium spiny neurons, other interneurons, and interactions with other synaptic regulators. Here, we underline the effects of intrastriatal and extrastriatal afferents onto cholinergic interneurons and discuss the activation of pre‐ and postsynaptic muscarinic and nicotinic receptors that participate in the modulation of intrastriatal neuronal interactions. We further address recent findings about corelease of other transmitters in cholinergic interneurons and actions of these interneurons in striosome and matrix compartments. In addition, we summarize recent evidence on acetylcholine‐mediated striatal synaptic plasticity and propose roles for cholinergic interneurons in normal striatal physiology. A short examination of their role in neurological disorders such as Parkinson's, Huntington's, and Tourette's pathologies and dystonia is also included.
Collapse
Affiliation(s)
| | | | | | - Gordon W Arbuthnott
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
8
|
Modulation of Excitability of Stellate Neurons in the Ventral Cochlear Nucleus of Mice by ATP-Sensitive Potassium Channels. J Membr Biol 2018; 251:163-178. [PMID: 29379989 DOI: 10.1007/s00232-017-0011-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 12/19/2017] [Indexed: 12/21/2022]
Abstract
Major voltage-activated ionic channels of stellate cells in the ventral part of cochlear nucleus (CN) were largely characterized previously. However, it is not known if these cells are equipped with other ion channels apart from the voltage-sensitive ones. In the current study, it was aimed to study subunit composition and function of ATP-sensitive potassium channels (KATP) in stellate cells of the ventral cochlear nucleus. Subunits of KATP channels, Kir6.1, Kir6.2, SUR1, and SUR2, were expressed at the mRNA level and at the protein level in the mouse VCN tissue. The specific and clearly visible bands for all subunits but that for Kir6.1 were seen in Western blot. Using immunohistochemical staining technique, stellate cells were strongly labeled with SUR1 and Kir6.2 antibodies and moderately labeled with SUR2 antibody, whereas the labeling signals for Kir6.1 were too weak. In patch clamp recordings, KATP agonists including cromakalim (50 µM), diazoxide (0.2 mM), 3-Amino-1,2,4-triazole (ATZ) (1 mM), 2,2-Dithiobis (5-nitro pyridine) (DTNP) (330 µM), 6-Chloro-3-isopropylamino- 4H-thieno[3,2-e]-1,2,4-thiadiazine 1,1-dioxide (NNC 55-0118) (1 µM), 6-chloro-3-(methylcyclopropyl)amino-4H-thieno[3,2-e]-1,2,4-thiadiazine 1,1-dioxide (NN414) (1 µM), and H2O2 (0.88 mM) induced marked responses in stellate cells, characterized by membrane hyperpolarization which were blocked by KATP antagonists. Blockers of KATP channels, glibenclamide (0.2 mM), tolbutamide (0.1 mM) as well as 5-hydroxydecanoic acid (1 mM), and catalase (500 IU/ml) caused depolarization of stellate cells, increasing spontaneous action potential firing. In conclusion, KATP channels seemed to be composed dominantly of Kir 6.2 subunit and SUR1 and SUR2 and activation or inhibition of KATP channels regulates firing properties of stellate cells by means of influencing resting membrane potential and input resistance.
Collapse
|
9
|
Nelson PT, Wang WX, Wilfred BR, Wei A, Dimayuga J, Huang Q, Ighodaro E, Artiushin S, Fardo DW. Novel human ABCC9/SUR2 brain-expressed transcripts and an eQTL relevant to hippocampal sclerosis of aging. J Neurochem 2015; 134:1026-39. [PMID: 26115089 DOI: 10.1111/jnc.13202] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 05/19/2015] [Accepted: 06/09/2015] [Indexed: 01/08/2023]
Abstract
ABCC9 genetic polymorphisms are associated with increased risk for various human diseases including hippocampal sclerosis of aging. The main goals of this study were 1 > to detect the ABCC9 variants and define the specific 3' untranslated region (3'UTR) for each variant in human brain, and 2 > to determine whether a polymorphism (rs704180) associated with risk for hippocampal sclerosis of aging pathology is also associated with variation in ABCC9 transcript expression and/or splicing. Rapid amplification of ABCC9 cDNA ends (3'RACE) provided evidence of novel 3' UTR portions of ABCC9 in human brain. In silico and experimental studies were performed focusing on the single nucleotide polymorphism, rs704180. Analyses from multiple databases, focusing on rs704180 only, indicated that this risk allele is a local expression quantitative trait locus (eQTL). Analyses of RNA from human brains showed increased ABCC9 transcript levels in individuals with the risk genotype, corresponding with enrichment for a shorter 3' UTR which may be more stable than variants with the longer 3' UTR. MicroRNA transfection experiments yielded results compatible with the hypothesis that miR-30c causes down-regulation of SUR2 transcripts with the longer 3' UTR. Thus we report evidence of complex ABCC9 genetic regulation in brain, which may be of direct relevance to human disease. ABCC9 gene variants are associated with increased risk for hippocampal sclerosis of aging (HS-Aging--a prevalent brain disease with symptoms that mimic Alzheimer's disease). We describe novel ABCC9 variants in human brain, corresponding to altered 3'UTR length, which could lead to targeting by miR-30c. We also determined that the HS-Aging risk mutation is associated with variation in ABCC9 transcript expression.
Collapse
Affiliation(s)
- Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| | - Wang-Xia Wang
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Bernard R Wilfred
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Angela Wei
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - James Dimayuga
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Qingwei Huang
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Eseosa Ighodaro
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Sergey Artiushin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
10
|
Gonzales KK, Smith Y. Cholinergic interneurons in the dorsal and ventral striatum: anatomical and functional considerations in normal and diseased conditions. Ann N Y Acad Sci 2015; 1349:1-45. [PMID: 25876458 DOI: 10.1111/nyas.12762] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Striatal cholinergic interneurons (ChIs) are central for the processing and reinforcement of reward-related behaviors that are negatively affected in states of altered dopamine transmission, such as in Parkinson's disease or drug addiction. Nevertheless, the development of therapeutic interventions directed at ChIs has been hampered by our limited knowledge of the diverse anatomical and functional characteristics of these neurons in the dorsal and ventral striatum, combined with the lack of pharmacological tools to modulate specific cholinergic receptor subtypes. This review highlights some of the key morphological, synaptic, and functional differences between ChIs of different striatal regions and across species. It also provides an overview of our current knowledge of the cellular localization and function of cholinergic receptor subtypes. The future use of high-resolution anatomical and functional tools to study the synaptic microcircuitry of brain networks, along with the development of specific cholinergic receptor drugs, should help further elucidate the role of striatal ChIs and permit efficient targeting of cholinergic systems in various brain disorders, including Parkinson's disease and addiction.
Collapse
Affiliation(s)
- Kalynda K Gonzales
- Yerkes National Primate Research Center, Department of Neurology and Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, Georgia.,Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Yoland Smith
- Yerkes National Primate Research Center, Department of Neurology and Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, Georgia
| |
Collapse
|
11
|
Lim SAO, Kang UJ, McGehee DS. Striatal cholinergic interneuron regulation and circuit effects. Front Synaptic Neurosci 2014; 6:22. [PMID: 25374536 PMCID: PMC4204445 DOI: 10.3389/fnsyn.2014.00022] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/05/2014] [Indexed: 01/11/2023] Open
Abstract
The striatum plays a central role in motor control and motor learning. Appropriate responses to environmental stimuli, including pursuit of reward or avoidance of aversive experience all require functional striatal circuits. These pathways integrate synaptic inputs from limbic and cortical regions including sensory, motor and motivational information to ultimately connect intention to action. Although many neurotransmitters participate in striatal circuitry, one critically important player is acetylcholine (ACh). Relative to other brain areas, the striatum contains exceptionally high levels of ACh, the enzymes that catalyze its synthesis and breakdown, as well as both nicotinic and muscarinic receptor types that mediate its postsynaptic effects. The principal source of striatal ACh is the cholinergic interneuron (ChI), which comprises only about 1-2% of all striatal cells yet sends dense arbors of projections throughout the striatum. This review summarizes recent advances in our understanding of the factors affecting the excitability of these neurons through acute effects and long term changes in their synaptic inputs. In addition, we discuss the physiological effects of ACh in the striatum, and how changes in ACh levels may contribute to disease states during striatal dysfunction.
Collapse
Affiliation(s)
| | - Un Jung Kang
- Department of Neurology, Columbia University New York, NY, USA
| | - Daniel S McGehee
- Committee on Neurobiology, University of Chicago Chicago, IL, USA ; Department of Anesthesia and Critical Care, University of Chicago Chicago, IL, USA
| |
Collapse
|
12
|
Acetylcholine encodes long-lasting presynaptic plasticity at glutamatergic synapses in the dorsal striatum after repeated amphetamine exposure. J Neurosci 2013; 33:10405-26. [PMID: 23785153 DOI: 10.1523/jneurosci.0014-13.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Locomotion and cue-dependent behaviors are modified through corticostriatal signaling whereby short-term increases in dopamine availability can provoke persistent changes in glutamate release that contribute to neuropsychiatric disorders, including Parkinson's disease and drug dependence. We found that withdrawal of mice from repeated amphetamine treatment caused a chronic presynaptic depression (CPD) in glutamate release that was most pronounced in corticostriatal terminals with a low probability of release and lasted >50 d in treated mice. An amphetamine challenge reversed CPD via a dopamine D1-receptor-dependent paradoxical presynaptic potentiation (PPP) that increased corticostriatal activity in direct pathway medium spiny neurons. This PPP was correlated with locomotor responses after a drug challenge, suggesting that it may underlie the sensitization process. Experiments in brain slices and in vivo indicated that dopamine regulation of acetylcholine release from tonically active interneurons contributes to CPD, PPP, locomotor sensitization, and cognitive ability. Therefore, a chronic decrease in corticostriatal activity during withdrawal is regulated around a new physiological range by tonically active interneurons and returns to normal upon reexposure to amphetamine, suggesting that this paradoxical return of striatal activity to a more stable, normalized state may represent an additional source of drug motivation during abstinence.
Collapse
|
13
|
Zhou M, He HJ, Tanaka O, Sekiguchi M, Kawahara K, Abe H. Localization of the ATP-sensitive K(+) channel regulatory subunits SUR2A and SUR2B in the rat brain. Neurosci Res 2012; 74:91-105. [PMID: 22960600 DOI: 10.1016/j.neures.2012.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 08/22/2012] [Accepted: 08/23/2012] [Indexed: 01/11/2023]
Abstract
ATP-sensitive K(+) (K(ATP)) channel subunits SUR2A and SUR2B in the rat brain were investigated by RT-PCR assay, western blot analysis, in situ hybridization histochemistry, and immunohistochemical staining. The results show that the mRNA and protein of SUR2A and SUR2B are expressed in whole rat brain extracts and selected regions. SUR2 mRNA is widely expressed in many neurons and glial cells as revealed by in situ hybridization histochemistry. Immunohistochemical staining shows SUR2A to be widely expressed in neurons of the brain, especially in the large pyramidal neurons and their main dendrites in the neocortex and in the Purkinje cells of the cerebellar cortex. In contrast to SUR2A, SUR2B is potently expressed in small cells in the corpus callosum and cerebellar white matter, but is also weakly expressed in some neurons. Double immunostaining shows SUR2B to be localized in astrocytes and oligodendrocytes, while SUR2A is only localized in oligodendrocytes. These results suggest that SUR2A might be mainly a regulatory subunit of the K(ATP) channel in most neurons and part of oligodendrocytes, while SUR2B might be mainly a regulatory subunit of the K(ATP) channel in astrocytes, oligodendrocytes, and some neurons.
Collapse
Affiliation(s)
- Ming Zhou
- Department of Anatomy, Akita University Graduate School of Medicine and Faculty of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan.
| | | | | | | | | | | |
Collapse
|
14
|
Patel JC, Witkovsky P, Coetzee WA, Rice ME. Subsecond regulation of striatal dopamine release by pre-synaptic KATP channels. J Neurochem 2011; 118:721-36. [PMID: 21689107 DOI: 10.1111/j.1471-4159.2011.07358.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
ATP-sensitive K(+) (K(ATP)) channels are composed of pore-forming subunits, typically Kir6.2 in neurons, and regulatory sulfonylurea receptor subunits. In dorsal striatum, activity-dependent H(2)O(2) produced from glutamate receptor activation inhibits dopamine release via K(ATP) channels. Sources of modulatory H(2)O(2) include striatal medium spiny neurons, but not dopaminergic axons. Using fast-scan cyclic voltammetry in guinea-pig striatal slices and immunohistochemistry, we determined the time window for H(2)O(2)/K(ATP)-channel-mediated inhibition and assessed whether modulatory K(ATP) channels are on dopaminergic axons. Comparison of paired-pulse suppression of dopamine release in the absence and presence of glibenclamide, a K(ATP)-channel blocker, or mercaptosuccinate, a glutathione peroxidase inhibitor that enhances endogenous H(2)O(2) levels, revealed a time window for inhibition of 500-1000 ms after stimulation. Immunohistochemistry demonstrated localization of Kir6.2 K(ATP)-channel subunits on dopaminergic axons. Consistent with the presence of functional K(ATP) channels on dopaminergic axons, K(ATP)-channel openers, diazoxide and cromakalim, suppressed single-pulse evoked dopamine release. Although cholinergic interneurons that tonically regulate dopamine release also express K(ATP) channels, diazoxide did not induce the enhanced frequency responsiveness of dopamine release seen with nicotinic-receptor blockade. Together, these studies reveal subsecond regulation of striatal dopamine release by endogenous H(2)O(2) acting at K(ATP) channels on dopaminergic axons, including a role in paired-pulse suppression.
Collapse
Affiliation(s)
- Jyoti C Patel
- Department of Neurosurgery, New York University School of Medicine, New York, New York 10016, USA.
| | | | | | | |
Collapse
|
15
|
Characterization of an ATP-sensitive K(+) channel in rat carotid body glomus cells. Respir Physiol Neurobiol 2011; 177:247-55. [PMID: 21536154 DOI: 10.1016/j.resp.2011.04.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 04/14/2011] [Accepted: 04/17/2011] [Indexed: 01/04/2023]
Abstract
Carotid body glomus (CB) cells express different types of K(+) channels such as TASK, BK, and Kv channels, and hypoxia has been shown to inhibit these channels. Here we report the presence of a ∼72-pS channel that has not been described previously in CB cells. In cell-attached patches with 150 mM K(+) in the pipette and bath solutions, TASK-like channels were present (∼15 and ∼36-pS). After formation of inside-out patches, a 72-pS channel became transiently active in ∼18% of patches. The 72-pS channel was K(+)-selective, inhibited by 2-4 mM ATP and 10-100 μM glybenclamide. The 72-pS channel was observed in CB cells isolated from newborn, 2-3 week and 10-12 week-old rats. Reverse transcriptase-PCR and immunocytochemistry showed that Kir6.1, Kir6.2, SUR1 and SUR2 were expressed in CB glomus cells as well as in non-glomus cells. Acute hypoxia (∼15 mmHg O(2)) inhibited TASK-like channels but failed to activate the 72-pS channel in cell-attached CB cells. K(+) channel openers (diazoxide, pinacidil, levcromakalim), sodium cyanide and removal of extracellular glucose also did not activate the 72-pS channel in the cell-attached state. The hypoxia-induced elevation of intracellular [Ca(2+)] was unchanged by glybenclamide or diazoxide. NaCN-induced increase in [Ca(2+)] was not affected by 10 μM glybenclamide but inhibited by 100 μM glybenclamide. Acute glucose deprivation did not elevate [Ca(2+)] in the presence or absence of glybenclamide. These results show that an ATP-sensitive K(+) channel is expressed in the plasma membrane of CB cells, but is not activated by short-term metabolic inhibition. The functional relevance of the 72-pS channel remains to be determined.
Collapse
|
16
|
Hibino H, Inanobe A, Furutani K, Murakami S, Findlay I, Kurachi Y. Inwardly rectifying potassium channels: their structure, function, and physiological roles. Physiol Rev 2010; 90:291-366. [PMID: 20086079 DOI: 10.1152/physrev.00021.2009] [Citation(s) in RCA: 1135] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inwardly rectifying K(+) (Kir) channels allow K(+) to move more easily into rather than out of the cell. They have diverse physiological functions depending on their type and their location. There are seven Kir channel subfamilies that can be classified into four functional groups: classical Kir channels (Kir2.x) are constitutively active, G protein-gated Kir channels (Kir3.x) are regulated by G protein-coupled receptors, ATP-sensitive K(+) channels (Kir6.x) are tightly linked to cellular metabolism, and K(+) transport channels (Kir1.x, Kir4.x, Kir5.x, and Kir7.x). Inward rectification results from pore block by intracellular substances such as Mg(2+) and polyamines. Kir channel activity can be modulated by ions, phospholipids, and binding proteins. The basic building block of a Kir channel is made up of two transmembrane helices with cytoplasmic NH(2) and COOH termini and an extracellular loop which folds back to form the pore-lining ion selectivity filter. In vivo, functional Kir channels are composed of four such subunits which are either homo- or heterotetramers. Gene targeting and genetic analysis have linked Kir channel dysfunction to diverse pathologies. The crystal structure of different Kir channels is opening the way to understanding the structure-function relationships of this simple but diverse ion channel family.
Collapse
Affiliation(s)
- Hiroshi Hibino
- Department of Pharmacology, Graduate School of Medicine and The Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
17
|
|
18
|
Abstract
Hypoglycaemia is a frequent and greatly feared side-effect of insulin therapy, and a major obstacle to achieving near-normal glucose control. This review will focus on the more recent developments in our understanding of the mechanisms that underlie the sensing of hypoglycaemia in both non-diabetic and diabetic individuals, and how this mechanism becomes impaired over time. The research focus of my own laboratory and many others is directed by three principal questions. Where does the body sense a falling glucose? How does the body detect a falling glucose? And why does this mechanism fail in Type 1 diabetes? Hypoglycaemia is sensed by specialized neurons found in the brain and periphery, and of these the ventromedial hypothalamus appears to play a major role. Neurons that react to fluctuations in glucose use mechanisms very similar to those that operate in pancreatic B- and A-cells, in particular in their use of glucokinase and the K(ATP) channel as key steps through which the metabolic signal is translated into altered neuronal firing rates. During hypoglycaemia, glucose-inhibited (GI) neurons may be regulated by the activity of AMP-activated protein kinase. This sensing mechanism is disturbed by recurrent hypoglycaemia, such that counter-regulatory defence responses are triggered at a lower glucose level. Why this should occur is not yet known, but it may involve increased metabolism or fuel delivery to glucose-sensing neurons or alterations in the mechanisms that regulate the stress response.
Collapse
Affiliation(s)
- R McCrimmon
- Yale University School of Medicine, Department of Internal Medicine, New Haven, CT 06520-8020, USA.
| |
Collapse
|
19
|
Gorostiza P, Isacoff E. Optical switches and triggers for the manipulation of ion channels and pores. MOLECULAR BIOSYSTEMS 2007; 3:686-704. [PMID: 17882331 DOI: 10.1039/b710287a] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Like fluorescence sensing techniques, methods to manipulate proteins with light have produced great advances in recent years. Ion channels have been one of the principal protein targets of photoswitched manipulation. In combination with fluorescence detection of cell signaling, this has enabled non-invasive, all-optical experiments on cell and tissue function, both in vitro and in vivo. Optical manipulation of channels has also provided insights into the mechanism of channel function. Optical control elements can be classified according to their molecular reversibility as non-reversible phototriggers where light breaks a chemical bond (e.g. caged ligands) and as photoswitches that reversibly photoisomerize. Synthetic photoswitches constitute nanoscale actuators that can alter channel function using three different strategies. These include (1) nanotoggles, which are tethered photoswitchable ligands that either activate channels (agonists) or inhibit them (blockers or antagonists), (2) nanokeys, which are untethered (freely diffusing) photoswitchable ligands, and (3) nanotweezers, which are photoswitchable crosslinkers. The properties of such photoswitches are discussed here, with a focus on tethered photoswitchable ligands. The recent literature on optical manipulation of ion channels is reviewed for the different channel families, with special emphasis on the understanding of ligand binding and gating processes, applications in nanobiotechnology, and with attention to future prospects in the field.
Collapse
Affiliation(s)
- Pau Gorostiza
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
20
|
van den Top M, Lyons DJ, Lee K, Coderre E, Renaud LP, Spanswick D. Pharmacological and molecular characterization of ATP-sensitive K(+) conductances in CART and NPY/AgRP expressing neurons of the hypothalamic arcuate nucleus. Neuroscience 2006; 144:815-24. [PMID: 17137725 DOI: 10.1016/j.neuroscience.2006.09.059] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Revised: 09/27/2006] [Accepted: 09/27/2006] [Indexed: 11/16/2022]
Abstract
The role of hypothalamic ATP-sensitive potassium channels in the maintenance of energy homeostasis has been extensively explored. However, how these channels are incorporated into the neuronal networks of the arcuate nucleus remains unclear. Whole-cell patch-clamp recordings from rat arcuate nucleus neurons in hypothalamic slice preparations revealed widespread expression of functional ATP-sensitive potassium channels within the nucleus. ATP-sensitive potassium channels were expressed in orexigenic neuropeptide Y/agouti-related protein (NPY/AgRP) and ghrelin-sensitive neurons and in anorexigenic cocaine-and-amphetamine regulated transcript (CART) neurons. In 70% of the arcuate nucleus neurons recorded, exposure to glucose-free bathing medium induced inhibition of electrical excitability, the response being characterized by membrane hyperpolarization, a reduction in neuronal input resistance and a reversal potential consistent with opening of potassium channels. These effects were reversible upon re-introduction of glucose to the bathing medium or upon exposure to the ATP-sensitive potassium channel blockers tolbutamide or glibenclamide. The potassium channel opener diazoxide, but not pinacidil, also induced a tolbutamide and glibenclamide-sensitive inhibition of electrical excitability. Single-cell reverse transcription-polymerase chain reaction revealed expression of mRNA for sulfonylurea receptor 1 but not sulfonylurea receptor 2 subunits of ATP-sensitive potassium channels. Thus, rat arcuate nucleus neurons, including those involved in functionally antagonistic orexigenic and anorexigenic pathways express functional ATP-sensitive potassium channels which include sulfonylurea receptor 1 subunits. These data indicate a crucial role for these ion channels in central sensing of metabolic and energy status. However, further studies are needed to clarify the differential roles of these channels, the organization of signaling pathways that regulate them and how they operate in functionally opposing cell types.
Collapse
Affiliation(s)
- M van den Top
- Division of Clinical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | | | | | | | | | | |
Collapse
|
21
|
Velasco M, García E, Onetti CG. Glucose deprivation activates diversity of potassium channels in cultured rat hippocampal neurons. Cell Mol Neurobiol 2006; 26:307-19. [PMID: 16767515 PMCID: PMC11520758 DOI: 10.1007/s10571-006-9000-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2005] [Accepted: 02/09/2006] [Indexed: 10/24/2022]
Abstract
1. Glucose is one of the most important substrates for generating metabolic energy required for the maintenance of cellular functions. Glucose-mediated changes in neuronal firing pattern have been observed in the central nervous system of mammals. K(+) channels directly regulated by intracellular ATP have been postulated as a linkage between cellular energetic metabolism and excitability; the functional roles ascribed to these channels include glucose-sensing to regulate energy homeostasis and neuroprotection under energy depletion conditions. The hippocampus is highly sensitive to metabolic insults and is the brain region most sensitive to ischemic damage. Because the identity of metabolically regulated potassium channels present in hippocampal neurons is obscure, we decided to study the biophysical properties of glucose-sensitive potassium channels in hippocampal neurons. 2. The dependence of membrane potential and the sensitivity of potassium channels to glucose and ATP in rat hippocampal neurons were studied in cell-attached and excised inside-out membrane patches. 3. We found that under hypoglycemic conditions, at least three types of potassium channels were activated; their unitary conductance values were 37, 147, and 241 pS in symmetrical K(+), and they were sensitive to ATP. For K(+) channels with unitary conductance of 37 and 241, when the membrane potential was depolarized the longer closed time constant diminished and this produced an increase in the open-state probability; nevertheless, the 147-pS channels were not voltage-dependent. 4. We propose that neuronal glucose-sensitive K(+) channels in rat hippocampus include subtypes of ATP-sensitive channels with a potential role in neuroprotection during short-term or prolonged metabolic stress.
Collapse
Affiliation(s)
- Myrian Velasco
- Centro de Investigaciones Biomédicas, Universidad de Colima, Apartado Postal 199, Colima, Col. 28000 México
| | - Esperanza García
- Centro de Investigaciones Biomédicas, Universidad de Colima, Apartado Postal 199, Colima, Col. 28000 México
| | - Carlos G. Onetti
- Centro de Investigaciones Biomédicas, Universidad de Colima, Apartado Postal 199, Colima, Col. 28000 México
| |
Collapse
|
22
|
Avshalumov MV, Chen BT, Koós T, Tepper JM, Rice ME. Endogenous hydrogen peroxide regulates the excitability of midbrain dopamine neurons via ATP-sensitive potassium channels. J Neurosci 2006; 25:4222-31. [PMID: 15858048 PMCID: PMC6725114 DOI: 10.1523/jneurosci.4701-04.2005] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ATP-sensitive K+ (K(ATP)) channels link metabolic state to cell excitability. Here, we examined regulation of K(ATP) channels in substantia nigra dopamine neurons by hydrogen peroxide (H2O2), which is produced in all cells during aerobic metabolism. Blockade of K(ATP) channels by glibenclamide (100 nM) or depletion of intracellular H2O2 by including catalase, a peroxidase enzyme, in the patch pipette increased the spontaneous firing rate of all dopamine neurons tested in guinea pig midbrain slices. Using fluorescence imaging with dichlorofluorescein to visualize intracellular H2O2, we found that moderate increases in H2O2 during partial inhibition of glutathione (GSH) peroxidase by mercaptosuccinate (0.1-0.3 mM) had no effect on dopamine neuron firing rate. However, with greater GSH inhibition (1 mM mercaptosuccinate) or application of exogenous H2O2, 50% of recorded cells showed K(ATP) channel-dependent hyperpolarization. Responsive cells also hyperpolarized with diazoxide, a selective opener for K(ATP) channels containing sulfonylurea receptor SUR1 subunits, but not with cromakalim, a selective opener for SUR2-based channels, indicating that SUR1-based K(ATP) channels conveyed enhanced sensitivity to elevated H2O2. In contrast, when endogenous H2O2 levels were increased after inhibition of catalase, the predominant peroxidase in the substantia nigra, with 3-amino-1,2,4-triazole (1 mM), all dopamine neurons responded with glibenclamide-reversible hyperpolarization. Fluorescence imaging of H2O2 indicated that catalase inhibition rapidly amplified intracellular H2O2, whereas inhibition of GSH peroxidase, a predominantly glial enzyme, caused a slower, smaller increase, especially in nonresponsive cells. Thus, endogenous H2O2 modulates neuronal activity via K(ATP) channel opening, thereby enhancing the reciprocal relationship between metabolism and excitability.
Collapse
Affiliation(s)
- Marat V Avshalumov
- Department of Neurosurgery, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | |
Collapse
|
23
|
Ohtsuka T, Ishiwa D, Kamiya Y, Itoh H, Nagata I, Saito Y, Yamada Y, Sumitomo M, Andoh T. Effects of barbiturates on ATP-sensitive K channels in rat substantia nigra. Neuroscience 2005; 137:573-81. [PMID: 16289884 DOI: 10.1016/j.neuroscience.2005.08.078] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2005] [Revised: 08/07/2005] [Accepted: 08/31/2005] [Indexed: 10/25/2022]
Abstract
ATP-sensitive K channels are widely expressed in cytoplasmic membranes of neurons, and they couple cell metabolism to excitability. They are thought to be involved in neuroprotection against cell damage during hypoxia, ischemia and excitotoxicity by hyperpolarizing neurons and reducing excitability. Although barbiturates are often used in patients with brain ischemia, the effects of these agents on neuronal ATP-sensitive K channels have not been clarified. We studied the effects of thiopental and pentobarbital on surface ATP-sensitive K channels in principal neurons of rat substantia nigra pars compacta. Whole cell voltage- and current-clamp recordings were made using rat midbrain slices. ATP-sensitive K channels were activated by intracellular dialysis with an ATP-free pipette solution during perfusion with a glucose-free solution. When the pipette solution contained 4mM ATP and the perfusing solution contained 25 mM glucose, the membrane current at -60 mV remained stable. When intracellular ATP was depleted, hyperpolarization and an outward current developed slowly. Although thiopental did not affect the membrane current in the presence of ATP and glucose, it reversibly inhibited the hyperpolarization and outward current induced by intracellular ATP depletion at 100 and 300 microM. Thiopental reduced the ATP depletion-induced outward current by 4.7%, 36.7% and 87% at 30, 100 and 300 microM, respectively. The high dose of pentobarbital also exhibited similar effects on ATP-sensitive K channels. These results suggest that barbiturates at high concentrations but not at clinically relevant concentrations inhibit ATP-sensitive K channels activated by intracellular ATP depletion in rat substantia nigra.
Collapse
Affiliation(s)
- T Ohtsuka
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Thomzig A, Laube G, Prüss H, Veh RW. Pore-forming subunits of K-ATP channels, Kir6.1 and Kir6.2, display prominent differences in regional and cellular distribution in the rat brain. J Comp Neurol 2005; 484:313-30. [PMID: 15739238 DOI: 10.1002/cne.20469] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
K-ATP channels consist of two structurally different subunits: a pore-forming subunit of the Kir6.0-family (Kir6.1 or Kir6.2) and a sulfonylurea receptor (SUR1, SUR2, SUR2A, SUR2B) with regulatory activity. The functional diversity of K-ATP channels in brain is broad and of fundamental importance for neuronal activity. Here, using immunocytochemistry with monospecific antibodies against the Kir6.1 and Kir6.2 subunits, we analyze the regional and cellular distribution of both proteins in the adult rat brain. We find Kir6.2 to be widely expressed in all brain regions, suggesting that the Kir6.2 subunit forms the pore of the K-ATP channels in most neurons, presumably protecting the cells during cellular stress conditions such as hypoglycemia or ischemia. Especially in hypothalamic nuclei, in particular the ventromedial and arcuate nucleus, neurons display Kir6.2 immunoreactivity only, suggesting that Kir6.2 is the pore-forming subunit of the K-ATP channels in the glucose-responsive neurons of the hypothalamus. In contrast, Kir6.1-like immunolabeling is restricted to astrocytes (Thomzig et al. [2001] Mol Cell Neurosci 18:671-690) in most areas of the rat brain and very weak or absent in neurons. Only in distinct nuclei or neuronal subpopulations is a moderate or even strong Kir6.1 staining detected. The biological functions of these K-ATP channels still need to be elucidated.
Collapse
Affiliation(s)
- Achim Thomzig
- Centrum für Anatomie, Charité, Universitätsmedizin Berlin, D-10115 Berlin, Germany
| | | | | | | |
Collapse
|
25
|
Pisani A, Bonsi P, Calabresi P. Calcium signaling and neuronal vulnerability to ischemia in the striatum. Cell Calcium 2005; 36:277-84. [PMID: 15261483 DOI: 10.1016/j.ceca.2004.02.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2004] [Accepted: 02/18/2004] [Indexed: 02/01/2023]
Abstract
Neurons express extremely different sensitivity to ischemic insults. The neuronal vulnerability is region-specific and the striatum is among the most susceptible areas to ischemic damage. Projecting GABAergic medium-sized neurons are very sensitive to energy metabolism impairment, whereas interneurons are selectively spared. However, the reasons for this differential vulnerability are largely unknown. Calcium ions (Ca2+) are important intracellular messengers enabling several physiological processes. However, excessive Ca2+ influx from the extracellular space or release from internal stores can elevate Ca2+ to levels that exceed the capacity of single neurons to appropriately buffer such overload. This capacity also appears to be a peculiar feature of single neuronal subtypes. This review will provide a brief survey of the ionic basis underlying the differential responses to in vitro ischemia of distinct striatal neuronal subtypes, mainly focusing on the role of Ca2+. The potential relevance of these findings in the development of therapeutic strategies for acute stroke will be discussed.
Collapse
Affiliation(s)
- Antonio Pisani
- Dipartimento di Neuroscienze, Clinica Neurologica, Università di Roma Tor Vergata, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | | | | |
Collapse
|
26
|
Ishiwa D, Kamiya Y, Itoh H, Saito Y, Ohtsuka T, Yamada Y, Andoh T. Effects of isoflurane and ketamine on ATP-sensitive K channels in rat substantia nigra. Neuropharmacology 2004; 46:1201-1212. [PMID: 15111027 DOI: 10.1016/j.neuropharm.2004.01.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2003] [Revised: 12/19/2003] [Accepted: 01/19/2004] [Indexed: 11/16/2022]
Abstract
Whole cell recordings were made using midbrain slices to examine the effects of two different anaesthetics on ATP-sensitive K (K(ATP)) channels in principle neurons of rat substantia nigra pars compacta. When neurons were dialyzed with an ATP-free pipette solution during perfusion with a glucose-free external solution, a hyperpolarization and an outward current developed slowly in a tolbutamide-inhibitable manner. The volatile anaesthetic 3% isoflurane slightly depolarised the neurons in the presence of ATP in the pipette solution and glucose in the external solution, but it did not affect the hyperpolarization or outward current in response to omission of ATP and glucose. Ketamine, an intravenous anaesthetic, did not change the membrane potential when ATP and glucose were included; however, it reversibly inhibited the hyperpolarization and outward current induced by intracellular ATP depletion in a dose-dependent manner. These effects of ketamine were not mimicked by AP-5, an NMDA receptor antagonist, or indatraline, an inhibitor of catecholamine uptake. These findings suggest that these anaesthetics have no stimulatory action on K(ATP) channels in these neurons when intracellular ATP is preserved and that ketamine but not isoflurane inhibits K(ATP) channels when the channels were activated by low intracellular ATP.
Collapse
Affiliation(s)
- Dai Ishiwa
- Department of Anaesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Nicholson R, Spanswick D, Lee K. Nitric oxide inhibits NMDA currents in a subpopulation of substantia gelatinosa neurons of the adult rat spinal cord. Neurosci Lett 2004; 359:180-4. [PMID: 15050693 DOI: 10.1016/j.neulet.2004.01.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2003] [Revised: 01/26/2004] [Accepted: 01/26/2004] [Indexed: 11/22/2022]
Abstract
In the present study, the actions of nitric oxide (NO) on NMDA mediated excitatory neurotransmission in substantia gelatinosa (SG) neurons of the adult rat spinal cord were investigated. Bath application of the NO donor sodium-nitroso-N-acetylpenicillamine (SNAP) reversibly inhibited NMDA receptor mediated excitatory postsynaptic currents evoked by electrical stimulation of the dorsal root. These effects of SNAP were prevented by pre-incubation with the NO scavenger 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (carboxy PTIO) or the guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-alpha]quinoxalin-1-one, and were mimicked by the membrane permeable cGMP analogue dibutyryl cGMP. Finally, these effects were shown to be mediated at a postsynaptic level using acutely applied exogenous NMDA. In conclusion, we demonstrate for the first time the inhibition of NMDA mediated excitatory neurotransmission by NO in spinal cord SG neurons, an action mediated at least in part via the activation of guanylate cyclase.
Collapse
Affiliation(s)
- Ruth Nicholson
- Department of Biological Sciences, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | | | | |
Collapse
|
28
|
de Rover M, Mansvelder HD, Lodder JC, Wardeh G, Schoffelmeer ANM, Brussaard AB. Long-lasting nicotinic modulation of GABAergic synaptic transmission in the rat nucleus accumbens associated with behavioural sensitization to amphetamine. Eur J Neurosci 2004; 19:2859-70. [PMID: 15147319 DOI: 10.1111/j.0953-816x.2004.03370.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A robust increase in dopaminergic transmission in the nucleus accumbens (NAc) shell has been reported to be consistently associated with the long-term expression of behavioural sensitization to drugs of abuse. However, little is known about how this affects the neuronal network of the NAc. We made cellular recordings in NAc slices of saline- and amphetamine-pretreated adult rats and found that expression of behavioural sensitization was associated with long-lasting changes in the basal firing pattern of cholinergic interneurons up to 3 weeks after the last drug injection. Consequently, upon amphetamine sensitization, an inhibiting effect of the nicotinic receptor blocker mecamylamine on the amplitudes of spontaneous GABAergic synaptic currents as well as on the failure rate of electrically evoked GABAergic currents was found that was not present under control conditions. Thus, behavioural sensitization to amphetamine is associated with an up-regulation of the endogenous activation of nicotinic receptors that, in turn, stimulate the GABAergic synaptic transmission within the NAc shell. This is a new mechanism by which drugs of abuse may induce alterations in the processing and integration of NAc inputs involved in psychomotor sensitization.
Collapse
Affiliation(s)
- Mischa de Rover
- Department of Experimental Neurophysiology, Institute for Neurosciences (INW), Centre for Neurogenomics and Cognitive Research, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
29
|
Héron-Milhavet L, Xue-Jun Y, Vannucci SJ, Wood TL, Willing LB, Stannard B, Hernandez-Sanchez C, Mobbs C, Virsolvy A, LeRoith D. Protection against hypoxic–ischemic injury in transgenic mice overexpressing Kir6.2 channel pore in forebrain. Mol Cell Neurosci 2004; 25:585-93. [PMID: 15080888 DOI: 10.1016/j.mcn.2003.10.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2002] [Revised: 09/23/2003] [Accepted: 10/21/2003] [Indexed: 11/26/2022] Open
Abstract
The role of the K-ATP channel pore-forming subunit Kir6.2 on protection from cerebral hypoxic-ischemic injury was assessed in transgenic mice overexpressing normal Kir6.2 or a dominant negative form (AFA) of this subunit in the forebrain. The resulting mice overexpress either the Kir6.2 or the AFA transgene mainly in the cerebral cortex and hippocampus. The Kir6.2 transgenic mice are resistant to hypoxic-ischemic injury showing a decreased region of cortical damage as compared to the dominant negative AFA and the wild-type mice. Moreover, the overexpression of Kir6.2 allowed an important silencing of the neurons present in forebrain regions thus protecting them from ischemic injury. Interestingly, the phenotype observed in Kir6.2 transgenic mice was observed without increased sulfonylurea binding. Taken together, these results indicate that the transgenic overexpression of Kir6.2 in forebrain significantly protects mice from hypoxic-ischemic injury and neuronal damage seen in stroke.
Collapse
Affiliation(s)
- Lisa Héron-Milhavet
- National Institutes of Health, Diabetes Branch, NIDDK, Bethesda, MD 20892-1758, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bonsi P, Calabresi P, De Persis C, Papa M, Centonze D, Bernardi G, Pisani A. Early ionic and membrane potential changes caused by the pesticide rotenone in striatal cholinergic interneurons. Exp Neurol 2004; 185:169-81. [PMID: 14697328 DOI: 10.1016/j.expneurol.2003.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mitochondrial metabolism impairment has been implicated in the pathogenesis of several neurodegenerative disorders. In the present work, we combined electrophysiological recordings and microfluorometric measurements from cholinergic interneurons obtained from a rat neostriatal slice preparation. Acute application of the mitochondrial complex I inhibitor rotenone produced an early membrane hyperpolarization coupled to a fall in input resistance, followed by a late depolarizing response. Current-voltage relationship showed a reversal potential of -80 +/- 3 mV, suggesting the involvement of a potassium (K+) current. Simultaneous measurement of intracellular sodium [Na+]i or calcium [Ca2+]i concentrations revealed a striking correlation between [Na+]i elevation and the early membrane hyperpolarization, whereas a significant [Ca2+]i rise matched the depolarizing phase. Interestingly, ion and membrane potential changes were mimicked by ouabain, inhibitor of the Na+-K+ATPase, and were insensitive to tetrodotoxin (TTX) or to a combination of glutamate receptor antagonists. The rotenone effects were partially reduced by blockers of ATP-sensitive K+ channels, glibenclamide and tolbutamide, and largely attenuated by a low Na+-containing solution. Morphological analysis of the rotenone effects on striatal slices showed a significant decrease in the number of choline acetyltransferase (ChAT) immunoreactive cells. These results suggest that rotenone rapidly disrupts the ATP content, leading to a decreased Na+-K+ATPase function and, therefore, to [Na+]i overload. In turn, the hyperpolarizing response might be generated both by the opening of ATP-sensitive K+ channels and by Na+-activated K+ conductances. The increase in [Ca2+]i occurs lately and does not seem to influence the early events.
Collapse
Affiliation(s)
- P Bonsi
- Clinica Neurologica, Dipartimento di Neuroscienze, Università di Roma Tor Vergata, Fondazione Santa Lucia, IRCCS, 00133 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
31
|
Thomzig A, Prüss H, Veh RW. The Kir6.1-protein, a pore-forming subunit of ATP-sensitive potassium channels, is prominently expressed by giant cholinergic interneurons in the striatum of the rat brain. Brain Res 2003; 986:132-8. [PMID: 12965237 DOI: 10.1016/s0006-8993(03)03222-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ATP-sensitive potassium channels comprise a complex of two structurally different proteins: a member of the inwardly rectifying Kir6 family (Kir6.1 or Kir6.2) and a sulfonylurea receptor (SUR1 or SUR2). Their regulation by intracellular ADP/ATP-concentrations and through various pharmacological agents has profound implications for the excitability of cells and, in the case of neurons, for neurotransmitter release. We previously showed that in rat brain, the Kir6.1 subunit is predominantly expressed in astrocytes in contrast to the Kir6.2 subunit, which is exclusively expressed in neurons. In this report we show, that in addition to the astrocytic expression, the Kir6.1 protein is also found in a small subset of neurons in distinct areas of the brain, like the hypothalamic supraoptic and paraventricular nuclei and the striatum. The Kir6.1-positive neurons in the striatum could be characterized as cholinergic interneurones, verified by immunofluorescence double staining. This complete colocalization of the Kir6.1 subunit in cholinergic interneurons is interesting with respect to the pharmacological potential of these channels. A selective modulation of the Kir6.1 subunit in the cholinergic striatal interneurons may eventually be of therapeutic value for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Achim Thomzig
- Institut für Anatomie, der Charité, Universitätsklinikum der Humboldt-Universität zu Berlin, Philippstrasse 12, D-10115 Berlin, Germany
| | | | | |
Collapse
|
32
|
Liu GJ, Simpson AM, Swan MA, Tao C, Tuch BE, Crawford RM, Jovanovic A, Martin DK. ATP-sensitive potassium channels induced in liver cells after transfection with insulin cDNA and the GLUT 2 transporter regulate glucose-stimulated insulin secretion. FASEB J 2003; 17:1682-4. [PMID: 12958175 PMCID: PMC2132862 DOI: 10.1096/fj.02-0051fje] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
As part of our research into the liver-directed gene therapy of Type I diabetes, we have engineered a human hepatoma cell line (HEPG2ins/g cells) to store and secrete insulin to a glucose stimulus. The aim of the present study was to determine whether HEPG2ins/g cells respond to glucose via signaling pathways that depend on ATP-sensitive potassium channels (KATP). Using patch-clamp electrophysiology with symmetrical KCl solutions, the single-channel conductance of KATP was 61pS. KATP was inhibited by ATP (1 mM) or cAMP (50 microM) applied to the cytosolic side of the membrane. Single KATP channels and macroscopic whole-cell currents were inhibited by glucose (20 mM) and glibenclamide (20 microM) and were activated by diazoxide (150 microM). Immunoprecipitation and Western blot analysis confirmed the presence of Kir6.2 KATP channel subunit protein in HEPG2ins/g and HEPG2ins cells. Using radioimmunoassay techniques, we report that exposure of the cells to tolbutamide (100 microM) resulted in an increase in insulin secretion from 0.3 +/- 0.05 to 1.8 +/- 0.2 pmol insulin/10(6) cells and glibenclamide (20 microM) from 0.4 +/- 0.06 to 2.1 +/- 0.3 (n=4), similar to what is seen on glucose (20 mM) stimulation. Diazoxide (150 microM) completely inhibited glucose-stimulated insulin release. Glucose 20 mM and glibenclamide 100 microM increased intracellular Ca2+ level in the HEPG2ins/g cells. However, glucose 20 mM did not stimulate a rise in intracellular Ca2+ in the un-transfected parent cell-line HEPG2. We used confocal microscopy to confirm that glucose (20 mM) stimulated the release of insulin from the fluorescently labeled secretion granules in the cells. Furthermore, glibenclamide (20 microM) also stimulated the release of insulin from fluorescently labeled secretion granules, and diazoxide (150 microM) blocked that stimulated release of insulin. Our results suggest that HEPG2ins/g cells respond to glucose via signaling pathways that depend on KATP, similar to a normal pancreatic beta cell.
Collapse
Affiliation(s)
- Guo Jun Liu
- Department of Health Sciences, University of Technology, Sydney, Broadway, NSW, Australia
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Marti M, Mela F, Ulazzi L, Hanau S, Stocchi S, Paganini F, Beani L, Bianchi C, Morari M. Differential responsiveness of rat striatal nerve endings to the mitochondrial toxin 3-nitropropionic acid: implications for Huntington's disease. Eur J Neurosci 2003; 18:759-67. [PMID: 12925002 DOI: 10.1046/j.1460-9568.2003.02806.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Rat striatal synaptosomes and slices were used to investigate the responsiveness of different populations of nerve terminals to 3-nitropropionic acid (3-NP), a suicide inhibitor of the mitochondrial enzyme succinate dehydrogenase, and to elucidate the ionic mechanisms involved. 3-NP (0.3-3 mm) stimulated spontaneous gamma-aminobutyric acid (GABA), glutamate and [3H]-dopamine efflux but left unchanged acetylcholine efflux from synaptosomes. This effect was associated with a >70% inhibition of succinate dehydrogenase, as measured in the whole synaptosomal population. The facilitation was not dependent on extracellular Ca2+ but relied on voltage-dependent Na+ channel opening, because it was prevented by tetrodotoxin and riluzole. 3-NP also elevated spontaneous glutamate efflux from slices but in a tetrodotoxin-insensitive way. To investigate whether energy depletion could change the responsiveness of nerve endings to a depolarizing stimulus, synaptosomes were pretreated with 3-NP and challenged with pulses of KCl evoking 'quasi-physiological' neurotransmitter release. 3-NP potentiated the K+-evoked GABA, glutamate and [3H]-dopamine release but inhibited the K+-evoked acetylcholine release. The 3-NP induced potentiation of GABA release was Ca2+-dependent and prevented by tetrodotoxin and riluzole whereas the 3-NP-induced inhibition of acetylcholine release was tetrodotoxin- and riluzole-insensitive but reversed by glipizide, an ATP-dependent K+ channel inhibitor. We conclude that the responsiveness of striatal nerve endings to 3-NP relies on activation of different ionic conductances, and suggest that the selective survival of striatal cholinergic interneurons following chronic 3-NP treatment (as in models of Huntington's disease) may rely on the opening of ATP-dependent K+ channels, which counteracts the fall in membrane potential as a result of mitochondrial impairment.
Collapse
Affiliation(s)
- Matteo Marti
- Department of Experimental and Clinical Medicine, Section of Pharmacology, via Fossato di Mortara 17-19, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Levin BE, Dunn-Meynell AA, Routh VH. CNS sensing and regulation of peripheral glucose levels. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 51:219-58. [PMID: 12420361 DOI: 10.1016/s0074-7742(02)51007-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is clear that the brain has evolved a mechanism for sensing levels of ambient glucose. Teleologically, this is likely to be a function of its requirement for glucose as a primary metabolic substrate. There is no question that the brain can sense and mount a counterregulatory response to restore very low levels of plasma and brain glucose. But it is less clear that the changes in glucose associated with normal diurnal rhythms and feeding cycles are sufficient to influence either ingestive behavior or the physiologic responses involved in regulating plasma glucose levels. Glucosensing neurons are clearly a distinct class of metabolic sensors with the capacity to respond to a variety of intero- and exteroceptive stimuli. This makes it likely that these glucosensing neurons do participate in physiologically relevant homeostatic mechanisms involving energy balance and the regulation of peripheral glucose levels. It is our challenge to identify the mechanisms by which these neurons sense and respond to these metabolic cues.
Collapse
Affiliation(s)
- Barry E Levin
- Neurology Service, VA Medical Center, East Orange, New Jersey 07018, USA
| | | | | |
Collapse
|
35
|
Smith MA, Herson PS, Lee K, Pinnock RD, Ashford MLJ. Hydrogen-peroxide-induced toxicity of rat striatal neurones involves activation of a non-selective cation channel. J Physiol 2003; 547:417-25. [PMID: 12562896 PMCID: PMC2342643 DOI: 10.1113/jphysiol.2002.034561] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Striatal neurones are particularly vulnerable to hypoxia/ischaemia-induced damage, and free radicals are thought to be prime mediators of this neuronal destruction. It has been shown that hydrogen peroxide (H2O2), through the production of free radicals, induces rat insulinoma cell death by activation of a non-selective cation channel, which leads to irreversible cell depolarization and unregulated Ca2+ entry into the cell. In the study presented here, we demonstrate that a subpopulation of striatal neurones (medium spiny neurones) is depolarized by H2O2 through the production of free radicals. Cell-attached recordings from rat cultured striatal neurones demonstrate that exposure to H2O2 opens a large-conductance channel that is characterized by extremely long open times (seconds). Inside-out recordings show that cytoplasmically applied beta-nicotinamide adenine dinucleotide activates a channel with little voltage dependence, a linear current-voltage relationship and a single-channel conductance of between 70 and 90 pS. This channel is permeable to Na+, K+ and Ca2+ ions. Fura-2 imaging from cultured striatal neurones reveals that H2O2 exposure induces a biphasic intracellular Ca2+ increase in a subpopulation of neurones, the second, later phase resulting in Ca2+ overload. This later component of the Ca2+ response is dependent on the presence of extracellular Ca2+ and is independent of synaptic activity or voltage-gated Ca2+ channel opening. Consequently, this channel may be an important contributor to free radical-induced selective striatal neurone destruction. These results are remarkably similar to those observed for insulinoma cells and suggest that this family of non-selective cation channels has a widespread distribution in mammalian tissues.
Collapse
Affiliation(s)
- M A Smith
- Department of Biomedical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | | | | | | | | |
Collapse
|
36
|
Abstract
ATP-sensitive potassium channels (K(ATP)) of vascular smooth muscle cells represent potential therapeutic targets for control of abnormal vascular contractility. The biophysical properties, regulation and pharmacology of these channels have received intense scrutiny during the past twenty years, however, the molecular basis of vascular K(ATP) channels remains ill-defined. This review summarizes the recent advancements made in our understanding of the molecular composition of vascular K(ATP) channels with a focus on the evidence that hetero-octameric complexes of Kir6.1 and SUR2B subunits constitute the vascular K(ATP) subtype responsible for control of arterial diameter by vasoactive agonists.
Collapse
Affiliation(s)
- William C Cole
- The Smooth Muscle Research Group, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | | |
Collapse
|
37
|
Apicella P. Tonically active neurons in the primate striatum and their role in the processing of information about motivationally relevant events. Eur J Neurosci 2002; 16:2017-26. [PMID: 12473069 DOI: 10.1046/j.1460-9568.2002.02262.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Analysis of recordings of single neuronal activity in the striatum of monkeys engaged in behavioural tasks has shown that tonically active neurons (TANs) can be distinguished by their distinct spontaneous firing and functional properties. As TANs are assumed to be cholinergic interneurons, the study of their physiological characteristics allows us to gain an insight into the role of a particular type of local-circuit neuron in the processing of information at the striatal level. In monkeys performing various behavioural tasks, the change in the activity of TANs, unlike the diversity of task-related activations exhibited by the phasically active population of striatal neurons, involves a transient depression of the tonic firing related to environmental events of motivational significance. Such events include primary rewards and stimuli that have acquired a reward value during associative learning. These neurons also respond to an aversive air puff, indicating that their responsiveness is not restricted to appetitive conditions. Another striking feature of the TANs is that their responses can be modulated by predictions about stimulus timing. Temporal variations in event occurrence have been found to favour the responses of TANs, whereas the responses are diminished or abolished in the presence of external cues that predict the time at which events will occur. These data suggest that the TANs respond as do detectors of motivationally relevant events, but they also demonstrate that these neurons are influenced by predictive information based on past experience with a given temporal context. TANs represent a unique subset of striatal neurons that might serve a modulatory function, monitoring for temporal relationships between environmental events.
Collapse
Affiliation(s)
- Paul Apicella
- Institut de Neurosciences Physiologiques et Cognitives, CNRS, 31 chemin Joseph Aiguier, 13402 Marseille cedex 20, France.
| |
Collapse
|
38
|
Dixon AK, Lee K, Richardson PJ, Bell MI, Skynner MJ. Single cell expression analysis--pharmacogenomic potential. Pharmacogenomics 2002; 3:809-22. [PMID: 12437482 DOI: 10.1517/14622416.3.6.809] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
A fundamental challenge in biology is to correlate physiology with gene expression in specific cell types. This can only be achieved by understanding gene expression at the level of the single cell because, in many systems, each cell has the capacity to express a unique set of genes. Therefore, each cell can be considered to be functionally distinct. A clearer understanding of gene expression differences at such a discrete level provides an opportunity to develop drugs with more targeted pharmacologies or with decreased side effects.
Collapse
Affiliation(s)
- A K Dixon
- Cambridge Biotechnology Ltd, Dept of Pharmacology, Tennis Court Road, UK
| | | | | | | | | |
Collapse
|
39
|
Müller M, Brockhaus J, Ballanyi K. ATP-independent anoxic activation of ATP-sensitive K+ channels in dorsal vagal neurons of juvenile mice in situ. Neuroscience 2002; 109:313-28. [PMID: 11801367 DOI: 10.1016/s0306-4522(01)00498-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The role of ATP in anoxic activation of ATP-sensitive K+ (KATP) channels was studied in dorsal vagal neurons of mouse brainstem slices. In the whole-cell configuration, cyanide-induced chemical anoxia evoked within 10 s a 300-pA outward current that gave rise to a hyperpolarization of 24 mV. These responses were mimicked by nitrogen-aerated saline, rotenone or diazoxide and abolished by tolbutamide. The cyanide-induced hyperpolarization was due to activation of 70 pS K(ATP) channels that were half-maximally blocked by 5 microM internal ATP. Dialyzing the cells with either 1, 20 or 0 mM ATP did not, however, affect the time to onset, the kinetics or the magnitude of the cyanide-induced hyperpolarization. Impairment of ATP consumption by ouabain, vanadate or reduced temperature had no effect either. Thus, anoxia-induced activation of these KATP channels cannot be explained by a fall of cellular ATP or a concomitant rise of ADP. Anoxia-related changes of the actin cytoskeleton or the composition of the plasma membrane are also not likely to be involved, as cytochalasin D did not affect the cyanide-evoked hyperpolarization and phosphatidylinositol 4,5-bisphosphate failed to decrease the ATP sensitivity of single KATP channels. Finally, because of a lack of effects of reduced/oxidized glutathione and the oxidase blocker diphenyliodonium on the cyanide-induced hyperpolarization, cellular redox state does not appear to be involved. Our results indicate that despite a high sensitivity to ATP in excised patches, anoxic activation of KATP channels is independent of cellular ATP. Rather the ATP block seems to be removed as a consequence of impaired mitochondrial function.
Collapse
Affiliation(s)
- M Müller
- II. Physiologisches Institut, Georg-August-Universität Göttingen, Humboldtalle 23, D-37073 Göttingen, Germany
| | | | | |
Collapse
|
40
|
Bell MI, Richardson PJ, Lee K. Functional and molecular characterization of metabotropic glutamate receptors expressed in rat striatal cholinergic interneurones. J Neurochem 2002; 81:142-9. [PMID: 12067226 DOI: 10.1046/j.1471-4159.2002.00815.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the present study we have used single-cell RT-PCR in conjunction with electrophysiology to examine the expression and functional properties of metabotropic glutamate receptors (mGluRs) expressed within biochemically identified cholinergic interneurones in the rat striatum. Using single-cell RT-PCR, it was possible to demonstrate the presence of mGluR1, mGluR2, mGluR3, mGluR5 and mGluR7 mRNAs within single cholinergic interneurones. Bath application of the non-selective mGluR agonist (1 S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid (1 S,3R-ACPD) or the group-I mGluR agonist 3,5-dihydroxyphenylglycine (DHPG) depolarized all cholinergic neurones tested by activation of an inward current at -60 mV. The effects of DHPG were partially inhibited by the mGluR5 selective antagonist 6-methyl-2-(pherazo)-3-pyridinol and by the non-selective group-I antagonist alpha-methyl-4-carboxyphenylglycine but were not mimicked by the group-II and group-III selective mGluR agonists 2-(2,3-dicarboxycyclopropyl)glycine (DCG-IV) and L-2-amino-4-phosphonobutanoate (L-AP4), respectively. Intrastriatal stimulation evoked an excitatory postsynaptic current within cholinergic neurones that was reversibly inhibited by bath application of the group-II and group-III selective mGluR agonists DCG-IV and L-AP4, respectively, via presynaptic actions. In summary, we have identified the mGluRs expressed by striatal cholinergic interneurones and demonstrated that their activation produces modulatory effects via both pre- and postsynaptic mechanisms.
Collapse
Affiliation(s)
- Matt I Bell
- Pfizer Global Research and Development, Cambridge University Forvie Site, UK
| | | | | |
Collapse
|
41
|
Cepeda C, Itri JN, Flores-Hernández J, Hurst RS, Calvert CR, Levine MS. Differential sensitivity of medium- and large-sized striatal neurons to NMDA but not kainate receptor activation in the rat. Eur J Neurosci 2001; 14:1577-89. [PMID: 11860453 DOI: 10.1046/j.0953-816x.2001.01783.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Infrared videomicroscopy and differential interference contrast optics were used to identify medium- and large-sized neurons in striatal slices from young rats. Whole-cell patch-clamp recordings were obtained to compare membrane currents evoked by application of N-methyl-d-aspartate (NMDA) and kainate. Inward currents and current densities induced by NMDA were significantly smaller in large- than in medium-sized striatal neurons. The negative slope conductance for NMDA currents was greater in medium- than in large-sized neurons and more depolarization was required to remove the Mg2+ blockade. In contrast, currents induced by kainate were significantly greater in large-sized neurons whilst current densities were approximately equal in both cell types. Spontaneous excitatory postsynaptic currents occurred frequently in medium-sized neurons but were relatively infrequent in large-sized neurons. Excitatory postsynaptic currents evoked by electrical stimulation were smaller in large- than in medium-sized neurons. A final set of experiments assessed a functional consequence of the differential sensitivity of medium- and large-sized neurons to NMDA. Cell swelling was used to examine changes in somatic area in both neuronal types after prolonged application of NMDA or kainate. NMDA produced a time-dependent increase in somatic area in medium-sized neurons whilst it produced only minimal changes in large interneurons. In contrast, application of kainate produced significant swelling in both medium- and large-sized cells. We hypothesize that reduced sensitivity to NMDA may be due to variations in receptor subunit composition and/or the relative density of receptors in the two cell types. These findings help define the conditions that put neurons at risk for excitotoxic damage in neurological disorders.
Collapse
Affiliation(s)
- C Cepeda
- Mental Retardation Research Center and Brain Research Institute, UCLA School of Medicine, 760 Westwood Plaza NPI Room 58-258, Los Angeles, CA 90024, USA
| | | | | | | | | | | |
Collapse
|
42
|
Centonze D, Marfia GA, Pisani A, Picconi B, Giacomini P, Bernardi G, Calabresi P. Ionic mechanisms underlying differential vulnerability to ischemia in striatal neurons. Prog Neurobiol 2001; 63:687-96. [PMID: 11165001 DOI: 10.1016/s0301-0082(00)00037-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Brain cells express extremely different sensitivity to ischemic insults. The reason for this differential vulnerability is still largely unknown. Here we discuss the ionic bases underlying the physiological responses to in vitro ischemia in two neostriatal neuronal subtypes exhibiting respectively high sensitivity and high resistance to energy deprivation. Vulnerable neostriatal neurons respond to ischemia with a membrane depolarization. This membrane depolarization mainly depends on the increased permeability to Na+ ions. In contrast, resistant neostriatal neurons respond to ischemia with a membrane hyperpolarization due to the opening of K+ channels. Interestingly, in both neuronal subtypes the ischemia-dependent membrane potential changes can be significantly enhanced or attenuated by a variety of pharmacological agents interfering with intracellular Ca2+ entry, ATP-dependent K+ channels opening, and Na+/Ca2+ exchanger functioning. The understanding of the ionic mechanisms underlying the differential membrane responses to ischemia represents the basis for the development of rational neuroprotective treatments during acute cerebrovascular insults.
Collapse
|
43
|
Hernández-Sánchez C, Basile AS, Fedorova I, Arima H, Stannard B, Fernandez AM, Ito Y, LeRoith D. Mice transgenically overexpressing sulfonylurea receptor 1 in forebrain resist seizure induction and excitotoxic neuron death. Proc Natl Acad Sci U S A 2001; 98:3549-54. [PMID: 11248115 PMCID: PMC30690 DOI: 10.1073/pnas.051012898] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The ability of the sulfonylurea receptor (SUR) 1 to suppress seizures and excitotoxic neuron damage was assessed in mice transgenically overexpressing this receptor. Fertilized eggs from FVB mice were injected with a construct containing SUR cDNA and a calcium-calmodulin kinase IIalpha promoter. The resulting mice showed normal gross anatomy, brain morphology and histology, and locomotor and cognitive behavior. However, they overexpressed the SUR1 transgene, yielding a 9- to 12-fold increase in the density of [(3)H]glibenclamide binding to the cortex, hippocampus, and striatum. These mice resisted kainic acid-induced seizures, showing a 36% decrease in average maximum seizure intensity and a 75% survival rate at a dose that killed 53% of the wild-type mice. Kainic acid-treated transgenic mice showed no significant loss of hippocampal pyramidal neurons or expression of heat shock protein 70, whereas wild-type mice lost 68-79% of pyramidal neurons in the CA1-3 subfields and expressed high levels of heat shock protein 70 after kainate administration. These results indicate that the transgenic overexpression of SUR1 alone in forebrain structures significantly protects mice from seizures and neuronal damage without interfering with locomotor or cognitive function.
Collapse
Affiliation(s)
- C Hernández-Sánchez
- Section on Molecular and Cellular Physiology, Clinical Endocrinology Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Intrinsic membrane properties underlying spontaneous tonic firing in neostriatal cholinergic interneurons. J Neurosci 2001. [PMID: 11069957 DOI: 10.1523/jneurosci.20-22-08493.2000] [Citation(s) in RCA: 213] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neostriatal cholinergic interneurons produce spontaneous tonic firing in the absence of synaptic input. Perforated patch recording and whole-cell recording combined with calcium imaging were used in vitro to identify the intrinsic membrane properties underlying endogenous excitability. Spontaneous firing was driven by the combined action of a sodium current and the hyperpolarization-activated cation current (I(h)), which together ensured that there was no zero current point in the subthreshold voltage range. Blockade of sodium channels or I(h) established a stable subthreshold resting membrane potential. A tetrodotoxin-sensitive region of negative slope conductance was observed between approximately -60 mV and threshold (approximately -50 mV) and the h-current was activated at all subthreshold voltages. Calcium imaging experiments revealed that there was minimal calcium influx at subthreshold membrane potentials but that action potentials produced elevations of calcium in both the soma and dendrites. Spike-triggered calcium entry shaped the falling phase of the action potential waveform and activated calcium-dependent potassium channels. Blockade of big-conductance channels caused spike broadening. Application of apamin, which blocks small-conductance channels, abolished the slow spike afterhyperpolarization (AHP) and caused a transition to burst firing. In the absence of synaptic input, a range of tonic firing patterns are observed, suggesting that the characteristic spike sequences described for tonically active cholinergic neurons (TANs) recorded in vivo are intrinsic in origin. The pivotal role of the AHP in regulating spike patterning indicates that burst firing of TANs in vivo could arise from direct or indirect modulation of the AHP without requiring phasic synaptic input.
Collapse
|
45
|
Bennett BD, Callaway JC, Wilson CJ. Intrinsic membrane properties underlying spontaneous tonic firing in neostriatal cholinergic interneurons. J Neurosci 2000; 20:8493-503. [PMID: 11069957 PMCID: PMC6773196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Neostriatal cholinergic interneurons produce spontaneous tonic firing in the absence of synaptic input. Perforated patch recording and whole-cell recording combined with calcium imaging were used in vitro to identify the intrinsic membrane properties underlying endogenous excitability. Spontaneous firing was driven by the combined action of a sodium current and the hyperpolarization-activated cation current (I(h)), which together ensured that there was no zero current point in the subthreshold voltage range. Blockade of sodium channels or I(h) established a stable subthreshold resting membrane potential. A tetrodotoxin-sensitive region of negative slope conductance was observed between approximately -60 mV and threshold (approximately -50 mV) and the h-current was activated at all subthreshold voltages. Calcium imaging experiments revealed that there was minimal calcium influx at subthreshold membrane potentials but that action potentials produced elevations of calcium in both the soma and dendrites. Spike-triggered calcium entry shaped the falling phase of the action potential waveform and activated calcium-dependent potassium channels. Blockade of big-conductance channels caused spike broadening. Application of apamin, which blocks small-conductance channels, abolished the slow spike afterhyperpolarization (AHP) and caused a transition to burst firing. In the absence of synaptic input, a range of tonic firing patterns are observed, suggesting that the characteristic spike sequences described for tonically active cholinergic neurons (TANs) recorded in vivo are intrinsic in origin. The pivotal role of the AHP in regulating spike patterning indicates that burst firing of TANs in vivo could arise from direct or indirect modulation of the AHP without requiring phasic synaptic input.
Collapse
Affiliation(s)
- B D Bennett
- Cajal Neuroscience Research Center, Division of Life Sciences, University of Texas, San Antonio, Texas 78249, USA
| | | | | |
Collapse
|
46
|
Pelletier MR, Pahapill PA, Pennefather PS, Carlen PL. Analysis of single K(ATP) channels in mammalian dentate gyrus granule cells. J Neurophysiol 2000; 84:2291-301. [PMID: 11067973 DOI: 10.1152/jn.2000.84.5.2291] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ATP-sensitive potassium (K(ATP)) channels are heteromultimer complexes of subunits from members of the inwardly rectifying K(+) channel and the ATP-binding cassette protein superfamilies. K(ATP) channels couple metabolic state to membrane excitability, are distributed widely, and participate in a variety of physiological functions. Understood best in pancreatic beta cells, where their activation inhibits insulin release, K(ATP) channels have been implicated also in postischemia cardio- and neuroprotection. The dentate gyrus (DG) is a brain region with a high density of K(ATP) channels and is relatively resistant to ischemia/reperfusion-induced cell death. Therefore we were interested in describing the characteristics of single K(ATP) channels in DG granule cells. We recorded single K(ATP) channels in 59/105 cell-attached patches from DG granule cells in acutely prepared hippocampal slices. Single-channel openings had an E(K) close to 0 mV (symmetrical K(+)) and were organized in bursts with a duration of 19.3 +/- 1.6 (SE) ms and a frequency of 3.5 +/- 0.8 Hz, a unitary slope conductance of 27 pS, and a low, voltage-independent, probability of opening (P(open), 0.04 +/- 0.01). Open and closed dwell-time histograms were fitted best with one (tau(open) = 1.3 +/- 0.2 ms) and the sum of two (tau(closed,fast) = 2.6 +/- 0.9 ms, tau(closed,slow) = 302.7 +/- 67. 7 ms) exponentials, respectively, consistent with a kinetic model having at least a single open and two closed states. The P(open) was reduced ostensibly to zero by the sulfonylureas, glybenclamide (500 nM, 2/6; 10 microM,11/14 patches) and tolbutamide (20 microM, 4/6; 100 microM, 4/4 patches). The blocking dynamics for glybenclamide included transition to a subconductance state (43.3 +/- 2.6% of control I(open channel)). Unlike glybenclamide, the blockade produced by tolbutamide was reversible. In 5/5 patches, application of diazoxide (100 microM) increased significantly P(open) (0.12 +/- 0.02), which was attributable to a twofold increase in the frequency of bursts (8.3 +/- 2.0 Hz). Diazoxide was without effect on tau(open) and tau(closed,fast) but decreased significantly tau(closed,slow) (24.4 +/- 2.6 ms). We observed similar effects in 6/7 patches after exposure to hypoxia/hypoglycemia, which increased significantly P(open) (0.09 +/- 0.03) and the frequency of bursts (7.1 +/- 1.7 Hz) and decreased significantly tau(closed,slow) (29.5 +/- 1.8 ms). We have presented convergent evidence consistent with single K(ATP) channel activity in DG granule cells. The subunit composition of K(ATP) channels native to DG granule cells is not known; however, the characteristics of the channel activity we recorded are representative of Kir6.1/SUR1, SUR2B-based channels.
Collapse
Affiliation(s)
- M R Pelletier
- Bloorview Epilepsy Research Laboratory, Division of Cellular and Molecular Biology, Toronto Western Research Institute, University of Toronto, University Health Network, Toronto, Ontario M5T 2S8, Canada
| | | | | | | |
Collapse
|
47
|
Bell MI, Richardson PJ, Lee K. Histamine depolarizes cholinergic interneurones in the rat striatum via a H(1)-receptor mediated action. Br J Pharmacol 2000; 131:1135-42. [PMID: 11082121 PMCID: PMC1572445 DOI: 10.1038/sj.bjp.0703692] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. Whole-cell patch clamp recordings were made from rat striatal cholinergic interneurones in slices of brain tissue in vitro. Bath application of histamine (EC(50) 6.3 microM) was found to rapidly and reversibly depolarize these neurones through the induction of an inward current at -60 mV. 2. The effects of histamine were mimicked by the H(1) receptor agonist 2-thiazolylethylamine (50 microM) and selectively inhibited by pre-incubation with the H(1) receptor antagonist triprolidine (1 microM). 3. Ion substitution experiments under voltage clamp conditions revealed that the histamine activated current was comprised of two components. One component was sensitive to the concentration of extracellular Na(+), whilst the other component was inhibited by intracellular Cs(+) or extracellular Ba(2+). 4. In situ hybridization experiments revealed that the majority of cholinergic interneurones in the rat striatum express the histamine H(1) receptor but few neurones express H(2) receptors. These findings were confirmed using single cell RT - PCR. 5. It is concluded that histamine depolarizes cholinergic interneurones in the rat striatum via a H(1)-receptor mediated mechanism.
Collapse
Affiliation(s)
- M I Bell
- Parke Davis Neuroscience Research Centre, Cambridge University Forvie Site, Cambridge, CB2 2QB
| | - P J Richardson
- Department of Pharmacology, Tennis Court Road, Cambridge, CB2 1QJ
| | - K Lee
- Parke Davis Neuroscience Research Centre, Cambridge University Forvie Site, Cambridge, CB2 2QB
- Author for correspondence:
| |
Collapse
|
48
|
Malhi H, Irani AN, Rajvanshi P, Suadicani SO, Spray DC, McDonald TV, Gupta S. KATP channels regulate mitogenically induced proliferation in primary rat hepatocytes and human liver cell lines. Implications for liver growth control and potential therapeutic targeting. J Biol Chem 2000; 275:26050-7. [PMID: 10862612 DOI: 10.1074/jbc.m001576200] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To determine whether K(ATP) channels control liver growth, we used primary rat hepatocytes and several human cancer cell lines for assays. K(ATP) channel openers (minoxidil, cromakalim, and pinacidil) increased cellular DNA synthesis, whereas K(ATP) channel blockers (quinidine and glibenclamide) attenuated DNA synthesis. The channel inhibitor glibenclamide decreased the clonogenicity of HepG2 cells without inducing cytotoxicity or apoptosis. To demonstrate the specificity of drugs for K(+) channels, whole-cell patch-clamp recordings were made. Hepatocytes revealed K(+) currents with K(ATP) channel properties. These K(+) currents were augmented by minoxidil and pinacidil and attenuated by glibenclamide as well as tetraethylammonium, in agreement with established responses of K(ATP) channels. Reverse transcription of total cellular RNA followed by polymerase chain reaction showed expression of K(ATP) channel-specific subunits in rat hepatocytes and human liver cell lines. Calcium fluxes were unperturbed in glibenclamide-treated HepG2 cells and primary rat hepatocytes following induction with ATP and hepatocyte growth factor, respectively, suggesting that the effect of K(ATP) channel activity upon hepatocyte proliferation was not simply due to indirect modulation of intracellular calcium. The regulation of mitogen-related hepatocyte proliferation by K(ATP) channels advances our insights into liver growth control. The findings have implications in mechanisms concerning liver development, regeneration, and oncogenesis.
Collapse
Affiliation(s)
- H Malhi
- Marion Bessin Liver Research Center, the Cancer Research Center, and the Departments of Medicine and Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Preston Z, Lee K, Widdowson L, Freeman TC, Dixon AK, Richardson PJ. Adenosine receptor expression and function in rat striatal cholinergic interneurons. Br J Pharmacol 2000; 130:886-90. [PMID: 10864896 PMCID: PMC1572128 DOI: 10.1038/sj.bjp.0703366] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Cholinergic neurons were identified in rat striatal slices by their size, membrane properties, sensitivity to the NK(1) receptor agonist (Sar(9), Met(O(2))(11)) Substance P, and expression of choline acetyltransferase mRNA. A(1) receptor mRNA was detected in 60% of the neurons analysed, and A(2A) receptor mRNA in 67% (n=15). The A(1) receptor agonist R-N(6)-(2-phenylisopropyl)adenosine (R-PIA) hyperpolarized cholinergic neurons in a concentration dependent manner sensitive to the A(1) antagonist 8-cyclopentyl-1, 3-dipropylxanthine (DPCPX, 100 nM). In dual stimulus experiments, the A(2A) receptor antagonist 8-(3-chlorostyryl)caffeine (CSC, 500 nM) decreased release of [(3)H]-acetylcholine from striatal slices (S2/S1 0.78+/-0.07 versus 0.95+/-0.05 in control), as did adenosine deaminase (S2/S1 ratio 0.69+/-0.05), whereas the A(1) receptor antagonist DPCPX (100 nM) had no effect (S2/S1 1.05+/-0.14). In the presence of adenosine deaminase the adenosine A(2A) receptor agonist 2-p-((carboxyethyl)phenylethylamino)-5'-N-ethylcarboxamidoadeno sin e (CGS21680, 10 nM) increased release (S2/S1 ratio 1.03+/-0.05 versus 0.88+/-0.05 in control), an effect blocked by the antagonist CSC (500 nM, S2/S1 0.68+/-0.05, versus 0.73+/-0.08 with CSC alone). The combined superfusion of bicuculline (10 microM), saclofen (1 microM) and naloxone (10 microM) had no effect on the stimulation by CGS21680 (S2/S1 ratio 0.99+/-0.04). The A(1) receptor agonist R-PIA (100 nM) inhibited the release of [(3)H]-acetylcholine (S2/S1 ratio 0.70+/-0.03), an effect blocked by DPCPX (S2/S1 ratio 1.06+/-0.07). It is concluded that both A(1) and A(2A) receptors are expressed on striatal cholinergic neurons where they are functionally active.
Collapse
Affiliation(s)
- Zoe Preston
- Parke-Davis, Cambridge University Forvie Site, Robinson Way, Cambridge, CB2 2QB
| | - Kevin Lee
- Parke-Davis, Cambridge University Forvie Site, Robinson Way, Cambridge, CB2 2QB
| | - Leon Widdowson
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QJ
| | - Tom C Freeman
- The Sanger Centre, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA
| | - Alistair K Dixon
- Parke-Davis, Cambridge University Forvie Site, Robinson Way, Cambridge, CB2 2QB
- The Sanger Centre, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA
| | - Peter J Richardson
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QJ
- Author for correspondence:
| |
Collapse
|
50
|
Preston Z, Lee K, Widdowson L, Richardson PJ, Pinnock RD. Tachykinins increase [3H]acetylcholine release in mouse striatum through multiple receptor subtypes. Neuroscience 2000; 95:367-76. [PMID: 10658616 DOI: 10.1016/s0306-4522(99)00440-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Tachykinins have been suggested to play a significant role in the mammalian striatum, at least in part by the control of acetylcholine release from cholinergic interneurons. In the present study, we have examined the ability of known tachykinin agonists and antagonists to modulate the activity of these interneurons in mouse striatal slices. Using whole-cell patch-clamp recordings, the selective neurokinin-1, neurokinin-2 and neurokinin-3 receptor agonists [sar9,Met(O2)11]substance P, [beta-ala8]neurokinin A(4-10) and senktide each produced a dose-dependent depolarization of visually identified cholinergic interneurons that was retained under conditions designed to interrupt synaptic transmission. The nature of these neurons and the expression of multiple tachykinin receptors was confirmed using single-cell reverse transcriptase-polymerase chain reaction analysis. Using in vitro superfusion techniques, the selective neurokinin-1, neurokinin-2 and neurokinin-3 receptor agonists [sar9,Met(O2)11]substance P, [beta-ala8]neurokinin A(4-10) and senktide, respectively, each produced a dose-dependent increase in acetylcholine release, the selectivity of which was confirmed using the neurokinin-1, neurokinin-2 and neurokinin-3 receptor antagonists SR140333, GR94800 and SR142801 (100 nM). U73122 (10 microM), a phospholipase C inhibitor, blocked [sar9,Met(O2)11]substance P- and senktide-induced acetylcholine release, but had no effect on [beta-ala8]neurokinin A(4-10)-induced release. The protein kinase C inhibitors chelerythrine and Ro-31-8220 (both 1 microM) significantly inhibited responses induced by all three agonists. These findings indicate that tachykinins modulate the activity of mouse striatal cholinergic interneurons. Furthermore, neurokinin-2 receptors are shown to perform a role in mouse that has not been identified previously in other species.
Collapse
MESH Headings
- 2-Amino-5-phosphonovalerate/pharmacology
- Acetylcholine/pharmacokinetics
- Alkaloids
- Animals
- Benzophenanthridines
- Choline O-Acetyltransferase/genetics
- Choline O-Acetyltransferase/metabolism
- Corpus Striatum/chemistry
- Corpus Striatum/metabolism
- Electrophysiology
- Enzyme Inhibitors/pharmacology
- Estrenes/pharmacology
- Excitatory Amino Acid Antagonists/pharmacology
- In Situ Hybridization
- Indoles/pharmacology
- Interneurons/chemistry
- Interneurons/enzymology
- Membrane Potentials/drug effects
- Membrane Potentials/physiology
- Mice
- Mice, Inbred C57BL
- Neurokinin A/analogs & derivatives
- Neurokinin A/pharmacology
- Oligopeptides/pharmacology
- Peptide Fragments/pharmacology
- Phenanthridines/pharmacology
- Phosphodiesterase Inhibitors/pharmacology
- Piperidines/pharmacology
- Pyrrolidinones/pharmacology
- Quinoxalines/pharmacology
- Quinuclidines/pharmacology
- RNA, Messenger/analysis
- Receptors, Neurokinin-1/genetics
- Receptors, Neurokinin-1/metabolism
- Receptors, Neurokinin-2/genetics
- Receptors, Neurokinin-2/metabolism
- Receptors, Neurokinin-3/antagonists & inhibitors
- Receptors, Neurokinin-3/genetics
- Receptors, Neurokinin-3/metabolism
- Receptors, Tachykinin/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Substance P/analogs & derivatives
- Substance P/pharmacology
- Tachykinins/metabolism
- Tetrodotoxin/pharmacology
- Tritium
- omega-N-Methylarginine/pharmacology
Collapse
Affiliation(s)
- Z Preston
- Parke-Davis Neuroscience Research Centre, Cambridge University Forvie Site, UK
| | | | | | | | | |
Collapse
|